1
|
Paquette B, Oweida A. Combination of radiotherapy and immunotherapy in duality with the protumoral action of radiation. Cancer Radiother 2024; 28:484-492. [PMID: 39304400 DOI: 10.1016/j.canrad.2024.07.002] [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: 05/24/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 09/22/2024]
Abstract
Radiotherapy is widely used to treat various cancers. Its combination with immune checkpoint inhibitors is intensively studied preclinically and clinically. Although the first results were very encouraging, the number of patients who respond positively remains low, and the therapeutic benefit is often temporary. This review summarizes how radiation can stimulate an antitumor immune response and its combination with immunotherapy based on inhibiting immune checkpoints. We will provide an overview of radiotherapy parameters that should be better controlled to avoid downregulating the antitumor immune response. The low response rate of combining radiotherapy and immunotherapy could, at least in part, be caused by the stimulation of cancer cell invasion and metastasis development that occur at similar doses and number of radiation fractions. To end on a positive note, we explore how a targeted inhibition of the inflammatory cytokines induced by radiation with a cyclooxygenase-2 inhibitor could both support an antitumor immune response and block radiation-induced metastasis formation.
Collapse
Affiliation(s)
- Benoît Paquette
- Centre for Research in Radiotherapy, Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, Sherbrooke, Quebec, Canada.
| | - Ayman Oweida
- Centre for Research in Radiotherapy, Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| |
Collapse
|
2
|
Elgohary S, Eissa RA, El Tayebi HM. Thymoquinone, a Novel Multi-Strike Inhibitor of Pro-Tumorigenic Breast Cancer (BC) Markers: CALR, NLRP3 Pathway and sPD-L1 in PBMCs of HR+ and TNBC Patients. Int J Mol Sci 2023; 24:14254. [PMID: 37762557 PMCID: PMC10531892 DOI: 10.3390/ijms241814254] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/19/2023] [Accepted: 06/25/2023] [Indexed: 09/29/2023] Open
Abstract
Breast cancer (BC) is not only a mass of malignant cells but also a systemic inflammatory disease. BC pro-tumorigenic inflammation has been shown to promote immune evasion and provoke BC progression. The NOD-like receptor (NLR) family pyrin domain-containing protein 3 (NLRP3) inflammasome is activated when pattern recognition receptors (PRRs) sense danger signals such as calreticulin (CALR) from damaged/dying cells, leading to the secretion of interleukin-1β (IL-1β). CALR is a novel BC biological marker, and its high levels are associated with advanced tumors. NLRP3 expression is strongly correlated with an elevated proliferative index Ki67, BC progression, metastasis, and recurrence in patients with hormone receptor-positive (HR+) and triple-negative BC (TNBC). Tumor-associated macrophages (TAMs) secrete high levels of IL-1β promoting endocrine resistance in HR+ BC. Recently, an immunosuppressive soluble form of programmed death ligand 1 (sPD-L1) has been identified as a novel prognostic biomarker in triple-negative breast cancer (TNBC) patients. Interestingly, IL-1β induces sPD-L1 release. BC Patients with elevated IL-1β and sPD-L1 levels show significantly short progression-free survival. For the first time, this study aims to investigate the inhibitory impact of thymoquinone (TQ) on CALR, the NLRP3 pathway and sPD-L1 in HR+ and TNBC. Blood samples were collected from 45 patients with BC. The effect of differing TQ concentrations for different durations on the expression of CALR, NLRP3 complex components and IL-1β as well as the protein levels of sPD-L1 and IL-1β were investigated in the peripheral blood mononuclear cells (PBMCs) and TAMs of TNBC and HR+ BC patients, respectively. The findings showed that TQ significantly downregulated the expression of CALR, NLRP3 components and IL-1β together with the protein levels of secreted IL-1β and sPD-L1. The current findings demonstrated novel immunomodulatory effects of TQ, highlighting its potential role not only as an excellent adjuvant but also as a possible immunotherapeutic agent in HR+ and TNBC patients.
Collapse
Affiliation(s)
- Sawsan Elgohary
- Clinical Pharmacology and Pharmacogenomics Research Group, Department of Pharmacology and Toxicology, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt;
| | - Reda A. Eissa
- Department of Surgery, Faculty of Medicine, Ain Shams University, Cairo 11591, Egypt;
| | - Hend M. El Tayebi
- Clinical Pharmacology and Pharmacogenomics Research Group, Department of Pharmacology and Toxicology, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt;
| |
Collapse
|
3
|
Elgohary S, El Tayebi HM. Inflammasomes in breast cancer: the ignition spark of progression and resistance? Expert Rev Mol Med 2023; 25:e22. [PMID: 37337426 DOI: 10.1017/erm.2023.14] [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] [Indexed: 06/21/2023]
Abstract
Inflammation and immune evasion are major key players in breast cancer (BC) progression. Recently, the FDA approved the use of anti-programmed death-ligand 1 antibody (anti-PD-L1) and phosphoinositide 3-kinase (PI3K) inhibitors against aggressive BC. Despite the paradigm shift in BC treatments, patients still suffer from resistance, recurrence and serious immune-related adverse events. These obstacles require unravelling of the hidden molecular contributors for such therapy failure hence yielding therapeutics that are at least as efficient yet safer. Inflammasome pathway is activated when the pattern recognition receptor senses danger signals (danger-associated molecular patterns) from damagedRdying cells or pathogen-associated molecular patterns found in microbes, leading to secretion of the active pro-inflammatory cytokines interleukin-1β (IL-1β) and interleukin-18 (IL-18). It has been shown throughout numerous studies that inflammasome pathway enhanced invasion, metastasis, provoked BC progression and therapy resistance. Additionally, inflammasomes upregulated the proliferative index ki67 and enhanced PD-L1 expression leading to immunotherapy resistance. IL-1β contributed to significant decrease in oestrogen receptor levels and promoted BC chemo-resistance. High levels of IL-18 in sera of BC patients were associated with worst prognosis. Stimulation of purinergic receptors and modulation of adipokines in obese subjects activated inflammasomes that evoked radiotherapy resistance and BC progression. The micro RNA miR-223-3p attenuated the inflammasome over-expression leading to lowered tumour volume and lessened angiogenesis in BC. This review sheds the light on the molecular pathways of inflammasomes and their impacts in distinct BC subtypes. In addition, it highlights novel strategies in treatment and prevention of BC.
Collapse
Affiliation(s)
- Sawsan Elgohary
- Clinical Pharmacology and Pharmacogenomics Research Group, Department of Pharmacology and Toxicology, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| | - Hend M El Tayebi
- Clinical Pharmacology and Pharmacogenomics Research Group, Department of Pharmacology and Toxicology, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| |
Collapse
|
4
|
Nigam M, Mishra AP, Deb VK, Dimri DB, Tiwari V, Bungau SG, Bungau AF, Radu AF. Evaluation of the association of chronic inflammation and cancer: Insights and implications. Biomed Pharmacother 2023; 164:115015. [PMID: 37321055 DOI: 10.1016/j.biopha.2023.115015] [Citation(s) in RCA: 78] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 06/02/2023] [Accepted: 06/11/2023] [Indexed: 06/17/2023] Open
Abstract
Among the most extensively researched processes in the development and treatment of cancer is inflammatory condition. Although acute inflammation is essential for the wound healing and reconstruction of tissues that have been damaged, chronic inflammation may contribute to the onset and growth of a number of diseases, including cancer. By disrupting the signaling processes of cells, which result in cancer induction, invasion, and development, a variety of inflammatory molecules are linked to the development of cancer. The microenvironment surrounding the tumor is greatly influenced by inflammatory cells and their subsequent secretions, which also contribute significantly to the tumor's growth, survivability, and potential migration. These inflammatory variables have been mentioned in several publications as prospective diagnostic tools for anticipating the onset of cancer. Targeting inflammation with various therapies can reduce the inflammatory response and potentially limit or block the proliferation of cancer cells. The scientific medical literature from the past three decades has been studied to determine how inflammatory chemicals and cell signaling pathways related to cancer invasion and metastasis are related. The current narrative review updates the relevant literature while highlighting the specifics of inflammatory signaling pathways in cancer and their possible therapeutic possibilities.
