1
|
Roozitalab MR, Prekete N, Allen M, Grose RP, Louise Jones J. The Microenvironment in DCIS and Its Role in Disease Progression. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1464:211-235. [PMID: 39821028 DOI: 10.1007/978-3-031-70875-6_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Ductal carcinoma in situ (DCIS) accounts for ~20% of all breast cancer diagnoses but whilst known to be a precursor of invasive breast cancer (IBC), evidence suggests only one in six patients will ever progress. A key challenge is to distinguish between those lesions that will progress and those that will remain indolent. Molecular analyses of neoplastic epithelial cells have not identified consistent differences between lesions that progressed and those that did not, and this has focused attention on the tumour microenvironment (ME).The DCIS ME is unique, complex and dynamic. Myoepithelial cells form the wall of the ductal-lobular tree and exhibit broad tumour suppressor functions. However, in DCIS they acquire phenotypic changes that bestow them with tumour promoter properties, an important evolution since they act as the primary barrier for invasion. Changes in the peri-ductal stromal environment also arise in DCIS, including transformation of fibroblasts into cancer-associated fibroblasts (CAFs). CAFs orchestrate other changes in the stroma, including the physical structure of the extracellular matrix (ECM) through altered protein synthesis, as well as release of a plethora of factors including proteases, cytokines and chemokines that remodel the ECM. CAFs can also modulate the immune ME as well as impact on tumour cell signalling pathways. The heterogeneity of CAFs, including recognition of anti-tumourigenic populations, is becoming evident, as well as heterogeneity of immune cells and the interplay between these and the adipocyte and vascular compartments. Knowledge of the impact of these changes is more advanced in IBC but evidence is starting to accumulate for a role in DCIS. Detailed in vitro, in vivo and tissue studies focusing on the interplay between DCIS epithelial cells and the ME should help to define features that can better predict DCIS behaviour.
Collapse
Affiliation(s)
- Mohammad Reza Roozitalab
- Centre for Tumour Biology, Barts Cancer Institute, John Vane Science Centre, Charterhouse Square, Queen Mary University of London, London, UK
| | - Niki Prekete
- Centre for Tumour Biology, Barts Cancer Institute, John Vane Science Centre, Charterhouse Square, Queen Mary University of London, London, UK
| | - Michael Allen
- Centre for Tumour Biology, Barts Cancer Institute, John Vane Science Centre, Charterhouse Square, Queen Mary University of London, London, UK
| | - Richard P Grose
- Centre for Tumour Biology, Barts Cancer Institute, John Vane Science Centre, Charterhouse Square, Queen Mary University of London, London, UK
| | - J Louise Jones
- Centre for Tumour Biology, Barts Cancer Institute, John Vane Science Centre, Charterhouse Square, Queen Mary University of London, London, UK.
| |
Collapse
|
2
|
Jalil SMA, Henry JC, Cameron AJM. Targets in the Tumour Matrisome to Promote Cancer Therapy Response. Cancers (Basel) 2024; 16:1847. [PMID: 38791926 PMCID: PMC11119821 DOI: 10.3390/cancers16101847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/03/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
The extracellular matrix (ECM) is composed of complex fibrillar proteins, proteoglycans, and macromolecules, generated by stromal, immune, and cancer cells. The components and organisation of the matrix evolves as tumours progress to invasive disease and metastasis. In many solid tumours, dense fibrotic ECM has been hypothesised to impede therapy response by limiting drug and immune cell access. Interventions to target individual components of the ECM, collectively termed the matrisome, have, however, revealed complex tumour-suppressor, tumour-promoter, and immune-modulatory functions, which have complicated clinical translation. The degree to which distinct components of the matrisome can dictate tumour phenotypes and response to therapy is the subject of intense study. A primary aim is to identify therapeutic opportunities within the matrisome, which might support a better response to existing therapies. Many matrix signatures have been developed which can predict prognosis, immune cell content, and immunotherapy responses. In this review, we will examine key components of the matrisome which have been associated with advanced tumours and therapy resistance. We have primarily focussed here on targeting matrisome components, rather than specific cell types, although several examples are described where cells of origin can dramatically affect tumour roles for matrix components. As we unravel the complex biochemical, biophysical, and intracellular transduction mechanisms associated with the ECM, numerous therapeutic opportunities will be identified to modify tumour progression and therapy response.
