1
|
Chen B, Huang L, Gui M, Torres-de la Roche LA, De Wilde RL, Shi W, Liu H, Gong Z. High Expression of Complement 3 Enhances the Efficacy of Neoadjuvant Chemotherapy Prior to Oncoplastic Surgery for HER2-Positive Breast Cancer. Cancer Biother Radiopharm 2025. [PMID: 40242863 DOI: 10.1089/cbr.2025.0038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2025] Open
Abstract
Background: Neoadjuvant chemotherapy for breast cancer (BC) improves patient prognosis, but its efficacy is hindered by the disease's high heterogeneity. This study enhances effectiveness of targeted therapy to improve clinical outcomes. Methods: This study enrolled 335 patients from three centers. Differentially expressed genes were identified using DESeq2, and Venn analysis was applied to identify hub Complement genes. Hub gene expression was validated through public databases and IHC in real-world samples. In addition, associations between these genes and clinical factors were evaluated. Survival analysis, using the log-rank test, assessed overall survival (OS), disease-specific survival (DSS), and progression-free interval (PFI) as end points. The authors also locate hub Complement 3 gene position by immunofluorescence. Results: The study identified C3 as a hub Complement gene associated with trastuzumab sensitivity. C3 shows higher expression in normal than tumor tissues. C3 was highly expressed in HER2-negative and early-stage BC, but showed no differences in lymph node or metastasis subgroups. High C3 expression correlated with better OS, DSS, and PFI, particularly in HER2+ patients. IHC analysis confirmed higher C3 expression in normal tissues with the lowest in triple-negative BC. Immunofluorescence findings suggest that C3 recruits complement receptor 2 to enhance trastuzumab efficacy in HER2+ patients. Conclusions: This finding highlights the potential of complement 3 to improve therapeutic outcomes and pave the way for more personalized treatment strategies in BC.
Collapse
Affiliation(s)
- Bo Chen
- Department of Medical Aesthetic Surgery, The First Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Lifen Huang
- Clinicopathological Diagnosis & Research Center, the Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
- Key Laboratory of Tumor Molecular Pathology of Guangxi Higher Education Institutes, Baise, China
| | - Morui Gui
- Department of Medical Aesthetic Surgery, The First Affiliated Hospital of Guilin Medical University, Guilin, China
| | | | - Rudy Leon De Wilde
- Pius-Hospital, University Hospital for Gynecology, University Medicine Oldenburg, Oldenburg, Germany
| | - Wenjie Shi
- Medical Faculty and University Hospital Magdeburg, Molecular and Experimental Surgery, Clinic for General, Visceral, Vascular and Transplantation Surgery, Otto-von-Guericke University, Magdeburg, Germany
| | - Hui Liu
- School of Public Health, Guilin Medical University, Guilin, China
| | - Zhenyu Gong
- Department of Medical Aesthetic Surgery, The First Affiliated Hospital of Guilin Medical University, Guilin, China
| |
Collapse
|
2
|
Cai J, Wang W, Cong D, Bai Y, Zhang W. Development of treatment strategies for advanced HER2-positive gastric cancer: Insights from clinical trials. Crit Rev Oncol Hematol 2025; 207:104617. [PMID: 39805409 DOI: 10.1016/j.critrevonc.2025.104617] [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: 04/05/2024] [Revised: 01/06/2025] [Accepted: 01/08/2025] [Indexed: 01/16/2025] Open
Abstract
HER2-positive gastric cancer (GC), a unique molecular subtype, has garnered significant interest in recent years. Here, we review clinical trial data on advanced HER2-positive GC from the past 15 years. Trastuzumab plus standard chemotherapy remain the first-line treatment. The initial survival benefits conferred by immune checkpoint inhibitors plus trastuzumab and standard chemotherapy are encouraging. The combination of ramucirumab and mono-chemotherapy, as well as the antibody conjugated drug trastuzumab deruxtecan, is the recommended second-line regimen. Treatment with immune checkpoint inhibitors plus ramucirumab and mono-chemotherapy shows promise. Despite the limited treatment options for third line and beyond, development of novel therapeutic strategies is expected. Although clinical cure of advanced HER2-positive GC is unlikely, current clinical studies offer valuable insight into regimens that prolong survival.
Collapse
Affiliation(s)
- Jing Cai
- Department of Pediatrics, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Wanning Wang
- Department of Nephrology, the First Hospital of Jilin University, Changchun 130021, China
| | - Dan Cong
- Department of Hematology and Oncology, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Yuansong Bai
- Department of Hematology and Oncology, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Wenlong Zhang
- Department of Hematology and Oncology, China-Japan Union Hospital of Jilin University, Changchun 130033, China.
| |
Collapse
|
3
|
Niu X, Liu W, Zhang Y, Liu J, Zhang J, Li B, Qiu Y, Zhao P, Wang Z, Wang Z. Cancer plasticity in therapy resistance: Mechanisms and novel strategies. Drug Resist Updat 2024; 76:101114. [PMID: 38924995 DOI: 10.1016/j.drup.2024.101114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 06/12/2024] [Accepted: 06/15/2024] [Indexed: 06/28/2024]
Abstract
Therapy resistance poses a significant obstacle to effective cancer treatment. Recent insights into cell plasticity as a new paradigm for understanding resistance to treatment: as cancer progresses, cancer cells experience phenotypic and molecular alterations, corporately known as cell plasticity. These alterations are caused by microenvironment factors, stochastic genetic and epigenetic changes, and/or selective pressure engendered by treatment, resulting in tumor heterogeneity and therapy resistance. Increasing evidence suggests that cancer cells display remarkable intrinsic plasticity and reversibly adapt to dynamic microenvironment conditions. Dynamic interactions between cell states and with the surrounding microenvironment form a flexible tumor ecosystem, which is able to quickly adapt to external pressure, especially treatment. Here, this review delineates the formation of cancer cell plasticity (CCP) as well as its manipulation of cancer escape from treatment. Furthermore, the intrinsic and extrinsic mechanisms driving CCP that promote the development of therapy resistance is summarized. Novel treatment strategies, e.g., inhibiting or reversing CCP is also proposed. Moreover, the review discusses the multiple lines of ongoing clinical trials globally aimed at ameliorating therapy resistance. Such advances provide directions for the development of new treatment modalities and combination therapies against CCP in the context of therapy resistance.
Collapse
Affiliation(s)
- Xing Niu
- China Medical University, Shenyang, Liaoning 110122, China; Experimental Center of BIOQGene, YuanDong International Academy Of Life Sciences, 999077, Hong Kong, China
| | - Wenjing Liu
- Medical Oncology Department of Thoracic Cancer (2), Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China
| | - Yinling Zhang
- Department of Oncology Radiotherapy 1, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, Shandong 266042, China
| | - Jing Liu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Jianjun Zhang
- Department of Gastric Surgery, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China
| | - Bo Li
- Department of Orthopedics, Beijing Luhe Hospital, Capital Medical University, Beijing 101149, China
| | - Yue Qiu
- Department of Digestive Diseases 1, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China
| | - Peng Zhao
- Department of Medical Imaging, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China
| | - Zhongmiao Wang
- Department of Digestive Diseases 1, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China.
| | - Zhe Wang
- Department of Digestive Diseases 1, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China.
| |
Collapse
|
4
|
Zhang R, Zhang D, Han F, Song X, Zhang Y, Zhang J, Zhu Q, Qin Y. The deubiquitinase USP7 and E3 ligase TRIM21 regulate vasculogenic mimicry and malignant progression of RMS by balancing SNAI2 homeostasis. J Exp Clin Cancer Res 2024; 43:135. [PMID: 38702792 PMCID: PMC11069146 DOI: 10.1186/s13046-024-03056-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 04/22/2024] [Indexed: 05/06/2024] Open
Abstract
BACKGROUND Rhabdomyosarcoma (RMS) is a rare malignancy and the most common soft tissue sarcoma in children. Vasculogenic mimicry (VM) is a novel tumor microcirculation model different from traditional tumor angiogenesis, which does not rely on endothelial cells to provide sufficient blood supply for tumor growth. In recent years, VM has been confirmed to be closely associated with tumor progression. However, the ability of RMS to form VM has not yet been reported. METHODS Immunohistochemistry, RT-qPCR and western blot were used to test the expression level of SNAI2 and its clinical significance. The biological function in regulating vasculogenic mimicry and malignant progression of SNAI2 was examined both in vitro and in vivo. Mass spectrometry, co-immunohistochemistry, immunofluorescence staining, and ubiquitin assays were performed to explore the regulatory mechanism of SNAI2. RESULTS Our study indicated that SNAI2 was abnormally expressed in patients with RMS and RMS cell lines and promoted the proliferation and metastasis of RMS. Through cell tubule formation experiments, nude mice Matrigel plug experiments, and immunohistochemistry (IHC), we confirmed that RMS can form VM and that SNAI2 promotes the formation of VM. Due to SNAI2 is a transcription factor that is not easily drugged, we used Co-IP combined with mass spectrometry to screen for the SNAI2-binding protein USP7 and TRIM21. USP7 depletion inhibited RMS VM formation, proliferation and metastasis by promoting SNAI2 degradation. We further demonstrated that TRIM21 is expressed at low levels in human RMS tissues and inhibits VM in RMS cells. TRIM21 promotes SNAI2 protein degradation through ubiquitination in the RMS. The deubiquitinase USP7 and E3 ligase TRIM21 function in an antagonistic rather than competitive mode and play a key role in controlling the stability of SNAI2 to determine the VM formation and progression of RMS. CONCLUSION Our findings reveal a previously unknown mechanism by which USP7 and TRIM21 balance the level of SNAI2 ubiquitination, determining RMS vasculogenic mimicry, proliferation, and migration. This new mechanism may provide new targeted therapies to inhibit the development of RMS by restoring TRIM21 expression or inhibiting USP7 expression in RMS patients with high SNAI2 protein levels.
Collapse
Affiliation(s)
- Ruyue Zhang
- Department of Clinical Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Daidi Zhang
- Department of Clinical Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Fugen Han
- Department of Otorhinolaryngology Head and Neck surgery, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450052, China
| | - Xiaorui Song
- Henan Province Engineering Research Center of Diagnosis and Treatment of Pediatric Infection and Critical Care, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450052, China
| | - Yaodong Zhang
- Henan Province Engineering Research Center of Diagnosis and Treatment of Pediatric Infection and Critical Care, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450052, China
| | - Jie Zhang
- Department of Otorhinolaryngology Head and Neck surgery, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450052, China.
- Department of Otorhinolaryngology Head and Neck surgery, National Center for Children's Health, Beijing Children's Hospital Capital Medical University, Beijing, 10045, China.
| | - Qingwen Zhu
- Department of Otorhinolaryngology Head and Neck surgery, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450052, China.
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Yanru Qin
- Department of Clinical Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
5
|
Zhang R, Yao Y, Gao H, Hu X. Mechanisms of angiogenesis in tumour. Front Oncol 2024; 14:1359069. [PMID: 38590656 PMCID: PMC10999665 DOI: 10.3389/fonc.2024.1359069] [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: 12/20/2023] [Accepted: 03/11/2024] [Indexed: 04/10/2024] Open
Abstract
Angiogenesis is essential for tumour growth and metastasis. Antiangiogenic factor-targeting drugs have been approved as first line agents in a variety of oncology treatments. Clinical drugs frequently target the VEGF signalling pathway during sprouting angiogenesis. Accumulating evidence suggests that tumours can evade antiangiogenic therapy through other angiogenesis mechanisms in addition to the vascular sprouting mechanism involving endothelial cells. These mechanisms include (1) sprouting angiogenesis, (2) vasculogenic mimicry, (3) vessel intussusception, (4) vascular co-option, (5) cancer stem cell-derived angiogenesis, and (6) bone marrow-derived angiogenesis. Other non-sprouting angiogenic mechanisms are not entirely dependent on the VEGF signalling pathway. In clinical practice, the conversion of vascular mechanisms is closely related to the enhancement of tumour drug resistance, which often leads to clinical treatment failure. This article summarizes recent studies on six processes of tumour angiogenesis and provides suggestions for developing more effective techniques to improve the efficacy of antiangiogenic treatment.
