151
|
Goliwas KF, Miller LM, Marshall LE, Berry JL, Frost AR. Preparation and Analysis of In Vitro Three Dimensional Breast Carcinoma Surrogates. J Vis Exp 2016. [PMID: 27214165 DOI: 10.3791/54004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Three dimensional (3D) culture is a more physiologically relevant method to model cell behavior in vitro than two dimensional culture. Carcinomas, including breast carcinomas, are complex 3D tissues composed of cancer epithelial cells and stromal components, including fibroblasts and extracellular matrix (ECM). Yet most in vitro models of breast carcinoma consist only of cancer epithelial cells, omitting the stroma and, therefore, the 3D architecture of a tumor in vivo. Appropriate 3D modeling of carcinoma is important for accurate understanding of tumor biology, behavior, and response to therapy. However, the duration of culture and volume of 3D models is limited by the availability of oxygen and nutrients within the culture. Herein, we demonstrate a method in which breast carcinoma epithelial cells and stromal fibroblasts are incorporated into ECM to generate a 3D breast cancer surrogate that includes stroma and can be cultured as a solid 3D structure or by using a perfusion bioreactor system to deliver oxygen and nutrients. Following setup and an initial growth period, surrogates can be used for preclinical drug testing. Alternatively, the cellular and matrix components of the surrogate can be modified to address a variety of biological questions. After culture, surrogates are fixed and processed to paraffin, in a manner similar to the handling of clinical breast carcinoma specimens, for evaluation of parameters of interest. The evaluation of one such parameter, the density of cells present, is explained, where ImageJ and CellProfiler image analysis software systems are applied to photomicrographs of histologic sections of surrogates to quantify the number of nucleated cells per area. This can be used as an indicator of the change in cell number over time or the change in cell number resulting from varying growth conditions and treatments.
Collapse
Affiliation(s)
- Kayla F Goliwas
- Department of Pathology, University of Alabama at Birmingham
| | - Lindsay M Miller
- Department of Biomedical Engineering, University of Alabama at Birmingham
| | - Lauren E Marshall
- Department of Biomedical Engineering, University of Alabama at Birmingham
| | - Joel L Berry
- Department of Biomedical Engineering, University of Alabama at Birmingham
| | - Andra R Frost
- Department of Pathology, University of Alabama at Birmingham;
| |
Collapse
|
152
|
3D extracellular matrix interactions modulate tumour cell growth, invasion and angiogenesis in engineered tumour microenvironments. Acta Biomater 2016; 36:73-85. [PMID: 26971667 DOI: 10.1016/j.actbio.2016.03.017] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Revised: 02/23/2016] [Accepted: 03/09/2016] [Indexed: 12/15/2022]
Abstract
UNLABELLED Interactions between tumour cells and extracellular matrix proteins of the tumour microenvironment play crucial roles in cancer progression. So far, however, there are only a few experimental platforms available that allow us to study these interactions systematically in a mechanically defined three-dimensional (3D) context. Here, we have studied the effect of integrin binding motifs found within common extracellular matrix (ECM) proteins on 3D breast (MCF-7) and prostate (PC-3, LNCaP) cancer cell cultures, and co-cultures with endothelial and mesenchymal stromal cells. For this purpose, matrix metalloproteinase-degradable biohybrid poly(ethylene) glycol-heparin hydrogels were decorated with the peptide motifs RGD, GFOGER (collagen I), or IKVAV (laminin-111). Over 14days, cancer spheroids of 100-200μm formed. While the morphology of poorly invasive MCF-7 and LNCaP cells was not modulated by any of the peptide motifs, the aggressive PC-3 cells exhibited an invasive morphology when cultured in hydrogels comprising IKVAV and GFOGER motifs compared to RGD motifs or nonfunctionalised controls. PC-3 (but not MCF-7 and LNCaP) cell growth and endothelial cell infiltration were also significantly enhanced in IKVAV and GFOGER presenting gels. Taken together, we have established a 3D culture model that allows for dissecting the effect of biochemical cues on processes relevant to early cancer progression. These findings provide a basis for more mechanistic studies that may further advance our understanding of how ECM modulates cancer cell invasion and how to ultimately interfere with this process. STATEMENT OF SIGNIFICANCE Threedimensional in vitro cancer models have generated great interest over the past decade. However, most models are not suitable to systematically study the effects of environmental cues on cancer development and progression. To overcome this limitation, we have developed an innovative hydrogel platform to study the interactions between breast and prostate cancer cells and extracellular matrix ligands relevant to the tumour microenvironment. Our results show that hydrogels with laminin- and collagen-derived adhesive peptides induce a malignant phenotype in a cell-line specific manner. Thus, we have identified a method to control the incorporation of biochemical cues within a three dimensional culture model and anticipate that it will help us in better understanding the effects of the tumour microenvironment on cancer progression.
Collapse
|
153
|
Georgoudaki AM, Prokopec K, Boura V, Hellqvist E, Sohn S, Östling J, Dahan R, Harris R, Rantalainen M, Klevebring D, Sund M, Brage S, Fuxe J, Rolny C, Li F, Ravetch J, Karlsson M. Reprogramming Tumor-Associated Macrophages by Antibody Targeting Inhibits Cancer Progression and Metastasis. Cell Rep 2016; 15:2000-11. [DOI: 10.1016/j.celrep.2016.04.084] [Citation(s) in RCA: 293] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 03/02/2016] [Accepted: 04/21/2016] [Indexed: 01/05/2023] Open
|
154
|
Pulz LH, Barra CN, Kleeb SR, Xavier JG, Catão-Dias JL, Sobral RA, Fukumasu H, Strefezzi RF. Increased expression of tissue inhibitor of metalloproteinase-1 correlates with improved outcome in canine cutaneous mast cell tumours. Vet Comp Oncol 2016; 15:606-614. [PMID: 27041588 DOI: 10.1111/vco.12204] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 11/29/2015] [Indexed: 01/21/2023]
Abstract
Canine mast cell tumour (MCT) is a biologically heterogeneous disease. The extracellular matrix degradation promoted by matrix metalloproteinases (MMPs) has been studied in an attempt to elucidate the mechanisms involved in the biological behaviour of tumours. The aim of this study was to characterize the expression of MMP-2 and -9 and tissue inhibitors of metalloproteinase (TIMP)-1 and -2 in canine cutaneous MCTs and to evaluate their prognostic values. Immunohistochemical staining for MMP-2, MMP-9, TIMP-2 and TIMP-1 was performed in 46 canine cases of MCTs. TIMP-1 expression showed an independent prognostic value for post-surgical survival and disease-related mortality. Dogs with MCTs showing less than 22.9% mast cell TIMP-1 positivity were more prone to die because of the disease and had a shorter post-surgical survival. This article suggests the involvement of TIMP-1 in MCT progression, by contributing to a good outcome in patients with MCTs.
Collapse
Affiliation(s)
- L H Pulz
- Departamento de Patologia, Faculdade de Medicina Veterinária e Zootecnia, Universidade de São Paulo, São Paulo, Brazil.,Laboratório de Oncologia Comparada e Translacional, Faculdade de Zootecnia e Engenharia de Alimentos, Universidade de São Paulo, Pirassununga, Brazil
| | - C N Barra
- Departamento de Patologia, Faculdade de Medicina Veterinária e Zootecnia, Universidade de São Paulo, São Paulo, Brazil.,Laboratório de Oncologia Comparada e Translacional, Faculdade de Zootecnia e Engenharia de Alimentos, Universidade de São Paulo, Pirassununga, Brazil
| | - S R Kleeb
- Universidade Metodista de São Paulo, São Bernardo do Campo, Brazil
| | - J G Xavier
- Universidade Paulista, São Paulo, Brazil
| | - J L Catão-Dias
- Departamento de Patologia, Faculdade de Medicina Veterinária e Zootecnia, Universidade de São Paulo, São Paulo, Brazil
| | - R A Sobral
- Onco Cane Veterinária, São Paulo, Brazil
| | - H Fukumasu
- Laboratório de Oncologia Comparada e Translacional, Faculdade de Zootecnia e Engenharia de Alimentos, Universidade de São Paulo, Pirassununga, Brazil
| | - R F Strefezzi
- Laboratório de Oncologia Comparada e Translacional, Faculdade de Zootecnia e Engenharia de Alimentos, Universidade de São Paulo, Pirassununga, Brazil
| |
Collapse
|
155
|
Holle AW, Young JL, Spatz JP. In vitro cancer cell-ECM interactions inform in vivo cancer treatment. Adv Drug Deliv Rev 2016; 97:270-9. [PMID: 26485156 DOI: 10.1016/j.addr.2015.10.007] [Citation(s) in RCA: 142] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 10/05/2015] [Accepted: 10/11/2015] [Indexed: 02/07/2023]
Abstract
The general progression of cancer drug development involves in vitro testing followed by safety and efficacy evaluation in clinical trials. Due to the expense of bringing candidate drugs to trials, in vitro models of cancer cells and tumor biology are required to screen drugs. There are many examples of drugs exhibiting cytotoxic behavior in cancer cells in vitro but losing efficacy in vivo, and in many cases, this is the result of poorly understood chemoresistant effects conferred by the cancer microenvironment. To address this, improved methods for culturing cancer cells in biomimetic scaffolds have been developed; along the way, a great deal about the nature of cancer cell-extracellular matrix (ECM) interactions has been discovered. These discoveries will continue to be leveraged both in the development of novel drugs targeting these interactions and in the fabrication of biomimetic substrates for efficient cancer drug screening in vitro.
Collapse
|
156
|
The extracellular matrix in breast cancer. Adv Drug Deliv Rev 2016; 97:41-55. [PMID: 26743193 DOI: 10.1016/j.addr.2015.12.017] [Citation(s) in RCA: 271] [Impact Index Per Article: 33.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 12/18/2015] [Accepted: 12/19/2015] [Indexed: 12/31/2022]
Abstract
The extracellular matrix (ECM) is increasingly recognized as an important regulator in breast cancer. ECM in breast cancer development features numerous changes in composition and organization when compared to the mammary gland under homeostasis. Matrix proteins that are induced in breast cancer include fibrillar collagens, fibronectin, specific laminins and proteoglycans as well as matricellular proteins. Growing evidence suggests that many of these induced ECM proteins play a major functional role in breast cancer progression and metastasis. A number of the induced ECM proteins have moreover been shown to be essential components of metastatic niches, promoting stem/progenitor signaling pathways and metastatic growth. ECM remodeling enzymes are also markedly increased, leading to major changes in the matrix structure and biomechanical properties. Importantly, several ECM components and ECM remodeling enzymes are specifically induced in breast cancer or during tissue regeneration while healthy tissues under homeostasis express exceedingly low levels. This may indicate that ECM and ECM-associated functions may represent promising drug targets against breast cancer, providing important specificity that could be utilized when developing therapies.
Collapse
|
157
|
Buchsbaum RJ, Oh SY. Breast Cancer-Associated Fibroblasts: Where We Are and Where We Need to Go. Cancers (Basel) 2016; 8:cancers8020019. [PMID: 26828520 PMCID: PMC4773742 DOI: 10.3390/cancers8020019] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 01/12/2016] [Accepted: 01/20/2016] [Indexed: 02/04/2023] Open
Abstract
Cancers are heterogeneous tissues comprised of multiple components, including tumor cells and microenvironment cells. The tumor microenvironment has a critical role in tumor progression. The tumor microenvironment is comprised of various cell types, including fibroblasts, macrophages and immune cells, as well as extracellular matrix and various cytokines and growth factors. Fibroblasts are the predominant cell type in the tumor microenvironment. However, neither the derivation of tissue-specific cancer-associated fibroblasts nor markers of tissue-specific cancer-associated fibroblasts are well defined. Despite these uncertainties it is increasingly apparent that cancer-associated fibroblasts have a crucial role in tumor progression. In breast cancer, there is evolving evidence showing that breast cancer-associated fibroblasts are actively involved in breast cancer initiation, proliferation, invasion and metastasis. Breast cancer-associated fibroblasts also play a critical role in metabolic reprogramming of the tumor microenvironment and therapy resistance. This review summarizes the current understanding of breast cancer-associated fibroblasts.
