1
|
Erin N, Tavşan E, Haksever S, Yerlikaya A, Riganti C. Targeting BMP-1 enhances anti-tumoral effects of doxorubicin in metastatic mammary cancer: common and distinct features of TGF-β inhibition. Breast Cancer Res Treat 2025; 210:563-574. [PMID: 39792296 PMCID: PMC11953206 DOI: 10.1007/s10549-024-07592-4] [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: 10/20/2024] [Accepted: 12/17/2024] [Indexed: 01/12/2025]
Abstract
PURPOSE Mammary carcinoma is comprised heterogeneous groups of cells with different metastatic potential. 4T1 mammary carcinoma cells metastasized to heart (4THM), liver (4TLM) and brain (4TBM) and demonstrate cancer-stem cell phenotype. Using these cancer cells we found thatTGF-β is the top upstream regulator of metastatic process. In addition, secretion of bone morphogenetic protein 1 (BMP-1), which is crucial for the proteolytic release of TGF-β, was markedly high in metastatic mammary cancer cells compared to non-metastatic cells. Although TGF-β inhibitors are in clinical trials, systemic inhibition of TGF-β may produce heavy side effects. We here hypothesize that inhibition of BMP-1 proteolytic activity inhibits TGF-β activity and induces anti-tumoral effects. METHOD AND RESULTS Effects of specific BMP-1 inhibitor on liver and brain metastatic murine mammary cancer cells (4TLM and 4TBM), as well as on human mammary cancer MDA-MB-231 and MCF-7 cells, were examined and compared with the results of TGF-β inhibition. Inhibition of BMP-1 activity markedly suppressed proliferation of cancer cells and enhanced anti-tumoral effects of doxorubicin. Inhibition of BMP-1 activity but not of TGF-β activity decreased colony and spheroid formation. Differential effects of BMP-1 and TGF-β inhibitors on TGF-β secretion was also observed. CONCLUSIONS These results demonstrated for the first time that the inhibition of BMP-1 activity has therapeutic potential for treatment of metastatic mammary cancer and enhances the anti-tumoral effects of doxorubicin.
Collapse
Affiliation(s)
- Nuray Erin
- Department of Medical Pharmacology, Faculty of Medicine, Akdeniz University, Antalya, Turkey.
| | - Esra Tavşan
- Department of Medical Pharmacology, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Seren Haksever
- Department of Medical Pharmacology, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Azmi Yerlikaya
- Department of Medical Biology, Faculty of Medicine, Kutahya Health Sciences University, Kutahya, Turkey
| | - Chiara Riganti
- Department of Oncology, University of Torino, Via Nizza 44, 10126, Turin, Italy
- Molecular Biotechnology Center "Guido Tarone", Via Nizza 44, 10126, Turin, Italy
- Interdepartmental Center "G.Scansetti" for the Study of Asbestos and Other Toxic Particulates, University of Torino, 10126, Turin, Italy
| |
Collapse
|
2
|
Jan A, Sofi S, Jan N, Mir MA. An update on cancer stem cell survival pathways involved in chemoresistance in triple-negative breast cancer. Future Oncol 2025; 21:715-735. [PMID: 39936282 PMCID: PMC11881842 DOI: 10.1080/14796694.2025.2461443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 01/29/2025] [Indexed: 02/13/2025] Open
Abstract
Triple-negative breast cancer (TNBC) presents a formidable global health challenge, marked by its aggressive behavior and significant treatment resistance. This subtype, devoid of estrogen, progesterone, and HER2 receptors, largely relies on breast cancer stem cells (BCSCs) for its progression, metastasis, and recurrence. BCSCs, characterized by their self-renewal capacity and resistance to conventional therapies, exploit key surface markers and critical signaling pathways like Wnt, Hedgehog, Notch, TGF-β, PI3K/AKT/mTOR and Hippo-YAP/TAZ to thrive. Their adaptability is underscored by mechanisms including drug efflux and enhanced DNA repair, contributing to poor prognosis and high recurrence rates. The tumor microenvironment (TME) further facilitates BCSC survival through complex interactions with stromal and immune cells. Emerging therapeutic strategies targeting BCSCs - ranging from immunotherapy and nanoparticle-based drug delivery systems to gene-editing technologies - aim to disrupt these resistant cells. Additionally, innovative approaches focusing on exosome-mediated signaling and metabolic reprogramming show promise in overcoming chemoresistance. By elucidating the distinct characteristics of BCSCs and their role in TNBC, researchers are paving the way for novel treatments that may effectively eradicate these resilient cells, mitigate metastasis, and ultimately improve patient outcomes. This review highlights the urgent need for targeted strategies that address the unique biology of BCSCs in the pursuit of more effective therapeutic interventions for TNBC.