Collapse
Affiliation(s)
| | | | | | - Xin Hu
- China–Japan Union Hospital of Jilin University, Jilin University, Changchun, China
| |
Collapse
|
6
|
Rönnlund C, Sifakis EG, Schagerholm C, Yang Q, Karlsson E, Chen X, Foukakis T, Weidler J, Bates M, Fredriksson I, Robertson S, Hartman J. Prognostic impact of HER2 biomarker levels in trastuzumab-treated early HER2-positive breast cancer. Breast Cancer Res 2024; 26:24. [PMID: 38321542 PMCID: PMC10848443 DOI: 10.1186/s13058-024-01779-9] [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: 11/24/2023] [Accepted: 01/24/2024] [Indexed: 02/08/2024] Open
Abstract
BACKGROUND Overexpression of human epidermal growth factor receptor 2 (HER2) caused by HER2 gene amplification is a driver in breast cancer tumorigenesis. We aimed to investigate the prognostic significance of manual scoring and digital image analysis (DIA) algorithm assessment of HER2 copy numbers and HER2/CEP17 ratios, along with ERBB2 mRNA levels among early-stage HER2-positive breast cancer patients treated with trastuzumab. METHODS This retrospective study comprised 371 early HER2-positive breast cancer patients treated with adjuvant trastuzumab, with HER2 re-testing performed on whole tumor sections. Digitized tumor tissue slides were manually scored and assessed with uPath HER2 Dual ISH image analysis, breast algorithm. Targeted ERBB2 mRNA levels were assessed by the Xpert® Breast Cancer STRAT4 Assay. HER2 copy number and HER2/CEP17 ratio from in situ hybridization assessment, along with ERBB2 mRNA levels, were explored in relation to recurrence-free survival (RFS). RESULTS The analysis showed that patients with tumors with the highest and lowest manually counted HER2 copy number levels had worse RFS than those with intermediate levels (HR = 2.7, CI 1.4-5.3, p = 0.003 and HR = 2.1, CI 1.1-3.9, p = 0.03, respectively). A similar trend was observed for HER2/CEP17 ratio, and the DIA algorithm confirmed the results. Moreover, patients with tumors with the highest and the lowest values of ERBB2 mRNA had a significantly worse prognosis (HR = 2.7, CI 1.4-5.1, p = 0.003 and HR = 2.8, CI 1.4-5.5, p = 0.004, respectively) compared to those with intermediate levels. CONCLUSIONS Our findings suggest that the association between any of the three HER2 biomarkers and RFS was nonlinear. Patients with tumors with the highest levels of HER2 gene amplification or ERBB2 mRNA were associated with a worse prognosis than those with intermediate levels, which is of importance to investigate in future clinical trials studying HER2-targeted therapy.
Collapse
Affiliation(s)
- Caroline Rönnlund
- Department of Oncology and Pathology, Karolinska Institutet, Visionsgatan 56, CCK R8:04, 17176, Stockholm, Sweden.
- Department of Clinical Pathology and Cancer Diagnostics, Karolinska University Hospital, Stockholm, Sweden.
| | - Emmanouil G Sifakis
- Department of Oncology and Pathology, Karolinska Institutet, Visionsgatan 56, CCK R8:04, 17176, Stockholm, Sweden
| | - Caroline Schagerholm
- Department of Oncology and Pathology, Karolinska Institutet, Visionsgatan 56, CCK R8:04, 17176, Stockholm, Sweden
| | - Qiao Yang
- Department of Oncology and Pathology, Karolinska Institutet, Visionsgatan 56, CCK R8:04, 17176, Stockholm, Sweden
| | - Emelie Karlsson
- Department of Oncology and Pathology, Karolinska Institutet, Visionsgatan 56, CCK R8:04, 17176, Stockholm, Sweden
| | - Xinsong Chen
- Department of Oncology and Pathology, Karolinska Institutet, Visionsgatan 56, CCK R8:04, 17176, Stockholm, Sweden
| | - Theodoros Foukakis
- Department of Oncology and Pathology, Karolinska Institutet, Visionsgatan 56, CCK R8:04, 17176, Stockholm, Sweden
- Breast Center, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - Jodi Weidler
- Medical and Scientific Affairs and Strategy, Oncology, Cepheid, Sunnyvale, CA, USA
| | - Michael Bates
- Medical and Scientific Affairs and Strategy, Oncology, Cepheid, Sunnyvale, CA, USA
| | - Irma Fredriksson
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Department of Breast-, Endocrine Tumors and Sarcoma, Karolinska University Hospital, Stockholm, Sweden
| | - Stephanie Robertson
- Department of Oncology and Pathology, Karolinska Institutet, Visionsgatan 56, CCK R8:04, 17176, Stockholm, Sweden
| | - Johan Hartman
- Department of Oncology and Pathology, Karolinska Institutet, Visionsgatan 56, CCK R8:04, 17176, Stockholm, Sweden
- Department of Clinical Pathology and Cancer Diagnostics, Karolinska University Hospital, Stockholm, Sweden
- Medtechlabs, Bioclinicum, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
7
|
Sarkar S, Deyoung T, Ressler H, Chandler W. Brain Tumors: Development, Drug Resistance, and Sensitization - An Epigenetic Approach. Epigenetics 2023; 18:2237761. [PMID: 37499114 PMCID: PMC10376921 DOI: 10.1080/15592294.2023.2237761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 06/26/2023] [Accepted: 07/11/2023] [Indexed: 07/29/2023] Open
Abstract
In this article, we describe contrasting developmental aspects of paediatric and adult brain tumours. We hypothesize that the formation of cancer progenitor cells, for both paediatric and adult, could be due to epigenetic events. However, the progression of adult brain tumours selectively involves more mutations compared to paediatric tumours. We further discuss epigenetic switches, comprising both histone modifications and DNA methylation, and how they can differentially regulate transcription and expression of oncogenes and tumour suppressor genes. Next, we summarize the currently available therapies for both types of brain tumours, explaining the merits and failures leading to drug resistance. We analyse different mechanisms of drug resistance and the role of epigenetics in this process. We then provide a rationale for combination therapy, which includes epigenetic drugs. In the end, we postulate a concept which describes how a combination therapy could be initiated. The timing, doses, and order of individual drug regimens will depend on the individual case. This type of combination therapy will be part of a personalized medicine which will differ from patient to patient.
Collapse
Affiliation(s)
- Sibaji Sarkar
- Division of Biotechnology, Quincy College, Quincy, MA, USA
- Division of Biology, STEM, MBC College, Wellesley, MA, USA
- Division of Biology, STEM, RC College Boston, Boston, MA, USA
| | - Tara Deyoung
- Division of Biotechnology, Quincy College, Quincy, MA, USA
| | - Hope Ressler
- Division of Biology, STEM, MBC College, Wellesley, MA, USA
| | | |
Collapse
|
8
|
Lin X, Long S, Yan C, Zou X, Zhang G, Zou J, Wu G. Therapeutic potential of vasculogenic mimicry in urological tumors. Front Oncol 2023; 13:1202656. [PMID: 37810976 PMCID: PMC10551447 DOI: 10.3389/fonc.2023.1202656] [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: 04/09/2023] [Accepted: 09/06/2023] [Indexed: 10/10/2023] Open
Abstract
Angiogenesis is an essential process in the growth and metastasis of cancer cells, which can be hampered by an anti-angiogenesis mechanism, thereby delaying the progression of tumors. However, the benefit of this treatment modality could be restricted, as most patients tend to develop acquired resistance during treatment. Vasculogenic mimicry (VM) is regarded as a critical alternative mechanism of tumor angiogenesis, where studies have demonstrated that patients with tumors supplemented with VM generally have a shorter survival period and a poorer prognosis. Inhibiting VM may be an effective therapeutic strategy to prevent cancer progression, which could prove helpful in impeding the limitations of lone use of anti-angiogenic therapy when performed concurrently with other anti-tumor therapies. This review summarizes the mechanism of VM signaling pathways in urological tumors, i.e., prostate cancer, clear cell renal cell carcinoma, and bladder cancer. Furthermore, it also summarizes the potential of VM as a therapeutic strategy for urological tumors.
Collapse
Affiliation(s)
- Xinyu Lin
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Sheng Long
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Congcong Yan
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Xiaofeng Zou
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Guoxi Zhang
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Junrong Zou
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Gengqing Wu
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| |
Collapse
|
9
|
Wu J, Zhang L, Li W, Wang L, Jia Q, Shi F, Li K, Liao L, Shi Y, Wu S. The role of TOP2A in immunotherapy and vasculogenic mimicry in non-small cell lung cancer and its potential mechanism. Sci Rep 2023; 13:10906. [PMID: 37407689 DOI: 10.1038/s41598-023-38117-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 07/03/2023] [Indexed: 07/07/2023] Open
Abstract
Type IIA topoisomerase (TOP2A) is significantly associated with malignant tumor development, invasion, treatment and its prognosis, and has been shown to be a therapeutic target against cancer. In contrast, the role of TOP2A in the immunotherapy of non-small cell lung cancer as well as in Vasculogenic mimicry (VM) formation and its potential mechanisms are unclear. The aim of this study was to investigate the role of TOP2A in proliferation, skeleton regulation, motility and VM production in non-small cell lung cancer and its mechanisms by using bioinformatics tools and molecular biology experiments. Subgroup analysis showed that the low-risk group had a better prognosis, while the high-risk group was positively correlated with high tumor mutational load, M1-type macrophage infiltration, immune checkpoint molecule expression, and immunotherapy efficacy. As confirmed by further clinical specimens, the presence of TOP2A and VM was significantly and positively correlated with poor prognosis. Our study established a model based on significant co-expression of TOP2A genes, which significantly correlated with mutational load and immunotherapy outcomes in patients with non-small cell lung cancer. Further mechanistic exploration suggests that TOP2A plays an important role in immunotherapy and VM formation in NSCLC through upregulation of Wnt3a and PD-L1 expression.
Collapse
Affiliation(s)
- Jiatao Wu
- Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, First Affiliated Hospital, Bengbu Medical College, 287 Changhuai Road, Bengbu, 233004, Anhui, China
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, People's Republic of China
| | - Lei Zhang
- Department of Oncology Surgery, The Second Affiliated Hospital of Bengbu Medical College, Bengbu, 233080, Anhui, China
| | - Wenjuan Li
- Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, First Affiliated Hospital, Bengbu Medical College, 287 Changhuai Road, Bengbu, 233004, Anhui, China
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, People's Republic of China
| | - Luyao Wang
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, People's Republic of China
| | - Qianhao Jia
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, People's Republic of China
| | - Fan Shi
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, People's Republic of China
| | - Kairui Li
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, People's Republic of China
| | - Lingli Liao
- Department of Radiation Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, Anhui, China
| | - Yuqi Shi
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, People's Republic of China
| | - Shiwu Wu
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, People's Republic of China.
- Department of Pathology, Bengbu Medical College, Bengbu, 233030, Anhui, China.