Collapse
Affiliation(s)
- Manisha Nigam
- Department of Biochemistry, Hemvati Nandan Bahuguna Garhwal University, 246174 Srinagar Garhwal, Uttarakhand, India
| | - Abhay Prakash Mishra
- Department of Pharmacology, Faculty of Health Science, University of Free State, 9300 Bloemfontein, South Africa.
| | - Vishal Kumar Deb
- Dietetics and Nutrition Technology Division, CSIR Institute of Himalayan Bioresource Technology, 176061 Palampur, Himanchal Pradesh, India
| | - Deen Bandhu Dimri
- Department of Biochemistry, Hemvati Nandan Bahuguna Garhwal University, 246174 Srinagar Garhwal, Uttarakhand, India
| | - Vinod Tiwari
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology BHU, Varanasi 221005, Uttar Pradesh, India
| | - Simona Gabriela Bungau
- Doctoral School of Biomedical Sciences, University of Oradea, 410087 Oradea, Romania; Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania.
| | - Alexa Florina Bungau
- Doctoral School of Biomedical Sciences, University of Oradea, 410087 Oradea, Romania
| | - Andrei-Flavius Radu
- Doctoral School of Biomedical Sciences, University of Oradea, 410087 Oradea, Romania; Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
| |
Collapse
|
5
|
Sun L, Wei P, Ge S, Zheng J, Ye S, Zhang Y. A nomogram based on hematological markers to predict radiosensitivity in patients with esophageal squamous cell carcinoma. Medicine (Baltimore) 2023; 102:e33282. [PMID: 36930089 PMCID: PMC10019115 DOI: 10.1097/md.0000000000033282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 02/23/2023] [Indexed: 03/18/2023] Open
Abstract
This study aimed to determine the predictive value of pretreatment levels of hematological markers on the radiosensitivity of patients with esophageal squamous cell carcinoma (ESCC). The specific hematological markers assessed included total lymphocyte count (TLC), neutrophil count, platelet count, monocyte count, neutrophil-lymphocyte ratio (NLR), platelet-lymphocyte ratio (PLR), and lymphocyte-monocyte ratio (LMR). A total of 353 ESCC patients who received radiotherapy (RT) alone or concurrent RT between 2015 and 2019 were reviewed. Pretreatment levels of hematological markers (NLR, PLR, LMR, and TLC) were used to assess the radiosensitivity of individual patients. Receiver operating characteristics curves were used to determine optimal cutoff values. Multivariate logistic models with radiosensitivity were established with meaningful results used for univariate analyses. Finally, a nomogram was developed and validated from the calibration curve and concordance index. One month after RT, 121 (34.3%) cases were shown to have low levels of radiosensitivity based on hematological markers. Univariate analyses showed that NLR, PLR, LMR, and TLC were associated with high levels of radiosensitivity (all markers, P < .05). Due to the collinearity between NLR, PLR, and LMR, these markers were separately evaluated by multivariate analysis. Multivariate analysis showed that high pretreatment NLP and PLR were independently associated with high radiosensitivity. In contrast, high pretreatment LMR and TLC were independent biomarkers associated with lower radiosensitivity. The concordance index of the nomogram was 0.737, and the calibration curves predicted by the nomogram were highly consistent with the observed experimental findings. Pretreatment hematologic markers (NLR, PLR, LMR, and TLC) can be used to predict the radiosensitivity of patients with ESCC accurately.
Collapse
Affiliation(s)
- Lijun Sun
- Affiliated Hospital of Jining Medical University, Jining, China
| | - Peng Wei
- Affiliated Hospital of Jining Medical University, Jining, China
| | - Shuang Ge
- Affiliated Hospital of Jining Medical University, Jining, China
| | - Jie Zheng
- Affiliated Hospital of Jining Medical University, Jining, China
| | - Shucheng Ye
- Affiliated Hospital of Jining Medical University, Jining, China
| | - Yanhui Zhang
- Affiliated Hospital of Jining Medical University, Jining, China
| |
Collapse
|
6
|
Oweida A, Paquette B. Reconciling two opposing effects of radiation therapy: stimulation of cancer cell invasion and activation of anti-cancer immunity. Int J Radiat Biol 2021; 99:951-963. [PMID: 34264178 DOI: 10.1080/09553002.2021.1956005] [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: 10/20/2022]
Abstract
PURPOSE The damage caused by radiation therapy to cancerous and normal cells inevitably leads to changes in the secretome profile of pro and anti-inflammatory mediators. The inflammatory response depends on the dose of radiation and its fractionation, while the inherent radiosensitivity of each patient dictates the intensity and types of adverse reactions. This review will present an overview of two apparently opposite reactions that may occur after radiation treatment: induction of an antitumor immune response and a protumoral response. Emphasis is placed on the molecular and cellular mechanisms involved. CONCLUSIONS By understanding how radiation changes the balance between anti- and protumoral effects, these forces can be manipulated to optimize radiation oncology treatments.
Collapse
Affiliation(s)
- Ayman Oweida
- Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, Universite de Sherbrooke, Sherbrooke, Canada
| | - Benoit Paquette
- Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, Universite de Sherbrooke, Sherbrooke, Canada
| |
Collapse
|
7
|
Parkins KM, Dubois VP, Kelly JJ, Chen Y, Knier NN, Foster PJ, Ronald JA. Engineering Circulating Tumor Cells as Novel Cancer Theranostics. Am J Cancer Res 2020; 10:7925-7937. [PMID: 32685030 PMCID: PMC7359075 DOI: 10.7150/thno.44259] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 06/10/2020] [Indexed: 12/13/2022] Open
Abstract
New ways to target and treat metastatic disease are urgently needed. Tumor “self-homing” describes the recruitment of circulating tumor cells (CTCs) back to a previously excised primary tumor location, contributing to tumor recurrence, as well as their migration to established metastatic lesions. Recently, self-homing CTCs have been exploited as delivery vehicles for anti-cancer therapeutics in preclinical primary tumor models. However, the ability of CTCs to self-home and treat metastatic disease is largely unknown. Methods: Here, we used bioluminescence imaging (BLI) to explore whether systemically administered CTCs home to metastatic lesions and if CTCs armed with both a reporter gene and a cytotoxic prodrug gene therapy can be used to visualize and treat metastatic disease. Results: BLI performed over time revealed a remarkable ability of CTCs to home to and treat tumors throughout the body. Excitingly, metastatic tumor burden in mice that received therapeutic CTCs was lower compared to mice receiving control CTCs. Conclusion: This study demonstrates the noteworthy ability of experimental CTCs to home to disseminated breast cancer lesions. Moreover, by incorporating a prodrug gene therapy system into our self-homing CTCs, we show exciting progress towards effective and targeted delivery of gene-based therapeutics to treat both primary and metastatic lesions.
Collapse
|
8
|
Repeated Fractions of X-Radiation to the Breast Fat Pads of Mice Augment Activation of the Autotaxin-Lysophosphatidate-Inflammatory Cycle. Cancers (Basel) 2019; 11:cancers11111816. [PMID: 31752313 PMCID: PMC6895803 DOI: 10.3390/cancers11111816] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 11/07/2019] [Accepted: 11/13/2019] [Indexed: 12/31/2022] Open
Abstract
Breast cancer patients are usually treated with multiple fractions of radiotherapy (RT) to the whole breast after lumpectomy. We hypothesized that repeated fractions of RT would progressively activate the autotaxin–lysophosphatidate-inflammatory cycle. To test this, a normal breast fat pad and a fat pad containing a mouse 4T1 tumor were irradiated with X-rays using a small-animal “image-guided” RT platform. A single RT dose of 7.5 Gy and three daily doses of 7.5 Gy increased ATX activity and decreased plasma adiponectin concentrations. The concentrations of IL-6 and TNFα in plasma and of VEGF, G-CSF, CCL11 and CXCL10 in the irradiated fat pad were increased, but only after three fractions of RT. In 4T1 breast tumor-bearing mice, three fractions of 7.5 Gy augmented tumor-induced increases in plasma ATX activity and decreased adiponectin levels in the tumor-associated mammary fat pad. There were also increased expressions of multiple inflammatory mediators in the tumor-associated mammary fat pad and in tumors, which was accompanied by increased infiltration of CD45+ leukocytes into tumor-associated adipose tissue. This work provides novel evidence that increased ATX production is an early response to RT and that repeated fractions of RT activate the autotaxin–lysophosphatidate-inflammatory cycle. This wound healing response to RT-induced damage could decrease the efficacy of further fractions of RT.