Collapse
Affiliation(s)
- Asma Jan
- Cancer Biology Laboratory, Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar, India
| | - Shazia Sofi
- Cancer Biology Laboratory, Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar, India
| | - Nusrat Jan
- Cancer Biology Laboratory, Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar, India
| | - Manzoor Ahmad Mir
- Cancer Biology Laboratory, Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar, India
| |
Collapse
|
3
|
Djamgoz MBA. Stemness of Cancer: A Study of Triple-negative Breast Cancer From a Neuroscience Perspective. Stem Cell Rev Rep 2025; 21:337-350. [PMID: 39531198 PMCID: PMC11872763 DOI: 10.1007/s12015-024-10809-0] [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] [Accepted: 10/21/2024] [Indexed: 11/16/2024]
Abstract
Stemness, giving cancer cells massive plasticity enabling them to survive in dynamic (e.g. hypoxic) environments and become resistant to treatment, especially chemotherapy, is an important property of aggressive tumours. Here, we review some essentials of cancer stemness focusing on triple-negative breast cancer (TNBC), the most aggressive form of all breast cancers. TNBC cells express a range of genes and mechanisms associated with stemness, including the fundamental four "Yamanaka factors". Most of the evidence concerns the transcription factor / oncogene c-Myc and an interesting case is the expression of the neonatal splice variant of voltage-gated sodium channel subtype Nav1.5. On the whole, measures that reduce the stemness make cancer cells less aggressive, reducing their invasive/metastatic potential and increasing/restoring their chemosensitivity. Such measures include gene silencing techniques, epigenetic therapies as well as novel approaches like optogenetics aiming to modulate the plasma membrane voltage. Indeed, simply hyperpolarizing their membrane potential can make stem cells differentiate. Finally, we give an overview of the clinical aspects and exploitation of cancer/TNBC stemness, including diagnostics and therapeutics. In particular, personalised mRNA-based therapies and mechanistically meaningful combinations are promising and the emerging discipline of 'cancer neuroscience' is providing novel insights to both fundamental issues and clinical applications.
Collapse
Affiliation(s)
- Mustafa B A Djamgoz
- Department of Life Sciences, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK.
| |
Collapse
|
4
|
Lv F, Si W, Xu X, He X, Wang Y, Li Y, Li F. RUNX2 prompts triple negative breast cancer drug resistance through TGF-β pathway regulating breast cancer stem cells. Neoplasia 2024; 48:100967. [PMID: 38219710 PMCID: PMC10826822 DOI: 10.1016/j.neo.2024.100967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 12/22/2023] [Accepted: 01/02/2024] [Indexed: 01/16/2024]
Abstract
Triple-negative breast cancer (TNBC) stands out as the most aggressive subtype within the spectrum of breast cancer. The current clinical guidelines propose treatment strategies involving cytotoxic agents like epirubicin or paclitaxel. However, the emergence of acquired resistance frequently precipitates secondary tumor recurrence or the spread of metastasis. In recent times, significant attention has been directed toward the transcription factor RUNX2, due to its pivotal role in both tumorigenesis and the progression of cancer. Previous researches suggest that RUNX2 might be intricately linked to the development of resistance against chemotherapy, with its mechanism of action possibly intertwined with the signaling of TGF-β. Nevertheless, the precise interplay between their effects and the exact molecular mechanisms underpinning chemoresistance in TNBC remain elusive. Therefore, we have taken a multifaceted approach from in vitro and in vivo experiments to validate the relationship between RUNX2 and TGF-β and to search for their pathogenic mechanisms in chemoresistance. In conclusion, we found that RUNX2 affects chemoresistance by regulating cancer cell stemness through direct binding to TGF-β, and that TGF-β dually regulates RUNX2 expression. The important finding will provide a new reference for clinical reversal of the development of chemoresistance in breast cancer.
Collapse
Affiliation(s)
- Fengxu Lv
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, PR China
| | - Wentao Si
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, PR China
| | - Xiaodan Xu
- Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, PR China
| | - Xiaogang He
- Medical Faculty Mannheim, University of Heidelberg, Mannheim, 68169, Germany
| | - Ying Wang
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, PR China
| | - Yetian Li
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, PR China.
| | - Feifei Li
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, PR China..
| |
Collapse
|
5
|
Guo Z, Han S. Targeting cancer stem cell plasticity in triple-negative breast cancer. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2023; 4:1165-1181. [PMID: 38213533 PMCID: PMC10776602 DOI: 10.37349/etat.2023.00190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 10/15/2023] [Indexed: 01/13/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive breast cancer subtype with limited treatment options. Cancer stem cells (CSCs) are thought to play a crucial role in TNBC progression and resistance to therapy. CSCs are a small subpopulation of cells within tumors that possess self-renewal and differentiation capabilities and are responsible for tumor initiation, maintenance, and metastasis. CSCs exhibit plasticity, allowing them to switch between states and adapt to changing microenvironments. Targeting CSC plasticity has emerged as a promising strategy for TNBC treatment. This review summarizes recent advances in understanding the molecular mechanisms underlying CSC plasticity in TNBC and discusses potential therapeutic approaches targeting CSC plasticity.