- Department of pathology, Anhui No.2 Provincial People's Hospital, Bengbu, China.
| |
Collapse
|
10
|
Contreras-Sanzón E, Carlos-Reyes Á, Sierra-Martínez M, Acosta-Altamirano G, Luna-Rivero C, Núñez-Corona D, García-Hernández AP, Ibarra-Sierra E, Vidrio-Morgado H, Alvarez-Sánchez ME, Marchat LA, López-Camarillo C. Metastatic breast tumors downregulate miR-145 regulating the hypoxia-induced vasculogenic mimicry. Transl Oncol 2023; 33:101680. [PMID: 37121177 PMCID: PMC10172997 DOI: 10.1016/j.tranon.2023.101680] [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: 01/06/2023] [Revised: 03/06/2023] [Accepted: 04/22/2023] [Indexed: 05/02/2023] Open
Abstract
Tumor cells grow in three-dimensional (3D) channels-like structures denoted as vasculogenic mimicry (VM), which provides a route for nutrients and oxygen acquisition. VM is activated by hypoxia and associated with metastasis and poor prognosis. MetastamiRs are microRNAs regulating metastasis, however, if they control VM in breast cancer remains poorly understood. The aim of this study was to evaluate the expression of VM-associated microRNAs in tumors of metastatic breast cancer patients. Firstly, we constructed microRNAs/mRNAs coregulation networks using expression data from TCGA databases. Dozens of microRNAs regulating genes involved in VM and metastasis were found. Of these, we selected 10 microRNAs for further characterization. The presence of VM in histological samples from patients with or without metastasis was evaluated using CD31-/PAS+ immunophenotyping. Remarkably, data showed that VM was significantly increased in tumors from patients with metastasis in comparison with no-metastatic group. Gene expression analysis indicated that miR-145, miR-142-3p, miR-31, miR-148a, miR-200b-3p and miR-526b were downregulated in primary tumors from patients with metastatic disease and positive for VM. Moreover, modulated microRNAs showed a predictive clinical value in overall survival in a cohort (n=1262) of breast cancer patients. Of these, we evaluated the role of miR-145 in formation of hypoxia-induced 3D channels-like using an in vitro model that recapitulates the early stages of VM. Data showed that miR-145 mimics was able to abolish the VM development in both metastatic Hs578t and MDA-MB-231 breast cancer cells. In conclusion, manipulation of miR-145 levels may represent a therapeutic approach in metastatic breast cancer patients that developed VM.
Collapse
Affiliation(s)
| | - Ángeles Carlos-Reyes
- Laboratorio de Onco-inmunobiologia, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas", CDMX, Mexico
| | - Mónica Sierra-Martínez
- Unidad de Investigación en Salud del Hospital Regional de Alta Especialidad de Ixtapaluca, Estado de Mexico, Mexico
| | - Gustavo Acosta-Altamirano
- Dirección de Planeación, Enseñanza e Investigación del Hospital Regional de Alta Especialidad de Ixtapaluca, Estado de Mexico, Mexico
| | - Cesar Luna-Rivero
- Servicio de Patología, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas", CDMX, Mexico
| | - David Núñez-Corona
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, CDMX, México
| | | | - Eloisa Ibarra-Sierra
- Departamento de Investigacion. Instituto Estatal de Cancerologia "Dr. Arturo Béltran Ortega", Acapulco, Guerrero, Mexico
| | - Horacio Vidrio-Morgado
- Departamento de Investigacion. Instituto Estatal de Cancerologia "Dr. Arturo Béltran Ortega", Acapulco, Guerrero, Mexico
| | | | - Laurence A Marchat
- Laboratorio de Biomedicina Molecular II, Programa en Biomedicina Molecular y Red de Biotecnologia, Instituto Politecnico Nacional, CDMX, Mexico
| | - César López-Camarillo
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, CDMX, México.
| |
Collapse
|
11
|
Cabello P, Torres-Ruiz S, Adam-Artigues A, Forés-Martos J, Martínez MT, Hernando C, Zazo S, Madoz-Gúrpide J, Rovira A, Burgués O, Rojo F, Albanell J, Lluch A, Bermejo B, Cejalvo JM, Eroles P. miR-146a-5p Promotes Angiogenesis and Confers Trastuzumab Resistance in HER2+ Breast Cancer. Cancers (Basel) 2023; 15:cancers15072138. [PMID: 37046799 PMCID: PMC10093389 DOI: 10.3390/cancers15072138] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 03/17/2023] [Accepted: 03/31/2023] [Indexed: 04/08/2023] Open
Abstract
Trastuzumab treatment has significantly improved the prognosis of HER2-positive breast cancer patients. Despite this, resistance to therapy still remains the main clinical challenge. In order to evaluate the implication of microRNAs in the trastuzumab response, we performed a microRNA array in parental and acquired trastuzumab-resistant HER2-positive breast cancer cell lines. Our results identified miR-146a-5p as the main dysregulated microRNA. Interestingly, high miR-146a-5p expression in primary tumor tissue significantly correlated with shorter disease-free survival in HER2-positive breast cancer patients. The gain- and loss-of-function of miR-146a-5p modulated the response to trastuzumab. Furthermore, the overexpression of miR-146a-5p increased migration and angiogenesis, and promoted cell cycle progression by reducing CDKN1A expression. Exosomes from trastuzumab-resistant cells showed a high level of miR-146a-5p expression compared with the parental cells. In addition, the co-culture with resistant cells’ exosomes was able to decrease in sensitivity and increase the migration capacities in trastuzumab-sensitive cells, as well as angiogenesis in HUVEC-2 cells. Collectively, these data support the role of miR-146a-5p in resistance to trastuzumab, and demonstrate that it can be transferred by exosomes conferring resistance properties to other cells.
Collapse
Affiliation(s)
- Paula Cabello
- Biomedical Research Institute INCLIVA, 46010 Valencia, Spain
- International University of Valencia—VIU, 46002 Valencia, Spain
| | | | | | | | - María Teresa Martínez
- Biomedical Research Institute INCLIVA, 46010 Valencia, Spain
- Department of Medical Oncology, University Clinical Hospital of Valencia, 46010 Valencia, Spain
| | - Cristina Hernando
- Biomedical Research Institute INCLIVA, 46010 Valencia, Spain
- Department of Medical Oncology, University Clinical Hospital of Valencia, 46010 Valencia, Spain
| | - Sandra Zazo
- Department of Pathology, Jiménez Díaz Foundation, 28040 Madrid, Spain
| | | | - Ana Rovira
- Center for Biomedical Network Research on Cancer (CIBERONC), 28040 Madrid, Spain
- Department of Medical Oncology, Hospital del Mar, 08003 Barcelona, Spain
- Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), 08003 Barcelona, Spain
| | - Octavio Burgués
- Biomedical Research Institute INCLIVA, 46010 Valencia, Spain
- Center for Biomedical Network Research on Cancer (CIBERONC), 28040 Madrid, Spain
- Department of Pathology, University Clinical Hospital of Valencia, 46010 Valencia, Spain
| | - Federico Rojo
- Department of Pathology, Jiménez Díaz Foundation, 28040 Madrid, Spain
- Center for Biomedical Network Research on Cancer (CIBERONC), 28040 Madrid, Spain
| | - Joan Albanell
- Center for Biomedical Network Research on Cancer (CIBERONC), 28040 Madrid, Spain
- Department of Medical Oncology, Hospital del Mar, 08003 Barcelona, Spain
- Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), 08003 Barcelona, Spain
| | - Ana Lluch
- Biomedical Research Institute INCLIVA, 46010 Valencia, Spain
- Department of Medical Oncology, University Clinical Hospital of Valencia, 46010 Valencia, Spain
- Center for Biomedical Network Research on Cancer (CIBERONC), 28040 Madrid, Spain
- Department of Medicine, University of Valencia, 46010 Valencia, Spain
| | - Begoña Bermejo
- Biomedical Research Institute INCLIVA, 46010 Valencia, Spain
- Department of Medical Oncology, University Clinical Hospital of Valencia, 46010 Valencia, Spain
- Center for Biomedical Network Research on Cancer (CIBERONC), 28040 Madrid, Spain
| | - Juan Miguel Cejalvo
- Biomedical Research Institute INCLIVA, 46010 Valencia, Spain
- Department of Medical Oncology, University Clinical Hospital of Valencia, 46010 Valencia, Spain
- Center for Biomedical Network Research on Cancer (CIBERONC), 28040 Madrid, Spain
| | - Pilar Eroles
- Biomedical Research Institute INCLIVA, 46010 Valencia, Spain
- Center for Biomedical Network Research on Cancer (CIBERONC), 28040 Madrid, Spain
- Department of Physiology, University of Valencia, 46010 Valencia, Spain
- Department of Biotechnology, Polytechnic University of Valencia, 46022 Valencia, Spain
| |
Collapse
|
12
|
Xu L, Han F, Zhu L, Ding W, Zhang K, Kan C, Hou N, Li Q, Sun X. Advances in understanding the role and mechanisms of tumor stem cells in HER2-positive breast cancer treatment resistance (Review). Int J Oncol 2023; 62:48. [PMID: 36866766 PMCID: PMC9990588 DOI: 10.3892/ijo.2023.5496] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 02/15/2023] [Indexed: 03/04/2023] Open
Abstract
Approximately 15-20% of breast carcinomas exhibit human epidermal growth factor receptor (HER2) protein overexpression. HER2-positive breast cancer (BC) is a heterogeneous and aggressive subtype with poor prognosis and high relapse risk. Although several anti-HER2 drugs have achieved substantial efficacy, certain patients with HER2-positive BC relapse due to drug resistance after a treatment period. There is increasing evidence that BC stem cells (BCSCs) drive therapeutic resistance and a high rate of BC recurrence. BCSCs may regulate cellular self-renewal and differentiation, as well as invasive metastasis and treatment resistance. Efforts to target BCSCs may yield new methods to improve patient outcomes. In the present review, the roles of BCSCs in the occurrence, development and management of BC treatment resistance were summarized; BCSC-targeted strategies for the treatment of HER2-positive BC were also discussed.
Collapse
Affiliation(s)
- Linfei Xu
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261031, P.R. China
- Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261031, P.R. China
- Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261031, P.R. China
| | - Fang Han
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261031, P.R. China
- Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261031, P.R. China
- Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261031, P.R. China
| | - Liang Zhu
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261031, P.R. China
- Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261031, P.R. China
| | - Wenli Ding
- Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261031, P.R. China
| | - Kexin Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261031, P.R. China
- Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261031, P.R. China
| | - Chengxia Kan
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261031, P.R. China
- Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261031, P.R. China
| | - Ningning Hou
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261031, P.R. China
- Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261031, P.R. China
| | - Qinying Li
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261031, P.R. China
- Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261031, P.R. China
| | - Xiaodong Sun
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261031, P.R. China
- Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261031, P.R. China
| |
Collapse
|
13
|
Microbiota-Derived Natural Products Targeting Cancer Stem Cells: Inside the Gut Pharma Factory. Int J Mol Sci 2023; 24:ijms24054997. [PMID: 36902427 PMCID: PMC10003410 DOI: 10.3390/ijms24054997] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/28/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
Cancer stem cells (CSCs) have drawn much attention as important tumour-initiating cells that may also be crucial for recurrence after chemotherapy. Although the activity of CSCs in various forms of cancer is complex and yet to be fully elucidated, opportunities for therapies targeting CSCs exist. CSCs are molecularly distinct from bulk tumour cells, so they can be targeted by exploiting their signature molecular pathways. Inhibiting stemness has the potential to reduce the risk posed by CSCs by limiting or eliminating their capacity for tumorigenesis, proliferation, metastasis, and recurrence. Here, we briefly described the role of CSCs in tumour biology, the mechanisms involved in CSC therapy resistance, and the role of the gut microbiota in cancer development and treatment, to then review and discuss the current advances in the discovery of microbiota-derived natural compounds targeting CSCs. Collectively, our overview suggests that dietary intervention, toward the production of those identified microbial metabolites capable of suppressing CSC properties, is a promising approach to support standard chemotherapy.
Collapse
|
14
|
Nakajima M, Kawahara R, Simizu S. Cofilin promotes vasculogenic mimicry by regulating the actin cytoskeleton in human breast cancer cells. FEBS Lett 2023; 597:1114-1124. [PMID: 36737242 DOI: 10.1002/1873-3468.14594] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/16/2023] [Accepted: 01/27/2023] [Indexed: 02/05/2023]
Abstract
Vasculogenic mimicry (VM) is the formation of microvascular channels by cancer cells. VM requires cellular processes that are regulated by changes in cellular migration and morphology. Cofilin (CFL), a key regulator of actin depolymerization, has been reported to affect malignant phenotypes of cancer. We show that treatment with inhibitors of actin dynamics suppresses VM in MDA-MB-231 human breast cancer cells. We established CFL-knockout (KO) MDA-MB-231 cells and found that VM was attenuated in CFL-KO cells. Although the re-expression of wild-type CFL restored VM in CFL-KO cells, inactive phosphomimetic CFL failed to do so. Collectively, our results demonstrate that CFL is a critical regulator of VM and implicate CFL as a novel therapeutic target for breast cancer.
