1
|
Zhang M, Zhou K, Wang Z, Liu T, Stevens LE, Lynce F, Chen WY, Peng S, Xie Y, Zhai D, Chen Q, Shi Y, Shi H, Yuan Z, Li X, Xu J, Cai Z, Guo J, Shao N, Lin Y. A Subpopulation of Luminal Progenitors Secretes Pleiotrophin to Promote Angiogenesis and Metastasis in Inflammatory Breast Cancer. Cancer Res 2024; 84:1781-1798. [PMID: 38507720 PMCID: PMC11148543 DOI: 10.1158/0008-5472.can-23-2640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 01/19/2024] [Accepted: 03/14/2024] [Indexed: 03/22/2024]
Abstract
Inflammatory breast cancer (IBC) is a highly aggressive subtype of breast cancer characterized by rapidly arising diffuse erythema and edema. Genomic studies have not identified consistent alterations and mechanisms that differentiate IBC from non-IBC tumors, suggesting that the microenvironment could be a potential driver of IBC phenotypes. Here, using single-cell RNA sequencing, multiplex staining, and serum analysis in patients with IBC, we identified enrichment of a subgroup of luminal progenitor (LP) cells containing high expression of the neurotropic cytokine pleiotrophin (PTN) in IBC tumors. PTN secreted by the LP cells promoted angiogenesis by directly interacting with the NRP1 receptor on endothelial tip cells located in both IBC tumors and the affected skin. NRP1 activation in tip cells led to recruitment of immature perivascular cells in the affected skin of IBC, which are correlated with increased angiogenesis and IBC metastasis. Together, these findings reveal a role for cross-talk between LPs, endothelial tip cells, and immature perivascular cells via PTN-NRP1 axis in the pathogenesis of IBC, which could lead to improved strategies for treating IBC. SIGNIFICANCE Nonmalignant luminal progenitor cells expressing pleiotrophin promote angiogenesis by activating NRP1 and induce a prometastatic tumor microenvironment in inflammatory breast cancer, providing potential therapeutic targets for this aggressive breast cancer subtype.
Collapse
Affiliation(s)
- Mengmeng Zhang
- Breast Disease Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Kaiwen Zhou
- Breast Disease Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zilin Wang
- Breast Disease Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ting Liu
- Breast Disease Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Laura E Stevens
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Filipa Lynce
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Wendy Y Chen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Sui Peng
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yubin Xie
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Duanyang Zhai
- Breast Disease Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qianjun Chen
- Department of Breast Oncology, Traditional Chinese Medicine Hospital of Guangdong Province, Guangzhou, Guangdong, China
| | - Yawei Shi
- Breast Disease Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Huijuan Shi
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhongyu Yuan
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiaoping Li
- Department of Breast Oncology, Jiangmen Central Hospital, Jiangmen, China
| | - Juan Xu
- Department of Breast Oncology, Maternal and Child Health Care Hospital of Guangdong Province, Guangzhou, China
| | - Zhenhai Cai
- Department of Breast Oncology, Jieyang People's Hospital, Jieyang, China
| | - Jianping Guo
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Nan Shao
- Breast Disease Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ying Lin
- Breast Disease Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
2
|
Tarek A, Mohamed HT, El-Sharkawy AA, El-Sayed SK, Hirshon JM, Woodward WA, El-Shinawi M, Mohamed MM. Differential Gene Expression of fresh tissue and patient-derived explants' matricellular proteins augment inflammatory breast cancer metastasis: the possible role of IL-6 and MCP-1. QJM 2023; 116:345-354. [PMID: 36592055 PMCID: PMC10226750 DOI: 10.1093/qjmed/hcac284] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 11/25/2022] [Accepted: 12/22/2022] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Matricellular proteins comprising matrisome and adhesome are responsible for structure integrity and interactions between cells in the tumour microenvironment of breast cancer. Changes in the gene expression of matrisome and adhesome augment metastasis. Since inflammatory breast cancer (IBC) is characterized by high metastatic behaviour. Herein, we compared the gene expression profile of matrisome and adhesome in non-IBC and IBC in fresh tissue and ex vivo patient-derived explants (PDEs) and we also compared the secretory inflammatory mediators of PDEs in non-IBC and IBC to identify secretory cytokines participate in cross-talk between cells via interactions with matrisome and adhisome. METHODS Fifty patients (31 non-IBC and 19 IBC) were enrolled in the present study. To test their validation in clinical studies, PDEs were cultured as an ex vivo model. Gene expression and cytokine array were used to identify candidate genes and cytokines contributing to metastasis in the examined fresh tissues and PDEs. Bioinformatics analysis was applied on identified differentially expressed genes using GeneMANIA and Metascape gene annotation and analysis resource to identify pathways involved in IBC metastasis. RESULTS Normal and cancer fresh tissues and PDEs of IBC were characterized by overexpression of CDH1 and MMP14 and downregulation of CTNNA1 and TIMP1 compared with non-IBC. The secretome of IBC cancer PDEs is characterized by significantly high expression of interleukin 6 and monocyte chemoattractant protein-1 (CCL2) compared with non-IBC. CONCLUSION Genes expressed by adhisome and matrisome play a significant role in IBC metastasis and should be considered novel target therapy.
Collapse
Affiliation(s)
- Alshaimaa Tarek
- From the Department of Zoology, Faculty of Science, Cairo University, Giza 12613, Egypt
| | - Hossam Taha Mohamed
- From the Department of Zoology, Faculty of Science, Cairo University, Giza 12613, Egypt
- Faculty of Biotechnology, October University for Modern Sciences and Arts, Giza 12451, Egypt
| | - Aya Ali El-Sharkawy
- From the Department of Zoology, Faculty of Science, Cairo University, Giza 12613, Egypt
| | | | - Jon Mark Hirshon
- School of Medicine, University of Maryland, Baltimore, MD 21201, USA
| | - Wendy A Woodward
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mohamed El-Shinawi
- Faculty of Medicine, Galala University, Suez 43511, Egypt
- Department of General Surgery, Faculty of Medicine, Ain Shams University, Cairo 11566, Egypt
| | - Mona Mostafa Mohamed
- From the Department of Zoology, Faculty of Science, Cairo University, Giza 12613, Egypt
| |
Collapse
|
3
|
Ortiz-Soto G, Babilonia-Díaz NS, Lacourt-Ventura MY, Rivera-Rodríguez DM, Quiñones-Rodríguez JI, Colón-Vargas M, Almodóvar-Rivera I, Ferrer-Torres LE, Suárez-Arroyo IJ, Martínez-Montemayor MM. Metadherin Regulates Inflammatory Breast Cancer Invasion and Metastasis. Int J Mol Sci 2023; 24:4694. [PMID: 36902125 PMCID: PMC10002532 DOI: 10.3390/ijms24054694] [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: 01/05/2023] [Revised: 02/25/2023] [Accepted: 02/26/2023] [Indexed: 03/05/2023] Open
Abstract
Inflammatory breast cancer (IBC) is one of the most lethal subtypes of breast cancer (BC), accounting for approximately 1-5% of all cases of BC. Challenges in IBC include accurate and early diagnosis and the development of effective targeted therapies. Our previous studies identified the overexpression of metadherin (MTDH) in the plasma membrane of IBC cells, further confirmed in patient tissues. MTDH has been found to play a role in signaling pathways related to cancer. However, its mechanism of action in the progression of IBC remains unknown. To evaluate the function of MTDH, SUM-149 and SUM-190 IBC cells were edited with CRISPR/Cas9 vectors for in vitro characterization studies and used in mouse IBC xenografts. Our results demonstrate that the absence of MTDH significantly reduces IBC cell migration, proliferation, tumor spheroid formation, and the expression of NF-κB and STAT3 signaling molecules, which are crucial oncogenic pathways in IBC. Furthermore, IBC xenografts showed significant differences in tumor growth patterns, and lung tissue revealed epithelial-like cells in 43% of wild-type (WT) compared to 29% of CRISPR xenografts. Our study emphasizes the role of MTDH as a potential therapeutic target for the progression of IBC.
Collapse
Affiliation(s)
- Gabriela Ortiz-Soto
- Department of Biochemistry, Universidad Central del Caribe-School of Medicine, Bayamón, PR 00960, USA
| | - Natalia S. Babilonia-Díaz
- Department of Biochemistry, Universidad Central del Caribe-School of Medicine, Bayamón, PR 00960, USA
| | | | | | - Jailenne I. Quiñones-Rodríguez
- Department of Clinical Anatomy, College of Osteopathic Medicine, Sam Houston State University, Conroe, TX 77304, USA
- Department of Anatomy and Cell Biology, School of Medicine, Universidad Central del Caribe, Bayamón, PR 00960, USA
| | - Mónica Colón-Vargas
- Department of Mathematical Sciences, University of Puerto Rico at Mayagüez, Mayagüez, PR 00681, USA
| | - Israel Almodóvar-Rivera
- Department of Mathematical Sciences, University of Puerto Rico at Mayagüez, Mayagüez, PR 00681, USA
| | - Luis E. Ferrer-Torres
- Department of Pathology and Laboratory Medicine, Hospital Interamericano de Medicina Avanzada (H.I.M.A.)—San Pablo Caguas, Caguas, PR 00725, USA
- Department of Immunopathology, Hato Rey Pathology Associates Inc. (HRPLABS), San Juan, PR 00936, USA
| | - Ivette J. Suárez-Arroyo
- Department of Biochemistry, Universidad Central del Caribe-School of Medicine, Bayamón, PR 00960, USA
| | | |
Collapse
|
4
|
Geometric tumor embolic budding characterizes inflammatory breast cancer. Breast Cancer Res Treat 2023; 197:461-478. [PMID: 36473978 PMCID: PMC9734724 DOI: 10.1007/s10549-022-06819-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 11/15/2022] [Indexed: 12/12/2022]
Abstract
PURPOSE Inflammatory breast cancer (IBC) is characterized by numerous tumor emboli especially within dermal lymphatics. The explanation remains a mystery. METHODS This study combines experimental studies with two different IBC xenografts with image algorithmic studies utilizing human tissue microarrays (TMAs) of IBC vs non-IBC cases to support a novel hypothesis to explain IBC's sina qua non signature of florid lymphovascular emboli. RESULTS In the human TMAs, compared to tumor features like nuclear grade (size), mitosis and Ki-67 immunoreactivity which show that IBC is only modestly more proliferative with larger nuclei than non-IBC, what really sets IBC apart is the markedly greater number of tumor emboli and distinctly smaller emboli whose numbers indicate geometric or exponential differences between IBC and non-IBC. In the experimental xenograft studies, Mary-X gives rise to tight spheroids in vitro which exhibit dynamic budding into smaller daughter spheroids whereas Karen-X exhibits only loose non-budding aggregates. Furthermore Mary-X emboli also bud dramatically into smaller daughter emboli in vivo. The mechanism that regulates this involves the generation of E-cad/NTF1, a calpain-mediated cleavage 100 kDa product of 120 kDa full length membrane E-cadherin. Inhibiting this calpain-mediated cleavage of E-cadherin by blocking either the calpain site of cleavage (SC) or the site of binding (SB) with specific decapeptides that both penetrate the cell membrane and mimic either the cleavage site or the binding site on E-cadherin, inhibits the generation of E-cad/NTF1 in a dose-dependent manner, reduces spheroid compactness and decreases budding. CONCLUSION Since E-cad/NFT1 retains the p120ctn binding site but loses the α-and β-catenin sites, promoting its 360° distribution around the cell's membrane, the vacilating levels of this molecule trigger budding of both the spheroids as well as the emboli. Recurrent and geometric budding of parental emboli into daughter emboli then would account for the plethora of emboli seen in IBC.
Collapse
|
5
|
Sannappa Gowda NG, Shiragannavar VD, Puttahanumantharayappa LD, Shivakumar AT, Dallavalasa S, Basavaraju CG, Bhat SS, Prasad SK, Vamadevaiah RM, Madhunapantula SV, Santhekadur PK. Quercetin activates vitamin D receptor and ameliorates breast cancer induced hepatic inflammation and fibrosis. Front Nutr 2023; 10:1158633. [PMID: 37153919 PMCID: PMC10157213 DOI: 10.3389/fnut.2023.1158633] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 03/24/2023] [Indexed: 05/10/2023] Open
Abstract
Aims To explore the hepatoprotective role of quercetin and its novel molecular mechanism of action on breast cancer associated hepatic inflammation and fibrosis via Vitamin D receptor (VDR). Main methods We used Ehrlich Ascites Carcinoma (mouse mammary carcinoma) model for our in-vivo experiments and human breast cancer cell lines for in-vitro assays. We inoculated 1.5 × 106 Ehrlich ascites carcinoma cells into female Swiss albino mice. Quercetin (50 mg/kg) was administered intraperitoneally for 15 days. Liver enzymes activity was determined using a spectrophotometric assay. The hallmarks of inflammation and fibrosis were determined using Immunohistochemistry. The effect of quercetin on tumor formation was elucidated using human breast cancer cell lines and chick chorioallantoic membrane assay. Docking study was performed to explore the binding mode of quercetin with VDR. Key findings In EAC tumor-bearing mice, cell numbers, tumor volume, body weight and liver weight were dramatically increased, while they significantly decreased in mice treated with quercetin. Additionally, the peritoneal neo-angiogenesis was also significantly suppressed in the quercetin-treated mice, compared to the control. In addition, quercetin treated EAC tumor bearing mice had lower levels of liver enzymes, decreased hepatic inflammation and fibrosis compared with EAC tumor bearing mice. Docking study confirmed VDR-quercetin interaction. Furthermore, in-vitro assays and chick chorioallantoic membrane assay revealed the Vitamin D mimicking effect of quercetin. Significance Dietary flavonoid, quercetin could act as a promising therapeutic drug to suppress the breast cancer induced tumor angiogenesis, hepatic inflammation, and fibrosis possibly via activation of VDR.
