1
|
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
|
2
|
Li J, Xie B, Wang H, Wang Q, Wu Y. Investigating MATN3 and ASPN as novel drivers of gastric cancer progression via EMT pathways. Hum Mol Genet 2024; 33:2035-2050. [PMID: 39301785 DOI: 10.1093/hmg/ddae129] [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: 06/24/2024] [Revised: 08/09/2024] [Accepted: 08/24/2024] [Indexed: 09/22/2024] Open
Abstract
Gastric cancer (GC) is a leading cause of cancer-related deaths globally, necessitating the identification of novel therapeutic targets. This study investigates the roles of MATN3 and ASPN in GC progression via the epithelial-mesenchymal transition (EMT) pathway. Analysis of the Cancer Genome Atlas-Stomach Adenocarcinoma (TCGA-STAD) dataset revealed that both MATN3 and ASPN are significantly upregulated in GC tissues and correlate with poor patient survival. Protein-protein interaction and co-expression analyses confirmed a direct interaction between MATN3 and ASPN, suggesting their synergistic role in EMT activation. Functional assays demonstrated that MATN3 promotes GC cell proliferation, migration, and invasion, while its knockdown inhibits these malignant behaviors and induces apoptosis. ASPN overexpression further amplified these oncogenic effects. In vivo, studies in a mouse model corroborated that co-overexpression of MATN3 and ASPN enhances tumor growth and metastasis. These findings highlight the MATN3-ASPN axis as a potential therapeutic target in GC, offering new insights into the molecular mechanisms driving GC progression.
Collapse
Affiliation(s)
- Jing Li
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Soochow University, No. 1055, Sanxiang Road, Suzhou 215004, Jiangsu Province, China
- Department of Oncological Surgery, The First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Longzihu District, Bengbu 233004, Anhui Province, China
| | - Bo Xie
- Department of Oncological Surgery, The First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Longzihu District, Bengbu 233004, Anhui Province, China
| | - Hu Wang
- Department of Oncological Surgery, The First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Longzihu District, Bengbu 233004, Anhui Province, China
| | - QingKang Wang
- Department of Oncological Surgery, The First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Longzihu District, Bengbu 233004, Anhui Province, China
| | - YongYou Wu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Soochow University, No. 1055, Sanxiang Road, Suzhou 215004, Jiangsu Province, China
| |
Collapse
|
3
|
Wu M, Lei S, Tang Y, He C, Zhang J, Lu X, Tan A, Wang H, Xie W. Real-World Outcomes of Pyrotinib-Based Therapy for HER2-Positive Breast Cancer With Brain Metastases: A Multicentre, Retrospective Analysis. Clin Breast Cancer 2024:S1526-8209(24)00300-8. [PMID: 39603902 DOI: 10.1016/j.clbc.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/27/2024] [Accepted: 11/02/2024] [Indexed: 11/29/2024]
Abstract
OBJECTIVE This study was designed to investigate the efficacy and safety of pyrotinib-based therapy for HER2-positive breast cancer with brain metastases (BM) in the real-world setting. METHODS Data of HER2-positive breast cancer patients with BM treated with pyrotinib-based therapy from a multicetre, registered, real-world study were analyzed. RESULTS Among 45 female patients, the overall objective response rate (ORR) was 62.2%, higher in 1st/2nd-line than ≥ 3rd-line (71.0% vs. 42.9%, P = .072). The objective response rate of intracranial lesions (CNS-ORR) was 71.1 %, with a significantly higher CNS-ORR observed in the 1st or 2nd-line subgroup compared to that of ≥ 3rd-line subgroup (83.9% vs. 42.9%, P < .05). By the end of follow-up, 20 patients (44.4%) died, and the 1-year survival rate was 73.3%. The median progression-free survival (PFS) was 9.1 months (95% CI 6.7-11.5). Patients with 1 or 2 BM had a longer median PFS of 12.0 months compared to 7.7 months for those with ≥ 3 BM (P = .01). In addition, 1- or 2-line therapy and full dose exposure of pyrotinib of 320mg-400mg/day were associated with improved median PFS (all P > .05). The median intracranial PFS (CNS-PFS) was 11.4 months (95% CI 7.5-15.3). However, local intervention plus systemic treatment seemed to prolong CNS-PFS compared with systemic treatment alone (13.7 vs. 9.1 months, P = .128). Diarrhea was most common (88.9%), 24.4% grade 3. CONCLUSIONS The pyrotinib-based therapy is effective for HER-2 positive breast cancer with BM, especially in 1st- or 2nd-line treatment, with tolerable adverse events. However, insufficient dosing of pyrotinib may impair efficacy outcomes.