Collapse
Affiliation(s)
- Zhengwang Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Integration of Chinese and Western Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Shuyan Han
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Integration of Chinese and Western Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| |
Collapse
|
6
|
Guo Q, Yu Y, Tang W, Zhou S, Lv X. Matrine exerts an anti-tumor effect via regulating HN1 in triple breast cancer both in vitro and in vivo. Chem Biol Drug Des 2023; 102:1469-1477. [PMID: 37674344 DOI: 10.1111/cbdd.14338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/25/2023] [Accepted: 08/18/2023] [Indexed: 09/08/2023]
Abstract
The treatment of triple-negative breast cancer (TNBC) cannot meet medical needs, and it is urgent to find new drugs for intervention. This study aimed to investigate the anti-tumor effect of matrine on the proliferation and apoptosis of TNBC cells based on HN1 regulation in vitro and in vivo. TNBC cell lines (MDA-MB-453 and HCC-1806) were treated with varying concentrations of matrine (0, 1.0, 2.0, 3.0, 4.0, and 5.0 mM). CCK-8, colony formation assay, transwell assay, and flow cytometry assay were employed to detect proliferation, clone formation, invasion, and apoptosis of TNBC cells. Western blot analysis was applied to detect the protein expression of apoptosis HN1. The effects of matrine on tumor growth, protein expression of HN1, and apoptosis in vivo were validated by xenograft tumor models and histology. It was found that matrine inhibited proliferation, colony formation, and invasion and promoted apoptosis of TNBC cells in vitro. HN1 expression was suppressed by matrine. HN1 overexpression perceptibly reversed the above-mentioned additive effect in vitro. In vivo experiments found that matrine inhibited tumor growth and the expression of HN1 protein but promoted the protein expression of Cleared-Caspase-3. Above all, this study demonstrated that matrine inhibited proliferation and promoted apoptosis of TNBC cells via suppressing HN1 expression. Targeting HN1 by matrine may provide new insights into the therapeutic management of patients with TNBC.
Collapse
Affiliation(s)
- Qiusheng Guo
- Department of Medical Oncology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Yuan Yu
- Zhejiang Cancer Research Institute, Cancer Hospital of The University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
| | - Wanfen Tang
- Department of Medical Oncology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Shishi Zhou
- Department of Medical Oncology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Xianmei Lv
- Department of Radiotherapy, Jinhua People's Hospital, Jinhua, China
| |
Collapse
|
7
|
Li YR, Fang Y, Lyu Z, Zhu Y, Yang L. Exploring the dynamic interplay between cancer stem cells and the tumor microenvironment: implications for novel therapeutic strategies. J Transl Med 2023; 21:686. [PMID: 37784157 PMCID: PMC10546755 DOI: 10.1186/s12967-023-04575-9] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 09/28/2023] [Indexed: 10/04/2023] Open
Abstract
Cancer stem cells (CSCs) have emerged as key contributors to tumor initiation, growth, and metastasis. In addition, CSCs play a significant role in inducing immune evasion, thereby compromising the effectiveness of cancer treatments. The reciprocal communication between CSCs and the tumor microenvironment (TME) is observed, with the TME providing a supportive niche for CSC survival and self-renewal, while CSCs, in turn, influence the polarization and persistence of the TME, promoting an immunosuppressive state. Consequently, these interactions hinder the efficacy of current cancer therapies, necessitating the exploration of novel therapeutic approaches to modulate the TME and target CSCs. In this review, we highlight the intricate strategies employed by CSCs to evade immune surveillance and develop resistance to therapies. Furthermore, we examine the dynamic interplay between CSCs and the TME, shedding light on how this interaction impacts cancer progression. Moreover, we provide an overview of advanced therapeutic strategies that specifically target CSCs and the TME, which hold promise for future clinical and translational studies in cancer treatment.
