1
|
Rey-Vargas L, Bejarano-Rivera LM, Ballen DF, Serrano-Gómez SJ. Characterization of HER2-Low Breast Tumors among a Cohort of Colombian Women. Cancers (Basel) 2024; 16:3141. [PMID: 39335113 PMCID: PMC11430567 DOI: 10.3390/cancers16183141] [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: 05/30/2024] [Revised: 06/20/2024] [Accepted: 07/01/2024] [Indexed: 09/30/2024] Open
Abstract
HER2-low tumors have shown promise in response to antibody-drug conjugates (ADCs) in recent clinical trials, underscoring the need to characterize this group's clinical phenotype. In this study, we aimed to explore the clinicopathological features, survival rates, and HER2 amplicon mRNA expression of women affected with HER2-low breast cancer, compared with HER2-negative and HER2-positive groups. We included 516 breast cancer patients from Colombia, for whom we compared clinicopathological features, mRNA expression of three HER2 amplicon genes (ERBB2, GRB7 and MIEN1), survival and risk of mortality between HER2-low cases (1+ or 2+ with negative in situ hybridization (ISH) result) with HER2-positive (3+ or 2+ with positive ISH test) and HER2-negative (0+) cases. A higher proportion of patients with better-differentiated tumors and a lower proliferation index were observed for HER2-low tumors compared to the HER2-positive group. Additionally, HER2-low tumors showed higher mRNA expression of the ERBB2 gene and longer overall survival rates compared to HER2-negative cases. Nonetheless, a Cox-adjusted model by ER status and clinical stage showed no statistically significant differences between these groups. Our results show differences in important clinicopathological features between HER2-low and both HER2-positive and negative tumors. Given this unique phenotype, it is crucial to evaluate the potential advantages of ADC therapies for this emerging subtype of breast cancer.
Collapse
Affiliation(s)
- Laura Rey-Vargas
- Cancer Biology Research Group, National Cancer Institute, Bogotá 111411, Colombia; (L.R.-V.); (L.M.B.-R.)
- Doctoral Program in Biological Sciences, Pontificia Universidad Javeriana, Bogotá 110231, Colombia
| | - Lina María Bejarano-Rivera
- Cancer Biology Research Group, National Cancer Institute, Bogotá 111411, Colombia; (L.R.-V.); (L.M.B.-R.)
| | | | - Silvia J. Serrano-Gómez
- Cancer Biology Research Group, National Cancer Institute, Bogotá 111411, Colombia; (L.R.-V.); (L.M.B.-R.)
- Research Support and Follow-Up Group, National Cancer Institute, Bogotá 111411, Colombia
| |
Collapse
|
2
|
Zhu M, Hwang J, Xu C. Meta-analysis identifies key genes and pathways implicated in Benzo[a]pyrene exposure response. CHEMOSPHERE 2024; 364:143121. [PMID: 39154768 PMCID: PMC11424241 DOI: 10.1016/j.chemosphere.2024.143121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 07/30/2024] [Accepted: 08/15/2024] [Indexed: 08/20/2024]
Abstract
INTRODUCTION Benzo[a]pyrene (B[a]P) is a carcinogenic polycyclic aromatic hydrocarbon that poses significant risks to human health. B[a]P influences cellular processes via intricate interactions; however, a comprehensive understanding of B[a]P's effects on the transcriptome remains elusive. This study aimed to conduct a comprehensive analysis focused on identifying relevant genes and signaling pathways affected by B[a]P exposure and their impact on human gene expression. METHODS We searched the Gene Expression Omnibus database and identified four studies involving B[a]P exposure in human cells (T lymphocytes, hepatocellular carcinoma cells, and C3A cells). We utilized two approaches for differential expression analysis: the LIMMA package and linear regression. A meta-analysis was utilized to combine log fold changes (FC) and p-values from the identified studies using a random effects model. We identified significant genes at a Bonferroni-adjusted significance level of 0.05 and determined overlapping genes across datasets. Pathway enrichment analysis elucidated key cellular processes modulated by B[a]P exposure. RESULTS The meta-analysis revealed significant upregulation of CYP1B1 (log FC = 1.15, 95% CI: 0.51-1.79, P < 0.05, I2 = 82%) and ASB2 (log FC = 0.44, 95% CI: 0.20-0.67, P < 0.05, I2 = 40%) in response to B[a]P exposure. Pathway analyses identified 26 significantly regulated pathways, with the top including Aryl Hydrocarbon Receptor Signaling (P = 0.00214) and Xenobiotic Metabolism Signaling (P = 0.00550). Key genes CYP1A1, CYP1B1, and CDKN1A were implicated in multiple pathways, highlighting their roles in xenobiotic metabolism, oxidative stress response, and cell cycle regulation. CONCLUSION The results provided insights into the mechanisms of B[a]P toxicity, highlighting CYP1B1's key role in B[a]P bioactivation. The findings underscored the complexity of B[a]P's mechanisms of action and their potential implications for human health. The identified genes and pathways provided a foundation for further exploration and enhanced our understanding of the multifaceted biological activities associated with B[a]P exposure.
Collapse
Affiliation(s)
- Mingze Zhu
- Department of Occupational and Environmental Health, Hudson College of Public Health, University of Oklahoma Health Sciences, Oklahoma City, OK, 73104, USA
| | - Jooyeon Hwang
- Department of Occupational and Environmental Health, Hudson College of Public Health, University of Oklahoma Health Sciences, Oklahoma City, OK, 73104, USA; Department of Environmental & Occupational Health Sciences, School of Public Health, University of Texas Health Science Center, Houston, TX, 77030, USA; Southwest Center for Occupational and Environmental Health, University of Texas Health Science Center, Houston, TX, 77030, USA.
| | - Chao Xu
- Department of Biostatistics and Epidemiology, Hudson College of Public Health, University of Oklahoma Health Sciences, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
3
|
Lane RE, Korbie D, Khanna KK, Mohamed A, Hill MM, Trau M. Defining the relationship between cellular and extracellular vesicle (EV) content in breast cancer via an integrative multi-omic analysis. Proteomics 2024; 24:e2300089. [PMID: 38168906 DOI: 10.1002/pmic.202300089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 11/16/2023] [Accepted: 11/17/2023] [Indexed: 01/05/2024]
Abstract
Much recent research has been dedicated to exploring the utility of extracellular vesicles (EVs) as circulating disease biomarkers. Underpinning this work is the assumption that the molecular cargo of EVs directly reflects the originating cell. Few attempts have been made, however, to empirically validate this on the -omic level. To this end, we have performed an integrative multi-omic analysis of a panel of breast cancer cell lines and corresponding EVs. Whole transcriptome analysis validated that the cellular transcriptome remained stable when cultured cells are transitioned to low serum or serum-free medium for EV collection. Transcriptomic profiling of the isolated EVs indicated a positive correlation between transcript levels in cells and EVs, including disease-associated transcripts. Analysis of the EV proteome verified that HER2 protein is present in EVs, however neither the estrogen (ER) nor progesterone (PR) receptor proteins are detected regardless of cellular expression. Using multivariate analysis, we derived an EV protein signature to infer cellular patterns of ER and HER2 expression, though the ER protein could not be directly detected. Integrative analyses affirmed that the EV proteome and transcriptome captured key phenotypic hallmarks of the originating cells, supporting the potential of EVs for non-invasive monitoring of breast cancers.
Collapse
Affiliation(s)
- Rebecca E Lane
- Australian Institute for Bioengineering and Nanotechnology, Centre for Personalised Nanomedicine, The University of Queensland, St Lucia, Queensland, Australia
| | - Darren Korbie
- Australian Institute for Bioengineering and Nanotechnology, Centre for Personalised Nanomedicine, The University of Queensland, St Lucia, Queensland, Australia
| | - Kum Kum Khanna
- QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Ahmed Mohamed
- QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Michelle M Hill
- QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Matt Trau
- Australian Institute for Bioengineering and Nanotechnology, Centre for Personalised Nanomedicine, The University of Queensland, St Lucia, Queensland, Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Queensland, Australia
| |
Collapse
|
4
|
Korucu AN, Inandiklioglu N. Is STARD3 A New Biomarker for Breast Cancer? Eur J Breast Health 2024; 20:89-93. [PMID: 38571685 PMCID: PMC10985577 DOI: 10.4274/ejbh.galenos.2024.2024-1-7] [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: 01/16/2024] [Accepted: 03/17/2024] [Indexed: 04/05/2024]
Abstract
Despite advances in diagnosis and treatment, breast cancer is still one of the three most common cancers in the world and a significant cause of morbidity and mortality. Lipids play a role in many basic physiological pathways in cells, from regulating cell homeostasis to energy expenditure. As in many types of cancer, changes in lipid metabolism and their relationship have been reported in breast cancer. The STARD3 gene encodes a member of the subfamily of lipid trafficking proteins. It is a sterol-binding protein that mediates the transport of cholesterol from the endoplasmic reticulum to endosomes. It has been shown that STARD3 is correlated with human epidermal growth factor receptor 2 (HER2) amplification since it has the same localization as HER2 in the chromosome. In this review, we aimed to emphasize that investigating lipid metabolism together with the STARD3 biomarker has great potential not only for subtype-specific strategies but also for patient-specific strategies.