Collapse
Affiliation(s)
- Nirmala G. Sannappa Gowda
- Department of Biochemistry, Center of Excellence in Molecular Biology and Regenerative Medicine, JSS Medical College, JSS Academy of Higher Education and Research, Mysore, India
| | - Varsha D. Shiragannavar
- Department of Biochemistry, Center of Excellence in Molecular Biology and Regenerative Medicine, JSS Medical College, JSS Academy of Higher Education and Research, Mysore, India
| | - Lakshana D. Puttahanumantharayappa
- Department of Biochemistry, Center of Excellence in Molecular Biology and Regenerative Medicine, JSS Medical College, JSS Academy of Higher Education and Research, Mysore, India
| | - Ashwini Tumkur Shivakumar
- Department of Conservative Dentistry and Endodontics, JSS Dental College and Hospital, Mysore, Karnataka, India
| | - Siva Dallavalasa
- Department of Biochemistry, Center of Excellence in Molecular Biology and Regenerative Medicine, JSS Medical College, JSS Academy of Higher Education and Research, Mysore, India
| | - Chaithanya G. Basavaraju
- Department of Biochemistry, Center of Excellence in Molecular Biology and Regenerative Medicine, JSS Medical College, JSS Academy of Higher Education and Research, Mysore, India
| | - Smitha S. Bhat
- Department of Biotechnology and Bioinformatics, JSS Academy of Higher Education and Research, Mysore, Karnataka, India
| | - Shashanka K. Prasad
- Department of Biotechnology and Bioinformatics, JSS Academy of Higher Education and Research, Mysore, Karnataka, India
- Bioactive Compound Laboratory, Faculty of Agriculture, Chiang Mai University, Chiang Mai, Thailand
| | | | - SubbaRao V. Madhunapantula
- Department of Biochemistry, Center of Excellence in Molecular Biology and Regenerative Medicine, JSS Medical College, JSS Academy of Higher Education and Research, Mysore, India
| | - Prasanna K. Santhekadur
- Department of Biochemistry, Center of Excellence in Molecular Biology and Regenerative Medicine, JSS Medical College, JSS Academy of Higher Education and Research, Mysore, India
- *Correspondence: Prasanna K. Santhekadur,
| |
Collapse
|
6
|
Elebiyo TC, Rotimi D, Evbuomwan IO, Maimako RF, Iyobhebhe M, Ojo OA, Oluba OM, Adeyemi OS. Reassessing vascular endothelial growth factor (VEGF) in anti-angiogenic cancer therapy. Cancer Treat Res Commun 2022; 32:100620. [PMID: 35964475 DOI: 10.1016/j.ctarc.2022.100620] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 06/02/2022] [Accepted: 08/05/2022] [Indexed: 05/23/2023]
Abstract
Vascularization is fundamental to the growth and spread of tumor cells to distant sites. As a consequence, angiogenesis, the sprouting of new blood vessels from existing ones, is a characteristic trait of cancer. In 1971, Judah Folkman postulated that tumour growth is angiogenesis dependent and that by cutting off blood supply, a neoplastic lesion could be potentially starved into remission. Decades of research have been devoted to understanding the role that vascular endothelial growth factor (VEGF) plays in tumor angiogenesis, and it has been identified as a significant pro-angiogenic factor that is frequently overexpressed within a tumor mass. Today, anti-VEGF drugs such as Sunitinib, Sorafenib, Axitinib, Tanibirumab, and Ramucirumab have been approved for the treatment of advanced and metastatic cancers. However, anti-angiogenic therapy has turned out to be more complex than originally thought. The failure of this therapeutic option calls for a reevaluation of VEGF as the major target in anti-angiogenic cancer therapy. The call for reassessment is based on two rationales: first, tumour blood vessels are abnormal, disorganized, and leaky; this not only prevents optimal drug delivery but it also promotes hypoxia and metastasis; secondly, tumour growth or regrowth might be blood vessel dependent and not angiogenesis dependent as tumour cells can acquire blood vessels via non-angiogenic mechanisms. Therefore, a critical assessment of VEGF, VEGFRs, and their inhibitors could glean newer options such as repurposing anti-VEGF drugs as vascular normalizing agents to enhance drug delivery of immune checkpoint inhibitors.
Collapse
Affiliation(s)
| | - Damilare Rotimi
- Department of Biochemistry, Landmark University, Omu-Aran, Nigeria
| | | | | | | | - Oluwafemi Adeleke Ojo
- Phytomedicine, Molecular Toxicology, and Computational Biochemistry Research Laboratory (PMTCB-RL), Department of Biochemistry, Bowen University, Iwo, 232101, Nigeria..
| | | | | |
Collapse
|
7
|
Alonso-Diez A, Affolter V, Sevane N, Dunner S, Valdivia G, Clemente M, De Andrés P, Illera J, Pérez-Alenza M, Peña L. Cell adhesion molecules E-cadherin and CADM1 are differently expressed in canine inflammatory mammary cancer. Res Vet Sci 2022; 152:307-313. [DOI: 10.1016/j.rvsc.2022.08.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 07/27/2022] [Accepted: 08/23/2022] [Indexed: 10/14/2022]
|
8
|
Decorin-mediated suppression of tumorigenesis, invasion, and metastasis in inflammatory breast cancer. Commun Biol 2021; 4:72. [PMID: 33452400 PMCID: PMC7811004 DOI: 10.1038/s42003-020-01590-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 12/11/2020] [Indexed: 12/15/2022] Open
Abstract
Inflammatory breast cancer (IBC) is a clinically distinct and highly aggressive form of breast cancer with rapid onset and a strong propensity to metastasize. The molecular mechanisms underlying the aggressiveness and metastatic propensity of IBC are largely unknown. Herein, we report that decorin (DCN), a small leucine-rich extracellular matrix proteoglycan, is downregulated in tumors from patients with IBC. Overexpression of DCN in IBC cells markedly decreased migration, invasion, and cancer stem cells in vitro and inhibited tumor growth and metastasis in IBC xenograft mouse models. Mechanistically, DCN functioned as a suppressor of invasion and tumor growth in IBC by destabilizing E-cadherin and inhibiting EGFR/ERK signaling. DCN physically binds E-cadherin in IBC cells and accelerates its degradation through an autophagy-linked lysosomal pathway. We established that DCN inhibits tumorigenesis and metastasis in IBC cells by negatively regulating the E-cadherin/EGFR/ERK axis. Our findings offer a potential therapeutic strategy for IBC, and provide a novel mechanism for IBC pathobiology. Xiaoding Hu et al. find that expression of the proteoglycan decorin is decreased in patients with inflammatory breast cancer compared to normal breast tissue and some other types of breast cancer. They demonstrate that decorin acts as a tumor suppressor in cancer cells and human xenograft mouse models by destabilizing the E-cadherin-EGFR signaling axis, and their findings suggest potential therapeutic strategies for this aggressive breast cancer.
Collapse
|
9
|
Shams A, Binothman N, Boudreault J, Wang N, Shams F, Hamam D, Tian J, Moamer A, Dai M, Lebrun JJ, Ali S. Prolactin receptor-driven combined luminal and epithelial differentiation in breast cancer restricts plasticity, stemness, tumorigenesis and metastasis. Oncogenesis 2021; 10:10. [PMID: 33446633 PMCID: PMC7809050 DOI: 10.1038/s41389-020-00297-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 12/09/2020] [Accepted: 12/11/2020] [Indexed: 02/06/2023] Open
Abstract
Dedifferentiation increased cellular plasticity and stemness are established derivers of tumor heterogeneity, metastasis and therapeutic failure resulting in incurable cancers. Therefore, it is essential to decipher pro/forward-differentiation mechanisms in cancer that may serve as therapeutic targets. We found that interfering with expression of the receptor for the lactogenic hormone prolactin (PRLR) in breast cancer cells representative of the luminal and epithelial breast cancer subtypes (hormone receptor positive (HR+) and HER2-enriched (HER2-E) resulted in loss of their differentiation state, enriched for stem-like cell subpopulations, and increased their tumorigenic capacity in a subtype-specific manner. Loss of PRLR expression in HR+ breast cancer cells caused their dedifferentiation generating a mesenchymal-basal-like phenotype enriched in CD44+ breast cancer stem-like cells (BCSCs) showing high tumorigenic and metastatic capacities and resistance to anti-hormonal therapy. Whereas loss of PRLR expression in HER2-E breast cancer cells resulted in loss of their luminal differentiation yet enriched for epithelial ALDH+ BCSC population showing elevated HER2-driven tumorigenic, multi-organ metastatic spread, and resistance to anti-HER2 therapy. Collectively, this study defines PRLR as a driver of precise luminal and epithelial differentiation limiting cellular plasticity, stemness, and tumorigenesis and emphasizing the function of pro/forward-differentiation pathways as a foundation for the discovery of anti-cancer therapeutic targets.
Collapse
Affiliation(s)
- Anwar Shams
- grid.63984.300000 0000 9064 4811Department of Medicine, Cancer Research Program, The Research Institute of the McGill University Health Centre, Montreal, QC Canada ,grid.412895.30000 0004 0419 5255Present Address: Department of Pharmacology, Faculty of Medicine, Taif University, Taif, Saudi Arabia
| | - Najat Binothman
- grid.63984.300000 0000 9064 4811Department of Medicine, Cancer Research Program, The Research Institute of the McGill University Health Centre, Montreal, QC Canada ,grid.412125.10000 0001 0619 1117Present Address: Department of Chemistry, College of Science and Arts, King Abdulaziz University, P.O. Box 344, Rabigh, 21911 Saudi Arabia
| | - Julien Boudreault
- grid.63984.300000 0000 9064 4811Department of Medicine, Cancer Research Program, The Research Institute of the McGill University Health Centre, Montreal, QC Canada
| | - Ni Wang
- grid.63984.300000 0000 9064 4811Department of Medicine, Cancer Research Program, The Research Institute of the McGill University Health Centre, Montreal, QC Canada
| | - Fuad Shams
- grid.415252.5Department of Pathology and Laboratory Medicine, King Abdulaziz Hospital, Mecca, Saudi Arabia
| | - Dana Hamam
- grid.63984.300000 0000 9064 4811Department of Medicine, Cancer Research Program, The Research Institute of the McGill University Health Centre, Montreal, QC Canada
| | - Jun Tian
- grid.63984.300000 0000 9064 4811Department of Medicine, Cancer Research Program, The Research Institute of the McGill University Health Centre, Montreal, QC Canada
| | - Alaa Moamer
- grid.63984.300000 0000 9064 4811Department of Medicine, Cancer Research Program, The Research Institute of the McGill University Health Centre, Montreal, QC Canada
| | - Meiou Dai
- grid.63984.300000 0000 9064 4811Department of Medicine, Cancer Research Program, The Research Institute of the McGill University Health Centre, Montreal, QC Canada
| | - Jean-Jacques Lebrun
- grid.63984.300000 0000 9064 4811Department of Medicine, Cancer Research Program, The Research Institute of the McGill University Health Centre, Montreal, QC Canada
| | - Suhad Ali
- grid.63984.300000 0000 9064 4811Department of Medicine, Cancer Research Program, The Research Institute of the McGill University Health Centre, Montreal, QC Canada
| |
Collapse
|
10
|
Bertucci A, Bertucci F, Zemmour C, Lerebours F, Pierga JY, Levy C, Dalenc F, Grenier J, Petit T, Berline M, Gonçalves A. PELICAN-IPC 2015-016/Oncodistinct-003: A Prospective, Multicenter, Open-Label, Randomized, Non-Comparative, Phase II Study of Pembrolizumab in Combination With Neo Adjuvant EC-Paclitaxel Regimen in HER2-Negative Inflammatory Breast Cancer. Front Oncol 2020; 10:575978. [PMID: 33330051 PMCID: PMC7732675 DOI: 10.3389/fonc.2020.575978] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 10/28/2020] [Indexed: 12/31/2022] Open
Abstract
Inflammatory breast cancer (IBC) is a highly aggressive entity with a poor outcome and relative resistance to treatment. Despite progresses achieved during the last decades, the survival remains significantly lower than non-IBC. Recent clinical trials assessing PD-1/PD-L1 inhibitors showed promising results in non-IBC. Pembrolizumab, an anti-PD-1 monoclonal antibody, revolutionized the treatment of different cancers. Several recent studies suggested a potential interest of targeting the immune system in IBC by revealing a more frequent PD-L1 expression and an enriched immune microenvironment when compared with non-IBC. Here, we describe the rationale and design of PELICAN-IPC 2015-016/Oncodistinct-003 trial, an open-label, randomized, non-comparative, phase II study assessing efficacy, and safety of pembrolizumab in combination with anthracycline-containing neoadjuvant chemotherapy in HER2-negative IBC. The trial is ongoing. The primary endpoint is the pCR rate (ypT0/Tis, ypN0) in overall population and the co-primary endpoint is safety profile during a run-in phase. Key secondary objectives include tolerability, invasive disease-free, event-free and overall survivals, as well as collection of tumor and blood samples for translational research. CLINICAL TRIAL REGISTRATION https://clinicaltrials.gov/ (NCT03515798).