Collapse
Affiliation(s)
- Yan-Ruide Li
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| | - Ying Fang
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Zibai Lyu
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Yichen Zhu
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Lili Yang
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
8
|
Zhang B, Zhao R, Wang Q, Zhang YJ, Yang L, Yuan ZJ, Yang J, Wang QJ, Yao L. An EMT-Related Gene Signature to Predict the Prognosis of Triple-Negative Breast Cancer. Adv Ther 2023; 40:4339-4357. [PMID: 37462865 PMCID: PMC10499992 DOI: 10.1007/s12325-023-02577-z] [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: 04/11/2023] [Accepted: 06/05/2023] [Indexed: 09/14/2023]
Abstract
INTRODUCTION Epithelial-mesenchymal transition (EMT) is an important biological process in tumor invasion and metastasis, and thus a potential indicator of the progression and drug resistance of breast cancer. This study comprehensively analyzed EMT-related genes in triple-negative breast cancer (TNBC) to develop an EMT-related prognostic gene signature. METHODS With the application of The Cancer Genome Atlas (TCGA) database, Molecular Taxonomy of Breast Cancer International Consortium (METABRIC), and the Genotype-Tissue Expression (GTEx) database, we identified EMT-related signature genes (EMGs) by Cox univariate regression and LASSO regression analysis. Risk scores were calculated and used to divide patients with TNBC into high-risk group and low-risk groups by the median value. Kaplan-Meier (K-M) and receiver operating characteristic (ROC) curve analyses were applied for model validation. Independent prognostic predictors were used to develop nomograms. Then, we assessed the risk model in terms of the immune microenvironment, genetic alteration and DNA methylation effects on prognosis, the probability of response to immunotherapy and chemotherapy, and small molecule drugs predicted by The Connectivity Map (Cmap) database. RESULTS Thirteen EMT-related genes with independent prognostic value were identified and used to stratify the patients with TNBC into high- and low-risk groups. The survival analysis revealed that patients in the high-risk group had significantly poorer overall survival than patients in the low-risk group. Populations of immune cells, including CD4 memory resting T cells, CD4 memory activated T cells, and activated dendritic cells, significantly differed between the high- and low-risk groups. Moreover, some therapeutic drugs to which the high-risk group might show sensitivity were identified. CONCLUSIONS Our research identified the significant impact of EMGs on prognosis in TNBC, providing new strategies for personalizing TNBC treatment and improving clinical outcomes.
Collapse
Affiliation(s)
- Bo Zhang
- Department of Breast Oncology, Shanxi Provincial Cancer Hospital, Taiyuan, China
| | - Rong Zhao
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Qi Wang
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China
| | - Ya-Jing Zhang
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Shanxi Medical University, Taiyuan, China
| | - Liu Yang
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Shanxi Medical University, Taiyuan, China
| | - Zhou-Jun Yuan
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Shanxi Medical University, Taiyuan, China
| | - Jun Yang
- Department of Breast Oncology, Shanxi Provincial Cancer Hospital, Taiyuan, China
| | - Qian-Jun Wang
- Department of Breast Oncology, Shanxi Provincial Cancer Hospital, Taiyuan, China
| | - Liang Yao
- Department of Breast Oncology, Shanxi Provincial Cancer Hospital, Taiyuan, China.
| |
Collapse
|
9
|
Yan C, Ma Y, Li J, Chen X, Ma J. Identification of key immune cell-related genes involved in tumorigenesis and prognosis of cervical squamous cell carcinoma. Hum Vaccin Immunother 2023; 19:2254239. [PMID: 37799074 PMCID: PMC10561582 DOI: 10.1080/21645515.2023.2254239] [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: 02/19/2023] [Accepted: 08/29/2023] [Indexed: 10/07/2023] Open
Abstract
The infiltration of immune cells can significantly affect the prognosis and immune therapy of patients with cervical squamous cell carcinoma (CSCC). This study aimed to explore key immune cell-related genes in the tumorigenesis and prognosis of CSCC. The module significantly related to immunity was screened by weighted gene co-expression network analysis (WGCNA) and ESTIMATE analysis, followed by correlation analysis with clinical traits. Key candidate genes were intersected with the protein-protein interaction (PPI) network genes for immune-related genes. The relationship between immune cell infiltration and key genes was analyzed. Tumor immune dysfunction and exclusion (TIDE) and immunophenoscore (IPS) predicted the response to immunotherapy in CSCC patients. Clinically, quantitative real-time polymerase chain reaction (qRT-PCR) and immunohistochemistry were manipulated for analyzing the changes in mRNA and protein expression of key genes in cancer. Western blot was conducted to assess the correlation between key genes and immune infiltration. The brown module was notably associated with the immune microenvironment of CSCC, from which three immune-related key genes (TYROBP, CCL5, and HLA-DRA) were obtained. High expression of these genes was significantly positively associated with the infiltration abundance of T cells, B cells, and other immune cells. High expression levels of three key genes were confirmed in para-cancer tissue and correlated with the abundance of immune cells. The high-expression group of key genes was more sensitive to immunotherapy. We provide a theoretical basis for searching for potential targets for effective treatment and diagnosis of CSCC and provide new ideas for developing novel immunotherapy strategies.