Collapse
Affiliation(s)
- Minami Nakajima
- Department of Applied Chemistry, Faculty of Science and Technology, Keio University, Yokohama, Japan
| | - Ryota Kawahara
- Department of Applied Chemistry, Faculty of Science and Technology, Keio University, Yokohama, Japan
| | - Siro Simizu
- Department of Applied Chemistry, Faculty of Science and Technology, Keio University, Yokohama, Japan
| |
Collapse
|
15
|
Resistance to Trastuzumab. Cancers (Basel) 2022; 14:cancers14205115. [PMID: 36291900 PMCID: PMC9600208 DOI: 10.3390/cancers14205115] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 10/12/2022] [Accepted: 10/17/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Trastuzumab is a humanized antibody that has significantly improved the management and treatment outcomes of patients with cancers that overexpress HER2. Many research groups, both in academia and industry, have contributed towards understanding the various mechanisms engaged by trastuzumab to mediate its anti-tumor effects. Nevertheless, data from several clinical studies have indicated that a significant proportion of patients exhibit primary or acquired resistance to trastuzumab therapy. In this article, we discuss underlying mechanisms that contribute towards to resistance. Furthermore, we discuss the potential strategies to overcome some of the mechanisms of resistance to enhance the therapeutic efficacy of trastuzumab and other therapies based on it. Abstract One of the most impactful biologics for the treatment of breast cancer is the humanized monoclonal antibody, trastuzumab, which specifically recognizes the HER2/neu (HER2) protein encoded by the ERBB2 gene. Useful for both advanced and early breast cancers, trastuzumab has multiple mechanisms of action. Classical mechanisms attributed to trastuzumab action include cell cycle arrest, induction of apoptosis, and antibody-dependent cell-mediated cytotoxicity (ADCC). Recent studies have identified the role of the adaptive immune system in the clinical actions of trastuzumab. Despite the multiple mechanisms of action, many patients demonstrate resistance, primary or adaptive. Newly identified molecular and cellular mechanisms of trastuzumab resistance include induction of immune suppression, vascular mimicry, generation of breast cancer stem cells, deregulation of long non-coding RNAs, and metabolic escape. These newly identified mechanisms of resistance are discussed in detail in this review, particularly considering how they may lead to the development of well-rationalized, patient-tailored combinations that improve patient survival.
Collapse
|
16
|
Hu H, Ma T, Liu N, Hong H, Yu L, Lyu D, Meng X, Wang B, Jiang X. Immunotherapy checkpoints in ovarian cancer vasculogenic mimicry: Tumor immune microenvironments, and drugs. Int Immunopharmacol 2022; 111:109116. [PMID: 35969899 DOI: 10.1016/j.intimp.2022.109116] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 07/27/2022] [Accepted: 07/29/2022] [Indexed: 02/09/2023]
Abstract
Vasculogenic mimicry (VM), a vessel-like structure independent of endothelial cells, commonly exists in solid tumors which requires blood vessels to grow. As a special source of blood supply for tumor progression to a more aggressive state, VM has been observed in a variety of human malignant tumors and is tightly associated with tumor proliferation, invasion, metastasis, and poor patient prognosis. So far, various factors, including immune cells and cytokines, were reported to regulate ovarian cancer progression by influencing VM formation. Herein, we review the mechanisms that regulate VM formation in ovarian cancer and the effect of cells, cytokines, and signaling molecules in the tumor microenvironment on VM formation, Furthermore, we summarize the current clinical application of drugs targeting VM formation.
Collapse
Affiliation(s)
- Haitao Hu
- Cancer Hospital of China Medical University, No. 44 Xiaoheyan Road, Dadong District, Shenyang 110042, Liaoning Province, PR China.
| | - Ting Ma
- Department of Biochemistry and Molecular Biology, College of Life Science, China Medical University, Shenyang 110122, Liaoning Province, PR China.
| | - Nanqi Liu
- Department of Biochemistry and Molecular Biology, College of Life Science, China Medical University, Shenyang 110122, Liaoning Province, PR China.
| | - Hong Hong
- Department of Geriatrics, The First Hospital of China Medical University, Shenyang 110001, Liaoning Province, PR China.
| | - Lujiao Yu
- Department of Geriatrics, The First Hospital of China Medical University, Shenyang 110001, Liaoning Province, PR China.
| | - Dantong Lyu
- Department of Biochemistry and Molecular Biology, College of Life Science, China Medical University, Shenyang 110122, Liaoning Province, PR China.
| | - Xin Meng
- Department of Biochemistry and Molecular Biology, College of Life Science, China Medical University, Shenyang 110122, Liaoning Province, PR China.
| | - Biao Wang
- Department of Biochemistry and Molecular Biology, College of Life Science, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning Province, PR China.
| | - Xuefeng Jiang
- Department of Immunology, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, PR China.
| |
Collapse
|
17
|
Bajbouj K, Al-Ali A, Shafarin J, Sahnoon L, Sawan A, Shehada A, Elkhalifa W, Saber-Ayad M, Muhammad J, Elmoselhi AB, Guraya S, Hamad M. Vitamin D Exerts Significant Antitumor Effects by Suppressing Vasculogenic Mimicry in Breast Cancer Cells. Front Oncol 2022; 12:918340. [PMID: 35747793 PMCID: PMC9210804 DOI: 10.3389/fonc.2022.918340] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/09/2022] [Indexed: 12/24/2022] Open
Abstract
BackgroundNumerous clinical and experimental observations have alluded to the substantial anti-neoplastic role of vitamin D in breast cancer (BC), primarily by inducing apoptosis and affecting metastasis. Tumor progression and resistance to chemotherapy have been linked to vasculogenic mimicry (VM), which represents the endothelial-independent formation of microvascular channels by cancer cells. However, the effect of vitamin D on VM formation in BC has not been thoroughly investigated. This study examined the impact of 1α,25-dihydroxyvitamin D3 (calcitriol), the active form of vitamin D, on the expression of major factors involved in BC migration, invasion, and VM formation.Experimental MethodsPublicly available transcriptomic datasets were used to profile the expression status of the key VM markers in vitamin D-treated BC cells. The in silico data were validated by examining the expression and activity of the key factors that are involved in tumor progression and MV formation in hormone-positive MCF-7 and aggressive triple‐negative MDA-MB-231 BC cells after treatment with calcitriol.Results and DiscussionsThe bioinformatics analysis showed that tumor VM formation-enriched pathways were differentially downregulated in vitamin D-treated cells when compared with control counterparts. Treatment of BC cells with calcitriol resulted in increased expression of tissue inhibitors of metalloproteinases (TIMPs 1 and 2) and decreased content and gelatinolytic activity of matrix metalloproteinases (MMPs 2 and 9). Furthermore, calcitriol treatment reduced the expression of several pro-MV formation regulators including vascular endothelial growth factor (VEGF), tumor growth factor (TGF-β1), and amphiregulin. Eventually, this process resulted in a profound reduction in cell migration and invasion following the treatment of BC cells with calcitriol when compared to the controls. Finally, the formation of VM was diminished in the aggressive triple‐negative MDA-MB-231 cancer cell line after calcitriol treatment.ConclusionOur findings demonstrate that vitamin D mediates its antitumor effects in BC cells by inhibiting and curtailing their potential for VM formation.
Collapse
Affiliation(s)
- Khuloud Bajbouj
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
- *Correspondence: Khuloud Bajbouj,
| | - Abeer Al-Ali
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Jasmin Shafarin
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Lina Sahnoon
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Ahmad Sawan
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Ahmed Shehada
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | | | - Maha Saber-Ayad
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
- Medical Pharmacology Department, Cairo University, Cairo, Egypt
| | - Jibran Sualeh Muhammad
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Adel B. Elmoselhi
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
- Department of Physiology, Michigan State University, East Lansing, MI, United States
| | - Salman Y. Guraya
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Mawieh Hamad
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
- College of Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
18
|
Vascular mimicry: A potential therapeutic target in breast cancer. Pathol Res Pract 2022; 234:153922. [DOI: 10.1016/j.prp.2022.153922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/19/2022] [Accepted: 04/24/2022] [Indexed: 10/18/2022]
|
19
|
Yang L, Higashisaka K, Shimoda M, Haga Y, Sekine N, Tsujino H, Nagano K, Shimazu K, Tsutsumi Y. Alpha-crystallin B chains in trastuzumab-resistant breast cancer cells promote endothelial cell tube formation through activating mTOR. Biochem Biophys Res Commun 2021; 588:175-181. [PMID: 34959190 DOI: 10.1016/j.bbrc.2021.12.056] [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: 11/05/2021] [Accepted: 12/15/2021] [Indexed: 01/18/2023]
Abstract
The specific human epidermal growth factor receptor 2 (HER2)-targeting monoclonal antibody trastuzumab shows considerable clinical efficacy in patients with HER2-overexpressing breast cancer. However, about 20% of patients who receive trastuzumab in the adjuvant setting relapse, and approximately half of patients with metastatic HER2-positive breast cancer develop resistance to trastuzumab within 1 year. Although the mechanism of trastuzumab resistance has been explored broadly, whether and how angiogenesis participates in trastuzumab resistance is unclear. Here, we examined the association between angiogenesis and trastuzumab resistance by using a trastuzumab-resistant cell line (SKBR3-TR). Compared with that from the parental trastuzumab-sensitive SKBR3 cells, the culture supernatant from SKBR3-TR cells significantly increased the sprouting of endothelial cells. To identify intercellular features that contribute to the induction of endothelial tube formation, proteomics revealed that α-crystallin B chain (αB-crystallin) was upregulated in SKBR3-TR cells. Moreover, silencing of αB-crystallin significantly repressed SKBR3-TR-induced tube formation, and knockdown of αB-crystallin in SKBR3-TR cells suppressed the activation of mechanistic target of rapamycin (mTOR) in endothelial cells. In addition, treatment with rapamycin, an inhibitor of mTOR, reversed the SKBR3-TR-induced promotion of tube formation. In summary, αB-crystallin enhanced the ability of SKBR3-TR cells to activate mTOR in endothelial cells and thus promote angiogenesis.
Collapse
Affiliation(s)
- Lili Yang
- Laboratory of Toxicology and Safety Science, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Kazuma Higashisaka
- Laboratory of Toxicology and Safety Science, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan; Institute for Advanced Co-Creation Studies, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Masafumi Shimoda
- Department of Breast and Endocrine Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Yuya Haga
- Laboratory of Toxicology and Safety Science, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Naoki Sekine
- Laboratory of Toxicology and Safety Science, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Hirofumi Tsujino
- Laboratory of Toxicology and Safety Science, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan; The Museum of Osaka University, 1-13 Machikaneyama, Toyonaka, Osaka, 560-0043, Japan.
| | - Kazuya Nagano
- Laboratory of Toxicology and Safety Science, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan; School of Pharmaceutical Sciences, Wakayama Medical University, 25-1 Shichibancho, Wakayama, Wakayama, 640-8156, Japan.
| | - Kenzo Shimazu
- Department of Breast and Endocrine Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Yasuo Tsutsumi
- Laboratory of Toxicology and Safety Science, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan; Global Center for Medical Engineering and Informatics, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
20
|
D’Andrea MR, Cereda V, Coppola L, Giordano G, Remo A, De Santis E. Propensity for Early Metastatic Spread in Breast Cancer: Role of Tumor Vascularization Features and Tumor Immune Infiltrate. Cancers (Basel) 2021; 13:5917. [PMID: 34885027 PMCID: PMC8657227 DOI: 10.3390/cancers13235917] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 11/17/2021] [Accepted: 11/22/2021] [Indexed: 02/05/2023] Open
Abstract
Breast cancer is a complex and highly heterogeneous disease consisting of various subtypes. It is classified into human epidermal growth receptor 2 (HER-2)-enriched, luminal A, luminal B and basal-like/triple negative (TNBC) breast cancer, based on histological and molecular features. At present, clinical decision-making in breast cancer is focused only on the assessment of tumor cells; nevertheless, it has been recognized that the tumor microenvironment (TME) plays a critical biologic role in breast cancer. This is constituted by a large group of immune and non-immune cells, but also by non-cellular components, such as several cytokines. TME is deeply involved in angiogenesis, immune-evasion strategies, and propensity for early metastatic spread, impacting on prognosis and prediction of response to specific treatments. In this review, we focused our attention on the early morphological changes of tumor microenvironment (tumor vasculature features, presence of immune and non-immune cells infiltrating the stroma, levels of cytokines) during breast cancer development. At the same time, we correlate these characteristics with early metastatic propensity (defined as synchronous metastasis or early recurrence) with particular attention to breast cancer subtypes.