Collapse
Affiliation(s)
- Alexandre Bertucci
- Département d’Oncologie Médicale, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM UMR1068, CNRS UMR7258, Aix-Marseille Université, Marseille, France
| | - François Bertucci
- Département d’Oncologie Médicale, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM UMR1068, CNRS UMR7258, Aix-Marseille Université, Marseille, France
- Laboratoire d’Oncologie Prédictive, CRCM, Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR7258, Aix-Marseille Université, Marseille, France
- Aix-Marseille Université, Faculté de Médecine, Marseille, France
| | - Christophe Zemmour
- Laboratoire d’Oncologie Prédictive, CRCM, Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR7258, Aix-Marseille Université, Marseille, France
| | | | - Jean-Yves Pierga
- Département d’Oncologie Médicale, Institut Curie, Saint-Cloud, France
- Département d’Oncologie Médicale, Institut Curie, Université de Paris, Paris, France
| | - Christelle Levy
- Breast Cancer Unit, François Baclesse Cancer Centre, Caen, France
| | - Florence Dalenc
- Département d’Oncologie Médicale, Institut Claudius Regaud, Institut Universitaire du Cancer, Oncopole Toulouse, France
| | - Julien Grenier
- Département d’Oncologie Médicale, Institut Sainte Catherine, Avignon, France
| | - Thierry Petit
- Département d’Oncologie Médicale, Centre Paul-Strauss, Strasbourg, France
| | - Marguerite Berline
- Laboratoire d’Oncologie Prédictive, CRCM, Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR7258, Aix-Marseille Université, Marseille, France
| | - Anthony Gonçalves
- Département d’Oncologie Médicale, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM UMR1068, CNRS UMR7258, Aix-Marseille Université, Marseille, France
- Laboratoire d’Oncologie Prédictive, CRCM, Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR7258, Aix-Marseille Université, Marseille, France
- Aix-Marseille Université, Faculté de Médecine, Marseille, France
- Oncodistinct Network, Bruxelles, Belgium
| |
Collapse
|
11
|
Bakir B, Chiarella AM, Pitarresi JR, Rustgi AK. EMT, MET, Plasticity, and Tumor Metastasis. Trends Cell Biol 2020; 30:764-776. [PMID: 32800658 PMCID: PMC7647095 DOI: 10.1016/j.tcb.2020.07.003] [Citation(s) in RCA: 544] [Impact Index Per Article: 136.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 07/02/2020] [Accepted: 07/10/2020] [Indexed: 01/06/2023]
Abstract
Cancer cell identity and plasticity are required in transition states, such as epithelial-mesenchymal transition (EMT) and mesenchymal-epithelial transition (MET), in primary tumor initiation, progression, and metastasis. The functional roles of EMT, MET, and the partial state (referred to as pEMT) may vary based on the type of tumor, the state of dissemination, and the degree of metastatic colonization. Herein, we review EMT, MET, pEMT, and plasticity in the context of tumor metastasis.
Collapse
Affiliation(s)
- Basil Bakir
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Anna M Chiarella
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10032, USA
| | - Jason R Pitarresi
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Anil K Rustgi
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
12
|
Gadde M, Phillips C, Ghousifam N, Sorace AG, Wong E, Krishnamurthy S, Syed A, Rahal O, Yankeelov TE, Woodward WA, Rylander MN. In vitro vascularized tumor platform for modeling tumor-vasculature interactions of inflammatory breast cancer. Biotechnol Bioeng 2020; 117:3572-3590. [PMID: 32648934 DOI: 10.1002/bit.27487] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 06/24/2020] [Accepted: 07/08/2020] [Indexed: 12/26/2022]
Abstract
Inflammatory breast cancer (IBC), a rare form of breast cancer associated with increased angiogenesis and metastasis, is largely driven by tumor-stromal interactions with the vasculature and the extracellular matrix (ECM). However, there is currently a lack of understanding of the role these interactions play in initiation and progression of the disease. In this study, we developed the first three-dimensional, in vitro, vascularized, microfluidic IBC platform to quantify the spatial and temporal dynamics of tumor-vasculature and tumor-ECM interactions specific to IBC. Platforms consisting of collagen type 1 ECM with an endothelialized blood vessel were cultured with IBC cells, MDA-IBC3 (HER2+) or SUM149 (triple negative), and for comparison to non-IBC cells, MDA-MB-231 (triple negative). Acellular collagen platforms with endothelialized blood vessels served as controls. SUM149 and MDA-MB-231 platforms exhibited a significantly (p < .05) higher vessel permeability and decreased endothelial coverage of the vessel lumen compared to the control. Both IBC platforms, MDA-IBC3 and SUM149, expressed higher levels of vascular endothelial growth factor (p < .05) and increased collagen ECM porosity compared to non-IBCMDA-MB-231 (p < .05) and control (p < .01) platforms. Additionally, unique to the MDA-IBC3 platform, we observed progressive sprouting of the endothelium over time resulting in viable vessels with lumen. The newly sprouted vessels encircled clusters of MDA-IBC3 cells replicating a key feature of in vivo IBC. The IBC in vitro vascularized platforms introduced in this study model well-described in vivo and clinical IBC phenotypes and provide an adaptable, high throughput tool for systematically and quantitatively investigating tumor-stromal mechanisms and dynamics of tumor progression.
Collapse
Affiliation(s)
- Manasa Gadde
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas
| | - Caleb Phillips
- Oden Institute for Computational and Engineering Sciences, The University of Texas at Austin, Austin, Texas
| | - Neda Ghousifam
- Department of Mechanical Engineering, The University of Texas at Austin, Austin, Texas
| | - Anna G Sorace
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, Alabama.,Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, Alabama.,O'Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, Alabama
| | - Enoch Wong
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas
| | - Savitri Krishnamurthy
- Department of Pathology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Anum Syed
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas
| | - Omar Rahal
- M.D. Anderson Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Experimental Radiation Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Thomas E Yankeelov
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas.,Oden Institute for Computational and Engineering Sciences, The University of Texas at Austin, Austin, Texas.,Departments of Diagnostic Medicine, The University of Texas at Austin, Austin, Texas.,Department of Oncology, The University of Texas at Austin, Austin, Texas.,Livestrong Cancer Institutes, The University of Texas at Austin, Austin, Texas
| | - Wendy A Woodward
- M.D. Anderson Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Experimental Radiation Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Marissa N Rylander
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas.,Oden Institute for Computational and Engineering Sciences, The University of Texas at Austin, Austin, Texas.,Department of Mechanical Engineering, The University of Texas at Austin, Austin, Texas
| |
Collapse
|
13
|
Inflammatory Breast Cancer: Diagnostic, Molecular and Therapeutic Considerations. CURRENT BREAST CANCER REPORTS 2019. [DOI: 10.1007/s12609-019-00337-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
14
|
Mizukoshi K, Okazawa Y, Haeno H, Koyama Y, Sulidan K, Komiyama H, Saeki H, Ohtsuji N, Ito Y, Kojima Y, Goto M, Habu S, Hino O, Sakamoto K, Orimo A. Metastatic seeding of human colon cancer cell clusters expressing the hybrid epithelial/mesenchymal state. Int J Cancer 2019; 146:2547-2562. [PMID: 31506938 DOI: 10.1002/ijc.32672] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 08/05/2019] [Accepted: 08/26/2019] [Indexed: 12/15/2022]
Abstract
Emerging evidence supports the theory that tumor cell clusters efficiently metastasize to distant organs. However, the roles of epithelial-to-mesenchymal transition (EMT) in metastasizing tumor cell clusters have not yet been fully elucidated. To investigate this issue, tumor fragments were dissected from 40 colorectal cancer (CRC) patients and implanted subcutaneously into immunodeficient mice. We observed that tumors developed from the tumor fragments obtained from 28 of the 40 CRC patients. The tumors were then dissociated into cell suspensions to be orthotopically injected into secondary mice. The tumors from 13 of the 28 patients progressed. Furthermore, metastases formed spontaneously in the liver and lungs from the tumor fragments obtained from 8 of these 13 patients. Moreover, employing a mathematical analysis, we showed that tumor cell clusters seeded these metastases significantly more often than did single tumor cells. Membrane E-cadherin- and nuclear ZEB1-positive tumor cells indicating the hybrid epithelial/mesenchymal state were also detected in primary tumors of various CRC patients, and in the corresponding patient-derived xenografts (PDXs) and circulating tumor cell clusters in the bloodstreams of mice. In contrast, ZEB1 staining was barely detectable in the patient-matched liver metastases presumably developing through mesenchymal-to-epithelial transition. Inhibition of E-cadherin or ZEB1 expression by shRNA notably prevented the PDX-derived tumor organoids from colonizing the liver, when injected intrasplenically into mice, indicating E-cadherin and ZEB1 expressions to be required for their metastatic colonization. Taken together, these findings suggest that the epithelial/mesenchymal state mediates metastatic seeding of human CRC cell clusters into distant organs.
Collapse
Affiliation(s)
- Kosuke Mizukoshi
- Department of Coloproctological Surgery, Juntendo University Faculty of Medicine, Tokyo, Japan.,Department of Molecular Pathogenesis, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Yu Okazawa
- Department of Coloproctological Surgery, Juntendo University Faculty of Medicine, Tokyo, Japan.,Department of Molecular Pathogenesis, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Hiroshi Haeno
- Division of Translational Genomics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Chiba, Japan.,Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Yu Koyama
- Department of Molecular Pathogenesis, Juntendo University Faculty of Medicine, Tokyo, Japan.,Department of Oral Pathobiological Science and Surgery, Tokyo Dental College, Tokyo, Japan
| | - Kaidiliayi Sulidan
- Department of Molecular Pathogenesis, Juntendo University Faculty of Medicine, Tokyo, Japan.,Department of Obstetrics and Gynecology, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Hiromitsu Komiyama
- Department of Coloproctological Surgery, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Harumi Saeki
- Department of Molecular Pathogenesis, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Naomi Ohtsuji
- Department of Molecular Pathogenesis, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Yasuhiko Ito
- Department of Molecular Pathogenesis, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Yutaka Kojima
- Department of Coloproctological Surgery, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Michitoshi Goto
- Department of Coloproctological Surgery, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Sonoko Habu
- Atopy Research Center, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Okio Hino
- Department of Molecular Pathogenesis, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Kazuhiro Sakamoto
- Department of Coloproctological Surgery, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Akira Orimo
- Department of Molecular Pathogenesis, Juntendo University Faculty of Medicine, Tokyo, Japan
| |
Collapse
|
15
|
Di Bonito M, Cantile M, Botti G. Pathological and molecular characteristics of inflammatory breast cancer. Transl Cancer Res 2019; 8:S449-S456. [PMID: 35117122 PMCID: PMC8798351 DOI: 10.21037/tcr.2019.03.24] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 03/18/2019] [Indexed: 11/11/2022]
Abstract
Inflammatory breast cancer (IBC) is a rare and aggressive form of breast cancer characterized by the presence of many dermal tumor emboli in the papillary and reticular dermis of the skin overlying the breast. IBC patients, compared to other breast cancer patients, have more frequently metastatic axillary lymph nodes. IBC is often high grade, negative for hormone receptors and presents with amplification of the HER2 gene. Invasive IBC is frequently of ductal phenotype, even if a specific histological distinction for these lesions has not been described. The pathogenesis and evolution of IBC are strongly dependent upon tumor microenvironment, characterized by several macrophages/monocytes and lymphocytes. The tumor and microenvironment cells are well molecularly characterized, showing the main contributor of inflammatory pathways in tumor biology of IBC. In addition, several molecular alterations are described in this tumor, such as mutations of ERBB2, KRAS, BRAF, EGFR, PIK3CA, PTEN, AKT1, and AKT3 genes that could suggest a therapeutic stratification of IBC patients with the combination of different biological target therapies.
Collapse
Affiliation(s)
- Maurizio Di Bonito
- Pathology Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| | - Monica Cantile
- Pathology Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| | - Gerardo Botti
- Scientific Direction, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| |
Collapse
|
16
|
Barreno L, Cáceres S, Alonso-Diez Á, Vicente-Montaña A, García ML, Clemente M, Illera JC, Peña L. Vasculogenic mimicry-associated ultrastructural findings in human and canine inflammatory breast cancer cell lines. BMC Cancer 2019; 19:750. [PMID: 31362745 PMCID: PMC6668131 DOI: 10.1186/s12885-019-5955-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 07/18/2019] [Indexed: 12/29/2022] Open
Abstract
Background Human inflammatory breast cancer (IBC) and canine inflammatory mammary cancer (IMC) are the most lethal mammary cancers. An exacerbated angiogenesis and the existence of vasculogenic mimicry (VM) are hallmarks of these tumors. The information regarding VM and ultrastructural characteristics of mammary cell lines is scant. Methods In this study, IBC cell line SUM149 and IMC cell line IPC-366 in adherent (2D) and non-adherent (3D) (mammospheres, cancer stem cells) conditions were analyzed by transmission and scanning electron microscopy (TEM and SEM, respectively). Results The TEM revealed round to oval shape cells with microvilli on the surface, high numbers of peroxisomes in close apposition to lipid droplets and some extracellular derived vesicles. The TEM and the SEM mammospheres revealed group of cells clumping together with a central lumen (resembling a mammary acini). The cells joint are tight junctions and zonula adherens. By SEM two cell morphologies were observed: spherical and flattened cells. There was evidence endothelial-like cells (ELCs), which is characteristic for this disease, showing several or unique cytoplasmic empty space. ELCs were more frequent in 3D than in 2D culture conditions and contained Weibel-Palade cytoplasmic bodies, which are exclusive structures of endothelial cells. Conclusions Both cell lines, IPC-366 and SUM-149, shared ultrastructural characteristics, further supporting canine IMC as a model for the human disease. To the best of our knowledge, this is the first study that demonstrate the morphological differentiation of cultured cancer stem cells from cancer epithelial cell lines into endothelial-like cells, confirming the vasculogenic mimicry phenomenon from an ultrastructural point of view.
