1
|
Dong H, Wang X, Zheng Y, Li J, Liu Z, Wang A, Shen Y, Wu D, Cui H. Mapping the rapid growth of multi-omics in tumor immunotherapy: Bibliometric evidence of technology convergence and paradigm shifts. Hum Vaccin Immunother 2025; 21:2493539. [PMID: 40275437 PMCID: PMC12026087 DOI: 10.1080/21645515.2025.2493539] [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: 11/08/2024] [Revised: 04/01/2025] [Accepted: 04/11/2025] [Indexed: 04/26/2025] Open
Abstract
This study aims to fill the knowledge gap in systematically mapping the evolution of omics-driven tumor immunotherapy research through a bibliometric lens. While omics technologies (genomics, transcriptomics, proteomics, metabolomics)provide multidimensional molecular profiling, their synergistic potential with immunotherapy remains underexplored in large-scale trend analyses. A comprehensive search was conducted using the Web of Science Core Collection for literature related to omics in tumor immunotherapy, up to August 2024. Bibliometric analyses, conducted using R version 4.3.3, VOSviewer 1.6.20, and Citespace 6.2, examined publication trends, country and institutional contributions, journal distributions, keyword co-occurrence, and citation bursts. This analysis of 9,494 publications demonstrates rapid growth in omics-driven tumor immunotherapy research since 2019, with China leading in output (63% of articles) yet exhibiting limited multinational collaboration (7.9% vs. the UK's 61.8%). Keyword co-occurrence and citation burst analyses reveal evolving frontiers: early emphasis on "PD-1/CTLA-4 blockade" has transitioned toward "machine learning," "multi-omics," and "lncRNA," reflecting a shift to predictive modeling and biomarker discovery. Multi-omics integration has facilitated the development of immune infiltration-based prognostic models, such as TIME subtypes, which have been validated across multiple tumor types, which inform clinical trial design (e.g. NCT06833723). Additionally, proteomic analysis of melanoma patients suggests that metabolic biomarkers, particularly oxidative phosphorylation and lipid metabolism, may stratify responders to PD-1 blockade therapy. Moreover, spatial omics has confirmed ENPP1 as a potential novel therapeutic target in Ewing sarcoma. Citation trends underscore clinical translation, particularly mutation-guided therapies. Omics technologies are transforming tumor immunotherapy by enhancing biomarker discovery and improving therapeutic predictions. Future advancements will necessitate longitudinal omics monitoring, AI-driven multi-omics integration, and international collaboration to accelerate clinical translation. This study presents a systematic framework for exploring emerging research frontiers and offers insights for optimizing precision-driven immunotherapy.
Collapse
Affiliation(s)
- Huijing Dong
- China-Japan Friendship Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Xinmeng Wang
- China-Japan Friendship Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Yumin Zheng
- China-Japan Friendship Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Jia Li
- China-Japan Friendship Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Zhening Liu
- China-Japan Friendship Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Aolin Wang
- China-Japan Friendship Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Yulei Shen
- China-Japan Friendship Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Daixi Wu
- China-Japan Friendship Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Huijuan Cui
- Department of Integrative Oncology, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
2
|
Wen J, Cui W, Yin X, Chen Y, Liu A, Wang Q, Meng X. Application and future prospects of bispecific antibodies in the treatment of non-small cell lung cancer. Cancer Biol Med 2025; 22:j.issn.2095-3941.2024.0470. [PMID: 40192238 PMCID: PMC12032835 DOI: 10.20892/j.issn.2095-3941.2024.0470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 03/05/2025] [Indexed: 04/29/2025] Open
Abstract
As the leading cause of cancer-related deaths, lung cancer remains a noteworthy threat to human health. Although immunotherapies, such as immune checkpoint inhibitors (ICIs), have significantly increased the efficacy of lung cancer treatment, a significant percentage of patients are not sensitive to immunotherapies and patients who initially respond to treatment can quickly develop acquired drug resistance. Bispecific antibodies (bsAbs) bind two different antigens or epitopes simultaneously and have been shown to enhance antitumor efficacy with suitable safety profiles, thus attracting increasing attention as novel antitumor therapies. At present, in addition to the approved bsAb, amivantamab, three novel bsAbs (KN046, AK112, and SHR-1701) are being evaluated in phase 3 clinical trials and many bsAbs are being evaluated in phase 1/2 clinical trials for patients with non-small cell lung cancer (NSCLC). Herein we present the structure, classification, and mechanism of action underlying bsAbs in NSCLC and introduce related clinical trials. Finally, we discuss challenges, potential solutions, and future prospects in the context of cancer treatment with bsAbs.
Collapse
Affiliation(s)
- Junxu Wen
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Wenxing Cui
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Xiaoyan Yin
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Yu Chen
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Ailing Liu
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Qian Wang
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Xiangjiao Meng
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| |
Collapse
|
3
|
Wu Y, Zhang W, Liang X, Zhang P, Zhang M, Jiang Y, Cui Y, Chen Y, Zhou W, Liang Q, Dai J, Zhang C, Xu J, Li J, Yu T, Zhang Z, Guo R. Habitat radiomics analysis for progression free survival and immune-related adverse reaction prediction in non-small cell lung cancer treated by immunotherapy. J Transl Med 2025; 23:393. [PMID: 40181378 PMCID: PMC11970015 DOI: 10.1186/s12967-024-06057-y] [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: 09/09/2024] [Accepted: 12/25/2024] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is highly heterogeneous, leading to varied treatment responses and immune-related adverse reactions (irAEs) among patients. Habitat radiomics allows non-invasive quantitative assessment of intratumor heterogeneity (ITH). Therefore, our objective is to employ habitat radiomics techniques to develop a robust approach for predicting the efficacy of Immune checkpoint inhibitors (ICIs) and the likelihood of irAEs in advanced NSCLC patients. METHODS In this retrospective two center study, two independent cohorts of patients with NSCLC were used to develop (n = 248) and validate signatures (n = 95). After applying four kinds of machine learning algorithms to select the key preoperative CT radiomic features, we used clinical, radiomics and habitat radiomic features to develop the clinical signature, radiomics signature and habitat radiomic signature for ICIs prognostics and irAEs prediction. By combining habitat radiomic features with corresponding clinicopathologic information, the nomogram signature was constructed in the training cohort. Next, the internal validation cohort (n = 75) of patients, and the external validation cohort (n = 20) of patients treated with ICIs were included to evaluate the predictive value of the four signatures, and their predictive performance was assessed by the area under operating characteristic curve (AUC). RESULTS Our study introduces a radiomic nomogram model that integrates clinical and habitat radiomic features to identify patients who may benefit from ICIs or experience irAEs. The Radiomics Nomogram model exhibited superior predictive performance in the training, validation, and external validation sets, with AUCs of 0.923, 0.817, and 0.899, respectively. This model outperformed both the Whole-tumor Radiomics Signature model (AUCs of 0.870, 0.736, and 0.626) and the Habitat Signature model (AUCs of 0.900, 0.804, and 0.808). The radiomics model focusing on tumor sub-regional habitat showed better predictive performance than the model derived from the entire tumor. Decision Curve Analysis (DCA) and calibration curves confirmed the nomogram's effectiveness. CONCLUSION By leveraging machine learning to predict the outcomes of ICIs, we can move closer to achieving tailored ICIs for lung cancer. This advancement will assist physicians in selecting and managing subsequent treatment strategies, thereby facilitating clinical decision-making.
Collapse
Affiliation(s)
- Yuemin Wu
- Department of Oncology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Wei Zhang
- Department of Radiology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Xiao Liang
- Department of Oncology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Pengpeng Zhang
- Department of Lung Cancer Surgery, Tianjin Lung Cancer Institute, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Mengzhe Zhang
- Department of Lung Cancer Surgery, Tianjin Lung Cancer Institute, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yuqin Jiang
- Department of Oncology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Yanan Cui
- Department of Radiology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Yi Chen
- Department of Oncology, Pukou Branch of Jiangsu People's Hospital, Nanjing Pukou District Central Hospital, Nanjing, China
| | - Wenxin Zhou
- Department of Oncology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Qi Liang
- Department of Oncology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Jiali Dai
- Department of Oncology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Chen Zhang
- Department of Oncology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Jiali Xu
- Department of Oncology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Jun Li
- Department of Oncology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Tongfu Yu
- Department of Radiology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Zhihong Zhang
- Department of Pathology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Renhua Guo
- Department of Radiology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
4
|
Xu W, Wang Y, Wang N, Liu J, Zhou L, Guo J. Spatial immune remodeling of the liver metastases: discovering the path to antimetastatic therapy. J Immunother Cancer 2025; 13:e011002. [PMID: 40107672 PMCID: PMC11927485 DOI: 10.1136/jitc-2024-011002] [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: 11/06/2024] [Accepted: 03/10/2025] [Indexed: 03/22/2025] Open
Abstract
The intrinsic characteristics of metastatic tumors are of great importance in terms of the development of antimetastatic treatment strategies. Elucidation from a spatial immune perspective has the potential to provide a more comprehensive understanding of the mechanisms underlying immune escape, effectively addressing the limitations of relying solely on the analysis of immune cell subpopulation transcriptional profiles. Advances in spatial omics technology enable researchers to precisely analyze precious liver metastasis samples in a high-throughput manner, revealing spatial alterations in immune cell distribution induced by metastasis and exploring the molecular basis of the remodeling process. The aggregation of specific cell subpopulations in distinct regions not only modifies local immune characteristics but also concurrently affects global biological behaviors of liver metastatic tumors. Identifying specific spatial immune characteristics in pretreatment or early-stage treatment tissue samples may achieve accurate clinical predictions. Moreover, developing strategies that target spatial immune remodeling is a promising avenue for future antimetastatic therapy.