Collapse
Affiliation(s)
| | | | - Angus J. M. Cameron
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK; (S.M.A.J.); (J.C.H.)
| |
Collapse
|
3
|
Llinàs-Arias P, Ensenyat-Mendez M, Íñiguez-Muñoz S, Orozco JIJ, Valdez B, Salomon MP, Matsuba C, Solivellas-Pieras M, Bedoya-López AF, Sesé B, Mezger A, Ormestad M, Unzueta F, Strand SH, Boiko AD, Hwang ES, Cortés J, DiNome ML, Esteller M, Lupien M, Marzese DM. Chromatin insulation orchestrates matrix metalloproteinase gene cluster expression reprogramming in aggressive breast cancer tumors. Mol Cancer 2023; 22:190. [PMID: 38017545 PMCID: PMC10683115 DOI: 10.1186/s12943-023-01906-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 11/17/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is an aggressive subtype that exhibits a high incidence of distant metastases and lacks targeted therapeutic options. Here we explored how the epigenome contributes to matrix metalloprotease (MMP) dysregulation impacting tumor invasion, which is the first step of the metastatic process. METHODS We combined RNA expression and chromatin interaction data to identify insulator elements potentially associated with MMP gene expression and invasion. We employed CRISPR/Cas9 to disrupt the CCCTC-Binding Factor (CTCF) binding site on an insulator element downstream of the MMP8 gene (IE8) in two TNBC cellular models. We characterized these models by combining Hi-C, ATAC-seq, and RNA-seq with functional experiments to determine invasive ability. The potential of our findings to predict the progression of ductal carcinoma in situ (DCIS), was tested in data from clinical specimens. RESULTS We explored the clinical relevance of an insulator element located within the Chr11q22.2 locus, downstream of the MMP8 gene (IE8). This regulatory element resulted in a topologically associating domain (TAD) boundary that isolated nine MMP genes into two anti-correlated expression clusters. This expression pattern was associated with worse relapse-free (HR = 1.57 [1.06 - 2.33]; p = 0.023) and overall (HR = 2.65 [1.31 - 5.37], p = 0.005) survival of TNBC patients. After CRISPR/Cas9-mediated disruption of IE8, cancer cells showed a switch in the MMP expression signature, specifically downregulating the pro-invasive MMP1 gene and upregulating the antitumorigenic MMP8 gene, resulting in reduced invasive ability and collagen degradation. We observed that the MMP expression pattern predicts DCIS that eventually progresses into invasive ductal carcinomas (AUC = 0.77, p < 0.01). CONCLUSION Our study demonstrates how the activation of an IE near the MMP8 gene determines the regional transcriptional regulation of MMP genes with opposing functional activity, ultimately influencing the invasive properties of aggressive forms of breast cancer.
Collapse
Affiliation(s)
- Pere Llinàs-Arias
- Cancer Epigenetics Laboratory, Health Research Institute of the Balearic Islands (IdISBa), Palma, 07120, Spain
| | - Miquel Ensenyat-Mendez
- Cancer Epigenetics Laboratory, Health Research Institute of the Balearic Islands (IdISBa), Palma, 07120, Spain
| | - Sandra Íñiguez-Muñoz
- Cancer Epigenetics Laboratory, Health Research Institute of the Balearic Islands (IdISBa), Palma, 07120, Spain
| | - Javier I J Orozco
- Saint John's Cancer Institute, Providence Saint John's Health Center, Santa Monica, CA, USA
| | - Betsy Valdez
- Saint John's Cancer Institute, Providence Saint John's Health Center, Santa Monica, CA, USA
| | - Matthew P Salomon
- Keck School of Medicine, USC Research Center for Liver Diseases, University of Southern California, Los Angeles, CA, USA
| | - Chikako Matsuba
- Keck School of Medicine, USC Research Center for Liver Diseases, University of Southern California, Los Angeles, CA, USA
| | - Maria Solivellas-Pieras
- Cancer Epigenetics Laboratory, Health Research Institute of the Balearic Islands (IdISBa), Palma, 07120, Spain
| | - Andrés F Bedoya-López
- Cancer Epigenetics Laboratory, Health Research Institute of the Balearic Islands (IdISBa), Palma, 07120, Spain
