1
|
Del Toro K, Licon-Munoz Y, Crabtree W, Oper T, Robbins C, Hines WC. Breast pericytes: a newly identified driver of tumor cell proliferation. Front Oncol 2024; 14:1455484. [PMID: 39741968 PMCID: PMC11685225 DOI: 10.3389/fonc.2024.1455484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 11/27/2024] [Indexed: 01/03/2025] Open
Abstract
Introduction Effective treatment of breast cancer remains a formidable challenge, partly due to our limited understanding of the complex microenvironmental factors that contribute to disease pathology. Among these factors are tissue-resident perivascular cells, which play crucial roles in shaping vascular basement membranes, maintaining vessel integrity, and communicating with adjacent endothelial cells. Despite their essential functions, perivascular cells have been relatively overlooked. Identifying them by immunostaining has been challenging due to their low abundance, inherent heterogeneity, and shared marker expression with other cell types. These challenges have hindered efforts to purify pericytes and generate primary cell models for studying their biology. Methods Using a recently developed FACS method, we successfully identified and purified each cell type from breast tissues, allowing us to deep-sequence their transcriptomes and generate primary cell models of each cell type-including pericytes. Here, we used these data to analyze cell-type-specific gene expression in tumors, which revealed a strong association between pericyte-specific genes and breast cancer patient mortality. To explore this association, we defined the heterogeneity of breast pericytes using single-cell RNA sequencing and identified a broad marker for visualizing perivascular cells in breast tumors. Results Remarkably, we discovered perivascular cells dissociated from vessels and emerged as a dominant mesenchymal cell type in a subset of breast tumors that contrasted with their normal perivascular location. Moreover, when we purified pericytes from the breast and cultured them alongside breast tumor cells, we discovered that they induced rapid tumor cell growth significantly greater than isogenic fibroblast controls. Discussion These findings identify perivascular cells as a key microenvironmental factor in breast cancer, highlighting the critical need for further research to explore their biology and identify specific stimulatory mechanisms that could be targeted therapeutically.
Collapse
Affiliation(s)
| | | | | | | | | | - William C. Hines
- Department of Biochemistry and Molecular Biology, University of New Mexico School of
Medicine, 1 University of New Mexico MSC08 4670, Albuquerque, NM, United States
| |
Collapse
|
2
|
Ye Q, Wang J, Ducatman B, Raese RA, Rogers JL, Wan YW, Dong C, Padden L, Pugacheva EN, Qian Y, Guo NL. Expression-Based Diagnosis, Treatment Selection, and Drug Development for Breast Cancer. Int J Mol Sci 2023; 24:10561. [PMID: 37445737 DOI: 10.3390/ijms241310561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/06/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
There is currently no gene expression assay that can assess if premalignant lesions will develop into invasive breast cancer. This study sought to identify biomarkers for selecting patients with a high potential for developing invasive carcinoma in the breast with normal histology, benign lesions, or premalignant lesions. A set of 26-gene mRNA expression profiles were used to identify invasive ductal carcinomas from histologically normal tissue and benign lesions and to select those with a higher potential for future cancer development (ADHC) in the breast associated with atypical ductal hyperplasia (ADH). The expression-defined model achieved an overall accuracy of 94.05% (AUC = 0.96) in classifying invasive ductal carcinomas from histologically normal tissue and benign lesions (n = 185). This gene signature classified cancer development in ADH tissues with an overall accuracy of 100% (n = 8). The mRNA expression patterns of these 26 genes were validated using RT-PCR analyses of independent tissue samples (n = 77) and blood samples (n = 48). The protein expression of PBX2 and RAD52 assessed with immunohistochemistry were prognostic of breast cancer survival outcomes. This signature provided significant prognostic stratification in The Cancer Genome Atlas breast cancer patients (n = 1100), as well as basal-like and luminal A subtypes, and was associated with distinct immune infiltration and activities. The mRNA and protein expression of the 26 genes was associated with sensitivity or resistance to 18 NCCN-recommended drugs for treating breast cancer. Eleven genes had significant proliferative potential in CRISPR-Cas9/RNAi screening. Based on this gene expression signature, the VEGFR inhibitor ZM-306416 was discovered as a new drug for treating breast cancer.
Collapse
Affiliation(s)
- Qing Ye
- West Virginia University Cancer Institute/Mary Babb Randolph Cancer Center, West Virginia University, Morgantown, WV 26506, USA
| | - Jiajia Wang
- West Virginia University Cancer Institute/Mary Babb Randolph Cancer Center, West Virginia University, Morgantown, WV 26506, USA
| | - Barbara Ducatman
- Department of Pathology, West Virginia University, Morgantown, WV 26506, USA
| | - Rebecca A Raese
- West Virginia University Cancer Institute/Mary Babb Randolph Cancer Center, West Virginia University, Morgantown, WV 26506, USA
| | - Jillian L Rogers
- West Virginia University Cancer Institute/Mary Babb Randolph Cancer Center, West Virginia University, Morgantown, WV 26506, USA
| | - Ying-Wooi Wan
- West Virginia University Cancer Institute/Mary Babb Randolph Cancer Center, West Virginia University, Morgantown, WV 26506, USA
| | - Chunlin Dong
- West Virginia University Cancer Institute/Mary Babb Randolph Cancer Center, West Virginia University, Morgantown, WV 26506, USA
| | - Lindsay Padden
- West Virginia University Cancer Institute/Mary Babb Randolph Cancer Center, West Virginia University, Morgantown, WV 26506, USA
| | - Elena N Pugacheva
- West Virginia University Cancer Institute/Mary Babb Randolph Cancer Center, West Virginia University, Morgantown, WV 26506, USA
- Department of Biochemistry and Molecular Medicine, School of Medicine, West Virginia University, Morgantown, WV 26506, USA
- Department of Radiation Oncology, School of Medicine, West Virginia University, Morgantown, WV 26506, USA
| | - Yong Qian
- Pathology and Physiology Research Branch, National Institute for Occupational Safety and Health, Morgantown, WV 26505, USA
| | - Nancy Lan Guo
- West Virginia University Cancer Institute/Mary Babb Randolph Cancer Center, West Virginia University, Morgantown, WV 26506, USA
- Department of Occupational and Environmental Health Sciences, School of Public Health, West Virginia University, Morgantown, WV 26506, USA
| |
Collapse
|
3
|
Abstract
Cancer is a complex disease and a significant cause of mortality worldwide. Over the course of nearly all cancer types, collagen within the tumor microenvironment influences emergence, progression, and metastasis. This review discusses collagen regulation within the tumor microenvironment, pathological involvement of collagen, and predictive values of collagen and related extracellular matrix components in main cancer types. A survey of predictive tests leveraging collagen assays using clinical cohorts is presented. A conclusion is that collagen has high predictive value in monitoring cancer processes and stratifying by outcomes. New approaches should be considered that continue to define molecular facets of collagen related to cancer.
Collapse
|
4
|
Uddin MN, Wang X. Identification of breast cancer subtypes based on gene expression profiles in breast cancer stroma. Clin Breast Cancer 2022; 22:521-537. [DOI: 10.1016/j.clbc.2022.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 03/21/2022] [Accepted: 04/01/2022] [Indexed: 11/16/2022]
|
5
|
Uddin MN, Wang X. Identification of key tumor stroma-associated transcriptional signatures correlated with survival prognosis and tumor progression in breast cancer. Breast Cancer 2022; 29:541-561. [PMID: 35020130 DOI: 10.1007/s12282-022-01332-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 01/05/2022] [Indexed: 12/21/2022]
Abstract
BACKGROUND The aberrant expression of stromal gene signatures in breast cancer has been widely studied. However, the association of stromal gene signatures with tumor immunity, progression, and clinical outcomes remains lacking. METHODS Based on eight breast tumor stroma (BTS) transcriptomics datasets, we identified differentially expressed genes (DEGs) between BTS and normal breast stroma. Based on the DEGs, we identified dysregulated pathways and prognostic hub genes, hub oncogenes, hub protein kinases, and other key marker genes associated with breast cancer. Moreover, we compared the enrichment levels of stromal and immune signatures between breast cancer patients with bad and good clinical outcomes. We also investigated the association between tumor stroma-related genes and breast cancer progression. RESULTS The DEGs included 782 upregulated and 276 downregulated genes in BTS versus normal breast stroma. The pathways significantly associated with the DEGs included cytokine-cytokine receptor interaction, chemokine signaling, T cell receptor signaling, cell adhesion molecules, focal adhesion, and extracellular matrix-receptor interaction. Protein-protein interaction network analysis identified the stromal hub genes with prognostic value in breast cancer, including two oncogenes (COL1A1 and IL21R), two protein kinases encoding genes (PRKACA and CSK), and a growth factor encoding gene (PLAU). Moreover, we observed that the patients with bad clinical outcomes were less enriched in stromal and antitumor immune signatures (CD8 + T cells and tumor-infiltrating lymphocytes) but more enriched in tumor cells and immunosuppressive signatures (MDSCs and CD4 + regulatory T cells) compared with the patients with good clinical outcomes. The ratios of CD8 + /CD4 + regulatory T cells were lower in the patients with bad clinical outcomes. Furthermore, we identified the tumor stroma-related genes, including MCM4, SPECC1, IMPA2, and AGO2, which were gradually upregulated through grade I, II, and III breast cancers. In contrast, COL14A1, ESR1, SLIT2, IGF1, CH25H, PRR5L, ABCA6, CEP126, IGDCC4, LHFP, MFAP3, PCSK5, RAB37, RBMS3, SETBP1, and TSPAN11 were gradually downregulated through grade I, II, and III breast cancers. It suggests that the expression of these stromal genes has an association with the progression of breast cancers. These progression-associated genes also displayed an expression association with recurrence-free survival in breast cancer patients. CONCLUSIONS This study identified tumor stroma-associated biomarkers correlated with deregulated pathways, tumor immunity, tumor progression, and clinical outcomes in breast cancer. Our findings provide new insights into the pathogenesis of breast cancer.
Collapse
Affiliation(s)
- Md Nazim Uddin
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
- Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
- Big Data Research Institute, China Pharmaceutical University, Nanjing, 211198, China
- Institute of Food Science and Technology, Bangladesh Council of Scientific and Industrial Research (BCSIR), Dhaka, 1205, Bangladesh
| | - Xiaosheng Wang
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
- Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
- Big Data Research Institute, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
6
|
Postpartum breast cancer has a distinct molecular profile that predicts poor outcomes. Nat Commun 2021; 12:6341. [PMID: 34732713 PMCID: PMC8566602 DOI: 10.1038/s41467-021-26505-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 10/06/2021] [Indexed: 12/21/2022] Open
Abstract
Young women's breast cancer (YWBC) has poor prognosis and known interactions with parity. Women diagnosed within 5-10 years of childbirth, defined as postpartum breast cancer (PPBC), have poorer prognosis compared to age, stage, and biologic subtype-matched nulliparous patients. Genomic differences that explain this poor prognosis remain unknown. In this study, using RNA expression data from clinically matched estrogen receptor positive (ER+) cases (n = 16), we observe that ER+ YWBC can be differentiated based on a postpartum or nulliparous diagnosis. The gene expression signatures of PPBC are consistent with increased cell cycle, T-cell activation and reduced estrogen receptor and TP53 signaling. When applied to a large YWBC cohort, these signatures for ER+ PPBC associate with significantly reduced 15-year survival rates in high compared to low expressing cases. Cumulatively these results provide evidence that PPBC is a unique entity within YWBC with poor prognostic phenotypes.
Collapse
|
7
|
Reed ER, Monti S. Multi-resolution characterization of molecular taxonomies in bulk and single-cell transcriptomics data. Nucleic Acids Res 2021; 49:e98. [PMID: 34226941 PMCID: PMC8464061 DOI: 10.1093/nar/gkab552] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 06/07/2021] [Accepted: 06/18/2021] [Indexed: 12/21/2022] Open
Abstract
As high-throughput genomics assays become more efficient and cost effective, their utilization has become standard in large-scale biomedical projects. These studies are often explorative, in that relationships between samples are not explicitly defined a priori, but rather emerge from data-driven discovery and annotation of molecular subtypes, thereby informing hypotheses and independent evaluation. Here, we present K2Taxonomer, a novel unsupervised recursive partitioning algorithm and associated R package that utilize ensemble learning to identify robust subgroups in a 'taxonomy-like' structure. K2Taxonomer was devised to accommodate different data paradigms, and is suitable for the analysis of both bulk and single-cell transcriptomics, and other '-omics', data. For each of these data types, we demonstrate the power of K2Taxonomer to discover known relationships in both simulated and human tissue data. We conclude with a practical application on breast cancer tumor infiltrating lymphocyte (TIL) single-cell profiles, in which we identified co-expression of translational machinery genes as a dominant transcriptional program shared by T cells subtypes, associated with better prognosis in breast cancer tissue bulk expression data.
Collapse
Affiliation(s)
- Eric R Reed
- Section of Computational Biomedicine, Boston University School of Medicine, Boston, MA 02118, USA
- Bioinformatics Program, College of Engineering, Boston University, Boston, MA 02118, USA
| | - Stefano Monti
- Section of Computational Biomedicine, Boston University School of Medicine, Boston, MA 02118, USA
- Bioinformatics Program, College of Engineering, Boston University, Boston, MA 02118, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA
| |
Collapse
|
8
|
Roles of the Immune/Methylation/Autophagy Landscape on Single-Cell Genotypes and Stroke Risk in Breast Cancer Microenvironment. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5633514. [PMID: 34457116 PMCID: PMC8397558 DOI: 10.1155/2021/5633514] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 06/29/2021] [Accepted: 07/14/2021] [Indexed: 12/16/2022]
Abstract
This study sought to perform integrative analysis of the immune/methylation/autophagy landscape on breast cancer prognosis and single-cell genotypes. Breast Cancer Recurrence Risk Score (BCRRS) and Breast Cancer Prognostic Risk Score (BCPRS) were determined based on 6 prognostic IMAAGs obtained from the TCGA-BRCA cohort. BCRRS and BCPRS, respectively, were used to construct a risk prediction model of overall survival and progression-free survival. Predictive capacity of the model was evaluated using clinical data. Analysis showed that BCRRS is associated with a high risk of stroke. In addition, PPI and drug-ceRNA networks based on differences in BCPRS were constructed. Single cells were genotyped through integrated scRNA-seq of the TNBC samples based on clustering results of BCPRS-related genes. The findings of this study show the potential regulatory effects of IMAAGs on breast cancer tumor microenvironment. High AUCs of 0.856 and 0.842 were obtained for the OS and PFS prognostic models, respectively. scRNA-seq analysis showed high expression levels of adipocytes and adipose tissue macrophages (ATMs) in high BCPRS clusters. Moreover, analysis of ligand-receptor interactions and potential regulatory mechanisms were performed. The LINC00276&MALAT1/miR-206/FZD4-Wnt7b pathway was also identified which may be useful in future research on targets against breast cancer metastasis and recurrence. Neural network-based deep learning models using BCPRS-related genes showed that these genes can be used to map the tumor microenvironment. In summary, analysis of IMAAGs, BCPRS, and BCRRS provides information on the breast cancer microenvironment at both the macro- and microlevels and provides a basis for development of personalized treatment therapy.
Collapse
|
9
|
Savino A, De Marzo N, Provero P, Poli V. Meta-Analysis of Microdissected Breast Tumors Reveals Genes Regulated in the Stroma but Hidden in Bulk Analysis. Cancers (Basel) 2021; 13:3371. [PMID: 34282769 PMCID: PMC8268805 DOI: 10.3390/cancers13133371] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/22/2021] [Accepted: 06/29/2021] [Indexed: 02/06/2023] Open
Abstract
Transcriptome data provide a valuable resource for the study of cancer molecular mechanisms, but technical biases, sample heterogeneity, and small sample sizes result in poorly reproducible lists of regulated genes. Additionally, the presence of multiple cellular components contributing to cancer development complicates the interpretation of bulk transcriptomic profiles. To address these issues, we collected 48 microarray datasets derived from laser capture microdissected stroma or epithelium in breast tumors and performed a meta-analysis identifying robust lists of differentially expressed genes. This was used to create a database with carefully harmonized metadata that we make freely available to the research community. As predicted, combining the results of multiple datasets improved statistical power. Moreover, the separate analysis of stroma and epithelium allowed the identification of genes with different contributions in each compartment, which would not be detected by bulk analysis due to their distinct regulation in the two compartments. Our method can be profitably used to help in the discovery of biomarkers and the identification of functionally relevant genes in both the stroma and the epithelium. This database was made to be readily accessible through a user-friendly web interface.
Collapse
Affiliation(s)
- Aurora Savino
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126 Turin, Italy;
| | - Niccolò De Marzo
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126 Turin, Italy;
| | - Paolo Provero
- Department of Neurosciences “Rita Levi Montalcini”, University of Turin, Corso Massimo D’Azeglio 52, 10126 Turin, Italy;
- Center for Omics Sciences, Ospedale San Raffaele IRCCS, Via Olgettina 60, 20132 Milan, Italy
| | - Valeria Poli
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126 Turin, Italy;
| |
Collapse
|
10
|
Rozova VS, Anwer AG, Guller AE, Es HA, Khabir Z, Sokolova AI, Gavrilov MU, Goldys EM, Warkiani ME, Thiery JP, Zvyagin AV. Machine learning reveals mesenchymal breast carcinoma cell adaptation in response to matrix stiffness. PLoS Comput Biol 2021; 17:e1009193. [PMID: 34297718 PMCID: PMC8336795 DOI: 10.1371/journal.pcbi.1009193] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 08/04/2021] [Accepted: 06/17/2021] [Indexed: 12/31/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) and its reverse process, mesenchymal-epithelial transition (MET), are believed to play key roles in facilitating the metastatic cascade. Metastatic lesions often exhibit a similar epithelial-like state to that of the primary tumour, in particular, by forming carcinoma cell clusters via E-cadherin-mediated junctional complexes. However, the factors enabling mesenchymal-like micrometastatic cells to resume growth and reacquire an epithelial phenotype in the target organ microenvironment remain elusive. In this study, we developed a workflow using image-based cell profiling and machine learning to examine morphological, contextual and molecular states of individual breast carcinoma cells (MDA-MB-231). MDA-MB-231 heterogeneous response to the host organ microenvironment was modelled by substrates with controllable stiffness varying from 0.2kPa (soft tissues) to 64kPa (bone tissues). We identified 3 distinct morphological cell types (morphs) varying from compact round-shaped to flattened irregular-shaped cells with lamellipodia, predominantly populating 2-kPa and >16kPa substrates, respectively. These observations were accompanied by significant changes in E-cadherin and vimentin expression. Furthermore, we demonstrate that the bone-mimicking substrate (64kPa) induced multicellular cluster formation accompanied by E-cadherin cell surface localisation. MDA-MB-231 cells responded to different substrate stiffness by morphological adaptation, changes in proliferation rate and cytoskeleton markers, and cluster formation on bone-mimicking substrate. Our results suggest that the stiffest microenvironment can induce MET.