Collapse
Affiliation(s)
- Lucía Barreno
- Veterinary Clinical Hospital, Pathology Service, Complutense University of Madrid, Madrid, Spain
| | - Sara Cáceres
- Department of animal Physiology, Complutense University of Madrid, Madrid, Spain
| | - Ángela Alonso-Diez
- Veterinary Clinical Hospital, Pathology Service, Complutense University of Madrid, Madrid, Spain
| | - Ana Vicente-Montaña
- National Center of Electron Microscopy, Complutense University of Madrid, Madrid, Spain
| | - María Luisa García
- National Center of Electron Microscopy, Complutense University of Madrid, Madrid, Spain
| | - Mónica Clemente
- Veterinary Clinical Hospital, Pathology Service, Complutense University of Madrid, Madrid, Spain
| | - Juan Carlos Illera
- Department of animal Physiology, Complutense University of Madrid, Madrid, Spain
| | - Laura Peña
- Veterinary Clinical Hospital, Pathology Service, Complutense University of Madrid, Madrid, Spain.
| |
Collapse
|
17
|
Velatooru LR, Vakamullu S, Penugurti V, S PR. Alpinoid c analog inhibits angiogenesis and induces apoptosis in COLO205 cell line. Chem Biol Interact 2019; 308:1-10. [PMID: 31071337 DOI: 10.1016/j.cbi.2019.05.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 04/26/2019] [Accepted: 05/05/2019] [Indexed: 01/05/2023]
Abstract
Diarylheptanoids display an array of biological and pharmacological properties. We previously reported the synthesis of a diarylheptanoid Alpinoid c and a series of its derivatives, evaluated their cytotoxicity against various human cancer cells. We found some of these derivatives were significantly more potent than Alpinoid c in preventing the proliferation of various cancer cell lines. Among these, (S, E)-1-(3, 4 dimethoxyphenyl)-6-hydroxy-7-phenylhept-4-en-3-one (DPHP) showed most potent cytotoxicity against COLO205 cells, however, the mechanism by which DPHP prevents the growth of these colon cancer cells remains unknown. In the current study, we investigated the molecular mechanism of DPHP on colon cancer cells. DPHP inhibited the proliferation of COLO205 (IC50 7.01 ± 0.62 μM) and A549 (IC50 20.03 ± 3.11 μM) cells more specifically than normal human colon epithelial cell line NCM460 (IC50 55.6 ± 4.02 μM). In COLO205 cells, DPHP induced cell shrinkage, membrane blebbing, chromatin condensation, phosphatidylserine externalization, and an accumulation of cells at sub-G1 phase. Further analysis these cells treated with DPHP revealed a decrease in mitochondrial membrane potential, an increase in Bax/Bcl2 ratio, the release of cytochrome c, activation of caspases -9, -3/7, and cleavage of the poly-ADP-ribose polymerase. DPHP treatment resulted in inhibition of hypoxia induced VEGF downstream signaling pathway in COLO205 cells is concurrent with inhibition of angiogenesis in CAM. Based on these data we suggest that DPHP significantly induced apoptosis possibly via intrinsic mitochondrial apoptosis pathway and inhibited angiogenesis. Our study suggests DPHP could be a therapeutic agent in treating colon cancer.
Collapse
Affiliation(s)
- Loka Reddy Velatooru
- Toxicology Unit, Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500607, Telangana, India; Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, 500046, Telangana, India.
| | - Sridhar Vakamullu
- Toxicology Unit, Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500607, Telangana, India
| | - Vasudevarao Penugurti
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, 500046, Telangana, India
| | - Purushotham Reddy S
- Division of Natural Product, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500607, Telangana, India
| |
Collapse
|
18
|
Provance OK, Lewis-Wambi J. Deciphering the role of interferon alpha signaling and microenvironment crosstalk in inflammatory breast cancer. Breast Cancer Res 2019; 21:59. [PMID: 31060575 PMCID: PMC6501286 DOI: 10.1186/s13058-019-1140-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Inflammatory breast cancer (IBC) is the most rare and aggressive subtype of breast cancer characterized by clusters of tumor cells invading lymph vessels, high rates of metastasis, and resistance to systemic chemotherapy. While significant progress has been made in understanding IBC, survival among IBC patients is still only one half that among patients with non-IBC. A major limitation to the development of more specific and effective treatments for IBC is a lack of identifiable molecular alterations that are specific to IBC. Emerging evidence suggests that the aggressive nature of IBC is not specific to IBC cells but instead driven by the interplay between autonomous signaling and context-dependent cytokine networks from the surrounding tumor microenvironment. Recently, the type I interferon, specifically the interferon alpha signature, has been identified as a pathway upregulated in IBC but few studies have addressed its role. Activation of the interferon alpha signaling pathway has been shown to contribute to apoptosis and cellular senescence but is also attributed to increased migration and drug resistance depending on the interferon-stimulated genes transcribed. The mechanisms promoting the increase in interferon alpha expression and the role interferon alpha plays in IBC remain speculative. Current hypotheses suggest that immune and stromal cells in the local tumor microenvironment contribute to the interferon alpha signaling cascade within the tumor cell and that this activation may further alter the immune and stromal cells within the microenvironment. This review serves as an overview of the role of interferon alpha signaling in IBC. Ideally, future experiments should investigate the mechanistic interplay of interferons in IBC to develop more efficacious treatment strategies for IBC patients.
Collapse
Affiliation(s)
- Olivia K Provance
- Department of Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Wahl Hall East 1031, Kansas City, KS, 66160, USA
| | - Joan Lewis-Wambi
- Department of Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Wahl Hall East 1031, Kansas City, KS, 66160, USA. .,The University of Kansas Cancer Center, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA.
| |
Collapse
|
19
|
Jolly MK, Somarelli JA, Sheth M, Biddle A, Tripathi SC, Armstrong AJ, Hanash SM, Bapat SA, Rangarajan A, Levine H. Hybrid epithelial/mesenchymal phenotypes promote metastasis and therapy resistance across carcinomas. Pharmacol Ther 2018; 194:161-184. [PMID: 30268772 DOI: 10.1016/j.pharmthera.2018.09.007] [Citation(s) in RCA: 202] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Cancer metastasis and therapy resistance are the major unsolved clinical challenges, and account for nearly all cancer-related deaths. Both metastasis and therapy resistance are fueled by epithelial plasticity, the reversible phenotypic transitions between epithelial and mesenchymal phenotypes, including epithelial-mesenchymal transition (EMT) and mesenchymal-epithelial transition (MET). EMT and MET have been largely considered as binary processes, where cells detach from the primary tumor as individual units with many, if not all, traits of a mesenchymal cell (EMT) and then convert back to being epithelial (MET). However, recent studies have demonstrated that cells can metastasize in ways alternative to traditional EMT paradigm; for example, they can detach as clusters, and/or occupy one or more stable hybrid epithelial/mesenchymal (E/M) phenotypes that can be the end point of a transition. Such hybrid E/M cells can integrate various epithelial and mesenchymal traits and markers, facilitating collective cell migration. Furthermore, these hybrid E/M cells may possess higher tumor-initiation and metastatic potential as compared to cells on either end of the EMT spectrum. Here, we review in silico, in vitro, in vivo and clinical evidence for the existence of one or more hybrid E/M phenotype(s) in multiple carcinomas, and discuss their implications in tumor-initiation, tumor relapse, therapy resistance, and metastasis. Together, these studies drive the emerging notion that cells in a hybrid E/M phenotype may occupy 'metastatic sweet spot' in multiple subtypes of carcinomas, and pathways linked to this (these) hybrid E/M state(s) may be relevant as prognostic biomarkers as well as a promising therapeutic targets.
Collapse
Affiliation(s)
- Mohit Kumar Jolly
- Center for Theoretical Biological Physics, Rice University, Houston, TX, USA.
| | - Jason A Somarelli
- Duke Cancer Institute and Department of Medicine, Duke University Medical Center, Durham, USA
| | - Maya Sheth
- Duke Cancer Institute and Department of Medicine, Duke University Medical Center, Durham, USA
| | - Adrian Biddle
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Satyendra C Tripathi
- Department of Clinical Cancer Prevention, UT MD Anderson Cancer Center, Houston, USA
| | - Andrew J Armstrong
- Duke Cancer Institute and Department of Medicine, Duke University Medical Center, Durham, USA
| | - Samir M Hanash
- Department of Clinical Cancer Prevention, UT MD Anderson Cancer Center, Houston, USA
| | - Sharmila A Bapat
- National Center for Cell Science, Savitribai Phule Pune University Campus, Ganeshkhind, Pune, India
| | - Annapoorni Rangarajan
- Department of Molecular Reproduction, Development & Genetics, Indian Institute of Science, Bangalore, India
| | - Herbert Levine
- Center for Theoretical Biological Physics, Rice University, Houston, TX, USA.
| |
Collapse
|
20
|
Cserni G, Charafe-Jauffret E, van Diest P. Inflammatory breast cancer: The pathologists' perspective. Eur J Surg Oncol 2018; 44:1128-1134. [DOI: 10.1016/j.ejso.2018.04.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 03/30/2018] [Accepted: 04/05/2018] [Indexed: 12/20/2022] Open
|
21
|
Lim B, Woodward WA, Wang X, Reuben JM, Ueno NT. Inflammatory breast cancer biology: the tumour microenvironment is key. Nat Rev Cancer 2018; 18:485-499. [PMID: 29703913 DOI: 10.1038/s41568-018-0010-y] [Citation(s) in RCA: 209] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Inflammatory breast cancer (IBC) is a rare and aggressive disease that accounts for ~2-4% of all breast cancers. However, despite its low incidence rate, IBC is responsible for 7-10% of breast cancer-related mortality in Western countries. Thus, the discovery of robust biological targets and the development of more effective therapeutics in IBC are crucial. Despite major international efforts to understand IBC biology, genomic studies have not led to the discovery of distinct biological mechanisms in IBC that can be translated into novel therapeutic strategies. In this Review, we discuss these molecular profiling efforts and highlight other important aspects of IBC biology. We present the intrinsic characteristics of IBC, including stemness, metastatic potential and hormone receptor positivity; the extrinsic features of the IBC tumour microenvironment (TME), including various constituent cell types; and lastly, the communication between these intrinsic and extrinsic components. We summarize the latest perspectives on the key biological features of IBC, with particular emphasis on the TME as an important contributor to the aggressive nature of IBC. On the basis of the current understanding of IBC, we hope to develop the next generation of translational studies, which will lead to much-needed survival improvements in patients with this deadly disease.
Collapse
Affiliation(s)
- Bora Lim
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Wendy A Woodward
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xiaoping Wang
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - James M Reuben
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Naoto T Ueno
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
22
|
Jeong J, Naab TJ, Fernandez AI, Ongkeko MS, Makambi KH, Blancato JK. Homeoprotein DLX4 expression is increased in inflammatory breast cancer cases from an urban African-American population. Oncotarget 2018; 9:31253-31263. [PMID: 30131852 PMCID: PMC6101289 DOI: 10.18632/oncotarget.25790] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 07/05/2018] [Indexed: 02/07/2023] Open
Abstract
Protein expression of Distal-less homeobox 4 (DLX4) was analyzed in inflammatory breast cancer (IBC) cases from an African-American (AA) population to determine if a) DLX4 gene over expression exists in this cohort and b) if the overexpression is associated with breast cancer clinicopathological characteristics (ER, PR, HER2, triple-negative). Twenty-nine blocks of formalin-fixed paraffin-embedded (FFPE) tissue from well-characterized human IBC cases were used for immunohistochemical staining (IHC). IHC results were assigned an intensity and percentage score. Percentage scores were assigned as 0, 1, 2, 3, or 4 and intensity scores were assigned 0, 1+, 2+ or 3+. For the analysis of the IHC, a percentage score of 3 or 4 and an intensity score of 2+ or 3+ were categorized as high. Chi-square or Fisher's exact tests were used to compare the high and low groups. In this cohort, 89.7% (26 out of 29) of IBC cases showed high percentages of positive cells staining for the DLX4 protein, while 40.0% (12 out of 30) of normal breast tissue from reduction mammoplasty cases demonstrated DLX4 expression (p < 0.01). In IBC patients, 65.5% of cases showed a high level of staining intensity, compared to 20.0% of normal breast tissues (test, p = 0.001). Intensity to DLX4 was higher in the HER2 negative status (78.3%) than the HER2 positive status (16.7%) (test, p = 0.011). DLX4 expression is higher in the IBC cases in this study of an urban AA population than in normal breast tissue cases. HER2 negative status is positively associated with high intensity of DLX4.