| | - Borja Sesé
- Cancer Epigenetics Laboratory, Health Research Institute of the Balearic Islands (IdISBa), Palma, 07120, Spain
| | - Anja Mezger
- Science for Life Laboratory, Solna, 17665, Sweden
| | | | - Fernando Unzueta
- Advanced Optical Microscopy Facility Scientific and Technological Centres of University of Barcelona, Barcelona, Spain
| | - Siri H Strand
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Alexander D Boiko
- Department of Medicine, Cedars-Sinai Medical Center, Samuel Oschin Comprehensive Cancer Institute, Los Angeles, CA, 90048, USA
| | - E Shelley Hwang
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Javier Cortés
- Pangaea Oncology, Quiron Group, International Breast Cancer Center (IBCC), Barcelona, 08017, Spain
- Medica Scientia Innovation Research SL (MEDSIR), Barcelona, 08018, Spain
- Department of Medicine, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, Madrid, 28670, Spain
| | - Maggie L DiNome
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Manel Esteller
- Josep Carreras Leukaemia Research Institute, Badalona, Barcelona, Catalonia, Spain
- Centro de Investigación Biomédica en Red Cancer (CIBERONC), Madrid, 28029, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Catalonia, Spain
- Physiological Sciences Department, School of Medicine and Health Sciences, University of Barcelona (UB), Barcelona, Catalonia, Spain
| | - Mathieu Lupien
- Princess Margaret Cancer Centre, Toronto, ON, M5G 1L7, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, M5G 1L7, Canada
- Ontario Institute for Cancer Research, Toronto, ON, M5G 0A3, Canada
| | - Diego M Marzese
- Cancer Epigenetics Laboratory, Health Research Institute of the Balearic Islands (IdISBa), Palma, 07120, Spain.
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
4
|
Eiro N, Cid S, Aguado N, Fraile M, de Pablo N, Fernández B, Domínguez F, González LO, Vizoso FJ. MMP1 and MMP11 Expression in Peripheral Blood Mononuclear Cells upon Their Interaction with Breast Cancer Cells and Fibroblasts. Int J Mol Sci 2020; 22:ijms22010371. [PMID: 33396463 PMCID: PMC7795480 DOI: 10.3390/ijms22010371] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/24/2020] [Accepted: 12/27/2020] [Indexed: 01/01/2023] Open
Abstract
Tumor-infiltrating immune cells phenotype is associated with tumor progression. However, little is known about the phenotype of the peripheral blood mononuclear cells (PBMC) from breast cancer patients. We investigated MMP1 and MMP11 expression in PBMC from breast cancer patients and we analyzed gene expression changes upon their interaction with cancer cells and cancer-associated fibroblasts (CAF). We measured the impact of PBMC on proinflammatory gene expression in breast cancer cells, normal fibroblast (NF), and CAF and the impact on proliferation and invasiveness capacity of breast cancer cells. Gene expression of MMP1 and MMP11 in PBMC from breast cancer patients (n = 54) and control (n = 28); expression of IL1A, IL6, IL17, IFNβ, and NFĸB in breast cancer cell lines (MCF-7 and MDA-MB-231); and, additionally, IL10 and MMP11 in CAF and NF were analyzed by qRT-PCR before and after co-culture. Our results show the existence of a subpopulation of breast cancer patients (25.9%) with very high levels of MMP11 gene expression in PBMC. Also, gene expression of MMP1 and MMP11 increases in PBMC after co-culture with breast cancer cell lines, NF or CAF. PBMC from healthy or breast cancer patients induce an increased proliferation rate on MCF-7 and an increased invasiveness capacity of MDA-MB-231. Finally, we show a differential expression profile of inflammatory genes in NF and CAF when co-cultured with control or breast cancer PBMC. We have observed that MMPs’ expression in PBMC is regulated by the microenvironment, while the expression of inflammatory genes in NF or CAF is differentially regulated by PBMC. These findings confirm the importance of the crosstalk between stromal cells and suggest that PBMC would play a role in promoting aggressive tumor behavior.