Collapse
Affiliation(s)
- Muxin Wu
- Department of Breast, Bone&Soft Tissue Oncology, Guangxi Medical University Cancer Hospital, China
| | - Sen Lei
- Department of Breast, Bone&Soft Tissue Oncology, Guangxi Medical University Cancer Hospital, China
| | - Yijing Tang
- Department of Breast, Bone&Soft Tissue Oncology, Guangxi Medical University Cancer Hospital, China
| | - Chengzu He
- Department of Oncology, Binyang People's Hospital, China
| | - Jian Zhang
- Department of Oncology, Laibin People's Hospital, China
| | - Xin'an Lu
- Department of Oncology, Hengzhou People's Hospital, China
| | - Aihua Tan
- Department of Breast, Bone&Soft Tissue Oncology, Guangxi Medical University Cancer Hospital, China
| | - Hongxue Wang
- Department of Breast, Bone&Soft Tissue Oncology, Guangxi Medical University Cancer Hospital, China
| | - Weimin Xie
- Department of Breast, Bone&Soft Tissue Oncology, Guangxi Medical University Cancer Hospital, China.
| |
Collapse
|
4
|
Svajda L, Ranđelović I, Surguta SE, Baranyi M, Cserepes M, Tóvári J. Targeting hypoxia in combination with paclitaxel to enhance therapeutic efficacy in breast and ovarian cancer. Biomed Pharmacother 2024; 180:117601. [PMID: 39476764 DOI: 10.1016/j.biopha.2024.117601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 10/11/2024] [Accepted: 10/21/2024] [Indexed: 11/14/2024] Open
Abstract
The poor vascularization of solid tumors results in oxygen-deprived areas within the tumor mass. This phenomenon is defined as tumor hypoxia and is considered to be a major contributor to tumor progression in breast and ovarian cancers due to hypoxia-cascade-promoted increased metastasizing capacity. Hence, targeting hypoxia is a strategic cancer treatment approach, however, the hypoxia-modulating drugs face several limitations in monotherapies. Here, we investigated the impact of the potent hypoxia-inducible factor inhibitory compound acriflavine on tumor cell proliferation, migration, and metabolism under hypoxic conditions. We identified that acriflavine inhibited the proliferation of breast and ovarian tumor cells. To model the potential benefits of additional hypoxia response inhibition next to standard chemotherapy, we combined acriflavine with a frequently used chemotherapeutic agent, paclitaxel. In most breast and ovarian cancer cell lines used, we identified additive effects between the two drugs. The most significant findings were detected in triple-negative breast cancer cell lines, where we observed synergism. The drug combination effectively impeded tumor growth and metastasis formation in an in vivo orthotopic triple-negative breast cancer model as well. Additionally, we demonstrated that an epithelial-mesenchymal transition inhibitory drug, rolipram, combined with acriflavine and paclitaxel, notably reduced the motility of hypoxic triple-negative breast cancer cells. In conclusion, we identified novel drug combinations that can potentially combat triple-negative breast cancer by inhibiting hypoxia signaling and hindering cell migration and metastasis formation.
Collapse
Affiliation(s)
- Laura Svajda
- Department of Experimental Pharmacology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, Hungary; Doctoral School of Semmelweis University, Budapest, Hungary.
| | - Ivan Ranđelović
- Department of Experimental Pharmacology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, Hungary
| | - Sára Eszter Surguta
- Department of Experimental Pharmacology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, Hungary
| | - Marcell Baranyi
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Mihály Cserepes
- Department of Experimental Pharmacology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, Hungary; Doctoral School of Semmelweis University, Budapest, Hungary
| | - József Tóvári
- Department of Experimental Pharmacology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, Hungary; Doctoral School of Semmelweis University, Budapest, Hungary
| |
Collapse
|
5
|
Ji L, Song G, Xiao M, Chen X, Li Q, Wang J, Fan Y, Luo Y, Li Q, Chen S, Ma F, Xu B, Zhang P. Subdivision of M1 category and prognostic stage for de novo metastatic breast cancer to enhance prognostic prediction and guide the selection of locoregional therapy. Thorac Cancer 2024; 15:2193-2205. [PMID: 39279162 PMCID: PMC11496194 DOI: 10.1111/1759-7714.15452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 08/19/2024] [Accepted: 09/03/2024] [Indexed: 09/18/2024] Open
Abstract
BACKGROUND Although de novo metastatic breast cancer (dnMBC) is acknowledged as a heterogeneous disease, the current staging systems do not distinguish between patients within the M1 or stage IV category. This study aimed to refine the M1 category and prognostic staging for dnMBC to enhance prognosis prediction and guide the choice of locoregional treatment. METHODS We selected patients with dnMBC from the SEER database (2010-2019), grouping them into training (N = 8048) and internal validation (N = 3450) cohorts randomly at a 7:3 ratio. An independent external validation cohort (N = 660) was enrolled from dnMBC patients (2010-2023) treated in three hospitals. Nomogram-based risk stratification was employed to refine the M1 category and prognostic stage, incorporating T/N stage, histologic grade, subtypes, and the location and number of metastatic sites. Both internal and external validation sets were used for validation analyses. RESULTS Brain, liver, or lung involvement and multiple metastases were independent prognostic factors for overall survival (OS). The nomogram-based stratification effectively divided M1 stage into three groups: M1a (bone-only involvement), M1b (liver or lung involvement only, with or without bone metastases), and M1c (brain metastasis or involvement of both liver and lung, regardless of other metastatic sites). Only subtype and M1 stage were included to define the final prognostic stage. Significant differences in OS were observed across M1 and prognostic subgroups. Patients with the M1c stage benefited less from primary tumor surgery in comparison with M1a stage. CONCLUSION Subdivision of the M1 and prognostic stage could serve as a supplement to the current staging guidelines for dnMBC and guide locoregional treatment.