Collapse
Affiliation(s)
- Chunxiao Yan
- School of Medicine, Department of Gynecology, The Second Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Yanyan Ma
- School of Medicine, Department of Gynecology, The Second Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Junyan Li
- School of Medicine, Department of Gynecology, The Second Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Xuejun Chen
- School of Medicine, Department of Gynecology, The Second Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Jiong Ma
- School of Medicine, Department of Gynecology, The Second Affiliated Hospital of Zhejiang University, Hangzhou, China
| |
Collapse
|
10
|
Ordaz-Ramos A, Tellez-Jimenez O, Vazquez-Santillan K. Signaling pathways governing the maintenance of breast cancer stem cells and their therapeutic implications. Front Cell Dev Biol 2023; 11:1221175. [PMID: 37492224 PMCID: PMC10363614 DOI: 10.3389/fcell.2023.1221175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 06/28/2023] [Indexed: 07/27/2023] Open
Abstract
Breast cancer stem cells (BCSCs) represent a distinct subpopulation of cells with the ability to self-renewal and differentiate into phenotypically diverse tumor cells. The involvement of CSC in treatment resistance and cancer recurrence has been well established. Numerous studies have provided compelling evidence that the self-renewal ability of cancer stem cells is tightly regulated by specific signaling pathways, which exert critical roles to maintain an undifferentiated phenotype and prevent the differentiation of CSCs. Signaling pathways such as Wnt/β-catenin, NF-κB, Notch, Hedgehog, TGF-β, and Hippo have been implicated in the promotion of self-renewal of many normal and cancer stem cells. Given the pivotal role of BCSCs in driving breast cancer aggressiveness, targeting self-renewal signaling pathways holds promise as a viable therapeutic strategy for combating this disease. In this review, we will discuss the main signaling pathways involved in the maintenance of the self-renewal ability of BCSC, while also highlighting current strategies employed to disrupt the signaling molecules associated with stemness.
Collapse
Affiliation(s)
- Alejandro Ordaz-Ramos
- Innovation in Precision Medicine Laboratory, Instituto Nacional de Medicina Genómica, Mexico City, México
- Posgrado en Ciencias Biológicas, Unidad de Posgrado, Circuito de Posgrados, Ciudad Universitaria, Coyoacán, México
| | - Olivia Tellez-Jimenez
- Innovation in Precision Medicine Laboratory, Instituto Nacional de Medicina Genómica, Mexico City, México
- Posgrado en Ciencias Biológicas, Unidad de Posgrado, Circuito de Posgrados, Ciudad Universitaria, Coyoacán, México
| | - Karla Vazquez-Santillan
- Innovation in Precision Medicine Laboratory, Instituto Nacional de Medicina Genómica, Mexico City, México
| |
Collapse
|
11
|
Xue W, Hao J, Zhang Q, Jin R, Luo Z, Yang X, Liu Y, Lu Q, Ouyang Y, Guo H. Chlorogenic Acid Inhibits Epithelial-Mesenchymal Transition and Invasion of Breast Cancer by Down-Regulating LRP6. J Pharmacol Exp Ther 2023; 384:254-264. [PMID: 36456194 DOI: 10.1124/jpet.122.001189] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 09/30/2022] [Accepted: 10/17/2022] [Indexed: 12/05/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a crucial biologic process for breast cancer metastasis, and inhibition of EMT could be an effective approach to suppress metastatic potential of mammary cancer. High expression of low-density lipoprotein receptor-related protein 6 (LRP6) is usually observed in breast carcinoma and predicts poor prognosis. In the present study, we investigated whether chlorogenic acid (CA) can inhibit the EMT of breast cancer cells and underlying molecular mechanism. We found that CA treatment transformed MCF-7 cell morphology from spindle shape (mesenchymal phenotype) to spherical shape (epithelial phenotype). CA clearly increased epithelial biomarkers' expression (E-cadherin and ZO-1) but decreased mesenchymal proteins' expression (ZEB1, N-cadherin, vimentin, snail, and slug). In addition, CA attenuated MMP-2 and MMP-9 activities and inhibited cell migration and invasion. CA downregulated the expression of LRP6 in MCF-7 cells. Knockdown LRP6 with siRNA repressed cell mobility and invasion, wheras overexpression of LRP6 promoted EMT and antagonized the EMT inhibitory effect of CA on MCF-7 cells. Furthermore, CA directly interacted with Wnt/β-catenin signaling coreceptor LRP6 and reduced LRP6, p-LRP6, and β-catenin expression levels in MCF-7 cells. In vivo study revealed that CA notably reduced tumor volume and tumor weight. CA decreased the expression of LRP6, N-cadherin, ZEB1, vimentin, MMP2, MMP9, and increased the expression of E-cadherin and ZO-1. In conclusion, CA inhibited EMT and invasion of breast cancer by targeting LRP6. SIGNIFICANCE STATEMENT: CA, the familiar polyphenol compound in traditional Chinese medicine, repressed EMT and weakened cellular mobility and invasion in MCF-7 cells. The mechanism studies demonstrated that CA could inhibit EMT and invasion of MCF-7 cells via targeting LRP6. Additionally, CA restrained tumor growth and xenograft tumor EMT in vivo. The EMT inhibitory property of CA warrants further studies of CA as a drug candidate for the therapy of metastatic breast carcinoma.