Collapse
Affiliation(s)
- Mario Rosario D’Andrea
- Clinical Oncology Unit, San Paolo Hospital, Largo Donatori del Sangue 1, Civitavecchia, 00053 Rome, Italy;
| | - Vittore Cereda
- Clinical Oncology Unit, San Paolo Hospital, Largo Donatori del Sangue 1, Civitavecchia, 00053 Rome, Italy;
| | - Luigi Coppola
- Unit of Anatomy, Pathological Histology and Diagnostic Cytology, Department of Diagnostic and Pharma-Ceutical Services, Sandro Pertini Hospital, 00157 Rome, Italy;
| | - Guido Giordano
- Unit of Medical Oncology and Biomolecular Therapy, Department of Medical and Surgical Sciences, University of Foggia, Policlinico Riuniti, 71122 Foggia, Italy;
| | - Andrea Remo
- Pathology Unit, Mater Salutis Hospital, ULSS9, Legnago, 37045 Verona, Italy;
| | - Elena De Santis
- Department of Anatomical, Histological, Forensic Medicine and Orthopedic Sciences, Sapienza University of Rome, 00185 Rome, Italy;
| |
Collapse
|
21
|
Qi D, Liu Y, Li J, Huang JH, Hu X, Wu E. Salinomycin as a potent anticancer stem cell agent: State of the art and future directions. Med Res Rev 2021; 42:1037-1063. [PMID: 34786735 PMCID: PMC9298915 DOI: 10.1002/med.21870] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 10/22/2021] [Accepted: 10/26/2021] [Indexed: 12/11/2022]
Abstract
Cancer stem cells (CSCs) are a small subpopulation of cells within a tumor that can both self‐renew and differentiate into other cell types forming the heterogeneous tumor bulk. Since CSCs are involved in all aspects of cancer development, including tumor initiation, cell proliferation, metastatic dissemination, therapy resistance, and recurrence, they have emerged as attractive targets for cancer treatment and management. Salinomycin, a widely used antibiotic in poultry farming, was identified by the Weinberg group as a potent anti‐CSC agent in 2009. As a polyether ionophore, salinomycin exerts broad‐spectrum activities, including the important anti‐CSC function. Studies on the mechanism of action of salinomycin against cancer have been continuously and rapidly published since then. Thus, it is imperative for us to update its literature of recent research findings in this area. We here summarize the notable work reported on salinomycin's anticancer activities, intracellular binding target(s), effects on tumor microenvironment, safety, derivatives, and tumor‐specific drug delivery; after that we also discuss the translational potential of salinomycin toward clinical application based on current multifaceted understandings.
Collapse
Affiliation(s)
- Dan Qi
- Department of Neurosurgery, Baylor Scott & White Health, Temple, Texas, USA.,Neuroscience Institute, Baylor Scott & White Health, Temple, Texas, USA
| | - Yunyi Liu
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Molecular Engineering for Theranostics, Hunan University, Changsha, China
| | - Juan Li
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Molecular Engineering for Theranostics, Hunan University, Changsha, China
| | - Jason H Huang
- Department of Neurosurgery, Baylor Scott & White Health, Temple, Texas, USA.,Neuroscience Institute, Baylor Scott & White Health, Temple, Texas, USA.,Department of Surgery, Texas A&M University College of Medicine, Temple, Texas, USA
| | - Xiaoxiao Hu
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Molecular Engineering for Theranostics, Hunan University, Changsha, China.,Shenzhen Research Institute, Hunan University, Shenzhen, Guangdong, China
| | - Erxi Wu
- Department of Neurosurgery, Baylor Scott & White Health, Temple, Texas, USA.,Neuroscience Institute, Baylor Scott & White Health, Temple, Texas, USA.,Department of Surgery, Texas A&M University College of Medicine, Temple, Texas, USA.,LIVESTRONG Cancer Institutes and Department of Oncology, Dell Medical School, The University of Texas at Austin, Austin, Texas, USA.,Department of Pharmaceutical Sciences, Texas A&M University College of Pharmacy, College Station, Texas, USA
| |
Collapse
|
22
|
Maroufi NF, Rashidi M, Vahedian V, Jahanbazi R, Mostafaei S, Akbarzadeh M, Kazemzadeh H, Nejabati HR, Isazadeh A, Rashidi MR, Nouri M. Effect of Apatinib plus melatonin on vasculogenic mimicry formation by cancer stem cells from breast cancer cell line. Breast Cancer 2021; 29:260-273. [PMID: 34725795 DOI: 10.1007/s12282-021-01310-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 10/23/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND AIM Vasculogenic mimicry (VM) is one of the most important causes of breast cancer metastasis and resistance against drugs. The cancer stem cells (CSCs) are known as essential factors for VM formation. In this study, the effects of melatonin, Apatinib, and a combination of Apatinib/melatonin on VM formation were investigated by breast CSCs from breast cancer cell line. MATERIALS AND METHODS The percentage of CSCs was determined in two breast cancer cell lines (MCF-7 and MDA-MB-231) by flow cytometry. The effects of Apatinib, melatonin, and a combination of Apatinib/melatonin were evaluated on proliferation and viability, migration and invasion, apoptosis, and VM formation in MDA-MB-231 cells. Moreover, expression levels of the involved proteins in cancer cell proliferation and viability, CSCs, migration and invasion, and VM formation were evaluated by real-time polymerase chain reaction (RT-PCR) and western blotting methods. RESULTS Results of the present study showed that melatonin and Apatinib reduced survival rate of CSCs in a dose- and time-dependent manner. Apatinib, melatonin, and a combination of Apatinib/melatonin inhibited proliferation of breast CSCs (P ≤ 0.001). Formation of VM was decreased in the MDA-MB-231 cancer cell line treated with Apatinib and combination of Apatinib/melatonin. Apatinib and combination of Apatinib/melatonin reduced invasion of breast CSCs (P ≤ 0.0001). Expression of vascular endothelial VE-cadherin, ephrinA2 receptor (EPHA2), p-PI3K/phosphoinositide-3 kinase (PI3K) and phospho-AKT (p-AKT)/AKT ratios was decreased under the influence of Apatinib and a combination of Apatinib/melatonin (P ≤ 0.01). CONCLUSION Apatinib or a combination of Apatinib/melatonin may be used to manage patients with breast cancer. However, further studies are needed to identify anti-cancer mechanisms of melatonin and Apatinib for better management of the patients with breast cancer.
Collapse
Affiliation(s)
- Nazila Fathi Maroufi
- Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohsen Rashidi
- Department of Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.,The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Vahid Vahedian
- Researchers Club of Tums Preclinical Core Facility (TPCF), Tehran University of Medical Science (TUMS), Tehran, Iran
| | - Raheleh Jahanbazi
- Department of Biology, Faculty of Science, Islamic Azad University, Falavarjan branch, Isfahan, Iran
| | | | - Maryam Akbarzadeh
- Department of Biochemistry, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Hamid Kazemzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamid-Reza Nejabati
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Isazadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad-Reza Rashidi
- Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mohammad Nouri
- Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
23
|
Zhang X, Zhang JG, Mu W, Zhou HM, Liu GL, Li Q. The role of daurisoline treatment in hepatocellular carcinoma: Inhibiting vasculogenic mimicry formation and enhancing sensitivity to sorafenib. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 92:153740. [PMID: 34600176 DOI: 10.1016/j.phymed.2021.153740] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 08/27/2021] [Accepted: 09/05/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Vasculogenic mimicry (VM) is a newly described tumor vascular phenomenon that is independent of traditional angiogenesis and provides an adequate blood supply for tumor growth. VM has been consistently observed in different cancer types. Hence, inhibition of VM may be considered a new anticancer therapeutic strategy. PURPOSE This study aimed to elucidate the potential anticancer effect of daurisoline (DS) on hepatocellular carcinoma (HCC) and the potential molecular mechanism by which DS inhibits VM. We also verified whether combination treatment with sorafenib and DS constitutes a novel therapeutic approach to prevent HCC progression. METHODS The effects of DS on proliferation were evaluated by Cell Counting Kit-8 (CCK-8), colony formation, and 5-ethynyl-2'-deoxyuridine (EdU) incorporation assays. 4',6-Diamidino-2-phenylindole (DAPI) staining and flow cytometric analysis were employed to investigate its effects on apoptosis. Western blot analysis, Matrigel tube formation assays, pulldown assays and immunofluorescence staining were applied to validate the potential mechanism by which DS inhibits VM. Mouse xenograft models were used to evaluate anticancer activities. RESULTS DS inhibited HCC cell proliferation, induced HCC cell apoptosis and repressed VM formation by inactivating RhoA/ROCK2-mediated AKT and ERK-p38 MAPK signaling. Additionally, DS dramatically sensitized HCC cell lines to sorafenib, a curative anticancer drug for patients with advanced HCC. CONCLUSIONS Our study provides insights into the molecular mechanisms underlying DS-induced inhibition of VM, which may facilitate the development of a novel clinical anti-HCC drug. Moreover, our findings suggest that the combination of DS and sorafenib constitutes a potential therapeutic strategy for HCC.
Collapse
Affiliation(s)
- Xue Zhang
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100 Haining Road, Shanghai, 200080, China; Department of Pharmacy, Shanghai Eye Diseases Prevention & Treatment Center, National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai General Hospital, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, 200040, China
| | - Ji-Gang Zhang
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100 Haining Road, Shanghai, 200080, China
| | - Wan Mu
- Department of Pharmacy, Shanghai Eye Diseases Prevention & Treatment Center, National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai General Hospital, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, 200040, China
| | - He-Ming Zhou
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100 Haining Road, Shanghai, 200080, China
| | - Gao-Lin Liu
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100 Haining Road, Shanghai, 200080, China; Department of Pharmacy, Shanghai Eye Diseases Prevention & Treatment Center, National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai General Hospital, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, 200040, China.
| | - Qin Li
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100 Haining Road, Shanghai, 200080, China.
| |
Collapse
|
24
|
Shin SU, Cho HM, Das R, Gil-Henn H, Ramakrishnan S, Al Bayati A, Carroll SF, Zhang Y, Sankar AP, Elledge C, Pimentel A, Blonska M, Rosenblatt JD. Inhibition of Vasculogenic Mimicry and Angiogenesis by an Anti-EGFR IgG1-Human Endostatin-P125A Fusion Protein Reduces Triple Negative Breast Cancer Metastases. Cells 2021; 10:cells10112904. [PMID: 34831127 PMCID: PMC8616280 DOI: 10.3390/cells10112904] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 10/08/2021] [Accepted: 10/19/2021] [Indexed: 12/16/2022] Open
Abstract
Triple negative breast cancer (TNBC) is an aggressive breast cancer subtype with limited therapeutic options. Metastasis is the major cause of TNBC mortality. Angiogenesis facilitates TNBC metastases. Many TNBCs also form vascular channels lined by tumor cells rather than endothelial cells, known as ‘vasculogenic mimicry’ (VM). VM has been linked to metastatic TNBC behavior and resistance to anti-angiogenic agents. Epidermal growth factor receptor (EGFR) is frequently expressed on TNBC, but anti-EGFR antibodies have limited efficacy. We synthesized an anti-EGFR antibody–endostatin fusion protein, αEGFR IgG1-huEndo-P125A (αEGFR-E-P125A), designed to deliver a mutant endostatin, huEndo-P125A (E-P125A), to EGFR expressing tumors, and tested its effects on angiogenesis, TNBC VM, and motility in vitro, and on the growth and metastasis of two independent human TNBC xenograft models in vivo. αEGFR-E-P125A completely inhibited the ability of human umbilical vein endothelial cells to form capillary-like structures (CLS) and of TNBC cells to engage in VM and form tubes in vitro. αEGFR-E-P125A treatment reduced endothelial and TNBC motility in vitro more effectively than E-P125A or cetuximab, delivered alone or in combination. Treatment of TNBC with αEGFR-E-P125A was associated with a reduction in cytoplasmic and nuclear β-catenin and reduced phosphorylation of vimentin. αEGFR-E-P125A treatment of TNBC xenografts in vivo inhibited angiogenesis and VM, reduced primary tumor growth and lung metastasis of orthotopically implanted MDA-MB-468 TNBC cells, and markedly decreased lung metastases following intravenous injection of MDA-MB-231-4175 lung-tropic TNBC cells. Combined inhibition of angiogenesis, VM, and TNBC motility mediated by αEGFR-E-P125A is a promising strategy for the prevention of TNBC metastases.