Collapse
Affiliation(s)
- Noemi Eiro
- Research Unit, Fundación Hospital de Jove, 33290 Gijón, Spain; (S.C.); (M.F.); (N.d.P.)
- Correspondence: (N.E.); (F.J.V.); Tel.: +34-985320050 (ext. 84216) (N.E.)
| | - Sandra Cid
- Research Unit, Fundación Hospital de Jove, 33290 Gijón, Spain; (S.C.); (M.F.); (N.d.P.)
| | - Nuria Aguado
- Department of Surgery, Hospital Universitario San Agustín, 33401 Avilés, Spain;
| | - María Fraile
- Research Unit, Fundación Hospital de Jove, 33290 Gijón, Spain; (S.C.); (M.F.); (N.d.P.)
| | - Nagore de Pablo
- Research Unit, Fundación Hospital de Jove, 33290 Gijón, Spain; (S.C.); (M.F.); (N.d.P.)
| | - Berta Fernández
- Department of Surgery, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain;
| | - Francisco Domínguez
- Department of Anatomical Pathology, Hospital Universitario de Cabueñes, 33394 Gijón, Spain;
| | - Luis O. González
- Department of Anatomical Pathology, Fundación Hospital de Jove, 33209 Gijón, Spain;
| | - Francisco J. Vizoso
- Research Unit, Fundación Hospital de Jove, 33290 Gijón, Spain; (S.C.); (M.F.); (N.d.P.)
- Department of Surgery, Fundación Hospital de Jove, 33290 Gijón, Spain
- Correspondence: (N.E.); (F.J.V.); Tel.: +34-985320050 (ext. 84216) (N.E.)
| |
Collapse
|
5
|
Toss M, Miligy I, Gorringe K, Mittal K, Aneja R, Ellis I, Green A, Rakha E. Prognostic significance of cathepsin V (CTSV/CTSL2) in breast ductal carcinoma in situ. J Clin Pathol 2019; 73:76-82. [PMID: 31444238 DOI: 10.1136/jclinpath-2019-205939] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 07/29/2019] [Accepted: 08/10/2019] [Indexed: 12/14/2022]
Abstract
AIMS Cathepsin V (CTSV/CTSL2) is a lysosomal cysteine proteinase and plays a role in extracellular matrix degradation. It is associated with poor prognosis in invasive breast cancer (IBC), but its role in breast ductal carcinoma in situ (DCIS) remains unclear. In this study, we aimed to evaluate the prognostic significance of CTSV in DCIS. METHODS CTSV protein expression was immunohistochemically assessed in a well-characterised and annotated cohort of DCIS comprising pure DCIS (n=776) and DCIS coexisting with IBC (n=239). CTSV expression was analysed in tumour cells and surrounding stroma, including its association with clinicopathological parameters and outcome. RESULTS In pure DCIS, high CTSV expression was observed in 29% of epithelial tumour cells and 20% of surrounding stroma. High expression in both components was associated with features of poor prognosis including higher nuclear grade, hormone receptor negativity and HER2 positivity. In addition, stromal CTSV expression was associated with larger DCIS size, comedo-type necrosis and high proliferation index. DCIS associated with IBC showed higher CTSV expression than pure DCIS either within the epithelial tumour cells or surrounding stroma (p<0.0001 and p=0.001, respectively). In DCIS/IBC, CTSV expression was higher in the invasive component than DCIS component either in tumour cells or surrounding stroma (both p<0.0001). CTSV stromal expression was associated with invasive recurrence independent of other prognostic factors in patients treated with breast conserving surgery (HR=3.0, p=0.005). CONCLUSION High expression of CTSV is associated with poor outcome in DCIS and is a potential marker to predict DCIS progression to invasive disease.
