1
|
Nie L, Zhang X, Wu J. PRSS3 is a potential prognostic biomarker for lung adenocarcinoma. Transl Cancer Res 2025; 14:1124-1140. [PMID: 40104712 PMCID: PMC11912035 DOI: 10.21037/tcr-24-1556] [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: 07/07/2024] [Accepted: 11/10/2024] [Indexed: 03/20/2025]
Abstract
Background Lung adenocarcinoma (LUAD) is a highly prevalent and deadly form of lung cancer and is a significant health concern worldwide. Although the expression of serine protease 3 (PRSS3) is elevated in certain cancers, its function in LUAD is yet unclear. The aim of this study was to investigate the mechanism of PRSS3 in lung adenocarcinoma, and validate PRSS3 as a reliable prognostic biomarker in lung adenocarcinoma. Methods The Cancer Genome Atlas (TCGA) provides RNA expression data and patient medical information for LUAD patients. To determine which genes are expressed differently in LUAD and normal lung tissues, we carefully examined these data. We then used Cox regression analysis to examine the expression and survival data to pinpoint the genes that are strongly associated with patient survival. The PRSS3 gene affects patient prognosis. Afterward, we divided LUAD patients into low- and high-expression groups on the basis of the median PRSS3 expression to examine the relationship between immune cells and PRSS3. The results of the CIBERSORT and CIBERSORTx studies revealed correlations between PRSS3 and the degree of infiltration of several immune cell types. After the groups with low and high PRSS3 expressions were compared, PRSS3-related genes were identified, and functional enrichment analysis was performed. Furthermore, a model was developed to predict patient prognosis according to clinical characteristics and PRSS3 expression. After the bioinformatics analyses were completed, we validated the differential expression of PRSS3 in samples obtained from our center via Western blotting and immunohistochemistry (IHC). Results We found that PRSS3 expression is highly upregulated in LUAD and that high PRSS3 expression is associated with a poorer prognosis in the TCGA database. Single-sample gene enrichment analysis revealed a strong correlation between PRSS3 and the immunological microenvironment. The clinical model developed on the basis of the PRSS3 showed great accuracy and can be used as a significant diagnostic indicator for LUAD. Western blotting and IHC confirmed a substantial increase in PRSS3 expression in LUAD. Herein, we analyzed an available dataset for a clinical cohort and revealed that elevated levels of PRSS3 are indicative of unfavorable outcomes in patients diagnosed with LUAD. Conclusions PRSS3 is significantly upregulated in LUAD and can be used as a marker for LUAD diagnosis and prognosis assessment. Further study of PRSS3 could provide valuable insight into the mechanisms underlying the occurrence and progression of LUAD.
Collapse
Affiliation(s)
- Lu Nie
- Department of Oncology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Xueqing Zhang
- Department of Oncology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Jie Wu
- Department of Oncology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
2
|
Izadi N, Solár P, Hašanová K, Zamani A, Akbar MS, Mrázová K, Bartošík M, Kazda T, Hrstka R, Joukal M. Breaking boundaries: role of the brain barriers in metastatic process. Fluids Barriers CNS 2025; 22:3. [PMID: 39780275 PMCID: PMC11708195 DOI: 10.1186/s12987-025-00618-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 01/02/2025] [Indexed: 01/11/2025] Open
Abstract
Brain metastases (BMs) are the most common intracranial tumors in adults and occur 3-10 times more frequently than primary brain tumors. Despite intensive multimodal therapies, including resection, radiotherapy, and chemotherapy, BMs are associated with poor prognosis and remain challenging to treat. BMs predominantly originate from primary lung (20-56%), breast (5-20%), and melanoma (7-16%) tumors, although they can arise from other cancer types less frequently. The metastatic cascade is a multistep process involving local invasion, intravasation into the bloodstream or lymphatic system, extravasation into normal tissue, and colonization of the distal site. After reaching the brain, circulating tumor cells (CTCs) breach the blood-brain barrier (BBB).The selective permeability of the BBB poses a significant challenge for therapeutic compounds, limiting the treatment efficacy of BMs. Understanding the mechanisms of tumor cell interactions with the BBB is crucial for the development of effective treatments. This review provides an in-depth analysis of the brain barriers, including the BBB, blood-spinal cord barrier, blood-meningeal barrier, blood-arachnoid barrier, and blood-cerebrospinal fluid barrier. It explores the molecular and cellular components of these barriers and their roles in brain metastasis, highlighting the importance of this knowledge for identifying druggable targets to prevent or limit BM formation.
Collapse
Affiliation(s)
- Nasim Izadi
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Zluty Kopec 7, 656 53, Brno, Czech Republic
| | - Peter Solár
- Department of Anatomy, Cellular and Molecular Neurobiology Research Group, Faculty of Medicine, Masaryk University, 625 00, Brno, Czech Republic
- Department of Neurosurgery, Faculty of Medicine, Masaryk University, St Anne University Hospital Brno, Pekařská 53, 656 91, Brno, Czech Republic
| | - Klaudia Hašanová
- Department of Anatomy, Cellular and Molecular Neurobiology Research Group, Faculty of Medicine, Masaryk University, 625 00, Brno, Czech Republic
| | - Alemeh Zamani
- Department of Anatomy, Cellular and Molecular Neurobiology Research Group, Faculty of Medicine, Masaryk University, 625 00, Brno, Czech Republic
| | - Maryam Shahidian Akbar
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Zluty Kopec 7, 656 53, Brno, Czech Republic
| | - Klára Mrázová
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Zluty Kopec 7, 656 53, Brno, Czech Republic
| | - Martin Bartošík
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Zluty Kopec 7, 656 53, Brno, Czech Republic
| | - Tomáš Kazda
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Zluty Kopec 7, 656 53, Brno, Czech Republic
| | - Roman Hrstka
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Zluty Kopec 7, 656 53, Brno, Czech Republic.
| | - Marek Joukal
- Department of Anatomy, Cellular and Molecular Neurobiology Research Group, Faculty of Medicine, Masaryk University, 625 00, Brno, Czech Republic.