Collapse
Affiliation(s)
- Wenchao Xu
- Department of General Surgery, Peking Union Medical College Hospital, Beijing, Beijing, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, Beijing, China
- National Infrastructures for Translational Medicine, Peking Union Medical College Hospital, Beijing, Beijing, China
- State Key Laboratory of Complex, Severe, and Rare Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, Beijing, China
| | - Yibo Wang
- Department of General Surgery, Peking Union Medical College Hospital, Beijing, Beijing, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, Beijing, China
- National Infrastructures for Translational Medicine, Peking Union Medical College Hospital, Beijing, Beijing, China
- State Key Laboratory of Complex, Severe, and Rare Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, Beijing, China
| | - Nanzhou Wang
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Jianzhou Liu
- Department of General Surgery, Peking Union Medical College Hospital, Beijing, Beijing, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, Beijing, China
- National Infrastructures for Translational Medicine, Peking Union Medical College Hospital, Beijing, Beijing, China
- State Key Laboratory of Complex, Severe, and Rare Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, Beijing, China
| | - Li Zhou
- Department of General Surgery, Peking Union Medical College Hospital, Beijing, Beijing, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, Beijing, China
- National Infrastructures for Translational Medicine, Peking Union Medical College Hospital, Beijing, Beijing, China
- State Key Laboratory of Complex, Severe, and Rare Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, Beijing, China
| | - Junchao Guo
- Department of General Surgery, Peking Union Medical College Hospital, Beijing, Beijing, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, Beijing, China
- National Infrastructures for Translational Medicine, Peking Union Medical College Hospital, Beijing, Beijing, China
- State Key Laboratory of Complex, Severe, and Rare Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, Beijing, China
| |
Collapse
|
5
|
Guan A, Quek C. Single-Cell Multi-Omics: Insights into Therapeutic Innovations to Advance Treatment in Cancer. Int J Mol Sci 2025; 26:2447. [PMID: 40141092 PMCID: PMC11942442 DOI: 10.3390/ijms26062447] [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: 02/06/2025] [Revised: 03/04/2025] [Accepted: 03/07/2025] [Indexed: 03/28/2025] Open
Abstract
Advances in single-cell multi-omics technologies have deepened our understanding of cancer biology by integrating genomic, transcriptomic, epigenomic, and proteomic data at single-cell resolution. These single-cell multi-omics technologies provide unprecedented insights into tumour heterogeneity, tumour microenvironment, and mechanisms of therapeutic resistance, enabling the development of precision medicine strategies. The emerging field of single-cell multi-omics in genomic medicine has improved patient outcomes. However, most clinical applications still depend on bulk genomic approaches, which fail to directly capture the genomic variations driving cellular heterogeneity. In this review, we explore the common single-cell multi-omics platforms and discuss key analytical steps for data integration. Furthermore, we highlight emerging knowledge in therapeutic resistance and immune evasion, and the potential of new therapeutic innovations informed by single-cell multi-omics. Finally, we discuss the future directions of the application of single-cell multi-omics technologies. By bridging the gap between technological advancements and clinical implementation, this review provides a roadmap for leveraging single-cell multi-omics to improve cancer treatment and patient outcomes.
Collapse
Affiliation(s)
- Angel Guan
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW 2065, Australia;
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia
| | - Camelia Quek
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW 2065, Australia;
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
6
|
Torre-Cea I, Berlana-Galán P, Guerra-Paes E, Cáceres-Calle D, Carrera-Aguado I, Marcos-Zazo L, Sánchez-Juanes F, Muñoz-Félix JM. Basement membranes in lung metastasis growth and progression. Matrix Biol 2025; 135:135-152. [PMID: 39719224 DOI: 10.1016/j.matbio.2024.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/19/2024] [Accepted: 12/20/2024] [Indexed: 12/26/2024]
Abstract
The lung is a highly vascularized tissue that often harbors metastases from various extrathoracic malignancies. Lung parenchyma consists of a complex network of alveolar epithelial cells and microvessels, structured within an architecture defined by basement membranes. Consequently, understanding the role of the extracellular matrix (ECM) in the growth of lung metastases is essential to uncover the biology of this pathology and developing targeted therapies. These basement membranes play a critical role in the progression of lung metastases, influencing multiple stages of the metastatic cascade, from the acquisition of an aggressive phenotype to intravasation, extravasation and colonization of secondary sites. This review examines the biological composition of basement membranes, focusing on their core components-collagens, fibronectin, and laminin-and their specific roles in cancer progression. Additionally, we discuss the function of integrins as primary mediators of cell adhesion and signaling between tumor cells, basement membranes and the extracellular matrix, as well as their implications for metastatic growth in the lung. We also explore vascular co-option (VCO) as a form of tumor growth resistance linked to basement membranes and tumor vasculature. Finally, the review covers current clinical therapies targeting tumor adhesion, extracellular matrix remodeling, and vascular development, aiming to improve the precision and effectiveness of treatments against lung metastases.
Collapse
Affiliation(s)
- Irene Torre-Cea
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Spain
| | - Patricia Berlana-Galán
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Spain
| | - Elena Guerra-Paes
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Spain
| | - Daniel Cáceres-Calle
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Spain
| | - Iván Carrera-Aguado
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Spain
| | - Laura Marcos-Zazo
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Spain
| | - Fernando Sánchez-Juanes
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Spain.
| | - José M Muñoz-Félix
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Spain.
| |
Collapse
|
7
|
Qiao T, Cheng Z, Duan Y. Innovative applications and future trends of multiparametric PET in the assessment of immunotherapy efficacy. Front Oncol 2025; 14:1530507. [PMID: 39902124 PMCID: PMC11788151 DOI: 10.3389/fonc.2024.1530507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 12/23/2024] [Indexed: 02/05/2025] Open
Abstract
Background The integration of multiparametric PET (Positron Emission Tomography.) imaging and multi-omics data has demonstrated significant clinical potential in predicting the efficacy of cancer immunotherapies. However, the specific predictive power and underlying mechanisms remain unclear. Objective This review systematically evaluates the application of multiparametric PET imaging metrics (e.g., SUVmax [Maximum Standardized Uptake Value], MTV [Metabolic Tumor Volume], and TLG [Total Lesion Glycolysis]) in predicting the efficacy of immunotherapies, including PD-1/PD-L1 inhibitors and CAR-T therapy, and explores their potential role in improving predictive accuracy when integrated with multi-omics data. Methods A systematic search of PubMed, Embase, and Web of Science databases identified studies evaluating the efficacy of immunotherapy using longitudinal PET/CT data and RECIST or iRECIST criteria. Only original prospective or retrospective studies were included for analysis. Review articles and meta-analyses were consulted for additional references but excluded from quantitative analysis. Studies lacking standardized efficacy evaluations were excluded to ensure data integrity and quality. Results Multiparametric PET imaging metrics exhibited high predictive capability for efficacy across various immunotherapies. Metabolic parameters such as SUVmax, MTV, and TLG were significantly correlated with treatment response rates, progression-free survival (PFS), and overall survival (OS). The integration of multi-omics data (including genomics and proteomics) with PET imaging enhanced the sensitivity and accuracy of efficacy prediction. Through integrated analysis, PET metabolic parameters demonstrated potential in predicting immune therapy response patterns, such as pseudo-progression and hyper-progression. Conclusion The integration of multiparametric PET imaging and multi-omics data holds broad potential for predicting the efficacy of immunotherapies and may support the development of personalized treatment strategies. Future validation using large-scale, multicenter datasets is needed to further advance precision medicine in cancer immunotherapy.
Collapse
Affiliation(s)
- Tingting Qiao
- Department of Nuclear Medicine, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
- Graduate School, Shandong First Medical University, Jinan, China
| | - Zhaoping Cheng
- Department of Nuclear Medicine, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Yanhua Duan
- Department of Nuclear Medicine, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| |
Collapse
|
8
|
Chen Z, Mei K, Tan F, Zhou Y, Du H, Wang M, Gu R, Huang Y. Integrative multi-omics analysis for identifying novel therapeutic targets and predicting immunotherapy efficacy in lung adenocarcinoma. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2025; 8:3. [PMID: 39935429 PMCID: PMC11810459 DOI: 10.20517/cdr.2024.91] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 12/18/2024] [Accepted: 12/31/2024] [Indexed: 02/13/2025]
Abstract
Aim: Lung adenocarcinoma (LUAD), the most prevalent subtype of non-small cell lung cancer (NSCLC), presents significant clinical challenges due to its high mortality and limited therapeutic options. The molecular heterogeneity and the development of therapeutic resistance further complicate treatment, underscoring the need for a more comprehensive understanding of its cellular and molecular characteristics. This study sought to delineate novel cellular subpopulations and molecular subtypes of LUAD, identify critical biomarkers, and explore potential therapeutic targets to enhance treatment efficacy and patient prognosis. Methods: An integrative multi-omics approach was employed to incorporate single-cell RNA sequencing (scRNA-seq), bulk transcriptomic analysis, and genome-wide association study (GWAS) data from multiple LUAD patient cohorts. Advanced computational approaches, including Bayesian deconvolution and machine learning algorithms, were used to comprehensively characterize the tumor microenvironment, classify LUAD subtypes, and develop a robust prognostic model. Results: Our analysis identified eleven distinct cellular subpopulations within LUAD, with epithelial cells predominating and exhibiting high mutation frequencies in Tumor Protein 53 (TP53) and Titin (TTN) genes. Two molecular subtypes of LUAD [consensus subtype (CS)1 and CS2] were identified, each showing distinct immune landscapes and clinical outcomes. The CS2 subtype, characterized by increased immune cell infiltration, demonstrated a more favorable prognosis and higher sensitivity to immunotherapy. Furthermore, a multi-omics-driven machine learning signature (MOMLS) identified ribonucleotide reductase M1 (RRM1) as a critical biomarker associated with chemotherapy response. Based on this model, several potential therapeutic agents targeting different subtypes were proposed. Conclusion: This study presents a comprehensive multi-omics framework for understanding the molecular complexity of LUAD, providing insights into cellular heterogeneity, molecular subtypes, and potential therapeutic targets. Differential sensitivity to immunotherapy among various cellular subpopulations was identified, paving the way for future immunotherapy-focused research.