Collapse
Affiliation(s)
- Rachel J Buchsbaum
- Molecular Oncology Research Institute and Department of Medicine, Division of Hematology-Oncology, Tufts Medical Center, Boston, MA 02111, USA.
| | - Sun Young Oh
- Department of Medicine, Division of Medical Oncology, Montefiore Medical Center, New York, NY 10467, USA.
| |
Collapse
|
158
|
Guo Z, Zhang T, Fang K, Dou J, Zhou N, Ma X, Gu N. The effects of macroporosity and stiffness of poly[(methyl vinyl ether)-alt-(maleic acid)] cross-linked egg white simulations of an aged extracellular matrix on the proliferation of ovarian cancer cells. RSC Adv 2016. [DOI: 10.1039/c6ra05134k] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The effects of macroporosity and stiffness of P(MVE-alt-MA) cross-linked EW simulations of an aged ECM on the proliferation of cancer cells.
Collapse
Affiliation(s)
- Zhenchao Guo
- State Key Laboratory of Bioelectronics
- Jiangsu Key Laboratory for Biomaterials and Devices
- School of Biological Science and Medical Engineering & Collaborative Innovation Center of Suzhou Nano Science and Technology
- Southeast University
- Nanjing 210096
| | - Tianzhu Zhang
- State Key Laboratory of Bioelectronics
- Jiangsu Key Laboratory for Biomaterials and Devices
- School of Biological Science and Medical Engineering & Collaborative Innovation Center of Suzhou Nano Science and Technology
- Southeast University
- Nanjing 210096
| | - Kun Fang
- State Key Laboratory of Bioelectronics
- Jiangsu Key Laboratory for Biomaterials and Devices
- School of Biological Science and Medical Engineering & Collaborative Innovation Center of Suzhou Nano Science and Technology
- Southeast University
- Nanjing 210096
| | - Jun Dou
- Medical School
- Southeast University
- Nanjing 210009
- China
| | - Naizhen Zhou
- State Key Laboratory of Bioelectronics
- Jiangsu Key Laboratory for Biomaterials and Devices
- School of Biological Science and Medical Engineering & Collaborative Innovation Center of Suzhou Nano Science and Technology
- Southeast University
- Nanjing 210096
| | - Xiaoe Ma
- State Key Laboratory of Bioelectronics
- Jiangsu Key Laboratory for Biomaterials and Devices
- School of Biological Science and Medical Engineering & Collaborative Innovation Center of Suzhou Nano Science and Technology
- Southeast University
- Nanjing 210096
| | - Ning Gu
- State Key Laboratory of Bioelectronics
- Jiangsu Key Laboratory for Biomaterials and Devices
- School of Biological Science and Medical Engineering & Collaborative Innovation Center of Suzhou Nano Science and Technology
- Southeast University
- Nanjing 210096
| |
Collapse
|
159
|
Keire PA, Bressler SL, Mulvihill ER, Starcher BC, Kang I, Wight TN. Inhibition of versican expression by siRNA facilitates tropoelastin synthesis and elastic fiber formation by human SK-LMS-1 leiomyosarcoma smooth muscle cells in vitro and in vivo. Matrix Biol 2015; 50:67-81. [PMID: 26723257 DOI: 10.1016/j.matbio.2015.12.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 12/17/2015] [Accepted: 12/19/2015] [Indexed: 12/23/2022]
Abstract
Versican is an extracellular matrix (ECM) molecule that interacts with other ECM components to influence ECM organization, stability, composition, and cell behavior. Versican is known to increase in a number of cancers, but little is known about how versican influences the amount and organization of the ECM components in the tumor microenvironment. In the present study, we modulated versican expression using siRNAs in the human leiomyosarcoma (LMS) smooth muscle cell line SK-LMS-1, and observed the formation of elastin and elastic fibers in vitro and also in vivo in a nude mouse tumor model. Constitutive siRNA-directed knockdown of versican in LMS cells resulted in increased levels of elastin, as shown by immunohistochemical staining of the cells in vitro, and by mRNA and protein analyses. Moreover, versican siRNA LMS cells, when injected into nude mice, generated smaller tumors that had significantly greater immunohistochemical and histochemical staining for elastin when compared to control tumors. Additionally, microarray analyses were used to determine the influence of versican isoform modulation on gene expression profiles, and to identify genes that influence and relate to the process of elastogenesis. cDNA microarray analysis and TaqMan low density array validation identified previously unreported genes associated with downregulation of versican and increased elastogenesis. These results highlight an important role for the proteoglycan versican in regulating the expression and assembly of elastin and the phenotype of LMS cells.
Collapse
Affiliation(s)
- Paul A Keire
- Matrix Biology Program, Benaroya Research Institute, 1201 Ninth Avenue, Seattle, WA 98101, USA; Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Steven L Bressler
- Matrix Biology Program, Benaroya Research Institute, 1201 Ninth Avenue, Seattle, WA 98101, USA
| | - Eileen R Mulvihill
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Barry C Starcher
- Department of Biochemistry, University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA
| | - Inkyung Kang
- Matrix Biology Program, Benaroya Research Institute, 1201 Ninth Avenue, Seattle, WA 98101, USA
| | - Thomas N Wight
- Matrix Biology Program, Benaroya Research Institute, 1201 Ninth Avenue, Seattle, WA 98101, USA; Department of Pathology, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
160
|
The extracellular matrix in breast cancer predicts prognosis through composition, splicing, and crosslinking. Exp Cell Res 2015; 343:73-81. [PMID: 26597760 DOI: 10.1016/j.yexcr.2015.11.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 11/11/2015] [Indexed: 12/19/2022]
Abstract
The extracellular matrix in the healthy breast has an important tumor suppressive role, whereas the abnormal ECM in tumors can promote aggressiveness, and has been linked to breast cancer relapse, survival and resistance to chemotherapy. This review article gives an overview of the elements of the ECM which have been linked to prognosis of breast cancers, including changes in ECM protein composition, splicing, and microstructure.
Collapse
|
161
|
Giussani M, Merlino G, Cappelletti V, Tagliabue E, Daidone MG. Tumor-extracellular matrix interactions: Identification of tools associated with breast cancer progression. Semin Cancer Biol 2015; 35:3-10. [PMID: 26416466 DOI: 10.1016/j.semcancer.2015.09.012] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 09/23/2015] [Indexed: 12/18/2022]
Abstract
Several evidences support the concept that cancer development and progression are not entirely cancer cell-autonomous processes, but may be influenced, and possibly driven, by cross-talk between cancer cells and the surrounding microenvironment in which, besides immune cells, stromal cells and extracellular matrix (ECM) play a major role in regulating distinct biologic processes. Stroma and ECM-related signatures proved to influence breast cancer progression, and to contribute to the identification of tumor phenotypes resistant to cytotoxic and hormonal treatments. The possible clinical implications of the interplay between tumor cells and the microenvironment, with special reference to ECM remodelling, will be discussed in this review.
Collapse
Affiliation(s)
- Marta Giussani
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via G.A. Amadeo, 42, 20133 Milan, Italy.
| | - Giuseppe Merlino
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via G.A. Amadeo, 42, 20133 Milan, Italy.
| | - Vera Cappelletti
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via G.A. Amadeo, 42, 20133 Milan, Italy.
| | - Elda Tagliabue
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via G.A. Amadeo, 42, 20133 Milan, Italy.
| | - Maria Grazia Daidone
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via G.A. Amadeo, 42, 20133 Milan, Italy.
| |
Collapse
|
162
|
Gandellini P, Andriani F, Merlino G, D'Aiuto F, Roz L, Callari M. Complexity in the tumour microenvironment: Cancer associated fibroblast gene expression patterns identify both common and unique features of tumour-stroma crosstalk across cancer types. Semin Cancer Biol 2015; 35:96-106. [PMID: 26320408 DOI: 10.1016/j.semcancer.2015.08.008] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Revised: 08/17/2015] [Accepted: 08/21/2015] [Indexed: 12/21/2022]
Abstract
Cancer is a complex disease, driven by the accumulation of several somatic aberrations but fostered by a two-way interaction between tumour cells and the surrounding microenvironment. Cancer associated fibroblasts (CAFs) represent one of the major players in tumour-stroma crosstalk. Recent in vitro and in vivo studies, often conducted by employing high throughput approaches, have started unravelling the key pathways involved in their functional effects. This review focus on open challenges in the study of CAF properties and function, highlighting at the same time the existence of common mechanisms as well as peculiarities in different cancer types (breast, prostate and lung cancer). Although still limited by current experimental models, which are unable to deal with the full level of complexity of the tumour microenvironment, a better understanding of these mechanisms may enable the identification of new biomarkers and therapeutic targets, to improve current strategies for cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Paolo Gandellini
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Francesca Andriani
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Giuseppe Merlino
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Francesca D'Aiuto
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Luca Roz
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Maurizio Callari
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
| |
Collapse
|
163
|
Kimbung S, Johansson I, Danielsson A, Veerla S, Egyhazi Brage S, Frostvik Stolt M, Skoog L, Carlsson L, Einbeigi Z, Lidbrink E, Linderholm B, Loman N, Malmström PO, Söderberg M, Walz TM, Fernö M, Hatschek T, Hedenfalk I. Transcriptional Profiling of Breast Cancer Metastases Identifies Liver Metastasis-Selective Genes Associated with Adverse Outcome in Luminal A Primary Breast Cancer. Clin Cancer Res 2015; 22:146-57. [PMID: 26276891 DOI: 10.1158/1078-0432.ccr-15-0487] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 07/26/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE The complete molecular basis of the organ-specificity of metastasis is elusive. This study aimed to provide an independent characterization of the transcriptional landscape of breast cancer metastases with the specific objective to identify liver metastasis-selective genes of prognostic importance following primary tumor diagnosis. EXPERIMENTAL DESIGN A cohort of 304 women with advanced breast cancer was studied. Associations between the site of recurrence and clinicopathologic features were investigated. Fine-needle aspirates of metastases (n = 91) were subjected to whole-genome transcriptional profiling. Liver metastasis-selective genes were identified by significance analysis of microarray (SAM) analyses and independently validated in external datasets. Finally, the prognostic relevance of the liver metastasis-selective genes in primary breast cancer was tested. RESULTS Liver relapse was associated with estrogen receptor (ER) expression (P = 0.002), luminal B subtype (P = 0.01), and was prognostic for an inferior postrelapse survival (P = 0.01). The major variation in the transcriptional landscape of metastases was also associated with ER expression and molecular subtype. However, liver metastases displayed unique transcriptional fingerprints, characterized by downregulation of extracellular matrix (i.e., stromal) genes. Importantly, we identified a 17-gene liver metastasis-selective signature, which was significantly and independently prognostic for shorter relapse-free (P < 0.001) and overall (P = 0.001) survival in ER-positive tumors. Remarkably, this signature remained independently prognostic for shorter relapse-free survival (P = 0.001) among luminal A tumors. CONCLUSIONS Extracellular matrix (stromal) genes can be used to partition breast cancer by site of relapse and may be used to further refine prognostication in ER positive primary breast cancer.