| |
Collapse
|
3
|
Tanzhu G, Chen L, Ning J, Xue W, Wang C, Xiao G, Yang J, Zhou R. Metastatic brain tumors: from development to cutting-edge treatment. MedComm (Beijing) 2025; 6:e70020. [PMID: 39712454 PMCID: PMC11661909 DOI: 10.1002/mco2.70020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 10/19/2024] [Accepted: 10/25/2024] [Indexed: 12/24/2024] Open
Abstract
Metastatic brain tumors, also called brain metastasis (BM), represent a challenging complication of advanced tumors. Tumors that commonly metastasize to the brain include lung cancer and breast cancer. In recent years, the prognosis for BM patients has improved, and significant advancements have been made in both clinical and preclinical research. This review focuses on BM originating from lung cancer and breast cancer. We briefly overview the history and epidemiology of BM, as well as the current diagnostic and treatment paradigms. Additionally, we summarize multiomics evidence on the mechanisms of tumor occurrence and development in the era of artificial intelligence and discuss the role of the tumor microenvironment. Preclinically, we introduce the establishment of BM models, detailed molecular mechanisms, and cutting-edge treatment methods. BM is primarily treated with a comprehensive approach, including local treatments such as surgery and radiotherapy. For lung cancer, targeted therapy and immunotherapy have shown efficacy, while in breast cancer, monoclonal antibodies, tyrosine kinase inhibitors, and antibody-drug conjugates are effective in BM. Multiomics approaches assist in clinical diagnosis and treatment, revealing the complex mechanisms of BM. Moreover, preclinical agents often need to cross the blood-brain barrier to achieve high intracranial concentrations, including small-molecule inhibitors, nanoparticles, and peptide drugs. Addressing BM is imperative.
Collapse
Affiliation(s)
- Guilong Tanzhu
- Department of OncologyXiangya HospitalCentral South UniversityChangshaChina
| | - Liu Chen
- Department of OncologyXiangya HospitalCentral South UniversityChangshaChina
| | - Jiaoyang Ning
- Department of OncologyXiangya HospitalCentral South UniversityChangshaChina
| | - Wenxiang Xue
- NHC Key Laboratory of RadiobiologySchool of Public HealthJilin UniversityChangchunJilinChina
| | - Ce Wang
- Department of RadiologyChina‐Japan Friendship HospitalBeijingChina
| | - Gang Xiao
- Department of OncologyXiangya HospitalCentral South UniversityChangshaChina
| | - Jie Yang
- Department of OncologyXiangya HospitalCentral South UniversityChangshaChina
- Department of DermatologyXiangya HospitalCentral South UniversityChangshaChina
| | - Rongrong Zhou
- Department of OncologyXiangya HospitalCentral South UniversityChangshaChina
- Xiangya Lung Cancer CenterXiangya HospitalCentral South UniversityChangshaChina
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan ProvinceChina
| |
Collapse
|
4
|
Zhao J, Zhong J, Chen Y, Chen Z, Yin H, He Y, Chen R, Guo R. Molecular features of NSCLC patients with liver metastasis. Ther Adv Med Oncol 2024; 16:17588359241275421. [PMID: 39346119 PMCID: PMC11437564 DOI: 10.1177/17588359241275421] [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: 01/24/2024] [Accepted: 07/30/2024] [Indexed: 10/01/2024] Open
Abstract
Background Metastasis is the primary cause of lung cancer-related death. Primary cancer cells invade through the lymphatic or blood vessels to distant sites. Recently, it was proposed that lymphatic metastasis was more a hallmark of tumor aggressiveness or metastatic potential than a gateway to metastases. Therefore, the underlying molecular mechanism of metastasis is not entirely clear. Objectives This study aimed to explore the genetic mechanisms underlying liver metastases from lung cancer and to evaluate the efficacy of different therapies in these patients. Design We retrospectively analyzed the mutation spectrum of different biopsy samples including primary lung tumors, liver, lymph node metastasis, and circulating tumor DNA (ctDNA) from 1090 non-small-cell lung cancer (NSCLC) patients with liver metastasis between the years 2017 and 2022. Methods Demographic and disease characteristics were summarized using descriptive parameters. Time to treatment discontinuation was used to analyze the clinical outcome. Results More liquid biopsies were performed than tissue biopsies, especially in the treated advanced NSCLC patients. Liver metastasis before treatment was associated with poor response to immune checkpoint inhibitors and targeted therapy. Liver and lymph node metastasis had higher levels of single nucleotide variants and copy number variants than primary lung tumors. In paired lung and liver, lymph nodes, and simultaneous ctDNA, we found actionable mutations were always shared, while metastasis samples had multiple private mutations. Serial ctDNA analysis identifies potential resistant mutations and describes the evolution of tumor cells. Conclusion Liver and lymph node metastasis in NSCLC showed shared actionable mutations. Of note, the discrepancy of private mutations in liver and lymph node metastases indicated that liver metastases are mainly seeded by the primary tumor rather than the earlier colonized lymph node metastases.
Collapse
Affiliation(s)
- Jun Zhao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department I of Thoracic Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Jia Zhong
- CAMS Key Laboratory of Translational Research on Lung Cancer, State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center
- National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences Peking Union Medical College, Beijing, China
| | - Yujie Chen
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Zipei Chen
- Medical Oncology Department 1, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Huan Yin
- Geneplus-Beijing, Beijing, China
| | | | - Rongrong Chen
- Geneplus-Beijing, 7 Science Road, Zhongguancun Life Science Park, Changping, Beijing 102206, China
| | - Renhua Guo
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| |
Collapse
|
5
|
Chen G, Yu Y, Qi Y, Li G, Li N, Meng F, Wang W, Shen R. Comparative analysis of PD-L1 expression and molecular alterations in primary versus metastatic lung adenocarcinoma: a real-world study in China. Front Oncol 2024; 14:1393686. [PMID: 39323996 PMCID: PMC11422015 DOI: 10.3389/fonc.2024.1393686] [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: 05/18/2024] [Accepted: 08/26/2024] [Indexed: 09/27/2024] Open
Abstract
Objectives Programmed death-ligand 1 (PD-L1) is the only Food and Drug Administration-approved biomarker for monitoring response to immune checkpoint inhibitor (ICI) therapy in patients with lung adenocarcinoma. Understanding the nuances of molecular phenotypes, clinical attributes, and PD-L1 expression levels in primary and metastatic lung adenocarcinoma may help predict response to therapy and assist in the clinical management of lung adenocarcinoma. Methods A total of 235 primary and metastatic lesion specimens from patients with non-small cell lung cancer (NSCLC) an institution in Shandong, China were analyzed. PD-L1 expression was assessed by immunohistochemistry using the 22C3 antibody, and the molecular phenotype was determined by next-generation sequencing of 450 genes. The molecular phenotypes of the primary and metastatic lesions were compared. Results Elevated PD-L1 expression was significantly associated with advanced and metastatic disease (P = 0.001). The distribution of PD-L1 expression varied based on the anatomical location, showing a higher frequency of elevated PD-L1 expression in distal metastases than in the primary tumor. Metastatic lesions exhibited a higher proportion of carcinogenic pathway gene alterations and a greater number of DNA damage-repair pathway gene alterations than the primary lesions. Notably, CDKN2A copy number deletions were more prevalent in metastatic lesions than in primary lesions. Clinical data stemming from research conducted at the Memorial Sloan Kettering Cancer Center revealed an association between the absence of CDKN2A expression and a poorer prognosis in stage I lung adenocarcinoma. Conclusion Samples of metastatic tumors exhibited a higher proportion of elevated PD-L1 expression, a greater number of pathway alterations, and a higher occurrence of CDKN2A copy number deletions than primary samples. This highlights the importance of reinforcing the clinical management and follow-up of patients with CDKN2A deficiency, particularly within the subset of stage I lung adenocarcinoma.