Collapse
Affiliation(s)
| | - Nihal Inandiklioglu
- Department of Medical Biology, Yozgat Bozok University Faculty of Medicine, Yozgat, Turkey
| |
Collapse
|
5
|
Bi Y, Hu J, Zeng L, Chen G, Cai H, Cao H, Ma Q, Wu X. Characteristics of HPV integration in cervical adenocarcinoma and squamous carcinoma. J Cancer Res Clin Oncol 2023; 149:17973-17986. [PMID: 37966613 PMCID: PMC10725361 DOI: 10.1007/s00432-023-05494-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 10/25/2023] [Indexed: 11/16/2023]
Abstract
PURPOSE HPV integration usually occurs in HPV-related cancer, and is the main cause of cancer. But the carcinogenic mechanism of HPV integration is unclear. The study aims to provide a theoretical basis for understanding the pathogenesis of cervical adenocarcinoma (AC) and cervical squamous carcinoma (SCC). METHODS We used HPV capture sequencing to obtain HPV integration sites in AC and SCC, and analyzed cytobands, distribution of genetic and genomic elements, identified integration hotspot genes, clinicopathological parameters, breakpoints of HPV16 and performed pathway analysis. Then we conducted immunohistochemical (IHC) assay to preliminarily verify the expression of most frequently integrated genes in AC, STARD3 and ERBB2. RESULTS The results revealed that the most frequently observed integrated cytoband was 17q12 in AC and 21p11.2 in SCC, respectively. The breakpoints in both AC and SCC were more tended to occur within gene regions, compared to intergenetic regions. Compared to SCC samples, AC samples had a higher prevalence of genomic elements. In AC, HPV integration has no significantly difference with clinicopathological parameters, but in SCC integration correlated with differentiation (P < 0.05). Breakpoints of HPV in SCC located in LCR more frequently compared to AC, which destroyed the activation of promoter p97. Hotspot genes of HPV integration were STARD3 and ERBB2 in AC, and RNA45S rDNA and MIR3648-1 in SCC, respectively. Meanwhile, we preliminarily proved that the expression of STARD3 and ERBB2, the most frequently integrated genes, would increase after integration. CONCLUSION These results suggested that HPV may utilize the powerful hosts' promoters to express viral oncogenes and overexpression of viral oncogenes plays a significant role in the carcinogenesis of SCC. In AC, HPV integration may affect hosts' oncogenes, and the dysregulation of oncogenes may primarily contribute to progression of AC.
Collapse
Affiliation(s)
- Yuxin Bi
- Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Medical Research Center for Gynecologic Malignancy, Wuhan, China
| | - Junbo Hu
- Department of Pathology, Maternal and Child Health Hospital of Hubei Province, Wuhan, China
| | - Ling Zeng
- Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Center for Medical Genetics, Wuhan, China
| | - Gang Chen
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hongning Cai
- Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Medical Research Center for Gynecologic Malignancy, Wuhan, China
| | - Huang Cao
- Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Medical Research Center for Gynecologic Malignancy, Wuhan, China
- Hubei Provincial Center for Medical Genetics, Wuhan, China
| | - Quanfu Ma
- Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Medical Research Center for Gynecologic Malignancy, Wuhan, China
| | - Xufeng Wu
- Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Clinical Medical Research Center for Gynecologic Malignancy, Wuhan, China.
| |
Collapse
|
6
|
Ben Hassen C, Goupille C, Vigor C, Durand T, Guéraud F, Silvente-Poirot S, Poirot M, Frank PG. Is cholesterol a risk factor for breast cancer incidence and outcome? J Steroid Biochem Mol Biol 2023; 232:106346. [PMID: 37321513 DOI: 10.1016/j.jsbmb.2023.106346] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 06/04/2023] [Accepted: 06/05/2023] [Indexed: 06/17/2023]
Abstract
Cholesterol plays important roles in many physiological processes, including cell membrane structure and function, hormone synthesis, and the regulation of cellular homeostasis. The role of cholesterol in breast cancer is complex, and some studies have suggested that elevated cholesterol levels may be associated with an increased risk of developing breast cancer, while others have found no significant association. On the other hand, other studies have shown that, for total cholesterol and plasma HDL-associated cholesterol levels, there was inverse association with breast cancer risk. One possible mechanism by which cholesterol may contribute to breast cancer risk is as a key precursor of estrogen. Other potential mechanisms by which cholesterol may contribute to breast cancer risk include its role in inflammation and oxidative stress, which have been linked to cancer progression. Cholesterol has also been shown to play a role in signaling pathways regulating the growth and proliferation of cancer cells. In addition, recent studies have shown that cholesterol metabolism can generate tumor promoters such as cholesteryl esters, oncosterone, 27-hydroxycholesterol but also tumor suppressor metabolites such as dendrogenin A. This review summarizes some of the most important clinical studies that have evaluated the role of cholesterol or its derivatives in breast cancer. It also addresses the role of cholesterol and its derivatives at the cellular level.
Collapse
Affiliation(s)
| | - Caroline Goupille
- INSERM N2C UMR1069, University of Tours, 37032 Tours, France; Department of Gynecology, CHRU Hôpital Bretonneau, boulevard Tonnellé, 37044 Tours, France
| | - Claire Vigor
- Institut des Biomolécules Max Mousseron, IBMM, Pôle Chimie Balard Recherche, Université de Montpellier, CNRS, ENSCM, 34293 CEDEX 5 Montpellier, France
| | - Thierry Durand
- Institut des Biomolécules Max Mousseron, IBMM, Pôle Chimie Balard Recherche, Université de Montpellier, CNRS, ENSCM, 34293 CEDEX 5 Montpellier, France
| | - Françoise Guéraud
- INRAE, Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Sandrine Silvente-Poirot
- Cancer Research Center of Toulouse (CRCT), Inserm, CNRS, University of Toulouse, Team INOV:"Cholesterol Metabolism and Therapeutic Innovations", Toulouse, France; Equipe labellisée par la Ligue Nationale contre le Cancer, France
| | - Marc Poirot
- Cancer Research Center of Toulouse (CRCT), Inserm, CNRS, University of Toulouse, Team INOV:"Cholesterol Metabolism and Therapeutic Innovations", Toulouse, France; Equipe labellisée par la Ligue Nationale contre le Cancer, France
| | - Philippe G Frank
- INSERM N2C UMR1069, University of Tours, 37032 Tours, France; SGS Health and Nutrition, Saint Benoît, France.
| |
Collapse
|
7
|
Wiedmann MK, Steinsvåg IV, Dinh T, Vigeland MD, Larsson PG, Hjorthaug H, Sheng Y, Mero IL, Selmer KK. Whole-exome sequencing in moyamoya patients of Northern-European origin identifies gene variants involved in Nitric Oxide metabolism: A pilot study. BRAIN & SPINE 2023; 3:101745. [PMID: 37383439 PMCID: PMC10293314 DOI: 10.1016/j.bas.2023.101745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 04/10/2023] [Accepted: 04/21/2023] [Indexed: 06/30/2023]
Abstract
Introduction Moyamoya disease (MMD) is a chronic cerebrovascular steno-occlusive disease of largely unknown etiology. Variants in the RNF213 gene are strongly associated with MMD in East-Asia. In MMD patients of Northern-European origin, no predominant susceptibility variants have been identified so far. Research question Are there specific candidate genes associated with MMD of Northern-European origin, including the known RNF213 gene? Can we establish a hypothesis for MMD phenotype and associated genetic variants identified for further research? Material and methods Adult patients of Northern-European origin, treated surgically for MMD at Oslo University Hospital between October 2018 to January 2019 were asked to participate. WES was performed, with subsequent bioinformatic analysis and variant filtering. The selected candidate genes were either previously reported in MMD or known to be involved in angiogenesis. The variant filtering was based on variant type, location, population frequency, and predicted impact on protein function. Results Analysis of WES data revealed nine variants of interest in eight genes. Five of those encode proteins involved in nitric oxide (NO) metabolism: NOS3, NR4A3, ITGAV, GRB7 and AGXT2. In the AGXT2 gene, a de novo variant was detected, not previously described in MMD. None harboured the p.R4810K missense variant in the RNF213 gene known to be associated with MMD in East-Asian patients. Discussion and conclusion Our findings suggest a role for NO regulation pathways in Northern-European MMD and introduce AGXT2 as a new susceptibility gene. This pilot study warrants replication in larger patient cohorts and further functional investigations.