Collapse
Affiliation(s)
- Siker Kimbung
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund, Lund University, Sweden. CREATE Health Strategic Center for Translational Cancer Research, Lund University, Lund, Sweden
| | - Ida Johansson
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund, Lund University, Sweden. CREATE Health Strategic Center for Translational Cancer Research, Lund University, Lund, Sweden
| | - Anna Danielsson
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Srinivas Veerla
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund, Lund University, Sweden. CREATE Health Strategic Center for Translational Cancer Research, Lund University, Lund, Sweden
| | - Suzanne Egyhazi Brage
- Department of Oncology and Pathology, Karolinska Institutet and Karolinska University Hospital, Sweden
| | - Marianne Frostvik Stolt
- Department of Oncology and Pathology, Karolinska Institutet and Karolinska University Hospital, Sweden
| | - Lambert Skoog
- Department of Oncology and Pathology, Karolinska Institutet and Karolinska University Hospital, Sweden
| | - Lena Carlsson
- Department of Oncology, Sundsvall Hospital, Sundsvall, Sweden
| | - Zakaria Einbeigi
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Elisabet Lidbrink
- Department of Oncology and Pathology, Karolinska Institutet and Karolinska University Hospital, Sweden
| | - Barbro Linderholm
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Niklas Loman
- Department of Oncology, Skåne University Hospital, Lund/Malmö, Sweden
| | | | - Martin Söderberg
- Department of Oncology, Skåne University Hospital, Lund/Malmö, Sweden
| | - Thomas M Walz
- Division of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden
| | - Mårten Fernö
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund, Lund University, Sweden
| | - Thomas Hatschek
- Department of Oncology and Pathology, Karolinska Institutet and Karolinska University Hospital, Sweden
| | - Ingrid Hedenfalk
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund, Lund University, Sweden. CREATE Health Strategic Center for Translational Cancer Research, Lund University, Lund, Sweden.
| | | |
Collapse
|
164
|
Sun Y. Tumor microenvironment and cancer therapy resistance. Cancer Lett 2015; 380:205-15. [PMID: 26272180 DOI: 10.1016/j.canlet.2015.07.044] [Citation(s) in RCA: 224] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2015] [Revised: 07/08/2015] [Accepted: 07/16/2015] [Indexed: 12/19/2022]
Abstract
Innate resistance to various therapeutic interventions is a hallmark of cancer. In recent years, however, acquired resistance has emerged as a daunting challenge to anticancer treatments including chemotherapy, radiation and targeted therapy, which abolishes the efficacy of otherwise successful regimens. Cancer cells gain resistance through a variety of mechanisms in both primary and metastatic sites, involving cell intrinsic and extrinsic factors, but the latter often remains overlooked. Mounting evidence suggests critical roles played by the tumor microenvironment (TME) in multiple aspects of cancer progression particularly therapeutic resistance. The TME decreases drug penetration, confers proliferative and antiapoptotic advantages to surviving cells, facilitates resistance without causing genetic mutations and epigenetic changes, collectively modifying disease modality and distorting clinical indices. Recent studies have set the baseline for future investigation on the intricate relationship between cancer resistance and the TME in pathological backgrounds. This review provides an updated outline of research advances in TME biology and highlights the prospect of targeting the TME as an essential strategy to overcome cancer resistance and improve therapeutic outcomes through precise intervention. In the long run, continued inputs into translational medicine remain highly desired to achieve durable responses in the current era of personalized clinical oncology.
Collapse
Affiliation(s)
- Yu Sun
- Key Lab of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiaotong University School of Medicine, Shanghai 200031, China; Collaborative Innovation Center of Systems Biomedicine, Shanghai Jiaotong University School of Medicine, Shanghai 200240, China; VA Seattle Medical Center, Seattle, WA 98108, USA; Department of Medicine, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
165
|
Abstract
PURPOSE OF REVIEW Combinatorial strategies in cancer medicine will not only target cancer cell-intrinsic pathways, but also cancer cell-extrinsic cells, pathways, and mediators of the tumor microenvironment. The aim of the present review is to define the roles of the tumor microenvironment in primary and metastatic breast cancer progression. RECENT FINDINGS The cancer microenvironment is composed of nontransformed host stromal cells, such as endothelial cells, fibroblasts, various immune cells, and a complex extracellular matrix secreted by both the normal and neoplastic cells embedded in it. The stromal constituents contribute to the core and emergent hallmarks of cancer. In particular, they can boost sustaining proliferative signaling, evading growth suppressors, resisting cell death, enabling replicative immortality, inducing angiogenesis, activating invasion and metastasis, reprogramming energy metabolism, and evading immune destruction. SUMMARY The stromal cells play a role in enabling or enhancing multiple hallmark capabilities in tumor microenvironment. This is a background for therapeutic-targeting strategies aimed to abrogate the stroma's contribution. Targeting tumor-associated fibroblasts, macrophages, angiogenesis and enhancing immune response may represent a paradigm-shifting approach to treating human cancer in the near future.
Collapse
|
166
|
Abstract
Traditionally, intertumour heterogeneity in breast cancer has been documented in terms of different histological subtypes, treatment sensitivity profiles, and clinical outcomes among different patients. Results of high-throughput molecular profiling studies have subsequently revealed the true extent of this heterogeneity. Further complicating this scenario, the heterogeneous expression of the oestrogen receptor (ER), progesterone receptor (PR), and HER2 has been reported in different areas of the same tumour. Furthermore, discordance, in terms of ER, PR and HER2 expression, has also been reported between primary tumours and their matched metastatic lesions. High-throughput molecular profiling studies have confirmed that spatial and temporal intratumour heterogeneity of breast cancers exist at a level beyond common expectations. We describe the different levels of tumour heterogeneity, and discuss the strategies that can be adopted by clinicians to tackle treatment response and resistance issues associated with such heterogeneity, including a rationally selected combination of agents that target driver mutations, the targeting of deleterious passenger mutations, identifying and eradicating the 'lethal' clone, targeting the tumour microenvironment, or using adaptive treatments and immunotherapy. The identification of the most-appropriate strategies and their implementation in the clinic will prove highly challenging and necessitate the adoption of radically new practices for the optimal clinical management of breast malignancies.
Collapse
Affiliation(s)
- Dimitrios Zardavas
- Breast International Group (BIG)-aisbl c/o Jules Bordet Institute, Boulevard de Waterloo 121, 1000 Brussels, Belgium
| | - Alexandre Irrthum
- Breast International Group (BIG)-aisbl c/o Jules Bordet Institute, Boulevard de Waterloo 121, 1000 Brussels, Belgium
| | - Charles Swanton
- University College London Cancer Institute, Cancer Research UK Lung Cancer Centre of Excellence, Paul O'Gorman Building, Huntley Street, London WC1E 6DD, UK
| | - Martine Piccart
- Jules Bordet Institute, Boulevard de Waterloo 121, 1000 Brussels, Belgium
| |
Collapse
|
167
|
Oh EY, Christensen SM, Ghanta S, Jeong JC, Bucur O, Glass B, Montaser-Kouhsari L, Knoblauch NW, Bertos N, Saleh SM, Haibe-Kains B, Park M, Beck AH. Extensive rewiring of epithelial-stromal co-expression networks in breast cancer. Genome Biol 2015; 16:128. [PMID: 26087699 PMCID: PMC4471934 DOI: 10.1186/s13059-015-0675-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 05/13/2015] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Epithelial-stromal crosstalk plays a critical role in invasive breast cancer pathogenesis; however, little is known on a systems level about how epithelial-stromal interactions evolve during carcinogenesis. RESULTS We develop a framework for building genome-wide epithelial-stromal co-expression networks composed of pairwise co-expression relationships between mRNA levels of genes expressed in the epithelium and stroma across a population of patients. We apply this method to laser capture micro-dissection expression profiling datasets in the setting of breast carcinogenesis. Our analysis shows that epithelial-stromal co-expression networks undergo extensive rewiring during carcinogenesis, with the emergence of distinct network hubs in normal breast, and estrogen receptor-positive and estrogen receptor-negative invasive breast cancer, and the emergence of distinct patterns of functional network enrichment. In contrast to normal breast, the strongest epithelial-stromal co-expression relationships in invasive breast cancer mostly represent self-loops, in which the same gene is co-expressed in epithelial and stromal regions. We validate this observation using an independent laser capture micro-dissection dataset and confirm that self-loop interactions are significantly increased in cancer by performing computational image analysis of epithelial and stromal protein expression using images from the Human Protein Atlas. CONCLUSIONS Epithelial-stromal co-expression network analysis represents a new approach for systems-level analyses of spatially localized transcriptomic data. The analysis provides new biological insights into the rewiring of epithelial-stromal co-expression networks and the emergence of epithelial-stromal co-expression self-loops in breast cancer. The approach may facilitate the development of new diagnostics and therapeutics targeting epithelial-stromal interactions in cancer.
Collapse
Affiliation(s)
- Eun-Yeong Oh
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Boston, MA, 02215, USA. .,Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA. .,Harvard Medical School, Boston, MA, 02215, USA.
| | - Stephen M Christensen
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Boston, MA, 02215, USA. .,Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA. .,Harvard Medical School, Boston, MA, 02215, USA.
| | - Sindhu Ghanta
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Boston, MA, 02215, USA. .,Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA. .,Harvard Medical School, Boston, MA, 02215, USA.
| | - Jong Cheol Jeong
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Boston, MA, 02215, USA. .,Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA. .,Harvard Medical School, Boston, MA, 02215, USA.
| | - Octavian Bucur
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Boston, MA, 02215, USA. .,Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA. .,Harvard Medical School, Boston, MA, 02215, USA.
| | - Benjamin Glass
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Boston, MA, 02215, USA. .,Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA. .,Harvard Medical School, Boston, MA, 02215, USA.
| | - Laleh Montaser-Kouhsari
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Boston, MA, 02215, USA. .,Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA. .,Harvard Medical School, Boston, MA, 02215, USA.
| | - Nicholas W Knoblauch
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Boston, MA, 02215, USA. .,Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA. .,Harvard Medical School, Boston, MA, 02215, USA.
| | - Nicholas Bertos
- Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada.
| | - Sadiq Mi Saleh
- Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada.
| | - Benjamin Haibe-Kains
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 1L7, Canada. .,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, M5G 1L7, Canada.
| | - Morag Park
- Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada.
| | - Andrew H Beck
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Boston, MA, 02215, USA. .,Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA. .,Harvard Medical School, Boston, MA, 02215, USA.
| |
Collapse
|
168
|
Abstract
The aim of the present review is to survey the accumulated knowledge on the extracellular matrix (ECM) of tumors referring to its putative utility as therapeutic target. Following the traditional observation on the extensive morphological alteration in the tumor-affected tissue, the well-documented aberrant cellular regulation indicated that ECM components have an active role in tumor progression. However, due to the diverse functions and variable expression of proteoglycans, matrix proteins, and integrins, it is rather difficult to identify a comprehensive therapeutic target among ECM components. At present, the elevated level of heparanase and the prominent expression of αvβ5 integrin are considered as promising therapeutic targets. The inhibition of glycosaminoglycan offers another promising approach in the treatment of those tumors which are stimulated by proteoglycans. It can be ascertained that a selective ECM inhibitor would be a great asset to control metastasis driven by ECM-mediated signaling.
Collapse
Affiliation(s)
- Revekka Harisi
- 1st Institute of Pathology and Experimental Cancer Research, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Andras Jeney
- 1st Institute of Pathology and Experimental Cancer Research, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| |
Collapse
|
169
|
Acquisition of epithelial-mesenchymal transition phenotype in the tamoxifen-resistant breast cancer cell: a new role for G protein-coupled estrogen receptor in mediating tamoxifen resistance through cancer-associated fibroblast-derived fibronectin and β1-integrin signaling pathway in tumor cells. Breast Cancer Res 2015; 17:69. [PMID: 25990368 PMCID: PMC4453053 DOI: 10.1186/s13058-015-0579-y] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2015] [Accepted: 05/11/2015] [Indexed: 12/21/2022] Open
Abstract
INTRODUCTION Acquired tamoxifen resistance remains the major obstacle to breast cancer endocrine therapy. β1-integrin was identified as one of the target genes of G protein-coupled estrogen receptor (GPER), a novel estrogen receptor recognized as an initiator of tamoxifen resistance. Here, we investigated the role of β1-integrin in GPER-mediated tamoxifen resistance in breast cancer. METHODS The expression of β1-integrin and biomarkers of epithelial-mesenchymal transition were evaluated immunohistochemically in 53 specimens of metastases and paired primary tumors. The function of β1-integrin was investigated in tamoxifen-resistant (MCF-7R) subclones, derived from parental MCF-7 cells, and MCF-7R β1-integrin-silenced subclones in MTT and Transwell assays. Involved signaling pathways were identified using specific inhibitors and Western blotting analysis. RESULTS GPER, β1-integrin and mesenchymal biomarkers (vimentin and fibronectin) expression in metastases increased compared to the corresponding primary tumors; a close expression pattern of β1-integrin and GPER were in metastases. Increased β1-integrin expression was also confirmed in MCF-7R cells compared with MCF-7 cells. This upregulation of β1-integrin was induced by agonists of GPER and blocked by both antagonist and knockdown of it in MCF-7R cells. Moreover, the epidermal growth factor receptor/extracellular regulated protein kinase (EGFR/ERK) signaling pathway was involved in this transcriptional regulation since specific inhibitors of these kinases also reduced the GPER-induced upregulation of β1-integrin. Interestingly, silencing of β1-integrin partially rescued the sensitivity of MCF-7R cells to tamoxifen and the α5β1-integrin subunit is probably responsible for this phenomenon. Importantly, the cell migration and epithelial-mesenchymal transition induced by cancer-associated fibroblasts, or the product of cancer-associated fibroblasts, fibronectin, were reduced by knockdown of β1-integrin in MCF-7R cells. In addition, the downstream kinases of β1-integrin including focal adhesion kinase, Src and AKT were activated in MCF-7R cells and may be involved in the interaction between cancer cells and cancer-associated fibroblasts. CONCLUSIONS GPER/EGFR/ERK signaling upregulates β1-integrin expression and activates downstream kinases, which contributes to cancer-associated fibroblast-induced cell migration and epithelial-mesenchymal transition, in MCF-7R cells. GPER probably contributes to tamoxifen resistance via interaction with the tumor microenvironment in a β1-integrin-dependent pattern. Thus, β1-integrin may be a potential target to improve anti-hormone therapy responses in breast cancer patients.