Collapse
Affiliation(s)
- Vlada S. Rozova
- ARC Centre of Excellence for Nanoscale Biophotonics, Macquarie University, Sydney, Australia
- Institute for Biology and Biomedicine, Lobachevsky State University, Nizhny Novgorod, Russia
| | - Ayad G. Anwer
- ARC Centre of Excellence for Nanoscale Biophotonics, Macquarie University, Sydney, Australia
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, Australia
| | - Anna E. Guller
- ARC Centre of Excellence for Nanoscale Biophotonics, Macquarie University, Sydney, Australia
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, Australia
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
| | | | - Zahra Khabir
- ARC Centre of Excellence for Nanoscale Biophotonics, Macquarie University, Sydney, Australia
| | - Anastasiya I. Sokolova
- Centre of Biomedical Engineering, Sechenov University, Moscow, Russia
- Laboratory of Medical Nanotechnologies, Federal Biomedical Agency, Moscow, Russia
| | - Maxim U. Gavrilov
- Centre of Biomedical Engineering, Sechenov University, Moscow, Russia
| | - Ewa M. Goldys
- ARC Centre of Excellence for Nanoscale Biophotonics, Macquarie University, Sydney, Australia
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, Australia
| | | | - Jean Paul Thiery
- Centre of Biomedical Engineering, Sechenov University, Moscow, Russia
- Bioland Laboratory, Guangzhou Regenerative Medicine and Health, Guangdong Laboratory, Guangzhou, China
| | - Andrei V. Zvyagin
- ARC Centre of Excellence for Nanoscale Biophotonics, Macquarie University, Sydney, Australia
- Centre of Biomedical Engineering, Sechenov University, Moscow, Russia
- Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
11
|
Parkes EE, Humphries MP, Gilmore E, Sidi FA, Bingham V, Phyu SM, Craig S, Graham C, Miller J, Griffin D, Salto-Tellez M, Madden SF, Kennedy RD, Bakhoum SF, McQuaid S, Buckley NE. The clinical and molecular significance associated with STING signaling in breast cancer. NPJ Breast Cancer 2021; 7:81. [PMID: 34172750 PMCID: PMC8233333 DOI: 10.1038/s41523-021-00283-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 05/27/2021] [Indexed: 12/22/2022] Open
Abstract
STING signaling in cancer is a crucial component of response to immunotherapy and other anti-cancer treatments. Currently, there is no robust method of measuring STING activation in cancer. Here, we describe an immunohistochemistry-based assay with digital pathology assessment of STING in tumor cells. Using this novel approach in estrogen receptor-positive (ER+) and ER- breast cancer, we identify perinuclear-localized expression of STING (pnSTING) in ER+ cases as an independent predictor of good prognosis, associated with immune cell infiltration and upregulation of immune checkpoints. Tumors with low pnSTING are immunosuppressed with increased infiltration of "M2"-polarized macrophages. In ER- disease, pnSTING does not appear to have a significant prognostic role with STING uncoupled from interferon responses. Importantly, a gene signature defining low pnSTING expression is predictive of poor prognosis in independent ER+ datasets. Low pnSTING is associated with chromosomal instability, MYC amplification and mTOR signaling, suggesting novel therapeutic approaches for this subgroup.
Collapse
Affiliation(s)
- Eileen E Parkes
- Department of Oncology, Medical Sciences Division, University of Oxford, Oxford, UK.
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, Northern Ireland, UK.
| | - Matthew P Humphries
- Precision Medicine Centre of Excellence, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Elaine Gilmore
- Precision Medicine Centre of Excellence, Queen's University Belfast, Belfast, Northern Ireland, UK
- School of Pharmacy, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Fatima A Sidi
- Precision Medicine Centre of Excellence, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Victoria Bingham
- Precision Medicine Centre of Excellence, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Su M Phyu
- Department of Oncology, Medical Sciences Division, University of Oxford, Oxford, UK
| | - Stephanie Craig
- Precision Medicine Centre of Excellence, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Catherine Graham
- Precision Medicine Centre of Excellence, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Joseph Miller
- Precision Medicine Centre of Excellence, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Daryl Griffin
- Precision Medicine Centre of Excellence, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Manuel Salto-Tellez
- Precision Medicine Centre of Excellence, Queen's University Belfast, Belfast, Northern Ireland, UK
- Department of Cellular Pathology, Belfast Health and Social Care Trust, Belfast, Northern Ireland, UK
- Integrated Pathology Programme, Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - Stephen F Madden
- Data Science Centre, RCSI University of Medicine and Health Sciences, Dublin, Ireland, UK
| | - Richard D Kennedy
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Samuel F Bakhoum
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Stephen McQuaid
- Precision Medicine Centre of Excellence, Queen's University Belfast, Belfast, Northern Ireland, UK
- Department of Cellular Pathology, Belfast Health and Social Care Trust, Belfast, Northern Ireland, UK
- Northern Ireland Biobank, Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Niamh E Buckley
- School of Pharmacy, Queen's University Belfast, Belfast, Northern Ireland, UK.
| |
Collapse
|
12
|
Tajbakhsh A, Gheibi Hayat SM, Movahedpour A, Savardashtaki A, Loveless R, Barreto GE, Teng Y, Sahebkar A. The complex roles of efferocytosis in cancer development, metastasis, and treatment. Biomed Pharmacother 2021; 140:111776. [PMID: 34062411 DOI: 10.1016/j.biopha.2021.111776] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 05/22/2021] [Accepted: 05/24/2021] [Indexed: 12/12/2022] Open
Abstract
When tumor cells are killed by targeted therapy, radiotherapy, or chemotherapy, they trigger their primary tumor by releasing pro-inflammatory cytokines. Microenvironmental interactions can also promote tumor heterogeneity and development. In this line, several immune cells within the tumor microenvironment, including macrophages, dendritic cells, regulatory T-cells, and CD8+ and CD4+ T cells, are involved in the clearance of apoptotic tumor cells through a process called efferocytosis. Although the efficiency of apoptotic tumor cell efferocytosis is positive under physiological conditions, there are controversies regarding its usefulness in treatment-induced apoptotic tumor cells (ATCs). Efferocytosis can show the limitation of cytotoxic treatments, such as chemotherapy and radiotherapy. Since cytotoxic treatments lead to extensive cell mortality, efferocytosis, and macrophage polarization toward an M2 phenotype, the immune response may get involved in tumor recurrence and metastasis. Tumor cells can use the anti-inflammatory effect of apoptotic tumor cell efferocytosis to induce an immunosuppressive condition that is tumor-tolerant. Since M2 polarization and efferocytosis are tumor-promoting processes, the receptors on macrophages act as potential targets for cancer therapy. Moreover, researchers have shown that efferocytosis-related molecules/pathways are potential targets for cancer therapy. These include phosphatidylserine and calreticulin, Tyro3, Axl, and Mer tyrosine kinase (MerTK), receptors of tyrosine kinase, indoleamine-2,3-dioxygenase 1, annexin V, CD47, TGF-β, IL-10, and macrophage phenotype switch are combined with conventional therapy, which can be more effective in cancer treatment. Thus, we set out to investigate the advantages and disadvantages of efferocytosis in treatment-induced apoptotic tumor cells.
Collapse
Affiliation(s)
- Amir Tajbakhsh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Mohammad Gheibi Hayat
- Department of Medical Biotechnology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Ahmad Movahedpour
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Savardashtaki
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Reid Loveless
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland; Health Research Institute, University of Limerick, Limerick, Ireland
| | - Yong Teng
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA 30912, USA; Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; Department of Medical Laboratory, Imaging and Radiologic Sciences, College of Allied Health, Augusta University, Augusta, GA 30912, USA
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
13
|
Vahidfar N, Aghanejad A, Ahmadzadehfar H, Farzanehfar S, Eppard E. Theranostic Advances in Breast Cancer in Nuclear Medicine. Int J Mol Sci 2021; 22:4597. [PMID: 33925632 PMCID: PMC8125561 DOI: 10.3390/ijms22094597] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 04/13/2021] [Accepted: 04/23/2021] [Indexed: 02/07/2023] Open
Abstract
The implication of 'theranostic' refers to targeting an identical receptor for diagnostic and therapeutic purposes, by the same radioligand, simultaneously or separately. In regard to extensive efforts, many considerable theranostic tracers have been developed in recent years. Emerging evidence strongly demonstrates the tendency of nuclear medicine towards therapies based on a diagnosis. This review is focused on the examples of targeted radiopharmaceuticals for the imaging and therapy of breast cancer.
Collapse
Affiliation(s)
- Nasim Vahidfar
- Department of Nuclear Medicine, Vali-Asr Hospital, Tehran University of Medical Sciences, Tehran 1419733141, Iran;
| | - Ayuob Aghanejad
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz 51368, Iran;
| | | | - Saeed Farzanehfar
- Department of Nuclear Medicine, Vali-Asr Hospital, Tehran University of Medical Sciences, Tehran 1419733141, Iran;
| | - Elisabeth Eppard
- Positronpharma SA. Rancagua 878, Santiago 7500621, Chile;
- Department of Nuclear Medicine, University Hospital Magdeburg, Leipziger Strass 44, 39120 Magdedurg, Germany
| |
Collapse
|
14
|
Jia Y, Wen W, Yang Y, Huang M, Ning Y, Jiao X, Liu S, Qin Y, Zhang M. The clinical role of combined serum C1q and hsCRP in predicting coronary artery disease. Clin Biochem 2021; 93:50-58. [PMID: 33861985 DOI: 10.1016/j.clinbiochem.2021.04.004] [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: 01/26/2021] [Revised: 03/19/2021] [Accepted: 04/06/2021] [Indexed: 10/21/2022]
Abstract
OBJECTIVE C1q has been shown to be associated with coronary heart disease (CAD) and can co-deposit with C-reactive protein (CRP) in atherosclerotic plaques. However, few studies have been conducted between C1q, CRP parameters and CAD. The aim of this study is to explore the relationship between C1q and CRP parameters and assess their clinical significance in CAD. METHODS 238 total patients who underwent coronary artery angiography were enrolled and divided into control group (n = 65), stable CAD group (n = 47) and unstable angina group (UA group, n = 126). Patients' data were collected from self-administered questionnaires and electrical medical records. The severity of coronary stenosis was presented by Gensini score. The relationship between C1q, CRP parameters and CAD were evaluated by multivariate regression analysis and their predicting performance were assessed by ROC analysis and odds ratio analysis. RESULTS Compared with control group, C1q was showed significantly lower in stable CAD (P = 0.004) and UA groups (P = 0.008), while hsCRP was higher in UA group (P = 0.024). Serum C1q was weakly positively associated with hsCRP (r = 0.24, P < 0.001) but not correlated with Gensini score. Logistic regression identified C1q (OR: 0.87 per 10 mg/L, 95% CI: 0.79-0.95, P = 0.001) and hsCRP (OR: 1.08 mg/L, 95% CI: 1.01-1.15, P = 0.032) as independent determinants of CAD. Furthermore, combined C1q and hsCRP level showed higher discriminatory accuracy in predicting CAD than C1q (AUC: 0.676 vs 0.585, P = 0.101; NRI: 10.4%, P = 0.049; IDI: 3.9%, P < 0.001) or hsCRP (AUC: 0.676 vs 0.585, P = 0.101; NRI: 16.7%, P = 0.006; IDI: 5.8%, P < 0.001). CONCLUSIONS Reduced serum C1q and increased hsCRP are independently associated with CAD and could be potential predictors for CAD diagnosis. Furthermore, combined C1q and hsCRP showed better performance in predicting CAD than using single one.
Collapse
Affiliation(s)
- Yifan Jia
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Wanwan Wen
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Yunxiao Yang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Mengling Huang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Yu Ning
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Xiaolu Jiao
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Sheng Liu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Yanwen Qin
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Ming Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China.
| |
Collapse
|
15
|
Jonasch E, Walker CL, Rathmell WK. Clear cell renal cell carcinoma ontogeny and mechanisms of lethality. Nat Rev Nephrol 2021; 17:245-261. [PMID: 33144689 PMCID: PMC8172121 DOI: 10.1038/s41581-020-00359-2] [Citation(s) in RCA: 328] [Impact Index Per Article: 82.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/22/2020] [Indexed: 02/07/2023]
Abstract
The molecular features that define clear cell renal cell carcinoma (ccRCC) initiation and progression are being increasingly defined. The TRACERx Renal studies and others that have described the interaction between tumour genomics and remodelling of the tumour microenvironment provide important new insights into the molecular drivers underlying ccRCC ontogeny and progression. Our understanding of common genomic and chromosomal copy number abnormalities in ccRCC, including chromosome 3p loss, provides a mechanistic framework with which to organize these abnormalities into those that drive tumour initiation events, those that drive tumour progression and those that confer lethality. Truncal mutations in ccRCC, including those in VHL, SET2, PBRM1 and BAP1, may engender genomic instability and promote defects in DNA repair pathways. The molecular features that arise from these defects enable categorization of ccRCC into clinically and therapeutically relevant subtypes. Consideration of the interaction of these subtypes with the tumour microenvironment reveals that specific mutations seem to modulate immune cell populations in ccRCC tumours. These findings present opportunities for disease prevention, early detection, prognostication and treatment.
Collapse
Affiliation(s)
- Eric Jonasch
- Department of Genitourinary Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Cheryl Lyn Walker
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA
| | - W Kimryn Rathmell
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
16
|
Yuan W, Yan J, Liu H, Li L, Wu B, Guo C, Zhang M. Identification of Prognostic Related Genes of Tumor Microenvironment Derived From Esophageal Cancer Patients. Pathol Oncol Res 2021; 27:589662. [PMID: 34257539 PMCID: PMC8262216 DOI: 10.3389/pore.2021.589662] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 03/05/2021] [Indexed: 12/29/2022]
Abstract
Background and Objective: Esophageal cancer (ESCA) is a commonly occurring cancer worldwide with poor survival and limited therapeutic options. Due to the lack of biomarkers that facilitate early detection, its treatment remains a great challenge. This study aims at identifying the tumor microenvironment (TME)-related genes, which might affect prognosis and accelerate clinical treatment for ESCA patients. Methods: We integrated the expression profiles from ESCA patients in The Cancer Genome Atlas. Then, we determined the stromal and immune scores of each sample using the R package. The Gene Expression Omnibus database was used to validate the expression profile of the key genes. Results: Tumor mutational burden showed a significant difference between the groups of ESCA patients with high and low ESTIMATE scores. We identified 859 intersection genes among patients with different immune and stromal scores. Moreover, gene ontology analysis demonstrated that these 859 intersection genes were closely related to adaptive immune response and regulation of lymphocyte activation. Kyoto Encyclopedia of Genes and Genomes showed the enrichment of cytokine-cytokine receptor interaction and chemokine signaling pathway in the TME. Furthermore, the protein–protein interaction network consisted of 175 nodes. We selected 35 hub genes, including ITGAM, CXCL10, CCR2, CCR5, and CCR1. Of these, 23 intersection genes predicted the overall survival rate. C1QA and FCER1G correlated with overall survival of the ESCA patients in the two databases. Conclusion: We identified a set of stromal and immune score-related prognostic differentially expressed genes that could influence the complexity of the TME. C1QA and FCER1G were identified and validated with respect to their role in the progression of ESCA.
Collapse
Affiliation(s)
- Wei Yuan
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,The Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jiaqin Yan
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongtao Liu
- College of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Ling Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - BoWen Wu
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,The Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Can Guo
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,The Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Mingzhi Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
17
|
Lin J, Guo Z, Wang S, Zheng X. Omission of Chemotherapy in HR+/HER2- Early Invasive Breast Cancer Based on Combined 6-IHC Score? Clin Breast Cancer 2021; 21:e565-e574. [PMID: 33674187 DOI: 10.1016/j.clbc.2021.01.011] [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: 09/04/2020] [Revised: 12/10/2020] [Accepted: 01/18/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Current methods of judging whether HR+/HER2- breast cancer (BC) require adjuvant therapy, such as Ki67 and multigene prognostic tests, cannot balance accuracy with the price most patients can afford. METHODS A retrospective analysis of 330 HR+/HER2- BC patients was conducted. Six BC-related genes (Cathepsin L2, MMP11, CyclinB1, Aurora A, Survivin, and Ki67) were screened using univariate and multivariate COX regression, and correlate clinical follow-up with immunohistochemical expression (designated as 6-IHC). All the included patients were divided randomly at a 7:3 ratio into training and testing cohorts. The cutoff prognosis index (PI) of 6-IHC was determined by multivariate Cox risk regression analysis after calculating the PI of each patient in training cohort and confirmed in testing cohort. Kaplan-Meier (KM) method was used to analyze Disease-free survival (DFS) and overall survival (OS). Six-IHC score and other factors associated with survival benefit of adjuvant chemotherapy were compared with Ki67 index. RESULTS The receiver operating characteristic curve analysis showed that the patients can be divided into 6-IHC score "High" and "Low" risk groups. The 8-year DFS and OS of the KM curves showed that chemotherapy did not significantly improve the DFS in the 6-IHC score "Low" risk group (P= 0.830), but significantly improved the DFS in the 6-IHC score "High" risk group (P = 0.012). CONCLUSIONS Combined 6-IHC score could be a reliable tool in predicting cancer-specific recurrences and survival in HR+/HER2-breast cancer patients, with additional advantages over using immunohistochemical expression of Ki67.
Collapse
Affiliation(s)
- Jiaman Lin
- Department of Breast Surgery, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Zihe Guo
- Department of Breast Surgery, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Shuo Wang
- Department of Breast Surgery, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Xinyu Zheng
- Department of Breast Surgery, First Affiliated Hospital, China Medical University, Shenyang, China; Lab 1, Cancer Institute, First Affiliated Hospital, China Medical University, Shenyang, China.
| |
Collapse
|
18
|
Park SB, Hwang KT, Chung CK, Roy D, Yoo C. Causal Bayesian gene networks associated with bone, brain and lung metastasis of breast cancer. Clin Exp Metastasis 2020; 37:657-674. [PMID: 33083937 DOI: 10.1007/s10585-020-10060-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 10/14/2020] [Indexed: 02/16/2023]
Abstract
Using a machine learning method, this study aimed to identify unique causal networks of genes associated with bone, brain, and lung metastasis of breast cancer. Bayesian network analysis identified differentially expressed genes in primary breast cancer tissues, in bone, brain, and lung breast cancer metastatic tissues, and the clinicopathological features of patients obtained from the Gene Expression Omnibus microarray datasets. We evaluated the causal Bayesian networks of breast metastasis to distant sites (bone, brain, or lung) by (i) measuring how well the structures of each specific type of breast cancer metastasis fit the data, (ii) comparing the structures with known experimental evidence, and (iii) reporting predictive capabilities of the structures. We report for the first time that the molecular gene signatures are specific to the different types of breast cancer metastasis. Several genes, including CHPF, ARC, ANGPTL4, NR2E1, SH2D1A, CTSW, POLR2J4, SPTLC1, ILK, ALDH3B1, PDE6A, SCTR, ADM, HEY1, KCNF1, and UVRAG, were found to be predictors of the risk for site-specific metastasis of breast cancer. Expression of POLR2JA, SPTLC1, ILK, ALDH3B1, and the estrogen receptor was significantly associated with breast cancer bone metastasis. Expression of PDE6A and NR2E1 was causally linked to breast cancer brain metastasis. Expression of HEY1, KCNF1, UVRAG, and the estrogen and progesterone receptors was strongly associated with breast cancer lung metastasis. The causal Bayesian network structures of these genes identify potential interactions among the genes in distant metastases of breast cancer, including to the bone, brain, and lung, and may serve as target candidates for treatment of breast cancer metastasis.
Collapse
Affiliation(s)
- Sung Bae Park
- Department of Neurosurgery, Seoul National University Boramae Medical Center, Seoul, Korea
| | - Ki-Tae Hwang
- Department of Surgery, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul, Korea
| | - Chun Kee Chung
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul, Korea.,Department of Neurosurgery, Clinical Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Deodutta Roy
- Department of Environmental Health Sciences, Stempel College of Public Health and Social Work, Florida International University, Miami, FL, USA.
| | - Changwon Yoo
- Department of Biostatistics, Robert Stempel College of Public Health and Social Work, Florida International University, 11200 SW 8th Street AHC5, Miami, FL, 33199, USA.
| |
Collapse
|
19
|
Anti gC1qR/p32/HABP1 Antibody Therapy Decreases Tumor Growth in an Orthotopic Murine Xenotransplant Model of Triple Negative Breast Cancer. Antibodies (Basel) 2020; 9:antib9040051. [PMID: 33036212 PMCID: PMC7709104 DOI: 10.3390/antib9040051] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 08/18/2020] [Accepted: 09/08/2020] [Indexed: 01/09/2023] Open
Abstract
gC1qR is highly expressed in breast cancer and plays a role in cancer cell proliferation. This study explored therapy with gC1qR monoclonal antibody 60.11, directed against the C1q binding domain of gC1qR, in a murine orthotopic xenotransplant model of triple negative breast cancer. MDA231 breast cancer cells were injected into the mammary fat pad of athymic nu/nu female mice. Mice were segregated into three groups (n = 5, each) and treated with the vehicle (group 1) or gC1qR antibody 60.11 (100 mg/kg) twice weekly, starting at day 3 post-implantation (group 2) or when the tumor volume reached 100 mm3 (group 3). At study termination (d = 35), the average tumor volume in the control group measured 895 ± 143 mm3, compared to 401 ± 48 mm3 and 701 ± 100 mm3 in groups 2 and 3, respectively (p < 0.05). Immunohistochemical staining of excised tumors revealed increased apoptosis (caspase 3 and TUNEL staining) in 60.11-treated mice compared to controls, and decreased angiogenesis (CD31 staining). Slightly decreased white blood cell counts were noted in 60.11-treated mice. Otherwise, no overt toxicities were observed. These data are the first to demonstrate an in vivo anti-tumor effect of 60.11 therapy in a mouse model of triple negative breast cancer.