Collapse
Affiliation(s)
- Wei Xue
- Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation & College of Pharmacy (W.X., J.H., Q.Z., R.J., Z.L., Y.L., Q.L., H.G.), Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education & Center for Translational Medicine (W.X, J.H., Q.Z., X.Y., H.G.), and Laboratory Animal Center (Y.O.), Guangxi Medical University, Nanning, China; Department of Pharmacy, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China (W.X.); and The First Affiliated Hospital of Guangxi Medical University, Nanning, China (Q.Z.)
| | - Jie Hao
- Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation & College of Pharmacy (W.X., J.H., Q.Z., R.J., Z.L., Y.L., Q.L., H.G.), Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education & Center for Translational Medicine (W.X, J.H., Q.Z., X.Y., H.G.), and Laboratory Animal Center (Y.O.), Guangxi Medical University, Nanning, China; Department of Pharmacy, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China (W.X.); and The First Affiliated Hospital of Guangxi Medical University, Nanning, China (Q.Z.)
| | - Qiuping Zhang
- Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation & College of Pharmacy (W.X., J.H., Q.Z., R.J., Z.L., Y.L., Q.L., H.G.), Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education & Center for Translational Medicine (W.X, J.H., Q.Z., X.Y., H.G.), and Laboratory Animal Center (Y.O.), Guangxi Medical University, Nanning, China; Department of Pharmacy, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China (W.X.); and The First Affiliated Hospital of Guangxi Medical University, Nanning, China (Q.Z.)
| | - Ronghua Jin
- Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation & College of Pharmacy (W.X., J.H., Q.Z., R.J., Z.L., Y.L., Q.L., H.G.), Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education & Center for Translational Medicine (W.X, J.H., Q.Z., X.Y., H.G.), and Laboratory Animal Center (Y.O.), Guangxi Medical University, Nanning, China; Department of Pharmacy, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China (W.X.); and The First Affiliated Hospital of Guangxi Medical University, Nanning, China (Q.Z.)
| | - Zhuo Luo
- Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation & College of Pharmacy (W.X., J.H., Q.Z., R.J., Z.L., Y.L., Q.L., H.G.), Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education & Center for Translational Medicine (W.X, J.H., Q.Z., X.Y., H.G.), and Laboratory Animal Center (Y.O.), Guangxi Medical University, Nanning, China; Department of Pharmacy, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China (W.X.); and The First Affiliated Hospital of Guangxi Medical University, Nanning, China (Q.Z.)
| | - Xin Yang
- Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation & College of Pharmacy (W.X., J.H., Q.Z., R.J., Z.L., Y.L., Q.L., H.G.), Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education & Center for Translational Medicine (W.X, J.H., Q.Z., X.Y., H.G.), and Laboratory Animal Center (Y.O.), Guangxi Medical University, Nanning, China; Department of Pharmacy, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China (W.X.); and The First Affiliated Hospital of Guangxi Medical University, Nanning, China (Q.Z.)
| | - Yanying Liu
- Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation & College of Pharmacy (W.X., J.H., Q.Z., R.J., Z.L., Y.L., Q.L., H.G.), Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education & Center for Translational Medicine (W.X, J.H., Q.Z., X.Y., H.G.), and Laboratory Animal Center (Y.O.), Guangxi Medical University, Nanning, China; Department of Pharmacy, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China (W.X.); and The First Affiliated Hospital of Guangxi Medical University, Nanning, China (Q.Z.)
| | - Qinpei Lu
- Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation & College of Pharmacy (W.X., J.H., Q.Z., R.J., Z.L., Y.L., Q.L., H.G.), Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education & Center for Translational Medicine (W.X, J.H., Q.Z., X.Y., H.G.), and Laboratory Animal Center (Y.O.), Guangxi Medical University, Nanning, China; Department of Pharmacy, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China (W.X.); and The First Affiliated Hospital of Guangxi Medical University, Nanning, China (Q.Z.)