Collapse
|
9
|
Holen I, Lefley DV, Francis SE, Rennicks S, Bradbury S, Coleman RE, Ottewell P. IL-1 drives breast cancer growth and bone metastasis in vivo. Oncotarget 2018; 7:75571-75584. [PMID: 27765923 PMCID: PMC5342762 DOI: 10.18632/oncotarget.12289] [Citation(s) in RCA: 160] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 09/15/2016] [Indexed: 01/19/2023] Open
Abstract
Background We have recently identified interleukin 1B (IL-1B) as a potential biomarker for predicting breast cancer patients at increased risk for developing bone metastasis. In mouse models, IL-1B and its receptor (IL-1R1) are upregulated in breast cancer cells that metastasise to bone compared with cells that do not. We have now investigated the functional role of IL-1 by blocking IL-1R signalling with the clinically licensed antagonist, anakinra. Methodology 6-week old female BALB/c mice received a subcutaneous or intra-venous injection of MDA-MB-231-IV or MCF7 cells. Anakinra (1mg/kg/day) or placebo was administered 3 days before (preventative) or 7 days later (treatment). Tumour volume, apoptosis (TUNEL, Caspase 3), proliferation (Ki67) and angiogenesis (CD34, VEGF and endothelin) were analysed. Effects on bone were measured by uCT, and TRAP, P1NP, IL-1B, TNF alpha and IL-6 ELISA. Results Anakinra significantly reduced growth of MDA-MB-231-IV tumours in bone from 6.50+/3.00mm2 (placebo) to 2.56+/−1.07mm2 (treatment) and 0.63+/−0.18mm2 (preventative). Anakinra also reduced the number of mice that developed bone metastasis from 90% (placebo) to 40% (treatment) and 10% (preventative). Anti-tumour effects were not confined to bone, subcutaneous tumour volumes reduced from 656.68mm3 (placebo) to 160.47mm3 (treatment) and 31.08mm3 (preventative). Anakinra did not increase tumour cell apoptosis but reduced proliferation and angiogenesis in addition to exerting significant effects on the tumour environment reducing bone turnover markers, IL-1B and TNF alpha. Conclusions Our novel data demonstrate a functional role of IL-1 signalling in breast tumour progression and metastasis, supporting that anakinra could be repurposed for the treatment of breast cancer bone metastasis.
Collapse
Affiliation(s)
- Ingunn Holen
- Academic Unit of Clinical Oncology, Department of Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Sheffield S10 2RX, UK
| | - Diane V Lefley
- Academic Unit of Clinical Oncology, Department of Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Sheffield S10 2RX, UK
| | - Sheila E Francis
- Department of Infection Immunity and Cardiovascular Disease, University of Sheffield, Sheffield S10 2RX, UK
| | - Sarah Rennicks
- Academic Unit of Clinical Oncology, Department of Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Sheffield S10 2RX, UK
| | - Steven Bradbury
- Academic Unit of Clinical Oncology, Department of Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Sheffield S10 2RX, UK
| | - Robert E Coleman
- Academic Unit of Clinical Oncology, Department of Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Sheffield S10 2RX, UK
| | - Penelope Ottewell
- Academic Unit of Clinical Oncology, Department of Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Sheffield S10 2RX, UK
| |
Collapse
|
10
|
Lemay R, Lepage M, Tremblay L, Therriault H, Charest G, Paquette B. Tumor Cell Invasion Induced by Radiation in Balb/C Mouse is Prevented by the Cox-2 Inhibitor NS-398. Radiat Res 2017; 188:605-614. [PMID: 28956695 DOI: 10.1667/rr14716.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Radiation stimulates the expression of inflammatory mediators known to increase cancer cell invasion. Therefore, it is important to determine whether anti-inflammatory drugs can prevent this adverse effect of radiation. Since cyclooxygenase-2 (COX-2) is a central player in the inflammatory response, we performed studies to determine whether the COX-2 inhibitor NS-398 can reduce the radiation enhancement of cancer cell invasion. Thighs of Balb/c mice treated with NS-398 were irradiated with either daily fractions of 7.5 Gy for five consecutive days or a single 30 Gy dose prior to subcutaneous injection of nonirradiated MC7-L1 mammary cancer cells. Five weeks later, tumor invasion, blood vessel permeability and interstitial volumes were assessed using magnetic resonance imaging (MRI). Matrix metalloproteinase-2 (MMP-2) was measured in tissues by zymography at 21 days postirradiation. Cancer cell invasion in the mouse thighs was increased by 12-fold after fractionated irradiations (5 × 7.5 Gy) and by 17-fold after a single 30 Gy dose of radiation. This stimulation of cancer cell invasion was accompanied by a significant increase in the interstitial volume and a higher level of the protease MMP-2. NS-398 treatment largely prevented the stimulation of cancer cell invasion, which was associated with a reduction in interstitial volume in the irradiated thighs and a complete suppression of MMP-2 stimulation. In conclusion, this animal model using MC7-L1 cells demonstrates that radiation-induced cancer cell invasion can be largely prevented with the COX-2 inhibitor NS-398.
Collapse
Affiliation(s)
| | - Martin Lepage
- b Centre d'imagerie moléculaire de Sherbrooke, Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada, J1H 5N4
| | - Luc Tremblay
- b Centre d'imagerie moléculaire de Sherbrooke, Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada, J1H 5N4
| | | | | | | |
Collapse
|
11
|
Oweida A, Sharifi Z, Halabi H, Xu Y, Sabri S, Abdulkarim B. Differential response to ablative ionizing radiation in genetically distinct non-small cell lung cancer cells. Cancer Biol Ther 2017; 17:390-9. [PMID: 27096542 DOI: 10.1080/15384047.2016.1139241] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Stereotactic ablative radiotherapy (SABR) has emerged as a highly promising treatment for medically inoperable early-stage non-small cell lung cancer patients. Treatment outcomes after SABR have been excellent compared to conventional fractionated radiotherapy (CFRT). However, the biological determinants of the response to ablative doses of radiation remain poorly characterized. Furthermore, there's little data on the cellular and molecular response of genetically distinct NSCLC subtypes to radiation. We assessed the response of 3 genetically distinct lung adenocarcinoma cell lines to ablative and fractionated ionizing radiation (AIR and FIR). We studied clonogenic survival, cell proliferation, migration, invasion, apoptosis and senescence. We also investigated the effect of AIR and FIR on the expression of pro-invasive proteins, epithelial-to-mesenchymal transition (EMT), extracellular signal-regulated kinases (ERK1/2) and the transmembrane receptor cMET. Our findings reveal that AIR significantly reduced cell proliferation and clonogenic survival compared to FIR in A549 cells only. This differential response was not observed in HCC827 or H1975 cells. AIR significantly enhanced the invasiveness of A549 cells, but not HCC827 or H1975 cells compared to FIR. Molecular analysis of pathways involved in cell proliferation and invasion revealed that AIR significantly reduced phosphorylation of ERK1/2 and upregulated cMET expression in A549 cells. Our results show a differential proliferative and invasive response to AIR that is dependent on genetic subtype and independent of intrinsic radioresistance. Further examination of these findings in a larger panel of NSCLC cell lines and in pre-clinical models is warranted for identification of biomarkers of tumor response to AIR.