Collapse
|
170
|
Giussani M, De Maria C, Michele V, Montemurro F, Triulzi T, Tagliabue E, Gelfi C, Vozzig G. Biomimicking of the Breast Tumor Microenvironment. ACTA ACUST UNITED AC 2015. [DOI: 10.1007/s40610-015-0014-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
171
|
Huang X, Dugo M, Callari M, Sandri M, De Cecco L, Valeri B, Carcangiu ML, Xue J, Bi R, Veneroni S, Daidone MG, Ménard S, Tagliabue E, Shao Z, Wu J, Orlandi R. Molecular portrait of breast cancer in China reveals comprehensive transcriptomic likeness to Caucasian breast cancer and low prevalence of luminal A subtype. Cancer Med 2015; 4:1016-30. [PMID: 25787708 PMCID: PMC4529340 DOI: 10.1002/cam4.442] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 12/29/2014] [Accepted: 01/13/2015] [Indexed: 01/02/2023] Open
Abstract
The recent dramatic increase in breast cancer incidence across China with progressive urbanization and economic development has signaled the urgent need for molecular and clinical detailing of breast cancer in the Chinese population. Our analyses of a unique transethnic collection of breast cancer frozen specimens from Shanghai Fudan Cancer Center (Chinese Han) profiled simultaneously with an analogous Caucasian Italian series revealed consistent transcriptomic data lacking in batch effects. The prevalence of Luminal A subtype was significantly lower in Chinese series, impacting the overall prevalence of estrogen receptor (ER)-positive disease in a large cohort of Chinese/Caucasian patients. Unsupervised and supervised comparison of gene and microRNA (miRNA) profiles of Chinese and Caucasian samples revealed extensive similarity in the comprehensive taxonomy of transcriptional elements regulating breast cancer biology. Partition of gene expression data using gene lists relevant to breast cancer as "intrinsic" and "extracellular matrix" genes identified Chinese and Caucasian subgroups with equivalent global gene and miRNA profiles. These findings indicate that in the Chinese and Caucasian groups, breast neoplasia and the surrounding stromal characteristics undergo the same differentiation and molecular processes. Transcriptional similarity across transethnic cohorts may simplify translational medicine approaches and clinical management of breast cancer patients worldwide.
Collapse
Affiliation(s)
- Xiaoyan Huang
- Department of Breast Surgery, Fudan University Shanghai Cancer Center and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Matteo Dugo
- Functional Genomics and Bioinformatics, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Maurizio Callari
- Biomarkers Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Marco Sandri
- Molecular Targeting Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Loris De Cecco
- Functional Genomics and Bioinformatics, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Barbara Valeri
- Department of Pathology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Maria Luisa Carcangiu
- Department of Pathology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Jingyan Xue
- Department of Breast Surgery, Fudan University Shanghai Cancer Center and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Rui Bi
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Silvia Veneroni
- Biomarkers Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Maria Grazia Daidone
- Biomarkers Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Sylvie Ménard
- Molecular Targeting Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Elda Tagliabue
- Molecular Targeting Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Zhimin Shao
- Department of Breast Surgery, Fudan University Shanghai Cancer Center and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Jiong Wu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Rosaria Orlandi
- Molecular Targeting Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
172
|
Wang ZL, Sun L, Li Y, Li N. Relationship between elasticity and collagen fiber content in breast disease: a preliminary report. ULTRASONICS 2015; 57:44-49. [PMID: 25465961 DOI: 10.1016/j.ultras.2014.10.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2014] [Revised: 10/13/2014] [Accepted: 10/20/2014] [Indexed: 06/04/2023]
Abstract
OBJECTIVE To investigate the differences in elasticity and collagen fiber content between malignant and benign breast lesions, and to study the relationship between shear wave elasticity and the content of collagen fiber in extracellular matrix (ECM). MATERIALS AND METHODS Between May 2012 to May 2013, 106 patients with 116 breast lesions who were referred to our center for ultrasound-guided biopsy of a sonographically apparent breast lesion underwent shear wave elasticity examination. The specimen underwent Van Gieson (VG) dye and Image-Pro Plus 5.1 software was used to quantitatively analyze area of collagen fiber. RESULTS Malignant lesions exhibited significantly higher maximum elasticity, mean elasticity, and elasticity ratio between lesions and surrounding parenchyma (140.43±70.16 kPa, 63.11±33.68 kPa, 3.49±1.95) than benign lesions (54.64±48.53 kPa, 34.52±25.23 kPa, 2.25±1.48) (t=5.329, t=4.382, t=4.487, P<0.001). The content of collagen fiber of malignant lesions was significantly higher than that of benign lesions (t=8.437, p=0.000). There was a positive correlation between max elasticity and the content of fiber collagen (r=0.746). CONCLUSION The elasticity of breast lesions has a close correlation with the content of collagen fiber, which might have an important impact on tissue stiffness of breast lesions.
Collapse
Affiliation(s)
- Zhi Li Wang
- Department of Ultrasound, Chinese People's Liberation Army General Hospital, 28 Fuxing Road, Beijing 100853, China.
| | - Lu Sun
- Department of Pathology, Chinese People's Liberation Army General Hospital, 28 Fuxing Road, Beijing 100853, China
| | - Ye Li
- Department of Ultrasound, Chinese People's Liberation Army General Hospital, 28 Fuxing Road, Beijing 100853, China
| | - Nan Li
- Department of Ultrasound, Chinese People's Liberation Army General Hospital, 28 Fuxing Road, Beijing 100853, China
| |
Collapse
|
173
|
Hopkins AM, DeSimone E, Chwalek K, Kaplan DL. 3D in vitro modeling of the central nervous system. Prog Neurobiol 2015; 125:1-25. [PMID: 25461688 PMCID: PMC4324093 DOI: 10.1016/j.pneurobio.2014.11.003] [Citation(s) in RCA: 148] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2014] [Revised: 10/12/2014] [Accepted: 11/15/2014] [Indexed: 12/15/2022]
Abstract
There are currently more than 600 diseases characterized as affecting the central nervous system (CNS) which inflict neural damage. Unfortunately, few of these conditions have effective treatments available. Although significant efforts have been put into developing new therapeutics, drugs which were promising in the developmental phase have high attrition rates in late stage clinical trials. These failures could be circumvented if current 2D in vitro and in vivo models were improved. 3D, tissue-engineered in vitro systems can address this need and enhance clinical translation through two approaches: (1) bottom-up, and (2) top-down (developmental/regenerative) strategies to reproduce the structure and function of human tissues. Critical challenges remain including biomaterials capable of matching the mechanical properties and extracellular matrix (ECM) composition of neural tissues, compartmentalized scaffolds that support heterogeneous tissue architectures reflective of brain organization and structure, and robust functional assays for in vitro tissue validation. The unique design parameters defined by the complex physiology of the CNS for construction and validation of 3D in vitro neural systems are reviewed here.
Collapse
Affiliation(s)
- Amy M Hopkins
- Department of Biomedical Engineering, Tufts University, Science & Technology Center, 4 Colby Street, Medford, MA 02155, USA
| | - Elise DeSimone
- Department of Biomedical Engineering, Tufts University, Science & Technology Center, 4 Colby Street, Medford, MA 02155, USA
| | - Karolina Chwalek
- Department of Biomedical Engineering, Tufts University, Science & Technology Center, 4 Colby Street, Medford, MA 02155, USA
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Science & Technology Center, 4 Colby Street, Medford, MA 02155, USA.
| |
Collapse
|
174
|
Eiró N, Fernandez-Garcia B, Vázquez J, Del Casar JM, González LO, Vizoso FJ. A phenotype from tumor stroma based on the expression of metalloproteases and their inhibitors, associated with prognosis in breast cancer. Oncoimmunology 2015; 4:e992222. [PMID: 26140253 DOI: 10.4161/2162402x.2014.992222] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Revised: 11/11/2014] [Accepted: 11/22/2014] [Indexed: 01/17/2023] Open
Abstract
The objective of the present work was to evaluate the impact of the phenotype of both mononuclear inflammatory cells (MICs) and cancer-associated fibroblast (CAFs) in early breast cancer patients, specifically assessed as to their expression of MMP/TIMP relative to their position within the tumor (i.e., localization at the tumor center or invasive front) and the occurrence of distant metastases.. An immunohistochemical study was performed using tissue arrays and specific antibodies against matrix metalloproteinase (MMP)-1, -2, -7, -9, -11, -13 and -14, tissue inhibitors of metalloproteinase (TIMP)-1, -2 and -3, both at tumor center and at invasive front, in 107 patients with primary ductal invasive breast tumors. Data were analyzed by unsupervised hierarchical clustering analysis. Our results indicated that MMP-11 expression by MICs, and TIMP-2 expression by CAFs at either the tumor center or the invasive front, were the most potent independent prognostic factors for predicting the clinical outcome of patients. Using the unsupervised hierarchical clustering analysis, we found well-defined clusters of cases identifying subgroups of tumors showing a high molecular profile of MMPs/TIMPs expression by stromal cells (CAFs and MICs), both at the tumor center and at the invasive front, which were strongly associated with a higher prevalence of distant metastasis. In addition, we found combinations of these clusters defining subpopulations of breast carcinomas differing widely in their clinical outcome. The results presented here identify biologic markers useful to categorize patients into different subgroups based on their tumor stroma, which may contribute to improved understanding of the prognosis of breast cancer patients.