Collapse
Affiliation(s)
- Zilu Chen
- Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
- Authors contributed equally
| | - Kun Mei
- Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
- Department of Cardiothoracic Surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213003, Jiangsu, China
- Authors contributed equally
| | - Foxing Tan
- Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| | - Yuheng Zhou
- Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| | - Haolin Du
- Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| | - Min Wang
- Department of Cardiothoracic Surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213003, Jiangsu, China
| | - Renjun Gu
- School of Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
- Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210046, Jiangsu, China
| | - Yan Huang
- Department of Ultrasound, Nanjing Hospital of Chinese Medicine Affiliated with Nanjing University of Chinese Medicine, Nanjing 210022, Jiangsu, China
| |
Collapse
|
9
|
Zeng F, Zhang Y, Luo T, Wang C, Fu D, Wang X. Daidzein Inhibits Non-small Cell Lung Cancer Growth by Pyroptosis. Curr Pharm Des 2025; 31:884-924. [PMID: 39623715 DOI: 10.2174/0113816128330530240918073721] [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: 07/01/2024] [Accepted: 08/08/2024] [Indexed: 04/24/2025]
Abstract
INTRODUCTION Non-small cell lung cancer (NSCLC) represents the leading cause of cancer deaths in the world. We previously found that daidzein, one of the key bioactivators in soy isoflavone, can inhibit NSCLC cell proliferation and migration, while the molecular mechanisms of daidzein in NSCLC remain unclear. METHODS We developed an NSCLC nude mouse model using H1299 cells and treated the mice with daidzein (30 mg/kg/day). Mass spectrometry analysis of tumor tissues from daidzein-treated mice identified 601 differentially expressed proteins (DEPs) compared to the vehicle-treated group. Gene enrichment analysis revealed that these DEPs were primarily associated with immune regulatory functions, including B cell receptor and chemokine pathways, as well as natural killer cell-mediated cytotoxicity. Notably, the NOD-like receptor signaling pathway, which is closely linked to pyroptosis, was significantly enriched. RESULTS Further analysis of key pyroptosis-related molecules, such as ASC, CASP1, GSDMD, and IL-1β, revealed differential expression in NSCLC versus normal tissues. High levels of ASC and CASP1 were associated with a favorable prognosis in NSCLC, suggesting that they may be critical effectors of daidzein's action. In NSCLC-bearing mice treated with daidzein, RT-qPCR and Western blot analyses showed elevated mRNA and protein levels of ASC, CASP1, and IL-1β but not GSDMD, which was consistent with the proteomic data. CONCLUSION In summary, this study demonstrated that daidzein inhibits NSCLC growth by inducing pyroptosis. Key pathway modulators ASC, CASP1, and IL-1β were identified as primary targets of daidzein. These findings offer insights into the molecular mechanisms underlying the anti-NSCLC effects of daidzein and could offer dietary recommendations for managing NSCLC.
Collapse
Affiliation(s)
- Fanfan Zeng
- Jiangxi Provincial Key Laboratory of Cell Precision Therapy, School of Basic Medical Sciences, Jiujiang University, Jiujiang, 332005, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Preventive Medicine, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yu Zhang
- Jiangxi Provincial Key Laboratory of Cell Precision Therapy, School of Basic Medical Sciences, Jiujiang University, Jiujiang, 332005, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Preventive Medicine, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Ting Luo
- Jiangxi Provincial Key Laboratory of Cell Precision Therapy, School of Basic Medical Sciences, Jiujiang University, Jiujiang, 332005, Jiangxi, China
- Department of Infection Control, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Chengman Wang
- Jiangxi Provincial Key Laboratory of Cell Precision Therapy, School of Basic Medical Sciences, Jiujiang University, Jiujiang, 332005, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Preventive Medicine, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Denggang Fu
- College of Medicine, Medical University of South Carolina, Columbia, Charleston, SC 29425, United States
| | - Xin Wang
- Jiangxi Provincial Key Laboratory of Cell Precision Therapy, School of Basic Medical Sciences, Jiujiang University, Jiujiang, 332005, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Preventive Medicine, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
10
|
Kiseleva OI, Arzumanian VA, Ikhalaynen YA, Kurbatov IY, Kryukova PA, Poverennaya EV. Multiomics of Aging and Aging-Related Diseases. Int J Mol Sci 2024; 25:13671. [PMID: 39769433 PMCID: PMC11677528 DOI: 10.3390/ijms252413671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 12/03/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
Despite their astonishing biological diversity, surprisingly few shared traits connect all or nearly all living organisms. Aging, i.e., the progressive and irreversible decline in the function of multiple cells and tissues, is one of these fundamental features of all organisms, ranging from single-cell creatures to complex animals, alongside variability, adaptation, growth, healing, reproducibility, mobility, and, finally, death. Age is a key determinant for many pathologies, shaping the risks of incidence, severity, and treatment outcomes for cancer, neurodegeneration, heart failure, sarcopenia, atherosclerosis, osteoporosis, and many other diseases. In this review, we aim to systematically investigate the age-related features of the development of several diseases through the lens of multiomics: from genome instability and somatic mutations to pathway alterations and dysregulated metabolism.
Collapse
Affiliation(s)
- Olga I. Kiseleva
- Institute of Biomedical Chemistry, Pogodinskaya Street, 10/8, 119121 Moscow, Russia; (V.A.A.); (Y.A.I.); (I.Y.K.); (P.A.K.); (E.V.P.)
| | - Viktoriia A. Arzumanian
- Institute of Biomedical Chemistry, Pogodinskaya Street, 10/8, 119121 Moscow, Russia; (V.A.A.); (Y.A.I.); (I.Y.K.); (P.A.K.); (E.V.P.)
| | - Yuriy A. Ikhalaynen
- Institute of Biomedical Chemistry, Pogodinskaya Street, 10/8, 119121 Moscow, Russia; (V.A.A.); (Y.A.I.); (I.Y.K.); (P.A.K.); (E.V.P.)
- Chemistry Department, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Ilya Y. Kurbatov
- Institute of Biomedical Chemistry, Pogodinskaya Street, 10/8, 119121 Moscow, Russia; (V.A.A.); (Y.A.I.); (I.Y.K.); (P.A.K.); (E.V.P.)
| | - Polina A. Kryukova
- Institute of Biomedical Chemistry, Pogodinskaya Street, 10/8, 119121 Moscow, Russia; (V.A.A.); (Y.A.I.); (I.Y.K.); (P.A.K.); (E.V.P.)
| | - Ekaterina V. Poverennaya
- Institute of Biomedical Chemistry, Pogodinskaya Street, 10/8, 119121 Moscow, Russia; (V.A.A.); (Y.A.I.); (I.Y.K.); (P.A.K.); (E.V.P.)
| |
Collapse
|
11
|
Lubo I, Hernandez S, Wistuba II, Solis Soto LM. Novel Spatial Approaches to Dissect the Lung Cancer Immune Microenvironment. Cancers (Basel) 2024; 16:4145. [PMID: 39766047 PMCID: PMC11674389 DOI: 10.3390/cancers16244145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/07/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
Lung cancer is a deadly disease with the highest rates of mortality. Over recent decades, a better understanding of the biological mechanisms implicated in its pathogenesis has led to the development of targeted therapies and immunotherapy, resulting in improvements in patient outcomes. To better understand lung cancer tumor biology and advance towards precision oncology, a comprehensive tumor profile is necessary. In recent years, novel in situ spatial multiomics approaches have emerged offering a more detailed view of the spatial location of tumor and tumor microenvironment cells, identifying their unique composition and functional status. In this sense, novel multiomics platforms have been developed to evaluate tumor heterogeneity, gene expression, metabolic reprogramming, signaling pathway activation, cell-cell interactions, and immune cell programs. In lung cancer research, several studies have used these spatial technologies to locate cells and associated them with histological features that are relevant to the pathogenesis of lung adenocarcinoma. These advancements may unveil further molecular and immune mechanisms in tumor biology that will lead to the discovery of biomarkers for treatment prediction and prognosis. In this review, we provide an overview of more widely used and emerging pathology-based approaches for spatial immune profiling in lung cancer and how they enhance our understanding of tumor biology and immune response.
Collapse
Affiliation(s)
| | | | | | - Luisa Maren Solis Soto
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (I.L.); (S.H.); (I.I.W.)
| |
Collapse
|
12
|
Qin S, Zhang H, Liu C, Yi M. Editorial: Investigating tumor immunotherapy responses in lung cancer using deep learning. Front Immunol 2024; 15:1529949. [PMID: 39691722 PMCID: PMC11649539 DOI: 10.3389/fimmu.2024.1529949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 11/19/2024] [Indexed: 12/19/2024] Open
Affiliation(s)
- Shuang Qin
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haoxiang Zhang
- Department of Hepatopancreatobiliary Surgery, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
| | - Chao Liu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Ming Yi
- Department of Breast Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
13
|
Xu J, Tang Z. Progress on angiogenic and antiangiogenic agents in the tumor microenvironment. Front Oncol 2024; 14:1491099. [PMID: 39629004 PMCID: PMC11611712 DOI: 10.3389/fonc.2024.1491099] [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: 09/04/2024] [Accepted: 10/31/2024] [Indexed: 12/06/2024] Open
Abstract
The development of tumors and their metastasis relies heavily on the process of angiogenesis. When the volume of a tumor expands, the resulting internal hypoxic conditions trigger the body to enhance the production of various angiogenic factors. These include vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF), platelet-derived growth factor (PDGF), and transforming growth factor-α (TGF-α), all of which work together to stimulate the activation of endothelial cells and catalyze angiogenesis. Antiangiogenic therapy (AAT) aims to normalize tumor blood vessels by inhibiting these angiogenic signals. In this review, we will explore the molecular mechanisms of angiogenesis within the tumor microenvironment, discuss traditional antiangiogenic drugs along with their limitations, examine new antiangiogenic drugs and the advantages of combination therapy, and consider future research directions in the field of antiangiogenic drugs. This comprehensive overview aims to provide insights that may aid in the development of more effective anti-tumor treatments.