Collapse
Affiliation(s)
- Ayman Oweida
- a Department of Oncology , McGill University , Montreal , Quebec , Canada
| | - Zeinab Sharifi
- a Department of Oncology , McGill University , Montreal , Quebec , Canada
| | - Hani Halabi
- a Department of Oncology , McGill University , Montreal , Quebec , Canada
| | - Yaoxian Xu
- a Department of Oncology , McGill University , Montreal , Quebec , Canada
| | - Siham Sabri
- a Department of Oncology , McGill University , Montreal , Quebec , Canada
| | - Bassam Abdulkarim
- a Department of Oncology , McGill University , Montreal , Quebec , Canada
| |
Collapse
|
12
|
Setrerrahmane S, Xu H. Tumor-related interleukins: old validated targets for new anti-cancer drug development. Mol Cancer 2017; 16:153. [PMID: 28927416 PMCID: PMC5606116 DOI: 10.1186/s12943-017-0721-9] [Citation(s) in RCA: 174] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 09/05/2017] [Indexed: 02/07/2023] Open
Abstract
In-depth knowledge of cancer molecular and cellular mechanisms have revealed a strong regulation of cancer development and progression by the inflammation which orchestrates the tumor microenvironment. Immune cells, residents or recruited, in the inflammation milieu can have rather contrasting effects during cancer development. Accumulated clinical and experimental data support the notion that acute inflammation could exert an immunoprotective effect leading to tumor eradication. However, chronic immune response promotes tumor growth and invasion. These reactions are mediated by soluble mediators or cytokines produced by either host immune cells or tumor cells themselves. Herein, we provide an overview of the current understanding of the role of the best-validated cytokines involved in tumor progression, IL-1, IL-4 and IL-6; in addition to IL-2 cytokines family, which is known to promote tumor eradication by immune cells. Furthermore, we summarize the clinical attempts to block or bolster the effect of these tumor-related interleukins in anti-cancer therapy development.
Collapse
Affiliation(s)
- Sarra Setrerrahmane
- The Engineering Research Center of Peptide Drug Discovery and Development, China Pharmaceutical University, Nanjing, Jiangsu, 210009, People's Republic of China
| | - Hanmei Xu
- The Engineering Research Center of Peptide Drug Discovery and Development, China Pharmaceutical University, Nanjing, Jiangsu, 210009, People's Republic of China. .,State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing, Jiangsu, 210009, People's Republic of China.
| |
Collapse
|
13
|
Ryan D, Sinha A, Bogan D, Davies J, Koziol J, ElShamy WM. A niche that triggers aggressiveness within BRCA1-IRIS overexpressing triple negative tumors is supported by reciprocal interactions with the microenvironment. Oncotarget 2017; 8:103182-103206. [PMID: 29262555 PMCID: PMC5732721 DOI: 10.18632/oncotarget.20892] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 08/15/2017] [Indexed: 12/12/2022] Open
Abstract
Production of metastasis capable precursors begins within the primary tumor. Here, we define the bidirectional interactions with stromal cells involved in promoting these precursors within BRCA1-IRIS (hereafter IRIS) overexpressing (IRISOE) TNBC tumors. We define an aggressiveness niche, functionally defined as the necrotic/hypoxic core of the tumor, in which metabolically stressed, hypoxic, and inflamed IRISOE TNBC cells secrete higher levels of cytokines, chemokines and growth factors. One cytokine; IL-1β attracts mesenchymal stem cells (MSCs) to the niche and activates them to secrete CXCL1 that entrains IRISOE cells to secrete higher levels of CCL2 and VEGF. CCL2 attracts macrophages (TAMs) to the niche and activates them to secrete S100A8, and VEGF attracts endothelial cells (ECs) and activates them to secrete IL-8. In concert, CXCL1, S100A8 and IL-8 entrain aggressiveness in IRISOE TNBC cells within the niche. Indeed, compared to IRISOE cells alone, tumors developed by co-injecting IRISOE cells admixed with MSCs (10:1) in athymic mice were bigger and more aggressive. They contained more TAMs and ECs, expressed higher-levels of basal, epithelial to mesenchymal transition, and stemness biomarkers, quickly progressed to lymph-node or visceral metastases, and were highly sensitive to the IL-1β inhibitor “Anakinra”. Our findings supported by human data show that breast cancer patients with high-levels of IL-1β, CXCL1, CCL2, S100A8, VEGF, and IL-8 would show worse clinical outcomes. Our findings argue that this cytokine set is a diagnostic biomarker for patients who may benefit from an IRIS inhibitor-based therapy, and is a blue print for translation of approaches to combining that therapy with inhibitors of these bidirectional interactions to overcome TNBC metastasis.
Collapse
Affiliation(s)
- Daniel Ryan
- Breast Cancer Program, San Diego Biomedical Research Institute, San Diego, CA, USA
| | | | - Danielle Bogan
- University of Mississippi Medical Center, Jackson, MS, USA
| | - Joanna Davies
- Breast Cancer Program, San Diego Biomedical Research Institute, San Diego, CA, USA
| | - Jim Koziol
- Department of Molecular and Experimental Medicine, Scripps Research Institute, San Diego, CA, USA
| | - Wael M ElShamy
- Breast Cancer Program, San Diego Biomedical Research Institute, San Diego, CA, USA
| |
Collapse
|
14
|
Zang C, Liu X, Li B, He Y, Jing S, He Y, Wu W, Zhang B, Ma S, Dai W, Li S, Peng Z. IL-6/STAT3/TWIST inhibition reverses ionizing radiation-induced EMT and radioresistance in esophageal squamous carcinoma. Oncotarget 2017; 8:11228-11238. [PMID: 28061440 PMCID: PMC5355260 DOI: 10.18632/oncotarget.14495] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 12/27/2016] [Indexed: 01/06/2023] Open
Abstract
The acquisition of radioresistance by esophageal squamous carcinoma (ESC) cells during radiotherapy may lead to cancer recurrence and poor survival. Previous studies have demonstrated that ionizing radiation (IR) induces epithelial–mesenchymal transition (EMT) of ESC cells accompanied by increased migration, invasion, and radioresistance. However, the underlying molecular mechanisms of IR-induced EMT and radioresistance are not well established, hampering the development of potential solutions. To address this issue, we investigated the role of the IL-6/STAT3/TWIST signaling pathway in IR-induced EMT. We found not only the pathway was activated during IR-induced EMT but also STAT3 inhibition or Twist depletion reversed the EMT process and attenuated radioresistance. These results improve our understanding of the underlying mechanisms involved in IR-induced EMT and suggest potential interventions to prevent EMT-induced acquisition of radioresistance.