Collapse
|
20
|
Elastosis in ERα-positive male breast cancer. Virchows Arch 2020; 478:257-263. [PMID: 32929565 PMCID: PMC7969537 DOI: 10.1007/s00428-020-02920-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 08/07/2020] [Accepted: 09/01/2020] [Indexed: 01/07/2023]
Abstract
In female breast cancer (BC), elastosis is strongly related to estrogen receptor alpha (ERα) expression. Male breast cancers almost invariably express ERα; so, the aim of this study was to investigate elastosis frequency in invasive male BC as well as clinicopathological correlations, in comparison with females. A total of 177 male BC cases and 135 female BC cases were included, all ERα-positive and invasive carcinoma of no special type. Elastosis on H&E-stained slides was scored in a four-tiered system as elastosis grade (EG) 0 (no elastosis) to EG3 (high amount of elastosis). EG scores in male BC were correlated to histopathological characteristics and overall surviva and compared with female BC EG scores. Male BC showed some degree of elastosis in 26/117 cases (22.2%) with none showing EG3, while female BC cases showed elastosis in 89/135 cases (65.9%) with 21.5% showing EG3 (p < 0.001). This difference retained its significance in multivariate logistic regression. In male BC cases, no significant correlations were found between the amount of elastosis and age, grade, mitotic activity index, and PgR. In addition, no significant prognostic value of elastosis was seen. In conclusion, despite high ERα expression, male BC showed significantly less elastosis than female BC. Elastosis did not show clinicopathological correlations or prognostic value. Therefore, elastosis seems to be a less useful ERα tissue biomarker with less clinical significance in male BC compared with females, pointing towards important BC sex differences.
Collapse
|
21
|
Larsson C, Ehinger A, Winslow S, Leandersson K, Klintman M, Dahl L, Vallon-Christersson J, Häkkinen J, Hegardt C, Manjer J, Saal L, Rydén L, Malmberg M, Borg Å, Loman N. Prognostic implications of the expression levels of different immunoglobulin heavy chain-encoding RNAs in early breast cancer. NPJ Breast Cancer 2020; 6:28. [PMID: 32656317 PMCID: PMC7338507 DOI: 10.1038/s41523-020-0170-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 06/02/2020] [Indexed: 12/22/2022] Open
Abstract
The extent and composition of the immune response in a breast cancer is one important prognostic factor for the disease. The aim of the current work was to refine the analysis of the humoral component of an immune response in breast tumors by quantifying mRNA expression of different immunoglobulin classes and study their association with prognosis. We used RNA-Seq data from two local population-based breast cancer cohorts to determine the expression of IGJ and immunoglobulin heavy (IGH) chain-encoding RNAs. The association with prognosis was investigated and public data sets were used to corroborate the findings. Except for IGHE and IGHD, mRNAs encoding heavy chains were generally detected at substantial levels and correlated with other immune-related genes. High IGHG1 mRNA was associated with factors related to poor prognosis such as estrogen receptor negativity, HER2 amplification, and high grade, whereas high IGHA2 mRNA levels were primarily associated with lower age at diagnosis. High IGHA2 and IGJ mRNA levels were associated with a more favorable prognosis both in univariable and multivariable Cox models. When adjusting for other prognostic factors, high IGHG1 mRNA levels were positively associated with improved prognosis. To our knowledge, these results are the first to demonstrate that expression of individual Ig class types has prognostic implications in breast cancer.
Collapse
Affiliation(s)
- Christer Larsson
- Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Anna Ehinger
- Oncology and Pathology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Sofia Winslow
- Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Karin Leandersson
- Cancer Immunology, Department of Translational Medicine, Lund University, Malmö, Sweden
| | - Marie Klintman
- Oncology and Pathology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Ludvig Dahl
- Oncology and Pathology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | | | - Jari Häkkinen
- Oncology and Pathology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Cecilia Hegardt
- Oncology and Pathology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Jonas Manjer
- Surgery, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Lao Saal
- Oncology and Pathology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Lisa Rydén
- Surgery, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Martin Malmberg
- Oncology and Pathology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Åke Borg
- Oncology and Pathology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Niklas Loman
- Oncology and Pathology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| |
Collapse
|
22
|
Terkelsen T, Russo F, Gromov P, Haakensen VD, Brunak S, Gromova I, Krogh A, Papaleo E. Secreted breast tumor interstitial fluid microRNAs and their target genes are associated with triple-negative breast cancer, tumor grade, and immune infiltration. Breast Cancer Res 2020; 22:73. [PMID: 32605588 PMCID: PMC7329449 DOI: 10.1186/s13058-020-01295-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 05/14/2020] [Indexed: 12/21/2022] Open
Abstract
Background Studies on tumor-secreted microRNAs point to a functional role of these in cellular communication and reprogramming of the tumor microenvironment. Uptake of tumor-secreted microRNAs by neighboring cells may result in the silencing of mRNA targets and, in turn, modulation of the transcriptome. Studying miRNAs externalized from tumors could improve cancer patient diagnosis and disease monitoring and help to pinpoint which miRNA-gene interactions are central for tumor properties such as invasiveness and metastasis. Methods Using a bioinformatics approach, we analyzed the profiles of secreted tumor and normal interstitial fluid (IF) microRNAs, from women with breast cancer (BC). We carried out differential abundance analysis (DAA), to obtain miRNAs, which were enriched or depleted in IFs, from patients with different clinical traits. Subsequently, miRNA family enrichment analysis was performed to assess whether any families were over-represented in the specific sets. We identified dysregulated genes in tumor tissues from the same cohort of patients and constructed weighted gene co-expression networks, to extract sets of co-expressed genes and co-abundant miRNAs. Lastly, we integrated miRNAs and mRNAs to obtain interaction networks and supported our findings using prediction tools and cancer gene databases. Results Network analysis showed co-expressed genes and miRNA regulators, associated with tumor lymphocyte infiltration. All of the genes were involved in immune system processes, and many had previously been associated with cancer immunity. A subset of these, BTLA, CXCL13, IL7R, LAMP3, and LTB, was linked to the presence of tertiary lymphoid structures and high endothelial venules within tumors. Co-abundant tumor interstitial fluid miRNAs within this network, including miR-146a and miR-494, were annotated as negative regulators of immune-stimulatory responses. One co-expression network encompassed differences between BC subtypes. Genes differentially co-expressed between luminal B and triple-negative breast cancer (TNBC) were connected with sphingolipid metabolism and predicted to be co-regulated by miR-23a. Co-expressed genes and TIF miRNAs associated with tumor grade were BTRC, CHST1, miR-10a/b, miR-107, miR-301a, and miR-454. Conclusion Integration of IF miRNAs and mRNAs unveiled networks associated with patient clinicopathological traits, and underlined molecular mechanisms, specific to BC sub-groups. Our results highlight the benefits of an integrative approach to biomarker discovery, placing secreted miRNAs within a biological context.
Collapse
Affiliation(s)
- Thilde Terkelsen
- Computational Biology Laboratory, Danish Cancer Society Research Center, Strandboulevarden 49, 2100, Copenhagen, Denmark
| | - Francesco Russo
- Computational Biology Laboratory, Danish Cancer Society Research Center, Strandboulevarden 49, 2100, Copenhagen, Denmark.,Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Pavel Gromov
- Breast Cancer Biology Group, Genome Integrity Unit, Danish Cancer Society Research Center, Strandboulevarden 49, 2100, Copenhagen, Denmark
| | - Vilde Drageset Haakensen
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Søren Brunak
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Irina Gromova
- Breast Cancer Biology Group, Genome Integrity Unit, Danish Cancer Society Research Center, Strandboulevarden 49, 2100, Copenhagen, Denmark
| | - Anders Krogh
- Unit of Computational and RNA Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Elena Papaleo
- Computational Biology Laboratory, Danish Cancer Society Research Center, Strandboulevarden 49, 2100, Copenhagen, Denmark. .,Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
23
|
Li B, Geng R, Wu Q, Yang Q, Sun S, Zhu S, Xu Z, Sun S. Alterations in Immune-Related Genes as Potential Marker of Prognosis in Breast Cancer. Front Oncol 2020; 10:333. [PMID: 32226776 PMCID: PMC7080956 DOI: 10.3389/fonc.2020.00333] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Accepted: 02/25/2020] [Indexed: 01/07/2023] Open
Abstract
The tumor microenvironment (TME) is a heterogeneous system that contributes to breast cancer progression. The Cancer Genome Atlas (TCGA) database provides global gene expression profiling data for further analysis of various malignancies, including breast cancer. Based on the ESTIMATE algorithm, immune and stromal scores were calculated according to immune or stromal components in the TME. We divided breast cancer cases into high- and low-score groups and identified differentially expressed genes (DEGs) that were significantly associated with overall survival. We performed enrichment analysis and constructed a protein-protein interaction network and found that the DEGs were mainly involved in primary immunodeficiency, T cell receptor signaling pathway and cytokine-cytokine receptor reaction. Furthermore, we explored the effect of aging on immune and stromal scores, which was validated by lower immune/stromal scores, lower infiltration of T cells and lower expression of immune checkpoints in the elder group. In conclusion, certain differentially expressed immune-related genes contribute to longer overall survival, and aging influences the immune microenvironment and immunotherapy efficacy by changing the tumor-infiltrating lymphocyte (TIL) abundance and checkpoint expression in breast cancer.
Collapse
Affiliation(s)
- Bei Li
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Rongxin Geng
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China.,Brain Tumor Clinical Center of Wuhan, Wuhan, China
| | - Qi Wu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, China.,Faculty of Medicine, University of Paris Sud-Saclay, Le Kremlin-Bicêtre, France
| | - Qian Yang
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Si Sun
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shan Zhu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhiliang Xu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shengrong Sun
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
24
|
Shaker H, Bundred NJ, Landberg G, Pritchard SA, Albadry H, Nicholson SL, Harries LJ, Heah JYE, Castle J, Kirwan CC. Breast cancer stromal clotting activation (Tissue Factor and thrombin): A pre-invasive phenomena that is prognostic in invasion. Cancer Med 2020; 9:1768-1778. [PMID: 31962001 PMCID: PMC7050075 DOI: 10.1002/cam4.2748] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 11/13/2019] [Accepted: 11/15/2019] [Indexed: 12/25/2022] Open
Abstract
Background Tumor stroma, of which fibroblasts are the most abundant cell, resembles a non‐healing wound, where a procoagulant environment creates a permissive milieu for cancer growth. We aimed to determine if tumor expression of coagulation factors (procoagulant phenotype), and systemic hypercoagulability, occur at the preinvasive (ductal carcinoma in situ; DCIS) stage and correlate with breast cancer subtype, disease‐free survival (DFS), and overall survival (OS). Methods In a prospective cohort of early breast cancer (DCIS, n = 76; invasive, n = 248) tumor, normal breast and plasma were examined. Fibroblast and epithelial expression of Tissue Factor (TF), thrombin, PAR1, PAR2, and plasma thrombin‐antithrombin (TAT) and D‐dimer were correlated with clinicopathological data, and 5‐year survival. Results Fibroblast expression of TF, thrombin, and PAR1 was increased in DCIS and invasive cancer compared to normal breast fibroblasts (P ≤ .003, all). Fibroblast TF, thrombin, PAR1, and PAR2 was increased in cancers with high Ki67, high grade, ER‐ (vs ER+), and HER2+ (vs HER2‐) (all P < .05). On univariate analysis, fibroblast TF expression was inversely associated with DFS (P = .04) and OS (P = .02). D‐dimer was higher in node positive (507 (CI: 411‐625) ng/mL, n = 68) vs negative patients (428 (CI: 387‐472) ng/mL, n = 171, P = .004) and inversely associated with OS (P = .047). On multivariate analysis, plasma TAT was associated with reduced OS (HR 3.26, CI 1.16‐3.1, P = .02), with a high plasma TAT (≥3.2 ng/mL) associated with > 3‐fold mortality risk compared to low TAT. Conclusion This demonstrates procoagulant phenotypic changes occur in fibroblasts at the preinvasive stage. Fibroblast procoagulant phenotype is associated with aggressive breast cancer subtypes and reduced survival. Coagulation may be a therapeutic target in breast cancer.
Collapse
Affiliation(s)
- Hudhaifah Shaker
- Faculty of Biology, Medicine and Health, Division of Cancer Sciences, School of Medical Sciences, Manchester Cancer Research Centre, University of Manchester, Manchester, UK
| | - Nigel J Bundred
- Faculty of Biology, Medicine and Health, Division of Cancer Sciences, School of Medical Sciences, Manchester Cancer Research Centre, University of Manchester, Manchester, UK
| | - Göran Landberg
- Department of Pathology, Institute for Biomedicine, Sahlgrenska Cancer Center, University of Gothenburg, Gothenburg, Sweden
| | - Susan A Pritchard
- Department of Histopathology, Manchester University NHS Foundation Trust, Wythenshawe, Manchester, UK
| | - Harith Albadry
- Department of Histopathology, Royal Liverpool and Broadgreen University Hospitals NHS Trust, Liverpool, UK
| | - Sarah L Nicholson
- Department of Histopathology, East Lancashire Hospitals NHS Trust, Blackburn, UK
| | - Lauren J Harries
- Department of Histopathology, Manchester University NHS Foundation Trust, Wythenshawe, Manchester, UK
| | - Jing Y E Heah
- The Nightingale Centre and Prevent Breast Cancer Research Centre, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| | - John Castle
- Faculty of Biology, Medicine and Health, Division of Cancer Sciences, School of Medical Sciences, Manchester Cancer Research Centre, University of Manchester, Manchester, UK
| | - Cliona C Kirwan
- Faculty of Biology, Medicine and Health, Division of Cancer Sciences, School of Medical Sciences, Manchester Cancer Research Centre, University of Manchester, Manchester, UK.,The Nightingale Centre and Prevent Breast Cancer Research Centre, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| |
Collapse
|
25
|
Targeting of the Cancer-Associated Fibroblast-T-Cell Axis in Solid Malignancies. J Clin Med 2019; 8:jcm8111989. [PMID: 31731701 PMCID: PMC6912330 DOI: 10.3390/jcm8111989] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 10/31/2019] [Accepted: 11/12/2019] [Indexed: 12/23/2022] Open
Abstract
The introduction of a wide range of immunotherapies in clinical practice has revolutionized the treatment of cancer in the last decade. The majority of these therapeutic modalities are centered on reinvigorating a tumor-reactive cytotoxic T-cell response. While impressive clinical successes are obtained, the majority of cancer patients still fail to show a clinical response, despite the fact that their tumors express antigens that can be recognized by the immune system. This is due to a series of other cellular actors, present in or attracted towards the tumor microenvironment, including regulatory T-cells, myeloid-derived suppressor cells and cancer-associated fibroblasts (CAFs). As the main cellular constituent of the tumor-associated stroma, CAFs form a heterogeneous group of cells which can drive cancer cell invasion but can also impair the migration and activation of T-cells through direct and indirect mechanisms. This singles CAFs out as an important next target for further optimization of T-cell based immunotherapies. Here, we review the recent literature on the role of CAFs in orchestrating T-cell activation and migration within the tumor microenvironment and discuss potential avenues for targeting the interactions between fibroblasts and T-cells.
Collapse
|
26
|
D’Angelo A, Sobhani N, Roviello G, Bagby S, Bonazza D, Bottin C, Giudici F, Zanconati F, De Manzini N, Guglielmi A, Generali D. Tumour infiltrating lymphocytes and immune-related genes as predictors of outcome in pancreatic adenocarcinoma. PLoS One 2019; 14:e0219566. [PMID: 31381571 PMCID: PMC6681957 DOI: 10.1371/journal.pone.0219566] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 06/26/2019] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND We investigated the correlation between pancreatic ductal adenocarcinoma patient prognosis and the presence of tumour infiltrating lymphocytes and expression of 521 immune system genes. METHODS Intratumoural CD3+, CD8+, and CD20+ lymphocytes were examined by immunohistochemistry in 12 PDAC patients with different outcomes who underwent pancreaticoduodenectomy. The results were correlated with gene expression profile using the digital multiplexed NanoString nCounter analysis system (NanoString Technologies, Seattle, WA, USA). RESULTS Twenty immune system genes were significantly differentially expressed in patients with a good prognosis relative to patients with a worse prognosis: TLR2 and TLR7 (Toll-like receptor superfamily); CD4, CD37, FOXP3, PTPRC (B cell and T cell signalling); IRF5, IRF8, STAT1, TFE3 (transcription factors); ANP32B, CCND3 (cell cycle); BTK (B cell development); TNF, TNFRF1A (TNF superfamily); HCK (leukocyte function); C1QA (complement system); BAX, PNMA1 (apoptosis); IKBKE (NFκB pathway). Differential expression was more than twice log 2 for TLR7, TNF, C1QA, FOXP3, and CD37. DISCUSSION Tumour infiltrating lymphocytes were present at higher levels in samples from patients with better prognosis. Our findings indicate that tumour infiltrating lymphocyte levels and expression level of the immune system genes listed above influence pancreatic ductal adenocarcinoma prognosis. This information could be used to improve selection of best responders to immune inhibitors.
Collapse
Affiliation(s)
- Alberto D’Angelo
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
- Department of Medical, Surgical, & Health Sciences, University of Trieste, Piazza Ospitale, Trieste, Italy
| | - Navid Sobhani
- Department of Medical, Surgical, & Health Sciences, University of Trieste, Piazza Ospitale, Trieste, Italy
- Breast Cancer Unit, ASST Cremona, Cremona, Italy
| | - Giandomenico Roviello
- Department of Medical, Surgical, & Health Sciences, University of Trieste, Piazza Ospitale, Trieste, Italy
| | - Stefan Bagby
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Deborah Bonazza
- Department of Medical Sciences, Ospedale di Cattinara, Università degli Studi di Trieste, Strada di Fiume, Trieste, Italy
| | - Cristina Bottin
- Department of Medical Sciences, Ospedale di Cattinara, Università degli Studi di Trieste, Strada di Fiume, Trieste, Italy
| | - Fabiola Giudici
- Department of Medical Sciences, Ospedale di Cattinara, Università degli Studi di Trieste, Strada di Fiume, Trieste, Italy
| | - Fabrizio Zanconati
- Department of Medical Sciences, Ospedale di Cattinara, Università degli Studi di Trieste, Strada di Fiume, Trieste, Italy
| | - Nicolo De Manzini
- Department of Medical Sciences, Ospedale di Cattinara, Università degli Studi di Trieste, Strada di Fiume, Trieste, Italy
| | - Alessandra Guglielmi
- Department of Medical, Surgical, & Health Sciences, University of Trieste, Piazza Ospitale, Trieste, Italy
| | - Daniele Generali
- Department of Medical, Surgical, & Health Sciences, University of Trieste, Piazza Ospitale, Trieste, Italy
- Breast Cancer Unit, ASST Cremona, Cremona, Italy
- Department of Medical Sciences, Ospedale di Cattinara, Università degli Studi di Trieste, Strada di Fiume, Trieste, Italy
| |
Collapse
|
27
|
Fox NS, Haider S, Harris AL, Boutros PC. Landscape of transcriptomic interactions between breast cancer and its microenvironment. Nat Commun 2019; 10:3116. [PMID: 31308365 PMCID: PMC6629667 DOI: 10.1038/s41467-019-10929-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 06/04/2019] [Indexed: 12/31/2022] Open
Abstract
Solid tumours comprise mixtures of tumour cells (TCs) and tumour-adjacent cells (TACs), and the intricate interconnections between these diverse populations shape the tumour’s microenvironment. Despite this complexity, clinical genomic profiling is typically performed from bulk samples, without distinguishing TCs from TACs. To better understand TC–TAC interactions, we computationally distinguish their transcriptomes in 1780 primary breast tumours. We show that TC and TAC mRNA abundances are divergently associated with clinical phenotypes, including tumour subtypes and patient survival. These differences reflect distinct responses of TCs and TACs to specific somatic driver mutations, particularly TP53. These data further elucidate how the molecular interplay between breast tumours and their microenvironment drives aggressive tumour phenotypes. The transcriptomic profile of tumour-adjacent cells provides important information about tumour context but its clinical utility is unclear. Here, in breast cancer, Fox et al. show that the mRNA abundances of tumour and tumour-adjacent cells hold prognostic information.