| | - Yiqiang Ouyang
- Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation & College of Pharmacy (W.X., J.H., Q.Z., R.J., Z.L., Y.L., Q.L., H.G.), Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education & Center for Translational Medicine (W.X, J.H., Q.Z., X.Y., H.G.), and Laboratory Animal Center (Y.O.), Guangxi Medical University, Nanning, China; Department of Pharmacy, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China (W.X.); and The First Affiliated Hospital of Guangxi Medical University, Nanning, China (Q.Z.)
| | - Hongwei Guo
- Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation & College of Pharmacy (W.X., J.H., Q.Z., R.J., Z.L., Y.L., Q.L., H.G.), Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education & Center for Translational Medicine (W.X, J.H., Q.Z., X.Y., H.G.), and Laboratory Animal Center (Y.O.), Guangxi Medical University, Nanning, China; Department of Pharmacy, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China (W.X.); and The First Affiliated Hospital of Guangxi Medical University, Nanning, China (Q.Z.)
| |
Collapse
|
12
|
Mahmoud R, Ordóñez-Morán P, Allegrucci C. Challenges for Triple Negative Breast Cancer Treatment: Defeating Heterogeneity and Cancer Stemness. Cancers (Basel) 2022; 14:cancers14174280. [PMID: 36077812 PMCID: PMC9454775 DOI: 10.3390/cancers14174280] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/12/2022] [Accepted: 08/26/2022] [Indexed: 11/25/2022] Open
Abstract
The Triple Negative Breast Cancer (TNBC) subtype is known to have a more aggressive clinical course compared to other breast cancer subtypes. Targeted therapies for this type of breast cancer are limited and patients are mostly treated with conventional chemo- and radio-therapies which are not specific and do not target resistant cells. Therefore, one of the major clinical challenges is to find compounds that target the drug-resistant cell populations which are responsible for reforming secondary tumours. The molecular profiling of the different TNBC subtypes holds a promise for better defining these resistant cells specific to each tumour. To this end, a better understanding of TNBC heterogeneity and cancer stemness is required, and extensive genomic analysis can help to understand the disease complexity and distinguish new molecular drivers that can be targeted in the clinics. The use of persister cancer cell-targeting therapies combined with other therapies may provide a big advance to improve TNBC patients' survival.
Collapse
Affiliation(s)
- Rinad Mahmoud
- Centre for Cancer Sciences, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
- Translational Medical Sciences Unit, School of Medicine, University of Nottingham, Nottingham NG7 2RD, UK
| | - Paloma Ordóñez-Morán
- Centre for Cancer Sciences, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
- Translational Medical Sciences Unit, School of Medicine, University of Nottingham, Nottingham NG7 2RD, UK
- Correspondence: (P.O.-M.); (C.A.)
| | - Cinzia Allegrucci
- Centre for Cancer Sciences, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
- Nottingham Breast Cancer Research Centre, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
- SVMS, University of Nottingham, Sutton Bonington Campus, Loughborough LE12 5RD, UK
- Correspondence: (P.O.-M.); (C.A.)
| |
Collapse
|
13
|
Sulaiman A, Chambers J, Chilumula SC, Vinod V, Kandunuri R, McGarry S, Kim S. At the Intersection of Cardiology and Oncology: TGFβ as a Clinically Translatable Therapy for TNBC Treatment and as a Major Regulator of Post-Chemotherapy Cardiomyopathy. Cancers (Basel) 2022; 14:1577. [PMID: 35326728 PMCID: PMC8946238 DOI: 10.3390/cancers14061577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/13/2022] [Accepted: 03/17/2022] [Indexed: 02/01/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is a subtype of breast cancer that accounts for the majority of breast cancer-related deaths due to the lack of specific targets for effective treatments. While there is immense focus on the development of novel therapies for TNBC treatment, a persistent and critical issue is the rate of heart failure and cardiomyopathy, which is a leading cause of mortality and morbidity amongst cancer survivors. In this review, we highlight mechanisms of post-chemotherapeutic cardiotoxicity exposure, evaluate how this is assessed clinically and highlight the transforming growth factor-beta family (TGF-β) pathway and its significance as a mediator of cardiomyopathy. We also highlight recent findings demonstrating TGF-β inhibition as a potent method to prevent cardiac remodeling, fibrosis and cardiomyopathy. We describe how dysregulation of the TGF-β pathway is associated with negative patient outcomes across 32 types of cancer, including TNBC. We then highlight how TGF-β modulation may be a potent method to target mesenchymal (CD44+/CD24-) and epithelial (ALDHhigh) cancer stem cell (CSC) populations in TNBC models. CSCs are associated with tumorigenesis, metastasis, relapse, resistance and diminished patient prognosis; however, due to plasticity and differential regulation, these populations remain difficult to target and continue to present a major barrier to successful therapy. TGF-β inhibition represents an intersection of two fields: cardiology and oncology. Through the inhibition of cardiomyopathy, cardiac damage and heart failure may be prevented, and through CSC targeting, patient prognoses may be improved. Together, both approaches, if successfully implemented, would target the two greatest causes of cancer-related morbidity in patients and potentially lead to a breakthrough therapy.