Collapse
Affiliation(s)
- Chunbao Zang
- Department of Radiological Medicine, Chongqing Medical University, Chonging, China
| | - Xujie Liu
- Department of Radiological Medicine, Chongqing Medical University, Chonging, China
| | - Bing Li
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chonqqing Medical University, Chongqing, China
| | - Yanqiong He
- Department of Radiological Medicine, Chongqing Medical University, Chonging, China
| | - Shen Jing
- Department of Radiological Medicine, Chongqing Medical University, Chonging, China
| | - Yujia He
- Department of Radiological Medicine, Chongqing Medical University, Chonging, China
| | - Wenli Wu
- Department of Radiological Medicine, Chongqing Medical University, Chonging, China
| | - Bingqian Zhang
- Department of Radiological Medicine, Chongqing Medical University, Chonging, China
| | - Shuhong Ma
- Department of Radiological Medicine, Chongqing Medical University, Chonging, China
| | - Weiwei Dai
- Department of Radiological Medicine, Chongqing Medical University, Chonging, China
| | - Shaolin Li
- Department of Radiological Medicine, Chongqing Medical University, Chonging, China
| | - Zhiping Peng
- Department of Radiological Medicine, Chongqing Medical University, Chonging, China
| |
Collapse
|
15
|
Gandhi S, Chandna S. Radiation-induced inflammatory cascade and its reverberating crosstalks as potential cause of post-radiotherapy second malignancies. Cancer Metastasis Rev 2017; 36:375-393. [DOI: 10.1007/s10555-017-9669-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
16
|
Zhang D, Xie M, Yang X, Zhang Y, Su Y, Wang Y, Huang H, Han H, Li W, Fu K, Su H, Xu W, Han Y, Wang R, Zhang P, Wu W, Huang Y, Chen D, Jin T, Wei J. Determination of IL-1B (rs16944) and IL-6 (rs1800796) genetic polymorphisms in IgA nephropathy in a northwest Chinese Han population. Oncotarget 2017; 8:71750-71758. [PMID: 29069743 PMCID: PMC5641086 DOI: 10.18632/oncotarget.17603] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 04/15/2017] [Indexed: 12/26/2022] Open
Abstract
IgA nephropathy (IgAN) is the most common form of primary glomerulonephritis worldwide, but etiology and pathogenesis continue to be poorly understood. Polymorphisms in the cytokine genes may play a role in the etiology and pathogenesis of IgAN. The incidence of different between diverse ethnic groups suggested important genetic influences on its pathogenesis. We genotype 10 single nucleotide polymorphisms (SNPs) in IL-1B and IL-6 gene using Sequenom Mass-ARRAY technology from 417 IgAN patients and 463 healthy controls of the Chinese Han population. We evaluated these SNPs associated with IgAN utilising the chi-square tests and genetic model analysis. We identified that the minor alleles of rs16944 (“A”), rs1800796 (“G”) in IL-1B, IL-6 were involved in an increasingly risk of IgAN in allelic model analysis, respectively. The rs16944 in IL-1B and rs1800796 in IL-6 were associated with 1.23-fold (95% CI, 1.02-1.48, P = 0.031) and 1.33-fold (95% CI, 1.11-1.66, P = 0.003) increases in the risk of developing IgAN, respectively. There was only rs1800796 still correlated with IgAN in the allelic model after adjustment by age and gender and the Bonferroni correction. In addition, Haplotype Grs1800796A rs2069837G rs2069840 (P = 0.037) and G rs1800796A rs2069837C rs2069840 (P = 0.042) in IL-6were considered to be associated with increased IgAN risk. This study verified the IL-6, IL-1B genetic variants polymorphisms contributed to IgAN susceptibility in a Chinese Han population. Although we identified SNPs susceptibility, however, replication studies and functional research are required to confirm the genetic contribution in IgAN.
Collapse
Affiliation(s)
- Daofa Zhang
- Department of Nephrology, Hainan General Hospital, Haikou, Hainan 570311, China
| | - Maowei Xie
- Department of Nephrology, Hainan General Hospital, Haikou, Hainan 570311, China
| | - Xiaohong Yang
- Department of Nephrology, Hainan General Hospital, Haikou, Hainan 570311, China
| | - Yin Zhang
- Department of Nephrology, Hainan General Hospital, Haikou, Hainan 570311, China
| | - Yan Su
- Department of Nephrology, Hainan General Hospital, Haikou, Hainan 570311, China
| | - Yanni Wang
- Department of Nephrology, Hainan General Hospital, Haikou, Hainan 570311, China
| | - Haiyang Huang
- Central Laboratory, Hainan General Hospital, Haikou, Hainan 570311, China
| | - Hui Han
- Department of Nephrology, Hainan General Hospital, Haikou, Hainan 570311, China
| | - Wenning Li
- Department of Nephrology, Hainan General Hospital, Haikou, Hainan 570311, China
| | - Keying Fu
- Central Laboratory, Hainan General Hospital, Haikou, Hainan 570311, China
| | - Huiluan Su
- Department of Nephrology, Hainan General Hospital, Haikou, Hainan 570311, China
| | - Wentan Xu
- Department of Nephrology, Hainan General Hospital, Haikou, Hainan 570311, China
| | - Yeguang Han
- Central Laboratory, Hainan General Hospital, Haikou, Hainan 570311, China
| | - Ru Wang
- Central Laboratory, Hainan General Hospital, Haikou, Hainan 570311, China
| | - Pei Zhang
- Central Laboratory, Hainan General Hospital, Haikou, Hainan 570311, China
| | - Wei Wu
- Department of Nephrology, Hainan General Hospital, Haikou, Hainan 570311, China
| | - Yun Huang
- Department of Nephrology, Hainan General Hospital, Haikou, Hainan 570311, China
| | - Daojun Chen
- Department of Nephrology, Hainan General Hospital, Haikou, Hainan 570311, China
| | - Tianbo Jin
- Key Laboratory of Resource Biology and Biotechnology in Western China, Northwest University, Ministry of Education, Xi'an, Shaanxi 710069, China.,Xi'an Tiangen Precision Medical Institute, Xi'an, Shaanxi 710075, China
| | - Jiali Wei
- Department of Nephrology, Hainan General Hospital, Haikou, Hainan 570311, China
| |
Collapse
|
17
|
Chung STM, Geerts D, Roseman K, Renaud A, Connelly L. Osteoprotegerin mediates tumor-promoting effects of Interleukin-1beta in breast cancer cells. Mol Cancer 2017; 16:27. [PMID: 28143606 PMCID: PMC5286681 DOI: 10.1186/s12943-017-0606-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 01/25/2017] [Indexed: 12/26/2022] Open
Abstract
Background It is widely recognized that inflammation promotes breast cancer invasion and metastasis. Given the complex nature of the breast tumor inflammatory microenvironment, much remains to be understood of the molecular mechanisms that govern these effects. We have previously shown that osteoprotegerin knockdown in breast cancer cells resulted in reduced invasion and metastasis. Here we present novel insight into the role of osteoprotegerin in inflammation-driven tumor progression in breast cancer by investigating the link between osteoprotegerin, macrophages and the potent pro-inflammatory cytokine Interleukin-1beta. Methods We used human breast cancer cell lines to investigate the effects of Interleukin-1beta treatment on osteoprotegerin secretion as measured by ELISA. We analyzed public datasets containing human breast cancer genome-wide mRNA expression data to reveal a significant and positive correlation between osteoprotegerin mRNA expression and the mRNA expression of Interleukin-1beta and of monocyte chemoattractant protein CC-chemokine ligand 2. Osteoprotegerin, Interleukin-1beta and CC-chemokine ligand 2 mRNA levels were also examined by qPCR on cDNA from normal and cancerous human breast tissue. We determined the effect of Interleukin-1beta–producing macrophages on osteoprotegerin expression by co-culturing breast cancer cells and differentiated THP-1 macrophages. Immunohistochemistry was performed on human breast tumor tissue microarrays to assess macrophage infiltration and osteoprotegerin expression. To demonstrate that osteoprotegerin mediated functional effects of Interleukin-1beta we performed cell invasion studies with control and OPG siRNA knockdown on Interleukin-1beta-treated breast cancer cells. Results We report that Interleukin-1beta induces osteoprotegerin secretion, independent of breast cancer subtype and basal osteoprotegerin levels. Co-culture of breast cancer cells with Interleukin-1beta-secreting macrophages resulted in a similar increase in osteoprotegerin secretion in breast cancer cells as Interleukin-1beta treatment. Macrophage infiltration correlates with osteoprotegerin secretion in human breast tumor tissue samples. We show that osteoprotegerin secretion is regulated by Interleukin-1beta in a p38- and p42/44-dependent manner. We also demonstrate that osteoprotegerin knockdown represses Interleukin-1beta expression, Interleukin-1beta-mediated breast cancer cell invasion and MMP3 expression. Conclusions These data indicate a novel role for osteoprotegerin as a mediator of inflammation- promoted breast cancer progression. Electronic supplementary material The online version of this article (doi:10.1186/s12943-017-0606-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Stephanie Tsang Mui Chung
- Department of Pharmaceutical Sciences, Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, Hilo, Hawaii, USA
| | - Dirk Geerts
- Department of Pediatric Oncology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Kim Roseman
- Department of Pharmaceutical Sciences, Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, Hilo, Hawaii, USA
| | - Ashleigh Renaud
- Department of Pharmaceutical Sciences, Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, Hilo, Hawaii, USA
| | - Linda Connelly
- Department of Pharmaceutical Sciences, Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, Hilo, Hawaii, USA.