Collapse
Affiliation(s)
- Gang Chen
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yang Yu
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Youchao Qi
- Department of Thoracic Surgery, The Second People's Hospital of Dezhou City, Dezhou, Shandong, China
| | - Guangxu Li
- Department of Thoracic Surgery, The Second People's Hospital of Dezhou City, Dezhou, Shandong, China
| | - Ning Li
- Department of Radiotherapy, The Second People's Hospital of Dezhou City, Dezhou, Shandong, China
| | - Fande Meng
- Department of Internal Medicine, Changle County Traditional Chinese Medicine (TMC) Hospital, Weifang, Shandong, China
| | - Wujie Wang
- Department of Interventional Medicine, The Second Hospital, Cheeloo College of Medicine, Institute of Tumor Intervention, Shandong University, Jinan, Shandong, China
| | - Rong Shen
- Department of Minimally Invasive Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
6
|
Koh YW, Han JH, Haam S, Lee HW. Changes in the expression of cell interaction-related pathways during brain metastasis in lung adenocarcinoma: Gene expression and immunohistochemical analysis. Pathol Res Pract 2024; 260:155375. [PMID: 38878665 DOI: 10.1016/j.prp.2024.155375] [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: 12/21/2023] [Accepted: 05/28/2024] [Indexed: 08/09/2024]
Abstract
BACKGROUND Brain metastasis (BM) is a prevalent prognostic event in the development of lung adenocarcinoma (LUAD) with a poor prognosis. Alterations in gene or protein expression during various phases of BM remain unclear. METHODS We performed gene expression and pathway analyses using a metastasis-related gene panel on 12 lung tissues from patients with confirmed BM, 12 lung tissues from patients without BM, and 12 matched brain tissues from patients with confirmed BM during follow-up after LUAD surgery. The results of the gene expression analysis were validated by immunohistochemistry. RESULTS Cell interaction-related pathways (such as focal adhesion, extracellular matrix-receptor interaction, and proteoglycans in cancer) showed the greatest differences among the three groups. Expression of the cell interaction-related pathway was highest in the lung sample of BM group and lowest in the matched brain tissue. Using a machine learning model, a signature of 20 genes from cell interaction-related pathways accurately predicted BM (area under the curve score of 0.792 and an accuracy rate of 0.875). Immunohistochemical analysis showed higher expression of proteins associated with cell interaction-related genes and a mesenchymal phenotype in the lung sample of BM group than in those without BM or matched brain tissue. CONCLUSIONS LUAD acquires the characteristics of the cell interaction-related pathway that leads to the development of BM, with a significant decrease in expression following brain colonization.
Collapse
Affiliation(s)
- Young Wha Koh
- Department of Pathology, Ajou University School of Medicine, Suwon-si, South Korea.
| | - Jae-Ho Han
- Department of Pathology, Ajou University School of Medicine, Suwon-si, South Korea
| | - Seokjin Haam
- Department of Thoracic and Cardiovascular Surgery, Ajou University School of Medicine, Suwon-si, South Korea
| | - Hyun Woo Lee
- Department of Hematology-Oncology, Ajou University School of Medicine, Suwon-si, South Korea
| |
Collapse
|
7
|
Du X, Yang S, Bian J, Zhang Y, Wang Y, Lv Z. Role of vascular endothelial growth factor D in lung adenocarcinoma immunotherapy response. Am J Transl Res 2024; 16:2263-2277. [PMID: 39006287 PMCID: PMC11236651 DOI: 10.62347/oxro7113] [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: 04/23/2024] [Accepted: 05/15/2024] [Indexed: 07/16/2024]
Abstract
OBJECTIVE To identify key genes associated with tumor-associated macrophages (TAMs), tumor immunotherapy, in the prognosis of lung adenocarcinoma (LUAD). METHODS The mRNA expression profiles of LUAD samples were obtained from The Cancer Genome Atlas (TCGA) database. The "CIBERSORT" R package was employed to calculate the proportion of innate immune cell infiltration in both tumor and adjacent normal tissues. TAM-associated genes in LUAD were identified to construct a prognostic risk model using weighted gene correlation network analysis (WGCNA), Least Absolute Shrinkage and Selection Operator (LASSO), and multivariate Cox regression analyses (COX). The IMvigor210 cohort was utilized to validate the roles of these genes as predictors of immunotherapy response. Tissue microarrays, immunofluorescence staining, and mRNA level detection methods were used to determine the correlation of risk factors in LUAD tissues. RESULTS CIBERSORT analysis revealed significant differences in innate immune cells between tumor and adjacent tissues. Seventy-four differential genes linked to these cells were identified from WGCNA. Four hub genes (endothelin receptor type B, vascular endothelial growth factor D (VEGFD), latent transforming growth factor beta binding protein 4 (LTBP4), and fibroblast growth factor receptor 4 (FGFR4)) in the TAM prognostic model were identified as independent prognostic risk factors (P < 0.05). VEGFD expression was identified as a low-risk factor for LUAD prognosis prediction (P < 0.05). Moreover, low-risk patients exhibited higher sensitivity to anti-PD-L1 therapy compared to high-risk patients (P < 0.05). VEGFD levels were negatively correlated with programmed cell death 1 (PD-1) levels (r = -0.363; P < 0.05), suggesting that VEGFD may serve as a predictor for anti-PD-1 treatment. CONCLUSIONS VEGFD is associated with innate immunity in LUAD, it can predict LUAD prognosis, and therefor may be a potential predictor for anti-PD-1 treatment in patients with LUAD.