Collapse
Affiliation(s)
- Jaehong Jeong
- Department of Oncology, Georgetown University Medical Center, Washington DC 20057, USA
- Comprehensive and Integrative Medicine Institute, Daegu 42473, South Korea
| | - Tammey J. Naab
- Department of Pathology, Howard University Hospital, Washington DC 20059, USA
| | - Aileen I. Fernandez
- Department of Oncology, Georgetown University Medical Center, Washington DC 20057, USA
| | - Martin S. Ongkeko
- Department of Pathology, Georgetown University Medical Center, Washington DC 20057, USA
| | - Kepher H. Makambi
- Department of Biostatistics, Bioinformatics, and Biomathematics, Georgetown University, Washington DC 20057, USA
| | - Jan K. Blancato
- Department of Oncology, Georgetown University Medical Center, Washington DC 20057, USA
| |
Collapse
|
23
|
Tripathi S, Jolly MK, Woodward WA, Levine H, Deem MW. Analysis of Hierarchical Organization in Gene Expression Networks Reveals Underlying Principles of Collective Tumor Cell Dissemination and Metastatic Aggressiveness of Inflammatory Breast Cancer. Front Oncol 2018; 8:244. [PMID: 30023340 PMCID: PMC6039554 DOI: 10.3389/fonc.2018.00244] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 06/18/2018] [Indexed: 01/06/2023] Open
Abstract
Clusters of circulating tumor cells (CTCs), despite being rare, may account for more than 90% of metastases. Cells in these clusters do not undergo a complete epithelial-to-mesenchymal transition (EMT), but retain some epithelial traits as compared to individually disseminating tumor cells. Determinants of single cell dissemination versus collective dissemination remain elusive. Inflammatory breast cancer (IBC), a highly aggressive breast cancer subtype that chiefly metastasizes via CTC clusters, is a promising model for studying mechanisms of collective tumor cell dissemination. Previous studies, motivated by a theory that suggests physical systems with hierarchical organization tend to be more adaptable, have found that the expression of metastasis-associated genes is more hierarchically organized in cases of successful metastases. Here, we used the cophenetic correlation coefficient (CCC) to quantify the hierarchical organization in the expression of two distinct gene sets, collective dissemination-associated genes and IBC-associated genes, in cancer cell lines and in tumor samples from breast cancer patients. Hypothesizing that a higher CCC for collective dissemination-associated genes and for IBC-associated genes would be associated with retention of epithelial traits enabling collective dissemination and with worse disease progression in breast cancer patients, we evaluated the correlation of CCC with different phenotypic groups. The CCC of both the abovementioned gene sets, the collective dissemination-associated genes and the IBC-associated genes, was higher in (a) epithelial cell lines as compared to mesenchymal cell lines and (b) tumor samples from IBC patients as compared to samples from non-IBC breast cancer patients. A higher CCC of both gene sets was also correlated with a higher rate of metastatic relapse in breast cancer patients. In contrast, neither the levels of CDH1 gene expression nor gene set enrichment analysis (GSEA) of the abovementioned gene sets could provide similar insights. These results suggest that retention of some epithelial traits in disseminating tumor cells as IBC progresses promotes successful breast cancer metastasis. The CCC provides additional information regarding the organizational complexity of gene expression in comparison to GSEA. We have shown that the CCC may be a useful metric for investigating the collective dissemination phenotype and a prognostic factor for IBC.
Collapse
Affiliation(s)
- Shubham Tripathi
- PhD Program in Systems, Synthetic, and Physical Biology, Rice University, Houston, TX, United States
- Center for Theoretical Biological Physics, Rice University, Houston, TX, United States
| | - Mohit Kumar Jolly
- Center for Theoretical Biological Physics, Rice University, Houston, TX, United States
| | - Wendy A. Woodward
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- MD Anderson Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Herbert Levine
- PhD Program in Systems, Synthetic, and Physical Biology, Rice University, Houston, TX, United States
- Center for Theoretical Biological Physics, Rice University, Houston, TX, United States
- Department of Bioengineering, Rice University, Houston, TX, United States
- Department of Physics and Astronomy, Rice University, Houston, TX, United States
| | - Michael W. Deem
- PhD Program in Systems, Synthetic, and Physical Biology, Rice University, Houston, TX, United States
- Center for Theoretical Biological Physics, Rice University, Houston, TX, United States
- Department of Bioengineering, Rice University, Houston, TX, United States
- Department of Physics and Astronomy, Rice University, Houston, TX, United States
| |
Collapse
|
24
|
Kai K, Iwamoto T, Zhang D, Shen L, Takahashi Y, Rao A, Thompson A, Sen S, Ueno NT. CSF-1/CSF-1R axis is associated with epithelial/mesenchymal hybrid phenotype in epithelial-like inflammatory breast cancer. Sci Rep 2018; 8:9427. [PMID: 29930294 PMCID: PMC6013474 DOI: 10.1038/s41598-018-27409-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 05/18/2018] [Indexed: 12/14/2022] Open
Abstract
Inflammatory breast cancer (IBC) is a rare subtype of breast cancer, accounting for 8–10% of breast cancer-associated deaths in the US. Clinical hallmarks of IBC include tumor emboli in lymphatic vessels and E-cadherin overexpression, which supports a type of metastasis referred to as cell cluster-based metastasis, prevalent in IBC. In contrast, we previously reported epithelial-to-mesenchymal transition (EMT)-based progression of IBC, utilizing in vivo xenografts and in vitro Matrigel culture models. To address these two contradictory concepts of IBC metastasis, we used Matrigel culture to induce EMT in a panel of IBC cells. Results revealed Matrigel culture induced vimentin expression in SUM149 and SUM190 IBC cells at the transcriptional and protein levels while maintaining the expression of E-cadherin, a phenomenon referred to as partial EMT. Transcriptional profiling revealed that expression of colony-stimulating factor 1 (CSF-1) was induced in Matrigel culture. When the receptor tyrosine kinase of CSF-1 (CSF-1R) was inhibited by CSF-1R inhibitor BLZ945, the partial EMT was reversed in a dose-dependent manner, indicating that the CSF-1/CSF-1R axis plays a key role in controlling partial EMT. This observation may help reconcile the two contradictory theories of IBC metastasis, EMT vs cell cluster-based metastasis.
Collapse
Affiliation(s)
- Kazuharu Kai
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Takayuki Iwamoto
- Department of Breast and Endocrine Surgery, Okayama University Hospital, Okayama, Japan
| | - Dongwei Zhang
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Li Shen
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yuko Takahashi
- Department of Breast and Endocrine Surgery, Okayama University Hospital, Okayama, Japan
| | - Arvind Rao
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Alastair Thompson
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Subrata Sen
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
| | - Naoto T Ueno
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA. .,Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
| |
Collapse
|
25
|
Curigliano G. Inflammatory breast cancer and chest wall disease: The oncologist perspective. Eur J Surg Oncol 2018; 44:1142-1147. [PMID: 30032791 DOI: 10.1016/j.ejso.2018.05.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 05/11/2018] [Indexed: 12/13/2022] Open
Abstract
Chest wall inflammatory and lymphangitic breast cancer represents a clinical spectrum and a model disease. Inflammation and the immune response have a role in the natural history of this special clinical presentation. Preclinical models and biomarker studies suggest that inflammatory breast cancer comprises a more important role for the tumour microenvironment, including immune cell infiltration and vasculogenesis, especially lympho-angiogenesis. Across this clinical continuum of the chest wall disease there is an important role of the inflammation cascade. The activation of mature dendritic cells (DCs) through toll like receptors (TLRs) or by inflammatory cytokines converts immature DCs into mature DCs that present specific antigen to T cells, thereby activating them. Maturation of DCs is accompanied by co-stimulatory molecules and secretion of inflammatory cytokines polarizing lymphocytic, macrophages and fibroblast infiltration. It is unknown whether immune cells associated to the IBC microenvironment play a role in this scenario to transiently promote epithelial to mesenchymal transition (EMT) in these cells. Immune and microenvirnment factors can induce phenotypic, morphological, and functional changes in breast cancer cells. We can hypothesize that similar inflammatory conditions in vivo may support both the rapid metastasis and tight tumor emboli that are characteristic of chest wall disease and that targeted anti-inflammatory therapy may play a role in this patient population. The current review will review biological and clinical data of this special condition.
Collapse
Affiliation(s)
- Giuseppe Curigliano
- University of Milano, Department of Oncology and Hemato-Oncology, Division of Early Drug Development for Innovative Therapies, Istituto Europeo di Oncologia, Via Ripamonti 435, 20141 Milano, Italy.
| |
Collapse
|
26
|
Li J, Xia Y, Wu Q, Zhu S, Chen C, Yang W, Wei W, Sun S. Outcomes of patients with inflammatory breast cancer by hormone receptor- and HER2-defined molecular subtypes: A population-based study from the SEER program. Oncotarget 2018; 8:49370-49379. [PMID: 28472761 PMCID: PMC5564775 DOI: 10.18632/oncotarget.17217] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 03/24/2017] [Indexed: 01/12/2023] Open
Abstract
Background The aim of this study was to evaluate the outcomes of patients with inflammatory breast cancer (IBC), with emphasis on the role of molecular subtypes and radiotherapy. Methods A retrospective cohort study to investigate overall survival (OS) and breast cancer-specific mortality (BCSM) in patients with IBC was conducted using data obtained by the Surveillance, Epidemiology, and End Results (SEER) program from 2010–2013. Cox multivariate regression was used to calculate the adjusted Hazard Ratios (aHR). Results 403 patients were eligible for this study. Patients in the group with hormone receptors (HR)+/HER2- subtype had an OS of 79.6% compared with 89.0 % in the group with (HR)+/HER2+ subtype and 76.8% in the HR-/HER2+ group and 62.9% in the triple-negative (TN) group. BCSM was 16.3% for the HR+/HER2- group, 9.8% for the HR+/HER2+ group, 21.7% for the HR-/HER2+ group, and 30.5% for the TN group. For distant metastases, the results showed that there was a high probability of bone metastasis in HR-positive groups, brain and liver metastasis in HER2-positive groups, and lung metastasis in the TN group. Multivariate analysis demonstrated that estrogen receptor and HER2 positivity were associated with better survival and that the TN subtype had a poorer OS and BCSM compared with other subtypes (P<0.05). Furthermore, patients who received radiotherapy were more likely to have improved survival (P< 0.05). Conclusion Inflammatory breast cancer appears to alter the prognosis in association with the receptor status and molecular subtypes. Radiotherapy was still considered to be a crucial treatment for patients with IBC.
Collapse
Affiliation(s)
- Juanjuan Li
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| | - Yue Xia
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| | - Qi Wu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| | - Shan Zhu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| | - Chuang Chen
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| | - Wen Yang
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| | - Wen Wei
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| | - Shengrong Sun
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| |
Collapse
|
27
|
Raposo TP, Arias-Pulido H, Chaher N, Fiering SN, Argyle DJ, Prada J, Pires I, Queiroga FL. Comparative aspects of canine and human inflammatory breast cancer. Semin Oncol 2018. [PMID: 29526258 DOI: 10.1053/j.seminoncol.2017.10.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Inflammatory breast cancer (IBC) in humans is the most aggressive form of mammary gland cancer and shares clinical, pathologic, and molecular patterns of disease with canine inflammatory mammary carcinoma (CIMC). Despite the use of multimodal therapeutic approaches, including targeted therapies, the prognosis for IBC/CIMC remains poor. The aim of this review is to critically analyze IBC and CIMC in terms of biology and clinical features. While rodent cancer models have formed the basis of our understanding of cancer biology, the translation of this knowledge into improved outcomes has been limited. However, it is possible that a comparative "one health" approach to research, using a natural canine model of the disease, may help advance our knowledge on the biology of the disease. This will translate into better clinical outcomes for both species. We propose that CIMC has the potential to be a useful model for developing and testing novel therapies for IBC. Further, this strategy could significantly improve and accelerate the design and establishment of new clinical trials to identify novel and improved therapies for this devastating disease in a more predictable way.
Collapse
Affiliation(s)
- Teresa P Raposo
- Division of Cancer and Stem Cells, Faculty of Medicine, University of Nottingham, United Kingdom
| | - Hugo Arias-Pulido
- Department of Microbiology and Immunology and Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire 03756, USA
| | - Nabila Chaher
- Department of Pathology, Centre Pierre et Marie Curie, 1, Avenue Battendier, Place May 1st, Algiers, Algeria
| | - Steven N Fiering
- Department of Microbiology and Immunology and Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire 03756, USA
| | - David J Argyle
- The Royal (Dick) School of Veterinary Studies and Roslin Institute, Easter Bush Campus, Midlothian, University of Edinburgh, United Kingdom
| | - Justina Prada
- Departament of Veterinary Sciences, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal; Animal and Veterinary research Centre (CECAV), University of Trás-os-Montes and Alto Douro, Vila Real, Portugal
| | - Isabel Pires
- Departament of Veterinary Sciences, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal; Animal and Veterinary research Centre (CECAV), University of Trás-os-Montes and Alto Douro, Vila Real, Portugal
| | - Felisbina Luísa Queiroga
- Departament of Veterinary Sciences, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal; Center for the Study of Animal Sciences, CECA-ICETA, University of Porto, Porto, Portugal; Center for Research and Technology of Agro-Environment and Biological Sciences, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal.
| |
Collapse
|
28
|
Prognostic and predictive role of spatially positioned tumour infiltrating lymphocytes in metastatic HER2 positive breast cancer treated with trastuzumab. Sci Rep 2017; 7:18027. [PMID: 29269742 PMCID: PMC5740084 DOI: 10.1038/s41598-017-18266-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 12/04/2017] [Indexed: 01/01/2023] Open
Abstract
Disease outcomes of HER2+ breast cancers have dramatically changed after targeted therapies, such as trastuzumab, came to clinical practice but predictive factors for trastuzumab sensitivity and resistance are frequently unknown. Current work included metastatic breast cancer patients (n = 48), who were treated with trastuzumab and had pre-treatment tumour samples available. The tumours were immunostained for T-cell (CD3, CD8), natural killer (NK)-cell (CD56) and macrophage (CD68) markers and quantitative analysis of the immune cells was carried out using a computer-assisted image analysis in different tumour locations. High number of CD3 and CD8 positive T-cells was associated with significant survival benefit in the center of the tumour (CT) (p = 0.007, p = 0.001) but not in the invasive margin. The number of NK-cells and macrophages in the CT showed non-significant tendency towards improved survival. In subgroup analyses, high density of CD8 CT cells was associated with significant survival benefit in non-bone only disease, in TX or T1-3, and in ER+ tumours (p = 0.006, p = 0.003, p = 0.001). Moreover, high CD8 CT cell density associated significantly with long trastuzumab interruption periods in response. The results suggest important prognostic and predictive role of tumour infiltrating lymphocytes in center of the tumours in metastatic HER2+ breast cancer.