| |
Collapse
|
18
|
Bouchard G, Therriault H, Geha S, Bujold R, Saucier C, Paquette B. Radiation-induced lung metastasis development is MT1-MMP-dependent in a triple-negative breast cancer mouse model. Br J Cancer 2017; 116:479-488. [PMID: 28103615 PMCID: PMC5318978 DOI: 10.1038/bjc.2016.448] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 12/05/2016] [Accepted: 12/15/2016] [Indexed: 12/02/2022] Open
Abstract
Background: The prognosis of triple-negative breast cancer (TNBC) is still difficult to establish. Some TNBC benefit from radiotherapy (RT) and are cured, while in other patients metastases appear during the first 3 years after treatment. In this study, an animal model of TNBC was used to determine whether the expression of the cell membrane protease MT1-MMP in cancer cells was associated with radiation-stimulated development of lung metastases. Methods: Using invasion chambers, irradiated fibroblasts were used as chemoattractants to assess the invasiveness of TNBC D2A1 cell lines showing downregulated expression of MT1-MMP, which were compared with D2A1-wt (wild-type) and D2A1 shMT1-mock (empty vector) cell lines. In a mouse model, a mammary gland was irradiated followed by the implantation of the downregulated MT1-MMP D2A1, D2A1-wt or D2A1 shMT1-mock cell lines. Migration of D2A1 cells in the mammary gland, number of circulating tumour cells and development of lung metastases were assessed. Results: The reduction of MT1-MMP expression decreased the invasiveness of D2A1 cells and blocked the radiation enhancement of cancer cell invasion. In BALB/c mice, irradiation of the mammary gland has stimulated the invasion of cancer cells, which was associated with a higher number of circulating tumour cells and of lung metastases. These adverse effects of radiation were prevented by downregulating the MT1-MMP. Conclusions: This study shows that the MT1-MMP is necessary for the radiation enhancement of lung metastasis development, and that its expression level and/or localisation could be evaluated as a biomarker for predicting the early recurrence observed in some TNBC patients.
Collapse
Affiliation(s)
- Gina Bouchard
- Centre for Research in Radiotherapy, Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, Sherbrooke, Québec, Canada
| | - Hélène Therriault
- Centre for Research in Radiotherapy, Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, Sherbrooke, Québec, Canada
| | - Sameh Geha
- Department of Pathology, Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Québec, Canada
| | - Rachel Bujold
- Centre for Research in Radiotherapy, Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, Sherbrooke, Québec, Canada.,Service of Radiation Oncology, Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Québec, Canada
| | - Caroline Saucier
- Department of Anatomy and Cellular Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Benoit Paquette
- Centre for Research in Radiotherapy, Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, Sherbrooke, Québec, Canada
| |
Collapse
|
19
|
Bouchard G, Therriault H, Bujold R, Saucier C, Paquette B. Induction of interleukin-1β by mouse mammary tumor irradiation promotes triple negative breast cancer cells invasion and metastasis development. Int J Radiat Biol 2017; 93:507-516. [PMID: 27935337 DOI: 10.1080/09553002.2017.1270471] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
PURPOSE Radiotherapy increases the level of inflammatory cytokines, some of which are known to promote metastasis. In a mouse model of triple negative breast cancer (TNBC), we determined whether irradiation of the mammary tumor increases the level of key cytokines and favors the development of lung metastases. MATERIALS AND METHODS D2A1 TNBC cells were implanted in the mammary glands of a Balb/c mouse and then 7 days old tumors were irradiated (4 × 6 Gy). The cytokines IL-1β, IL-4, IL-6, IL-10, IL-17 and MIP-2 were quantified in plasma before, midway and after irradiation. The effect of tumor irradiation on the invasion of cancer cells, the number of circulating tumor cells (CTC) and lung metastases were also measured. RESULTS TNBC tumor irradiation significantly increased the plasma level of IL-1β, which was associated with a greater number of CTC (3.5-fold) and lung metastases (2.3-fold), compared to sham-irradiated animals. Enhancement of D2A1 cell invasion in mammary gland was associated with an increase of the matrix metalloproteinases-2 and -9 activity (MMP-2, -9). The ability of IL-1β to stimulate the invasiveness of irradiated D2A1 cells was confirmed by in vitro invasion chamber assays. CONCLUSION Irradiation targeting a D2A1 tumor and its microenvironment increased the level of the inflammatory cytokine IL-1β and was associated with the promotion of cancer cell invasion and lung metastasis development.
Collapse
Affiliation(s)
- Gina Bouchard
- a Centre for Research in Radiotherapy, Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences , Université de Sherbrooke , Sherbrooke , Québec , Canada
| | - Hélène Therriault
- a Centre for Research in Radiotherapy, Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences , Université de Sherbrooke , Sherbrooke , Québec , Canada
| | - Rachel Bujold
- b Service of Radiation Oncology , Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke , Sherbrooke , Québec , Canada
| | - Caroline Saucier
- c Department of Anatomy and Cellular Biology, Faculty of Medicine and Health Sciences , Université de Sherbrooke , Sherbrooke , Québec , Canada
| | - Benoit Paquette
- a Centre for Research in Radiotherapy, Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences , Université de Sherbrooke , Sherbrooke , Québec , Canada
| |
Collapse
|
20
|
Vilalta M, Rafat M, Graves EE. Effects of radiation on metastasis and tumor cell migration. Cell Mol Life Sci 2016; 73:2999-3007. [PMID: 27022944 PMCID: PMC4956569 DOI: 10.1007/s00018-016-2210-5] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 02/25/2016] [Accepted: 03/22/2016] [Indexed: 12/25/2022]
Abstract
It is well known that tumor cells migrate from the primary lesion to distant sites to form metastases and that these lesions limit patient outcome in a majority of cases. However, the extent to which radiation influences this process and to which migration in turn alters radiation response remains controversial. There are preclinical and clinical reports showing that focal radiotherapy can both increase the development of distant metastasis, as well as that it can induce the regression of established metastases through the abscopal effect. More recently, preclinical studies have suggested that radiation can attract migrating tumor cells and may, thereby, facilitate tumor recurrence. In this review, we summarize these phenomena and their potential mechanisms of action, and evaluate their significance for modern radiation therapy strategies.
Collapse
Affiliation(s)
- Marta Vilalta
- Department of Radiation Oncology, Stanford University, 269 Campus Dr., CCSR South Rm. 1255A, Stanford, CA, 94305-5152, USA
| | - Marjan Rafat
- Department of Radiation Oncology, Stanford University, 269 Campus Dr., CCSR South Rm. 1255A, Stanford, CA, 94305-5152, USA
| | - Edward E Graves
- Department of Radiation Oncology, Stanford University, 269 Campus Dr., CCSR South Rm. 1255A, Stanford, CA, 94305-5152, USA.