Collapse
Affiliation(s)
- Michael Toss
- Histopathology, University of Nottingham School of Medicine, Nottingham, UK
| | - Islam Miligy
- Histopathology, University of Nottingham School of Medicine, Nottingham, UK
| | | | | | | | - Ian Ellis
- Histopathology, University of Nottingham School of Medicine, Nottingham, UK
| | - Andrew Green
- Histopathology, University of Nottingham School of Medicine, Nottingham, UK
| | - Emad Rakha
- Histopathology, University of Nottingham School of Medicine, Nottingham, UK
| |
Collapse
|
6
|
Toss MS, Miligy IM, Gorringe KL, McCaffrey L, AlKawaz A, Abidi A, Ellis IO, Green AR, Rakha EA. Legumain is an independent predictor for invasive recurrence in breast ductal carcinoma in situ. Mod Pathol 2019; 32:639-649. [PMID: 30429518 DOI: 10.1038/s41379-018-0180-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 11/01/2018] [Accepted: 11/01/2018] [Indexed: 12/21/2022]
Abstract
Legumain is a proteolytic enzyme that plays a role in the regulation of cell proliferation in invasive breast cancer. Studies evaluating its role in ductal carcinoma in situ (DCIS) are lacking. Here, we aimed to characterize legumain protein expression in DCIS and evaluate its prognostic significance. Legumain was assessed immunohistochemically in a tissue microarray of a well-characterized cohort of DCIS (n = 776 pure DCIS and n = 239 DCIS associated with invasive breast cancer (DCIS-mixed)). Legumain immunoreactivity was scored in tumor cells and surrounding stroma and related to clinicopathological parameters and patient outcome. High legumain expression was observed in 23% of pure DCIS and was associated with features of high-risk DCIS including higher nuclear grade, comedo necrosis, hormone receptor negativity, HER2 positivity, and higher proliferation index. Legumain expression was higher in DCIS associated with invasive breast cancer than in pure DCIS (p < 0.0001). In the DCIS-mixed cohort, the invasive component showed higher legumain expression than the DCIS component (p < 0.0001). Legumain was an independent predictor of shorter local recurrencefree interval for all recurrences (p = 0.0003) and for invasive recurrences (p = 0.002). When incorporated with other risk factors, legumain provided better patient risk stratification. High legumain expression is associated with poor prognosis in DCIS and could be a potential marker to predict DCIS progression to invasive disease.
Collapse
Affiliation(s)
- Michael S Toss
- Department of Histopathology, Nottingham Breast Cancer Research Center, Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham, Nottingham University Hospital NHS Trust, Nottingham City Hospital, Nottingham, UK.,Histopathology department, South Egypt Cancer Institute, Assiut University, Assiut, Egypt
| | - Islam M Miligy
- Department of Histopathology, Nottingham Breast Cancer Research Center, Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham, Nottingham University Hospital NHS Trust, Nottingham City Hospital, Nottingham, UK.,Histopathology department, Faculty of Medicine, Menoufia University, Shebeen El-Kom, Egypt
| | - Kylie L Gorringe
- Cancer Genomics Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| | - L McCaffrey
- Department of Oncology, Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada
| | - Abdulbaqi AlKawaz
- Department of Histopathology, Nottingham Breast Cancer Research Center, Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham, Nottingham University Hospital NHS Trust, Nottingham City Hospital, Nottingham, UK.,College of dentistry, Al Mustansiriya University, Baghdad, Iraq
| | - Asima Abidi
- Department of Histopathology, Nottingham Breast Cancer Research Center, Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham, Nottingham University Hospital NHS Trust, Nottingham City Hospital, Nottingham, UK
| | - Ian O Ellis
- Department of Histopathology, Nottingham Breast Cancer Research Center, Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham, Nottingham University Hospital NHS Trust, Nottingham City Hospital, Nottingham, UK
| | - Andrew R Green
- Department of Histopathology, Nottingham Breast Cancer Research Center, Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham, Nottingham University Hospital NHS Trust, Nottingham City Hospital, Nottingham, UK
| | - Emad A Rakha
- Department of Histopathology, Nottingham Breast Cancer Research Center, Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham, Nottingham University Hospital NHS Trust, Nottingham City Hospital, Nottingham, UK. .,Histopathology department, Faculty of Medicine, Menoufia University, Shebeen El-Kom, Egypt.