Collapse
|
29
|
Costa R, Santa-Maria CA, Rossi G, Carneiro BA, Chae YK, Gradishar WJ, Giles FJ, Cristofanilli M. Developmental therapeutics for inflammatory breast cancer: Biology and translational directions. Oncotarget 2017; 8:12417-12432. [PMID: 27926493 PMCID: PMC5355355 DOI: 10.18632/oncotarget.13778] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 11/07/2016] [Indexed: 12/16/2022] Open
Abstract
Inflammatory breast cancer (IBC) is a rare and aggressive form of breast cancer, which accounts for approximately 3% of cases of breast malignancies. Diagnosis relies largely on its clinical presentation, and despite a characteristic phenotype, underlying molecular mechanisms are poorly understood. Unique clinical presentation indicates that IBC is a distinct clinical and biological entity when compared to non-IBC. Biological understanding of non-IBC has been extrapolated into IBC and targeted therapies for HER2 positive (HER2+) and hormonal receptor positive non-IBC led to improved patient outcomes in the recent years. This manuscript reviews recent discoveries related to the underlying biology of IBC, clinical progress to date and suggests rational approaches for investigational therapies.
Collapse
Affiliation(s)
- Ricardo Costa
- Developmental Therapeutics Program, Division of Hematology/Oncology, Feinberg School of Medicine, Chicago, United States of America
| | - Cesar A Santa-Maria
- Developmental Therapeutics Program, Division of Hematology/Oncology, Feinberg School of Medicine, Chicago, United States of America.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, United States of America
| | - Giovanna Rossi
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, United States of America
| | - Benedito A Carneiro
- Developmental Therapeutics Program, Division of Hematology/Oncology, Feinberg School of Medicine, Chicago, United States of America.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, United States of America
| | - Young Kwang Chae
- Developmental Therapeutics Program, Division of Hematology/Oncology, Feinberg School of Medicine, Chicago, United States of America.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, United States of America
| | - William J Gradishar
- Developmental Therapeutics Program, Division of Hematology/Oncology, Feinberg School of Medicine, Chicago, United States of America.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, United States of America
| | - Francis J Giles
- Developmental Therapeutics Program, Division of Hematology/Oncology, Feinberg School of Medicine, Chicago, United States of America.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, United States of America
| | - Massimo Cristofanilli
- Developmental Therapeutics Program, Division of Hematology/Oncology, Feinberg School of Medicine, Chicago, United States of America.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, United States of America
| |
Collapse
|
30
|
Abstract
Metastases claim more than 90% of cancer-related patient deaths and are usually seeded by a subset of circulating tumor cells shed off from the primary tumor. In circulation, circulating tumor cells are found both as single cells and as clusters of cells. The clusters of circulating tumor cells, although many fewer in number, possess much higher metastatic potential as compared to that of individual circulating tumor cells. In this review, we highlight recent insights into molecular mechanisms that can enable the formation of these clusters—(a) hybrid epithelial/mesenchymal phenotype of cells that couples their ability to migrate and adhere, and (b) intercellular communication that can spatially coordinate the cluster formation and provide survival signals to cancer cells. Building upon these molecular mechanisms, we also offer a possible mechanistic understanding of why clusters are endowed with a higher metastatic potential. Finally, we discuss the highly aggressive Inflammatory Breast Cancer as an example of a carcinoma that can metastasize via clusters and corroborates the proposed molecular mechanisms.
Collapse
|
31
|
Gonçalves A, Monneur A, Viens P, Bertucci F. The use of systemic therapies to prevent progression of inflammatory breast cancer: which targeted therapies to add on cytotoxic combinations? Expert Rev Anticancer Ther 2017; 17:593-606. [PMID: 28506194 DOI: 10.1080/14737140.2017.1330655] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
INTRODUCTION Inflammatory breast cancer is a rare but frequently fatal disease, essentially because of its high ability to develop distant metastases. Even though the prognosis of IBC was significantly improved by multimodal management, including the systematic use of cytotoxic-based induction, the prognosis remains largely dismal. Areas covered: This review presents the main achievements in the systemic treatment of IBC during the past 30 years. It focuses more specifically on recent results obtained with targeted therapies, including anti-HER2 and anti-angiogenic agents. Novel approaches under investigation are presented. Expert commentary: Current management of IBC is subtype-specific and the largest benefit has been achieved in HER2-positive disease. The identification of breakthrough therapeutic advances is eagerly awaited and will require the development of IBC-specific clinical trials. Future clinical investigations should not only aim to increase the pathological response rate but also to eradicate distant metastases, which ultimately lead to patient death.
Collapse
Affiliation(s)
- Anthony Gonçalves
- a Department of Medical Oncology, Institut Paoli-Calmettes, Aix Marseille Univ , CNRS U7258, INSERM U1068, CRCM , Marseille , France
| | - Audrey Monneur
- a Department of Medical Oncology, Institut Paoli-Calmettes, Aix Marseille Univ , CNRS U7258, INSERM U1068, CRCM , Marseille , France
| | - Patrice Viens
- a Department of Medical Oncology, Institut Paoli-Calmettes, Aix Marseille Univ , CNRS U7258, INSERM U1068, CRCM , Marseille , France
| | - François Bertucci
- a Department of Medical Oncology, Institut Paoli-Calmettes, Aix Marseille Univ , CNRS U7258, INSERM U1068, CRCM , Marseille , France
| |
Collapse
|
32
|
Challenging a Misnomer? The Role of Inflammatory Pathways in Inflammatory Breast Cancer. Mediators Inflamm 2017; 2017:4754827. [PMID: 28607534 PMCID: PMC5457777 DOI: 10.1155/2017/4754827] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 04/18/2017] [Indexed: 12/26/2022] Open
Abstract
Inflammatory breast cancer is a rare, yet highly aggressive form of breast cancer, which accounts for less than 5% of all locally advanced presentations. The clinical presentation of inflammatory breast cancer often differs significantly from that of noninflammatory breast cancer; however, immunohistochemistry reveals few, if any, distinguishing features. The more aggressive triple-negative and HER2-positive breast cancer subtypes are overrepresented in inflammatory breast cancer compared with noninflammatory breast cancer, with a poorer prognosis in response to conventional therapies. Despite its name, there remains some controversy regarding the role of inflammation in inflammatory breast cancer. This review summarises the current molecular evidence suggesting that inflammatory signaling pathways are upregulated in this disease, including NF-κB activation and excessive IL-6 production among others, which may provide an avenue for novel therapeutics. The role of the tumor microenvironment, through tumor-associated macrophages, infiltrating lymphocytes, and cancer stem cells is also discussed, suggesting that these tumor extrinsic factors may help account for the differences in behavior between inflammatory breast cancer and noninflammatory breast cancer. While there are various novel treatment strategies already underway in clinical trials, the need for further development of preclinical models of this rare but aggressive disease is paramount.
Collapse
|
33
|
Su C, Zhang P, Liu J, Cao Y. Erianin inhibits indoleamine 2, 3-dioxygenase -induced tumor angiogenesis. Biomed Pharmacother 2017; 88:521-528. [PMID: 28129624 DOI: 10.1016/j.biopha.2017.01.090] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 01/13/2017] [Accepted: 01/14/2017] [Indexed: 01/17/2023] Open
Abstract
Tumor angiogenesis is the key process in tumor growth and metastasis, and transfers essential nutrients for solid tumor. Inhibition of tumor angiogenesis has been recognized as a more effective anti-cancer strategy for NSCLC and has acquired certain therapeutic effects. IDO has non-immune functions including regulating tumor angiogenesis and IDO dysregulation in cancer pathogenesis has been valued. Erianin is a natural product isolated from Dendrobium chrysotoxum Lindl. The antitumor activity of erianin in many kinds of cancers had been demonstrated in previous studies. In this study, we demonstrated that IDO could promote the attachment of 2LL cells, the ability of migration, invasion and VM formation, as well as the tubules forming ability of HUVECs. We also find that erianin suppressed expression and enzyme ability of IDO and erianin could inhibit IDO-induced metastasis and invasion ability of 2LL cells significantly. Erianin not only blocked IDO-induced tube formation of HUVECs, but also suppressed VM formation of 2LL-IDO cells. What's more, we examined that Erianin might play its role in angiogenesis through down-regulating phosphorylation of JAK2/STAT3, inhibiting its downstream target genes MMP-2/-9 and some inflammatory mediators (COX-2, HIF-1α and IL-6), which were all induced by IDO. All these results indicated that erianin had anti-angiogenesis ability and could inhibit the expresison of IDO to prevent and treat the malignant tumors.
Collapse
Affiliation(s)
- Chang Su
- Minhang Hospital, Fudan University, China
| | - Peng Zhang
- School of Pharmacy, East China University of Science and Technology, China
| | - Jianwen Liu
- School of Pharmacy, East China University of Science and Technology, China.
| | - Yiou Cao
- Minhang Hospital, Fudan University, China.
| |
Collapse
|
34
|
Anti-angiogenic treatment in breast cancer: Facts, successes, failures and future perspectives. Cancer Treat Rev 2017; 53:98-110. [PMID: 28088074 DOI: 10.1016/j.ctrv.2016.12.009] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 12/20/2016] [Accepted: 12/21/2016] [Indexed: 01/06/2023]
Abstract
Angiogenesis is one of the hallmarks of cancer and a crucial requisite in the development of tumors. Interrupting this process by blocking the vascular endothelial growth factor (VEGF) with the monoclonal antibody bevacizumab has been considered a possible breakthrough in the treatment of various types of cancer, especially for advanced disease. However in breast cancer, studies have shown ambivalent results causing debate about the value of this drug. In this article, we review the evidence for anti-angiogenic treatment options for breast cancer, as well as discuss the possible factors limiting the effectiveness of anti-angiogenic agents and offer a recommendation regarding the future research on these therapies for the treatment of breast cancer.
Collapse
|
35
|
Curigliano G. Chest Wall Disease: The Clinical Continuum Between Inflammatory and Lymphangitic Breast Cancer. Breast Cancer 2017. [DOI: 10.1007/978-3-319-48848-6_60] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
36
|
Roggiani F, Mezzanzanica D, Rea K, Tomassetti A. Guidance of Signaling Activations by Cadherins and Integrins in Epithelial Ovarian Cancer Cells. Int J Mol Sci 2016; 17:ijms17091387. [PMID: 27563880 PMCID: PMC5037667 DOI: 10.3390/ijms17091387] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 08/11/2016] [Accepted: 08/13/2016] [Indexed: 12/12/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is the deadliest tumor among gynecological cancer in the industrialized countries. The EOC incidence and mortality have remained unchanged over the last 30 years, despite the progress in diagnosis and treatment. In order to develop novel and more effective therapeutic approaches, the molecular mechanisms involved in EOC progression have been thoroughly investigated in the last few decades. At the late stage, peritoneal metastases originate from the attachment of small clusters of cancer cells that shed from the primary site and carried by the ascites adhere to the abdominal peritoneum or omentum. This behavior suggests that cell–cell or cell–matrix adhesion mechanisms regulate EOC growth and dissemination. Complex downstream signalings, which might be influenced by functional cross-talk between adhesion molecules and co-expressed and activated signaling proteins, can affect the proliferation/survival and the migration/invasion of EOC cells. This review aimed to define the impact of the mechanisms of cell–cell, through cadherins, and cell–extracellular matrix adhesion, through integrins, on the signaling cascades induced by membrane receptors and cytoplasmic proteins known to have a role in the proliferation, migration and invasion of EOC cells. Finally, some novel approaches using peptidomimetic ligands to cadherin and integrins are summarized.
Collapse
Affiliation(s)
- Francesca Roggiani
- Unit of Molecular Therapies, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, Milan 20133, Italy.
| | - Delia Mezzanzanica
- Unit of Molecular Therapies, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, Milan 20133, Italy.
| | - Katia Rea
- Unit of Molecular Therapies, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, Milan 20133, Italy.
| | - Antonella Tomassetti
- Unit of Molecular Therapies, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, Milan 20133, Italy.
| |
Collapse
|
37
|
Deng Y, Sriwiriyajan S, Tedasen A, Hiransai P, Graidist P. Anti-cancer effects of Piper nigrum via inducing multiple molecular signaling in vivo and in vitro. JOURNAL OF ETHNOPHARMACOLOGY 2016; 188:87-95. [PMID: 27155135 DOI: 10.1016/j.jep.2016.04.047] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 04/23/2016] [Accepted: 04/25/2016] [Indexed: 06/05/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Piper nigrum is widely used as a folk medicine including usage for pain relief, fevers, as well as an anti-cancer agent. However the crude extract of piperine free P. nigrum (PFPE), which inhibits breast cancer, and its mechanisms are still being kept secret. This research aims to elucidate the anti-cancer effects of PFPE and its mechanisms. MATERIALS AND METHODS Anti-cancer effects of PFPE were investigated in N-nitroso-N-methylurea (NMU)-induced mammary tumorigenesis rats and breast cancer cell lines MCF-7 and ZR-75-1. Furthermore, the cancer prevention effects of PFPE were investigated in rats. Western blotting was employed to study protein levels induced by PFPE. RESULTS PFPE was found to up-regulate p53, and down-regulate estrogen receptor (ER), E-cadherin (E-cad), matrix metalloproteinase 9 (MMP-9), matrix metalloproteinase 2 (MMP-2), c-Myc, and vascular endothelial growth factor (VEGF) levels in breast cancer rats. Moreover, PFPE decreased protein levels of E-cad, c-Myc, and VEGF in MCF-7 cells. These results suggest that PFPE can enhance breast cancer cell response to phytochemicals, then induce cell cycle arrest, and inhibit cancer cell proliferation resulting in tumor size decrease in the PFPE treated group. It further suggests that PFPE may suppress tumor cell invasion, migration, and angiogenesis. In addition, PFPE possessed cancer prevention effects through generation of reactive oxygen species (ROS) to higher cancer cell cellular stress. CONCLUSIONS PFPE may possess anti-cancer and cancer prevention effects; hence, it deserves further investigation as a novel candidate for breast cancer treatment.