Collapse
Affiliation(s)
- Seung-Uon Shin
- Sylvester Comprehensive Cancer Center, Department of Medicine, Division of Hematology, University of Miami Miller School of Medicine (UMMSOM), Miami, FL 33136, USA; (S.-U.S.); (H.-M.C.); (A.A.B.); (Y.Z.); (M.B.)
| | - Hyun-Mi Cho
- Sylvester Comprehensive Cancer Center, Department of Medicine, Division of Hematology, University of Miami Miller School of Medicine (UMMSOM), Miami, FL 33136, USA; (S.-U.S.); (H.-M.C.); (A.A.B.); (Y.Z.); (M.B.)
| | - Rathin Das
- Synergys Biotherapeutics Inc., Alamo, CA 94507, USA; (R.D.); (S.F.C.)
| | - Hava Gil-Henn
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel;
| | - Sundaram Ramakrishnan
- Department of Surgery, University of Miami Miller School of Medicine (UMMSOM), Miami, FL 33136, USA;
| | - Ahmed Al Bayati
- Sylvester Comprehensive Cancer Center, Department of Medicine, Division of Hematology, University of Miami Miller School of Medicine (UMMSOM), Miami, FL 33136, USA; (S.-U.S.); (H.-M.C.); (A.A.B.); (Y.Z.); (M.B.)
- Kentucky Clinic, University of Kentucky, Lexington, KY 40536, USA
| | | | - Yu Zhang
- Sylvester Comprehensive Cancer Center, Department of Medicine, Division of Hematology, University of Miami Miller School of Medicine (UMMSOM), Miami, FL 33136, USA; (S.-U.S.); (H.-M.C.); (A.A.B.); (Y.Z.); (M.B.)
| | - Ankita P. Sankar
- Sheila and David Fuente Graduate Program in Cancer Biology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine (UMMSOM), Miami, FL 33136, USA; (A.P.S.); (C.E.)
| | - Christian Elledge
- Sheila and David Fuente Graduate Program in Cancer Biology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine (UMMSOM), Miami, FL 33136, USA; (A.P.S.); (C.E.)
| | - Augustin Pimentel
- Sylvester Comprehensive Cancer Center, Department of Medicine, Division of Medical Oncology, University of Miami Miller School of Medicine (UMMSOM), Miami, FL 33136, USA;
| | - Marzenna Blonska
- Sylvester Comprehensive Cancer Center, Department of Medicine, Division of Hematology, University of Miami Miller School of Medicine (UMMSOM), Miami, FL 33136, USA; (S.-U.S.); (H.-M.C.); (A.A.B.); (Y.Z.); (M.B.)
| | - Joseph D. Rosenblatt
- Sylvester Comprehensive Cancer Center, Department of Medicine, Division of Hematology, University of Miami Miller School of Medicine (UMMSOM), Miami, FL 33136, USA; (S.-U.S.); (H.-M.C.); (A.A.B.); (Y.Z.); (M.B.)
- Correspondence: ; Tel.: +1-305-243-4618; Fax: +1-305-243-9161
| |
Collapse
|
25
|
Andreucci E, Laurenzana A, Peppicelli S, Biagioni A, Margheri F, Ruzzolini J, Bianchini F, Fibbi G, Del Rosso M, Nediani C, Serratì S, Fucci L, Guida M, Calorini L. uPAR controls vasculogenic mimicry ability expressed by drug-resistant melanoma cells. Oncol Res 2021; 28:873-884. [PMID: 34315564 PMCID: PMC8790129 DOI: 10.3727/096504021x16273798026651] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Malignant melanoma is a highly aggressive skin cancer characterized by an elevated grade of tumor cell plasticity. Such plasticity allows melanoma cells adaptation to different hostile conditions and guarantees tumor survival and disease progression, including aggressive features such as drug resistance. Indeed, almost 50% of melanoma rapidly develop resistance to the BRAFV600E inhibitor vemurafenib, with fast tumor dissemination, a devastating consequence for patients' outcomes. Vasculogenic mimicry (VM), the ability of cancer cells to organize themselves in perfused vascular-like channels, might sustain tumor spread by providing vemurafenib-resistant cancer cells with supplementary ways to enter into circulation and disseminate. Thus, this research aims to determine if vemurafenib resistance goes with the acquisition of VM ability by aggressive melanoma cells, and identify a driving molecule for both vemurafenib resistance and VM. We used two independent experimental models of drug-resistant melanoma cells, the first one represented by a chronic adaptation of melanoma cells to extracellular acidosis, known to drive a particularly aggressive and vemurafenib-resistant phenotype, the second one generated with chronic vemurafenib exposure. By performing in vitro tube formation assay and evaluating the expression levels of the VM markers EphA2 and VE-cadherin by western blotting and flow cytometer analyses, we demonstrated that vemurafenib-resistant cells obtained by both models are characterized by an increased ability to perform VM. Moreover, by exploiting the CRISPR-Cas9 technique and using the urokinase plasminogen activator receptor (uPAR) inhibitor M25, we identified uPAR as a driver of VM expressed by vemurafenib-resistant melanoma cells. Thus, uPAR targeting may be successfully leveraged as a new complementary therapy to inhibit VM in drug-resistant melanoma patients, to counteract the rapid progression and dissemination of the disease.
Collapse
Affiliation(s)
- Elena Andreucci
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134, Florence, Italy
| | - Anna Laurenzana
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134, Florence, Italy
| | - Silvia Peppicelli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134, Florence, Italy
| | - Alessio Biagioni
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134, Florence, Italy
| | - Francesca Margheri
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134, Florence, Italy
| | - Jessica Ruzzolini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134, Florence, Italy
| | - Francesca Bianchini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134, Florence, Italy
| | - Gabriella Fibbi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134, Florence, Italy
| | - Mario Del Rosso
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134, Florence, Italy.,Center of Excellence for Research, Transfer and High Education DenoTHE University of Florence, 50134, Florence, Italy
| | - Chiara Nediani
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134, Florence, Italy
| | - Simona Serratì
- Laboratory of Nanotecnology, IRCCS Istituto Tumori "Giovanni Paolo II", 70124, Bari, Italy
| | - Livia Fucci
- Pathology Department, IRCCS IstitutoTumori "Giovanni Paolo II", 70124, Bari, Italy
| | - Michele Guida
- Rare tumors and Melnaoma Unit, IRCCS IstitutoTumori "Giovanni Paolo II", 70124, Bari, Italy
| | - Lido Calorini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134, Florence, Italy.,Center of Excellence for Research, Transfer and High Education DenoTHE University of Florence, 50134, Florence, Italy
| |
Collapse
|
26
|
Wang H, Zhang H, Zhu Y, Wu Z, Cui C, Cai F. Anticancer Mechanisms of Salinomycin in Breast Cancer and Its Clinical Applications. Front Oncol 2021; 11:654428. [PMID: 34381705 PMCID: PMC8350729 DOI: 10.3389/fonc.2021.654428] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 06/28/2021] [Indexed: 01/11/2023] Open
Abstract
Breast cancer (BC) is the most frequent cancer among women worldwide and is the leading cause of cancer-related deaths in women. Cancer cells with stem cell-like features and tumor-initiating potential contribute to drug resistance, tumor recurrence, and metastasis. To achieve better clinical outcomes, it is crucial to eradicate both bulk BC cells and breast cancer stem cells (BCSCs). Salinomycin, a monocarboxylic polyether antibiotic isolated from Streptomyces albus, can precisely kill cancer stem cells (CSCs), particularly BCSCs, by various mechanisms, including apoptosis, autophagy, and necrosis. There is increasing evidence that salinomycin can inhibit cell proliferation, invasion, and migration in BC and reverse the immune-inhibitory microenvironment to prevent tumor growth and metastasis. Therefore, salinomycin is a promising therapeutic drug for BC. In this review, we summarize established mechanisms by which salinomycin protects against BC and discuss its future clinical applications.
Collapse
Affiliation(s)
- Hui Wang
- Laboratory of Tumor Molecular Biology, School of Basic Medical Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Hongyi Zhang
- Department of Breast Surgery, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yihao Zhu
- Laboratory of Tumor Molecular Biology, School of Basic Medical Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Zhonghang Wu
- Department of Scientific Research, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China.,Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Chunhong Cui
- Laboratory of Tumor Molecular Biology, School of Basic Medical Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, China.,Department of Scientific Research, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Fengfeng Cai
- Department of Breast Surgery, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
27
|
Morales-Guadarrama G, García-Becerra R, Méndez-Pérez EA, García-Quiroz J, Avila E, Díaz L. Vasculogenic Mimicry in Breast Cancer: Clinical Relevance and Drivers. Cells 2021; 10:cells10071758. [PMID: 34359928 PMCID: PMC8304745 DOI: 10.3390/cells10071758] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/07/2021] [Accepted: 07/08/2021] [Indexed: 12/24/2022] Open
Abstract
In solid tumors, vasculogenic mimicry (VM) is the formation of vascular structures by cancer cells, allowing to generate a channel-network able to transport blood and tumor cells. While angiogenesis is undertaken by endothelial cells, VM is assumed by cancer cells. Besides the participation of VM in tumor neovascularization, the clinical relevance of this process resides in its ability to favor metastasis and to drive resistance to antiangiogenic therapy. VM occurs in many tumor types, including breast cancer, where it has been associated with a more malignant phenotype, such as triple-negative and HER2-positive tumors. The latter may be explained by known drivers of VM, like hypoxia, TGFB, TWIST1, EPHA2, VEGF, matrix metalloproteinases, and other tumor microenvironment-derived factors, which altogether induce the transformation of tumor cells to a mesenchymal phenotype with a high expression rate of stemness markers. This review analyzes the current literature in the field, including the participation of some microRNAs and long noncoding RNAs in VM-regulation and tumorigenesis of breast cancer. Considering the clinical relevance of VM and its association with the tumor phenotype and clinicopathological parameters, further studies are granted to target VM in the clinic.
Collapse
Affiliation(s)
- Gabriela Morales-Guadarrama
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México 14080, Mexico; (G.M.-G.); (E.A.M.-P.); (J.G.-Q.); (E.A.)
| | - Rocío García-Becerra
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico;
| | - Edgar Armando Méndez-Pérez
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México 14080, Mexico; (G.M.-G.); (E.A.M.-P.); (J.G.-Q.); (E.A.)
| | - Janice García-Quiroz
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México 14080, Mexico; (G.M.-G.); (E.A.M.-P.); (J.G.-Q.); (E.A.)
| | - Euclides Avila
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México 14080, Mexico; (G.M.-G.); (E.A.M.-P.); (J.G.-Q.); (E.A.)
| | - Lorenza Díaz
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México 14080, Mexico; (G.M.-G.); (E.A.M.-P.); (J.G.-Q.); (E.A.)