Collapse
Affiliation(s)
- Markus K.H. Wiedmann
- Department of Neurosurgery, The National Hospital, Oslo University Hospital, Oslo, Norway
| | - Ingunn V. Steinsvåg
- Department of Medical Genetics, Oslo University Hospital and the University of Oslo, Oslo, Norway
| | - Tovy Dinh
- Department of Neurosurgery, The National Hospital, Oslo University Hospital, Oslo, Norway
| | - Magnus D. Vigeland
- Department of Medical Genetics, Oslo University Hospital and the University of Oslo, Oslo, Norway
| | - Pål G. Larsson
- Department of Neurosurgery, The National Hospital, Oslo University Hospital, Oslo, Norway
| | - Hanne Hjorthaug
- Department of Medical Genetics, Oslo University Hospital and the University of Oslo, Oslo, Norway
| | - Ying Sheng
- Department of Medical Genetics, Oslo University Hospital and the University of Oslo, Oslo, Norway
| | - Inger-Lise Mero
- Department of Medical Genetics, Oslo University Hospital and the University of Oslo, Oslo, Norway
| | - Kaja K. Selmer
- Department of Research and Innovation, Division of Clinical Neuroscience, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
8
|
Yang T, Kang L, Li D, Song Y. Immunotherapy for HER-2 positive breast cancer. Front Oncol 2023; 13:1097983. [PMID: 37007133 PMCID: PMC10061112 DOI: 10.3389/fonc.2023.1097983] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 03/03/2023] [Indexed: 03/18/2023] Open
Abstract
Immunotherapy is a developing treatment for advanced breast cancer. Immunotherapy has clinical significance for the treatment of triple-negative breast cancers and human epidermal growth factor receptor-2 positive (HER2+) breast cancers. As a proved effective passive immunotherapy, clinical application of the monoclonal antibodies trastuzumab, pertuzumab and T-DM1 (ado-trastuzumab emtansine) has significantly improved the survival of patients with HER2+ breast cancers. Immune checkpoint inhibitors that block programmed death receptor-1 and its ligand (PD-1/PD-L1) have also shown benefits for breast cancer in various clinical trials. Adoptive T-cell immunotherapies and tumor vaccines are emerging as novel approaches to treating breast cancer, but require further study. This article reviews recent advances in immunotherapy for HER2+ breast cancers.
Collapse
|
9
|
Pries K, Krüger S, Heckl S, Behrens HM, Röcken C. SMARCA4 and SMARCE1 in gastric cancer: Correlation with ARID1A, and microsatellite stability, and SMARCE1/ERBB2 co-amplification. Cancer Med 2023; 12:10423-10437. [PMID: 36916408 DOI: 10.1002/cam4.5776] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 02/05/2023] [Accepted: 02/24/2023] [Indexed: 03/15/2023] Open
Abstract
BACKGROUND Recent studies have shown an association between certain subunits of the SWI/SNF complex with specific tumor characteristics in gastric cancer (GC). In an earlier study, we applied multiregional whole exome sequencing on multiple primary tumor samples and found alterations of the SWI/SNF complex in 78% of the cases. ERBB2, which encodes for Her2/neu, is a well-known predictive biomarker used to guide the treatment of GC in the palliative setting. SMARCE1, which encodes for a subunit of the SWI/SNF complex, is localized in close genetic proximity to ERBB2. AIM As little is known about the significance of the SWI/SNF complex in GC biology and the potential relationship between ERBB2 and SMARCE1 upregulation, we examined the expression patterns of SMARCA4 and SMARCE1, two mutually exclusive catalytic ATPase subunits of the SWI/SNF complex, in a well characterized GC cohort. MATERIALS AND METHODS The expression of SMARCA4 and SMARCE1 was studied by immunohistochemistry in connection with clinicopathological patient characteristics in a cohort of 468 GCs. Digital droplet polymerase chain reaction was performed for amplification analysis on ERBB2 and SMARCE1. RESULTS Immunohistochemical staining of whole-mount tissue sections found a diffusely "gray scale" expression of SMARCA4 in 446 (95.2%) GCs and of SMARCE1 in 463 (98.8%) GCs. The expression of SMARCA4 and SMARCE1 correlated significantly with ARID1A, p53, and microsatellite status. No correlation was found with the patient prognosis. The amplification analysis of SMARCE1 showed amplification in 4 of 34 cases. In 3 of 34 cases, SMARCE1 was co-amplified with ERBB2. We also found a co-expression of SMARCE1 and Her2/neu in a subset of patients. CONCLUSION While the effect of a co-amplification is currently unknown, synergistic effects of SMARCE1 and Her2/neu overexpression should be explored in future studies, holding potential for an improved treatment of GC.
Collapse
Affiliation(s)
- Katharina Pries
- Department of Pathology, Christian-Albrechts-University, University-Hospital Schleswig-Holstein, Kiel, Germany
| | - Sandra Krüger
- Department of Pathology, Christian-Albrechts-University, University-Hospital Schleswig-Holstein, Kiel, Germany
| | - Steffen Heckl
- Department of Internal Medicine II, Christian-Albrechts-University, University-Hospital Schleswig-Holstein, Kiel, Germany
| | - Hans-Michael Behrens
- Department of Pathology, Christian-Albrechts-University, University-Hospital Schleswig-Holstein, Kiel, Germany
| | - Christoph Röcken
- Department of Pathology, Christian-Albrechts-University, University-Hospital Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
10
|
STARD3: A New Biomarker in HER2-Positive Breast Cancer. Cancers (Basel) 2023; 15:cancers15020362. [PMID: 36672312 PMCID: PMC9856516 DOI: 10.3390/cancers15020362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 12/23/2022] [Accepted: 12/29/2022] [Indexed: 01/09/2023] Open
Abstract
Pathological complete response (pCR) after neoadjuvant systemic treatment (NST) is an important prognostic factor in HER2-positive breast cancer. The majority of HER2-positive breast cancers are amplified at the HER2 gene locus, several genes are co-amplified with HER2, and a subset of them are co-expressed. The STARD3 gene belongs to the HER2 amplicon, and its role as a predictive marker was never addressed. The objective of this study was to investigate the predictive value of STARD3 protein expression on NST pathological response in HER2-positive breast cancer. In addition, we studied the prognostic value of this marker. METHODS We conducted a retrospective study between 2007 and 2020 on 112 patients with non-metastatic HER2-positive breast cancer treated by NST and then by surgery. We developed an immunohistochemistry assay for STARD3 expression and subcellular localization and determined a score for STARD3-positivity. As STARD3 is an endosomal protein, its expression was considered positive if the intracellular signal pattern was granular. RESULTS In this series, pCR was achieved in half of the patients. STARD3 was positive in 86.6% of cases and was significantly associated with pCR in univariate analysis (p = 0.013) and after adjustment on other known pathological parameters (p = 0.044). Performances on pCR prediction showed high sensitivity (96%) and negative predictive value (87%), while specificity was 23% and positive predictive value was 56%. Overall, specific, relapse-free, and distant metastasis-free survivals were similar among STARD3 positive and negative groups, independently of other prognosis factors. CONCLUSION NST is an opportunity for HER2-positive cancers. In this series of over a hundred HER2-positive and non-metastatic patients, a STARD3-negative score was associated with the absence of pathological complete response. This study suggests that determining STARD3 overexpression status on initial biopsies of HER2-positive tumors is an added value for the management of a subset of patients with high probability of no pathological response.
Collapse
|
11
|
Cheng X, Huang Z, Pan A, Long D. ORLNC1 Suppresses Cell Growth in HER2-Positive Breast Cancer via miRNA-296 Sponging. Curr Mol Med 2023; 23:289-299. [PMID: 35658886 DOI: 10.2174/1566524022666220603113550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/08/2022] [Accepted: 03/17/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Accumulating research has demonstrated that aberrant levels of long noncoding RNAs (LncRNAs) are related to cancer progression. The effects of ORLNC1 in HER2+ breast cancer have yet to be explored. METHODS Real-time PCR was used to examine the expression of LncRNA ORLNC1 in HER+ breast cancer. CCK-8, wound healing and cell invasion assays were used to examine the effect of LncRNA ORLNC1 on HER+ breast cancer cells. Luciferase reporter assay was utilized to determine the regulatory relationship between LncRNA ORLNC1 and miR-296. Western blotting was used to measure the expression of PTEN. Xenograft mouse model was used to examine the effect of LncRNA ORLNC1 on tumor progression in vivo. RESULTS In this study, our findings revealed downregulation of ORLNC1 in HER2+ breast cancer specimens and cell lines. Low levels of ORLNC1 were related to poor prognosis and advanced cancer stage. Using gain- and loss-of-function assays, the ability of these tumor cells to proliferate was found to be inhibited by ORLNC1 in vitro and in vivo. Further analyses revealed that miR-296/PTEN axis is directly targeted by ORLNC1. Consequently, over-expression of miR-296 efficiently abrogated the upregulation of PTEN induced by ORLNC1, suggesting that ORLNC1 positively regulates PTEN expression by competitively binding to miR-296. CONCLUSION Our results indicate that lncRNA ORLNC1 acts as a tumor suppressor by regulating the miR-296/PTEN axis in HER2+ breast cancer.