Collapse
Affiliation(s)
- Xiaoling Du
- Department of Pharmacy, North Sichuan Medical College Nanchong 637000, Sichuan, P. R. China
- Department of Cardiology, Affiliated Hospital of North Sichuan Medical College Nanchong 637000, Sichuan, P. R. China
| | - Sha Yang
- Department of Pharmacy, North Sichuan Medical College Nanchong 637000, Sichuan, P. R. China
| | - Jiaojiao Bian
- Department of Pharmacy, North Sichuan Medical College Nanchong 637000, Sichuan, P. R. China
| | - Ying Zhang
- Department of Pharmacy, North Sichuan Medical College Nanchong 637000, Sichuan, P. R. China
| | - Yuquan Wang
- Department of Cardiology, Affiliated Hospital of North Sichuan Medical College Nanchong 637000, Sichuan, P. R. China
| | - Zhan Lv
- Department of Cardiology, Affiliated Hospital of North Sichuan Medical College Nanchong 637000, Sichuan, P. R. China
| |
Collapse
|
8
|
Yubin LI, Xiaoyan LI. [Advances in Clinical Application of Cerebrospinal Fluid Circulating Tumor DNA
in Leptomeningeal Metastasis of Non-small Cell Lung Cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2024; 27:376-382. [PMID: 38880925 PMCID: PMC11183312 DOI: 10.3779/j.issn.1009-3419.2024.102.16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Indexed: 06/18/2024]
Abstract
Leptomeningeal metastasis (LM) is a lethal complication of malignant tumors, with an incidence rate of 3%-5% among patients with non-small cell lung cancer (NSCLC). LM poses significant challenges in diagnosis, has poor prognosis, limited treatment options, and lacks standardized criteria for evaluating therapeutic efficacy, making it a difficult aspect of NSCLC management. Circulating tumor DNA (ctDNA), shed from tumor cells and carrying cancer-related information, holds significant value in precision oncology. Cerebrospinal fluid (CSF), present in the subarachnoid space of the brain, the spinal cord, and the central canal, and in direct contact with meningeal tissues, serves as the fluid medium that best reflects the genetic characteristics of LM. In recent years, CSF ctDNA has become a focal point due to its multi-omics features, playing a crucial role in the management of central nervous system (CNS) metastatic tumors. Its applications span the entire continuum of care, including aiding in diagnosis, assessing treatment response, predicting prognosis, and analyzing resistance mechanisms. This article provides a concise overview of CSF ctDNA detection techniques and their clinical applications in patients with NSCLC-LM.
.
Collapse
|
9
|
Chen J, Hu C, Yang H, Wang L, Chu X, Yu X, Zhang S, Li X, Zhao C, Cheng L, Hong W, Liu D, Wen L, Su C. PMS2 amplification contributes brain metastasis from lung cancer. Biol Proced Online 2024; 26:12. [PMID: 38714954 PMCID: PMC11075212 DOI: 10.1186/s12575-024-00238-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 04/16/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Lung adenocarcinoma metastasizing to the brain results in a notable increase in patient mortality. The high incidence and its impact on survival presents a critical unmet need to develop an improved understanding of its mechanisms. METHODS To identify genes that drive brain metastasis of tumor cells, we collected cerebrospinal fluid samples and paired plasma samples from 114 lung adenocarcinoma patients with brain metastasis and performed 168 panel-targeted gene sequencing. We examined the biological behavior of PMS2 (PMS1 Homolog 2)-amplified lung cancer cell lines through wound healing assays and migration assays. In vivo imaging techniques are used to detect fluorescent signals that colonize the mouse brain. RNA sequencing was used to compare differentially expressed genes between PMS2 amplification and wild-type lung cancer cell lines. RESULTS We discovered that PMS2 amplification was a plausible candidate driver of brain metastasis. Via in vivo and in vitro assays, we validated that PMS2 amplified PC-9 and LLC lung cancer cells had strong migration and invasion capabilities. The functional pathway of PMS2 amplification of lung cancer cells is mainly enriched in thiamine, butanoate, glutathione metabolism. CONCLUSION Tumor cells elevated expression of PMS2 possess the capacity to augment the metastatic potential of lung cancer and establish colonies within the brain through metabolism pathways.
Collapse
Affiliation(s)
- Jianing Chen
- Department of Medical Oncology, Shanghai Pulmonary Hospital &, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Congli Hu
- Department of Medical Oncology, Shanghai Pulmonary Hospital &, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Hainan Yang
- Department of Medical Oncology, Shanghai Pulmonary Hospital &, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
- Department of Critical Care Medicine, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Li Wang
- Department of Medical Oncology, Shanghai Pulmonary Hospital &, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Xiangling Chu
- Department of Medical Oncology, Shanghai Pulmonary Hospital &, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Xin Yu
- Department of Medical Oncology, Shanghai Pulmonary Hospital &, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Shiji Zhang
- Department of Medical Oncology, Shanghai Pulmonary Hospital &, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Xuefei Li
- Department of Medical Oncology, Shanghai Pulmonary Hospital &, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Chao Zhao
- Department of Medical Oncology, Shanghai Pulmonary Hospital &, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Lei Cheng
- Department of Medical Oncology, Shanghai Pulmonary Hospital &, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Weiping Hong
- Department of Oncology, Guangdong Sanjiu Brain Hospital, Guangzhou, China
| | - Da Liu
- Department of Neurosurgery, Guangdong Sanjiu Brain Hospital, Guangzhou, China
| | - Lei Wen
- Department of Radiation Oncology, Zhujiang Hospital, Southern Medical University, 253 Gongye Dadao, Guangdong, 510280, Guangzhou, China.
| | - Chunxia Su
- Department of Medical Oncology, Shanghai Pulmonary Hospital &, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China.