- Correspondence: ; Tel.: +52-(55)-5487-0900
| |
Collapse
|
28
|
Treps L, Faure S, Clere N. Vasculogenic mimicry, a complex and devious process favoring tumorigenesis – Interest in making it a therapeutic target. Pharmacol Ther 2021; 223:107805. [DOI: 10.1016/j.pharmthera.2021.107805] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
29
|
Alternative Vascularization Mechanisms in Tumor Resistance to Therapy. Cancers (Basel) 2021; 13:cancers13081912. [PMID: 33921099 PMCID: PMC8071410 DOI: 10.3390/cancers13081912] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/12/2021] [Accepted: 04/13/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Tumors rely on blood vessels to grow and metastasize. Malignant tumors can employ different strategies to create a functional vascular network. Tumor cells can use normal processes of vessel formation but can also employ cancer-specific mechanisms, by co-opting normal vessels present in tissues or by turning themselves into vascular cells. These different types of tumor vessels have specific molecular and functional characteristics that profoundly affect tumor behavior and response to therapies, including drugs targeting the tumor vasculature (antiangiogenic therapies). In this review, we discuss how vessels formed by different mechanisms affect the intrinsic sensitivity of tumors to therapy and, on the other hand, how therapies can affect tumor vessel formation, leading to resistance to drugs, cancer recurrence, and treatment failure. Potential strategies to avoid vessel-mediated resistance to antineoplastic therapies will be discussed. Abstract Blood vessels in tumors are formed through a variety of different mechanisms, each generating vessels with peculiar structural, molecular, and functional properties. This heterogeneity has a major impact on tumor response or resistance to antineoplastic therapies and is now emerging as a promising target for strategies to prevent drug resistance and improve the distribution and efficacy of antineoplastic treatments. This review presents evidence of how different mechanisms of tumor vessel formation (vasculogenesis, glomeruloid proliferation, intussusceptive angiogenesis, vasculogenic mimicry, and vessel co-option) affect tumor responses to antiangiogenic and antineoplastic therapies, but also how therapies can promote alternative mechanisms of vessel formation, contributing to tumor recurrence, malignant progression, and acquired drug resistance. We discuss the possibility of tailoring treatment strategies to overcome vasculature-mediated drug resistance or to improve drug distribution and efficacy.
Collapse
|
30
|
He X, You J, Ding H, Zhang Z, Cui L, Shen X, Bian X, Liu Y, Chen J. Vasculogenic mimicry, a negative indicator for progression free survival of lung adenocarcinoma irrespective of first line treatment and epithelial growth factor receptor mutation status. BMC Cancer 2021; 21:132. [PMID: 33549061 PMCID: PMC7866877 DOI: 10.1186/s12885-021-07863-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 02/01/2021] [Indexed: 12/19/2022] Open
Abstract
Background Vascular mimicry (VM) was associated with the prognosis of cancers. The aim of the study was to explore the association between VM and anticancer therapy response in patients with lung adenocarcinoma. Methods This was a single-center retrospective study of patients with lung adenocarcinoma between March 1st, 2013, to April 1st, 2019, at the Second People’s Hospital of Taizhou City. All included patients were divided into the VM and no-VM groups according to whether VM was observed or not in the specimen. Vessels with positive PAS and negative CD34 staining were confirmed as VM. The main outcome was progression-free survival (PFS). Results Sixty-six (50.4%) patients were male. Eighty-one patients received chemotherapy as the first-line treatment, and 50 patients received TKIs. Forty-five (34.4%) patients were confirmed with VM. There was no difference regarding the first-line treatment between the VM and no-VM groups (P = 0.285). The 86 patients without VM had a median PFS of 279 (range, 90–1095) days, and 45 patients with VM had a median PFS of 167 (range, 90–369) days (P < 0.001). T stage (hazard ratio (HR) = 1.37, 95% confidence interval (CI): 1.10–1.71), N stage (HR = 1.43, 95%CI: 1.09–1.86), M stage (HR = 2.85, 95%CI: 1.76–4.61), differentiation (HR = 1.85, 95%CI: 1.29–2.65), therapy (HR = 0.32, 95%CI: 0.21–0.49), VM (HR = 2.12, 95%CI: 1.33–3.37), and ECOG (HR = 1.41, 95%CI: 1.09–1.84) were independently associated with PFS. Conclusion The benefits of first-line TKIs for NSCLC with EGFR mutation are possibly better than those of platinum-based regimens in patients without VM, but there is no difference in the benefit of chemotherapy or target therapy for VM-positive NSCLC harboring EGFR mutations.
Collapse
Affiliation(s)
- Xuejun He
- Oncology Department, the Second People's Hospital of Taizhou affiliated to Medical College of Yangzhou University, No. 27, Jiangyan District, Taizhou, 225500, China
| | - Jijun You
- Orthopaedic Department, the Second People's Hospital of Taizhou Affiliated to Medical College of Yangzhou University, Taizhou, China
| | - Haibing Ding
- Orthopaedic Department, the Second People's Hospital of Taizhou Affiliated to Medical College of Yangzhou University, Taizhou, China
| | - Zhisheng Zhang
- Oncology Department, the Second People's Hospital of Taizhou affiliated to Medical College of Yangzhou University, No. 27, Jiangyan District, Taizhou, 225500, China
| | - Lin Cui
- Oncology Department, the Second People's Hospital of Taizhou affiliated to Medical College of Yangzhou University, No. 27, Jiangyan District, Taizhou, 225500, China
| | - Xiaomei Shen
- Oncology Department, the Second People's Hospital of Taizhou affiliated to Medical College of Yangzhou University, No. 27, Jiangyan District, Taizhou, 225500, China
| | - Xiaoxia Bian
- Oncology Department, the Second People's Hospital of Taizhou affiliated to Medical College of Yangzhou University, No. 27, Jiangyan District, Taizhou, 225500, China
| | - Yanqing Liu
- Institute of Medicine, Yangzhou University, No. 88, South Daxue Road, Yangzhou, 225001, China
| | - Jue Chen
- Oncology Department, the Second People's Hospital of Taizhou affiliated to Medical College of Yangzhou University, No. 27, Jiangyan District, Taizhou, 225500, China. .,Institute of Medicine, Yangzhou University, No. 88, South Daxue Road, Yangzhou, 225001, China. .,Respiratory Department, the Second People's Hospital of Taizhou Affiliated to Medical College of Yangzhou University, Taizhou, China.
| |
Collapse
|
31
|
Yu S, Ruan X, Liu X, Zhang F, Wang D, Liu Y, Yang C, Shao L, Liu Q, Zhu L, Lin Y, Xue Y. HNRNPD interacts with ZHX2 regulating the vasculogenic mimicry formation of glioma cells via linc00707/miR-651-3p/SP2 axis. Cell Death Dis 2021; 12:153. [PMID: 33542193 PMCID: PMC7862279 DOI: 10.1038/s41419-021-03432-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 12/15/2020] [Accepted: 12/29/2020] [Indexed: 12/16/2022]
Abstract
Studies have found that RNA-binding proteins (RBPs) are dysfunctional and play a significant regulatory role in the development of glioma. Based on The Cancer Genome Atlas database and the previous studies, we selected heterogeneous nuclear ribonucleoprotein (HNRNPD) as the research candidate and sought its downstream targeted genes. In the present study, HNRNPD, linc00707, and specific protein 2 (SP2) were highly expressed, while zinc fingers and homeboxes 2 (ZHX2) and miR-651-3p were remarkedly downregulated in glioma tissues and cells. HNRNPD, linc00707, and SP2 knockdown or ZHX2 and miR-651-3p overexpression suppressed glioma cells proliferation, migration, and invasion and vasculogenic mimicry (VM) formation. Knockdown of HNRNPD increased the stability of ZHX2 mRNA. ZHX2 bound to the promoter region of linc00707 and negatively regulate its expression. Linc00707 could bind with miR-651-3p, while miR-651-3p bound to the 3' untranslated region (3'UTR) of SP2 mRNA to negatively regulate its expression. The transcription factor SP2 directly bound to the promoter regions of the VM formation-related proteins MMP2, MMP9, and VE-cadherin, playing a role in promoting transcription in order to regulate the VM formation ability of glioma cells.
Collapse
Affiliation(s)
- Sifei Yu
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, People's Republic of China
- Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, 110122, People's Republic of China
- Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, 110122, People's Republic of China
| | - Xuelei Ruan
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, People's Republic of China
- Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, 110122, People's Republic of China
- Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, 110122, People's Republic of China
| | - Xiaobai Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
- Liaoning Research Center for Translational Medicine in Nervous System Disease, Shenyang, 110004, People's Republic of China
- Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, People's Republic of China
| | - Fangfang Zhang
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, People's Republic of China
- Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, 110122, People's Republic of China
- Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, 110122, People's Republic of China
| | - Di Wang
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
- Liaoning Research Center for Translational Medicine in Nervous System Disease, Shenyang, 110004, People's Republic of China
- Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, People's Republic of China
| | - Yunhui Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
- Liaoning Research Center for Translational Medicine in Nervous System Disease, Shenyang, 110004, People's Republic of China
- Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, People's Republic of China
| | - Chunqing Yang
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
- Liaoning Research Center for Translational Medicine in Nervous System Disease, Shenyang, 110004, People's Republic of China
- Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, People's Republic of China
| | - Lianqi Shao
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, People's Republic of China
- Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, 110122, People's Republic of China
- Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, 110122, People's Republic of China
| | - Qianshuo Liu
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, People's Republic of China
- Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, 110122, People's Republic of China
- Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, 110122, People's Republic of China
| | - Lu Zhu
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, People's Republic of China
- Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, 110122, People's Republic of China
- Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, 110122, People's Republic of China
| | - Yang Lin
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, People's Republic of China
- Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, 110122, People's Republic of China
- Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, 110122, People's Republic of China
| | - Yixue Xue
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, People's Republic of China.
- Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, 110122, People's Republic of China.
- Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, 110122, People's Republic of China.
| |
Collapse
|
32
|
Wei X, Chen Y, Jiang X, Peng M, Liu Y, Mo Y, Ren D, Hua Y, Yu B, Zhou Y, Liao Q, Wang H, Xiang B, Zhou M, Li X, Li G, Li Y, Xiong W, Zeng Z. Mechanisms of vasculogenic mimicry in hypoxic tumor microenvironments. Mol Cancer 2021; 20:7. [PMID: 33397409 PMCID: PMC7784348 DOI: 10.1186/s12943-020-01288-1] [Citation(s) in RCA: 260] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 11/24/2020] [Indexed: 02/08/2023] Open
Abstract
Background Vasculogenic mimicry (VM) is a recently discovered angiogenetic process found in many malignant tumors, and is different from the traditional angiogenetic process involving vascular endothelium. It involves the formation of microvascular channels composed of tumor cells; therefore, VM is considered a new model for the formation of new blood vessels in aggressive tumors, and can provide blood supply for tumor growth. Many studies have pointed out that in recent years, some clinical treatments against angiogenesis have not been satisfactory possibly due to the activation of VM. Although the mechanisms underlying VM have not been fully elucidated, increasing research on the soil “microenvironment” for tumor growth suggests that the initial hypoxic environment in solid tumors is inseparable from VM. Main body In this review, we describe that the stemness and differentiation potential of cancer stem cells are enhanced under hypoxic microenvironments, through hypoxia-induced epithelial-endothelial transition (EET) and extracellular matrix (ECM) remodeling to form the specific mechanism of vasculogenic mimicry; we also summarized some of the current drugs targeting VM through these processes, suggesting a new reference for the clinical treatment of tumor angiogenesis. Conclusion Overall, the use of VM inhibitors in combination with conventional anti-angiogenesis treatments is a promising strategy for improving the effectiveness of targeted angiogenesis treatments; further, considering the importance of hypoxia in tumor invasion and metastasis, drugs targeting the hypoxia signaling pathway seem to achieve good results.