Collapse
Affiliation(s)
- Natalie S Fox
- Ontario Institute for Cancer Research, Toronto, ON, M5G 0A3, Canada. .,Department of Medical Biophysics, University of Toronto, Toronto, ON, M5G 1L7, Canada.
| | - Syed Haider
- Ontario Institute for Cancer Research, Toronto, ON, M5G 0A3, Canada.,Department of Oncology, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK.,The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW7 3RP, UK
| | - Adrian L Harris
- Department of Oncology, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Paul C Boutros
- Ontario Institute for Cancer Research, Toronto, ON, M5G 0A3, Canada. .,Department of Medical Biophysics, University of Toronto, Toronto, ON, M5G 1L7, Canada. .,Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, M5S 1A8, Canada. .,Department of Human Genetics, University of California, Los Angeles, CA, 90095, USA. .,Department of Urology, University of California, Los Angeles, CA, 90024, USA. .,Broad Stem Cell Research Center, University of California, Los Angeles, CA, 90095, USA. .,Institute for Precision Health, University of California, Los Angeles, CA, 90095, USA. .,Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, 90024, USA.
| |
Collapse
|
28
|
Schlafen-11 expression is associated with immune signatures and basal-like phenotype in breast cancer. Breast Cancer Res Treat 2019; 177:335-343. [PMID: 31222709 DOI: 10.1007/s10549-019-05313-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 06/05/2019] [Indexed: 12/21/2022]
Abstract
PURPOSE Breast cancer (BC) is a heterogeneous disorder, with variable response to systemic chemotherapy. Likewise, BC shows highly complex immune activation patterns, only in part reflecting classical histopathological subtyping. Schlafen-11 (SLFN11) is a nuclear protein we independently described as causal factor of sensitivity to DNA damaging agents (DDA) in cancer cell line models. SLFN11 has been reported as a predictive biomarker for DDA and PARP inhibitors in human neoplasms. SLFN11 has been implicated in several immune processes such as thymocyte maturation and antiviral response through the activation of interferon signaling pathway, suggesting its potential relevance as a link between immunity and cancer. In the present work, we investigated the transcriptional landscape of SLFN11, its potential prognostic value, and the clinico-pathological associations with its variability in BC. METHODS We assessed SLFN11 determinants in a gene expression meta-set of 5061 breast cancer patients annotated with clinical data and multigene signatures. RESULTS We found that 537 transcripts are highly correlated with SLFN11, identifying "immune response", "lymphocyte activation", and "T cell activation" as top Gene Ontology processes. We established a strong association of SLFN11 with stromal signatures of basal-like phenotype and response to chemotherapy in estrogen receptor negative (ER-) BC. We identified a distinct subgroup of patients, characterized by high SLFN11 levels, ER- status, basal-like phenotype, immune activation, and younger age. Finally, we observed an independent positive predictive role for SLFN11 in BC. CONCLUSIONS Our findings are suggestive of a relevant role for SLFN11 in BC and its immune and molecular variability.
Collapse
|
29
|
Mangogna A, Agostinis C, Bonazza D, Belmonte B, Zacchi P, Zito G, Romano A, Zanconati F, Ricci G, Kishore U, Bulla R. Is the Complement Protein C1q a Pro- or Anti-tumorigenic Factor? Bioinformatics Analysis Involving Human Carcinomas. Front Immunol 2019; 10:865. [PMID: 31130944 PMCID: PMC6509152 DOI: 10.3389/fimmu.2019.00865] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 04/04/2019] [Indexed: 01/04/2023] Open
Abstract
C1q is the first subcomponent of the classical pathway of the complement system and belongs to the C1q/Tumor Necrosis Factor superfamily. C1q can perform a diverse range of immune and non-immune functions in a complement-dependent as well as -independent manner. Being a pattern recognition molecule of the innate immunity, C1q can recognize a number of self, non-self and altered-self ligands and bring about effector mechanisms designed to clear pathogens via opsonisation and inflammatory response. C1q is locally synthesized by macrophages and dendritic cells, and thus, can get involved in a range of biological processes, such as angiogenesis and tissue remodeling, immune modulation, and immunologic tolerance. The notion of C1q involvement in the pathogenesis of cancer is still evolving. C1q appears to have a dual role in cancer: tumor promoting as well as tumor-protective, depending on the context of the disease. In the current study, we performed a bioinformatics analysis to investigate whether C1q can serve as a potential prognostic marker for human carcinoma. We used the Oncomine database and the survival analysis platforms Kaplan-Meier plotter. Our results showed that high levels of C1q have a favorable prognostic index in basal-like breast cancer for disease-free survival, and in HER2-positive breast cancer for overall survival, while it showed a pro-tumorigenic role of C1q in lung adenocarcinoma, and in clear cell renal cell carcinoma. This in silico study, if validated via a retrospective study, can be a step forward in establishing C1q as a new tool as a prognostic biomarker for various carcinoma.
Collapse
Affiliation(s)
| | - Chiara Agostinis
- Institute for Maternal and Child Health, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Burlo Garofolo, Trieste, Italy
| | - Deborah Bonazza
- Department of Medical, Surgical and Health Science, University of Trieste, Trieste, Italy
| | - Beatrice Belmonte
- Tumor Immunology Unit, Human Pathology Section, Department of Health Sciences, University of Palermo, Palermo, Italy
| | - Paola Zacchi
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Gabriella Zito
- Institute for Maternal and Child Health, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Burlo Garofolo, Trieste, Italy
| | - Andrea Romano
- Department of Medical, Surgical and Health Science, University of Trieste, Trieste, Italy
| | - Fabrizio Zanconati
- Department of Medical, Surgical and Health Science, University of Trieste, Trieste, Italy
| | - Giuseppe Ricci
- Institute for Maternal and Child Health, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Burlo Garofolo, Trieste, Italy.,Department of Medical, Surgical and Health Science, University of Trieste, Trieste, Italy
| | - Uday Kishore
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Roberta Bulla
- Department of Life Sciences, University of Trieste, Trieste, Italy
| |
Collapse
|
30
|
Yu-Qing Y, Lei W, Mei-Ling H, Jing-Jing X, Mei-Chen W, Jiang W, Jun-Sheng H, Rui L, Nan-Lin L. Clinical significance of 21-gene recurrence score assay for hormone receptor-positive, lymph node-negative breast cancer in early stage. Exp Mol Pathol 2019; 108:150-155. [PMID: 31026440 DOI: 10.1016/j.yexmp.2019.04.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 04/12/2019] [Accepted: 04/19/2019] [Indexed: 12/23/2022]
Abstract
OBJECTIVE To identify the relationship between clinical pathological characteristics and the recurrence score (RS) on a 21-gene expression assay in patients with hormone receptor-positive, node-negative breast cancer, as well as the effect of RS on adjuvant decision-making. METHODS The retrospective study was conducted among luminal breast cancer patients admitted to Xijing Hospital between October 10, 2016, and September 14, 2018. Real-time PCR was used for 21-genome detection. Based on the calculated RS, participants were classified into low-risk, moderate-risk, and high-risk groups. Single-factor analysis and multiple logistic regression analysis were performed to explore independent predictors of high RS. Moreover, the effect of RS on adjuvant decision-making was studied. RESULTS Two hundred twenty-two patients with luminal breast cancer, aged 48.3 ± 9.66, were enrolled. Among them, 33.8% had low (13 ± 3.34), 45.5% intermediate (23 ± 3.65), and 20.7% high (37 ± 3.44) RS. According to the single-factor analysis, age, tumor size, Ki-67, molecular subtype, CK5/6 expression, E-cadherin level, and histological grade were positively associated with high RS. Multiple logistic analyses showed that tumor size and histological grade were independent variables that might predict high RS in patients with hormone receptor-positive, node-negative breast cancer. For adjuvant decision-making, the proportion of adjuvant chemotherapy in the intermediate-/high-risk groups was higher than that in the low-risk group, P < 0.001. Compared with the data worldwide, the changes of treatment selection in the present study were similar to those in Japan (23.0% vs. 26%) and America (23.0% vs. 23.0%). Considering the pathology types, 14.3% of patients with invasive breast cancer with lower RS changed treatment recommendations, predominantly from chemo-endocrine to endocrine treatment alone, whereas the percentage in intermediate/high RS groups was 8.1%. CONCLUSIONS Tumor size and histological grade were independent variables, predicting high risk in patients with hormone receptor-positive, node-negative breast cancer; 21-gene RS assessment was potentially a critical tool in guiding adjuvant decision-making in China.
Collapse
Affiliation(s)
- Yang Yu-Qing
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Air Force Medical University, Xi'an 710032, Shaanxi, China
| | - Wang Lei
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Air Force Medical University, Xi'an 710032, Shaanxi, China
| | - Huang Mei-Ling
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Air Force Medical University, Xi'an 710032, Shaanxi, China
| | - Xiao Jing-Jing
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Air Force Medical University, Xi'an 710032, Shaanxi, China
| | - Wei Mei-Chen
- Department of Pathology, Xijing Hospital, Air Force Medical University, Xi'an 710032, Shaanxi, China
| | - Wu Jiang
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Air Force Medical University, Xi'an 710032, Shaanxi, China
| | - Hao Jun-Sheng
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Air Force Medical University, Xi'an 710032, Shaanxi, China
| | - Ling Rui
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Air Force Medical University, Xi'an 710032, Shaanxi, China.
| | - Li Nan-Lin
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Air Force Medical University, Xi'an 710032, Shaanxi, China.
| |
Collapse
|
31
|
Santucci-Pereira J, Zeleniuch-Jacquotte A, Afanasyeva Y, Zhong H, Slifker M, Peri S, Ross EA, López de Cicco R, Zhai Y, Nguyen T, Sheriff F, Russo IH, Su Y, Arslan AA, Bordas P, Lenner P, Åhman J, Landström Eriksson AS, Johansson R, Hallmans G, Toniolo P, Russo J. Genomic signature of parity in the breast of premenopausal women. Breast Cancer Res 2019; 21:46. [PMID: 30922380 PMCID: PMC6438043 DOI: 10.1186/s13058-019-1128-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 03/14/2019] [Indexed: 12/17/2022] Open
Abstract
Background Full-term pregnancy (FTP) at an early age confers long-term protection against breast cancer. Previously, we reported that a FTP imprints a specific gene expression profile in the breast of postmenopausal women. Herein, we evaluated gene expression changes induced by parity in the breast of premenopausal women. Methods Gene expression profiling of normal breast tissue from 30 nulliparous (NP) and 79 parous (P) premenopausal volunteers was performed using Affymetrix microarrays. In addition to a discovery/validation analysis, we conducted an analysis of gene expression differences in P vs. NP women as a function of time since last FTP. Finally, a laser capture microdissection substudy was performed to compare the gene expression profile in the whole breast biopsy with that in the epithelial and stromal tissues. Results Discovery/validation analysis identified 43 differentially expressed genes in P vs. NP breast. Analysis of expression as a function of time since FTP revealed 286 differentially expressed genes (238 up- and 48 downregulated) comparing all P vs. all NP, and/or P women whose last FTP was less than 5 years before biopsy vs. all NP women. The upregulated genes showed three expression patterns: (1) transient: genes upregulated after FTP but whose expression levels returned to NP levels. These genes were mainly related to immune response, specifically activation of T cells. (2) Long-term changing: genes upregulated following FTP, whose expression levels decreased with increasing time since FTP but did not return to NP levels. These were related to immune response and development. (3) Long-term constant: genes that remained upregulated in parous compared to nulliparous breast, independently of time since FTP. These were mainly involved in development/cell differentiation processes, and also chromatin remodeling. Lastly, we found that the gene expression in whole tissue was a weighted average of the expression in epithelial and stromal tissues. Conclusions Genes transiently activated by FTP may have a role in protecting the mammary gland against neoplastically transformed cells through activation of T cells. Furthermore, chromatin remodeling and cell differentiation, represented by the genes that are maintained upregulated long after the FTP, may be responsible for the lasting preventive effect against breast cancer. Electronic supplementary material The online version of this article (10.1186/s13058-019-1128-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Julia Santucci-Pereira
- The Irma H. Russo, MD Breast Cancer Research Laboratory, Fox Chase Cancer Center - Temple University Health System, 333 Cottman Ave, P2051, Philadelphia, PA, 19111, USA.
| | - Anne Zeleniuch-Jacquotte
- Division of Epidemiology, Department of Population Health, New York University School of Medicine, New York, NY, 10016, USA.,New York University Perlmutter Cancer Center, New York, NY, 10016, USA
| | - Yelena Afanasyeva
- Division of Epidemiology, Department of Population Health, New York University School of Medicine, New York, NY, 10016, USA
| | - Hua Zhong
- Division of Epidemiology, Department of Population Health, New York University School of Medicine, New York, NY, 10016, USA
| | - Michael Slifker
- Department of Biostatistics and Bioinformatics, Fox Chase Cancer Center - Temple University Health System, Philadelphia, PA, 19111, USA
| | - Suraj Peri
- Department of Biostatistics and Bioinformatics, Fox Chase Cancer Center - Temple University Health System, Philadelphia, PA, 19111, USA
| | - Eric A Ross
- Department of Biostatistics and Bioinformatics, Fox Chase Cancer Center - Temple University Health System, Philadelphia, PA, 19111, USA
| | - Ricardo López de Cicco
- The Irma H. Russo, MD Breast Cancer Research Laboratory, Fox Chase Cancer Center - Temple University Health System, 333 Cottman Ave, P2051, Philadelphia, PA, 19111, USA
| | - Yubo Zhai
- The Irma H. Russo, MD Breast Cancer Research Laboratory, Fox Chase Cancer Center - Temple University Health System, 333 Cottman Ave, P2051, Philadelphia, PA, 19111, USA
| | - Theresa Nguyen
- The Irma H. Russo, MD Breast Cancer Research Laboratory, Fox Chase Cancer Center - Temple University Health System, 333 Cottman Ave, P2051, Philadelphia, PA, 19111, USA
| | - Fathima Sheriff
- The Irma H. Russo, MD Breast Cancer Research Laboratory, Fox Chase Cancer Center - Temple University Health System, 333 Cottman Ave, P2051, Philadelphia, PA, 19111, USA
| | - Irma H Russo
- The Irma H. Russo, MD Breast Cancer Research Laboratory, Fox Chase Cancer Center - Temple University Health System, 333 Cottman Ave, P2051, Philadelphia, PA, 19111, USA
| | - Yanrong Su
- The Irma H. Russo, MD Breast Cancer Research Laboratory, Fox Chase Cancer Center - Temple University Health System, 333 Cottman Ave, P2051, Philadelphia, PA, 19111, USA
| | - Alan A Arslan
- Division of Epidemiology, Department of Population Health, New York University School of Medicine, New York, NY, 10016, USA.,Department of Obstetrics and Gynecology, New York University School of Medicine, New York, NY, 10016, USA
| | - Pal Bordas
- Sunderby Hospital, Luleå and the Norrbotten Mammography Screening Program, Luleå, Sweden.,Departments of Radiation Sciences and Oncology, Umeå University, Umeå, Sweden
| | - Per Lenner
- Departments of Radiation Sciences and Oncology, Umeå University, Umeå, Sweden
| | - Janet Åhman
- Sunderby Hospital, Luleå and the Norrbotten Mammography Screening Program, Luleå, Sweden
| | | | | | - Göran Hallmans
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Paolo Toniolo
- Department of Obstetrics and Gynecology, New York University School of Medicine, New York, NY, 10016, USA
| | - Jose Russo
- The Irma H. Russo, MD Breast Cancer Research Laboratory, Fox Chase Cancer Center - Temple University Health System, 333 Cottman Ave, P2051, Philadelphia, PA, 19111, USA
| |
Collapse
|
32
|
Six novel immunoglobulin genes as biomarkers for better prognosis in triple-negative breast cancer by gene co-expression network analysis. Sci Rep 2019; 9:4484. [PMID: 30872752 PMCID: PMC6418134 DOI: 10.1038/s41598-019-40826-w] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 02/22/2019] [Indexed: 02/06/2023] Open
Abstract
Gene co-expression network analysis (GCNA) can detect alterations in regulatory activities in case/control comparisons. We propose a framework to detect novel genes and networks for predicting breast cancer recurrence. Thirty-four prognosis candidate genes were selected based on a literature review. Four Gene Expression Omnibus Series (GSE) microarray datasets (n = 920) were used to create gene co-expression networks based on these candidates. We applied the framework to four comparison groups according to node (+/−) and recurrence (+/−). We identified a sub-network containing two candidate genes (LST1 and IGHM) and six novel genes (IGHA1, IGHD, IGHG1, IGHG3, IGLC2, and IGLJ3) related to B cell-specific immunoglobulin. These novel genes were correlated with recurrence under the control of node status and were found to function as tumor suppressors; higher mRNA expression indicated a lower risk of recurrence (hazard ratio, HR = 0.87, p = 0.001). We created an immune index score by performing principle component analysis and divided the genes into low and high groups. This discrete index significantly predicted relapse-free survival (RFS) (high: HR = 0.77, p = 0.019; low: control). Public tool KM Plotter and TCGA-BRCA gene expression data were used to validate. We confirmed these genes are correlated with RFS and distal metastasis-free survival (DMFS) in triple-negative breast cancer (TNBC) and general breast cancer.
Collapse
|
33
|
Escala-Garcia M, Guo Q, Dörk T, Canisius S, Keeman R, Dennis J, Beesley J, Lecarpentier J, Bolla MK, Wang Q, Abraham J, Andrulis IL, Anton-Culver H, Arndt V, Auer PL, Beckmann MW, Behrens S, Benitez J, Bermisheva M, Bernstein L, Blomqvist C, Boeckx B, Bojesen SE, Bonanni B, Børresen-Dale AL, Brauch H, Brenner H, Brentnall A, Brinton L, Broberg P, Brock IW, Brucker SY, Burwinkel B, Caldas C, Caldés T, Campa D, Canzian F, Carracedo A, Carter BD, Castelao JE, Chang-Claude J, Chanock SJ, Chenevix-Trench G, Cheng TYD, Chin SF, Clarke CL, Cordina-Duverger E, Couch FJ, Cox DG, Cox A, Cross SS, Czene K, Daly MB, Devilee P, Dunn JA, Dunning AM, Durcan L, Dwek M, Earl HM, Ekici AB, Eliassen AH, Ellberg C, Engel C, Eriksson M, Evans DG, Figueroa J, Flesch-Janys D, Flyger H, Gabrielson M, Gago-Dominguez M, Galle E, Gapstur SM, García-Closas M, García-Sáenz JA, Gaudet MM, George A, Georgoulias V, Giles GG, Glendon G, Goldgar DE, González-Neira A, Alnæs GIG, Grip M, Guénel P, Haeberle L, Hahnen E, Haiman CA, Håkansson N, Hall P, Hamann U, Hankinson S, Harkness EF, Harrington PA, Hart SN, Hartikainen JM, Hein A, Hillemanns P, Hiller L, Holleczek B, Hollestelle A, Hooning MJ, Hoover RN, Hopper JL, Howell A, Huang G, Humphreys K, Hunter DJ, Janni W, John EM, Jones ME, Jukkola-Vuorinen A, Jung A, Kaaks R, Kabisch M, Kaczmarek K, Kerin MJ, Khan S, Khusnutdinova E, Kiiski JI, Kitahara CM, Knight JA, Ko YD, Koppert LB, Kosma VM, Kraft P, Kristensen VN, Krüger U, Kühl T, Lambrechts D, Le Marchand L, Lee E, Lejbkowicz F, Li L, Lindblom A, Lindström S, Linet M, Lissowska J, Lo WY, Loibl S, Lubiński J, Lux MP, MacInnis RJ, Maierthaler M, Maishman T, Makalic E, Mannermaa A, Manoochehri M, Manoukian S, Margolin S, Martinez ME, Mavroudis D, McLean C, Meindl A, Middha P, Miller N, Milne RL, Moreno F, Mulligan AM, Mulot C, Nassir R, Neuhausen SL, Newman WT, Nielsen SF, Nordestgaard BG, Norman A, Olsson H, Orr N, Pankratz VS, Park-Simon TW, Perez JIA, Pérez-Barrios C, Peterlongo P, Petridis C, Pinchev M, Prajzendanc K, Prentice R, Presneau N, Prokofieva D, Pylkäs K, Rack B, Radice P, Ramachandran D, Rennert G, Rennert HS, Rhenius V, Romero A, Roylance R, Saloustros E, Sawyer EJ, Schmidt DF, Schmutzler RK, Schneeweiss A, Schoemaker MJ, Schumacher F, Schwentner L, Scott RJ, Scott C, Seynaeve C, Shah M, Simard J, Smeets A, Sohn C, Southey MC, Swerdlow AJ, Talhouk A, Tamimi RM, Tapper WJ, Teixeira MR, Tengström M, Terry MB, Thöne K, Tollenaar RAEM, Tomlinson I, Torres D, Truong T, Turman C, Turnbull C, Ulmer HU, Untch M, Vachon C, van Asperen CJ, van den Ouweland AMW, van Veen EM, Wendt C, Whittemore AS, Willett W, Winqvist R, Wolk A, Yang XR, Zhang Y, Easton DF, Fasching PA, Nevanlinna H, Eccles DM, Pharoah PDP, Schmidt MK. Genome-wide association study of germline variants and breast cancer-specific mortality. Br J Cancer 2019; 120:647-657. [PMID: 30787463 PMCID: PMC6461853 DOI: 10.1038/s41416-019-0393-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 01/02/2019] [Accepted: 01/14/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND We examined the associations between germline variants and breast cancer mortality using a large meta-analysis of women of European ancestry. METHODS Meta-analyses included summary estimates based on Cox models of twelve datasets using ~10.4 million variants for 96,661 women with breast cancer and 7697 events (breast cancer-specific deaths). Oestrogen receptor (ER)-specific analyses were based on 64,171 ER-positive (4116) and 16,172 ER-negative (2125) patients. We evaluated the probability of a signal to be a true positive using the Bayesian false discovery probability (BFDP). RESULTS We did not find any variant associated with breast cancer-specific mortality at P < 5 × 10-8. For ER-positive disease, the most significantly associated variant was chr7:rs4717568 (BFDP = 7%, P = 1.28 × 10-7, hazard ratio [HR] = 0.88, 95% confidence interval [CI] = 0.84-0.92); the closest gene is AUTS2. For ER-negative disease, the most significant variant was chr7:rs67918676 (BFDP = 11%, P = 1.38 × 10-7, HR = 1.27, 95% CI = 1.16-1.39); located within a long intergenic non-coding RNA gene (AC004009.3), close to the HOXA gene cluster. CONCLUSIONS We uncovered germline variants on chromosome 7 at BFDP < 15% close to genes for which there is biological evidence related to breast cancer outcome. However, the paucity of variants associated with mortality at genome-wide significance underpins the challenge in providing genetic-based individualised prognostic information for breast cancer patients.