Collapse
Affiliation(s)
- Xueyuan Cheng
- Department of General Surgery, Beihai People's Hospital, Beihai, Guangxi, 536000, China
| | - Zhong Huang
- Department of General Surgery, Beihai People's Hospital, Beihai, Guangxi, 536000, China
| | - Anchao Pan
- Department of Gastrointestinal Surgery, Wuming Hospital of Guangxi Medical University, Nanning, Guangxi, 530199, China
| | - Di Long
- Department of Gastrointestinal Surgery, Wuming Hospital of Guangxi Medical University, Nanning, Guangxi, 530199, China
| |
Collapse
|
12
|
Gámez-Chiachio M, Sarrió D, Moreno-Bueno G. Novel Therapies and Strategies to Overcome Resistance to Anti-HER2-Targeted Drugs. Cancers (Basel) 2022; 14:4543. [PMID: 36139701 PMCID: PMC9496705 DOI: 10.3390/cancers14184543] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/09/2022] [Accepted: 09/15/2022] [Indexed: 11/17/2022] Open
Abstract
The prognosis and quality of life of HER2 breast cancer patients have significantly improved due to the crucial clinical benefit of various anti-HER2 targeted therapies. However, HER2 tumors can possess or develop several resistance mechanisms to these treatments, thus leaving patients with a limited set of additional therapeutic options. Fortunately, to overcome this problem, in recent years, multiple different and complementary approaches have been developed (such as antibody-drug conjugates (ADCs)) that are in clinical or preclinical stages. In this review, we focus on emerging strategies other than on ADCs that are either aimed at directly target the HER2 receptor (i.e., novel tyrosine kinase inhibitors) or subsequent intracellular signaling (e.g., PI3K/AKT/mTOR, CDK4/6 inhibitors, etc.), as well as on innovative approaches designed to attack other potential tumor weaknesses (such as immunotherapy, autophagy blockade, or targeting of other genes within the HER2 amplicon). Moreover, relevant technical advances such as anti-HER2 nanotherapies and immunotoxins are also discussed. In brief, this review summarizes the impact of novel therapeutic approaches on current and future clinical management of aggressive HER2 breast tumors.
Collapse
Affiliation(s)
- Manuel Gámez-Chiachio
- Biochemistry Department, Medicine Faculty, Universidad Autónoma Madrid-CSIC, IdiPaz, 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red-Oncología (CIBERONC), 28029 Madrid, Spain
| | - David Sarrió
- Biochemistry Department, Medicine Faculty, Universidad Autónoma Madrid-CSIC, IdiPaz, 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red-Oncología (CIBERONC), 28029 Madrid, Spain
| | - Gema Moreno-Bueno
- Biochemistry Department, Medicine Faculty, Universidad Autónoma Madrid-CSIC, IdiPaz, 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red-Oncología (CIBERONC), 28029 Madrid, Spain
- MD Anderson International Foundation, 28033 Madrid, Spain
| |
Collapse
|
13
|
Novel 2,6,9-Trisubstituted Purines as Potent CDK Inhibitors Alleviating Trastuzumab-Resistance of HER2-Positive Breast Cancers. Pharmaceuticals (Basel) 2022; 15:ph15091041. [PMID: 36145262 PMCID: PMC9506414 DOI: 10.3390/ph15091041] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 08/19/2022] [Indexed: 11/16/2022] Open
Abstract
HER2-positive (HER2+) breast cancer is defined by HER2 oncogene amplification on chromosome 17q12 and accounts for 15−20% population of breast-cancer patients. Therapeutic anti-HER2 antibody such as trastuzumab is used as the first-line therapy for HER2-positive breast cancers. However, more than 50% of the patients respond poorly to trastuzumab, illustrating that novel therapy is warranted to overcome the resistance. We previously reported that in the majority of HER2+ breast-cancer patients, CDK12 is co-amplified on 17q12 and involved in developing tumors and trastuzumab resistance, proposing CDK12 as a potential drug target for HER2+ breast cancers. Here, we designed and synthesized novel 2,6,9-trisubstituted purines as potent CDK12 inhibitors showing strong, equipotent antiproliferative activity against trastuzumab-sensitive HER2+ SK-Br3 cells and trastuzumab-resistant HER2+ HCC1954 cells (GI50 values < 50 nM) both of which express a high level of CDK12. Two potent analogue 30d and 30e at 40, 200 nM greatly downregulated the levels of cyclinK and Pol II p-CTD (Ser2), as well as the expression of CDK12 downstream genes (IRS1 and WNT1) in a dose-dependent manner. We also observed structure-property relationship for a subset of potent analogues, and found that 30e is highly stable in liver microsomes with lack of CYP inhibition. In addition, 30d exhibited a synergy with trastuzumab in the both cells, suggesting that our inhibitors could be applied to alleviate trastuzumab-resistance of HER2+ breast cancers and escalate the efficacy of trastuzumab as well. Our study may provide insight into developing a novel therapy for HER2+ breast cancers.
Collapse
|
14
|
Moon J, Song J, Jang H, Kang H, Huh YM, Son HY, Rho HW, Park M, Talwar CS, Park KH, Woo E, Lim J, Lim EK, Jung J, Jung Y, Park HG, Kang T. Ligation-free isothermal nucleic acid amplification. Biosens Bioelectron 2022; 209:114256. [DOI: 10.1016/j.bios.2022.114256] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/29/2022] [Accepted: 04/04/2022] [Indexed: 12/19/2022]
|
15
|
Tokura M, Nakayama J, Prieto-Vila M, Shiino S, Yoshida M, Yamamoto T, Watanabe N, Takayama S, Suzuki Y, Okamoto K, Ochiya T, Kohno T, Yatabe Y, Suto A, Yamamoto Y. Single-Cell Transcriptome Profiling Reveals Intratumoral Heterogeneity and Molecular Features of Ductal Carcinoma In Situ. Cancer Res 2022; 82:3236-3248. [DOI: 10.1158/0008-5472.can-22-0090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 04/25/2022] [Accepted: 07/11/2022] [Indexed: 11/16/2022]
Abstract
Abstract
Ductal carcinoma in situ (DCIS) is a precursor to invasive breast cancer. The frequency of DCIS is increasing because of routine mammography; however, the biological features and intratumoral heterogeneity of DCIS remain obscure. To address this deficiency, we performed single-cell transcriptomic profiling of DCIS and invasive ductal carcinoma (IDC). DCIS was found to be composed of several transcriptionally distinct subpopulations of cancer cells with specific functions. Several transcripts, including long noncoding RNAs, were highly expressed in IDC compared to DCIS and might be related to the invasive phenotype. Closeness centrality analysis revealed extensive heterogeneity in DCIS, and the prediction model for cell-to-cell interactions implied that the interaction network among luminal cells and immune cells in DCIS was comparable to that in IDC. Additionally, transcriptomic profiling of HER2+ luminal DCIS indicated HER2 genomic amplification at the DCIS stage. These data provide novel insight into the intratumoral heterogeneity and molecular features of DCIS, which exhibit properties similar to IDC.