Collapse
Affiliation(s)
| | - Zhihua Tang
- Department of Pharmacy, Shaoxing People’s Hospital, Shaoxing, China
| |
Collapse
|
14
|
Kwak Y, Nam SK, Park Y, Suh YS, Ahn SH, Kong SH, Park DJ, Lee HJ, Kim HH, Yang HK, Lee HS. Distinctive Phenotypic and Microenvironmental Characteristics of Neuroendocrine Carcinoma and Adenocarcinoma Components in Gastric Mixed Adenoneuroendocrine Carcinoma. Mod Pathol 2024; 37:100568. [PMID: 39029904 DOI: 10.1016/j.modpat.2024.100568] [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: 04/02/2024] [Revised: 06/30/2024] [Accepted: 07/11/2024] [Indexed: 07/21/2024]
Abstract
This study aimed to conduct an in-depth examination of gene expression and microenvironmental profiles of gastric neuroendocrine carcinoma (NEC) and mixed adeno-NEC (MANEC). Tissue microarrays from 55 patients with gastric MANEC (N = 32) or NEC (N = 23) were analyzed using digital spatial profiling (GeoMx DSP, NanoString Technologies). Representative regions of interest were selected from the adenocarcinoma (ADC) portion (ADC-MANEC) and the NEC portion (NEC-MANEC) of the MANEC cores, and pure NEC (pNEC) cores. All regions of interest were separated into epithelial components and stromal components using the masking procedure in the GeoMx platform, followed by transcriptome analysis. Comparison of gene expression between ADC-MANEC and NEC-MANEC/pNEC identified several differentially expressed genes in the epithelial (including PEG10, MAP1B, STMN3, and AKT3) and stromal (FN1, COL1A1, SPARC, and BGN) components. Gene set enrichment analysis revealed that pathways related to the E2F target and G2M checkpoint were more enriched in NEC-MANEC and pNEC than in ADC-MANEC. Deconvolution analysis showed that the microenvironmental profile varied according to histologic differentiation. In ADC-MANEC, intraepithelial infiltrating immune cells were relatively more numerous, whereas fibroblasts in the stroma were more abundant in NEC-MANEC and pNEC. This study confirmed the distinct expression profile of each histologic component of MANEC according to its tumor vs stromal compartment using the DSP platform. Although each component of MANEC shares the same genetic origin, distinctive phenotypes should not be overlooked when managing patients with MANEC. This study provides a useful validation data set for future studies.
Collapse
Affiliation(s)
- Yoonjin Kwak
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
| | - Soo Kyung Nam
- Department of Interdisciplinary Program in Cancer Biology, Seoul National University College of Medicine, Seoul, Korea
| | - Yujun Park
- Department of Pathology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Korea
| | - Yun-Suhk Suh
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Sang-Hoon Ahn
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Seong-Ho Kong
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea; Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Do Joong Park
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea; Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Hyuk-Joon Lee
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea; Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Hyung-Ho Kim
- Department of Surgery, Chung-Ang University College of Medicine, Seoul, Korea
| | - Han-Kwang Yang
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea; Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Hye Seung Lee
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea; Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
15
|
Wu M, Tao H, Xu T, Zheng X, Wen C, Wang G, Peng Y, Dai Y. Spatial proteomics: unveiling the multidimensional landscape of protein localization in human diseases. Proteome Sci 2024; 22:7. [PMID: 39304896 DOI: 10.1186/s12953-024-00231-2] [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: 04/29/2024] [Accepted: 09/01/2024] [Indexed: 09/22/2024] Open
Abstract
Spatial proteomics is a multidimensional technique that studies the spatial distribution and function of proteins within cells or tissues across both spatial and temporal dimensions. This field multidimensionally reveals the complex structure of the human proteome, including the characteristics of protein spatial distribution, dynamic protein translocation, and protein interaction networks. Recently, as a crucial method for studying protein spatial localization, spatial proteomics has been applied in the clinical investigation of various diseases. This review summarizes the fundamental concepts and characteristics of tissue-level spatial proteomics, its research progress in common human diseases such as cancer, neurological disorders, cardiovascular diseases, autoimmune diseases, and anticipates its future development trends. The aim is to highlight the significant impact of spatial proteomics on understanding disease pathogenesis, advancing diagnostic methods, and developing potential therapeutic targets in clinical research.
Collapse
Affiliation(s)
- Mengyao Wu
- School of Medicine, Anhui University of Science & Technology, Huainan, China
| | - Huihui Tao
- School of Medicine, Anhui University of Science & Technology, Huainan, China.
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, Huainan, China.
- Anhui Province Engineering Laboratory of Occupational Health and Safety, Huainan, China.
| | - Tiantian Xu
- School of Medicine, Anhui University of Science & Technology, Huainan, China
| | - Xuejia Zheng
- The First Hospital of Anhui University of Science and Technology, Huainan, China
| | - Chunmei Wen
- School of Medicine, Anhui University of Science & Technology, Huainan, China
| | - Guoying Wang
- School of Medicine, Anhui University of Science & Technology, Huainan, China
| | - Yali Peng
- School of Medicine, Anhui University of Science & Technology, Huainan, China
| | - Yong Dai
- School of Medicine, Anhui University of Science & Technology, Huainan, China
- The First Hospital of Anhui University of Science and Technology, Huainan, China
- Joint Research Center for Occupational Medicine and Health of IHM, Anhui University of Science and Technology, Huainan, China
| |
Collapse
|
16
|
Liang W, Zhu Z, Xu D, Wang P, Guo F, Xiao H, Hou C, Xue J, Zhi X, Ran R. The burgeoning spatial multi-omics in human gastrointestinal cancers. PeerJ 2024; 12:e17860. [PMID: 39285924 PMCID: PMC11404479 DOI: 10.7717/peerj.17860] [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: 03/27/2024] [Accepted: 07/14/2024] [Indexed: 09/19/2024] Open
Abstract
The development and progression of diseases in multicellular organisms unfold within the intricate three-dimensional body environment. Thus, to comprehensively understand the molecular mechanisms governing individual development and disease progression, precise acquisition of biological data, including genome, transcriptome, proteome, metabolome, and epigenome, with single-cell resolution and spatial information within the body's three-dimensional context, is essential. This foundational information serves as the basis for deciphering cellular and molecular mechanisms. Although single-cell multi-omics technology can provide biological information such as genome, transcriptome, proteome, metabolome, and epigenome with single-cell resolution, the sample preparation process leads to the loss of spatial information. Spatial multi-omics technology, however, facilitates the characterization of biological data, such as genome, transcriptome, proteome, metabolome, and epigenome in tissue samples, while retaining their spatial context. Consequently, these techniques significantly enhance our understanding of individual development and disease pathology. Currently, spatial multi-omics technology has played a vital role in elucidating various processes in tumor biology, including tumor occurrence, development, and metastasis, particularly in the realms of tumor immunity and the heterogeneity of the tumor microenvironment. Therefore, this article provides a comprehensive overview of spatial transcriptomics, spatial proteomics, and spatial metabolomics-related technologies and their application in research concerning esophageal cancer, gastric cancer, and colorectal cancer. The objective is to foster the research and implementation of spatial multi-omics technology in digestive tumor diseases. This review will provide new technical insights for molecular biology researchers.
Collapse
Affiliation(s)
- Weizheng Liang
- Central Laboratory, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei province, China
| | - Zhenpeng Zhu
- Department of Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei Province, China
- Hebei North University, Zhangjiakou, Hebei Province, China
| | - Dandan Xu
- Central Laboratory, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei province, China
| | - Peng Wang
- Department of Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei Province, China
- Hebei North University, Zhangjiakou, Hebei Province, China
| | - Fei Guo
- Department of Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei Province, China
| | - Haoshan Xiao
- Department of Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei Province, China
- Hebei North University, Zhangjiakou, Hebei Province, China
| | - Chenyang Hou
- Department of Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei Province, China
- Hebei North University, Zhangjiakou, Hebei Province, China
| | - Jun Xue
- Department of Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei Province, China
| | - Xuejun Zhi
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei province, China
| | - Rensen Ran
- Central Laboratory, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei province, China
- Department of Chemical Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
17
|
Chang SH, Mezzano-Robinson V, Zhou H, Moreira A, Pillai R, Ramaswami S, Loomis C, Heguy A, Tsirigos A, Pass HI. Digital spatial profiling to predict recurrence in grade 3 stage I lung adenocarcinoma. J Thorac Cardiovasc Surg 2024; 168:648-657.e8. [PMID: 37890657 DOI: 10.1016/j.jtcvs.2023.10.047] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 09/18/2023] [Accepted: 10/21/2023] [Indexed: 10/29/2023]
Abstract
OBJECTIVE Early-stage lung adenocarcinoma is treated with local therapy alone, although patients with grade 3 stage I lung adenocarcinoma have a 50% 5-year recurrence rate. Our objective is to determine if analysis of the tumor microenvironment can create a predictive model for recurrence. METHODS Thirty-four patients with grade 3 stage I lung adenocarcinoma underwent surgical resection. Digital spatial profiling was used to perform genomic (n = 31) and proteomic (n = 34) analyses of pancytokeratin positive and negative tumor cells. K-means clustering was performed on the top 50 differential genes and top 20 differential proteins, with Kaplan-Meier recurrence curves based on patient clustering. External validation of high-expression genes was performed with Kaplan-Meier plotter. RESULTS There were no significant clinicopathologic differences between patients who did (n = 14) and did not (n = 20) have recurrence. Median time to recurrence was 806 days; median follow-up with no recurrence was 2897 days. K-means clustering of pancytokeratin positive genes resulted in a model with a Kaplan-Meier curve with concordance index of 0.75. K-means clustering for pancytokeratin negative genes was less successful at differentiating recurrence (concordance index 0.6). Genes upregulated or downregulated for recurrence were externally validated using available public databases. Proteomic data did not reach statistical significance but did internally validate the genomic data described. CONCLUSIONS Genomic difference in lung adenocarcinoma may be able to predict risk of recurrence. After further validation, stratifying patients by this risk may help guide who will benefit from adjuvant therapy.