Collapse
Key Words
- Breast cancer
- CAF, cancer-associated fibroblast
- CI, confidence interval
- ECM, extracellular matrix
- EGF, epidermal growth factor
- EMT, epithelial-mesenchymal transition
- ER, estrogen receptor
- HER2, human epidermal growth factor receptor 2
- HGF, hepatocyte growth factor
- IGFBPs, IGF binding protein
- IGFs, insulin growth factors
- IL, interleukin
- MIC, mononuclear inflammatory cell
- MMP, matrix metalloprotease
- NFkB, nuclear factor kappa B
- PgR, progesterone receptor, TA, tissue array
- TGFß, transforming growth factor ß
- TIMP, tissue inhibitors of metalloproteases
- bFGF, fibroblast growth factor
- cancer-associated fibroblast
- matrix-metalloproteases
- mononuclear inflammatory cell
- tumor stroma
Collapse
Affiliation(s)
- Noemí Eiró
- Unidad de Investigación, Fundación Hospital de Jove , Gijón, Asturias, Spain
| | | | - Julio Vázquez
- Servicio de Ginecología, Hospital Álvarez-Buylla , Mieres, Asturias, Spain
| | - José M Del Casar
- Servicio de Cirugía General, Fundación Hospital de Jove , Gijón, Spain
| | - Luis O González
- Unidad de Investigación, Fundación Hospital de Jove , Gijón, Asturias, Spain ; Servicio de Anatomía Patológica, Fundación Hospital de Jove, Gijón , Asturias, Spain
| | - Francisco J Vizoso
- Unidad de Investigación, Fundación Hospital de Jove , Gijón, Asturias, Spain ; Servicio de Cirugía General, Fundación Hospital de Jove , Gijón, Spain
| |
Collapse
|
175
|
Bonnans C, Chou J, Werb Z. Remodelling the extracellular matrix in development and disease. Nat Rev Mol Cell Biol 2015; 15:786-801. [PMID: 25415508 DOI: 10.1038/nrm3904] [Citation(s) in RCA: 2776] [Impact Index Per Article: 308.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The extracellular matrix (ECM) is a highly dynamic structure that is present in all tissues and continuously undergoes controlled remodelling. This process involves quantitative and qualitative changes in the ECM, mediated by specific enzymes that are responsible for ECM degradation, such as metalloproteinases. The ECM interacts with cells to regulate diverse functions, including proliferation, migration and differentiation. ECM remodelling is crucial for regulating the morphogenesis of the intestine and lungs, as well as of the mammary and submandibular glands. Dysregulation of ECM composition, structure, stiffness and abundance contributes to several pathological conditions, such as fibrosis and invasive cancer. A better understanding of how the ECM regulates organ structure and function and of how ECM remodelling affects disease progression will contribute to the development of new therapeutics.
Collapse
Affiliation(s)
- Caroline Bonnans
- 1] Department of Anatomy, University of California, 513 Parnassus Avenue, San Francisco, California 94143-0452, USA. [2] Oncology Department, INSERM U661, Functional Genomic Institute, 141 rue de la Cardonille, 34094 Montpellier, France
| | - Jonathan Chou
- 1] Department of Anatomy, University of California, 513 Parnassus Avenue, San Francisco, California 94143-0452, USA. [2] Department of Medicine, University of California, 513 Parnassus Avenue, San Francisco, California 94143-0452, USA
| | - Zena Werb
- Department of Anatomy, University of California, 513 Parnassus Avenue, San Francisco, California 94143-0452, USA
| |
Collapse
|
176
|
Chen Y, Klingen TA, Wik E, Aas H, Vigeland E, Liestøl K, Garred Ø, Mæhlen J, Akslen LA, Lømo J. Breast cancer stromal elastosis is associated with mammography screening detection, low Ki67 expression and favourable prognosis in a population-based study. Diagn Pathol 2014; 9:230. [PMID: 25522915 PMCID: PMC4300053 DOI: 10.1186/s13000-014-0230-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 12/10/2014] [Indexed: 01/01/2023] Open
Abstract
Background Mammography screen-detected breast cancers have a better prognosis than predicted from established prognostic markers. A search for additional features that are characteristic for these tumours and their prognosis is needed to reduce overtreatment, a recognized challenge in breast cancer patient management today. Here, we have investigated the occurrence and importance of tumour elastosis. Methods We performed a population based retrospective study of breast cancers detected in the Norwegian Breast Cancer Screening Programme in Vestfold County during 2004–2009. In total, 197 invasive screen-detected cancers and 75 interval cancers in patients aged 50–69 years were compared with regard to standard clinico-pathological parameters and tumour shape, as well as ER, PR, HER2 and Ki67 expression. In particular, the presence of elastotic material in tumours was graded on a 4-tiered scale (score 0–3). Results Screen-detected cancers had a significantly higher content of stromal elastosis than interval cancers (p < 0.001). High content of elastosis (score 3) correlated strongly with stellate tumour shape, low histological grade, and ER+/HER2- status. Further, high elastosis score was significantly associated with lower Ki67 expression. In survival analyses, cases with high elastosis demonstrated increased recurrence free (p = 0.03) and disease-specific survival (p = 0.11) compared to cases with low elastosis. Conclusion There is a strong correlation between the presence of tumour elastosis, stellate tumour shape and mammography detection of breast cancers. To our knowledge, this is the first time elastosis has been studied in relation to breast cancer detection method. Presence of elastosis is associated with low tumour cell proliferation (Ki67) and a good prognosis. Virtual Slides The virtual slide(s) for this article can be found here: http://www.diagnosticpathology.diagnomx.eu/vs/13000_2014_230
Collapse
Affiliation(s)
- Ying Chen
- Department of Pathology, Vestfold Hospital, Tønsberg, Norway. .,Department of Pathology, Akershus University Hospital, Lørenskog, Norway.
| | - Tor A Klingen
- Department of Pathology, Vestfold Hospital, Tønsberg, Norway.
| | - Elisabeth Wik
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, Section for Pathology, University of Bergen, Bergen, Norway. .,Department of Pathology, Haukeland University Hospital, Bergen, Norway.
| | - Hans Aas
- Department of Surgery, Vestfold Hospital, Tønsberg, Norway.
| | - Einar Vigeland
- Department of Radiology, Vestfold Hospital, Tønsberg, Norway.
| | - Knut Liestøl
- Institute of Informatics, University of Oslo, Oslo, Norway.
| | - Øystein Garred
- Department of Pathology, Oslo University Hospital, 0424, Oslo, Norway.
| | - Jan Mæhlen
- Department of Pathology, Oslo University Hospital, 0424, Oslo, Norway.
| | - Lars A Akslen
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, Section for Pathology, University of Bergen, Bergen, Norway. .,Department of Pathology, Haukeland University Hospital, Bergen, Norway.
| | - Jon Lømo
- Department of Pathology, Oslo University Hospital, 0424, Oslo, Norway.
| |
Collapse
|
177
|
Neuzillet C, Tijeras-Raballand A, Cros J, Faivre S, Hammel P, Raymond E. Stromal expression of SPARC in pancreatic adenocarcinoma. Cancer Metastasis Rev 2014; 32:585-602. [PMID: 23690170 DOI: 10.1007/s10555-013-9439-3] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) stands as the poorest prognostic tumor of the digestive tract, with a 5-year survival rate of less than 5%. Therapeutic options for unresectable PDAC are extremely limited and there is a pressing need for expanded therapeutic approaches to improve current options available with gemcitabine-based regimens. With PDAC displaying one of the most prominent desmoplastic stromal reactions of all carcinomas, recent research has focused on the microenvironment surrounding PDAC cells. Secreted protein acid and rich in cysteine (SPARC), which is overexpressed in PDAC, may display tumor suppressor functions in several cancers (e.g., in colorectal, ovarian, prostate cancers, and acute myelogenous leukemia) but also appears to be overexpressed in other tumor types (e.g., breast cancer, melanoma, and glioblastoma). The apparent contradictory functions of SPARC may yield inhibition of angiogenesis via inhibition of vascular endothelial growth factor, while promoting epithelial-to-mesenchymal transition and invasion through matrix metalloprotease expression. This feature is of particular interest in PDAC where SPARC overexpression in the stroma stands along with inhibition of angiogenesis and promotion of cancer cell invasion and metastasis. Several therapeutic strategies to deplete stromal tissue have been developed. In this review, we focused on key preclinical and clinical data describing the role of SPARC in PDAC biology, the properties, and mechanisms of delivery of drugs that interact with SPARC and discuss the proof-of-concept clinical trials using nab-paclitaxel.
Collapse
Affiliation(s)
- Cindy Neuzillet
- Department of Medical Oncology (INSERM U728-PRES Paris 7 Diderot), Beaujon University Hospital, Assistance Publique-Hôpitaux de Paris, 100 boulevard du Général Leclerc, 92110, Clichy-La-Garenne, France
| | | | | | | | | | | |
Collapse
|
178
|
Yamada T, Ohno S, Kitamura N, Sasabe E, Yamamoto T. SPARC is associated with carcinogenesis of oral squamous epithelium and consistent with cell competition. Med Mol Morphol 2014; 48:129-37. [PMID: 25311631 DOI: 10.1007/s00795-014-0089-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 09/22/2014] [Indexed: 11/24/2022]
Abstract
The matricellular protein, secreted protein acidic and rich in cysteine (SPARC) is thought to be involved in cell competition. The objective of this study is to investigate the role of SPARC in cancerization of oral squamous epithelium. Clinical specimens from 57 pre- and early cancerous lesion, 66 invasive squamous cell carcinoma (SCC) and controls were immunostained with SPARC. Clinical features and SPARC expression were evaluated. Furthermore, effects of SPARC knockdown and overexpression were examined in oral cancer and keratinocyte cell lines. Leukoplakia, carcinoma in situ, and early invasive SCC had more SPARC-positive cells than normal mucous epithelium. However, there were no significant differences between leukoplakia, carcinoma in situ, and early SCC, and there were no correlations between SPARC immunoreactivity and prognosis of invasive oral SCCs. Cell proliferation was down-regulated by SPARC siRNA, and enhanced by SPARC transformed keratinocytes. But SPARC overexpression did not enhance cell migration activity. SPARC is induced by dysplastic cells in the early stage of cancerization, and may improve survival capability, but is not involved in malignancy. SPARC may act to escape from elimination by cell competition.
Collapse
Affiliation(s)
- Tomohiro Yamada
- Section of Oral and Maxillofacial Surgery, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan.
| | - Seiji Ohno
- Department of Oral and Maxillofacial Surgery, Kochi Medical School, Kochi University, Kohasu, Oko-cho, Nankoku, Kochi, 783-8505, Japan
| | - Naoya Kitamura
- Department of Oral and Maxillofacial Surgery, Kochi Medical School, Kochi University, Kohasu, Oko-cho, Nankoku, Kochi, 783-8505, Japan
| | - Eri Sasabe
- Department of Oral and Maxillofacial Surgery, Kochi Medical School, Kochi University, Kohasu, Oko-cho, Nankoku, Kochi, 783-8505, Japan
| | - Tetsuya Yamamoto
- Department of Oral and Maxillofacial Surgery, Kochi Medical School, Kochi University, Kohasu, Oko-cho, Nankoku, Kochi, 783-8505, Japan
| |
Collapse
|
179
|
Knower KC, Chand AL, Eriksson N, Takagi K, Miki Y, Sasano H, Visvader JE, Lindeman GJ, Funder JW, Fuller PJ, Simpson ER, Tilley WD, Leedman PJ, Graham JD, Muscat GEO, Clarke CL, Clyne CD. Distinct nuclear receptor expression in stroma adjacent to breast tumors. Breast Cancer Res Treat 2014; 142:211-23. [PMID: 24122391 DOI: 10.1007/s10549-013-2716-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2013] [Accepted: 09/26/2013] [Indexed: 12/21/2022]
Abstract
The interaction between breast tumor epithelial and stromal cells is vital for initial and recurrent tumor growth. While breast cancer-associated stromal cells provide a favorable environment for proliferation and metastasis, the molecular mechanisms contributing to this process are not fully understood. Nuclear receptors (NRs) are intracellular transcription factors that directly regulate gene expression. Little is known about the status of NRs in cancer-associated stroma. Nuclear Receptor Low-Density Taqman Arrays were used to compare the gene expression profiles of all 48 NR family members in a collection of primary cultured cancer-associated fibroblasts (CAFs) obtained from estrogen receptor (ER)α positive breast cancers (n = 9) and normal breast adipose fibroblasts (NAFs) (n = 7). Thirty-three of 48 NRs were expressed in both the groups, while 11 NRs were not detected in either. Three NRs (dosage-sensitive sex reversal, adrenal hypoplasia critical region, on chromosome X, gene 1 (DAX-1); estrogen-related receptor beta (ERR-β); and RAR-related orphan receptor beta (ROR-β)) were only detected in NAFs, while one NR (liver receptor homolog-1 (LRH-1)) was unique to CAFs. Of the NRs co-expressed, four were significantly down-regulated in CAFs compared with NAFs (RAR-related orphan receptor-α (ROR-α); Thyroid hormone receptor-β (TR-β); vitamin D receptor (VDR); and peroxisome proliferator-activated receptor-γ (PPAR-γ)). Quantitative immunohistochemistry for LRH-1, TR-β, and PPAR-γ proteins in stromal fibroblasts from an independent panel of breast cancers (ER-positive (n = 15), ER-negative (n = 15), normal (n = 14)) positively correlated with mRNA expression profiles. The differentially expressed NRs identified in tumor stroma are key mediators in aromatase regulation and subsequent estrogen production. Our findings reveal a distinct pattern of NR expression that therefore fits with a sustained and increased local estrogen microenvironment in ER-positive tumors. NRs in CAFs may provide a new avenue for the development of intratumoral-targeted therapies in breast cancer.