Collapse
Affiliation(s)
- Momoko Tokura
- National Cancer Center Research Institute, Tokyo, Japan
| | - Jun Nakayama
- National Cancer Center Research Institute, Tokyo, Japan
| | - Marta Prieto-Vila
- Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Sho Shiino
- National Cancer Center Hospital, Keio University School of Medicine, Tokyo, Japan
| | | | | | | | | | | | - Koji Okamoto
- National Cancer Center Research Institute, Tokyo, Japan
| | | | - Takashi Kohno
- National Cancer Center Research Institute, Tokyo, Japan
| | | | | | | |
Collapse
|
16
|
Linking Late Endosomal Cholesterol with Cancer Progression and Anticancer Drug Resistance. Int J Mol Sci 2022; 23:ijms23137206. [PMID: 35806209 PMCID: PMC9267071 DOI: 10.3390/ijms23137206] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 06/22/2022] [Accepted: 06/25/2022] [Indexed: 11/16/2022] Open
Abstract
Cancer cells undergo drastic metabolic adaptions to cover increased bioenergetic needs, contributing to resistance to therapies. This includes a higher demand for cholesterol, which often coincides with elevated cholesterol uptake from low-density lipoproteins (LDL) and overexpression of the LDL receptor in many cancers. This implies the need for cancer cells to accommodate an increased delivery of LDL along the endocytic pathway to late endosomes/lysosomes (LE/Lys), providing a rapid and effective distribution of LDL-derived cholesterol from LE/Lys to other organelles for cholesterol to foster cancer growth and spread. LDL-cholesterol exported from LE/Lys is facilitated by Niemann–Pick Type C1/2 (NPC1/2) proteins, members of the steroidogenic acute regulatory-related lipid transfer domain (StARD) and oxysterol-binding protein (OSBP) families. In addition, lysosomal membrane proteins, small Rab GTPases as well as scaffolding proteins, including annexin A6 (AnxA6), contribute to regulating cholesterol egress from LE/Lys. Here, we summarize current knowledge that links upregulated activity and expression of cholesterol transporters and related proteins in LE/Lys with cancer growth, progression and treatment outcomes. Several mechanisms on how cellular distribution of LDL-derived cholesterol from LE/Lys influences cancer cell behavior are reviewed, some of those providing opportunities for treatment strategies to reduce cancer progression and anticancer drug resistance.
Collapse
|
17
|
Yin C, Cao Y, Sun P, Zhang H, Li Z, Xu Y, Sun H. Molecular Subtyping of Cancer Based on Robust Graph Neural Network and Multi-Omics Data Integration. Front Genet 2022; 13:884028. [PMID: 35646077 PMCID: PMC9137453 DOI: 10.3389/fgene.2022.884028] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 03/31/2022] [Indexed: 11/13/2022] Open
Abstract
Accurate molecular subtypes prediction of cancer patients is significant for personalized cancer diagnosis and treatments. Large amount of multi-omics data and the advancement of data-driven methods are expected to facilitate molecular subtyping of cancer. Most existing machine learning–based methods usually classify samples according to single omics data, fail to integrate multi-omics data to learn comprehensive representations of the samples, and ignore that information transfer and aggregation among samples can better represent them and ultimately help in classification. We propose a novel framework named multi-omics graph convolutional network (M-GCN) for molecular subtyping based on robust graph convolutional networks integrating multi-omics data. We first apply the Hilbert–Schmidt independence criterion least absolute shrinkage and selection operator (HSIC Lasso) to select the molecular subtype-related transcriptomic features and then construct a sample–sample similarity graph with low noise by using these features. Next, we take the selected gene expression, single nucleotide variants (SNV), and copy number variation (CNV) data as input and learn the multi-view representations of samples. On this basis, a robust variant of graph convolutional network (GCN) model is finally developed to obtain samples’ new representations by aggregating their subgraphs. Experimental results of breast and stomach cancer demonstrate that the classification performance of M-GCN is superior to other existing methods. Moreover, the identified subtype-specific biomarkers are highly consistent with current clinical understanding and promising to assist accurate diagnosis and targeted drug development.
Collapse
Affiliation(s)
- Chaoyi Yin
- School of Artificial Intelligence, Jilin University, Changchun, China
| | - Yangkun Cao
- School of Artificial Intelligence, Jilin University, Changchun, China
| | - Peishuo Sun
- School of Artificial Intelligence, Jilin University, Changchun, China
| | - Hengyuan Zhang
- School of Artificial Intelligence, Jilin University, Changchun, China
| | - Zhi Li
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang, China
- *Correspondence: Zhi Li, ; Huiyan Sun,
| | - Ying Xu
- Computational Systems Biology Lab, Department of Biochemistry and Molecular Biology and Institute of Bioinformatics, University of Georgia, Athens, GA, United States
| | - Huiyan Sun
- School of Artificial Intelligence, Jilin University, Changchun, China
- *Correspondence: Zhi Li, ; Huiyan Sun,
| |
Collapse
|
18
|
Asif K, Memeo L, Palazzolo S, Frión-Herrera Y, Parisi S, Caligiuri I, Canzonieri V, Granchi C, Tuccinardi T, Rizzolio F. STARD3: A Prospective Target for Cancer Therapy. Cancers (Basel) 2021; 13:4693. [PMID: 34572920 PMCID: PMC8472075 DOI: 10.3390/cancers13184693] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 09/10/2021] [Indexed: 12/24/2022] Open
Abstract
Cancer is one of the major causes of death in developed countries and current therapies are based on surgery, chemotherapeutic agents, and radiation. To overcome side effects induced by chemo- and radiotherapy, in recent decades, targeted therapies have been proposed in second and even first lines. Targeted drugs act on the essential pathways involved in tumor induction, progression, and metastasis, basically all the hallmark of cancers. Among emerging pathways, the cholesterol metabolic pathway is a strong candidate for this purpose. Cancer cells have an accelerated metabolic rate and require a continuous supply of cholesterol for cell division and membrane renewal. Steroidogenic acute regulatory related lipid transfer (START) proteins are a family of proteins involved in the transfer of lipids and some of them are important in non-vesicular cholesterol transportation within the cell. The alteration of their expression levels is implicated in several diseases, including cancers. In this review, we report the latest discoveries on StAR-related lipid transfer protein domain 3 (STARD3), a member of the START family, which has a potential role in cancer, focusing on the structural and biochemical characteristics and mechanisms that regulate its activity. The role of the STARD3 protein as a molecular target for the development of cancer therapies is also discussed. As STARD3 is a key protein in the cholesterol movement in cancer cells, it is of interest to identify inhibitors able to block its activity.
Collapse
Affiliation(s)
- Kanwal Asif
- Department of Molecular Sciences and Nanosystems, PhD School in Science and Technology of Bio and Nanomaterials, Ca’ Foscari University of Venice, 30172 Venice, Italy;
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (C.R.O.) IRCCS, 33081 Aviano, Italy; (S.P.); (S.P.); (V.C.)
| | - Lorenzo Memeo
- Department of Experimental Oncology, Mediterranean Institute of Oncology, 95029 Catania, Italy;
| | - Stefano Palazzolo
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (C.R.O.) IRCCS, 33081 Aviano, Italy; (S.P.); (S.P.); (V.C.)
| | - Yahima Frión-Herrera
- Department of Molecular Sciences and Nanosystems, Ca’ Foscari University of Venice, 30172 Venice, Italy; or
| | - Salvatore Parisi
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (C.R.O.) IRCCS, 33081 Aviano, Italy; (S.P.); (S.P.); (V.C.)
| | - Isabella Caligiuri
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (C.R.O.) IRCCS, 33081 Aviano, Italy; (S.P.); (S.P.); (V.C.)
| | - Vincenzo Canzonieri
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (C.R.O.) IRCCS, 33081 Aviano, Italy; (S.P.); (S.P.); (V.C.)
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34149 Trieste, Italy
| | - Carlotta Granchi
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (C.G.); (T.T.)
| | - Tiziano Tuccinardi
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (C.G.); (T.T.)
| | - Flavio Rizzolio
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (C.R.O.) IRCCS, 33081 Aviano, Italy; (S.P.); (S.P.); (V.C.)
- Department of Molecular Sciences and Nanosystems, Ca’ Foscari University of Venice, 30172 Venice, Italy; or
| |
Collapse
|
19
|
Rubio AJ, Bencomo-Alvarez AE, Young JE, Velazquez VV, Lara JJ, Gonzalez MA, Eiring AM. 26S Proteasome Non-ATPase Regulatory Subunits 1 (PSMD1) and 3 (PSMD3) as Putative Targets for Cancer Prognosis and Therapy. Cells 2021; 10:2390. [PMID: 34572038 PMCID: PMC8472613 DOI: 10.3390/cells10092390] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 08/20/2021] [Accepted: 09/08/2021] [Indexed: 12/30/2022] Open
Abstract
Ever since the ubiquitin proteasome system was characterized, efforts have been made to manipulate its function to abrogate the progression of cancer. As a result, the anti-cancer drugs bortezomib, carfilzomib, and ixazomib targeting the 26S proteasome were developed to treat multiple myeloma, mantle cell lymphoma, and diffuse large B-cell lymphoma, among others. Despite success, adverse side effects and drug resistance are prominent, raising the need for alternative therapeutic options. We recently demonstrated that knockdown of the 19S regulatory components, 26S proteasome non-ATPase subunits 1 (PSMD1) and 3 (PSMD3), resulted in increased apoptosis of chronic myeloid leukemia (CML) cells, but had no effect on normal controls, suggesting they may be good targets for therapy. Therefore, we hypothesized that PSMD1 and PSMD3 are potential targets for anti-cancer therapeutics and that their relevance stretches beyond CML to other types of cancers. In the present study, we analyzed PSMD1 and PSMD3 mRNA and protein expression in cancerous tissue versus normal controls using data from The Cancer Genome Atlas (TCGA) and the Clinical Proteomic Tumor Analysis Consortium (CPTAC), comparing expression with overall survival. Altogether, our data suggest that PSMD1 and PSMD3 may be novel putative targets for cancer prognosis and therapy that are worthy of future investigation.