| |
Collapse
|
7
|
Toss MS, Miligy IM, Haj-Ahmad R, Gorringe KL, AlKawaz A, Mittal K, Ellis IO, Green AR, Rakha EA. The prognostic significance of lysosomal protective protein (cathepsin A) in breast ductal carcinoma in situ. Histopathology 2019; 74:1025-1035. [PMID: 30725481 DOI: 10.1111/his.13835] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 02/03/2019] [Indexed: 12/14/2022]
Abstract
AIMS Cathepsin A (CTSA) is a key regulatory enzyme for galactoside metabolism. Additionally, it has a distinct proteolytic activity and plays a role in tumour progression. CTSA is differentially expressed at the mRNA level between breast ductal carcinoma in situ (DCIS) and invasive breast carcinoma (IBC). In this study, we aimed to characterise CTSA protein expression in DCIS and evaluate its prognostic significance. METHODS AND RESULTS A large cohort of DCIS [n = 776 for pure DCIS and n = 239 for DCIS associated with IBC (DCIS/IBC)] prepared as a tissue microarray was immunohistochemically stained for CTSA. High CTSA expression was observed in 48% of pure DCIS. High expression was associated with features of poor DCIS prognosis, including younger age at diagnosis (<50 years), higher nuclear grade, hormone receptor negativity, HER2 positivity, high proliferative index and high hypoxia inducible factor 1 alpha expression. High CTSA expression was associated with shorter recurrence-free interval (RFI) (P = 0.0001). In multivariate survival analysis for patients treated with breast conserving surgery, CTSA was an independent predictor of shorter RFI (P = 0.015). DCIS associated with IBC showed higher CTSA expression than pure DCIS (P = 0.04). In the DCIS/IBC cohort, CTSA expression was higher in the invasive component than the DCIS component (P < 0.0001). CONCLUSION CTSA is not only associated with aggressive behaviour and poor outcome in DCIS but also a potential marker to predict co-existing invasion in DCIS.
Collapse
Affiliation(s)
- Michael S Toss
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham, Nottingham City Hospital, Notts, UK.,Histopathology Department, South Egypt Cancer Institute, Assiut University, Assiut, Egypt
| | - Islam M Miligy
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham, Nottingham City Hospital, Notts, UK.,Histopathology Department, Faculty of Medicine, Menoufia University, Menoufia, Egypt
| | - Rita Haj-Ahmad
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham, Nottingham City Hospital, Notts, UK
| | - Kylie L Gorringe
- Cancer Genomics Program, Peter MacCallum Cancer Centre, Melbourne, Australia.,The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| | - Abdulbaqi AlKawaz
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham, Nottingham City Hospital, Notts, UK.,College of Dentistry, Al Mustansiriya University, Baghdad, Iraq
| | | | - Ian O Ellis
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham, Nottingham City Hospital, Notts, UK
| | - Andrew R Green
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham, Nottingham City Hospital, Notts, UK
| | - Emad A Rakha
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham, Nottingham City Hospital, Notts, UK.,Histopathology Department, Faculty of Medicine, Menoufia University, Menoufia, Egypt
| |
Collapse
|
8
|
Gorringe KL, Fox SB. Ductal Carcinoma In Situ Biology, Biomarkers, and Diagnosis. Front Oncol 2017; 7:248. [PMID: 29109942 PMCID: PMC5660056 DOI: 10.3389/fonc.2017.00248] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 10/02/2017] [Indexed: 12/21/2022] Open
Abstract
Ductal carcinoma in situ (DCIS) is an often-diagnosed breast disease and a known, non-obligate, precursor to invasive breast carcinoma. In this review, we explore the clinical and pathological features of DCIS, fundamental elements of DCIS biology including gene expression and genetic events, the relationship of DCIS with recurrence and invasive breast cancer, and the interaction of DCIS with the microenvironment. We also survey how these various elements are being used to solve the clinical conundrum of how to optimally treat a disease that has potential to progress, and yet is also likely over-treated in a significant proportion of cases.