Collapse
|
180
|
Abstract
The extracellular matrix (ECM) is composed of highly variable and dynamic components that regulate cell behavior. The protein composition and physical properties of the ECM govern cell fate through biochemical and biomechanical mechanisms. This requires a carefully orchestrated and thorough regulation considering that a disturbed ECM can have serious consequences and lead to pathological conditions like cancer. In breast cancer, many ECM proteins are significantly deregulated and specific matrix components promote tumor progression and metastatic spread. Intriguingly, several ECM proteins that are associated with breast cancer development, overlap substantially with a group of ECM proteins induced during the state of tissue remodeling such as mammary gland involution. Fibrillar collagens, fibronectin, hyaluronan and matricellular proteins are matrix components that are common to both involution and cancer. Moreover, some of these proteins have in recent years been identified as important constituents of metastatic niches in breast cancer. In addition, specific ECM molecules, their receptors or enzymatic modifiers are significantly involved in resistance to therapeutic intervention. Further analysis of these ECM proteins and the downstream ECM mediated signaling pathways may provide a range of possibilities to identify druggable targets against advanced breast cancer.
Collapse
Affiliation(s)
- Thordur Oskarsson
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Im Neuenheimer Feld 280, D-69120 Heidelberg, Germany; Divison of Stem Cells and Cancer, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, D-69120 Heidelberg, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany.
| |
Collapse
|
181
|
Guzman A, Ziperstein MJ, Kaufman LJ. The effect of fibrillar matrix architecture on tumor cell invasion of physically challenging environments. Biomaterials 2014; 35:6954-63. [PMID: 24835043 DOI: 10.1016/j.biomaterials.2014.04.086] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Accepted: 04/18/2014] [Indexed: 11/30/2022]
Abstract
Local invasion by and dissemination of cancer cells from a primary tumor are key initial steps of metastasis, the most lethal aspect of cancer. To study these processes in vitro, the invasion of cells from multicellular breast cancer aggregates embedded in three-dimensional (3D) extracellular matrix culture systems was studied. This work showed that in 3D fibrillar environments composed of collagen I, pore size--not the viscoelastic properties of the matrix--was the biophysical characteristic controlling breast cancer cell invasion efficiency. Furthermore, it was shown that fibrillar matrix architecture is a crucial factor that allows for efficient 3D invasion. In a 3D non-fibrillar environment composed of basement membrane extract (BME), invasion efficiency was greatly diminished, the mesenchymal individual mode of 3D invasion was abolished, and establishment of cell polarity and protrusions was compromised. These effects were seen even though the BME matrix has invasion permissive viscoelasticity and suitable adhesion ligands. The altered and limited invasive behavior observed in BME was rescued through introduction of fibrillar collagen into the non-fibrillar matrix. The biophysical cues of fibrillar collagen facilitated efficient invasion of sterically disadvantageous environments through assisting cell polarization and formation of stable cell protrusions. Finally, we suggest the composite matrices employed in this study consisting of fibrillar collagen I and BME in either a liquid-like or gelled state are suitable for a wide range of 3D cell studies, as these matrices combine fibrillar features that require cells to deploy integrin-dependent mechanotransduction machinery and a tunable non-fibrillar component that may require cells to adopt alternative migratory modes.
Collapse
Affiliation(s)
- Asja Guzman
- Department of Chemistry, Columbia University, New York, NY 10027, USA
| | | | - Laura J Kaufman
- Department of Chemistry, Columbia University, New York, NY 10027, USA.
| |
Collapse
|
182
|
Le Guellec S, Duprez-Paumier R, Lacroix-Triki M. Microenvironnement tumoral. Med Sci (Paris) 2014; 30:372-7. [DOI: 10.1051/medsci/20143004009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
183
|
Naba A, Clauser KR, Lamar JM, Carr SA, Hynes RO. Extracellular matrix signatures of human mammary carcinoma identify novel metastasis promoters. eLife 2014; 3:e01308. [PMID: 24618895 PMCID: PMC3944437 DOI: 10.7554/elife.01308] [Citation(s) in RCA: 257] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The extracellular matrix (ECM) is a major component of tumors and a significant contributor to cancer progression. In this study, we use proteomics to investigate the ECM of human mammary carcinoma xenografts and show that primary tumors of differing metastatic potential differ in ECM composition. Both tumor cells and stromal cells contribute to the tumor matrix and tumors of differing metastatic ability differ in both tumor- and stroma-derived ECM components. We define ECM signatures of poorly and highly metastatic mammary carcinomas and these signatures reveal up-regulation of signaling pathways including TGFβ and VEGF. We further demonstrate that several proteins characteristic of highly metastatic tumors (LTBP3, SNED1, EGLN1, and S100A2) play causal roles in metastasis, albeit at different steps. Finally we show that high expression of LTBP3 and SNED1 correlates with poor outcome for ER−/PR−breast cancer patients. This study thus identifies novel biomarkers that may serve as prognostic and diagnostic tools. DOI:http://dx.doi.org/10.7554/eLife.01308.001 Metastasis is the process whereby tumor cells spread within the body and is the cause of most deaths from cancer. This complex process involves several steps: first the cancer cells invade the tissues that surround the tumor; second, the cancer cells enter the blood stream and travel throughout the body; and third, the cancer cells seed the growth of new tumors in distant organs. Within tissues, the extracellular matrix forms a complex scaffold of proteins that surrounds cells, to support and organize them: it also provides signals that control how much cells can multiply, how likely cells are to stick together or migrate, and even a cell’s chances of survival. Pathologists have used an accumulation of extracellular matrix proteins in tumors as a sign that the outcome of the disease will likely be unfavorable for a patient, and that treatment will be challenging. However, we still do not have a clear picture of the composition of the tumor extracellular matrix and we do not know all the details of how it affects tumor growth and metastasis. Now, Naba et al. have explored these questions by injecting different types of human breast tumor cells into mice. Some of the cells were capable of spreading throughout the body and were said to have a high ‘metastatic potential’; others were less capable of spreading and were said to have a low metastatic potential. Naba et al. then analyzed the proteins that made up the extracellular matrix of the tumors that grew in the mice. Some proteins were found in both types of tumor; whereas some proteins were only found in the tumors with low metastatic potential and some were only found in the highly metastatic tumors. Naba et al. also demonstrated that both cancer cells and non-cancer cells—which are also found within the tumors—contributed to the production of the extracellular matrix in the tumor. Moreover, and somewhat surprisingly, the contributions from the non-cancer cells in the two types of tumors were also different. Computational analysis predicted that the production of several extracellular matrix proteins in the highly metastatic tumors was under the control of signaling pathways that are involved in cancer progression. Furthermore, Naba et al. also demonstrated that several of the extracellular matrix proteins specific to highly metastatic tumors were required for the cancer to spread. These proteins are involved in different stages of the metastatic process, and some of them are commonly over-produced in tumors from patients with some of the worst chances of recovery. If similar results are consistently observed in clinical samples from humans, the work of Naba et al. could help doctors to discriminate between tumors that will spread and those that will not, which should lead to improved patient care. The proteins and pathways associated with the highly metastatic tumors could be also investigated as potential drug targets. DOI:http://dx.doi.org/10.7554/eLife.01308.002
Collapse
Affiliation(s)
- Alexandra Naba
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, United States
| | | | | | | | | |
Collapse
|
184
|
West RB, van de Rijn M, Chen JL. Stromal responses among carcinomas--response. Clin Cancer Res 2014; 20:1397. [PMID: 24590889 DOI: 10.1158/1078-0432.ccr-13-3238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Robert B West
- Authors' Affiliation: Stanford University, Stanford, California
| | | | | |
Collapse
|
185
|
Triulzi T, Orlandi R, Tagliabue E. Stromal responses among carcinomas--letter. Clin Cancer Res 2014; 20:1396. [PMID: 24590888 DOI: 10.1158/1078-0432.ccr-13-3005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Tiziana Triulzi
- Authors' Affiliation: Molecular Targeting Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | | |
Collapse
|
186
|
Glass K, Girvan M. Annotation enrichment analysis: an alternative method for evaluating the functional properties of gene sets. Sci Rep 2014; 4:4191. [PMID: 24569707 PMCID: PMC3935204 DOI: 10.1038/srep04191] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Accepted: 01/28/2014] [Indexed: 12/18/2022] Open
Abstract
Gene annotation databases (compendiums maintained by the scientific community that describe the biological functions performed by individual genes) are commonly used to evaluate the functional properties of experimentally derived gene sets. Overlap statistics, such as Fishers Exact test (FET), are often employed to assess these associations, but don't account for non-uniformity in the number of genes annotated to individual functions or the number of functions associated with individual genes. We find FET is strongly biased toward over-estimating overlap significance if a gene set has an unusually high number of annotations. To correct for these biases, we develop Annotation Enrichment Analysis (AEA), which properly accounts for the non-uniformity of annotations. We show that AEA is able to identify biologically meaningful functional enrichments that are obscured by numerous false-positive enrichment scores in FET, and we therefore suggest it be used to more accurately assess the biological properties of gene sets.
Collapse
Affiliation(s)
- Kimberly Glass
- 1] Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA, USA [2] Department of Biostatistics, Harvard School of Public Health, Boston, MA, USA [3] Department of Physics, University of Maryland, College Park, MD, USA
| | - Michelle Girvan
- 1] Department of Physics, University of Maryland, College Park, MD, USA [2] Institute for Physical Science and Technology, University of Maryland, College Park, MD, USA [3] Santa Fe Institute, Santa Fe, NM
| |
Collapse
|
187
|
Paulsson J, Micke P. Prognostic relevance of cancer-associated fibroblasts in human cancer. Semin Cancer Biol 2014; 25:61-8. [PMID: 24560651 DOI: 10.1016/j.semcancer.2014.02.006] [Citation(s) in RCA: 197] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 02/11/2014] [Indexed: 12/12/2022]
Abstract
Prognostication is an integral part of cancer diagnostic and helps oncologists to guide treatment decisions and therapy intensity. Accumulating evidence suggest that the stroma compartment also contains independent prognostic information, best exemplified by the impact of immune cells and cells of the vasculature on cancer progression. Similarly, strong experimental evidence exist that stromal fibroblasts, often designated cancer associated fibroblasts (CAFs), are actively involved in tumorigenesis. Thus, it can be anticipated that the molecular repertoire of CAFs is likewise important for the clinical behavior of the tumor. In this review we present recent studies addressing the prognostic impact of CAFs, with the focus on human lung and breast cancer. Several single markers have been suggested, either CAF specific or CAF derived, that in immunohistochemical studies have demonstrated independent association with survival. This includes members of the platelet derived growth factor receptor (PDGFR) family, CAF-markers like podoplanin and fibroblast activation protein (FAP) as well as transcription factors (FoxF1) and secreted factors (matrix metalloproteinases (MMPs), SPARC). However, most studies are based on explorative evaluations on single patient cohorts and require further validation. Using a more comprehensive approach, microarray studies have been employed to create gene expression signatures that detect an activated fibroblast state. These "stroma signatures" have been applied to identify specific CAF features associated with prognosis in several independent data sets of breast and lung cancer patients. Early studies in breast cancer have also demonstrated that fibroblast features influence therapy response. Thus, many strategies have been used to present encouraging proof-of-concept findings that CAFs could be exploited for prognostication. However, these studies also highlight the difficulties to conclusively define an "activated stroma" and to identify the individual factors involved in clinically relevant tumor-stroma interactions.