Collapse
Affiliation(s)
- Andres J Rubio
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center at El Paso, El Paso, TX 79905, USA
| | - Alfonso E Bencomo-Alvarez
- Center of Emphasis in Cancer, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center at El Paso, El Paso, TX 79905, USA
| | - James E Young
- Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center at El Paso, El Paso, TX 79905, USA
| | - Vanessa V Velazquez
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center at El Paso, El Paso, TX 79905, USA
| | - Joshua J Lara
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center at El Paso, El Paso, TX 79905, USA
| | - Mayra A Gonzalez
- Center of Emphasis in Cancer, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center at El Paso, El Paso, TX 79905, USA
| | - Anna M Eiring
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center at El Paso, El Paso, TX 79905, USA
- Center of Emphasis in Cancer, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center at El Paso, El Paso, TX 79905, USA
- Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center at El Paso, El Paso, TX 79905, USA
| |
Collapse
|
20
|
Orfanou IM, Argyros O, Papapetropoulos A, Tseleni-Balafouta S, Vougas K, Tamvakopoulos C. Discovery and Pharmacological Evaluation of STEAP4 as a Novel Target for HER2 Overexpressing Breast Cancer. Front Oncol 2021; 11:608201. [PMID: 33842315 PMCID: PMC8034292 DOI: 10.3389/fonc.2021.608201] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 03/08/2021] [Indexed: 01/11/2023] Open
Abstract
Breast cancer (BC) is a highly heterogeneous disease encompassing multiple subtypes with different molecular and histopathological features, disease prognosis, and therapeutic responses. Among these, the Triple Negative BC form (TNBC) is an aggressive subtype with poor prognosis and therapeutic outcome. With respect to HER2 overexpressing BC, although advanced targeted therapies have improved the survival of patients, disease relapse and metastasis remains a challenge for therapeutic efficacy. In this study the aim was to identify key membrane-associated proteins which are overexpressed in these aggressive BC subtypes and can serve as potential biomarkers or drug targets. We leveraged on the development of a membrane enrichment protocol in combination with the global profiling GeLC-MS/MS technique, and compared the proteomic profiles of a HER2 overexpressing (HCC-1954) and a TNBC (MDA-MB-231) cell line with that of a benign control breast cell line (MCF-10A). An average of 2300 proteins were identified from each cell line, of which approximately 600 were membrane-associated proteins. Our global proteomic methodology in tandem with invigoration by Western blot and Immunofluorescence analysis, readily detected several previously-established BC receptors like HER2 and EPHA2, but importantly STEAP4 and CD97 emerged as novel potential candidate markers. This is the first time that the mitochondrial iron reductase STEAP4 protein up-regulation is linked to BC (HER2+ subtype), while for CD97, its role in BC has been previously described, but never before by a global proteomic technology in TNBC. STEAP4 was selected for further detailed evaluation by the employment of Immunohistochemical analysis of BC xenografts and clinical tissue microarray studies. Results showed that STEAP4 expression was evident only in malignant breast tissues whereas all the benign breast cases had no detectable levels. A functional role of STEAP4 intervention was established in HER2 overexpressing BC by pharmacological studies, where blockage of the STEAP4 pathway with an iron chelator (Deferiprone) in combination with the HER2 inhibitor Lapatinib led to a significant reduction in cell growth in vitro. Furthermore, siRNA mediated knockdown of STEAP4 also suppressed cell proliferation and enhanced the inhibition of Lapatinib in HER2 overexpressing BC, confirming its potential oncogenic role in BC. In conclusion, STEAP4 may represent a novel BC related biomarker and a potential pharmacological target for the treatment of HER2 overexpressing BC.
Collapse
Affiliation(s)
- Ioanna-Maria Orfanou
- Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Orestis Argyros
- Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Andreas Papapetropoulos
- Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.,Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Sofia Tseleni-Balafouta
- Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Konstantinos Vougas
- Proteomics Laboratory, Division of Biotechnology, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Constantin Tamvakopoulos
- Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| |
Collapse
|
21
|
Uysal D, Kowalewski KF, Kriegmair MC, Wirtz R, Popovic ZV, Erben P. A comprehensive molecular characterization of the 8q22.2 region reveals the prognostic relevance of OSR2 mRNA in muscle invasive bladder cancer. PLoS One 2021; 16:e0248342. [PMID: 33711044 PMCID: PMC7954304 DOI: 10.1371/journal.pone.0248342] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 02/25/2021] [Indexed: 12/27/2022] Open
Abstract
Technological advances in molecular profiling have enabled the comprehensive identification of common regions of gene amplification on chromosomes (amplicons) in muscle invasive bladder cancer (MIBC). One such region is 8q22.2, which is largely unexplored in MIBC and could harbor genes with potential for outcome prediction or targeted therapy. To investigate the prognostic role of 8q22.2 and to compare different amplicon definitions, an in-silico analysis of 357 patients from The Cancer Genome Atlas, who underwent radical cystectomy for MIBC, was performed. Amplicons were generated using the GISTIC2.0 algorithm for copy number alterations (DNA_Amplicon) and z-score normalization for mRNA gene overexpression (RNA_Amplicon). Kaplan-Meier survival analysis, univariable, and multivariable Cox proportional hazard ratios were used to relate amplicons, genes, and clinical parameters to overall (OS) and disease-free survival (DFS). Analyses of the biological functions of 8q22.2 genes and genomic events in MIBC were performed to identify potential targets. Genes with prognostic significance from the in silico analysis were validated using RT-qPCR of MIBC tumor samples (n = 46). High 8q22.2 mRNA expression (RNA-AMP) was associated with lymph node metastases. Furthermore, 8q22.2 DNA and RNA amplified patients were more likely to show a luminal subtype (DNA_Amplicon_core: p = 0.029; RNA_Amplicon_core: p = 0.01). Overexpression of the 8q22.2 gene OSR2 predicted shortened DFS in univariable (HR [CI] 1.97 [1.2; 3.22]; p = 0.01) and multivariable in silico analysis (HR [CI] 1.91 [1.15; 3.16]; p = 0.01) and decreased OS (HR [CI] 6.25 [1.37; 28.38]; p = 0.0177) in RT-qPCR data analysis. Alterations in different levels of the 8q22.2 region are associated with manifestation of different clinical characteristics in MIBC. An in-depth comprehensive molecular characterization of genomic regions involved in cancer should include multiple genetic levels, such as DNA copy number alterations and mRNA gene expression, and could lead to a better molecular understanding. In this study, OSR2 is identified as a potential biomarker for survival prognosis.
Collapse
Affiliation(s)
- Daniel Uysal
- Department of Urology and Urosurgery, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Karl-Friedrich Kowalewski
- Department of Urology and Urosurgery, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | | | - Ralph Wirtz
- STRATIFYER Molecular Pathology GmbH, Köln, Germany
| | - Zoran V. Popovic
- Institute of Pathology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Philipp Erben
- Department of Urology and Urosurgery, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- * E-mail:
| |
Collapse
|
22
|
Yang Y, Leonard M, Luo Z, Yeo S, Bick G, Hao M, Cai C, Charif M, Wang J, Guan JL, Lower EE, Zhang X. Functional cooperation between co-amplified genes promotes aggressive phenotypes of HER2-positive breast cancer. Cell Rep 2021; 34:108822. [PMID: 33691110 PMCID: PMC8050805 DOI: 10.1016/j.celrep.2021.108822] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 12/21/2020] [Accepted: 02/12/2021] [Indexed: 12/21/2022] Open
Abstract
MED1 (mediator subunit 1)co-amplifies with HER2, but its role in HER2-driven mammary tumorigenesis is still unknown. Here, we generate MED1 mammary-specific overexpression mice and cross them with mouse mammary tumor virus (MMTV)-HER2 mice. We observe significantly promoted onset, growth, metastasis, and multiplicity of HER2 tumors by MED1 overexpression. Further studies reveal critical roles for MED1 in epithelial-mesenchymal transition, cancer stem cell formation, and response to anti-HER2 therapy. Mechanistically, RNA sequencing (RNA-seq) transcriptome analyses and clinical sample correlation studies identify Jab1, a component of the COP9 signalosome complex, as the key direct target gene of MED1 contributing to these processes. Further studies reveal that Jab1 can also reciprocally regulate the stability and transcriptional activity of MED1. Together, our findings support a functional cooperation between these co-amplified genes in HER2+ mammary tumorigenesis and their potential usage as therapeutic targets for the treatment of HER2+ breast cancers. In this study, Yang et al. generate a more clinically relevant MMTV-HER2/MMTV-MED1 mammary tumor mouse model and discover the critical roles and molecular mechanisms of MED1 overexpression in mediating the aggressive phenotypes of HER2+ tumor progression, metastasis, cancer stem cell formation, and therapy resistance.