- Clinical Research Center, Shanghai Pulmonary Hospital, Shanghai, China.
| |
Collapse
|
10
|
Li W, Zhang K, Wang W, Liu Y, Huang J, Zheng M, Li L, Zhang X, Xu M, Chen G, Wang L, Zhang S. Combined inhibition of HER2 and VEGFR synergistically improves therapeutic efficacy via PI3K-AKT pathway in advanced ovarian cancer. J Exp Clin Cancer Res 2024; 43:56. [PMID: 38403634 PMCID: PMC10895844 DOI: 10.1186/s13046-024-02981-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 02/11/2024] [Indexed: 02/27/2024] Open
Abstract
BACKGROUND Ovarian cancer (OC) is a prevalent malignancy in the female reproductive system, and developing effective targeted therapies for this disease remains challenging. The aim of this study was to use clinically-relevant OC models to evaluate the therapeutic effectiveness of RC48, an antibody-drug conjugate (ADC) targeting HER2, either alone or in combination with the VEGFR inhibitor Cediranib Maleate (CM), for the treatment of advanced OC. METHODS OC tumor specimens and cell lines were analyzed to determine HER2 and VEGFR expression by Western blot, immunocytochemistry and immunofluorescence. Moreover, the OC cell lines, cell-derived xenograft (CDX) and patient-derived xenograft (PDX) models were treated with RC48 and/or CM and then subjected to cell proliferation, viability, apoptosis, and tumor growth analyses to evaluate the feasibility of combination therapy for OC both in vitro and in vivo. Additionally, RNA-Seq was performed to investigate the critical mechanism underlying the combination therapy of RC48 and CM. RESULTS Our results demonstrated that RC48 alone effectively targeted and inhibited the growth of HER2-positive OC tumors in both cell lines and PDX models. Furthermore, the combination of RC48 and CM synergistically induced tumor regression in human OC cell lines, as well as CDX and PDX models. Mechanistically, we observed that the combination treatment inhibited the growth of OC cells involved inducing apoptosis and suppressing cell motility. RNA-seq analysis provided further mechanistic insights and revealed that co-administration of RC48 and CM downregulated multiple cancer-related pathways, including the AKT/mTOR pathway, cell cycle, and cell proliferation. Notably, our data further confirmed that the PI3K-AKT pathway played a key role in the inhibition of proliferation triggered by combinational treatment of RC48 and CM in OC cells. CONCLUSIONS These findings provide a preclinical framework supporting the potential of dual targeting HER2 and VEGFR as a promising therapeutic strategy to improve outcomes in patients with OC.
Collapse
Affiliation(s)
- Weisong Li
- Department of Pathology, First Affiliated Hospital, Gannan Medical University, Ganzhou, 341000, China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases (Ministry of Education), Gannan Medical University, 1 Hexie Road, Rongjiang New District, Ganzhou, 341000, China
| | - Kai Zhang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases (Ministry of Education), Gannan Medical University, 1 Hexie Road, Rongjiang New District, Ganzhou, 341000, China
- School of Basic Medicine, Gannan Medical University, Ganzhou, 341000, China
| | - Wenjun Wang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases (Ministry of Education), Gannan Medical University, 1 Hexie Road, Rongjiang New District, Ganzhou, 341000, China
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Yuanyuan Liu
- Department of Gynaecology and Obstetrics, Ganzhou People's Hospital (The Affiliated Ganzhou Hospital of Nanchang University), Ganzhou, 341000, China
| | - Jianming Huang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases (Ministry of Education), Gannan Medical University, 1 Hexie Road, Rongjiang New District, Ganzhou, 341000, China
- School of Basic Medicine, Gannan Medical University, Ganzhou, 341000, China
| | - Meihong Zheng
- Department of Pathology, First Affiliated Hospital, Gannan Medical University, Ganzhou, 341000, China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases (Ministry of Education), Gannan Medical University, 1 Hexie Road, Rongjiang New District, Ganzhou, 341000, China
| | - Ling Li
- Department of Pathology, First Affiliated Hospital, Gannan Medical University, Ganzhou, 341000, China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases (Ministry of Education), Gannan Medical University, 1 Hexie Road, Rongjiang New District, Ganzhou, 341000, China
| | - Xinyu Zhang
- Department of Pathology, First Affiliated Hospital, Gannan Medical University, Ganzhou, 341000, China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases (Ministry of Education), Gannan Medical University, 1 Hexie Road, Rongjiang New District, Ganzhou, 341000, China
| | - Minjuan Xu
- Department of Gynaecology and Obstetrics, Ganzhou People's Hospital (The Affiliated Ganzhou Hospital of Nanchang University), Ganzhou, 341000, China.
| | - Guofang Chen
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Tongji University, Shanghai, 200092, China.
| | - Liefeng Wang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases (Ministry of Education), Gannan Medical University, 1 Hexie Road, Rongjiang New District, Ganzhou, 341000, China.
- School of Basic Medicine, Gannan Medical University, Ganzhou, 341000, China.
| | - Shuyong Zhang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases (Ministry of Education), Gannan Medical University, 1 Hexie Road, Rongjiang New District, Ganzhou, 341000, China.
- School of Basic Medicine, Gannan Medical University, Ganzhou, 341000, China.
| |
Collapse
|
11
|
Deng Q, Wang F, Song L, Chen L, Huang Y, Guo Z, Yang H. Proteomics-based Model for Predicting the Risk of Brain Metastasis in Patients with Resected Lung Adenocarcinoma carrying the EGFR Mutation. Int J Med Sci 2024; 21:765-774. [PMID: 38464823 PMCID: PMC10920840 DOI: 10.7150/ijms.92993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 02/13/2024] [Indexed: 03/12/2024] Open
Abstract
Introduction: Epidermal growth factor receptor (EGFR) mutation is common in Chinese patients with lung adenocarcinoma (LUAD). Brain metastases (BMs) is high and associated with poor prognosis. Identification of EGFR-mutant patients at high risk of developing BMs is important to reduce or delay the incidence of BMs. Currently, there is no literature on the prediction and modeling of EGFR brain metastasis at the proteinomics level. Methods: We conducted a retrospective study of BMs in postoperative recurrent LUAD with EGFR mutation in the First Affiliated Hospital of Guangzhou Medical University. Tissue proteomic analysis was applied in the primary tumors of resected LUAD in this study using liquid chromatography-mass spectrometry (LC-MS/MS). To identify potential markers for predicting LUAD BM, comparative analyses were performed on different groups to evaluate proteins associated with high risk of BMs. Results: A combination of three potential marker proteins was found to discriminate well between distal metastasis (DM) and local recurrence (LR) of postoperative LUAD with EGFR mutation. Gene Ontology (GO) analysis of significantly altered proteins between BM and non-BM (NBM) indicated that lipid metabolism and cell cycle-related pathways were involved in BMs of LUAD. And the enriched pathways correlated with BMs were found to be quite different in the comparison groups of postoperative adjuvant therapy, tyrosine kinase inhibitor (TKI), and chemotherapy groups. Finally, we developed a random forest algorithm model with eight proteins (RRS1, CPT1A, DNM1, SRCAP, MLYCD, PCID2, IMPAD1 and FILIP1), which showed excellent predictive value (AUC: 0.9401) of BM in patients with LUAD harboring EGFR mutation. Conclusions: A predictive model based on protein markers was developed to accurately predict postoperative BM in operable LUAD harboring EGFR mutation.