Collapse
Affiliation(s)
- Stephanie H Chang
- Department of Cardiothoracic Surgery, NYU Langone Health, New York, NY.
| | - Valeria Mezzano-Robinson
- Experimental Pathology Research Laboratory, Department of Pathology, NYU Langone Health, New York, NY
| | - Hua Zhou
- Department of Pathology, Applied Bioinformatics Laboratory, NYU Langone Health, New York, NY
| | - Andre Moreira
- Department of Pathology, Center for Biomarker Research and Development, NYU Langone Health, New York, NY
| | - Raymond Pillai
- Division of Pulmonary Critical Care, Department of Medicine, NYU Langone Health, New York, NY
| | - Sitharam Ramaswami
- Department of Pathology, Genome Technology Center, NYU Langone Health, New York, NY
| | - Cynthia Loomis
- Experimental Pathology Research Laboratory, Department of Pathology, NYU Langone Health, New York, NY
| | - Adriana Heguy
- Department of Pathology, Genome Technology Center, NYU Langone Health, New York, NY
| | - Aristotelis Tsirigos
- Department of Pathology, Applied Bioinformatics Laboratory, NYU Langone Health, New York, NY
| | - Harvey I Pass
- Department of Cardiothoracic Surgery, NYU Langone Health, New York, NY
| |
Collapse
|
18
|
Charoensuksira S, Tantiwong S, Pongklaokam J, Hanvivattanakul S, Surinlert P, Krajarng A, Thanasarnaksorn W, Hongeng S, Ponnikorn S. Disturbance of Immune Microenvironment in Androgenetic Alopecia through Spatial Transcriptomics. Int J Mol Sci 2024; 25:9031. [PMID: 39201715 PMCID: PMC11354591 DOI: 10.3390/ijms25169031] [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: 06/03/2024] [Revised: 08/11/2024] [Accepted: 08/15/2024] [Indexed: 09/03/2024] Open
Abstract
Androgenetic alopecia (AGA) is characterized by microinflammation and abnormal immune responses, particularly in the upper segment of hair follicles (HFs). However, the precise patterns of immune dysregulation remain unclear, partly due to limitations in current analysis techniques to preserve tissue architecture. The infundibulum, a major part of the upper segment of HFs, is associated with significant clusters of immune cells. In this study, we investigated immune cells around the infundibulum, referred to as peri-infundibular immune infiltration (PII). We employed spatial transcriptome profiling, a high-throughput analysis technology, to investigate the immunological disruptions within the PII region. Our comprehensive analysis included an evaluation of overall immune infiltrates, gene set enrichment analysis (GSEA), cellular deconvolution, differential expression analysis, over-representation analysis, protein-protein interaction (PPI) networks, and upstream regulator analysis to identify cell types and molecular dysregulation in immune cells. Our results demonstrated significant differences in immune signatures between the PII of AGA patients (PII-A) and the PII of control donors (PII-C). Specifically, PII-A exhibited an enrichment of CD4+ helper T cells, distinct immune response patterns, and a bias toward a T helper (Th) 2 response. Immunohistochemistry revealed disruptions in T cell subpopulations, with more CD4+ T cells displaying an elevated Th2 response and a reduced Th1-cytotoxic response compared to PII-C. These findings reveal the unique immune landscapes of PII-A and PII-C, suggesting potential for the development of innovative treatment approaches.
Collapse
Affiliation(s)
- Sasin Charoensuksira
- Division of Dermatology, Chulabhorn International College of Medicine, Thammasat University, Pathum Thani 12120, Thailand; (S.C.); (S.T.); (J.P.); (W.T.)
| | - Supasit Tantiwong
- Division of Dermatology, Chulabhorn International College of Medicine, Thammasat University, Pathum Thani 12120, Thailand; (S.C.); (S.T.); (J.P.); (W.T.)
| | - Juthapa Pongklaokam
- Division of Dermatology, Chulabhorn International College of Medicine, Thammasat University, Pathum Thani 12120, Thailand; (S.C.); (S.T.); (J.P.); (W.T.)
| | - Sirashat Hanvivattanakul
- Chulabhorn International College of Medicine, Thammasat University, Pathum Thani 12120, Thailand; (S.H.); (P.S.); (A.K.)
| | - Piyaporn Surinlert
- Chulabhorn International College of Medicine, Thammasat University, Pathum Thani 12120, Thailand; (S.H.); (P.S.); (A.K.)
- Research Unit in Synthesis and Applications of Graphene, Thammasat University, Pathum Thani 12120, Thailand
| | - Aungkana Krajarng
- Chulabhorn International College of Medicine, Thammasat University, Pathum Thani 12120, Thailand; (S.H.); (P.S.); (A.K.)
| | - Wilai Thanasarnaksorn
- Division of Dermatology, Chulabhorn International College of Medicine, Thammasat University, Pathum Thani 12120, Thailand; (S.C.); (S.T.); (J.P.); (W.T.)
- Division of Dermatology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Suradej Hongeng
- Division of Hematology and Oncology, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand;
| | - Saranyoo Ponnikorn
- Division of Dermatology, Chulabhorn International College of Medicine, Thammasat University, Pathum Thani 12120, Thailand; (S.C.); (S.T.); (J.P.); (W.T.)
- Chulabhorn International College of Medicine, Thammasat University, Pathum Thani 12120, Thailand; (S.H.); (P.S.); (A.K.)
- Thammasat University, Pattaya Campus, Bang Lamung 20150, Thailand
| |
Collapse
|
19
|
Williams HL, Frei AL, Koessler T, Berger MD, Dawson H, Michielin O, Zlobec I. The current landscape of spatial biomarkers for prediction of response to immune checkpoint inhibition. NPJ Precis Oncol 2024; 8:178. [PMID: 39138341 PMCID: PMC11322473 DOI: 10.1038/s41698-024-00671-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 08/05/2024] [Indexed: 08/15/2024] Open
Abstract
Enabling the examination of cell-cell relationships in tissue, spatially resolved omics technologies have revolutionised our perspectives on cancer biology. Clinically, the development of immune checkpoint inhibitors (ICI) has advanced cancer therapeutics. However, a major challenge of effective implementation is the identification of predictive biomarkers of response. In this review we examine the potential added predictive value of spatial biomarkers of response to ICI beyond current clinical benchmarks.
Collapse
Affiliation(s)
- Hannah L Williams
- Institute for Tissue Medicine and Pathology, University of Bern, Bern, Switzerland.
| | - Ana Leni Frei
- Institute for Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Thibaud Koessler
- Medical Oncology Department, Geneva University Hospitals, 4 rue Gabrielle-Perret-Gentil, 1205, Geneva, Switzerland
- Swiss Cancer Centre Léman, Lausanne, Geneva, Switzerland
- University of Geneva, Faculty of Medicine, Geneva, Switzerland
| | - Martin D Berger
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Heather Dawson
- Institute for Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| | - Olivier Michielin
- Medical Oncology Department, Geneva University Hospitals, 4 rue Gabrielle-Perret-Gentil, 1205, Geneva, Switzerland
- Swiss Cancer Centre Léman, Lausanne, Geneva, Switzerland
- University of Geneva, Faculty of Medicine, Geneva, Switzerland
| | - Inti Zlobec
- Institute for Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| |
Collapse
|
20
|
Qi C, Li Y, Zeng H, Wei Q, Tan S, Zhang Y, Li W, Tian P. Current status and progress of PD-L1 detection: guiding immunotherapy for non-small cell lung cancer. Clin Exp Med 2024; 24:162. [PMID: 39026109 PMCID: PMC11258158 DOI: 10.1007/s10238-024-01404-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 06/14/2024] [Indexed: 07/20/2024]
Abstract
Non-small cell lung cancer (NSCLC) is the leading cause of cancer-related deaths and represents a substantial disease burden worldwide. Immune checkpoint inhibitors combined with chemotherapy are the standard first-line therapy for advanced NSCLC without driver mutations. Programmed death-ligand 1 (PD-L1) is currently the only approved immunotherapy marker. PD-L1 detection methods are diverse and have developed rapidly in recent years, such as improved immunohistochemical detection methods, the application of liquid biopsy in PD-L1 detection, genetic testing, radionuclide imaging, and the use of machine learning methods to construct PD-L1 prediction models. This review focuses on the detection methods and challenges of PD-L1 from different sources, and discusses the influencing factors of PD-L1 detection and the value of combined biomarkers. Provide support for clinical screening of immunotherapy-advantage groups and formulation of personalized treatment decisions.