Collapse
Affiliation(s)
- Maria Escala-Garcia
- The Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Division of Molecular Pathology, Amsterdam, The Netherlands
| | - Qi Guo
- University of Cambridge, Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, Cambridge, UK.
| | - Thilo Dörk
- Hannover Medical School, Gynaecology Research Unit, Hannover, Germany
| | - Sander Canisius
- The Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Division of Molecular Pathology, Amsterdam, The Netherlands
- The Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Division of Molecular Carcinogenesis, Amsterdam, The Netherlands
| | - Renske Keeman
- The Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Division of Molecular Pathology, Amsterdam, The Netherlands
| | - Joe Dennis
- University of Cambridge, Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, Cambridge, UK
| | - Jonathan Beesley
- QIMR Berghofer Medical Research Institute, Department of Genetics and Computational Biology, Brisbane, Queensland, Australia
| | - Julie Lecarpentier
- University of Cambridge, Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, Cambridge, UK
| | - Manjeet K Bolla
- University of Cambridge, Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, Cambridge, UK
| | - Qin Wang
- University of Cambridge, Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, Cambridge, UK
| | - Jean Abraham
- University of Cambridge, Centre for Cancer Genetic Epidemiology, Department of Oncology, Cambridge, UK
- Cambridge Experimental Cancer Medicine Centre, Cambridge, UK
- University of Cambridge NHS Foundation Hospitals, Cambridge Breast Unit and NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - Irene L Andrulis
- Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, Fred A. Litwin Center for Cancer Genetics, Toronto, ON, Canada
- University of Toronto, Department of Molecular Genetics, Toronto, ON, Canada
| | - Hoda Anton-Culver
- University of California Irvine, Department of Epidemiology, Genetic Epidemiology Research Institute, Irvine, CA, USA
| | - Volker Arndt
- German Cancer Research Center (DKFZ), Division of Clinical Epidemiology and Aging Research, Heidelberg, Germany
| | - Paul L Auer
- Fred Hutchinson Cancer Research Center, Cancer Prevention Program, Seattle, WA, USA
- University of Wisconsin-Milwaukee, Zilber School of Public Health, Milwaukee, WI, USA
| | - Matthias W Beckmann
- University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Department of Gynecology and Obstetrics, Comprehensive Cancer Center ER-EMN, Erlangen, Germany
| | - Sabine Behrens
- German Cancer Research Center (DKFZ), Division of Cancer Epidemiology, Heidelberg, Germany
| | - Javier Benitez
- Spanish National Cancer Research Centre (CNIO), Human Cancer Genetics Programme, Madrid, Spain
- Biomedical Network on Rare Diseases (CIBERER), Madrid, Spain
| | - Marina Bermisheva
- Ufa Scientific Center of Russian Academy of Sciences, Institute of Biochemistry and Genetics, Ufa, Russia
| | - Leslie Bernstein
- Beckman Research Institute of City of Hope, Department of Population Sciences, Duarte, CA, USA
| | - Carl Blomqvist
- University of Helsinki, Department of Oncology, Helsinki University Hospital, Helsinki, Finland
- Örebro University Hospital, Department of Oncology, Örebro, Sweden
| | - Bram Boeckx
- VIB, VIB Center for Cancer Biology, Leuven, Belgium
- University of Leuven, Laboratory for Translational Genetics, Department of Human Genetics, Leuven, Belgium
| | - Stig E Bojesen
- Copenhagen University Hospital, Copenhagen General Population Study, Herlevand Gentofte Hospital, Herlev, Denmark
- Copenhagen University Hospital, Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Herlev, Denmark
- University of Copenhagen, Faculty of Health and Medical Sciences, Copenhagen, Denmark
| | - Bernardo Bonanni
- Division of Cancer Prevention and Genetics, IEO, European Institute of Oncology IRCCS Milan, Milan, 20141, Italy
| | - Anne-Lise Børresen-Dale
- Oslo University Hospital-Radiumhospitalet, Department of Cancer Genetics, Institute for Cancer Research, Oslo, Norway
- University of Oslo, Institute of Clinical Medicine, Faculty of Medicine, Oslo, Norway
- Department of Research, Vestre Viken Hospital, Drammen, Norway; Section for Breast- and Endocrine Surgery, Department of Cancer, Division of Surgery, Cancer and Transplantation Medicine, Oslo University Hospital-Ullevål, Oslo, Norway
- Department of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
- Department of Pathology at Akershus University hospital, Lørenskog, Norway
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Department of Oncology, Division of Surgery and Cancer and Transplantation Medicine, Oslo University Hospital-Radiumhospitalet, Oslo, Norway
- National Advisory Unit on Late Effects after Cancer Treatment, Department of Oncology, Oslo University Hospital, Oslo, Norway
- Department of Oncology, Akershus University Hospital, Lørenskog, Norway
- Breast Cancer Research Consortium, Oslo University Hospital, Oslo, Norway
| | - Hiltrud Brauch
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tübingen, Tübingen, Germany
- German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Hermann Brenner
- German Cancer Research Center (DKFZ), Division of Clinical Epidemiology and Aging Research, Heidelberg, Germany
- German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany
- German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Division of Preventive Oncology, Heidelberg, Germany
| | - Adam Brentnall
- Queen Mary University of London, Centre for Cancer Prevention, Wolfson Institute of Preventive Medicine, London, UK
| | - Louise Brinton
- National Cancer Institute, Division of Cancer Epidemiology and Genetics, Bethesda, MD, USA
| | - Per Broberg
- Lund University, Department of Cancer Epidemiology, Clinical Sciences, Lund, Sweden
| | - Ian W Brock
- University of Sheffield, Sheffield Institute for Nucleic Acids (SInFoNiA), Department of Oncology and Metabolism, Sheffield, UK
| | - Sara Y Brucker
- University of Tübingen, Department of Gynecology and Obstetrics, Tübingen, Germany
| | - Barbara Burwinkel
- University of Heidelberg, Department of Obstetrics and Gynecology, Heidelberg, Germany
- German Cancer Research Center (DKFZ), Molecular Epidemiology Group, C080, Heidelberg, Germany
| | - Carlos Caldas
- Cambridge Experimental Cancer Medicine Centre, Cambridge, UK
- University of Cambridge NHS Foundation Hospitals, Cambridge Breast Unit and NIHR Cambridge Biomedical Research Centre, Cambridge, UK
- The Institute of Cancer Research, Section of Cancer Genetics, London, UK
| | - Trinidad Caldés
- Instituto de Investigación Sanitaria San Carlos (IdISSC), Centro Investigación Biomédica en Red de Cáncer (CIBERONC), Medical Oncology Department, Hospital Cl'nico San Carlos, Madrid, Spain
| | - Daniele Campa
- German Cancer Research Center (DKFZ), Division of Cancer Epidemiology, Heidelberg, Germany
- University of Pisa, Department of Biology, Pisa, Italy
| | - Federico Canzian
- German Cancer Research Center (DKFZ), Molecular Epidemiology Group, C080, Heidelberg, Germany
| | - Angel Carracedo
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Genomic Medicine Group, Galician Foundation of Genomic Medicine, SERGAS, Santiago de Compostela, Spain
- Universidad de Santiago de Compostela, Centro de Investigación en Red de Enfermedades Raras (CIBERER), Santiago De Compostela, Spain
- King Abdulaziz University, Center of Excellence in Genomic Medicine, Jeddah, Kingdom of Saudi Arabia
| | - Brian D Carter
- American Cancer Society, Epidemiology Research Program, Atlanta, GA, USA
| | - Jose E Castelao
- Instituto de Investigación Sanitaria Galicia Sur (IISGS), Xerencia de Xestion Integrada de Vigo-SERGAS, Oncology and Genetics Unit, Vigo, Spain
| | - Jenny Chang-Claude
- German Cancer Research Center (DKFZ), Division of Cancer Epidemiology, Heidelberg, Germany
- University Medical Center Hamburg-Eppendorf, Cancer Epidemiology Group, University Cancer Center Hamburg (UCCH), Hamburg, Germany
| | - Stephen J Chanock
- National Cancer Institute, Division of Cancer Epidemiology and Genetics, Bethesda, MD, USA
| | - Georgia Chenevix-Trench
- QIMR Berghofer Medical Research Institute, Department of Genetics and Computational Biology, Brisbane, Queensland, Australia
| | - Ting-Yuan David Cheng
- Roswell Park Cancer Institute, Division of Cancer Prevention and Control, Buffalo, NY, USA
| | - Suet-Feung Chin
- University of Cambridge, Cancer Research UK Cambridge Institute, Cambridge, UK
| | - Christine L Clarke
- University of Sydney, Westmead Institute for Medical Research, Sydney, NSW, Australia
| | - Emilie Cordina-Duverger
- INSERM, University Paris-Sud, University Paris-Saclay, Cancer & Environment Group, Center for Research in Epidemiology and Population Health (CESP), Villejuif, France
| | - Fergus J Couch
- Mayo Clinic, Department of Laboratory Medicine and Pathology, Rochester, MN, USA
| | - David G Cox
- Imperial College London, Department of Epidemiology and Biostatistics, School of Public Health, London, UK
- Cancer Research Center of Lyon, INSERM U1052, Lyon, France
| | - Angela Cox
- University of Sheffield, Sheffield Institute for Nucleic Acids (SInFoNiA), Department of Oncology and Metabolism, Sheffield, UK
| | - Simon S Cross
- University of Sheffield, Academic Unit of Pathology, Department of Neuroscience, Sheffield, UK
| | - Kamila Czene
- Karolinska Institutet, Department of Medical Epidemiology and Biostatistics, Stockholm, Sweden
| | - Mary B Daly
- Fox Chase Cancer Center, Department of Clinical Genetics, Philadelphia, PA, USA
| | - Peter Devilee
- Leiden University Medical Center, Department of Pathology, Leiden, The Netherlands
- Leiden University Medical Center, Department of Human Genetics, Leiden, The Netherlands
| | - Janet A Dunn
- University of Warwick, Warwick Clinical Trials Unit, Coventry, UK
| | - Alison M Dunning
- University of Cambridge, Centre for Cancer Genetic Epidemiology, Department of Oncology, Cambridge, UK
| | - Lorraine Durcan
- University of Southampton, Southampton Clinical Trials Unit, Faculty of Medicine, Southampton, UK
- University of Southampton, Cancer Sciences Academic Unit, Faculty of Medicine, Southampton, UK
| | - Miriam Dwek
- University of Westminster, Department of Biomedical Sciences, Faculty of Science and Technology, London, UK
| | - Helena M Earl
- University of Cambridge NHS Foundation Hospitals, Cambridge Breast Unit and NIHR Cambridge Biomedical Research Centre, Cambridge, UK
- University of Cambridge, Department of Oncology, Cambridge, UK
| | - Arif B Ekici
- Friedrich-Alexander University Erlangen-Nuremberg, Comprehensive Cancer Center Erlangen-EMN, Institute of Human Genetics, University Hospital Erlangen, Erlangen, Germany
| | - A Heather Eliassen
- Harvard Medical School, Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard T.H. Chan School of Public Health, Department of Epidemiology, Boston, MA, USA
| | - Carolina Ellberg
- Lund University, Department of Cancer Epidemiology, Clinical Sciences, Lund, Sweden
| | - Christoph Engel
- University of Leipzig, Institute for Medical Informatics, Statistics and Epidemiology, Leipzig, Germany
- University of Leipzig, LIFE - Leipzig Research Centre for Civilization Diseases, Leipzig, Germany
| | - Mikael Eriksson
- Karolinska Institutet, Department of Medical Epidemiology and Biostatistics, Stockholm, Sweden
| | - D Gareth Evans
- University of Manchester, Manchester Academic Health Science Centre, Division of Evolution and Genomic Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester, UK
- St Marys Hospital, Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester Centre for Genomic Medicine, Manchester, UK
| | - Jonine Figueroa
- National Cancer Institute, Division of Cancer Epidemiology and Genetics, Bethesda, MD, USA
- The University of Edinburgh Medical School, Usher Institute of Population Health Sciences and Informatics, Edinburgh, UK
- Cancer Research UK Edinburgh Centre, Edinburgh, UK
| | - Dieter Flesch-Janys
- University Medical Centre Hamburg-Eppendorf, Institute for Medical Biometrics and Epidemiology, Hamburg, Germany
- University Medical Centre Hamburg-Eppendorf, Department of Cancer Epidemiology, Clinical Cancer Registry, Hamburg, Germany
| | - Henrik Flyger
- Copenhagen University Hospital, Department of Breast Surgery, Herlev and Gentofte Hospital, Herlev, Denmark
| | - Marike Gabrielson
- Karolinska Institutet, Department of Medical Epidemiology and Biostatistics, Stockholm, Sweden
| | - Manuela Gago-Dominguez
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Genomic Medicine Group, Galician Foundation of Genomic Medicine, SERGAS, Santiago de Compostela, Spain
- University of California San Diego, Moores Cancer Center, La Jolla, CA, USA
| | - Eva Galle
- VIB, VIB Center for Cancer Biology, Leuven, Belgium
- University of Leuven, Laboratory for Translational Genetics, Department of Human Genetics, Leuven, Belgium
| | - Susan M Gapstur
- American Cancer Society, Epidemiology Research Program, Atlanta, GA, USA
| | - Montserrat García-Closas
- National Cancer Institute, Division of Cancer Epidemiology and Genetics, Bethesda, MD, USA
- Institute of Cancer Research, Division of Genetics and Epidemiology, London, UK
| | - José A García-Sáenz
- Instituto de Investigación Sanitaria San Carlos (IdISSC), Centro Investigación Biomédica en Red de Cáncer (CIBERONC), Medical Oncology Department, Hospital Cl'nico San Carlos, Madrid, Spain
| | - Mia M Gaudet
- American Cancer Society, Epidemiology Research Program, Atlanta, GA, USA
| | - Angela George
- The Institute of Cancer Research, Division of Genetics and Epidemiology, London, UK
- The Royal Marsden NHS Foundation Trust, Cancer Genetics Unit, London, UK
| | | | - Graham G Giles
- Cancer Council Victoria, Cancer Epidemiology & Intelligence Division, Melbourne, VIC, Australia
- The University of Melbourne, Melbourne School of Population and Global Health, Centre for Epidemiology and Biostatistics, Melbourne, VIC, Australia
- Monash University, Department of Epidemiology and Preventive Medicine, Melbourne, VIC, Australia
| | - Gord Glendon
- Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, Fred A. Litwin Center for Cancer Genetics, Toronto, ON, Canada
| | - David E Goldgar
- Huntsman Cancer Institute, University of Utah School of Medicine, Department of Dermatology, Salt Lake City, UT, USA
| | - Anna González-Neira
- Spanish National Cancer Research Centre (CNIO), Human Cancer Genetics Programme, Madrid, Spain
| | - Grethe I Grenaker Alnæs
- Oslo University Hospital-Radiumhospitalet, Department of Cancer Genetics, Institute for Cancer Research, Oslo, Norway
| | - Mervi Grip
- University of Oulu, Department of Surgery, Oulu University Hospital, Oulu, Finland
| | - Pascal Guénel
- INSERM, University Paris-Sud, University Paris-Saclay, Cancer & Environment Group, Center for Research in Epidemiology and Population Health (CESP), Villejuif, France
| | - Lothar Haeberle
- Friedrich-Alexander University Erlangen-Nuremberg, Comprehensive Cancer Centre Erlangen-EMN, Department of Gynaecology and Obstetrics, University Hospital Erlangen, Erlangen, Germany
| | - Eric Hahnen
- University Hospital of Cologne, Centre for Hereditary Breast and Ovarian Cancer, Cologne, Germany
- University of Cologne, Centre for Molecular Medicine Cologne (CMMC), Cologne, Germany
| | - Christopher A Haiman
- University of Southern California, Department of Preventive Medicine, Keck School of Medicine, Los Angeles, CA, USA
| | - Niclas Håkansson
- Karolinska Institutet, Institute of Environmental Medicine, Stockholm, Sweden
| | - Per Hall
- Karolinska Institutet, Department of Medical Epidemiology and Biostatistics, Stockholm, Sweden
- South General Hospital, Department of Oncology, Stockholm, Sweden
| | - Ute Hamann
- German Cancer Research Centre (DKFZ), Molecular Genetics of Breast Cancer, Heidelberg, Germany
| | - Susan Hankinson
- University of Massachusetts, Amherst, Department of Biostatistics & Epidemiology, Amherst, MA, USA
| | - Elaine F Harkness
- University of Manchester, Manchester Academic Health Science Centre, Division of Informatics, Imaging and Data Sciences, Faculty of Biology, Medicine and Health, Manchester, UK
- Wythenshawe Hospital, Manchester University NHS Foundation Trust, Nightingale Breast Screening Centre, Manchester, UK
- Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, NIHR Manchester Biomedical Research Unit, Manchester, UK
| | - Patricia A Harrington
- University of Cambridge, Centre for Cancer Genetic Epidemiology, Department of Oncology, Cambridge, UK
| | - Steven N Hart
- Mayo Clinic, Department of Health Sciences Research, Rochester, MN, USA
| | - Jaana M Hartikainen
- University of Eastern Finland, Translational Cancer Research Area, Kuopio, Finland
- University of Eastern Finland, Institute of Clinical Medicine, Pathology and Forensic Medicine, Kuopio, Finland
- Kuopio University Hospital, Imaging Centre, Department of Clinical Pathology, Kuopio, Finland
| | - Alexander Hein
- University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Department of Gynecology and Obstetrics, Comprehensive Cancer Center ER-EMN, Erlangen, Germany
| | - Peter Hillemanns
- Hannover Medical School, Gynaecology Research Unit, Hannover, Germany
| | - Louise Hiller
- University of Warwick, Warwick Clinical Trials Unit, Coventry, UK
| | | | - Antoinette Hollestelle
- Erasmus MC Cancer Institute, Department of Medical Oncology, Family Cancer Clinic, Rotterdam, The Netherlands
| | - Maartje J Hooning
- Erasmus MC Cancer Institute, Department of Medical Oncology, Family Cancer Clinic, Rotterdam, The Netherlands
| | - Robert N Hoover
- National Cancer Institute, Division of Cancer Epidemiology and Genetics, Bethesda, MD, USA
| | - John L Hopper
- The University of Melbourne, Melbourne School of Population and Global Health, Centre for Epidemiology and Biostatistics, Melbourne, VIC, Australia
| | - Anthony Howell
- University of Manchester, Institute of Cancer studies, Manchester, UK
| | - Guanmengqian Huang
- German Cancer Research Centre (DKFZ), Molecular Genetics of Breast Cancer, Heidelberg, Germany
| | - Keith Humphreys
- Karolinska Institutet, Department of Medical Epidemiology and Biostatistics, Stockholm, Sweden
| | - David J Hunter
- Harvard T.H. Chan School of Public Health, Department of Epidemiology, Boston, MA, USA
- Harvard T.H. Chan School of Public Health, Program in Genetic Epidemiology and Statistical Genetics, Boston, MA, USA
- University of Oxford, Nuffield Department of Population Health, Oxford, UK
| | | | - Esther M John
- Cancer Prevention Institute of California, Department of Epidemiology, Fremont, CA, USA
- Stanford University School of Medicine, Department of Health Research and Policy - Epidemiology, Stanford, CA, USA
- Stanford University School of Medicine, Department of Biomedical Data Science, Stanford, CA, USA
| | - Michael E Jones
- Institute of Cancer Research, Division of Genetics and Epidemiology, London, UK
| | | | - Audrey Jung
- German Cancer Research Center (DKFZ), Division of Cancer Epidemiology, Heidelberg, Germany
| | - Rudolf Kaaks
- German Cancer Research Center (DKFZ), Division of Cancer Epidemiology, Heidelberg, Germany
| | - Maria Kabisch
- German Cancer Research Centre (DKFZ), Molecular Genetics of Breast Cancer, Heidelberg, Germany
| | - Katarzyna Kaczmarek
- Pomeranian Medical University, Department of Genetics and Pathology, Szczecin, Poland
| | - Michael J Kerin
- National University of Ireland, Surgery, School of Medicine, Galway, Ireland
| | - Sofia Khan
- University of Helsinki, Department of Obstetrics and Gynaecology, Helsinki University Hospital, Helsinki, Finland
| | - Elza Khusnutdinova
- Ufa Scientific Center of Russian Academy of Sciences, Institute of Biochemistry and Genetics, Ufa, Russia
- Bashkir State University, Department of Genetics and Fundamental Medicine, Ufa, Russia
| | - Johanna I Kiiski
- University of Helsinki, Department of Obstetrics and Gynaecology, Helsinki University Hospital, Helsinki, Finland
| | - Cari M Kitahara
- National Cancer Institute, Radiation Epidemiology Branch, Division of Cancer Epidemiology and Genetics, Bethesda, MD, USA
| | - Julia A Knight
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Prosserman Centre for Population Health Research, Toronto, ON, Canada