Collapse
Affiliation(s)
- Qiuhua Deng
- Department of Clinical Laboratory, the First Affiliated Hospital of Guangzhou Medical University, National Center for Respiratory Medicine, Guangzhou 510120, China
| | - Fengnan Wang
- Department of Thoracic Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Diseases, National Clinical Research Center of Respiratory Disease, Guangzhou 510120, China
| | - Lei Song
- Tianjin Key Laboratory of Clinical Multi-Omics, Tianjin 300308, China
| | - Liangyu Chen
- Tianjin Key Laboratory of Clinical Multi-Omics, Tianjin 300308, China
| | - Ying Huang
- Department of Thoracic Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Diseases, National Clinical Research Center of Respiratory Disease, Guangzhou 510120, China
| | - Zhihua Guo
- Department of Thoracic Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Diseases, National Clinical Research Center of Respiratory Disease, Guangzhou 510120, China
| | - Haihong Yang
- Department of Thoracic Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Diseases, National Clinical Research Center of Respiratory Disease, Guangzhou 510120, China
| |
Collapse
|
12
|
Hickman RA, Miller AM, Arcila ME. Cerebrospinal fluid: A unique source of circulating tumor DNA with broad clinical applications. Transl Oncol 2023; 33:101688. [PMID: 37196447 DOI: 10.1016/j.tranon.2023.101688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 04/27/2023] [Accepted: 05/08/2023] [Indexed: 05/19/2023] Open
Abstract
Malignancies involving the central nervous system present unique challenges for diagnosis and monitoring due to the difficulties and risks of direct biopsies and the low specificity and/or sensitivity of other techniques for assessment. In recent years, liquid biopsy of the cerebrospinal fluid (CSF) has emerged as a convenient alternative that combines minimal invasiveness with the ability to detect disease-defining or therapeutically actionable genetic alterations from circulating tumor DNA (ctDNA). Since CSF can be obtained by lumbar puncture, or an established ventricular access device at multiple time points, ctDNA analysis enables initial molecular characterization and longitudinal monitoring throughout a patient's disease course, promoting optimization of treatment regimens. This review outlines some of the key aspects of ctDNA from CSF as a highly suitable approach for clinical assessment, the benefits and drawbacks, testing methods, as well as potential future advancements in this field. We anticipate wider adoption of this practice as technologies and pipelines improve and envisage significant improvements for cancer care.
Collapse
Affiliation(s)
- Richard A Hickman
- Human Oncology and Pathogenesis Program, Sloan Kettering Institute, New York, NY, United States; Murtha Cancer Center Research Program, Uniformed Services University of the Health Sciences, Bethesda, MD, United States; Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Alexandra M Miller
- Human Oncology and Pathogenesis Program, Sloan Kettering Institute, New York, NY, United States; Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Maria E Arcila
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, United States.
| |
Collapse
|
13
|
Pala L, Bagnardi V, Tettamanzi F, Barberis M, Mazzarol G, Casali C, De Pas T, Pennacchioli E, Coppola S, Baldini F, Cocorocchio E, Ferrucci P, Patane' D, Saponara M, Queirolo P, Conforti F. Genetic Alterations of Melanoma Brain Metastases: A Systematic Review and Meta-Analysis. Mol Diagn Ther 2023; 27:5-13. [PMID: 36401787 DOI: 10.1007/s40291-022-00623-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2022] [Indexed: 11/20/2022]
Abstract
BACKGROUND Data on molecular alterations harbored by melanoma brain metastases (MBMs) are limited, and this has hampered the development of more effective therapeutic strategies. We conducted a systematic review and meta-analysis of all the studies reporting DNA sequencing data of MBMs, in order to identify recurrently mutated genes and molecular pathways significantly enriched for genetic alterations. METHODS We searched PubMed, Embase and Scopus for articles published from the inception of each database to June 30, 2021. We included in the analysis all the studies that reported individual patient data on DNA sequencing of MBMs, assessing single nucleotide variants (SNVs) and/or gene copy number variations (CNVs) in at least five tumor samples. Meta-analysis was performed for genes evaluated for SNVs and/or CNVs in at least two studies. Pooled proportions of samples with SNVs and/or CNVs was calculated by applying random-effect models based on the DerSimonian-Laird method. Gene-set enrichment analysis (GSEA) was performed to identify molecular pathways significantly enriched for mutated genes. RESULTS Ten studies fulfilled the inclusion criteria and were included in the analysis, for a total of 531 samples of MBMs evaluated. Twenty-seven genes were found recurrently mutated with a meta-analytic rate of SNVs higher than 5%. GSEA conducted on the list of these 27 recurrently mutated genes revealed vascular endothelial growth factor-activated receptor activity and transmembrane receptor protein tyrosine kinase activity to be among the top 10 gene ontology (GO) molecular functions significantly enriched for mutated genes, while regulation of apoptosis and cell proliferation were among the top 10 significantly enriched GO biological processes. Notably, a high meta-analytic rate of SNVs was found in several actionable cancer-associated genes, such as all the vascular endothelial growth factor (VEGF) receptor isoforms (i.e., Flt1 and Flt2 genes, for both SNV rate: 0.22, 95% CI 0.04-0.49; KDR gene, SNV rate: 0.1, 95% CI 0.05-0.16). Finally, two tumor suppressor genes were characterized by a high meta-analytic rate of CNVs: CDKN2A/B (CNV rate: 0.59, 95% CI 0.23-0.90) and PTEN (CNV rate: 0.31, 95% CI 0.02-0.95). CONCLUSION MBMs harbored actionable molecular alterations that could be exploited as therapeutic targets to improve the poor prognosis of patients.