Collapse
Affiliation(s)
- Chang Qi
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Respiratory Health and Multimorbidity, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Center/Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yalun Li
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Respiratory Health and Multimorbidity, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Center/Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hao Zeng
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Respiratory Health and Multimorbidity, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Center/Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qi Wei
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Respiratory Health and Multimorbidity, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Center/Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Sihan Tan
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Respiratory Health and Multimorbidity, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Center/Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuanyuan Zhang
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Respiratory Health and Multimorbidity, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Center/Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Lung Cancer Center/Lung Cancer Institute, 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, Institute of Respiratory Health and Multimorbidity, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Center/Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Panwen Tian
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Respiratory Health and Multimorbidity, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Center/Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
21
|
Jin Y, Zuo Y, Li G, Liu W, Pan Y, Fan T, Fu X, Yao X, Peng Y. Advances in spatial transcriptomics and its applications in cancer research. Mol Cancer 2024; 23:129. [PMID: 38902727 PMCID: PMC11188176 DOI: 10.1186/s12943-024-02040-9] [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: 04/28/2024] [Accepted: 06/10/2024] [Indexed: 06/22/2024] Open
Abstract
Malignant tumors have increasing morbidity and high mortality, and their occurrence and development is a complicate process. The development of sequencing technologies enabled us to gain a better understanding of the underlying genetic and molecular mechanisms in tumors. In recent years, the spatial transcriptomics sequencing technologies have been developed rapidly and allow the quantification and illustration of gene expression in the spatial context of tissues. Compared with the traditional transcriptomics technologies, spatial transcriptomics technologies not only detect gene expression levels in cells, but also inform the spatial location of genes within tissues, cell composition of biological tissues, and interaction between cells. Here we summarize the development of spatial transcriptomics technologies, spatial transcriptomics tools and its application in cancer research. We also discuss the limitations and challenges of current spatial transcriptomics approaches, as well as future development and prospects.
Collapse
Affiliation(s)
- Yang Jin
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yuanli Zuo
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Gang Li
- Department of Thoracic Surgery, The Public Health Clinical Center of Chengdu, Chengdu, 610061, China
| | - Wenrong Liu
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yitong Pan
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ting Fan
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xin Fu
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiaojun Yao
- Department of Thoracic Surgery, The Public Health Clinical Center of Chengdu, Chengdu, 610061, China.
| | - Yong Peng
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Frontier Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610212, China.
| |
Collapse
|
22
|
Ye G, Wu G, Zhang C, Wang M, Liu H, Song E, Zhuang Y, Li K, Qi Y, Liao Y. CT-based quantification of intratumoral heterogeneity for predicting pathologic complete response to neoadjuvant immunochemotherapy in non-small cell lung cancer. Front Immunol 2024; 15:1414954. [PMID: 38933281 PMCID: PMC11199789 DOI: 10.3389/fimmu.2024.1414954] [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: 04/09/2024] [Accepted: 05/29/2024] [Indexed: 06/28/2024] Open
Abstract
Objectives To investigate the prediction of pathologic complete response (pCR) in patients with non-small cell lung cancer (NSCLC) undergoing neoadjuvant immunochemotherapy (NAIC) using quantification of intratumoral heterogeneity from pre-treatment CT image. Methods This retrospective study included 178 patients with NSCLC who underwent NAIC at 4 different centers. The training set comprised 108 patients from center A, while the external validation set consisted of 70 patients from center B, center C, and center D. The traditional radiomics model was contrasted using radiomics features. The radiomics features of each pixel within the tumor region of interest (ROI) were extracted. The optimal division of tumor subregions was determined using the K-means unsupervised clustering method. The internal tumor heterogeneity habitat model was developed using the habitats features from each tumor sub-region. The LR algorithm was employed in this study to construct a machine learning prediction model. The diagnostic performance of the model was evaluated using criteria such as area under the receiver operating characteristic curve (AUC), accuracy, specificity, sensitivity, positive predictive value (PPV), and negative predictive value (NPV). Results In the training cohort, the traditional radiomics model achieved an AUC of 0.778 [95% confidence interval (CI): 0.688-0.868], while the tumor internal heterogeneity habitat model achieved an AUC of 0.861 (95% CI: 0.789-0.932). The tumor internal heterogeneity habitat model exhibits a higher AUC value. It demonstrates an accuracy of 0.815, surpassing the accuracy of 0.685 achieved by traditional radiomics models. In the external validation cohort, the AUC values of the two models were 0.723 (CI: 0.591-0.855) and 0.781 (95% CI: 0.673-0.889), respectively. The habitat model continues to exhibit higher AUC values. In terms of accuracy evaluation, the tumor heterogeneity habitat model outperforms the traditional radiomics model, achieving a score of 0.743 compared to 0.686. Conclusion The quantitative analysis of intratumoral heterogeneity using CT to predict pCR in NSCLC patients undergoing NAIC holds the potential to inform clinical decision-making for resectable NSCLC patients, prevent overtreatment, and enable personalized and precise cancer management.
Collapse
Affiliation(s)
- Guanchao Ye
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Thoracic Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Guangyao Wu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chunyang Zhang
- Department of Thoracic Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Mingliang Wang
- Department of Thoracic Surgery, Henan Provincial People’s Hospital, Zhengzhou University, Zhengzhou, China
| | - Hong Liu
- School of Computer Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Enmin Song
- School of Computer Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Yuzhou Zhuang
- School of Computer Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Kuo Li
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Qi
- Department of Thoracic Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Yongde Liao
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
23
|
Díaz-Campos MÁ, Vasquez-Arriaga J, Ochoa S, Hernández-Lemus E. Functional impact of multi-omic interactions in lung cancer. Front Genet 2024; 15:1282241. [PMID: 38389572 PMCID: PMC10881857 DOI: 10.3389/fgene.2024.1282241] [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: 08/23/2023] [Accepted: 01/23/2024] [Indexed: 02/24/2024] Open
Abstract
Lung tumors are a leading cause of cancer-related death worldwide. Lung cancers are highly heterogeneous on their phenotypes, both at the cellular and molecular levels. Efforts to better understand the biological origins and outcomes of lung cancer in terms of this enormous variability often require of high-throughput experimental techniques paired with advanced data analytics. Anticipated advancements in multi-omic methodologies hold potential to reveal a broader molecular perspective of these tumors. This study introduces a theoretical and computational framework for generating network models depicting regulatory constraints on biological functions in a semi-automated way. The approach successfully identifies enriched functions in analyzed omics data, focusing on Adenocarcinoma (LUAD) and Squamous cell carcinoma (LUSC, a type of NSCLC) in the lung. Valuable information about novel regulatory characteristics, supported by robust biological reasoning, is illustrated, for instance by considering the role of genes, miRNAs and CpG sites associated with NSCLC, both novel and previously reported. Utilizing multi-omic regulatory networks, we constructed robust models elucidating omics data interconnectedness, enabling systematic generation of mechanistic hypotheses. These findings offer insights into complex regulatory mechanisms underlying these cancer types, paving the way for further exploring their molecular complexity.
Collapse
Affiliation(s)
| | - Jorge Vasquez-Arriaga
- Computational Genomics Division, National Institute of Genomic Medicine, Mexico City, Mexico
| | - Soledad Ochoa
- Computational Genomics Division, National Institute of Genomic Medicine, Mexico City, Mexico
- Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Enrique Hernández-Lemus
- Computational Genomics Division, National Institute of Genomic Medicine, Mexico City, Mexico
- Center for Complexity Sciences, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
24
|
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
|
25
|
Guo T, Zhu W, Zhao S, Qiu W, Wu Y, Li X, Ke F, Cheng H. Long‑term survival of a patient with advanced lung cancer treated with targeted therapy and anti‑PD‑1 immunotherapy as multi‑line therapy: A case report. Oncol Lett 2024; 27:32. [PMID: 38108071 PMCID: PMC10722554 DOI: 10.3892/ol.2023.14166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 09/14/2023] [Indexed: 12/19/2023] Open
Abstract
Lung cancer is the most common type of cancer worldwide. Lung adenocarcinoma, a type of non-small cell lung cancer (NSCLC), is a common type of lung cancer. In recent years, immunotherapy has become the primary method of treatment for several solid cancers, including NSCLC. In the present study, the case of a patient with NSCLC following left superior lobectomy is reported. Different systemic therapies failed, such as a pemetrexed + carboplatin regimen, paclitaxel liposome + cisplatin and pembrolizumab, and albumin-bound paclitaxel + toripalimab, but long-term survival was achieved following targeted therapy and anti-programmed cell death protein-1 (PD-1) immunotherapy. The patient survived for >4 years following lung cancer progression, which is notably longer than expected for patients with advanced lung cancer. In conclusion, the present case demonstrated the efficacy of targeted therapy and anti-PD-1 immunotherapy for the treatment of advanced lung cancer following the occurrence of drug resistance and progressive disease.