Collapse
Affiliation(s)
- Janna Paulsson
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institute, Stockholm, Sweden.
| | - Patrick Micke
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| |
Collapse
|
188
|
Moustakas A, Heldin P. TGFβ and matrix-regulated epithelial to mesenchymal transition. Biochim Biophys Acta Gen Subj 2014; 1840:2621-34. [PMID: 24561266 DOI: 10.1016/j.bbagen.2014.02.004] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2014] [Accepted: 02/05/2014] [Indexed: 12/14/2022]
Abstract
BACKGROUND The progression of cancer through stages that guide a benign hyperplastic epithelial tissue towards a fully malignant and metastatic carcinoma, is driven by genetic and microenvironmental factors that remodel the tissue architecture. The concept of epithelial-mesenchymal transition (EMT) has evolved to emphasize the importance of plastic changes in tissue architecture, and the cross-communication of tumor cells with various cells in the stroma and with specific molecules in the extracellular matrix (ECM). SCOPE OF THE REVIEW Among the multitude of ECM-embedded cytokines and the regulatory potential of ECM molecules, this article focuses on the cytokine transforming growth factor β (TGFβ) and the glycosaminoglycan hyaluronan, and their roles in cancer biology and EMT. For brevity, we concentrate our effort on breast cancer. MAJOR CONCLUSIONS Both normal and abnormal TGFβ signaling can be detected in carcinoma and stromal cells, and TGFβ-induced EMT requires the expression of hyaluronan synthase 2 (HAS2). Correspondingly, hyaluronan is a major constituent of tumor ECM and aberrant levels of both hyaluronan and TGFβ are thought to promote a wounding reaction to the local tissue homeostasis. The link between EMT and metastasis also involves the mesenchymal-epithelial transition (MET). ECM components, signaling networks, regulatory non-coding RNAs and epigenetic mechanisms form the network of regulation during EMT-MET. GENERAL SIGNIFICANCE Understanding the mechanism that controls epithelial plasticity in the mammary gland promises the development of valuable biomarkers for the prognosis of breast cancer progression and even provides new ideas for a more integrative therapeutic approach against disease. This article is part of a Special Issue entitled Matrix-mediated cell behaviour and properties.
Collapse
Affiliation(s)
- Aristidis Moustakas
- Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, Box 595, SE-751 24 Uppsala, Sweden; Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Box 582, SE-751 23 Uppsala, Sweden.
| | - Paraskevi Heldin
- Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, Box 595, SE-751 24 Uppsala, Sweden; Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Box 582, SE-751 23 Uppsala, Sweden.
| |
Collapse
|
189
|
Wolf DM, Lenburg ME, Yau C, Boudreau A, van ‘t Veer LJ. Gene co-expression modules as clinically relevant hallmarks of breast cancer diversity. PLoS One 2014; 9:e88309. [PMID: 24516633 PMCID: PMC3917875 DOI: 10.1371/journal.pone.0088309] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Accepted: 01/06/2014] [Indexed: 12/25/2022] Open
Abstract
Co-expression modules are groups of genes with highly correlated expression patterns. In cancer, differences in module activity potentially represent the heterogeneity of phenotypes important in carcinogenesis, progression, or treatment response. To find gene expression modules active in breast cancer subpopulations, we assembled 72 breast cancer-related gene expression datasets containing ∼5,700 samples altogether. Per dataset, we identified genes with bimodal expression and used mixture-model clustering to ultimately define 11 modules of genes that are consistently co-regulated across multiple datasets. Functionally, these modules reflected estrogen signaling, development/differentiation, immune signaling, histone modification, ERBB2 signaling, the extracellular matrix (ECM) and stroma, and cell proliferation. The Tcell/Bcell immune modules appeared tumor-extrinsic, with coherent expression in tumors but not cell lines; whereas most other modules, interferon and ECM included, appeared intrinsic. Only four of the eleven modules were represented in the PAM50 intrinsic subtype classifier and other well-established prognostic signatures; although the immune modules were highly correlated to previously published immune signatures. As expected, the proliferation module was highly associated with decreased recurrence-free survival (RFS). Interestingly, the immune modules appeared associated with RFS even after adjustment for receptor subtype and proliferation; and in a multivariate analysis, the combination of Tcell/Bcell immune module down-regulation and proliferation module upregulation strongly associated with decreased RFS. Immune modules are unusual in that their upregulation is associated with a good prognosis without chemotherapy and a good response to chemotherapy, suggesting the paradox of high immune patients who respond to chemotherapy but would do well without it. Other findings concern the ECM/stromal modules, which despite common themes were associated with different sites of metastasis, possibly relating to the “seed and soil” hypothesis of cancer dissemination. Overall, co-expression modules provide a high-level functional view of breast cancer that complements the “cancer hallmarks” and may form the basis for improved predictors and treatments.
Collapse
Affiliation(s)
- Denise M. Wolf
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, California, United States of America
- * E-mail:
| | - Marc E. Lenburg
- Department of Medicine, Section of Computational Biomedicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Christina Yau
- Department of Surgery, University of California San Francisco, San Francisco, California, United States of America
- Buck Institute for Research on Aging, Novato, California, United States of America
| | - Aaron Boudreau
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Laura J. van ‘t Veer
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, California, United States of America
| |
Collapse
|
190
|
Morandi A, Chiarugi P. Metabolic implication of tumor:stroma crosstalk in breast cancer. J Mol Med (Berl) 2014; 92:117-26. [PMID: 24458539 DOI: 10.1007/s00109-014-1124-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Accepted: 01/10/2014] [Indexed: 12/14/2022]
Abstract
The metabolic properties of cancer cells significantly differ from those of normal cells. In particular, cancer cells are largely dependent on aerobic glycolysis, a phenomenon that has been exploited clinically by using labelled glucose for positron emission tomography imaging. Importantly, cancer-associated alterations in metabolism are not merely due to the resulting response to cell proliferation and survival. Indeed, direct metabolic regulation could be driven by tumor oncogenes and/or suppressors, as demonstrated in several solid tumors, including breast cancer. Despite the fact that most breast cancer studies have focused on the intrinsic characteristics of breast tumor cells, it is now widely accepted that tumor microenvironment plays an important role in defining and reprogramming cancer cell metabolism. Tumor:stroma crosstalk, as well as inflammatory cues, concurs to outlining the cancer metabolism, impact on cancer aggressiveness and ultimately on patient survival and therapeutic responses. The aim of this review is to (i) gather the most recent data regarding the metabolic alterations in breast cancer, (ii) describe the role of tumor microenvironment in breast cancer cell metabolic reprogramming, and (iii) contemplate how targeting metabolic pathways aberrantly activated in breast cancer could help current therapeutic regimens.
Collapse
Affiliation(s)
- Andrea Morandi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, viale GB Morgagni 50, Florence, I-50134, Italy
| | | |
Collapse
|
191
|
Quail DF, Joyce JA. Microenvironmental regulation of tumor progression and metastasis. Nat Med 2014; 19:1423-37. [PMID: 24202395 DOI: 10.1038/nm.3394] [Citation(s) in RCA: 5301] [Impact Index Per Article: 530.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Accepted: 10/09/2013] [Indexed: 02/07/2023]
Abstract
Cancers develop in complex tissue environments, which they depend on for sustained growth, invasion and metastasis. Unlike tumor cells, stromal cell types within the tumor microenvironment (TME) are genetically stable and thus represent an attractive therapeutic target with reduced risk of resistance and tumor recurrence. However, specifically disrupting the pro-tumorigenic TME is a challenging undertaking, as the TME has diverse capacities to induce both beneficial and adverse consequences for tumorigenesis. Furthermore, many studies have shown that the microenvironment is capable of normalizing tumor cells, suggesting that re-education of stromal cells, rather than targeted ablation per se, may be an effective strategy for treating cancer. Here we discuss the paradoxical roles of the TME during specific stages of cancer progression and metastasis, as well as recent therapeutic attempts to re-educate stromal cells within the TME to have anti-tumorigenic effects.
Collapse
Affiliation(s)
- Daniela F Quail
- Cancer Biology and Genetics Program, Memorial Sloan-Kettering Cancer Center, New York, New York, USA
| | | |
Collapse
|
192
|
Mao Y, Keller ET, Garfield DH, Shen K, Wang J. Stromal cells in tumor microenvironment and breast cancer. Cancer Metastasis Rev 2013; 32:303-15. [PMID: 23114846 DOI: 10.1007/s10555-012-9415-3] [Citation(s) in RCA: 497] [Impact Index Per Article: 45.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cancer is a systemic disease encompassing multiple components of both tumor cells themselves and host stromal cells. It is now clear that stromal cells in the tumor microenvironment play an important role in cancer development. Molecular events through which reactive stromal cells affect cancer cells can be defined so that biomarkers and therapeutic targets can be identified. Cancer-associated fibroblasts (CAFs) make up the bulk of cancer stroma and affect the tumor microenvironment such that they promote cancer initiation, angiogenesis, invasion, and metastasis. In breast cancer, CAFs not only promote tumor progression but also induce therapeutic resistance. Accordingly, targeting CAFs provides a novel way to control tumors with therapeutic resistance. This review summarizes the current understandings of tumor stroma in breast cancer with a particular emphasis on the role of CAFs and the therapeutic implications of CAFs. In addition, the effects of other stromal components such as endothelial cells, macrophages, and adipocytes in breast cancer are also discussed. Finally, we describe the biologic markers to categorize patients into a specific and confirmed subtype for personalized treatment.
Collapse
Affiliation(s)
- Yan Mao
- Shanghai Ruijin Hospital, Comprehensive Breast Health Center, Shanghai, China
| | | | | | | | | |
Collapse
|
193
|
Guo X, Zhu SX, Brunner AL, van de Rijn M, West RB. Next generation sequencing-based expression profiling identifies signatures from benign stromal proliferations that define stromal components of breast cancer. Breast Cancer Res 2013; 15:R117. [PMID: 24342436 PMCID: PMC3978842 DOI: 10.1186/bcr3586] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Accepted: 12/06/2013] [Indexed: 12/19/2022] Open
Abstract
Introduction Multiple studies have shown that the tumor microenvironment (TME) of carcinomas can play an important role in the initiation, progression, and metastasis of cancer. Here we test the hypothesis that specific benign fibrous soft tissue tumor gene expression profiles may represent distinct stromal fibroblastic reaction types that occur in different breast cancers. The discovered stromal profiles could classify breast cancer based on the type of stromal reaction patterns in the TME. Methods Next generation sequencing-based gene expression profiling (3SEQ) was performed on formalin fixed, paraffin embedded (FFPE) samples of 10 types of fibrous soft tissue tumors. We determined the extent to which these signatures could identify distinct subsets of breast cancers in four publicly available breast cancer datasets. Results A total of 53 fibrous tumors were sequenced by 3SEQ with an average of 29 million reads per sample. Both the gene signatures derived from elastofibroma (EF) and fibroma of tendon sheath (FOTS) demonstrated robust outcome results for survival in the four breast cancer datasets. The breast cancers positive for the EF signature (20-33% of the cohort) demonstrated significantly better outcome for survival. In contrast, the FOTS signature-positive breast cancers (11-35% of the cohort) had a worse outcome. Conclusions We defined and validated two new stromal signatures in breast cancer (EF and FOTS), which are significantly associated with prognosis. Our group has previously identified novel cancer stromal gene expression signatures associated with outcome differences in breast cancer by gene expression profiling of three soft tissue tumors, desmoid-type fibromatosis (DTF), solitary fibrous tumor (SFT), and tenosynovial giant cell tumor (TGCT/CSF1), as surrogates for stromal expression patterns. By combining the stromal signatures of EF and FOTS, with our previously identified DTF and TGCT/CSF1 signatures we can now characterize clinically relevant stromal expression profiles in the TME for between 74% to 90% of all breast cancers.