Collapse
Affiliation(s)
- Laura Pala
- Division of Melanoma, Sarcomas and Rare Tumors, European Institute of Oncology IRCCS, via Ripamonti 435, 20141, Milan, Italy. .,Oncology Unit, Humanitas Gavazzeni, Via M.Gavazzeni 21, 24125, Bergamo, Italy.
| | - Vincenzo Bagnardi
- Department of Statistics and Quantitative Methods, University of Milan-Bicocca, Milan, Italy
| | | | - Massimo Barberis
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Giovanni Mazzarol
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Cecilia Casali
- Department of Neurological Surgery, IRCCS Foundation Neurological Institute "Carlo Besta", Milan, Italy
| | - Tommaso De Pas
- Division of Melanoma, Sarcomas and Rare Tumors, European Institute of Oncology IRCCS, via Ripamonti 435, 20141, Milan, Italy.,Oncology Unit, Humanitas Gavazzeni, Via M.Gavazzeni 21, 24125, Bergamo, Italy
| | - Elisabetta Pennacchioli
- Division of Melanoma, Sarcomas and Rare Tumors, European Institute of Oncology IRCCS, via Ripamonti 435, 20141, Milan, Italy
| | - Sara Coppola
- Division of Melanoma, Sarcomas and Rare Tumors, European Institute of Oncology IRCCS, via Ripamonti 435, 20141, Milan, Italy
| | - Federica Baldini
- Division of Melanoma, Sarcomas and Rare Tumors, European Institute of Oncology IRCCS, via Ripamonti 435, 20141, Milan, Italy
| | - Emilia Cocorocchio
- Division of Medical Oncology, European Institute of Oncology, IRCCS, Milan, Italy
| | - Pierfrancesco Ferrucci
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Damiano Patane'
- Division of Melanoma, Sarcomas and Rare Tumors, European Institute of Oncology IRCCS, via Ripamonti 435, 20141, Milan, Italy
| | - Maristella Saponara
- Division of Melanoma, Sarcomas and Rare Tumors, European Institute of Oncology IRCCS, via Ripamonti 435, 20141, Milan, Italy
| | - Paola Queirolo
- Division of Melanoma, Sarcomas and Rare Tumors, European Institute of Oncology IRCCS, via Ripamonti 435, 20141, Milan, Italy
| | - Fabio Conforti
- Division of Melanoma, Sarcomas and Rare Tumors, European Institute of Oncology IRCCS, via Ripamonti 435, 20141, Milan, Italy.,Oncology Unit, Humanitas Gavazzeni, Via M.Gavazzeni 21, 24125, Bergamo, Italy
| |
Collapse
|
14
|
Fernandes MB, Barata JT. IL-7 and IL-7R in health and disease: An update through COVID times. Adv Biol Regul 2023; 87:100940. [PMID: 36503870 DOI: 10.1016/j.jbior.2022.100940] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 11/24/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022]
Abstract
The role of IL-7 and IL-7R for normal lymphoid development and an adequately functioning immune system has been recognized for long, with severe immune deficiency and lymphoid leukemia as extreme examples of the consequences of deregulation of the IL-7-IL-7R axis. In this review, we provide an update (focusing on the past couple of years) on IL-7 and IL-7R in health and disease. We highlight the findings on IL-7/IL-7R signaling mechanisms and the, sometimes controversial, impact of IL-7 and its receptor on leukocyte biology, COVID-19, acute lymphoblastic leukemia, and different solid tumors, as well as their relevance as therapeutic tools or targets.
Collapse
Affiliation(s)
- Marta B Fernandes
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028, Lisbon, Portugal
| | - João T Barata
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028, Lisbon, Portugal.
| |
Collapse
|
15
|
Yang Y, Zhang J, Li JY, Xu L, Wang SN, Zhang JQ, Xun Z, Xia Y, Cao JB, Liu Y, Shi LY, Li W, Shi YL, He YG, Gu DJ, Yu ZY, Chen K, Lan J. The ctDNA-based postoperative molecular residual disease status in different subtypes of early-stage breast cancer. Gland Surg 2022; 11:1924-1935. [PMID: 36654951 PMCID: PMC9840987 DOI: 10.21037/gs-22-634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 12/12/2022] [Indexed: 12/28/2022]
Abstract
Background Breast cancer is a highly heterogeneous disease. Early-stage, non-metastatic breast cancer is considered curable after definitive treatment. Early detection of tumor recurrence and metastasis through sensitive biomarkers is helpful for guiding clinical decision-making and early intervention in second-line treatment, which could improve patient prognosis and survival. Methods In this real-world study, we retrospectively analyzed 82 patients with stages I to III breast cancer who had been analyzed by molecular residual disease (MRD) assay. A total of 82 tumor tissues and 224 peripheral blood samples were collected and detected by next-generation sequencing (NGS) based on a 1,021-gene panel in this study. Results MRD positivity was detected in 18 of 82 patients (22.0%). The hormone receptor-/human epidermal growth factor receptor 2+ (HR-/HER2+) subgroup had the highest postoperative MRD detection rate at 30.8% (4/13). The BRCA2 and SLX4 genes were significantly enriched in all patients in the MRD positive group and FGFR1 amplification was significantly enriched in the MRD negative group with HR+/HER2-. The number of single nucleotide variants (SNVs) in tissue samples of MRD-positive patients was higher than that of MRD-negative patients (11.94 vs. 8.50 SNVs/sample). Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis showed that there was a similar biological function of the tumor-mutated genes in the 2 MRD status groups. Conclusions This real-world study confirmed that patient samples of primary tumor tissue with different MRD status and molecular subtypes had differential genetic features, which may be used to predict patients at high risk for recurrence.