Collapse
Affiliation(s)
- Xiaoxu Wei
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital, Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yunhua Chen
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital, Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xianjie Jiang
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Miao Peng
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Yiduo Liu
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Yongzhen Mo
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Daixi Ren
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Yuze Hua
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Boyao Yu
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Yujuan Zhou
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Qianjin Liao
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Hui Wang
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Bo Xiang
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital, Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ming Zhou
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital, Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaoling Li
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital, Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Guiyuan Li
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital, Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yong Li
- Department of Medicine, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital, Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhaoyang Zeng
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital, Xiangya School of Medicine, Central South University, Changsha, Hunan, China. .,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China. .,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
33
|
Involvement of Actin and Actin-Binding Proteins in Carcinogenesis. Cells 2020; 9:cells9102245. [PMID: 33036298 PMCID: PMC7600575 DOI: 10.3390/cells9102245] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 09/18/2020] [Accepted: 10/05/2020] [Indexed: 12/13/2022] Open
Abstract
The actin cytoskeleton plays a crucial role in many cellular processes while its reorganization is important in maintaining cell homeostasis. However, in the case of cancer cells, actin and ABPs (actin-binding proteins) are involved in all stages of carcinogenesis. Literature has reported that ABPs such as SATB1 (special AT-rich binding protein 1), WASP (Wiskott-Aldrich syndrome protein), nesprin, and villin take part in the initial step of carcinogenesis by regulating oncogene expression. Additionally, changes in actin localization promote cell proliferation by inhibiting apoptosis (SATB1). In turn, migration and invasion of cancer cells are based on the formation of actin-rich protrusions (Arp2/3 complex, filamin A, fascin, α-actinin, and cofilin). Importantly, more and more scientists suggest that microfilaments together with the associated proteins mediate tumor vascularization. Hence, the presented article aims to summarize literature reports in the context of the potential role of actin and ABPs in all steps of carcinogenesis.
Collapse
|
34
|
Zhang Y. The root cause of drug resistance in HER2-positive breast cancer and the therapeutic approaches to overcoming the resistance. Pharmacol Ther 2020; 218:107677. [PMID: 32898548 DOI: 10.1016/j.pharmthera.2020.107677] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 08/28/2020] [Indexed: 12/19/2022]
Abstract
HER2 is a well-known oncogenic receptor tyrosine kinase. HER2 gene amplification occurs in about 20% of breast cancer (BC), which leads to overexpression of HER2 protein, known as HER2-positive BC. Inhibitors of HER2 have significantly improved the prognosis of patients with this subset of BC. Since 1998, seven HER2 inhibitors have been developed to treat this disease. However, drug resistance is common and remains a major unresolved clinical problem. Patients typically show disease progression after some time on treatment. This review discusses the complexity and diversified nature of HER2 signaling, the mechanisms of actions and therapeutic activities of all HER2 inhibitors, the roles of HER2 and other signaling proteins in HER2-positive BC resistant to the inhibitors, the non-cell-autonomous mechanisms of drug resistance, and the heterogeneity of tumor HER2 expression. The review presents the concept that drug resistance in HER2-positive BC results primarily from the inability of HER2 inhibitors to deplete HER2. Emerging therapeutics that are promising for overcoming drug resistance are also discussed.
Collapse
Affiliation(s)
- Yuesheng Zhang
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, USA.
| |
Collapse
|
35
|
Wechman SL, Emdad L, Sarkar D, Das SK, Fisher PB. Vascular mimicry: Triggers, molecular interactions and in vivo models. Adv Cancer Res 2020; 148:27-67. [PMID: 32723566 DOI: 10.1016/bs.acr.2020.06.001] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Vascular mimicry is induced by a wide array of genes with functions related to cancer stemness, hypoxia, angiogenesis and autophagy. Vascular mimicry competent (VM-competent) cells that form de novo blood vessels are common in solid tumors facilitating tumor cell survival and metastasis. VM-competent cells display increased levels of vascular mimicry selecting for stem-like cells in an O2-gradient-dependent manner in deeply hypoxic tumor regions, while also aiding in maintaining tumor cell metabolism and stemness. Three of the principal drivers of vascular mimicry are EphA2, Nodal and HIF-1α, however, directly or indirectly many of these molecules affect VE-Cadherin (VE-Cad), which forms gap-junctions to bind angiogenic blood vessels together. During vascular mimicry, the endothelial-like functions of VM-competent cancer stem cells co-opt VE-Cad to bind cancer cells together to create cancer cell-derived blood conducting vessels. This process potentially compensates for the lack of access to blood and nutrient in avascular tumors, simultaneously providing nutrients and enhancing cancer invasion and metastasis. Current evidence also supports that vascular mimicry promotes cancer malignancy and metastasis due to the cooperation of oncogenic signaling molecules driving cancer stemness and autophagy. While a number of currently used cancer therapeutics are effective inhibitors of vascular mimicry, developing a new class of vascular mimicry specific inhibitors could allow for the treatment of angiogenesis-resistant tumors, inhibit cancer metastasis and improve patient survival. In this review, we describe the principal vascular mimicry pathways in addition to emphasizing the roles of hypoxia, autophagy and select proangiogenic oncogenes in this process.
Collapse
Affiliation(s)
- Stephen L Wechman
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Luni Emdad
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Devanand Sarkar
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Swadesh K Das
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Paul B Fisher
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States.
| |
Collapse
|
36
|
Li P, Yang L, Li T, Bin S, Sun B, Huang Y, Yang K, Shan D, Gu H, Li H. The Third Generation Anti-HER2 Chimeric Antigen Receptor Mouse T Cells Alone or Together With Anti-PD1 Antibody Inhibits the Growth of Mouse Breast Tumor Cells Expressing HER2 in vitro and in Immune Competent Mice. Front Oncol 2020; 10:1143. [PMID: 32766150 PMCID: PMC7381237 DOI: 10.3389/fonc.2020.01143] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 06/05/2020] [Indexed: 12/18/2022] Open
Abstract
Chimeric Antigen Receptor (CAR)-T cells have great efficacy against CD19+ leukemia but little success for solid tumors. This study explored the effectiveness of third generation anti-HER2 CAR-T cells alone or in combination with anti-PD1 antibody on breast tumor cells expressing HER2 in vitro and in immune competent mouse model. The PDL1-positive mouse mammary tumor cell line 4T1 engineered to express luciferase and human HER2 was used as the target cell line (4T1-Luc-HER2). Anti-HER2 CAR-T cells were generated by transducing mouse spleen T cells with recombinant lentiviruses. ELISA analysis showed that IL-2 and IFN-γ secretion was increased in CAR-T cells co-cultured with the target cells, and the secretion of these two cytokines was increased further with the addition of anti-PD1 antibody. Lactate dehydrogenase assay revealed that CAR-T cells displayed a potent cytotoxicity against the target cells, and the addition of anti-PD1 antibody further enhanced the cytotoxicity. At the effector: target ratio of 16:1, cytotoxicity was 39.8% with CAR-T cells alone, and increased to 49.5% with the addition of anti-PD1 antibody. In immune competent syngeneic mouse model, CAR-T cells were found to be present in tumor stroma, inhibited tumor growth and increased tumor apoptosis significantly. Addition of anti-PD1 antibody further enhanced these anti-tumor activities. Twenty-one days after treatment, tumor weight was reduced by 50.0% and 73.3% in CAR-T group and CAR-T plus anti-PD1 group compared with blank T group. Our results indicate that anti-PD1 antibody can greatly increase the efficacy of anti-HER2 CAR-T against HER2-positive solid tumors.
Collapse
Affiliation(s)
- Panyuan Li
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Medical University, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Lingcong Yang
- The Third People's Hospital of Dalian, Dalian, China
| | - Tong Li
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Medical University, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Shufang Bin
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Medical University, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Bohao Sun
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Medical University, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yuting Huang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Medical University, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Kaiyan Yang
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Daming Shan
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Haihua Gu
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Medical University, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Hongzhi Li
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Medical University, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
37
|
Vasculogenic mimicry in carcinogenesis and clinical applications. J Hematol Oncol 2020; 13:19. [PMID: 32169087 PMCID: PMC7071697 DOI: 10.1186/s13045-020-00858-6] [Citation(s) in RCA: 191] [Impact Index Per Article: 38.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 03/05/2020] [Indexed: 12/12/2022] Open
Abstract
Distinct from classical tumor angiogenesis, vasculogenic mimicry (VM) provides a blood supply for tumor cells independent of endothelial cells. VM has two distinct types, namely tubular type and patterned matrix type. VM is associated with high tumor grade, tumor progression, invasion, metastasis, and poor prognosis in patients with malignant tumors. Herein, we discuss the recent studies on the role of VM in tumor progression and the diverse mechanisms and signaling pathways that regulate VM in tumors. Furthermore, we also summarize the latest findings of non-coding RNAs, such as lncRNAs and miRNAs in VM formation. In addition, we review application of molecular imaging technologies in detection of VM in malignant tumors. Increasing evidence suggests that VM is significantly associated with poor overall survival in patients with malignant tumors and could be a potential therapeutic target.
Collapse
|
38
|
Prado MCM, Macedo SDAL, Guiraldelli GG, de Faria Lainetti P, Leis-Filho AF, Kobayashi PE, Laufer-Amorim R, Fonseca-Alves CE. Investigation of the Prognostic Significance of Vasculogenic Mimicry and Its Inhibition by Sorafenib in Canine Mammary Gland Tumors. Front Oncol 2019; 9:1445. [PMID: 31921690 PMCID: PMC6930929 DOI: 10.3389/fonc.2019.01445] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 12/03/2019] [Indexed: 12/27/2022] Open
Abstract
Canine mammary gland tumor (CMT) is one of the most important tumors in intact female dogs, and due its similarity to human breast cancer (BC), it is considered a model in comparative oncology. A subset of mammary gland tumors can show aggressive behavior, and a recurrent histological finding is the presence of vasculogenic mimicry (VM). VM is a process in which highly aggressive cancer cells fuse, forming fluid-conducting channels without endothelial cells. Although, VM has been described in canine inflammatory carcinoma, no previous studies have investigated the prognostic and predictive significance of VM in CMT. Thus, this research aimed to investigate the prognostic significance of VM in vivo and the capacity of sorafenib to inhibit VM in vitro. VM was identified in situ in formalin-fixed paraffin-embedded CMT samples (n = 248) using CD31/PAS double staining. VM was identified in 33% of tumors (82/248). The presence of VM was more strongly related to tumor grade than to histological subtype. Patients with positive VM experienced shorter survival times than dogs without VM (P < 0.0001). Due to the importance of the VEGF-A/VEGFR-2 autocrine feed-forward loop in epithelial tumors, we investigated the association between VEGF-A and VEGFR-2 expression by neoplastic tumor cells and the associations of VEGF-A or VEGFR-2 expression with VM. Among the VM-positive samples, all (n = 82) showed high scores (3 or 4) for VEGF-A and VEGFR-2, indicating that VM was a common finding in tumors overexpressing VEGF-A and VEGFR-2. Thus, we cultured two CMT primary cell lines with VM abilities (CM9 and CM60) in vitro and evaluated the anti-tumoral effect of sorafenib. The CM9 cell line showed a half maximal inhibitory concentration (IC50) of 2.61 μM, and the CM60 cell line showed an IC50 of 1.34 μM. We performed a VM assay in vitro and treated each cell line with an IC50 dose of sorafenib, which was able to inhibit VM in vitro. Overall, our results indicated that VM was a prognostic factor for dogs bearing CMT and that sorafenib had an inhibitory effect on VM in CMT cancer cells in vitro.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Renee Laufer-Amorim
- Department of Veterinary Clinic, São Paulo State University—UNESP, Botucatu, Brazil
| | - Carlos Eduardo Fonseca-Alves
- Department of Veterinary Surgery and Anesthesiology, São Paulo State University—UNESP, Botucatu, Brazil
- Institute of Health Sciences, Universidade Paulista—UNIP, Bauru, Brazil
| |
Collapse
|