Collapse
Affiliation(s)
- Tianhao Guo
- Institute of Health and Regimen, Jiangsu Open University, Nanjing, Jiangsu 210036, P.R. China
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing, Jiangsu 210023, P.R. China
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Wenjian Zhu
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing, Jiangsu 210023, P.R. China
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Shuoqi Zhao
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing, Jiangsu 210023, P.R. China
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Wenli Qiu
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, P.R. China
| | - Yan Wu
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, P.R. China
| | - Xuan Li
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, P.R. China
| | - Fei Ke
- Department of Pathology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, P.R. China
| | - Haibo Cheng
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing, Jiangsu 210023, P.R. China
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
26
|
Zhang S, Li N, Wang F, Liu H, Zhang Y, Xiao J, Qiu W, Zhang C, Fan X, Qiu M, Li M, Tang H, Fan S, Wang J, Luo H, Li X, Lin J, Huang Y, Liang L. Characterization of the tumor microenvironment and identification of spatially predictive biomarkers associated with beneficial neoadjuvant chemoradiotherapy in locally advanced rectal cancer. Pharmacol Res 2023; 197:106974. [PMID: 37898442 DOI: 10.1016/j.phrs.2023.106974] [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: 08/26/2023] [Revised: 10/11/2023] [Accepted: 10/25/2023] [Indexed: 10/30/2023]
Abstract
Neoadjuvant chemoradiotherapy (nCRT) has become the standard treatment for patients with locally advanced rectal cancer (LARC). However, 20-40% of patients with LARC show little to no response to nCRT. Thus, comprehensively understanding the tumor microenvironment (TME), which might influence therapeutic efficacy, and identifying robust predictive biomarkers is urgently needed. Pre-treatment tumor biopsy specimens from patients with LARC were evaluated in detail through digital spatial profiling (DSP), public RNA sequencing datasets, and multiplex immunofluorescence (mIF). DSP analysis revealed distinct characteristics of the tumor stroma compared to the normal stroma and tumor compartments. We identified high levels of human leukocyte antigen-DR/major histocompatibility complex class II (HLA-DR/MHC-II) in the tumor compartment and B cells in the stroma as potential spatial predictors of nCRT efficacy in the Discovery cohort. Public datasets validated their predictive capacity for clinical outcomes. Using mIF in an independent nCRT cohort and/or the total cohort, we validated that a high density of HLA-DR/MHC-II+ cells in the tumor and CD20 + B cells in the stroma was associated with nCRT efficacy (all p ≤ 0.021). Spatial profiling successfully characterized the LARC TME and identified robust biomarkers with the potential to accurately predict nCRT response. These findings have important implications for individualized therapy.
Collapse
Affiliation(s)
- Shifen Zhang
- Department of Pathology, Nanfang Hospital/School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou 510515, China; Department of Pathology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, China
| | - Na Li
- Shenzhen Engineering Center for Translational Medicine of Precision Cancer Immunodiagnosis and Therapy, YuceBio Technology Co., Ltd, Shenzhen 518000, China.
| | - Feifei Wang
- Department of Pathology, Nanfang Hospital/School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou 510515, China
| | - Hailing Liu
- Department of Pathology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou 510655, China
| | - Yuhan Zhang
- Shenzhen Engineering Center for Translational Medicine of Precision Cancer Immunodiagnosis and Therapy, YuceBio Technology Co., Ltd, Shenzhen 518000, China
| | - Jinyuan Xiao
- Shenzhen Engineering Center for Translational Medicine of Precision Cancer Immunodiagnosis and Therapy, YuceBio Technology Co., Ltd, Shenzhen 518000, China
| | - Weihao Qiu
- Department of Pathology, Nanfang Hospital/School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou 510515, China
| | - Ceng Zhang
- Department of Pathology, Nanfang Hospital/School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou 510515, China
| | - Xinjuan Fan
- Department of Pathology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou 510655, China
| | - Mingxin Qiu
- Department of Pathology, Nanfang Hospital/School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou 510515, China
| | - Mingzhou Li
- Department of Pathology, Nanfang Hospital/School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou 510515, China
| | - Hongzhen Tang
- Shenzhen Engineering Center for Translational Medicine of Precision Cancer Immunodiagnosis and Therapy, YuceBio Technology Co., Ltd, Shenzhen 518000, China
| | - Shiheng Fan
- Shenzhen Engineering Center for Translational Medicine of Precision Cancer Immunodiagnosis and Therapy, YuceBio Technology Co., Ltd, Shenzhen 518000, China
| | - Jiaqian Wang
- Shenzhen Engineering Center for Translational Medicine of Precision Cancer Immunodiagnosis and Therapy, YuceBio Technology Co., Ltd, Shenzhen 518000, China
| | - Haitao Luo
- Shenzhen Engineering Center for Translational Medicine of Precision Cancer Immunodiagnosis and Therapy, YuceBio Technology Co., Ltd, Shenzhen 518000, China
| | - Xiangzhao Li
- Department of Pathology, Nanfang Hospital/School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou 510515, China
| | - Jie Lin
- Department of Pathology, Nanfang Hospital/School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou 510515, China; Jinfeng Laboratory, Chongqing 401329, China
| | - Yan Huang
- Department of Pathology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou 510655, China.
| | - Li Liang
- Department of Pathology, Nanfang Hospital/School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou 510515, China; Jinfeng Laboratory, Chongqing 401329, China.
| |
Collapse
|
27
|
Gray S, Ottensmeier CH. Advancing Understanding of Non-Small Cell Lung Cancer with Multiplexed Antibody-Based Spatial Imaging Technologies. Cancers (Basel) 2023; 15:4797. [PMID: 37835491 PMCID: PMC10571797 DOI: 10.3390/cancers15194797] [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: 08/21/2023] [Revised: 09/22/2023] [Accepted: 09/27/2023] [Indexed: 10/15/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) remains a cause of significant morbidity and mortality, despite significant advances made in its treatment using immune checkpoint inhibitors (ICIs) over the last decade; while a minority experience prolonged responses with ICIs, benefit is limited for most patients. The development of multiplexed antibody-based (MAB) spatial tissue imaging technologies has revolutionised analysis of the tumour microenvironment (TME), enabling identification of a wide range of cell types and subtypes, and analysis of the spatial relationships and interactions between them. Such study has the potential to translate into a greater understanding of treatment susceptibility and resistance, factors influencing prognosis and recurrence risk, and identification of novel therapeutic approaches and rational treatment combinations to improve patient outcomes in the clinic. Herein we review studies that have leveraged MAB technologies to deliver novel insights into the TME of NSCLC.
Collapse
Affiliation(s)
- Simon Gray
- Department of Molecular and Clinical Cancer Medicine, Faculty of Health and Life Sciences, University of Liverpool, Ashton St., Liverpool L69 3GB, UK
- Department of Medical Oncology, The Clatterbridge Cancer Centre NHS Foundation Trust, Pembroke Pl., Liverpool L7 8YA, UK
| | - Christian H. Ottensmeier
- Department of Molecular and Clinical Cancer Medicine, Faculty of Health and Life Sciences, University of Liverpool, Ashton St., Liverpool L69 3GB, UK
- Department of Medical Oncology, The Clatterbridge Cancer Centre NHS Foundation Trust, Pembroke Pl., Liverpool L7 8YA, UK
| |
Collapse
|
28
|
Wang WJ, Chu LX, He LY, Zhang MJ, Dang KT, Gao C, Ge QY, Wang ZG, Zhao XW. Spatial transcriptomics: recent developments and insights in respiratory research. Mil Med Res 2023; 10:38. [PMID: 37592342 PMCID: PMC10433685 DOI: 10.1186/s40779-023-00471-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 07/24/2023] [Indexed: 08/19/2023] Open
Abstract
The respiratory system's complex cellular heterogeneity presents unique challenges to researchers in this field. Although bulk RNA sequencing and single-cell RNA sequencing (scRNA-seq) have provided insights into cell types and heterogeneity in the respiratory system, the relevant specific spatial localization and cellular interactions have not been clearly elucidated. Spatial transcriptomics (ST) has filled this gap and has been widely used in respiratory studies. This review focuses on the latest iterative technology of ST in recent years, summarizing how ST can be applied to the physiological and pathological processes of the respiratory system, with emphasis on the lungs. Finally, the current challenges and potential development directions are proposed, including high-throughput full-length transcriptome, integration of multi-omics, temporal and spatial omics, bioinformatics analysis, etc. These viewpoints are expected to advance the study of systematic mechanisms, including respiratory studies.
Collapse
Affiliation(s)
- Wen-Jia Wang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Liu-Xi Chu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Li-Yong He
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Ming-Jing Zhang
- Orthopaedic Bioengineering Research Group, Division of Surgery and Interventional Science, University College London, London, HA7 4LP, UK
| | - Kai-Tong Dang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Chen Gao
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Qin-Yu Ge
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Zhou-Guang Wang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| | - Xiang-Wei Zhao
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China.
| |
Collapse
|
29
|
Yang L, Zhang Z, Dong J, Zhang Y, Yang Z, Guo Y, Sun X, Li J, Xing P, Ying J, Zhou M. Multi-dimensional characterization of immunological profiles in small cell lung cancer uncovers clinically relevant immune subtypes with distinct prognoses and therapeutic vulnerabilities. Pharmacol Res 2023; 194:106844. [PMID: 37392900 DOI: 10.1016/j.phrs.2023.106844] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 06/27/2023] [Accepted: 06/28/2023] [Indexed: 07/03/2023]
Abstract
Small-cell lung cancer (SCLC) is generally considered a 'homogenous' disease, with little documented inter-tumor heterogeneity in treatment guidance or prognosis evaluation. The precise identification of clinically relevant molecular subtypes remains incomplete and their translation into clinical practice is limited. In this retrospective cohort study, we comprehensively characterized the immune microenvironment in SCLC by integrating transcriptional and protein profiling of formalin-fixation-and-paraffin-embedded (FFPE) samples from 29 patients. We identified two distinct disease subtypes: immune-enriched (IE-subtype) and immune-deprived (ID-subtype), displaying heterogeneity in immunological, biological, and clinical features. The IE-subtype was characterized by abundant immune infiltrate and elevated levels of interferon-alpha/gamma (IFNα/IFNγ) and inflammatory response, while the ID-subtype featured a complete lack of immune infiltration and a more proliferative phenotype. These two immune subtypes are associated with clinical benefits in SCLC patients treated with adjuvant therapy, with the IE-subtype exhibiting a more favorable response leading to improved survival and reduced disease recurrence risk. Additionally, we identified and validated a personalized prognosticator of immunophenotyping, the CCL5/CXCL9 chemokine index (CCI), using machine learning. The CCI demonstrated superior predictive abilities for prognosis and clinical benefits in SCLC patients, validated in our institute immunohistochemistry cohort and multicenter bulk transcriptomic data cohorts. In conclusion, our study provides a comprehensive and multi-dimensional characterization of the immune architecture of SCLC using clinical FFPE samples and proposes a new immune subtyping conceptual framework enabling risk stratification and the appropriate selection of individualized therapy.