Collapse
Affiliation(s)
- Yang Yang
- Medical College of Soochow University, Suzhou, China
| | - Jie Zhang
- Department of Gynecology & Obstetrics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jiao-Yang Li
- Department of Ultrasound, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Lu Xu
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Si-Ning Wang
- Medical College of Soochow University, Suzhou, China
| | - Jun-Qi Zhang
- Medical College of Soochow University, Suzhou, China
| | - Zhou Xun
- Medical College of Soochow University, Suzhou, China
| | - Yu Xia
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jian-Bo Cao
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yang Liu
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Li-Yan Shi
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Wei Li
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | | | - Yuan-Ge He
- Geneplus-Beijing Institute, Beijing, China
| | - De-Jian Gu
- Geneplus-Beijing Institute, Beijing, China
| | - Zheng-Yuan Yu
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Kai Chen
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jing Lan
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
16
|
Wilcox JA, Li MJ, Boire AA. Leptomeningeal Metastases: New Opportunities in the Modern Era. Neurotherapeutics 2022; 19:1782-1798. [PMID: 35790709 PMCID: PMC9723010 DOI: 10.1007/s13311-022-01261-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/03/2022] [Indexed: 02/07/2023] Open
Abstract
Leptomeningeal metastases arise from cancer cell entry into the subarachnoid space, inflicting significant neurologic morbidity and mortality across a wide range of malignancies. The modern era of cancer therapeutics has seen an explosion of molecular-targeting agents and immune-mediated strategies for patients with breast, lung, and melanoma malignancies, with meaningful extracranial disease control and improvement in patient survival. However, the clinical efficacy of these agents in those with leptomeningeal metastases remains understudied, due to the relative rarity of this patient population, the investigational challenges associated with studying this dynamic disease state, and brisk disease pace. Nevertheless, retrospective studies, post hoc analyses, and small prospective trials in the last two decades provide a glimmer of hope for patients with leptomeningeal metastases, suggesting that several cancer-directed strategies are not only active in the intrathecal space but also improve survival against historical odds. The continued development of clinical trials devoted to patients with leptomeningeal metastases is critical to establish robust efficacy outcomes in this patient population, define drug pharmacokinetics in the intrathecal space, and uncover new avenues for treatment in the face of leptomeningeal therapeutic resistance.
Collapse
Affiliation(s)
- Jessica A Wilcox
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Min Jun Li
- Brain Tumor Center, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Adrienne A Boire
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Brain Tumor Center, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
17
|
Tsai C, Nguyen B, Luthra A, Chou JF, Feder L, Tang LH, Strong VE, Molena D, Jones DR, Coit DG, Ilson DH, Ku GY, Cowzer D, Cadley J, Capanu M, Schultz N, Beal K, Moss NS, Janjigian YY, Maron SB. Outcomes and Molecular Features of Brain Metastasis in Gastroesophageal Adenocarcinoma. JAMA Netw Open 2022; 5:e2228083. [PMID: 36001319 PMCID: PMC9403772 DOI: 10.1001/jamanetworkopen.2022.28083] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
IMPORTANCE Brain metastasis (BrM) in gastroesophageal adenocarcinoma (GEA) is a rare and poorly understood phenomenon associated with poor prognosis. OBJECTIVES To examine the clinical and genomic features of patients with BrM from GEA and evaluate factors associated with survival. DESIGN, SETTING, AND PARTICIPANTS In this single-institution retrospective cohort study, 68 patients with BrM from GEA diagnosed between January 1, 2008, and December 31, 2020, were identified via review of billing codes and imaging reports from the electronic medical record with follow-up through November 3, 2021. Genomic data were derived from the Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets clinical sequencing platform. EXPOSURES Treatment with BrM resection and/or radiotherapy. MAIN OUTCOMES AND MEASURES Overall survival after BrM diagnosis. RESULTS Sixty-eight patients (median age at diagnosis, 57.4 years [IQR, 49.8-66.4 years]; 59 [86.8%] male; 55 [85.9%] White) participated in the study. A total of 57 (83.8%) had primary tumors in the distal esophagus or gastroesophageal junction. Median time from initial diagnosis to BrM diagnosis was 16.9 months (IQR, 8.5-27.7 months). Median survival from BrM diagnosis was 8.7 months (95% CI, 5.5-11.5 months). Overall survival was 35% (95% CI, 25%-48%) at 1 year and 24% (95% CI, 16%-37%) at 2 years. In a multivariable analysis, an Eastern Cooperative Oncology Group performance status of 2 or greater (hazard ratio [HR], 4.66; 95% CI, 1.47-14.70; P = .009) and lack of surgical or radiotherapeutic intervention (HR, 7.71; 95% CI, 2.01-29.60; P = .003) were associated with increased risk of all-cause mortality, whereas 3 or more extracranial sites of disease (HR, 1.85; 95% CI, 0.64-5.29; P = .25) and 4 or more BrMs (HR, 2.15; 95% CI, 0.93-4.98; P = .07) were not statistically significant. A total of 31 patients (45.6%) had ERBB2 (formerly HER2 or HER2/neu)-positive tumors, and alterations in ERBB2 were enriched in BrM relative to primary tumors (8 [47.1%] vs 7 [20.6%], P = .05), as were alterations in PTPRT (7 [41.2%] vs 4 [11.8%], P = .03). CONCLUSIONS AND RELEVANCE This study suggests that that a notable proportion of patients with BrM from GEA achieve survival exceeding 1 and 2 years from BrM diagnosis, a more favorable prognosis than previously reported. Good performance status and treatment with combination surgery and radiotherapy were associated with the best outcomes. ERBB2 positivity and amplification as well as PTPRT alterations were enriched in BrM tissue compared with primary tumors; therefore, further study should be pursued to identify whether these variables represent genomic risk factors for BrM development.
Collapse
Affiliation(s)
- Charlton Tsai
- Department of Medicine, New York Presbyterian/Weill Cornell Medicine, New York, New York
| | - Bastien Nguyen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Anisha Luthra
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Joanne F. Chou
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Lara Feder
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Laura H. Tang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Vivian E. Strong
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Daniela Molena
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - David R. Jones
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Daniel G. Coit
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - David H. Ilson
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Geoffrey Y. Ku
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Darren Cowzer
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - John Cadley
- Department of Digital Informatics and Technology Solutions, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Marinela Capanu
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nikolaus Schultz
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kathryn Beal
- Department of Radiation Oncology and Brain Metastasis Center, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nelson S. Moss
- Department of Neurosurgery and Brain Metastasis Center, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Yelena Y. Janjigian
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Steven B. Maron
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|