Collapse
Affiliation(s)
- Yan Deng
- Department of Biomedical Sciences, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand; Yunnan Institute of Parasitic Diseases, Puer, Yunnan 65900, PR China.
| | - Somchai Sriwiriyajan
- Department of Biomedical Sciences, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand; Department of Pharmacology, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand.
| | - Aman Tedasen
- Department of Biomedical Sciences, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand.
| | - Poonsit Hiransai
- Molecular Medicine and Cancer Biology Research Unit, School of Allied Health Sciences and Public Health, Walailak University, Nakhon Si Thammarat 80161, Thailand.
| | - Potchanapond Graidist
- Department of Biomedical Sciences, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand; The Excellent Research Laboratory of Cancer Molecular Biology, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand.
| |
Collapse
|
38
|
Raluca BA, Cimpean AM, Cioca A, Cretu O, Mederle O, Ciolofan A, Gaje P, Raica M. Endothelial Cell Proliferation and Vascular Endothelial Growth Factor Expression in Primary Colorectal Cancer and Corresponding Liver Metastases. Asian Pac J Cancer Prev 2016; 16:4549-53. [PMID: 26107202 DOI: 10.7314/apjcp.2015.16.11.4549] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND . Colorectal carcinoma (CRC) is one of the major causes of cancer death worldwide. Data from the literature indicate differences between the proliferation rate of endothelial cells relative to the morphology growth type, possibly due to origin of specimens (autopsy material, surgery fragments) or quantification methods. Vascular endothelial growth factor (VEGF) is a factor that stimulates the proliferation of endothelial cells. It is expressed in more than 90% of cases of metastatic CRC. AIM The aim of this study was to evaluate the endothelial cell proliferation and VEGF expression in primary tumors and corresponding liver metastases. MATERIALS AND METHODS Our study included 24 recent biopsies of primary tumors and corresponding liver metastases of CRC cases. CD34/ Ki67 double immunostaining and RNA scope assay for VEGF were performed. RESULTS In the primary tumors analysis of VEGFmRNA expression indicated no significant correlation with differentiation grade, proliferative and non-proliferative vessels in the intratumoral and peritumoral areas. In contrast, in the corresponding liver metastases, VEGFmRNA expression significantly correlated with the total number of non- proliferative vessels and total number of vessels. CD34/ Ki67 double immunostaining in the cases with poorly differentiated carcinoma indicated a high number of proliferating endothelial cells in the peritumoral area and a low number in the intratumoral area for the primary tumor. Moderately differentiated carcinomas of colon showed no proliferating endothelial cells in the intratumoral area in half of the cases included in the study, for both, primary tumor and liver metastasis. In well differentiated CRCs, in primary tumors, a high proliferation rate of endothelial cells in the intratumoral area and a lower proliferation rate in the peritumoral area were found. A low value was found in corresponding liver metastasis. CONCLUSIONS The absence of proliferative endothelial cells in half of the cases for the primary tumors and liver metastases in moderately differentiated carcinoma suggest a vascular mimicry phenomenon. The mismatch between the total number of vessels and endothelial proliferation in primary tumors indicate that a functional vascular network is already formed or the existence of some mechanisms influenced by other angiogenic factors.
Collapse
Affiliation(s)
- Balica Amalia Raluca
- Department of Histology, Angiogenesis Research Center, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania E-mail :
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Woodward WA. Inflammatory breast cancer: unique biological and therapeutic considerations. Lancet Oncol 2016; 16:e568-e576. [PMID: 26545845 DOI: 10.1016/s1470-2045(15)00146-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Revised: 06/29/2015] [Accepted: 07/06/2015] [Indexed: 12/29/2022]
Abstract
Through the concerted efforts of many patients, health-care providers, legislators, and other supporters, the past decade has seen the development of the first clinics dedicated to the care of patients with inflammatory breast cancer in the USA and other countries. Together with social networking, advocacy, and education, a few specialised centres have had substantial increases in patient numbers (in some cases ten times higher), which has further expanded the community of science and advocacy and increased the understanding of the disease process. Although inflammatory breast cancer is considered rare, constituting only 2-4% of breast cancer cases, poor prognosis means that patients with the disease account for roughly 10% of breast cancer mortality annually in the USA. I propose that the unique presentation of inflammatory breast cancer might require specific, identifiable changes in the breast parenchyma that occur before the tumour-initiating event. This would make the breast tissue itself a tumour-promoting medium that should be treated as a component of the pathology in multidisciplinary treatment and should be further studied for complementary targets to inhibit the pathobiology that is specific to inflammatory breast cancer.
Collapse
Affiliation(s)
- Wendy A Woodward
- Department of Radiation Oncology and MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
40
|
Jhaveri K, Teplinsky E, Silvera D, Valeta-Magara A, Arju R, Giashuddin S, Sarfraz Y, Alexander M, Darvishian F, Levine PH, Hashmi S, Zolfaghari L, Hoffman HJ, Singh B, Goldberg JD, Hochman T, Formenti S, Esteva FJ, Moran MS, Schneider RJ. Hyperactivated mTOR and JAK2/STAT3 Pathways: Molecular Drivers and Potential Therapeutic Targets of Inflammatory and Invasive Ductal Breast Cancers After Neoadjuvant Chemotherapy. Clin Breast Cancer 2015; 16:113-22.e1. [PMID: 26774497 DOI: 10.1016/j.clbc.2015.11.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 11/09/2015] [Accepted: 11/12/2015] [Indexed: 01/02/2023]
Abstract
INTRODUCTION Inflammatory breast cancer (IBC) is an aggressive and rare cancer with a poor prognosis and a need for novel targeted therapeutic strategies. Preclinical IBC data showed strong activation of the phosphatidylinositide-3-kinase/mammalian target of rapamycin (mTOR) and Janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathways, and expression of inflammatory cytokines and tumor-associated macrophages (TAMs). PATIENTS AND METHODS Archival tumor tissue from 3 disease types (IBC treated with neoadjuvant chemotherapy [NAC], n = 45; invasive ductal carcinoma [IDC] treated with NAC [n = 24; 'treated IDC'; and untreated IDC [n = 27; 'untreated IDC']) was analyzed for the expression of biomarkers phospho-S6 (pS6) (mTOR), phospho-JAK2 (pJAK2), pSTAT3, interleukin (IL)-6, CD68 (monocytes, macrophages), and CD163 (TAMs). Surrounding nontumor tissue was also analyzed. RESULTS Biomarker levels and surrogate activity according to site-specific phosphorylation were shown in the tumor tissue of all 3 disease types but were greatest in IBC and treated IDC and least in untreated IDC for pS6, pJAK2, pSTAT3, and IL-6. Of 37 IBC patients with complete biomarker data available, 100% were pS6-positive and 95% were pJAK2-positive. In nontumor tissue, biomarker levels were observed in all groups but were generally greatest in untreated IDC and least in IBC, except for JAK2. CONCLUSION IBC and treated IDC display similar levels of mTOR and JAK2 biomarker activation, which suggests a potential mechanism of resistance after NAC. Biomarker levels in surrounding nontumor tissue suggested that the stroma might be activated by chemotherapy and resembles the oncogenic tumor-promoting environment. Activation of pS6 and pJAK2 in IBC might support dual targeting of the mTOR and JAK/STAT pathways, and the need for prospective studies to investigate combined targeted therapies in IBC.
Collapse
Affiliation(s)
- Komal Jhaveri
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY.
| | - Eleonora Teplinsky
- Division of Hematology & Medical Oncology, Department of Medicine, New York University School of Medicine, New York, NY
| | - Deborah Silvera
- Department of Microbiology, New York University School of Medicine, New York, NY
| | - Amanda Valeta-Magara
- Department of Microbiology, New York University School of Medicine, New York, NY
| | - Rezina Arju
- Department of Microbiology, New York University School of Medicine, New York, NY
| | - Shah Giashuddin
- Department of Pathology, The Brooklyn Hospital Center, Brooklyn, NY
| | - Yasmeen Sarfraz
- Department of Microbiology, New York University School of Medicine, New York, NY
| | | | | | - Paul H Levine
- Department of Epidemiology and Biostatistics, George Washington University, Washington, DC
| | - Salman Hashmi
- Division of Biostatistics, New York University School of Medicine, New York, NY
| | - Ladan Zolfaghari
- Division of Biostatistics, New York University School of Medicine, New York, NY
| | - Heather J Hoffman
- Division of Biostatistics, New York University School of Medicine, New York, NY
| | - Baljit Singh
- Department of Epidemiology and Biostatistics, George Washington University, Washington, DC
| | - Judith D Goldberg
- Division of Biostatistics, New York University School of Medicine, New York, NY
| | - Tsivia Hochman
- Division of Biostatistics, New York University School of Medicine, New York, NY
| | - Silvia Formenti
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY
| | - Francisco J Esteva
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY
| | - Meena S Moran
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT
| | - Robert J Schneider
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY; Department of Microbiology, New York University School of Medicine, New York, NY
| |
Collapse
|
41
|
Abstract
Locally advanced breast cancer (LABC) constitutes a heterogeneous entity that includes advanced-stage primary tumours, cancers with extensive nodal involvement and inflammatory breast carcinomas. Although the definition of LABC can be broadened to include some large operable breast tumours, we use this term to strictly refer to inoperable cancers that are included in the above-mentioned categories. The prognosis of such tumours is often unfavourable; despite aggressive treatment, many patients eventually develop distant metastases and die from the disease. Advances in systemic therapy, including radiation treatment, surgical techniques and the development of new targeted agents have significantly improved clinical outcomes for patients with this disease. Notwithstanding these advances, LABC remains an important clinical problem, particularly in developing countries and those without widely adapted breast cancer awareness programmes. The optimal management of LABC requires a multidisciplinary approach, a well-coordinated treatment schedule and close cooperation between medical, surgical and radiation oncologists. In this Review, we discuss the current state of the art and possible future treatment strategies for patients with LABC.
Collapse
|
42
|
Mego M, Giordano A, De Giorgi U, Masuda H, Hsu L, Giuliano M, Fouad TM, Dawood S, Ueno NT, Valero V, Andreopoulou E, Alvarez RH, Woodward WA, Hortobagyi GN, Cristofanilli M, Reuben JM. Circulating tumor cells in newly diagnosed inflammatory breast cancer. Breast Cancer Res 2015; 17:2. [PMID: 25572591 PMCID: PMC4318180 DOI: 10.1186/s13058-014-0507-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 12/17/2014] [Indexed: 11/25/2022] Open
Abstract
Introduction Circulating tumor cells (CTCs) are an independent prognostic factor for progression-free survival (PFS) and overall survival (OS) in patients with metastatic breast cancer. Inflammatory breast cancer (IBC) is one of the most aggressive forms of breast cancer. The prognostic value of a CTC count in newly diagnosed IBC has not been established. The aim of this study was to assess the prognostic value of a baseline CTC count in patients with newly diagnosed IBC. Methods This retrospective study included 147 patients with newly diagnosed IBC (77 with locally advanced and 70 with metastatic IBC) treated with neoadjuvant therapy or first-line chemotherapy during the period from January 2004 through December 2012 at The University of Texas MD Anderson Cancer Center. CTCs were detected and enumerated by using the CellSearch system before patients were started with chemotherapy. Results The proportion of patients with ≥1 CTC was lower among patients with stage III than among patients with metastatic IBC (54.5% versus 84.3%; P = 0.0002); the proportion of patients with ≥5 CTCs was also lower for stage III than for metastatic IBC (19.5% versus 47.1%; P = 0.0004). Patients with fewer than five CTCs had significantly better progression-free survival (PFS) (hazard ratio (HR) = 0.60; P = 0.02) and overall survival (HR = 0.59; P = 0.03) than patients with five or more CTCs. Among patients with stage III IBC, there was a nonsignificant difference in PFS (HR = 0.66; 95% confidence interval (CI), 0.31 to 1.39; P = 0.29) and OS (HR = 0.54; 95% CI, 0.24 to 1.26; P = 0.48) in patients with no CTCs compared with patients with one or more CTCs. In multivariate analysis, CTC was prognostic for PFS and OS independent of clinical stage. Conclusions CTCs can be detected in a large proportion of patients with newly diagnosed IBC and are a strong predictor of worse prognosis in patients with newly diagnosed IBC.
Collapse
Affiliation(s)
- Michal Mego
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA. .,Department of Medical Oncology, Comenius University, School of Medicine, Bratislava, Slovakia. .,Present affiliation: Breast Center, Thomas Jefferson University-Kimmel Cancer Center, Philadelphia, PA, USA.
| | - Antonio Giordano
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.
| | - Ugo De Giorgi
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA. .,Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) - IRCCS, Meldola, FC, Italy.
| | - Hiroko Masuda
- Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Limin Hsu
- Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Mario Giuliano
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA. .,Department of Clinical Medicine and Surgery, University Federico II, Naples, Italy.
| | - Tamer M Fouad
- Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Shaheenah Dawood
- Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. .,Medical Oncology Department, Dubai Hospital, Dubai, UAE.
| | - Naoto T Ueno
- Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. .,Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Vicente Valero
- Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. .,Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Eleni Andreopoulou
- Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. .,Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Ricardo H Alvarez
- Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. .,Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Wendy A Woodward
- Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Gabriel N Hortobagyi
- Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Massimo Cristofanilli
- Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. .,Present affiliation: Breast Center, Thomas Jefferson University-Kimmel Cancer Center, Philadelphia, PA, USA.
| | - James M Reuben
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA. .,Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
43
|
|
44
|
Li XJ, Zha QB, Xu XY, Xia L, Zhang Z, Ren ZJ, Tang JH. Lack of prognostic value of human epidermal growth factor-like receptor 2 status in inflammatory breast cancer (IBC): a meta-analysis. Asian Pac J Cancer Prev 2014; 15:9615-9. [PMID: 25520077 DOI: 10.7314/apjcp.2014.15.22.9615] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Inflammatory breast cancer (IBC) is a rare, aggressive form of breast cancer which is more likely to be her-2/ neu amplified. While the her-2/neu status has been utilised to predict prognosis, the published data are inconsistent. The present meta-analysis was conducted to determine whether the her-2/neu status predicts outcomes. Papers were selected from the PubMed database based on defined inclusion and exclusion criteria. Parameters such as total patients, follow-up time and outcome statistics (i.e. overall survival (OS), relapse-free survival (RFS) were collected. The analysis included 6 studies with 2,838 IBC patients. The summary hazards ratio (HR) estimating the association of OS with HER-2-positive disease was 0.96 (95% confidence interval (95%CI: 0.85-1.10)), with similar findings for RFS (HR=0.81, 95%CI: 0.61-1.09). No obvious statistical heterogeneity was detected. This meta-analysis suggests that HER-2-positive status is not an independent adverse prognostic factor for survival among IBC patient cases.