Collapse
Affiliation(s)
- Yongguang Yang
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Marissa Leonard
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; Graduate Program in Cancer and Cell Biology, Vontz Center for Molecular Studies, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Zhenhua Luo
- The Liver Care Center and Divisions of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Syn Yeo
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Gregory Bick
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Mingang Hao
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Chunmiao Cai
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Mahmoud Charif
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Jiang Wang
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Jun-Lin Guan
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Elyse E Lower
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Xiaoting Zhang
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; Graduate Program in Cancer and Cell Biology, Vontz Center for Molecular Studies, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; University of Cincinnati Cancer Center, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA.
| |
Collapse
|
23
|
Moody L, Chen H, Pan YX. Considerations for feature selection using gene pairs and applications in large-scale dataset integration, novel oncogene discovery, and interpretable cancer screening. BMC Med Genomics 2020; 13:148. [PMID: 33087122 PMCID: PMC7579924 DOI: 10.1186/s12920-020-00778-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Background Advancements in transcriptomic profiling have led to the emergence of new challenges regarding data integration and interpretability. Variability between measurement platforms makes it difficult to compare between cohorts, and large numbers of gene features have encouraged the use black box methods that are not easily translated into biologically and clinically meaningful findings. We propose that gene rankings and algorithms that rely on relative expression within gene pairs can address such obstacles. Methods We implemented an innovative process to evaluate the performance of five feature selection methods on simulated gene-pair data. Along with TSP, we consider other methods that retain more information in their score calculations, including the magnitude of gene expression change as well as within-class variation. Tree-based rule extraction was also applied to serum microRNA (miRNA) pairs in order to devise a noninvasive screening tool for pancreatic and ovarian cancer. Results Gene pair data were simulated using different types of signal and noise. Pairs were filtered using feature selection approaches, including top-scoring pairs (TSP), absolute differences between gene ranks, and Fisher scores. Methods that retain more information, such as the magnitude of expression change and within-class variance, yielded higher classification accuracy using a random forest model. We then demonstrate two powerful applications of gene pairs by first performing large-scale integration of 52 breast cancer datasets consisting of 10,350 patients. Not only did we confirm known oncogenes, but we also propose novel tumorigenic genes, such as BSDC1 and U2AF1, that could distinguish between tumor subtypes. Finally, circulating miRNA pairs were filtered and salient rules were extracted to build simplified tree ensemble learners (STELs) for four types of cancer. These accessible clinical frameworks detected pancreatic and ovarian cancer with 84.8 and 93.6% accuracy, respectively. Conclusion Rank-based gene pair classification benefits from careful feature selection methods that preserve maximal information. Gene pairs enable dataset integration for greater statistical power and discovery of robust biomarkers as well as facilitate construction of user-friendly clinical screening tools.
Collapse
Affiliation(s)
- Laura Moody
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, 461 Bevier Hall, 905 South Goodwin Avenue, Urbana, IL, 61801, USA
| | - Hong Chen
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, 461 Bevier Hall, 905 South Goodwin Avenue, Urbana, IL, 61801, USA.,Department of Food Science and Human Nutrition, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Yuan-Xiang Pan
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, 461 Bevier Hall, 905 South Goodwin Avenue, Urbana, IL, 61801, USA. .,Department of Food Science and Human Nutrition, University of Illinois Urbana-Champaign, Urbana, IL, USA. .,Illinois Informatics Institute, University of Illinois Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
24
|
Yuan F, Hung RJ, Walsh N, Zhang H, Platz EA, Wheeler W, Song L, Arslan AA, Beane Freeman LE, Bracci P, Canzian F, Du M, Gallinger S, Giles GG, Goodman PJ, Kooperberg C, Le Marchand L, Neale RE, Rosendahl J, Scelo G, Shu XO, Visvanathan K, White E, Zheng W, Albanes D, Amiano P, Andreotti G, Babic A, Bamlet WR, Berndt SI, Brennan P, Bueno-de-Mesquita B, Buring JE, Campbell PT, Chanock SJ, Fuchs CS, Gaziano JM, Goggins MG, Hackert T, Hartge P, Hassan MM, Holly EA, Hoover RN, Katzke V, Kirsten H, Kurtz RC, Lee IM, Malats N, Milne RL, Murphy N, Ng K, Oberg AL, Porta M, Rabe KG, Real FX, Rothman N, Sesso HD, Silverman DT, Thompson IM, Wactawski-Wende J, Wang X, Wentzensen N, Wilkens LR, Yu H, Zeleniuch-Jacquotte A, Shi J, Duell EJ, Amundadottir LT, Li D, Petersen GM, Wolpin BM, Risch HA, Yu K, Klein AP, Stolzenberg-Solomon R. Genome-Wide Association Study Data Reveal Genetic Susceptibility to Chronic Inflammatory Intestinal Diseases and Pancreatic Ductal Adenocarcinoma Risk. Cancer Res 2020; 80:4004-4013. [PMID: 32641412 PMCID: PMC7861352 DOI: 10.1158/0008-5472.can-20-0447] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 04/27/2020] [Accepted: 07/02/2020] [Indexed: 12/20/2022]
Abstract
Registry-based epidemiologic studies suggest associations between chronic inflammatory intestinal diseases and pancreatic ductal adenocarcinoma (PDAC). As genetic susceptibility contributes to a large proportion of chronic inflammatory intestinal diseases, we hypothesize that the genomic regions surrounding established genome-wide associated variants for these chronic inflammatory diseases are associated with PDAC. We examined the association between PDAC and genomic regions (±500 kb) surrounding established common susceptibility variants for ulcerative colitis, Crohn's disease, inflammatory bowel disease, celiac disease, chronic pancreatitis, and primary sclerosing cholangitis. We analyzed summary statistics from genome-wide association studies data for 8,384 cases and 11,955 controls of European descent from two large consortium studies using the summary data-based adaptive rank truncated product method to examine the overall association of combined genomic regions for each inflammatory disease group. Combined genomic susceptibility regions for ulcerative colitis, Crohn disease, inflammatory bowel disease, and chronic pancreatitis were associated with PDAC at P values < 0.05 (0.0040, 0.0057, 0.011, and 3.4 × 10-6, respectively). After excluding the 20 PDAC susceptibility regions (±500 kb) previously identified by GWAS, the genomic regions for ulcerative colitis, Crohn disease, and inflammatory bowel disease remained associated with PDAC (P = 0.0029, 0.0057, and 0.0098, respectively). Genomic regions for celiac disease (P = 0.22) and primary sclerosing cholangitis (P = 0.078) were not associated with PDAC. Our results support the hypothesis that genomic regions surrounding variants associated with inflammatory intestinal diseases, particularly, ulcerative colitis, Crohn disease, inflammatory bowel disease, and chronic pancreatitis are associated with PDAC. SIGNIFICANCE: The joint effects of common variants in genomic regions containing susceptibility loci for inflammatory bowel disease and chronic pancreatitis are associated with PDAC and may provide insights to understanding pancreatic cancer etiology.