- University of Toronto, Division of Epidemiology, Dalla Lana School of Public Health, Toronto, ON, Canada
| | - Yon-Dschun Ko
- Johanniter Krankenhaus, Department of Internal Medicine, Evangelische Kliniken Bonn gGmbH, Bonn, Germany
| | - Linetta B Koppert
- Erasmus MC Cancer Institute, Department of Surgical Oncology, Family Cancer Clinic, Rotterdam, The Netherlands
| | - Veli-Matti Kosma
- University of Eastern Finland, Translational Cancer Research Area, Kuopio, Finland
- University of Eastern Finland, Institute of Clinical Medicine, Pathology and Forensic Medicine, Kuopio, Finland
- Kuopio University Hospital, Imaging Centre, Department of Clinical Pathology, Kuopio, Finland
| | - Peter Kraft
- Harvard T.H. Chan School of Public Health, Department of Epidemiology, Boston, MA, USA
- Harvard T.H. Chan School of Public Health, Program in Genetic Epidemiology and Statistical Genetics, Boston, MA, USA
| | - Vessela N Kristensen
- Oslo University Hospital-Radiumhospitalet, Department of Cancer Genetics, Institute for Cancer Research, Oslo, Norway
- University of Oslo, Institute of Clinical Medicine, Faculty of Medicine, Oslo, Norway
- Department of Research, Vestre Viken Hospital, Drammen, Norway; Section for Breast- and Endocrine Surgery, Department of Cancer, Division of Surgery, Cancer and Transplantation Medicine, Oslo University Hospital-Ullevål, Oslo, Norway
- Department of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
- Department of Pathology at Akershus University hospital, Lørenskog, Norway
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Department of Oncology, Division of Surgery and Cancer and Transplantation Medicine, Oslo University Hospital-Radiumhospitalet, Oslo, Norway
- National Advisory Unit on Late Effects after Cancer Treatment, Department of Oncology, Oslo University Hospital, Oslo, Norway
- Department of Oncology, Akershus University Hospital, Lørenskog, Norway
- Breast Cancer Research Consortium, Oslo University Hospital, Oslo, Norway
| | - Ute Krüger
- Lund University, Department of Cancer Epidemiology, Clinical Sciences, Lund, Sweden
| | - Tabea Kühl
- University Medical Center Hamburg-Eppendorf, Cancer Epidemiology Group, University Cancer Center Hamburg (UCCH), Hamburg, Germany
| | - Diether Lambrechts
- VIB, VIB Center for Cancer Biology, Leuven, Belgium
- University of Leuven, Laboratory for Translational Genetics, Department of Human Genetics, Leuven, Belgium
| | - Loic Le Marchand
- University of Hawaii Cancer Center, Epidemiology Program, Honolulu, HI, USA
| | - Eunjung Lee
- University of Southern California, Department of Preventive Medicine, Keck School of Medicine, Los Angeles, CA, USA
| | - Flavio Lejbkowicz
- Carmel Medical Center and Technion Faculty of Medicine, Clalit National Cancer Control Center, Haifa, Israel
| | - Lian Li
- Tianjin Medical University Cancer Institute and Hospital, Department of Epidemiology, Tianjin, China
| | - Annika Lindblom
- Karolinska Institutet, Department of Molecular Medicine and Surgery, Stockholm, Sweden
| | - Sara Lindström
- University of Washington School of Public Health, Department of Epidemiology, Seattle, WA, USA
- Fred Hutchinson Cancer Research Center, Public Health Sciences Division, Seattle, WA, USA
| | - Martha Linet
- National Cancer Institute, Radiation Epidemiology Branch, Division of Cancer Epidemiology and Genetics, Bethesda, MD, USA
| | - Jolanta Lissowska
- M. Sklodowska-Curie Cancer Centre, Oncology Institute, Department of Cancer Epidemiology and Prevention, Warsaw, Poland
| | - Wing-Yee Lo
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tübingen, Tübingen, Germany
| | | | - Jan Lubiński
- Pomeranian Medical University, Department of Genetics and Pathology, Szczecin, Poland
| | - Michael P Lux
- Friedrich-Alexander University Erlangen-Nuremberg, Comprehensive Cancer Centre Erlangen-EMN, Department of Gynaecology and Obstetrics, University Hospital Erlangen, Erlangen, Germany
| | - Robert J MacInnis
- Cancer Council Victoria, Cancer Epidemiology & Intelligence Division, Melbourne, VIC, Australia
- The University of Melbourne, Melbourne School of Population and Global Health, Centre for Epidemiology and Biostatistics, Melbourne, VIC, Australia
| | - Melanie Maierthaler
- German Cancer Research Center (DKFZ), Molecular Epidemiology Group, C080, Heidelberg, Germany
| | - Tom Maishman
- University of Southampton, Southampton Clinical Trials Unit, Faculty of Medicine, Southampton, UK
- University of Southampton, Cancer Sciences Academic Unit, Faculty of Medicine, Southampton, UK
| | - Enes Makalic
- The University of Melbourne, Melbourne School of Population and Global Health, Centre for Epidemiology and Biostatistics, Melbourne, VIC, Australia
| | - Arto Mannermaa
- University of Eastern Finland, Translational Cancer Research Area, Kuopio, Finland
- University of Eastern Finland, Institute of Clinical Medicine, Pathology and Forensic Medicine, Kuopio, Finland
- Kuopio University Hospital, Imaging Centre, Department of Clinical Pathology, Kuopio, Finland
| | - Mehdi Manoochehri
- German Cancer Research Centre (DKFZ), Molecular Genetics of Breast Cancer, Heidelberg, Germany
| | - Siranoush Manoukian
- Fondazione IRCCS (Istituto Di Ricovero e Cura a Carattere Scientifico) Istituto Nazionale dei Tumori (INT), Unit of Medical Genetics, Department of Medical Oncology and Haematology, Milan, Italy
| | - Sara Margolin
- Karolinska Institutet, Department of Clinical Science and Education, Sšdersjukhuset, Stockholm, Sweden
| | - Maria Elena Martinez
- University of California San Diego, Moores Cancer Center, La Jolla, CA, USA
- University of California San Diego, Department of Family Medicine and Public Health, La Jolla, CA, USA
| | - Dimitrios Mavroudis
- University Hospital of Heraklion, Department of Medical Oncology, Heraklion, Greece
| | - Catriona McLean
- The Alfred Hospital, Anatomical Pathology, Melbourne, VIC, Australia
| | - Alfons Meindl
- Ludwig Maximilian University of Munich, Department of Gynaecology and Obstetrics, Munich, Germany
| | - Pooja Middha
- German Cancer Research Center (DKFZ), Division of Cancer Epidemiology, Heidelberg, Germany
- University of Heidelberg, Faculty of Medicine, Heidelberg, Germany
| | - Nicola Miller
- National University of Ireland, Surgery, School of Medicine, Galway, Ireland
| | - Roger L Milne
- Cancer Council Victoria, Cancer Epidemiology & Intelligence Division, Melbourne, VIC, Australia
- The University of Melbourne, Melbourne School of Population and Global Health, Centre for Epidemiology and Biostatistics, Melbourne, VIC, Australia
| | - Fernando Moreno
- Instituto de Investigación Sanitaria San Carlos (IdISSC), Centro Investigación Biomédica en Red de Cáncer (CIBERONC), Medical Oncology Department, Hospital Cl'nico San Carlos, Madrid, Spain
| | - Anna Marie Mulligan
- University of Toronto, Department of Laboratory Medicine and Pathobiology, Toronto, ON, Canada
- University Health Network, Laboratory Medicine Program, Toronto, ON, Canada
| | - Claire Mulot
- INSERM UMR-S1147, Université Paris Sorbonne Cité, Paris, France
| | - Rami Nassir
- University of California Davis, Department of Biochemistry and Molecular Medicine, Davis, CA, USA
| | - Susan L Neuhausen
- Beckman Research Institute of City of Hope, Department of Population Sciences, Duarte, CA, USA
| | - William T Newman
- University of Manchester, Manchester Academic Health Science Centre, Division of Evolution and Genomic Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester, UK
- St Marys Hospital, Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester Centre for Genomic Medicine, Manchester, UK
| | - Sune F Nielsen
- Copenhagen University Hospital, Copenhagen General Population Study, Herlevand Gentofte Hospital, Herlev, Denmark
- Copenhagen University Hospital, Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Herlev, Denmark
| | - Børge G Nordestgaard
- Copenhagen University Hospital, Copenhagen General Population Study, Herlevand Gentofte Hospital, Herlev, Denmark
- Copenhagen University Hospital, Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Herlev, Denmark
- University of Copenhagen, Faculty of Health and Medical Sciences, Copenhagen, Denmark
| | - Aaron Norman
- Mayo Clinic, Department of Health Sciences Research, Rochester, MN, USA
| | - Håkan Olsson
- Lund University, Department of Cancer Epidemiology, Clinical Sciences, Lund, Sweden
| | - Nick Orr
- Queen's University Belfast, Centre for Cancer Research and Cell Biology, Belfast, Ireland, UK
| | - V Shane Pankratz
- University of New Mexico, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | | | - Jose I A Perez
- Hospital Monte Naranco, Servicio de Cirug'a General y Especialidades, Oviedo, Spain
| | - Clara Pérez-Barrios
- Hospital Universitario Puerta de Hierro, Medical Oncology Department, Madrid, Spain
| | - Paolo Peterlongo
- The FIRC (Italian Foundation for Cancer Research) Institute of Molecular Oncology, IFOM, Milan, Italy
| | - Christos Petridis
- King's College London, Research Oncology, Guy's Hospital, London, UK
| | - Mila Pinchev
- Carmel Medical Center and Technion Faculty of Medicine, Clalit National Cancer Control Center, Haifa, Israel
| | - Karoliona Prajzendanc
- Pomeranian Medical University, Department of Genetics and Pathology, Szczecin, Poland
| | - Ross Prentice
- Fred Hutchinson Cancer Research Center, Cancer Prevention Program, Seattle, WA, USA
| | - Nadege Presneau
- University of Westminster, Department of Biomedical Sciences, Faculty of Science and Technology, London, UK
| | - Darya Prokofieva
- Bashkir State University, Department of Genetics and Fundamental Medicine, Ufa, Russia
| | - Katri Pylkäs
- University of Oulu, Laboratory of Cancer Genetics and Tumour Biology, Cancer and Translational Medicine Research Unit, Biocentre Oulu, Oulu, Finland
- Northern Finland Laboratory Centre Oulu, Laboratory of Cancer Genetics and Tumour Biology, Oulu, Finland
| | - Brigitte Rack
- Ludwig Maximilian University of Munich, Department of Gynaecology and Obstetrics, Munich, Germany
| | - Paolo Radice
- Fondazione IRCCS (Istituto Di Ricovero e Cura a Carattere Scientifico) Istituto Nazionale dei Tumori (INT), Unit of Molecular Bases of Genetic Risk and Genetic Testing, Department of Research, Milan, Italy
| | | | - Gadi Rennert
- Carmel Medical Center and Technion Faculty of Medicine, Clalit National Cancer Control Center, Haifa, Israel
| | - Hedy S Rennert
- Carmel Medical Center and Technion Faculty of Medicine, Clalit National Cancer Control Center, Haifa, Israel
| | - Valerie Rhenius
- University of Cambridge, Centre for Cancer Genetic Epidemiology, Department of Oncology, Cambridge, UK
| | - Atocha Romero
- Hospital Universitario Puerta de Hierro, Medical Oncology Department, Madrid, Spain
| | | | | | - Elinor J Sawyer
- King's College London, Research Oncology, Guy's Hospital, London, UK
| | - Daniel F Schmidt
- The University of Melbourne, Melbourne School of Population and Global Health, Centre for Epidemiology and Biostatistics, Melbourne, VIC, Australia
| | - Rita K Schmutzler
- University Hospital of Cologne, Centre for Hereditary Breast and Ovarian Cancer, Cologne, Germany
- University of Cologne, Centre for Molecular Medicine Cologne (CMMC), Cologne, Germany
| | - Andreas Schneeweiss
- University of Heidelberg, Department of Obstetrics and Gynecology, Heidelberg, Germany
- University of Heidelberg, National Centre for Tumour Diseases, Heidelberg, Germany
| | - Minouk J Schoemaker
- The Institute of Cancer Research, Division of Genetics and Epidemiology, London, UK
| | - Fredrick Schumacher
- Case Western Reserve University, Department of Population and Quantitative Health Sciences, Cleveland, OH, USA
| | | | - Rodney J Scott
- John Hunter Hospital, Division of Molecular Medicine, Pathology North, Newcastle, NSW, Australia
- University of Newcastle, Discipline of Medical Genetics, School of Biomedical Sciences and Pharmacy, Faculty of Health, Callaghan, NSW, Australia
- John Hunter Hospital, Hunter Medical Research Institute, Newcastle, NSW, Australia
- University of Newcastle, Centre for Information Based Medicine, Callaghan, Newcastle, NSW, Australia
| | - Christopher Scott
- Mayo Clinic, Department of Health Sciences Research, Rochester, MN, USA
| | - Caroline Seynaeve
- Erasmus MC Cancer Institute, Department of Medical Oncology, Family Cancer Clinic, Rotterdam, The Netherlands
| | - Mitul Shah
- University of Cambridge, Centre for Cancer Genetic Epidemiology, Department of Oncology, Cambridge, UK
| | - Jacques Simard
- Centre Hospitalier Universitaire de Québec - Université Laval Research Centre, Genomics Centre, Québec City, QC, Canada
| | - Ann Smeets
- University Hospitals Leuven, Department of Surgical Oncology, Leuven, Belgium
| | - Christof Sohn
- University of Heidelberg, National Centre for Tumour Diseases, Heidelberg, Germany
| | - Melissa C Southey
- Monash University, Precision Medicine, School of Clinical Sciences at Monash Health, Clayton, Victoria, Australia
- The University of Melbourne, Department of Clinical Pathology, Melbourne, VIC, Australia
| | - Anthony J Swerdlow
- The Institute of Cancer Research, Division of Genetics and Epidemiology, London, UK
- The Institute of Cancer Research, Division of Breast Cancer Research, London, UK
| | - Aline Talhouk
- BC Cancer Agency and University of British Columbia, British Columbia's Ovarian Cancer Research (OVCARE) Program, Vancouver General Hospital, Vancouver, BC, Canada
- University of British Columbia, Department of Pathology and Laboratory Medicine, Vancouver, BC, Canada
- University of British Columbia, Department of Obstetrics and Gynaecology, Vancouver, BC, Canada
| | - Rulla M Tamimi
- Harvard Medical School, Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard T.H. Chan School of Public Health, Department of Epidemiology, Boston, MA, USA
- Harvard T.H. Chan School of Public Health, Program in Genetic Epidemiology and Statistical Genetics, Boston, MA, USA
| | | | - Manuel R Teixeira
- Portuguese Oncology Institute, Department of Genetics, Porto, Portugal
- University of Porto, Biomedical Sciences Institute (ICBAS), Porto, Portugal
| | - Maria Tengström
- University of Eastern Finland, Translational Cancer Research Area, Kuopio, Finland
- Kuopio University Hospital, Cancer Centre, Kuopio, Finland
- University of Eastern Finland, Institute of Clinical Medicine, Oncology, Kuopio, Finland
| | - Mary Beth Terry
- Columbia University, Department of Epidemiology, Mailman School of Public Health, New York, NY, USA
| | - Kathrin Thöne
- University Medical Center Hamburg-Eppendorf, Cancer Epidemiology Group, University Cancer Center Hamburg (UCCH), Hamburg, Germany
| | - Rob A E M Tollenaar
- Leiden University Medical Centre, Department of Surgery, Leiden, The Netherlands
| | - Ian Tomlinson
- University of Birmingham, Institute of Cancer and Genomic Sciences, Birmingham, UK
- University of Oxford, Wellcome Trust Centre for Human Genetics and Oxford NIHR Biomedical Research Centre, Oxford, UK
| | - Diana Torres
- German Cancer Research Centre (DKFZ), Molecular Genetics of Breast Cancer, Heidelberg, Germany
- Pontificia Universidad Javeriana, Institute of Human Genetics, Bogota, Colombia
| | - Thérèse Truong
- INSERM, University Paris-Sud, University Paris-Saclay, Cancer & Environment Group, Center for Research in Epidemiology and Population Health (CESP), Villejuif, France
| | - Constance Turman
- Harvard T.H. Chan School of Public Health, Department of Epidemiology, Boston, MA, USA
| | - Clare Turnbull
- The Institute of Cancer Research, Division of Genetics and Epidemiology, London, UK
| | | | - Michael Untch
- Helios Clinics Berlin-Buch, Department of Gynaecology and Obstetrics, Berlin, Germany
| | - Celine Vachon
- Mayo Clinic, Department of Health Sciences Research, Rochester, MN, USA
| | - Christi J van Asperen
- Leiden University Medical Centre, Department of Clinical Genetics, Leiden, The Netherlands
| | | | - Elke M van Veen
- University of Manchester, Manchester Academic Health Science Centre, Division of Evolution and Genomic Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester, UK
- St Marys Hospital, Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester Centre for Genomic Medicine, Manchester, UK
| | - Camilla Wendt
- Karolinska Institutet, Department of Clinical Science and Education, Södersjukhuset, Stockholm, Sweden
| | - Alice S Whittemore
- Stanford University School of Medicine, Department of Health Research and Policy - Epidemiology, Stanford, CA, USA
- Stanford University School of Medicine, Department of Biomedical Data Science, Stanford, CA, USA
| | - Walter Willett
- Harvard T.H. Chan School of Public Health, Department of Epidemiology, Boston, MA, USA
- Harvard T.H. Chan School of Public Health, Department of Nutrition, Boston, MA, USA
- Brigham and Women's Hospital and Harvard Medical School, Channing Division of Network Medicine, Boston, MA, USA
| | - Robert Winqvist
- University of Oulu, Laboratory of Cancer Genetics and Tumour Biology, Cancer and Translational Medicine Research Unit, Biocentre Oulu, Oulu, Finland
- Northern Finland Laboratory Centre Oulu, Laboratory of Cancer Genetics and Tumour Biology, Oulu, Finland
| | - Alicja Wolk
- Karolinska Institutet, Department of Environmental Medicine, Division of Nutritional Epidemiology, Stockholm, Sweden
| | - Xiaohong R Yang
- National Cancer Institute, Division of Cancer Epidemiology and Genetics, Bethesda, MD, USA
| | - Yan Zhang
- German Cancer Research Center (DKFZ), Division of Clinical Epidemiology and Aging Research, Heidelberg, Germany
- German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Douglas F Easton
- University of Cambridge, Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, Cambridge, UK
- University of Cambridge, Centre for Cancer Genetic Epidemiology, Department of Oncology, Cambridge, UK
| | - Peter A Fasching
- University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Department of Gynecology and Obstetrics, Comprehensive Cancer Center ER-EMN, Erlangen, Germany
- University of California at Los Angeles, David Geffen School of Medicine, Department of Medicine Division of Hematology and Oncology, Los Angeles, CA, USA
| | - Heli Nevanlinna
- University of Helsinki, Department of Obstetrics and Gynaecology, Helsinki University Hospital, Helsinki, Finland
| | - Diana M Eccles
- University of Southampton, Cancer Sciences Academic Unit, Faculty of Medicine, Southampton, UK
| | - Paul D P Pharoah
- University of Cambridge, Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, Cambridge, UK
- University of Cambridge, Centre for Cancer Genetic Epidemiology, Department of Oncology, Cambridge, UK
| | - Marjanka K Schmidt
- The Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Division of Molecular Pathology, Amsterdam, The Netherlands
- The Netherlands Cancer Institute - Antoni van Leeuwenhoek hospital, Division of Psychosocial Research and Epidemiology, Amsterdam, The Netherlands
| |
Collapse
|
34
|
Louault K, Bonneaud TL, Séveno C, Gomez-Bougie P, Nguyen F, Gautier F, Bourgeois N, Loussouarn D, Kerdraon O, Barillé-Nion S, Jézéquel P, Campone M, Amiot M, Juin PP, Souazé F. Interactions between cancer-associated fibroblasts and tumor cells promote MCL-1 dependency in estrogen receptor-positive breast cancers. Oncogene 2019; 38:3261-3273. [PMID: 30631150 PMCID: PMC6756023 DOI: 10.1038/s41388-018-0635-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 11/22/2018] [Accepted: 11/23/2018] [Indexed: 12/24/2022]
Abstract
Selective inhibition of BCL-2 is expected to enhance therapeutic vulnerability in luminal estrogen receptor-positive breast cancers. We show here that the BCL-2 dependency of luminal tumor cells is nevertheless mitigated by breast cancer-associated fibroblasts (bCAFs) in a manner that defines MCL-1 as another critical therapeutic target. bCAFs favor MCL-1 expression and apoptotic resistance in luminal cancer cells in a IL-6 dependent manner while their own, robust, survival also relies on MCL-1. Studies based on ex vivo cultures of human luminal breast cancer tissues further argue that the contribution of stroma-derived signals to MCL-1 expression shapes BCL-2 dependency. Thus, MCL-1 inhibitors are beneficial for targeted apoptosis of breast tumor ecosystems, even in a subtype where MCL-1 dependency is not intrinsically driven by oncogenic pathways.