Collapse
Affiliation(s)
- Xiu-Juan Li
- Department of General Surgery, Jiangsu Cancer Hospital, The Affiliated Hospital of Nanjing Medical University, Nanjing, China E-mail : ,
| | | | | | | | | | | | | |
Collapse
|
45
|
van Uden DJP, van Laarhoven HWM, Westenberg AH, de Wilt JHW, Blanken-Peeters CFJM. Inflammatory breast cancer: an overview. Crit Rev Oncol Hematol 2014; 93:116-26. [PMID: 25459672 DOI: 10.1016/j.critrevonc.2014.09.003] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Revised: 08/08/2014] [Accepted: 09/30/2014] [Indexed: 12/14/2022] Open
Abstract
Inflammatory breast cancer (IBC) is the most aggressive entity of breast cancer. Management involves coordination of multidisciplinary management and usually includes neoadjuvant chemotherapy, ablative surgery if a tumor-free resection margin is expected and locoregional radiotherapy. This multimodal therapeutic approach has significantly improved patient survival. However, the median overall survival among women with IBC is still poor. By elucidating the biologic characteristics of IBC, new treatment options may become available. We performed a comprehensive review of the English-language literature on IBC through computerized literature searches. The objective of the current review is to present an overview of the literature related to the biology, imaging and multidisciplinary treatment of inflammatory breast cancer.
Collapse
Affiliation(s)
- D J P van Uden
- Department of Surgery, Canisius Wilhelmina Hospital, Postbus 9015, 6500 GS Nijmegen, The Netherlands.
| | - H W M van Laarhoven
- Medical Oncology, Academic Medical Center, University of Amsterdam, Postbus 22660, 1100 DD Amsterdam, The Netherlands
| | - A H Westenberg
- Institute for Radiation Oncology, Postbus 60160, 6800 JD Arnhem, The Netherlands
| | - J H W de Wilt
- Department of Surgery, Radboud University Medical Center, Geert Grooteplein-Zuid 10, 6525 GA Nijmegen, The Netherlands
| | | |
Collapse
|
46
|
Cheung KJ, Ewald AJ. Illuminating breast cancer invasion: diverse roles for cell-cell interactions. Curr Opin Cell Biol 2014; 30:99-111. [PMID: 25137487 DOI: 10.1016/j.ceb.2014.07.003] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 07/26/2014] [Accepted: 07/28/2014] [Indexed: 02/08/2023]
Abstract
Metastasis begins when tumors invade into surrounding tissues. In breast cancer, the study of cell interactions has provided fundamental insights into this complex process. Powerful intravital and 3D organoid culture systems have emerged that enable biologists to model the complexity of cell interactions during cancer invasion in real-time. Recent studies utilizing these techniques reveal distinct mechanisms through which multiple cancer cell and stromal cell subpopulations interact, including paracrine signaling, direct cell-cell adhesion, and remodeling of the extracellular matrix. Three cell interaction mechanisms have emerged to explain how breast tumors become invasive: epithelial-mesenchymal transition, collective invasion, and the macrophage-tumor cell feedback loop. Future work is needed to distinguish whether these mechanisms are mutually exclusive or whether they cooperate to drive metastasis.
Collapse
Affiliation(s)
- Kevin J Cheung
- Department of Cell Biology, Center for Cell Dynamics, School of Medicine, Johns Hopkins University, 855 N. Wolfe St, 452 Rangos Bldg, Baltimore, MD 21205, USA; Department of Oncology, School of Medicine, Johns Hopkins University, 855 N. Wolfe St, 452 Rangos Bldg, Baltimore, MD 21205, USA.
| | - Andrew J Ewald
- Department of Cell Biology, Center for Cell Dynamics, School of Medicine, Johns Hopkins University, 855 N. Wolfe St, 452 Rangos Bldg, Baltimore, MD 21205, USA; Department of Oncology, School of Medicine, Johns Hopkins University, 855 N. Wolfe St, 452 Rangos Bldg, Baltimore, MD 21205, USA.
| |
Collapse
|
47
|
Bertucci F, Finetti P, Vermeulen P, Van Dam P, Dirix L, Birnbaum D, Viens P, Van Laere S. Genomic profiling of inflammatory breast cancer: a review. Breast 2014; 23:538-45. [PMID: 24998451 DOI: 10.1016/j.breast.2014.06.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2013] [Revised: 06/02/2014] [Accepted: 06/08/2014] [Indexed: 01/04/2023] Open
Abstract
Inflammatory breast cancer (IBC) is a rare but aggressive form of breast cancer. Despite efforts in the past decade to delineate the molecular biology of IBC by applying high-throughput molecular profiling technologies to clinical samples, IBC remains insufficiently characterized. The reasons for that include limited sizes of the study population, heterogeneity with respect to the composition of the IBC and non-IBC control groups and technological differences across studies. In 2008, the World IBC Consortium was founded to foster collaboration between research groups focusing on IBC. One of the initial projects was to redefine the molecular profile of IBC using an unprecedented number of samples and search for gene signatures associated with survival and response to neo-adjuvant chemotherapy. Here, we provide an overview of all the molecular profiling studies that have been performed on IBC clinical samples to date.
Collapse
Affiliation(s)
- François Bertucci
- Département d'Oncologie Moléculaire, Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes and UMR1068 Inserm, Marseille, France; Département d'Oncologie Médicale, Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes and UMR1068 Inserm, Marseille, France; Faculté de Médecine, Université de la Méditerranée, Marseille, France.
| | - Pascal Finetti
- Département d'Oncologie Moléculaire, Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes and UMR1068 Inserm, Marseille, France
| | - Peter Vermeulen
- Translational Cancer Research Unit, GZA Hospitals Sint-Augustinus, Wilrijk, Belgium; Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Peter Van Dam
- Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Luc Dirix
- Translational Cancer Research Unit, GZA Hospitals Sint-Augustinus, Wilrijk, Belgium; Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Daniel Birnbaum
- Département d'Oncologie Moléculaire, Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes and UMR1068 Inserm, Marseille, France
| | - Patrice Viens
- Département d'Oncologie Médicale, Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes and UMR1068 Inserm, Marseille, France; Faculté de Médecine, Université de la Méditerranée, Marseille, France
| | - Steven Van Laere
- Translational Cancer Research Unit, GZA Hospitals Sint-Augustinus, Wilrijk, Belgium; Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium; Department of Oncology, KU Leuven, Leuven, Belgium.
| |
Collapse
|
48
|
Marsan M, Van den Eynden G, Limame R, Neven P, Hauspy J, Van Dam PA, Vergote I, Dirix LY, Vermeulen PB, Van Laere SJ. A core invasiveness gene signature reflects epithelial-to-mesenchymal transition but not metastatic potential in breast cancer cell lines and tissue samples. PLoS One 2014; 9:e89262. [PMID: 24586640 PMCID: PMC3931724 DOI: 10.1371/journal.pone.0089262] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 01/15/2014] [Indexed: 01/07/2023] Open
Abstract
Introduction Metastases remain the primary cause of cancer-related death. The acquisition of invasive tumour cell behaviour is thought to be a cornerstone of the metastatic cascade. Therefore, gene signatures related to invasiveness could aid in stratifying patients according to their prognostic profile. In the present study we aimed at identifying an invasiveness gene signature and investigated its biological relevance in breast cancer. Methods & Results We collected a set of published gene signatures related to cell motility and invasion. Using this collection, we identified 16 genes that were represented at a higher frequency than observed by coincidence, hereafter named the core invasiveness gene signature. Principal component analysis showed that these overrepresented genes were able to segregate invasive and non-invasive breast cancer cell lines, outperforming sets of 16 randomly selected genes (all P<0.001). When applied onto additional data sets, the expression of the core invasiveness gene signature was significantly elevated in cell lines forced to undergo epithelial-mesenchymal transition. The link between core invasiveness gene expression and epithelial-mesenchymal transition was also confirmed in a dataset consisting of 2420 human breast cancer samples. Univariate and multivariate Cox regression analysis demonstrated that CIG expression is not associated with a shorter distant metastasis free survival interval (HR = 0.956, 95%C.I. = 0.896–1.019, P = 0.186). Discussion These data demonstrate that we have identified a set of core invasiveness genes, the expression of which is associated with epithelial-mesenchymal transition in breast cancer cell lines and in human tissue samples. Despite the connection between epithelial-mesenchymal transition and invasive tumour cell behaviour, we were unable to demonstrate a link between the core invasiveness gene signature and enhanced metastatic potential.
Collapse
Affiliation(s)
- Melike Marsan
- Translational Cancer Research Unit, Oncology Center, GZA Hospitals Sint-Augustinus, Antwerp, Belgium
- Department of oncology, KU Leuven, Leuven, Belgium
- * E-mail:
| | - Gert Van den Eynden
- Translational Cancer Research Unit, Oncology Center, GZA Hospitals Sint-Augustinus, Antwerp, Belgium
| | - Ridha Limame
- Laboratory for Cancer Research and Clinical Oncology, University of Antwerp, Antwerp, Belgium
| | | | - Jan Hauspy
- Translational Cancer Research Unit, Oncology Center, GZA Hospitals Sint-Augustinus, Antwerp, Belgium
| | | | | | - Luc Y. Dirix
- Translational Cancer Research Unit, Oncology Center, GZA Hospitals Sint-Augustinus, Antwerp, Belgium
| | - Peter B. Vermeulen
- Translational Cancer Research Unit, Oncology Center, GZA Hospitals Sint-Augustinus, Antwerp, Belgium
| | - Steven J. Van Laere
- Translational Cancer Research Unit, Oncology Center, GZA Hospitals Sint-Augustinus, Antwerp, Belgium
- Department of oncology, KU Leuven, Leuven, Belgium
| |
Collapse
|
49
|
Adams A, van Brussel ASA, Vermeulen JF, Mali WPTM, van der Wall E, van Diest PJ, Elias SG. The potential of hypoxia markers as target for breast molecular imaging--a systematic review and meta-analysis of human marker expression. BMC Cancer 2013; 13:538. [PMID: 24206539 PMCID: PMC3903452 DOI: 10.1186/1471-2407-13-538] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Accepted: 10/23/2013] [Indexed: 02/07/2023] Open
Abstract
Background Molecular imaging of breast cancer is a promising emerging technology, potentially able to improve clinical care. Valid imaging targets for molecular imaging tracer development are membrane-bound hypoxia-related proteins, expressed when tumor growth outpaces neo-angiogenesis. We performed a systematic literature review and meta-analysis of such hypoxia marker expression rates in human breast cancer to evaluate their potential as clinically relevant molecular imaging targets. Methods We searched MEDLINE and EMBASE for articles describing membrane-bound proteins that are related to hypoxia inducible factor 1α (HIF-1α), the key regulator of the hypoxia response. We extracted expression rates of carbonic anhydrase-IX (CAIX), glucose transporter-1 (GLUT1), C-X-C chemokine receptor type-4 (CXCR4), or insulin-like growth factor-1 receptor (IGF1R) in human breast disease, evaluated by immunohistochemistry. We pooled study results using random-effects models and applied meta-regression to identify associations with clinicopathological variables. Results Of 1,705 identified articles, 117 matched our selection criteria, totaling 30,216 immunohistochemistry results. We found substantial between-study variability in expression rates. Invasive cancer showed pooled expression rates of 35% for CAIX (95% confidence interval (CI): 26-46%), 51% for GLUT1 (CI: 40-61%), 46% for CXCR4 (CI: 33-59%), and 46% for IGF1R (CI: 35-70%). Expression rates increased with tumor grade for GLUT1, CAIX, and CXCR4 (all p < 0.001), but decreased for IGF1R (p < 0.001). GLUT1 showed the highest expression rate in grade III cancers with 58% (45-69%). CXCR4 showed the highest expression rate in small T1 tumors with 48% (CI: 28-69%), but associations with size were only significant for CAIX (p < 0.001; positive association) and IGF1R (p = 0.047; negative association). Although based on few studies, CAIX, GLUT1, and CXCR4 showed profound lower expression rates in normal breast tissue and benign breast disease (p < 0.001), and high rates in carcinoma in situ. Invasive lobular carcinoma consistently showed lower expression rates (p < 0.001). Conclusions Our results support the potential of hypoxia-related markers as breast cancer molecular imaging targets. Although specificity is promising, combining targets would be necessary for optimal sensitivity. These data could help guide the choice of imaging targets for tracer development depending on the envisioned clinical application.
Collapse
Affiliation(s)
- Arthur Adams
- Department of Radiology, University Medical Center Utrecht, Utrecht, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
50
|
Yeh ED, Jacene HA, Bellon JR, Nakhlis F, Birdwell RL, Georgian-Smith D, Giess CS, Hirshfield-Bartek J, Overmoyer B, Van den Abbeele AD. What Radiologists Need to Know about Diagnosis and Treatment of Inflammatory Breast Cancer: A Multidisciplinary Approach. Radiographics 2013; 33:2003-17. [DOI: 10.1148/rg.337135503] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|