Collapse
Affiliation(s)
- Lei Ji
- Department of Medical OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Ge Song
- Department of Medical OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Min Xiao
- Department of Medical OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xi Chen
- Department of Medical OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Qing Li
- Department of Medical OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Jiayu Wang
- Department of Medical OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Ying Fan
- Department of Medical OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yang Luo
- Department of Medical OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Qiao Li
- Department of Medical OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Shanshan Chen
- Department of Medical OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Fei Ma
- Department of Medical OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Binghe Xu
- Department of Medical OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Pin Zhang
- Department of Medical OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
6
|
Gonsalves D, Ciérvide R, Couñago F. Bridging the gap: Predicting brain metastasis in breast cancer. World J Clin Oncol 2024; 15:356-359. [PMID: 38455134 PMCID: PMC10915941 DOI: 10.5306/wjco.v15.i2.356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/04/2024] [Accepted: 01/30/2024] [Indexed: 02/20/2024] Open
Abstract
Chen et al explored clinicopathological features and prognostic factors, revealing advanced tumor stage, lung metastases, HER-2 overexpression, and triple-negative status as key contributors. Recent research connects astrocytes' role in brain metastasis with signaling pathways and the impact of Trastuzumab on HER-2 tumor survival. Factors such as positive HER2 status, lack of estrogen receptor expression, and liver metastasis are identified as additional risk factors. The routine use of magnetic resonance imaging, insights into gene mutations associated with metastasis, and the role of radiotherapy, including prophylaxis possibilities, is controversial in clinical practice. Understanding these risk factors in a multidisciplinary collaboration is precise for local treatments and targeted therapies, particularly for HER2+ tumors, impacting directly on longer survival.
Collapse
Affiliation(s)
- Daniela Gonsalves
- Department of Radiation Oncology, GenesisCare Madrid, Madrid 28043, Spain
- Facultad de Medicina Salud y Deporte, Universidad Europea de Madrid, Madrid 28670, Spain
| | - Raquel Ciérvide
- Department of Radiation Oncology, HM Hospitales, Madrid 28050, Spain
| | - Felipe Couñago
- Department of Radiation Oncology, GenesisCare Madrid, Madrid 28043, Spain
- Facultad de Medicina Salud y Deporte, Universidad Europea de Madrid, Madrid 28670, Spain
| |
Collapse
|
7
|
Liu Z, Dong S, Liu M, Liu Y, Ye Z, Zeng J, Yao M. Experimental models for cancer brain metastasis. CANCER PATHOGENESIS AND THERAPY 2024; 2:15-23. [PMID: 38328712 PMCID: PMC10846332 DOI: 10.1016/j.cpt.2023.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 10/17/2023] [Accepted: 10/25/2023] [Indexed: 02/09/2024]
Abstract
Brain metastases are a leading cause of cancer-related mortality. However, progress in their treatment has been limited over the past decade, due to an incomplete understanding of the underlying biological mechanisms. Employing accurate in vitro and in vivo models to recapitulate the complexities of brain metastasis offers the most promising approach to unravel the intricate cellular and physiological processes involved. Here, we present a comprehensive review of the currently accessible models for studying brain metastasis. We introduce a diverse array of in vitro and in vivo models, including cultured cells using the Transwell system, organoids, microfluidic models, syngeneic models, xenograft models, and genetically engineered models. We have also provided a concise summary of the merits and limitations inherent to each model while identifying the optimal contexts for their effective utilization. This review serves as a comprehensive resource, aiding researchers in making well-informed decisions regarding model selection that align with specific research questions.
Collapse
Affiliation(s)
- Zihao Liu
- Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease, Guangzhou, Guangdong 510182, China
| | - Shanshan Dong
- Department of Medical Genetics and Cell Biology, GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Mengjie Liu
- Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease, Guangzhou, Guangdong 510182, China
| | - Yuqiang Liu
- Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease, Guangzhou, Guangdong 510182, China
| | - Zhiming Ye
- Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease, Guangzhou, Guangdong 510182, China
| | - Jianhao Zeng
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Maojin Yao
- Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease, Guangzhou, Guangdong 510182, China
| |
Collapse
|