| |
Collapse
|
21
|
Bouchard G, Therriault H, Geha S, Bérubé-Lauzière Y, Bujold R, Saucier C, Paquette B. Stimulation of triple negative breast cancer cell migration and metastases formation is prevented by chloroquine in a pre-irradiated mouse model. BMC Cancer 2016; 16:361. [PMID: 27282478 PMCID: PMC4901430 DOI: 10.1186/s12885-016-2393-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 06/01/2016] [Indexed: 12/04/2022] Open
Abstract
Background Some triple negative breast cancer (TNBC) patients are at higher risk of recurrence in the first three years after treatment. This rapid relapse has been suggested to be associated with inflammatory mediators induced by radiation in healthy tissues that stimulate cancer cell migration and metastasis formation. In this study, the ability of chloroquine (CQ) to inhibit radiation-stimulated development of metastasis was assessed. Methods The capacity of CQ to prevent radiation-enhancement of cancer cell invasion was assessed in vitro with the TNBC cell lines D2A1, 4T1 and MDA-MB-231 and the non-TNBC cell lines MC7-L1, and MCF-7. In Balb/c mice, a single mammary gland was irradiated with four daily doses of 6 Gy. After the last irradiation, irradiated and control mammary glands were implanted with D2A1 cells. Mice were treated with CQ (vehicle, 40 or 60 mg/kg) 3 h before each irradiation and then every 72 h for 3 weeks. Migration of D2A1 cells in the mammary gland, the number of circulating tumor cells and lung metastasis were quantified, and also the expression of some inflammatory mediators. Results Irradiated fibroblasts have increased the invasiveness of the TNBC cell lines only, a stimulation that was prevented by CQ. On the other hand, invasiveness of the non-TNBC cell lines, which was not enhanced by irradiated fibroblasts, was also not significantly modified by CQ. In Balb/c mice, treatment with CQ prevented the stimulation of D2A1 TNBC cell migration in the pre-irradiated mammary gland, and reduced the number of circulating tumor cells and lung metastases. This protective effect of CQ was associated with a reduced expression of the inflammatory mediators interleukin-1β, interleukin-6, and cyclooxygenase-2, while the levels of matrix metalloproteinases-2 and −9 were not modified. CQ also promoted a blocking of autophagy. Conclusion CQ prevented radiation-enhancement of TNBC cell invasion and reduced the number of lung metastases in a mouse model. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2393-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Gina Bouchard
- Centre for Research in Radiotherapy, Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, 3001, 12e Avenue Nord, Sherbrooke, Québec, J1H 5 N4, Canada
| | - Hélène Therriault
- Centre for Research in Radiotherapy, Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, 3001, 12e Avenue Nord, Sherbrooke, Québec, J1H 5 N4, Canada
| | - Sameh Geha
- Department of Pathology, Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Canada
| | - Yves Bérubé-Lauzière
- Department of Electrical and Computer Engineering, Centre d'imagerie moléculaire de Sherbrooke, Sherbrooke, Québec, Canada
| | - Rachel Bujold
- Centre for Research in Radiotherapy, Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, 3001, 12e Avenue Nord, Sherbrooke, Québec, J1H 5 N4, Canada.,Service of Radiation Oncology, Université de Sherbrooke, Sherbrooke, Canada
| | - Caroline Saucier
- Department of Anatomy and Cellular Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Canada
| | - Benoit Paquette
- Centre for Research in Radiotherapy, Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, 3001, 12e Avenue Nord, Sherbrooke, Québec, J1H 5 N4, Canada.
| |
Collapse
|
22
|
Desmarais G, Charest G, Therriault H, Shi M, Fortin D, Bujold R, Mathieu D, Paquette B. Infiltration of F98 glioma cells in Fischer rat brain is temporary stimulated by radiation. Int J Radiat Biol 2016; 92:444-50. [DOI: 10.1080/09553002.2016.1175682] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Guillaume Desmarais
- Center for Research in Radiotherapy, Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Gabriel Charest
- Department of Radiation Oncology, Stanford School of Medicine, Stanford University, Palo Alto, California, USA
| | - Hélène Therriault
- Center for Research in Radiotherapy, Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Minghan Shi
- Center for Research in Radiotherapy, Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - David Fortin
- Department of Surgery, Service of Neurosurgery/Neuro-oncology, Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Québec, Canada
| | - Rachel Bujold
- Center for Research in Radiotherapy, Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
- Service of Radiation Oncology, Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Québec, Canada
| | - David Mathieu
- Department of Radiation Oncology, Stanford School of Medicine, Stanford University, Palo Alto, California, USA
| | - Benoit Paquette
- Center for Research in Radiotherapy, Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
| |
Collapse
|
23
|
Zhang Y, Liu S, Wang H, Yang W, Li F, Yang F, Yu D, Ramsey FV, Tuszyski GP, Hu W. Elevated NIBP/TRAPPC9 mediates tumorigenesis of cancer cells through NFκB signaling. Oncotarget 2016; 6:6160-78. [PMID: 25704885 PMCID: PMC4467429 DOI: 10.18632/oncotarget.3349] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 01/20/2015] [Indexed: 12/21/2022] Open
Abstract
Regulatory mechanisms underlying constitutive and inducible NFκB activation in cancer remain largely unknown. Here we investigated whether a novel NIK- and IKK2-binding protein (NIBP) is required for maintaining malignancy of cancer cells in an NFκB-dependent manner. Real-time polymerase chain reaction analysis of a human cancer survey tissue-scan cDNA array, immunostaining of a human frozen tumor tissue array and immunoblotting of a high-density reverse-phase cancer protein lysate array showed that NIBP is extensively expressed in most tumor tissues, particularly in breast and colon cancer. Lentivirus-mediated NIBP shRNA knockdown significantly inhibited the growth/proliferation, invasion/migration, colony formation and xenograft tumorigenesis of breast (MDA-MB-231) or colon (HCT116) cancer cells. NIBP overexpression in HCT116 cells promoted cell proliferation, migration and colony formation. Mechanistically, NIBP knockdown in cancer cells inhibited cytokine-induced activation of NFκB luciferase reporter, thus sensitizing the cells to TNFα-induced apoptosis. Endogenous NIBP bound specifically to the phosphorylated IKK2 in a TNFα-dependent manner. NIBP knockdown transiently attenuated TNFα-stimulated phosphorylation of IKK2/p65 and degradation of IκBα. In contrast, NIBP overexpression enhanced TNFα-induced NFκB activation, thus inhibiting constitutive and TNFα-induced apoptosis. Collectively, our data identified important roles of NIBP in promoting tumorigenesis via NFκΒ signaling, spotlighting NIBP as a promising target in cancer therapeutic intervention.
Collapse
Affiliation(s)
- Yonggang Zhang
- Department of Neuroscience, Temple University School of Medicine, Philadelphia, PA, USA
| | - Shu Liu
- Department of Biotherapy, The Forth Affiliated Hospital, China Medical University, Shenyang, Liaoning, China
| | - Hong Wang
- Department of Neuroscience, Temple University School of Medicine, Philadelphia, PA, USA
| | - Wensheng Yang
- Department of Neuroscience, Temple University School of Medicine, Philadelphia, PA, USA
| | - Fang Li
- Department of Neuroscience, Temple University School of Medicine, Philadelphia, PA, USA
| | - Fan Yang
- Department of Neuroscience, Temple University School of Medicine, Philadelphia, PA, USA
| | - Daohai Yu
- Department of Clinical Sciences, Temple University School of Medicine, Philadelphia, PA, USA
| | - Frederick V Ramsey
- Department of Clinical Sciences, Temple University School of Medicine, Philadelphia, PA, USA
| | - George P Tuszyski
- Department of Neuroscience, Temple University School of Medicine, Philadelphia, PA, USA
| | - Wenhui Hu
- Department of Neuroscience, Temple University School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
24
|
Desmarais G, Charest G, Fortin D, Bujold R, Mathieu D, Paquette B. Cyclooxygenase-2 inhibitor prevents radiation-enhanced infiltration of F98 glioma cells in brain of Fischer rat. Int J Radiat Biol 2015; 91:624-33. [PMID: 25912457 DOI: 10.3109/09553002.2015.1043756] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
PURPOSE Radiation induces a neuro-inflammation that is characterized by the expression of genes known to increase the invasion of cancer cells. In Fischer rats, brain irradiation increases the infiltration of cancer cells and reduced the median survival of the animals. In this study, we have determined whether these adverse effects of radiation can be prevented with the cyclooxygenase-2 (COX-2) inhibitor meloxicam. MATERIALS AND METHODS Brain of Fischer rats treated or not with meloxicam were irradiated (15 Gy) and then implanted with the F98 glioma cells. The median survival of the animals, the infiltration of F98 cells, and the expression of inflammatory cytokines and pro-migration molecules were measured. RESULTS Meloxicam reduced by 75% the production of prostaglandin E2 (bioproduct of COX-2) in irradiated brains validating its anti-inflammatory effect. Median survival was increased to control levels by the treatment of meloxicam following brain irradiation. This protective effect was associated with a reduction of the infiltration of F98 cells in the brain, a complete inhibition of radiation-enhancement of matrix metalloproteinase-2, and a significant reduction of tumor necrosis factor α (TNF-α) and tumor growth factor β1 (TGF-β1) expression. Using invasion chambers, interleukin-1β (IL-1β) stimulated by 5-fold the invasiveness of F98 cells, but this stimulation was completely inhibited by meloxicam. This suggests that a cooperation between IL-1β and COX-2 are involved in radiation-enhancement of F98 cell invasion. CONCLUSIONS Our results indicate the importance of reducing the inflammatory response of normal brain tissue following irradiation in an effort to extend median survival in F98 tumor-bearing rats.