Collapse
Affiliation(s)
- Kylie L. Gorringe
- Cancer Genomics Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| | - Stephen B. Fox
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| |
Collapse
|
9
|
Collagen as a double-edged sword in tumor progression. Tumour Biol 2013; 35:2871-82. [PMID: 24338768 PMCID: PMC3980040 DOI: 10.1007/s13277-013-1511-7] [Citation(s) in RCA: 397] [Impact Index Per Article: 33.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Accepted: 12/03/2013] [Indexed: 12/11/2022] Open
Abstract
It has been recognized that cancer is not merely a disease of tumor cells, but a disease of imbalance, in which stromal cells and tumor microenvironment play crucial roles. Extracellular matrix (ECM) as the most abundant component in tumor microenvironment can regulate tumor cell behaviors and tissue tension homeostasis. Collagen constitutes the scaffold of tumor microenvironment and affects tumor microenvironment such that it regulates ECM remodeling by collagen degradation and re-deposition, and promotes tumor infiltration, angiogenesis, invasion and migration. While collagen was traditionally regarded as a passive barrier to resist tumor cells, it is now evident that collagen is also actively involved in promoting tumor progression. Collagen changes in tumor microenvironment release biomechanical signals, which are sensed by both tumor cells and stromal cells, trigger a cascade of biological events. In this work, we discuss how collagen can be a double-edged sword in tumor progression, both inhibiting and promoting tumor progression at different stages of cancer development.
Collapse
|
10
|
Abstract
Many studies investigated the relationship between matrix metalloproteinase 2 (MMP-2) overexpression and survival in patients with colorectal cancer (CRC), but yielded inconsistent results. To derive a more precise estimate of the prognostic significance of MMP-2 overexpression, we reviewed published studies and carried out a meta-analysis. Eligible articles were identified for the period up to March 2012 in electronic databases. To evaluate the correlation between MMP-2 overexpression and the prognosis in CRC, pooled hazard ratio (HR) and its 95 % confidence interval (95 % CI) for poorer overall and progression-free survival were appropriately derived from fixed-effects or random-effects models using standard meta-analysis techniques. Thirteen studies with a total of 1,919 CRC patients stratifying overall survival (OS) and/or progression-free survival in CRC patients by MMP-2 expression status were eligible for analysis. Ten studies investigated the OS in a total of 1,612 cases with CRC, and five studies investigated the progression-free survival in a total of 508 patients CRC. The combined HR estimate for OS and progression-free survival was 1.74 (95 % CI, 1.34-2.26) and 1.35 (95 % CI, 1.07-1.80), respectively. Both subgroup analyses and sensitivity analysis further identified the prognostic role of MMP-2 overexpression in patients with CRC. There was no evidence for publication bias. In conclusion, MMP-2 overexpression is associated with poorer overall and progression-free survival in patients with CRC.
Collapse
|
11
|
Torzilli PA, Bourne JW, Cigler T, Vincent CT. A new paradigm for mechanobiological mechanisms in tumor metastasis. Semin Cancer Biol 2012; 22:385-95. [PMID: 22613484 PMCID: PMC3445741 DOI: 10.1016/j.semcancer.2012.05.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Accepted: 05/13/2012] [Indexed: 12/24/2022]
Abstract
Tumor metastases and epithelial to mesenchymal transition (EMT) involve tumor cell invasion and migration through the dense collagen-rich extracellular matrix surrounding the tumor. Little is neither known about the mechanobiological mechanisms involved in this process, nor the role of the mechanical forces generated by the cells in their effort to invade and migrate through the stroma. In this paper we propose a new fundamental mechanobiological mechanism involved in cancer growth and metastasis, which can be both protective or destructive depending on the magnitude of the forces generated by the cells. This new mechanobiological mechanism directly challenges current paradigms that are focused mainly on biological and biochemical mechanisms associated with tumor metastasis. Our new mechanobiological mechanism describes how tumor expansion generates mechanical forces within the stroma to not only resist tumor expansion but also inhibit or enhance tumor invasion by, respectively, inhibiting or enhancing matrix metalloproteinase (MMP) degradation of the tensed interstitial collagen. While this mechanobiological mechanism has not been previously applied to the study of tumor metastasis and EMT, it may have the potential to broaden our understanding of the tumor invasive process and assist in developing new strategies for preventing or treating cancer metastasis.
Collapse
Affiliation(s)
- Peter A Torzilli
- Tissue Engineering, Regeneration and Repair Program, Hospital for Special Surgery, New York, NY 10021, United States.
| | | | | | | |
Collapse
|