Collapse
Affiliation(s)
- K Louault
- CRCINA, Team 8, INSERM, Université d'Angers, Université de Nantes, Nantes, France.,SIRIC ILIAD, Angers, Nantes, France
| | - T L Bonneaud
- CRCINA, Team 8, INSERM, Université d'Angers, Université de Nantes, Nantes, France.,SIRIC ILIAD, Angers, Nantes, France
| | - C Séveno
- CRCINA, Team 8, INSERM, Université d'Angers, Université de Nantes, Nantes, France.,SIRIC ILIAD, Angers, Nantes, France
| | - P Gomez-Bougie
- SIRIC ILIAD, Angers, Nantes, France.,CRCINA, Team 10, INSERM, Université d'Angers, Université de Nantes, Nantes, France
| | - F Nguyen
- CRCINA, Team 8, INSERM, Université d'Angers, Université de Nantes, Nantes, France.,ONIRIS, Nantes Atlantic College of Veterinary Medicine Food Science and Engineering, Animal Cancers, Nantes, France
| | - F Gautier
- CRCINA, Team 8, INSERM, Université d'Angers, Université de Nantes, Nantes, France.,SIRIC ILIAD, Angers, Nantes, France.,ICO René Gauducheau, Saint Herblain, France
| | - N Bourgeois
- CRCINA, Team 8, INSERM, Université d'Angers, Université de Nantes, Nantes, France.,SIRIC ILIAD, Angers, Nantes, France
| | - D Loussouarn
- Service d'Anatomie Pathologique, CHU Nantes, Nantes, France
| | - O Kerdraon
- SIRIC ILIAD, Angers, Nantes, France.,ICO René Gauducheau, Saint Herblain, France
| | - S Barillé-Nion
- CRCINA, Team 8, INSERM, Université d'Angers, Université de Nantes, Nantes, France.,SIRIC ILIAD, Angers, Nantes, France
| | - P Jézéquel
- CRCINA, Team 8, INSERM, Université d'Angers, Université de Nantes, Nantes, France.,SIRIC ILIAD, Angers, Nantes, France.,ICO René Gauducheau, Saint Herblain, France
| | - M Campone
- CRCINA, Team 8, INSERM, Université d'Angers, Université de Nantes, Nantes, France.,SIRIC ILIAD, Angers, Nantes, France.,ICO René Gauducheau, Saint Herblain, France
| | - M Amiot
- SIRIC ILIAD, Angers, Nantes, France.,CRCINA, Team 10, INSERM, Université d'Angers, Université de Nantes, Nantes, France
| | - P P Juin
- CRCINA, Team 8, INSERM, Université d'Angers, Université de Nantes, Nantes, France. .,SIRIC ILIAD, Angers, Nantes, France. .,ICO René Gauducheau, Saint Herblain, France. .,CNRS GDR3697 Micronit, Tours, France.
| | - F Souazé
- CRCINA, Team 8, INSERM, Université d'Angers, Université de Nantes, Nantes, France. .,SIRIC ILIAD, Angers, Nantes, France. .,CNRS GDR3697 Micronit, Tours, France.
| |
Collapse
|
35
|
Winslow S, Scholz A, Rappl P, Brauß TF, Mertens C, Jung M, Weigert A, Brüne B, Schmid T. Macrophages attenuate the transcription of CYP1A1 in breast tumor cells and enhance their proliferation. PLoS One 2019; 14:e0209694. [PMID: 30615637 PMCID: PMC6322746 DOI: 10.1371/journal.pone.0209694] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 12/10/2018] [Indexed: 12/31/2022] Open
Abstract
While aberrant cells are routinely recognized and removed by immune cells, tumors eventually escape innate immune responses. Infiltrating immune cells are even corrupted by the tumor to acquire a tumor-supporting phenotype. In line, tumor-associated macrophages are well-characterized to promote tumor progression and high levels of tumor-infiltrating macrophages are a poor prognostic marker in breast cancer. Here, we aimed to further decipher the influence of macrophages on breast tumor cells and determined global gene expression changes in three-dimensional tumor spheroids upon infiltration of macrophages. While various tumor-associated mRNAs were upregulated, expression of the cytochrome P450 family member CYP1A1 was markedly attenuated. Repression of CYP1A1 in tumor cells was elicited by a macrophage-shaped tumor microenvironment rather than by direct tumor cell-macrophage contacts. In line with changes in RNA expression profiles, macrophages enhanced proliferation of the tumor cells. Enhanced proliferation and macrophage presence further correlated with reduced CYP1A1 expression in patient tumors when compared with normal tissue. These findings are of interest in the context of combinatory therapeutic approaches involving cytotoxic and immune-modulatory compounds.
Collapse
Affiliation(s)
- Sofia Winslow
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany
| | - Anica Scholz
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany
| | - Peter Rappl
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany
| | - Thilo F. Brauß
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany
| | - Christina Mertens
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany
| | - Michaela Jung
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany
| | - Andreas Weigert
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany
| | - Bernhard Brüne
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany
| | - Tobias Schmid
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany
- * E-mail:
| |
Collapse
|
36
|
Webber JT, Kaushik S, Bandyopadhyay S. Integration of Tumor Genomic Data with Cell Lines Using Multi-dimensional Network Modules Improves Cancer Pharmacogenomics. Cell Syst 2018; 7:526-536.e6. [PMID: 30414925 DOI: 10.1016/j.cels.2018.10.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 08/01/2018] [Accepted: 10/04/2018] [Indexed: 02/08/2023]
Abstract
Leveraging insights from genomic studies of patient tumors is limited by the discordance between these tumors and the cell line models used for functional studies. We integrate omics datasets using functional networks to identify gene modules reflecting variation between tumors and show that the structure of these modules can be evaluated in cell lines to discover clinically relevant biomarkers of therapeutic responses. Applied to breast cancer, we identify 219 gene modules that capture recurrent alterations and subtype patients and quantitate various cell types within the tumor microenvironment. Comparison of modules between tumors and cell lines reveals that many modules composed primarily of gene expression and methylation are poorly preserved. In contrast, preserved modules are highly predictive of drug responses in a manner that is robust and clinically relevant. This work addresses a fundamental challenge in pharmacogenomics that can only be overcome by the joint analysis of patient and cell line data.
Collapse
Affiliation(s)
- James T Webber
- Department of Bioengineering and Therapeutic Sciences, Institute for Computational Health Sciences, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Swati Kaushik
- Department of Bioengineering and Therapeutic Sciences, Institute for Computational Health Sciences, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Sourav Bandyopadhyay
- Department of Bioengineering and Therapeutic Sciences, Institute for Computational Health Sciences, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
37
|
Wang T, Lu R, Kapur P, Jaiswal BS, Hannan R, Zhang Z, Pedrosa I, Luke JJ, Zhang H, Goldstein LD, Yousuf Q, Gu YF, McKenzie T, Joyce A, Kim MS, Wang X, Luo D, Onabolu O, Stevens C, Xie Z, Chen M, Filatenkov A, Torrealba J, Luo X, Guo W, He J, Stawiski E, Modrusan Z, Durinck S, Seshagiri S, Brugarolas J. An Empirical Approach Leveraging Tumorgrafts to Dissect the Tumor Microenvironment in Renal Cell Carcinoma Identifies Missing Link to Prognostic Inflammatory Factors. Cancer Discov 2018; 8:1142-1155. [PMID: 29884728 PMCID: PMC6125163 DOI: 10.1158/2159-8290.cd-17-1246] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 03/01/2018] [Accepted: 06/07/2018] [Indexed: 01/05/2023]
Abstract
By leveraging tumorgraft (patient-derived xenograft) RNA-sequencing data, we developed an empirical approach, DisHet, to dissect the tumor microenvironment (eTME). We found that 65% of previously defined immune signature genes are not abundantly expressed in renal cell carcinoma (RCC) and identified 610 novel immune/stromal transcripts. Using eTME, genomics, pathology, and medical record data involving >1,000 patients, we established an inflamed pan-RCC subtype (IS) enriched for regulatory T cells, natural killer cells, TH1 cells, neutrophils, macrophages, B cells, and CD8+ T cells. IS is enriched for aggressive RCCs, including BAP1-deficient clear-cell and type 2 papillary tumors. The IS subtype correlated with systemic manifestations of inflammation such as thrombocytosis and anemia, which are enigmatic predictors of poor prognosis. Furthermore, IS was a strong predictor of poor survival. Our analyses suggest that tumor cells drive the stromal immune response. These data provide a missing link between tumor cells, the TME, and systemic factors.Significance: We undertook a novel empirical approach to dissect the renal cell carcinoma TME by leveraging tumorgrafts. The dissection and downstream analyses uncovered missing links between tumor cells, the TME, systemic manifestations of inflammation, and poor prognosis. Cancer Discov; 8(9); 1142-55. ©2018 AACR.This article is highlighted in the In This Issue feature, p. 1047.
Collapse
Affiliation(s)
- Tao Wang
- Quantitative Biomedical Research Center, Department of Clinical Sciences, The University of Texas Southwestern Medical Center, Dallas, Texas.
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas
- Center for the Genetics of Host Defense, The University of Texas Southwestern Medical Center, Texas
| | - Rong Lu
- Quantitative Biomedical Research Center, Department of Clinical Sciences, The University of Texas Southwestern Medical Center, Dallas, Texas
- Bioinformatics Core Facility, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Payal Kapur
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Bijay S Jaiswal
- Molecular Biology Department, Genentech, Inc., South San Francisco, California
| | - Raquibul Hannan
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Radiation Oncology, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Ze Zhang
- Quantitative Biomedical Research Center, Department of Clinical Sciences, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Ivan Pedrosa
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Radiology, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jason J Luke
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - He Zhang
- Bioinformatics Core Facility, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Leonard D Goldstein
- Molecular Biology Department, Genentech, Inc., South San Francisco, California
| | - Qurratulain Yousuf
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Yi-Feng Gu
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Tiffani McKenzie
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Allison Joyce
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Min S Kim
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas
- Bioinformatics Core Facility, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Xinlei Wang
- Department of Statistical Science, Southern Methodist University, Dallas, Texas
| | - Danni Luo
- Bioinformatics Core Facility, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Oreoluwa Onabolu
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Christina Stevens
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Zhiqun Xie
- Quantitative Biomedical Research Center, Department of Clinical Sciences, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Mingyi Chen
- Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Alexander Filatenkov
- Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jose Torrealba
- Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Xin Luo
- Bioinformatics Core Facility, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Wenbin Guo
- Quantitative Biomedical Research Center, Department of Clinical Sciences, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jingxuan He
- Quantitative Biomedical Research Center, Department of Clinical Sciences, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Eric Stawiski
- Molecular Biology Department, Genentech, Inc., South San Francisco, California
- Bioinformatics and Computational Biology Department, Genentech, Inc., South San Francisco, California
| | - Zora Modrusan
- Molecular Biology Department, Genentech, Inc., South San Francisco, California
| | - Steffen Durinck
- Molecular Biology Department, Genentech, Inc., South San Francisco, California
- Bioinformatics and Computational Biology Department, Genentech, Inc., South San Francisco, California
| | - Somasekar Seshagiri
- Molecular Biology Department, Genentech, Inc., South San Francisco, California.
| | - James Brugarolas
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas.
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
38
|
McCarthy JB, El-Ashry D, Turley EA. Hyaluronan, Cancer-Associated Fibroblasts and the Tumor Microenvironment in Malignant Progression. Front Cell Dev Biol 2018; 6:48. [PMID: 29868579 PMCID: PMC5951929 DOI: 10.3389/fcell.2018.00048] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 04/13/2018] [Indexed: 12/16/2022] Open
Abstract
This review summarizes the roles of CAFs in forming a “cancerized” fibrotic stroma favorable to tumor initiation and dissemination, in particular highlighting the functions of the extracellular matrix component hyaluronan (HA) in these processes. The structural complexity of the tumor and its host microenvironment is now well appreciated to be an important contributing factor to malignant progression and resistance-to-therapy. There are multiple components of this complexity, which include an extensive remodeling of the extracellular matrix (ECM) and associated biomechanical changes in tumor stroma. Tumor stroma is often fibrotic and rich in fibrillar type I collagen and hyaluronan (HA). Cancer-associated fibroblasts (CAFs) are a major source of this fibrotic ECM. CAFs organize collagen fibrils and these biomechanical alterations provide highways for invading carcinoma cells either under the guidance of CAFs or following their epithelial to mesenchymal transition (EMT). The increased HA metabolism of a tumor microenvironment instructs carcinoma initiation and dissemination by performing multiple functions. The key effects of HA reviewed here are its role in activating CAFs in pre-malignant and malignant stroma, and facilitating invasion by promoting motility of both CAFs and tumor cells, thus facilitating their invasion. Circulating CAFs (cCAFs) also form heterotypic clusters with circulating tumor cells (CTC), which are considered to be pre-cursors of metastatic colonies. cCAFs are likely required for extravasation of tumors cells and to form a metastatic niche suitable for new tumor colony growth. Therapeutic interventions designed to target both HA and CAFs in order to limit tumor spread and increase response to current therapies are discussed.
Collapse
Affiliation(s)
- James B McCarthy
- Department of Laboratory Medicine and Pathology, Masonic Comprehensive Cancer Center, Minneapolis, MN, United States
| | - Dorraya El-Ashry
- Department of Laboratory Medicine and Pathology, Masonic Comprehensive Cancer Center, Minneapolis, MN, United States
| | - Eva A Turley
- London Regional Cancer Program, Department of Oncology, Biochemistry and Surgery, Schulich School of Medicine and Dentistry, Lawson Health Research Institute, Western University, London, ON, Canada
| |
Collapse
|
39
|
Reid KBM. Complement Component C1q: Historical Perspective of a Functionally Versatile, and Structurally Unusual, Serum Protein. Front Immunol 2018; 9:764. [PMID: 29692784 PMCID: PMC5902488 DOI: 10.3389/fimmu.2018.00764] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 03/27/2018] [Indexed: 12/28/2022] Open
Abstract
Complement component C1q plays an important recognition role in adaptive, and innate, immunity through its ability to interact, via its six globular head regions, with both immunoglobulin and non-immunoglobulin activators of the complement system, and also in the clearance of cell debris, and by playing a role in regulation of cellular events by interacting with a wide range of cell surface molecules. The presence of collagen-like triple-helical structures within C1q appears crucial to the presentation, and multivalent binding, of the globular heads of C1q to targets, and also to its association with the proenzyme complex of C1r2–C1s2, to yield the C1 complex. The possible role that movement of these collagen-like structures may play in the activation of the C1 complex is a controversial area, with there still being no definitive answer as to how the first C1r proenzyme molecule becomes activated within the C1 complex, thus allowing it to activate proenzyme C1s, and initiate and the consequent cascade of events in the activation of the classical pathway of complement. The globular heads of C1q are similar to domains found within the tumor necrosis factor (TNF) superfamily of proteins, and have been shown to bind to a very wide range of ligands. In addition to its well-defined roles in infection and immunity, a variety of other functions associated with C1q include possible roles, in the development of problems in the central nervous system, which occur with aging, and perhaps in the regulation of tumor growth.
Collapse
Affiliation(s)
- Kenneth B M Reid
- Green Templeton College, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
40
|
Zhao J, Cheng F, Jia P, Cox N, Denny JC, Zhao Z. An integrative functional genomics framework for effective identification of novel regulatory variants in genome-phenome studies. Genome Med 2018; 10:7. [PMID: 29378629 PMCID: PMC5789733 DOI: 10.1186/s13073-018-0513-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 01/04/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Genome-phenome studies have identified thousands of variants that are statistically associated with disease or traits; however, their functional roles are largely unclear. A comprehensive investigation of regulatory mechanisms and the gene regulatory networks between phenome-wide association study (PheWAS) and genome-wide association study (GWAS) is needed to identify novel regulatory variants contributing to risk for human diseases. METHODS In this study, we developed an integrative functional genomics framework that maps 215,107 significant single nucleotide polymorphism (SNP) traits generated from the PheWAS Catalog and 28,870 genome-wide significant SNP traits collected from the GWAS Catalog into a global human genome regulatory map via incorporating various functional annotation data, including transcription factor (TF)-based motifs, promoters, enhancers, and expression quantitative trait loci (eQTLs) generated from four major functional genomics databases: FANTOM5, ENCODE, NIH Roadmap, and Genotype-Tissue Expression (GTEx). In addition, we performed a tissue-specific regulatory circuit analysis through the integration of the identified regulatory variants and tissue-specific gene expression profiles in 7051 samples across 32 tissues from GTEx. RESULTS We found that the disease-associated loci in both the PheWAS and GWAS Catalogs were significantly enriched with functional SNPs. The integration of functional annotations significantly improved the power of detecting novel associations in PheWAS, through which we found a number of functional associations with strong regulatory evidence in the PheWAS Catalog. Finally, we constructed tissue-specific regulatory circuits for several complex traits: mental diseases, autoimmune diseases, and cancer, via exploring tissue-specific TF-promoter/enhancer-target gene interaction networks. We uncovered several promising tissue-specific regulatory TFs or genes for Alzheimer's disease (e.g. ZIC1 and STX1B) and asthma (e.g. CSF3 and IL1RL1). CONCLUSIONS This study offers powerful tools for exploring the functional consequences of variants generated from genome-phenome association studies in terms of their mechanisms on affecting multiple complex diseases and traits.