Collapse
Affiliation(s)
- Guillaume Desmarais
- a Center for Research in Radiotherapy, Department of Nuclear Medicine and Radiobiology , Faculty of Medicine and Health Sciences, Université de Sherbrooke , Québec , Canada
| | - Gabriel Charest
- a Center for Research in Radiotherapy, Department of Nuclear Medicine and Radiobiology , Faculty of Medicine and Health Sciences, Université de Sherbrooke , Québec , Canada
| | - David Fortin
- b Department of Surgery , Division of Neurosurgery/Neuro-oncology , Québec , Canada
| | - Rachel Bujold
- a Center for Research in Radiotherapy, Department of Nuclear Medicine and Radiobiology , Faculty of Medicine and Health Sciences, Université de Sherbrooke , Québec , Canada.,c Division of Radiation Oncology, Centre Hospitalier Universitaire de Sherbrooke , Sherbrooke, Québec , Canada
| | - David Mathieu
- b Department of Surgery , Division of Neurosurgery/Neuro-oncology , Québec , Canada
| | - Benoit Paquette
- a Center for Research in Radiotherapy, Department of Nuclear Medicine and Radiobiology , Faculty of Medicine and Health Sciences, Université de Sherbrooke , Québec , Canada
| |
Collapse
|
25
|
Di Maggio FM, Minafra L, Forte GI, Cammarata FP, Lio D, Messa C, Gilardi MC, Bravatà V. Portrait of inflammatory response to ionizing radiation treatment. J Inflamm (Lond) 2015; 12:14. [PMID: 25705130 PMCID: PMC4336767 DOI: 10.1186/s12950-015-0058-3] [Citation(s) in RCA: 196] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 01/29/2015] [Indexed: 01/05/2023] Open
Abstract
Ionizing radiation (IR) activates both pro-and anti-proliferative signal pathways producing an imbalance in cell fate decision. IR is able to regulate several genes and factors involved in cell-cycle progression, survival and/or cell death, DNA repair and inflammation modulating an intracellular radiation-dependent response. Radiation therapy can modulate anti-tumour immune responses, modifying tumour and its microenvironment. In this review, we report how IR could stimulate inflammatory factors to affect cell fate via multiple pathways, describing their roles on gene expression regulation, fibrosis and invasive processes. Understanding the complex relationship between IR, inflammation and immune responses in cancer, opens up new avenues for radiation research and therapy in order to optimize and personalize radiation therapy treatment for each patient.
Collapse
Affiliation(s)
- Federica Maria Di Maggio
- />Department of Pathobiology and Medical and Forensic Biotechnologies, University of Palermo, Palermo, Italy
- />IBFM CNR – LATO, Contrada Pietrapollastra Pisciotto, Cefalù, PA Italy
| | - Luigi Minafra
- />IBFM CNR – LATO, Contrada Pietrapollastra Pisciotto, Cefalù, PA Italy
| | - Giusi Irma Forte
- />IBFM CNR – LATO, Contrada Pietrapollastra Pisciotto, Cefalù, PA Italy
| | | | - Domenico Lio
- />Department of Pathobiology and Medical and Forensic Biotechnologies, University of Palermo, Palermo, Italy
| | - Cristina Messa
- />IBFM CNR – LATO, Contrada Pietrapollastra Pisciotto, Cefalù, PA Italy
- />Department of Health Sciences, Tecnomed Foundation, University of Milano-Bicocca, Milan, Italy
- />Nuclear Medicine Center, San Gerardo Hospital, Monza, Italy
| | - Maria Carla Gilardi
- />IBFM CNR – LATO, Contrada Pietrapollastra Pisciotto, Cefalù, PA Italy
- />Department of Health Sciences, Tecnomed Foundation, University of Milano-Bicocca, Milan, Italy
- />Nuclear Medicine, San Raffaele Scientific Institute, Milan, Italy
| | - Valentina Bravatà
- />IBFM CNR – LATO, Contrada Pietrapollastra Pisciotto, Cefalù, PA Italy
| |
Collapse
|
26
|
Vilalta M, Rafat M, Giaccia AJ, Graves EE. Recruitment of circulating breast cancer cells is stimulated by radiotherapy. Cell Rep 2014; 8:402-9. [PMID: 25017065 DOI: 10.1016/j.celrep.2014.06.011] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Revised: 05/07/2014] [Accepted: 06/10/2014] [Indexed: 12/31/2022] Open
Abstract
Radiotherapy (RT) is a localized therapy that is highly effective in killing primary tumor cells located within the field of the radiation beam. We present evidence that irradiation of breast tumors can attract migrating breast cancer cells. Granulocyte-macrophage colony stimulating factor (GM-CSF) produced by tumor cells in response to radiation stimulates the recruitment of migrating tumor cells to irradiated tumors, suggesting a mechanism of tumor recurrence after radiation facilitated by transit of unirradiated, viable circulating tumor cells to irradiated tumors. Data supporting this hypothesis are presented through in vitro invasion assays and in vivo orthotopic models of breast cancer. Our work provides a mechanism for tumor recurrence in which RT attracts cells outside the radiation field to migrate to the site of treatment.
Collapse
Affiliation(s)
- Marta Vilalta
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA
| | - Marjan Rafat
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA
| | - Amato J Giaccia
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA
| | - Edward E Graves
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA; Division of Radiation Physics, Department of Radiation Oncology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
27
|
Han J, Bae SY, Oh SJ, Lee J, Lee JH, Lee HC, Lee SK, Kil WH, Kim SW, Nam SJ, Kim S, Lee JE. Zerumbone suppresses IL-1β-induced cell migration and invasion by inhibiting IL-8 and MMP-3 expression in human triple-negative breast cancer cells. Phytother Res 2014; 28:1654-60. [PMID: 24890258 DOI: 10.1002/ptr.5178] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 05/01/2014] [Indexed: 12/19/2022]
Abstract
Inflammation is a key regulatory process in cancer development. Prolonged exposure of breast tumor cells to inflammatory cytokines leads to epithelial-mesenchymal transition, which is the principal mechanism involved in metastasis and tumor invasion. Interleukin (IL)-1β is a major inflammatory cytokine in a variety of tumors. To date, the regulatory mechanism of IL-1β-induced cell migration and invasion has not been fully elucidated. Here, we investigated the effect of zerumbone (ZER) on IL-1β-induced cell migration and invasion in breast cancer cells. The levels of IL-8 and matrix metalloproteinase (MMP)-3 mRNA were analyzed by real-time polymerase chain reaction. The levels of secreted IL-8 and MMP-3 protein were analyzed by enzyme-linked immunosorbent assay and western blot analysis, respectively. Cell invasion and migration was detected by Boyden chamber assay. The levels of IL-8 and MMP-3 expression were significantly increased by IL-1β treatment in Hs578T and MDA-MB231 cells. On the other hand, IL-1β-induced IL-8 and MMP-3 expression was decreased by ZER. Finally, IL-1β-induced cell migration and invasion were decreased by ZER in Hs578T and MDA-MB231 cells. ZER suppresses IL-1β-induced cell migration and invasion by inhibiting IL-8 expression and MMP-3 expression in TNBC cells. ZER could be a promising therapeutic drug for treatment of triple-negative breast cancer patients.
Collapse
Affiliation(s)
- Jeonghun Han
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, 50 Irwon-dong, Gangnam-gu, Seoul, Korea; Department of Life Science, Hanyang University, Seoul, 133-791, Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|