Collapse
|
194
|
Markers of breast cancer stromal fibroblasts in the primary tumour site associated with lymph node metastasis: a systematic review including our case series. Biosci Rep 2013; 33:BSR20130060. [PMID: 24229053 PMCID: PMC3860578 DOI: 10.1042/bsr20130060] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
CAFs (cancer-associated fibroblasts), the most abundant cell type in breast cancer stroma, produce a plethora of chemokines, growth factors and ECM (extracellular matrix) proteins, that may contribute to dissemination and metastasis. Axillary nodes are the first metastatic site in breast cancer; however, to the present date, there is no consensus of which specific proteins, synthesized by CAFs, might be related with lymph node involvement. The purpose of this study was to perform a systematic review of CAF biomarkers associated with the presence of regional metastasis. PubMed was searched using the words: ‘breast cancer’ and ‘lymph node’ and fibroblast or stroma or microenvironment. After exclusions, eight studies evaluating biomarkers immunoexpression in CAFs and lymph node status were selected. Biomarkers evaluated in these studies may be divided in two groups, according to their ontology: extracellular matrix components [MMP13 (matrix metalloproteinase 13), TIMP2 (tissue inhibitor of metalloproteinases-2), THBS1 (thrombospondin 1), LGALS1 (lectin, galactoside-binding, soluble, 1)] and response to wounding [PDPN (podoplanin), PLAU (plasminogen activator, urokinase), PLAUR (plasminogen activator, urokinase receptor), CAV1 (caveolin 1), THBS1, LGALS1]. A positive expression of MMP13 and LGALS1 in CAFs was associated with enhanced OR (odds ratio) for regional metastasis. Contrariwise, CAV1 positive staining of fibroblasts was associated with decreased OR for nodal involvement. Expression of MMP13, PDPN and CAV1 was further tested in a new series of 65 samples of invasive ductal breast carcinomas by immunohistochemistry and no association between biomarkers expression in CAFs and nodal status was found. It was suggested that breast cancer subtypes may differentially affect CAFs behaviour. It would be interesting to evaluate the prognostic significance of these biomarkers in CAFs from different tumour types.
Collapse
|
195
|
Triulzi T, Ratti M, Tortoreto M, Ghirelli C, Aiello P, Regondi V, Di Modica M, Cominetti D, Carcangiu ML, Moliterni A, Balsari A, Casalini P, Tagliabue E. Maspin influences response to doxorubicin by changing the tumor microenvironment organization. Int J Cancer 2013; 134:2789-97. [PMID: 24242003 DOI: 10.1002/ijc.28608] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Revised: 09/30/2013] [Accepted: 10/22/2013] [Indexed: 12/14/2022]
Abstract
Altered degradation and deposition of extracellular matrix are hallmarks of tumor progression and response to therapy. From a microarray supervised analysis on a dataset of chemotherapy-treated breast carcinoma patients, maspin, a member of the serpin protease inhibitor family, has been the foremost variable identified in non-responsive versus responsive tumors. Accordingly, in a series of 52 human breast carcinomas, we detected high maspin expression in tumors that progressed under doxorubicin (DXR)-based chemotherapy. Our analysis of the role of maspin in response to chemotherapy in human MCF7 and MDAMB231 breast and SKOV3 ovarian carcinoma cells transfected to overexpress maspin and injected into mice showed that maspin overexpression led to DXR resistance through the maspin-induced collagen-enriched microenvironment and that an anti-maspin neutralizing monoclonal antibody reversed the collagen-dependent DXR resistance. Impaired diffusion and decreased DXR activity were also found in tumors derived from Matrigel-embedded cells, where abundant collagen fibers characterize the tumor matrix. Conversely, liposome-based DXR reached maspin-overexpressing tumor cells despite the abundant extracellular matrix and was more efficient in reducing tumor growth. Our results identify maspin-induced accumulation of collagen fibers as a cause of disease progression under DXR chemotherapy for breast cancer. Use of a more hydrophilic DXR formulation or of a maspin inhibitor in combination with chemotherapy holds the promise of more consistent responses to maspin-overexpressing tumors and dense-matrix tumors in general.
Collapse
Affiliation(s)
- Tiziana Triulzi
- Molecular Targeting Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
196
|
Heldin P, Basu K, Olofsson B, Porsch H, Kozlova I, Kahata K. Deregulation of hyaluronan synthesis, degradation and binding promotes breast cancer. J Biochem 2013; 154:395-408. [PMID: 24092768 DOI: 10.1093/jb/mvt085] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Clinical and experimental data indicate that hyaluronan accumulates in breast cancer compared with normal breast epithelium, which correlates to poor prognosis. In this review, we discuss the expression of genes encoding enzymes that synthesize or degrade hyaluronan, i.e. hyaluronan synthases and hyaluronidases or bind hyaluronan, i.e. CD44 and receptor for hyaluronan-mediated motility (RHAMM, also designated as HMMR or CD168), in relation to breast cancer progression. Hyaluronan and hyaluronan receptors have multi-faceted roles in signalling events in breast cancer. A better understanding of the molecular mechanisms underlying these signalling pathways is highly warranted and may lead to improvement of cancer treatment.
Collapse
Affiliation(s)
- Paraskevi Heldin
- Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, Biomedical Center, Box 595, SE-75124 Uppsala, Sweden
| | | | | | | | | | | |
Collapse
|
197
|
Breast fibroblasts modulate early dissemination, tumorigenesis, and metastasis through alteration of extracellular matrix characteristics. Neoplasia 2013; 15:249-62. [PMID: 23479504 DOI: 10.1593/neo.121950] [Citation(s) in RCA: 145] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Revised: 01/16/2013] [Accepted: 01/18/2013] [Indexed: 12/22/2022] Open
Abstract
A wealth of evidence has now demonstrated that the microenvironment in which a tumorigenic cell evolves is as critical to its evolution as the genetic mutations it accrues. However, there is still relatively little known about how signals from the microenvironment contribute to the early events in the progression to malignancy. To address this question, we used a premalignant mammary model to examine how fibroblasts, and the extracellular matrix (ECM) proteins they secrete, influence progression to malignancy. Their effect on metastatic malignant cells was also assessed for comparison. We found that carcinoma-associated fibroblasts, and the distinct aligned ECM they deposit, can cause both premalignant and malignant mammary epithelial cells to assume a mesenchymal morphology that is associated with increased dissemination and metastasis, while benign reduction mammoplasty fibroblasts favor the maintenance of an epithelial morphology and constrain early dissemination, tumor growth, and metastasis. Our results suggest that normalizing the organization of the ECM could be effective in limiting systemic dissemination and tumor growth.
Collapse
|
198
|
Olsson N, Carlsson P, James P, Hansson K, Waldemarson S, Malmström P, Fernö M, Ryden L, Wingren C, Borrebaeck CAK. Grading breast cancer tissues using molecular portraits. Mol Cell Proteomics 2013; 12:3612-23. [PMID: 23982162 PMCID: PMC3861711 DOI: 10.1074/mcp.m113.030379] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Tumor progression and prognosis in breast cancer patients are difficult to assess using current clinical and laboratory parameters, where a pathological grading is indicative of tumor aggressiveness. This grading is based on assessments of nuclear grade, tubule formation, and mitotic rate. We report here the first protein signatures associated with histological grades of breast cancer, determined using a novel affinity proteomics approach. We profiled 52 breast cancer tissue samples by combining nine antibodies and label-free LC-MS/MS, which generated detailed quantified proteomic maps representing 1,388 proteins. The results showed that we could define in-depth molecular portraits of histologically graded breast cancer tumors. Consequently, a 49-plex candidate tissue protein signature was defined that discriminated between histological grades 1, 2, and 3 of breast cancer tumors with high accuracy. Highly biologically relevant proteins were identified, and the differentially expressed proteins indicated further support for the current hypothesis regarding remodeling of the tumor microenvironment during tumor progression. The protein signature was corroborated using meta-analysis of transcriptional profiling data from an independent patient cohort. In addition, the potential for using the markers to estimate the likelihood of long-term metastasis-free survival was also indicated. Taken together, these molecular portraits could pave the way for improved classification and prognostication of breast cancer.
Collapse
Affiliation(s)
- Niclas Olsson
- Department of Immunotechnology and CREATE HEALTH, Lund University, Medicon Village, SE-223 81 Lund, Sweden
| | | | | | | | | | | | | | | | | | | |
Collapse
|
199
|
Wood SL, Westbrook JA, Brown JE. Omic-profiling in breast cancer metastasis to bone: implications for mechanisms, biomarkers and treatment. Cancer Treat Rev 2013; 40:139-52. [PMID: 23958309 DOI: 10.1016/j.ctrv.2013.07.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Revised: 07/16/2013] [Accepted: 07/21/2013] [Indexed: 01/25/2023]
Abstract
Despite well-recognised advances in breast cancer treatment, there remain substantial numbers of patients who develop metastatic disease, of which up to 70% involves spread to bone, resulting in skeletal complications which have a major negative impact on mortality and quality of life. Bisphosphonates and newer bone-targeted agents have reduced the prevalence of skeletal complications, yet there remains significant unmet clinical need, particularly for the development of more specific therapies for the prevention and treatment of metastatic bone disease, for the prediction of risk of its development in individual patients and for the prediction of response to treatments. Modern 'omic' strategies can potentially make a major contribution to meeting this need. Technological advances in the field of nucleic acid sequencing, mass spectrometry and metabolic profiling have driven progress in genomics, transcriptomics (functional genomics), proteomics and metabolomics. This review appraises the recent application of these approaches to studies of breast cancer metastasis (particularly to bone), with a focus on understanding how omic approaches may lead to new therapeutic options and to novel biomarker molecules or molecular signatures with potential value in clinical practise. The increasingly recognised need for rigorous sample quality control and both pre-clinical and clinical validation to meet the ultimate goals of clinical utility and patient benefit is discussed. Future directions of omic driven research in breast cancer metastasis are considered, in particular micro-RNAs and their role in the post-transcriptional regulation of gene function and the possible role of cancer-stem cells and epigenetic modifications in the development of distant metastases.
Collapse
Affiliation(s)
- Steven L Wood
- Wolfson Molecular Imaging Centre, University of Manchester, Manchester M20 3LJ, UK.
| | | | | |
Collapse
|
200
|
Chen JLY, Espinosa I, Lin AY, Liao OYW, van de Rijn M, West RB. Stromal responses among common carcinomas correlated with clinicopathologic features. Clin Cancer Res 2013; 19:5127-35. [PMID: 23804424 DOI: 10.1158/1078-0432.ccr-12-3127] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
PURPOSE We have previously characterized a tumor stroma expression signature in a subset of breast tumors that correlates with better clinical outcome. The purpose of this study is to determine whether this stromal signature, termed the "DTF fibroblast" (desmoid-type fibromatosis) signature, is specific to breast cancer or is a common stromal response found in different types of cancer. EXPERIMENTAL DESIGNS The DTF fibroblast signature was applied to gene expression profiles from five ovarian, five lung, two colon, and three prostate cancer expression microarray datasets. In addition, two different tissue microarrays of 204 ovarian tumors and 140 colon tumors were examined for the expression of previously characterized protein markers of DTF fibroblast signature. The DTF fibroblast stromal response was then correlated with clinicopathologic features. RESULTS The DTF fibroblast signature is robustly present in ovarian, lung, and colon carcinomas. Both expression microarray data and immunohistochemistry show that the subset of ovarian tumors with strong DTF fibroblast signature expression has statistically significant, worse survival outcomes. No reproducible survival differences were found in either the lung or the colon cancers. The prostate cancers failed to show a DTF fibroblast signature. Multivariant analysis showed that DTF fibroblast signature was significantly more prognostic than the proliferation status in ovarian carcinomas. CONCLUSIONS Our results suggest that the DTF fibroblast signature is a common tumor stroma signature in different types of cancer, including ovarian, lung, and colon carcinomas. Our findings provide further insight into the DTF fibroblast stromal responses across different types of carcinomas and their potential as prognostic and therapeutic targets.
Collapse
Affiliation(s)
- Julia L-Y Chen
- Authors' Affiliations: Departments of Pathology and Medicine, Stanford University Medical Center; Department of Statistics, Stanford University, Stanford; Department of Medicine, Santa Clara Valley Medical Center, San Jose, California; and Department of Pathology, Hospital de la Santa Creu i Sant Pau, Institute of Biomedical Research (IIB Sant Pau), Autonomous University of Barcelona, Barcelona, Spain
| | | | | | | | | | | |
Collapse
|