Collapse
Affiliation(s)
- Andrew Sulaiman
- Department of Basic Science, Kansas City University, 1750 Independence Ave, Kansas City, MO 64106, USA; (S.C.C.); (V.V.); (R.K.); (S.K.)
| | - Jason Chambers
- Schulich School of Medicine, Western University, London, ON N6A5C1, Canada;
| | - Sai Charan Chilumula
- Department of Basic Science, Kansas City University, 1750 Independence Ave, Kansas City, MO 64106, USA; (S.C.C.); (V.V.); (R.K.); (S.K.)
| | - Vishak Vinod
- Department of Basic Science, Kansas City University, 1750 Independence Ave, Kansas City, MO 64106, USA; (S.C.C.); (V.V.); (R.K.); (S.K.)
| | - Rohith Kandunuri
- Department of Basic Science, Kansas City University, 1750 Independence Ave, Kansas City, MO 64106, USA; (S.C.C.); (V.V.); (R.K.); (S.K.)
| | - Sarah McGarry
- Children’s Mercy Hospital Kansas City, 2401 Gillham Rd, Kansas City, MO 64108, USA;
| | - Sung Kim
- Department of Basic Science, Kansas City University, 1750 Independence Ave, Kansas City, MO 64106, USA; (S.C.C.); (V.V.); (R.K.); (S.K.)
| |
Collapse
|
14
|
Yang R, Li Y, Wang H, Qin T, Yin X, Ma X. Therapeutic progress and challenges for triple negative breast cancer: targeted therapy and immunotherapy. MOLECULAR BIOMEDICINE 2022; 3:8. [PMID: 35243562 PMCID: PMC8894518 DOI: 10.1186/s43556-022-00071-6] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 02/09/2022] [Indexed: 02/08/2023] Open
Abstract
Triple negative breast cancer (TNBC) is a subtype of breast cancer, with estrogen receptor, human epidermal growth factor receptor 2 and progesterone receptor negative. TNBC is characterized by high heterogeneity, high rates of metastasis, poor prognosis, and lack of therapeutic targets. Now the treatment of TNBC is still based on surgery and chemotherapy, which is effective only in initial stage but almost useless in advanced stage. And due to the lack of hormone target, hormonal therapies have little beneficial effects. In recent years, signaling pathways and receptor-specific targets have been reported to be effective in TNBC patients under specific clinical conditions. Now targeted therapies have been approved for many other cancers and even other subtypes of breast cancer, but treatment options for TNBC are still limited. Most of TNBC patients showed no response, which may be related to the heterogeneity of TNBC, therefore more effective treatments and predictive biomarkers are needed. In the present review, we summarize potential treatment opinions for TNBC based on the dysregulated receptors and signaling pathways, which play a significant role in multiple stages of TNBC development. We also focus on the application of immunotherapy in TNBC, and summarize the preclinical and clinical trials of therapy for patients with TNBC. We hope to accelerate the research and development of new drugs for TNBC by understanding the relevant mechanisms, and to improve survival.
Collapse
Affiliation(s)
- Ruoning Yang
- Department of Biotherapy, State Key Laboratory of Biotherapy,Cancer Center, West China Hospital, 37 Guoxue Alley, Chengdu, 610041, PR, China.,Department of Breast Surgery, Clinical Research Center for Breast, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yueyi Li
- Department of Biotherapy, State Key Laboratory of Biotherapy,Cancer Center, West China Hospital, 37 Guoxue Alley, Chengdu, 610041, PR, China
| | - Hang Wang
- Department of Biotherapy, State Key Laboratory of Biotherapy,Cancer Center, West China Hospital, 37 Guoxue Alley, Chengdu, 610041, PR, China
| | - Taolin Qin
- West China Hospital, West China Medical School Sichuan University, Chengdu, PR, China
| | - Xiaomeng Yin
- Department of Biotherapy, State Key Laboratory of Biotherapy,Cancer Center, West China Hospital, 37 Guoxue Alley, Chengdu, 610041, PR, China
| | - Xuelei Ma
- Department of Biotherapy, State Key Laboratory of Biotherapy,Cancer Center, West China Hospital, 37 Guoxue Alley, Chengdu, 610041, PR, China.
| |
Collapse
|