Collapse
Affiliation(s)
- Junfei Zhao
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, 7000 Fannin St. Suite 820, Houston, TX, 77030, USA
| | - Feixiong Cheng
- Center for Cancer Systems Biology and Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02215, USA
- Center for Complex Networks Research, Northeastern University, Boston, MA, 02215, USA
| | - Peilin Jia
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, 7000 Fannin St. Suite 820, Houston, TX, 77030, USA
| | - Nancy Cox
- Vanderbilt Genetics Institute, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Joshua C Denny
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Zhongming Zhao
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, 7000 Fannin St. Suite 820, Houston, TX, 77030, USA.
- Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
| |
Collapse
|
41
|
Agostinis C, Vidergar R, Belmonte B, Mangogna A, Amadio L, Geri P, Borelli V, Zanconati F, Tedesco F, Confalonieri M, Tripodo C, Kishore U, Bulla R. Complement Protein C1q Binds to Hyaluronic Acid in the Malignant Pleural Mesothelioma Microenvironment and Promotes Tumor Growth. Front Immunol 2017; 8:1559. [PMID: 29209316 PMCID: PMC5701913 DOI: 10.3389/fimmu.2017.01559] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 10/31/2017] [Indexed: 01/08/2023] Open
Abstract
C1q is the first recognition subcomponent of the complement classical pathway, which acts toward the clearance of pathogens and apoptotic cells. C1q is also known to modulate a range of functions of immune and non-immune cells, and has been shown to be involved in placental development and sensorial synaptic pruning. We have recently shown that C1q can promote tumor by encouraging their adhesion, migration, and proliferation in addition to angiogenesis and metastasis. In this study, we have examined the role of human C1q in the microenvironment of malignant pleural mesothelioma (MPM), a rare form of cancer commonly associated with exposure to asbestos. We found that C1q was highly expressed in all MPM histotypes, particularly in epithelioid rather than in sarcomatoid histotype. C1q avidly bound high and low molecular weight hyaluronic acid (HA) via its globular domain. C1q bound to HA was able to induce adhesion and proliferation of mesothelioma cells (MES) via enhancement of ERK1/2, SAPK/JNK, and p38 phosphorylation; however, it did not activate the complement cascade. Consistent with the modular organization of the globular domain, we demonstrated that C1q may bind to HA through ghA module, whereas it may interact with human MES through the ghC. In conclusion, C1q highly expressed in MPM binds to HA and enhances the tumor growth promoting cell adhesion and proliferation. These data can help develop novel diagnostic markers and molecular targets for MPM.
Collapse
Affiliation(s)
- Chiara Agostinis
- Institute for Maternal and Child Health, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Burlo Garofolo, Trieste, Italy
| | - Romana Vidergar
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Beatrice Belmonte
- Department of Human Pathology, University of Palermo, Palermo, Italy
| | | | - Leonardo Amadio
- Institute for Maternal and Child Health, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Burlo Garofolo, Trieste, Italy
| | - Pietro Geri
- Department of Medical, Surgical and Health Science, University of Trieste, Trieste, Italy
| | - Violetta Borelli
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Fabrizio Zanconati
- Department of Medical, Surgical and Health Science, University of Trieste, Trieste, Italy
| | - Francesco Tedesco
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Istituto Auxologico Italiano, Milan, Italy
| | - Marco Confalonieri
- Department of Medical, Surgical and Health Science, University of Trieste, Trieste, Italy
| | - Claudio Tripodo
- Department of Human Pathology, University of Palermo, Palermo, Italy
| | - Uday Kishore
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Roberta Bulla
- Department of Life Sciences, University of Trieste, Trieste, Italy
| |
Collapse
|
42
|
Tissue and plasma levels of galectins in patients with high grade serous ovarian carcinoma as new predictive biomarkers. Sci Rep 2017; 7:13244. [PMID: 29038585 PMCID: PMC5643335 DOI: 10.1038/s41598-017-13802-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 10/02/2017] [Indexed: 12/25/2022] Open
Abstract
Galectins are moving closer to center stage in detecting glycosylation aberration in cancer cells. Here, we have investigated the expression of galectins in ovarian cancer (OC) and examined their potential as biomarkers in tissues and blood plasma samples of high grade serous ovarian carcinoma (HGSC) patients. In tissues, we found that increased protein expression of stromal gal-1 and epithelial gal-8/9 was associated with a poor response to treatment of HGSC patients. Gal-8/9 were both independent predictors of chemoresistance and overall survival (OS), respectively. This galectin signature increased the predictive value of the cancer antigen 125 (CA125) on 5-year disease-free survival (DFS), post-chemotherapy treatment and 5-year OS. In CA125LOW patients, epithelial gal-9 was associated with a lower 5-year OS while stromal gal-1 and epithelial gal-8 were both associated with a lower 5-year DFS. Such negative predictive value of gal-8 and gal-9 was also found using plasma samples. In both cases, high plasma levels of gal-8 and gal-9 was associated with a lower OS and DFS. Overall, these data suggest that galectins may be promising biomarkers to identify subgroups of HGSC patients with poorer prognosis. Our study also contributes to better define the heterogeneity of the disease.
Collapse
|
43
|
Tyekucheva S, Bowden M, Bango C, Giunchi F, Huang Y, Zhou C, Bondi A, Lis R, Van Hemelrijck M, Andrén O, Andersson SO, Watson RW, Pennington S, Finn SP, Martin NE, Stampfer MJ, Parmigiani G, Penney KL, Fiorentino M, Mucci LA, Loda M. Stromal and epithelial transcriptional map of initiation progression and metastatic potential of human prostate cancer. Nat Commun 2017; 8:420. [PMID: 28871082 PMCID: PMC5583238 DOI: 10.1038/s41467-017-00460-4] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 06/29/2017] [Indexed: 01/02/2023] Open
Abstract
While progression from normal prostatic epithelium to invasive cancer is driven by molecular alterations, tumor cells and cells in the cancer microenvironment are co-dependent and co-evolve. Few human studies to date have focused on stroma. Here, we performed gene expression profiling of laser capture microdissected normal non-neoplastic prostate epithelial tissue and compared it to non-transformed and neoplastic low-grade and high-grade prostate epithelial tissue from radical prostatectomies, each with its immediately surrounding stroma. Whereas benign epithelium in prostates with and without tumor were similar in gene expression space, stroma away from tumor was significantly different from that in prostates without cancer. A stromal gene signature reflecting bone remodeling and immune-related pathways was upregulated in high compared to low-Gleason grade cases. In validation data, the signature discriminated cases that developed metastasis from those that did not. These data suggest that the microenvironment may influence prostate cancer initiation, maintenance, and metastatic progression.Stromal cells contribute to tumor development but the mechanisms regulating this process are still unclear. Here the authors analyze gene expression profiles in the prostate and show that stromal gene signature changes ahead of the epithelial gene signature as prostate cancer initiates and progresses.
Collapse
Affiliation(s)
- Svitlana Tyekucheva
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02215, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA, 02115, USA
| | - Michaela Bowden
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02215, USA
| | - Clyde Bango
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02215, USA
| | - Francesca Giunchi
- Department of Pathology, Addarii Institute of Oncology, S.Orsola-Malpighi Teaching Hospital, University of Bologna, Viale Ercolani 4/2, 40138, Bologna, Italy
| | - Ying Huang
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02215, USA
| | - Chensheng Zhou
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02215, USA
| | - Arrigo Bondi
- Department of Surgical Pathology, Maggiore Hospital, Largo Nigrisoli 2, 40133, Bologna, Italy
| | - Rosina Lis
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02215, USA
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St, Boston, MA, 02115, USA
| | - Mieke Van Hemelrijck
- King's College London, Division of Cancer Studies, Translational Oncology & Urology Research, Guy's Hospital, London, SE1 9RT, UK
| | - Ove Andrén
- Department of Urology, School of Health and Medical Sciences, Örebro University Hospital, Örebro, SE 701 85, Sweden
| | - Sven-Olof Andersson
- Department of Urology, School of Health and Medical Sciences, Örebro University Hospital, Örebro, SE 701 85, Sweden
| | - R William Watson
- School of Medicine, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin, 4, Ireland
| | - Stephen Pennington
- School of Medicine, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin, 4, Ireland
| | - Stephen P Finn
- Department of Histopathology and Morbid Anatomy, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Neil E Martin
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St, Boston, MA, 02115, USA
| | - Meir J Stampfer
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 181 Longwood Ave, Boston, MA, 02115, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA, 02115, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, 02215, USA
| | - Giovanni Parmigiani
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02215, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA, 02115, USA
| | - Kathryn L Penney
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 181 Longwood Ave, Boston, MA, 02115, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA, 02115, USA
| | - Michelangelo Fiorentino
- Department of Pathology, Addarii Institute of Oncology, S.Orsola-Malpighi Teaching Hospital, University of Bologna, Viale Ercolani 4/2, 40138, Bologna, Italy
| | - Lorelei A Mucci
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 181 Longwood Ave, Boston, MA, 02115, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA, 02115, USA
| | - Massimo Loda
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02215, USA.
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St, Boston, MA, 02115, USA.
- The Broad Institute, 415 Main St, Cambridge, MA, 02142, USA.
| |
Collapse
|
44
|
Guo J, Gong G, Zhang B. Screening and identification of potential biomarkers in triple-negative breast cancer by integrated analysis. Oncol Rep 2017; 38:2219-2228. [PMID: 28849078 DOI: 10.3892/or.2017.5911] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 06/29/2017] [Indexed: 11/06/2022] Open
Abstract
Triple-negative breast cancer (TNBC) has attracted great attention due to its unique biology, poor prognosis, and aggressiveness. TNBC patients are more likely to suffer from metastasis. We screened and identified the TNBC-specific genes as potential biomarkers. A total of 167 breast cancer samples (45 TNBC and 122 non-TNBC) were used in the integrated analysis. Gene expression microarrays were used to screen the differentially expressed genes. We identified 65 core DEGs. According to the GO and KEGG analysis, the gene function enrichment in TNBC was revealed, such as basal cell carcinoma, prostate cancer, oocyte meiosis and choline metabolism in cancer pathways. Moreover, the PPI network reconstruction would benefit the screening of hubs. A RFS analysis of TNBC-specific genes was also conducted. RT-PCR was used to validate the expression pattern of hubs in TNBC. Finally, nine genes were identified and all of them were novel, specific and higher dysregulation expressed genes in TNBC. Such that, these genes will serve as potential biomarkers in TNBC and benefit further research in TNBC.
Collapse
Affiliation(s)
- Jilong Guo
- Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for Nationalities, Tongliao, Inner Mongolia 028000, P.R. China
| | - Guohua Gong
- Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for Nationalities, Tongliao, Inner Mongolia 028000, P.R. China
| | - Bin Zhang
- Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for Nationalities, Tongliao, Inner Mongolia 028000, P.R. China
| |
Collapse
|
45
|
Guo J, Gong G, Zhang B. Identification and prognostic value of anterior gradient protein 2 expression in breast cancer based on tissue microarray. Tumour Biol 2017; 39:1010428317713392. [PMID: 28671019 DOI: 10.1177/1010428317713392] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Breast cancer has attracted substantial attention as one of the major cancers causing death in women. It is crucial to find potential biomarkers of prognostic value in breast cancer. In this study, the expression pattern of anterior gradient protein 2 in breast cancer was identified based on the main molecular subgroups. Through analysis of 69 samples from the Gene Expression Omnibus database, we found that anterior gradient protein 2 expression was significantly higher in non-triple-negative breast cancer tissues compared with normal tissues and triple-negative breast cancer tissues (p < 0.05). The data from a total of 622 patients from The Cancer Genome Atlas were analysed. The data from The Cancer Genome Atlas and results from quantitative reverse transcription polymerase chain reaction also verified the anterior gradient protein 2 expression pattern. Furthermore, we performed immunohistochemical analysis. The quantification results revealed that anterior gradient protein 2 is highly expressed in non-triple-negative breast cancer (grade 3 excluded) and grade 1 + 2 (triple-negative breast cancer excluded) tumours compared with normal tissues. Anterior gradient protein 2 was significantly highly expressed in non-triple-negative breast cancer (grade 3 excluded) and non-triple-negative breast cancer tissues compared with triple-negative breast cancer tissues (p < 0.01). In addition, anterior gradient protein 2 was significantly highly expressed in grade 1 + 2 (triple-negative breast cancer excluded) and grade 1 + 2 tissues compared with grade 3 tissues (p < 0.05). Analysis by Fisher's exact test revealed that anterior gradient protein 2 expression was significantly associated with histologic type, histological grade, oestrogen status and progesterone status. Univariate analysis of clinicopathological variables showed that anterior gradient protein 2 expression, tumour size and lymph node status were significantly correlated with overall survival in patients with grade 1 and 2 tumours. Cox multivariate analysis revealed anterior gradient protein 2 as a putative independent indicator of unfavourable outcomes (p = 0.031). All these data clearly showed that anterior gradient protein 2 is highly expressed in breast cancer and can be regarded as a putative biomarker for breast cancer prognosis.
Collapse
Affiliation(s)
- Jilong Guo
- 1 Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for Nationalities, Tongliao, Inner Mongolia, China.,2 Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao, Inner Mongolia, China
| | - Guohua Gong
- 1 Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for Nationalities, Tongliao, Inner Mongolia, China.,2 Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao, Inner Mongolia, China.,3 Affiliated Hospital of Inner Mongolia University for Nationalities, Institute of Mongolia and Western Medicinal treatment, Tongliao, Inner Mongolia, China
| | - Bin Zhang
- 1 Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for Nationalities, Tongliao, Inner Mongolia, China.,2 Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao, Inner Mongolia, China.,3 Affiliated Hospital of Inner Mongolia University for Nationalities, Institute of Mongolia and Western Medicinal treatment, Tongliao, Inner Mongolia, China
| |
Collapse
|
46
|
Merlino G, Miodini P, Callari M, D'Aiuto F, Cappelletti V, Daidone MG. Prognostic and functional role of subtype-specific tumor-stroma interaction in breast cancer. Mol Oncol 2017; 11:1399-1412. [PMID: 28672102 PMCID: PMC5623822 DOI: 10.1002/1878-0261.12107] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 06/16/2017] [Accepted: 06/20/2017] [Indexed: 12/22/2022] Open
Abstract
None of the clinically relevant gene expression signatures available for breast cancer were specifically developed to capture the influence of the microenvironment on tumor cells. Here, we attempted to build subtype‐specific signatures derived from an in vitro model reproducing tumor cell modifications after interaction with activated or normal stromal cells. Gene expression signatures derived from HER2+, luminal, and basal breast cancer cell lines (treated by normal fibroblasts or cancer‐associated fibroblasts conditioned media) were evaluated in clinical tumors by in silico analysis on published gene expression profiles (GEPs). Patients were classified as microenvironment‐positive (μENV+ve), that is, with tumors showing molecular profiles suggesting activation by the stroma, or microenvironment‐negative (μENV−ve) based on correlation of their tumors' GEP with the respective subtype‐specific signature. Patients with estrogen receptor alpha (ER)+/HER2−/μENV+ve tumors were characterized by 2.5‐fold higher risk of developing distant metastases (HR = 2.546; 95% CI: 1.751–3.701, P = 9.84E‐07), while μENV status did not affect, or only suggested the risk of distant metastases, in women with HER2+ (HR = 1.541; 95% CI: 0.788–3.012, P = 0.206) or ER‐/HER2− tumors (HR = 1.894; 95% CI: 0.938–3.824; P = 0.0747), respectively. In ER+/HER2− tumors, the μENV status remained significantly associated with metastatic progression (HR = 2.098; CI: 1.214–3.624; P = 0.00791) in multivariable analysis including size, age, and Genomic Grade Index. Validity of our in vitro model was also supported by in vitro biological endpoints such as cell growth (MTT assay) and migration/invasion (Transwell assay). In vitro‐derived gene signatures tracing the bidirectional interaction with cancer activated fibroblasts are subtype‐specific and add independent prognostic information to classical prognostic variables in women with ER+/HER2− tumors.
Collapse
Affiliation(s)
- Giuseppe Merlino
- Biomarker Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Patrizia Miodini
- Biomarker Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Maurizio Callari
- Biomarker Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Francesca D'Aiuto
- Biomarker Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Vera Cappelletti
- Biomarker Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Maria Grazia Daidone
- Biomarker Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| |
Collapse
|
47
|
Ades F, Tryfonidis K, Zardavas D. The past and future of breast cancer treatment-from the papyrus to individualised treatment approaches. Ecancermedicalscience 2017; 11:746. [PMID: 28690677 PMCID: PMC5481194 DOI: 10.3332/ecancer.2017.746] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Indexed: 12/16/2022] Open
Abstract
Cancer is one of the oldest diseases ever described, since ancient Egypt there have always been attempts to treat and cure this illness. The growing body of knowledge about breast cancer biology and improvements in surgical and medical treatments has been built over time with contributions from many talented and enthusiastic physicians and researchers. Medical advances have changed the approach from a previously incurable condition, into a surgical disease. Further improvements in cancer biology have allowed the development of systemic treatments, hormonal therapies, and targeted drugs. The description of the molecular intrinsic subtypes of breast cancer clarified the understanding of breast cancer as a group of heterogeneous diseases, associated with different clinical outcomes, and therapeutic opportunities. This paper reviews how breast cancer treatment has improved since the earliest descriptions, in ancient times, and how future approaches, such as gene signatures, molecular profiling, and liquid biopsies, aim to further develop individualised treatments and improve treatment outcomes.
Collapse
Affiliation(s)
- Felipe Ades
- Hospital Albert Einstein, Avenida Albert Einstein, 627 - Morumbi, São Paulo - SP, 05652-900 Brazil
| | - Konstantinos Tryfonidis
- European Organisation for Research and Treatment of Cancer, Avenue E. Mounier 83/11, 1200 Brussels, Belgium
| | - Dimitrios Zardavas
- Breast International Group (BIG), Boulevard de Waterloo 76, Brussels 1000, Belgium
| |
Collapse
|
48
|
Thielens NM, Tedesco F, Bohlson SS, Gaboriaud C, Tenner AJ. C1q: A fresh look upon an old molecule. Mol Immunol 2017; 89:73-83. [PMID: 28601358 DOI: 10.1016/j.molimm.2017.05.025] [Citation(s) in RCA: 154] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 05/27/2017] [Accepted: 05/29/2017] [Indexed: 12/14/2022]
Abstract
Originally discovered as part of C1, the initiation component of the classical complement pathway, it is now appreciated that C1q regulates a variety of cellular processes independent of complement activation. C1q is a complex glycoprotein assembled from 18 polypeptide chains, with a C-terminal globular head region that mediates recognition of diverse molecular structures, and an N-terminal collagen-like tail that mediates immune effector mechanisms. C1q mediates a variety of immunoregulatory functions considered important in the prevention of autoimmunity such as the enhancement of phagocytosis, regulation of cytokine production by antigen presenting cells, and subsequent alteration in T-lymphocyte maturation. Furthermore, recent advances indicate additional roles for C1q in diverse physiologic and pathologic processes including pregnancy, tissue repair, and cancer. Finally, C1q is emerging as a critical component of neuronal network refinement and homeostatic regulation within the central nervous system. This review summarizes the classical functions of C1q and reviews novel discoveries within the field.
Collapse
Affiliation(s)
| | - Francesco Tedesco
- Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Auxologico Italiano, Milan, Italy
| | | | | | | |
Collapse
|
49
|
Urda D, Luque-Baena RM, Franco L, Jerez JM, Sanchez-Marono N. Machine learning models to search relevant genetic signatures in clinical context. 2017 INTERNATIONAL JOINT CONFERENCE ON NEURAL NETWORKS (IJCNN) 2017:1649-1656. [DOI: 10.1109/ijcnn.2017.7966049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
50
|
Rakha EA, Green AR. Molecular classification of breast cancer: what the pathologist needs to know. Pathology 2017; 49:111-119. [DOI: 10.1016/j.pathol.2016.10.012] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 10/31/2016] [Indexed: 12/20/2022]
|