Collapse
Affiliation(s)
- Lin Yang
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, PR China
| | - Zicheng Zhang
- School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, PR China
| | - Jiyan Dong
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, PR China
| | - Yibo Zhang
- School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, PR China
| | - Zijian Yang
- School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, PR China
| | - Yiying Guo
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, PR China
| | - Xujie Sun
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, PR China
| | - Junling Li
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, PR China
| | - Puyuan Xing
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, PR China.
| | - Jianming Ying
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, PR China.
| | - Meng Zhou
- School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, PR China.
| |
Collapse
|
30
|
Ma Y, Xue J, Zhao Y, Zhang Y, Huang Y, Yang Y, Fang W, Guo Y, Li Q, Ge X, Sun J, Zhang B, Zhang Y, Xiao J, Zhang L, Zhao H. Phase I trial of KN046, a novel bispecific antibody targeting PD-L1 and CTLA-4 in patients with advanced solid tumors. J Immunother Cancer 2023; 11:jitc-2022-006654. [PMID: 37263673 DOI: 10.1136/jitc-2022-006654] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2023] [Indexed: 06/03/2023] Open
Abstract
BACKGROUND KN046 is a novel bispecific antibody targeting programmed death ligand 1 (PD-L1) and cytotoxic T lymphocyte-associated protein 4 (CTLA-4). This multicenter phase I trial investigated the safety, tolerability, pharmacokinetics (PK), and efficacy of KN046 in patients with advanced solid tumors. METHODS Patients who failed standard treatment were included. KN046 was administered at doses of 1, 3, and 5 mg/kg every 2 weeks (Q2W), 5 mg/kg every 3 weeks (Q3W), and 300 mg Q3W based on the modified toxicity probability interval method in the dose-escalation phase; the recommended dose was used in the expansion phase. Primary objectives were maximum tolerated dose (MTD) and recommended phase II dose (RP2D) in escalation and preliminary efficacy in expansion. Secondary objectives included PK, pharmacodynamics, safety, and tolerability of KN046. We also explored biomarkers based on PD-L1 expression, multiplex immunofluorescence (mIF) staining, and RNAseq-derived nCounter platform. RESULTS Totally, 100 eligible patients were enrolled, including 59 with nasopharyngeal carcinoma (NPC), 36 with epidermal growth factor receptor (EGFR)-mutated non-small-cell lung cancer (NSCLC), and those with other advanced solid tumors. The most common treatment-related adverse events (TRAEs) were rash (33.0%), pruritus (31.0%), and fatigue (20.0%). Grade ≥3 TRAEs were observed in 14.0% of participants. No dose-limiting toxicity occurred in the dose-escalation phase, and the MTD was not reached. The RP2D was determined as 5 mg/kg Q2W according to the pharmacokinetic-pharmacodynamic model, the preliminary exposure-response analysis, and the overall safety profile. Among 88 efficacy-evaluable participants, the objective response rate (ORR) was 12.5%, and the median duration of response was 16.6 months. In the NPC subgroup, the ORR was 15.4%, and the median overall survival (OS) was 24.7 (95% CI 16.3 to not estimable) months. In the EGFR-mutant NSCLC subgroup, the ORR was 6.3%. mIF analysis results showed patients with high CD8 expression showed longer median OS (27.1 vs 9.2 months, p=0.02); better prognosis was observed in patients with high CD8 and PD-L1 expression. CONCLUSIONS KN046 was well tolerated and showed promising antitumor efficacy in advanced solid tumors, especially in patients with NPC. The combination of both CD8 and PD-L1 expression improved the prediction of KN046 response. TRIAL REGISTRATION NUMBERS NCT03733951 .
Collapse
Affiliation(s)
- Yuxiang Ma
- Department of Clinical Research, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Jinhui Xue
- Department of Clinical Research, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Yuanyuan Zhao
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Yang Zhang
- Department of Clinical Research, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Yan Huang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Yunpeng Yang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Wenfeng Fang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Ye Guo
- Department of Oncology, Shanghai East Hospital,School of Medicine, Tongji University, Shanghai, China
| | - Qun Li
- Department of Oncology, Shanghai East Hospital,School of Medicine, Tongji University, Shanghai, China
| | - Xiaoxiao Ge
- Department of Oncology, Shanghai East Hospital,School of Medicine, Tongji University, Shanghai, China
| | - Jie Sun
- Department of Clinical Medicine, Jiangsu Alphamab Biopharmaceuticals Co.,Ltd, Jiangsu, China
| | - Bangyong Zhang
- Department of Clinical Operations, Jiangsu Alphamab Biopharmaceuticals Co.,Ltd, Jiangsu, China
| | - Yuhan Zhang
- Department of Translational Medicine, YuceBio Technology Co., Ltd, Shenzhen, China
| | - Jinyuan Xiao
- Department of Translational Medicine, YuceBio Technology Co., Ltd, Shenzhen, China
| | - Li Zhang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Hongyun Zhao
- Department of Clinical Research, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| |
Collapse
|
31
|
Schirizzi A, De Leonardis G, Lorusso V, Donghia R, Rizzo A, Vallarelli S, Ostuni C, Troiani L, Lolli IR, Giannelli G, Ricci AD, D'Alessandro R, Lotesoriere C. Targeting Angiogenesis in the Era of Biliary Tract Cancer Immunotherapy: Biological Rationale, Clinical Implications, and Future Research Avenues. Cancers (Basel) 2023; 15:cancers15082376. [PMID: 37190304 DOI: 10.3390/cancers15082376] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/14/2023] [Accepted: 04/18/2023] [Indexed: 05/17/2023] Open
Abstract
Although biliary tract cancers are traditionally considered rare in Western countries, their incidence and mortality rates are rising worldwide. A better knowledge of the genomic landscape of these tumor types has broadened the number of molecular targeted therapies, including angiogenesis inhibitors. The role of immune checkpoint inhibitors (ICIs) could potentially change the first-line therapeutic approach, but monotherapy with ICIs has shown disappointing results in CCA. Several clinical trials are evaluating combination strategies that include immunotherapy together with other anticancer agents with a synergistic activity. The tumor microenvironment (TME) composition plays a pivotal role in the prognosis of BTC patients. The accumulation of immunosuppressive cell types, such as tumor-associated macrophages (TAMs) and regulatory T-cells, together with the poor infiltration of cytotoxic CD8+ T-cells, is known to predispose to a poor prognosis owing to the establishment of resistance mechanisms. Likewise, angiogenesis is recognized as a major player in modulating the TME in an immunosuppressive manner. This is the mechanistic rationale for combination treatment schemes blocking both immunity and angiogenesis. In this scenario, this review aims to provide an overview of the most recent completed or ongoing clinical trials combining immunotherapy and angiogenesis inhibitors with/without a chemotherapy backbone.
Collapse
Affiliation(s)
- Annalisa Schirizzi
- Laboratory of Experimental Oncology, National Institute of Gastroenterology-IRCCS "Saverio de Bellis", 70013 Castellana Grotte, Italy
| | - Giampiero De Leonardis
- Laboratory of Experimental Oncology, National Institute of Gastroenterology-IRCCS "Saverio de Bellis", 70013 Castellana Grotte, Italy
| | - Vincenza Lorusso
- Clinical Trial Unit, National Institute of Gastroenterology-IRCCS "Saverio de Bellis", 70013 Castellana Grotte, Italy
| | - Rossella Donghia
- Data Science Unit, National Institute of Gastroenterology-IRCCS "Saverio de Bellis", 70013 Castellana Grotte, Italy
| | - Alessandro Rizzo
- Struttura Semplice Dipartimentale di Oncologia Medica per la Presa in Carico Globale del Paziente Oncologico "Don Tonino Bello", I.R.C.C.S. Istituto Tumori "Giovanni Paolo II", Viale Orazio Flacco 65, 70124 Bari, Italy
| | - Simona Vallarelli
- Medical Oncology Unit, National Institute of Gastroenterology-IRCCS "Saverio de Bellis", 70013 Castellana Grotte, Italy
| | - Carmela Ostuni
- Medical Oncology Unit, National Institute of Gastroenterology-IRCCS "Saverio de Bellis", 70013 Castellana Grotte, Italy
| | - Laura Troiani
- Medical Oncology Unit, National Institute of Gastroenterology-IRCCS "Saverio de Bellis", 70013 Castellana Grotte, Italy
| | - Ivan Roberto Lolli
- Medical Oncology Unit, National Institute of Gastroenterology-IRCCS "Saverio de Bellis", 70013 Castellana Grotte, Italy
| | - Gianluigi Giannelli
- Scientific Direction, National Institute of Gastroenterology-IRCCS "Saverio de Bellis", 70013 Castellana Grotte, Italy
| | - Angela Dalia Ricci
- Medical Oncology Unit, National Institute of Gastroenterology-IRCCS "Saverio de Bellis", 70013 Castellana Grotte, Italy
| | - Rosalba D'Alessandro
- Laboratory of Experimental Oncology, National Institute of Gastroenterology-IRCCS "Saverio de Bellis", 70013 Castellana Grotte, Italy
| | - Claudio Lotesoriere
- Medical Oncology Unit, National Institute of Gastroenterology-IRCCS "Saverio de Bellis", 70013 Castellana Grotte, Italy
| |
Collapse
|