Collapse
Affiliation(s)
- Fangcheng Yuan
- Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Rayjean J Hung
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System and University of Toronto, Toronto, Ontario, Canada
| | - Naomi Walsh
- National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin, Ireland
| | - Han Zhang
- Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland
| | - Elizabeth A Platz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - William Wheeler
- Information Management Services, Inc., Silver Spring, Maryland
| | - Lei Song
- Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland
| | - Alan A Arslan
- Department of Obstetrics and Gynecology, New York University School of Medicine, New York, New York, USA
- Department of Population Health, New York University School of Medicine, New York, New York
- Department of Environmental Medicine, New York University School of Medicine, New York, New York
- Perlmutter Cancer Center, New York University School of Medicine, New York, New York
| | | | - Paige Bracci
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California
| | - Federico Canzian
- Genomic Epidemiology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mengmeng Du
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Steven Gallinger
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System and University of Toronto, Toronto, Ontario, Canada
| | - Graham G Giles
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Victoria, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Victoria, Australia
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Victoria, Australia
| | - Phyllis J Goodman
- SWOG Statistical Center, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Loic Le Marchand
- Cancer Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - Rachel E Neale
- Department of Population Health, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Jonas Rosendahl
- Department of Internal Medicine I, Martin Luther University, Halle, Germany
| | | | - Xiao-Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Kala Visvanathan
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Emily White
- Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Demetrius Albanes
- Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland
| | - Pilar Amiano
- Public Health Division of Gipuzkoa, Ministry of Health of the Basque Government, Donostia-San Sebastian, Spain
- Biodonostia Health Research Institute, Donostia-San Sebastian, Spain
- CIBER Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | | | - Ana Babic
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - William R Bamlet
- Department of Health Sciences Research, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Sonja I Berndt
- Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland
| | - Paul Brennan
- International Agency for Research on Cancer, Lyon, France
| | - Bas Bueno-de-Mesquita
- Department for Determinants of Chronic Diseases (DCD), National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
- Department of Gastroenterology and Hepatology, University Medical Centre, Utrecht, the Netherlands
- Department of Epidemiology and Biostatistics, The School of Public Health, Imperial College London, London, UK
- Department of Social and Preventive Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Julie E Buring
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Division of Preventive Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Peter T Campbell
- Behavioral and Epidemiology Research Group, American Cancer Society, Atlanta, Georgia
| | - Stephen J Chanock
- Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland
| | - Charles S Fuchs
- Yale Cancer Center, New Haven, Connecticut
- Department of Medicine, Yale School of Medicine, New Haven, Connecticut
- Smilow Cancer Hospital, New Haven, Connecticut
| | - J Michael Gaziano
- Division of Preventive Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
- Boston Veteran Affairs Healthcare System, Boston, Massachusetts
| | - Michael G Goggins
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Thilo Hackert
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Patricia Hartge
- Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland
| | - Manal M Hassan
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Elizabeth A Holly
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California
| | - Robert N Hoover
- Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland
| | - Verena Katzke
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Holger Kirsten
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
- LIFE-Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany
| | - Robert C Kurtz
- Gastroenterology, Hepatology, and Nutrition Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - I-Min Lee
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Division of Preventive Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Nuria Malats
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre, Madrid, Spain
| | - Roger L Milne
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Victoria, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Victoria, Australia
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Victoria, Australia
| | - Neil Murphy
- Section of Nutrition and Metabolism, International Agency for Research on Cancer, Lyon, France
| | - Kimmie Ng
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ann L Oberg
- Department of Health Sciences Research, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Miquel Porta
- CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
- Hospital del Mar Institute of Medical Research (IMIM), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Kari G Rabe
- Department of Health Sciences Research, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Francisco X Real
- CIBERONC, Madrid, Spain
- Epithelial Carcinogenesis Group, Spanish National Cancer Research Centre, Madrid, Spain
- Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Spain
| | - Nathaniel Rothman
- Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland
| | - Howard D Sesso
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Division of Preventive Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Debra T Silverman
- Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland
| | - Ian M Thompson
- CHRISTUS Santa Rosa Hospital - Medical Center, San Antonio, Texas
| | - Jean Wactawski-Wende
- Department of Epidemiology and Environmental Health, University at Buffalo, Buffalo, New York
| | - Xiaoliang Wang
- Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Nicolas Wentzensen
- Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland
| | - Lynne R Wilkens
- Cancer Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - Herbert Yu
- Cancer Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - Anne Zeleniuch-Jacquotte
- Department of Population Health, New York University School of Medicine, New York, New York
- Department of Environmental Medicine, New York University School of Medicine, New York, New York
- Perlmutter Cancer Center, New York University School of Medicine, New York, New York
| | - Jianxin Shi
- Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland
| | - Eric J Duell
- Unit of Nutrition and Cancer, Cancer Epidemiology Research Program, Bellvitge Biomedical Research Institute (IDIBELL), Catalan Institute of Oncology (ICO), Barcelona, Spain
| | | | - Donghui Li
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gloria M Petersen
- Department of Health Sciences Research, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Brian M Wolpin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Harvey A Risch
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, Connecticut
| | - Kai Yu
- Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland
| | - Alison P Klein
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | | |
Collapse
|
25
|
Yang SR, Bouhlal Y, De La Vega FM, Ballard M, Kuo CJ, Vilborg A, Jensen G, Allison K. Integrated genomic characterization of ERBB2/HER2 alterations in invasive breast carcinoma: a focus on unusual FISH groups. Mod Pathol 2020; 33:1546-1556. [PMID: 32161378 DOI: 10.1038/s41379-020-0504-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 01/11/2020] [Accepted: 01/30/2020] [Indexed: 01/29/2023]
Abstract
In patients with invasive breast cancer, fluorescence in situ hybridization (FISH) testing for HER2 typically demonstrates the clear presence or lack of ERBB2 (HER2) amplification (i.e., groups 1 or 5). However, a small subset of patients can present with unusual HER2 FISH patterns (groups 2-4), resulting in diagnostic confusion. To provide clarity, the 2018 CAP/ASCO HER2 testing guideline recommends additional testing using HER2 immunohistochemistry (IHC) for determining the final HER2 status. Despite this effort, the genomic correlates of unusual HER2 FISH groups remain poorly understood. Here, we used droplet digital PCR (ddPCR) and targeted next-generation sequencing (NGS) to characterize the genomic features of both usual and unusual HER2 FISH groups. In this study, 51 clinical samples were selected to represent FISH groups 1-5. Furthermore, group 1 was subdivided into two groups, with groups 1A and 1B corresponding to cases with HER2 signals/cell ≥6.0 and 4-6, respectively. Overall, our findings revealed a wide range of copy number alterations in HER2 across the different FISH groups. As expected, groups 1A and 5 showed the clear presence and lack of HER2 copy number gain, respectively, as measured by ddPCR and NGS. In contrast, group 1B and other uncommon FISH groups (groups 2-4) were characterized by a broader range of HER2 copy levels with only a few select cases showing high-level gain. Notably, these cases with increased HER2 copy levels also showed HER2 overexpression by IHC, thus highlighting the correlation between HER2 copy number and HER2 protein expression. Given the concordance between the genomic and protein results, our findings suggest that HER2 IHC may inform HER2 copy number status in patients with unusual FISH patterns. Hence, our results support the current recommendation for using IHC to resolve HER2 status in FISH groups 2-4.
Collapse
Affiliation(s)
- Soo-Ryum Yang
- Department of Pathology, School of Medicine, Stanford University, Stanford, CA, USA
| | | | - Francisco M De La Vega
- TOMA Biosciences, Holland, MI, USA.,Department of Biomedical Data Science, School of Medicine, Stanford University, Stanford, CA, USA
| | - Morgan Ballard
- Department of Pathology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Calvin J Kuo
- Department of Medicine, School of Medicine, Stanford University, Stanford, CA, USA
| | | | | | - Kimberly Allison
- Department of Pathology, School of Medicine, Stanford University, Stanford, CA, USA.
| |
Collapse
|
26
|
Lee JY, Joo HS, Choi HJ, Jin S, Kim HY, Jeong GY, An HW, Park MK, Lee SE, Kim WS, Son T, Min KW, Oh YH, Kong G. Role of MEL-18 Amplification in Anti-HER2 Therapy of Breast Cancer. J Natl Cancer Inst 2020; 111:609-619. [PMID: 30265336 DOI: 10.1093/jnci/djy151] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 06/23/2018] [Accepted: 08/03/2018] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Resistance to HER2-targeted therapy with trastuzumab still remains a major challenge in HER2-amplified tumors. Here we investigated the potential role of MEL-18, a polycomb group gene, as a novel prognostic marker for trastuzumab resistance in HER2-positive (HER2+) breast cancer. METHODS The genetic alteration of MEL-18 and its clinical relevance were examined in multiple breast cancer cohorts including METABRIC (n = 1,980), TCGA (n = 825), and our clinical specimens (n = 213, trastuzumab-treated HER2+ cases). MEL-18 amplification was validated by fluorescence in situ hybridization (FISH) analysis. The MEL-18 effect on trastuzumab response was confirmed by in vitro cell viability assays and an in vivo xenograft experiment (n = 7 per group). Gene expression microarray and receptor tyrosine kinase array were performed to identify the trastuzumab resistance mechanism by MEL-18 loss. All statistical tests were two-sided. RESULTS MEL-18 was exclusively amplified in approximately 30-50% of HER2+ breast tumors and was associated with a favorable clinical outcome (disease-free survival: P = .02 in HER2+ cases, METABRIC; P = .04 in patients receiving trastuzumab). In MEL-18-amplified HER2+ breast cancer, MEL-18 depletion induced trastuzumab resistance by increasing ADAM sheddase-mediated ErbB ligand production and receptor heterodimerization. MEL-18 epigenetically silenced ADAM10/17 expression in cooperation with polycomb-repressive complex (PRC) 1 and PRC2. Combination treatment with an ADAM10/17 inhibitor and trastuzumab could overcome MEL-18 loss-mediated trastuzumab resistance in vivo (BT474/shMEL-18 xenograft: trastuzumab, mean [SD] tumor volume = 406.1 [50.1] mm3, vs trastuzumab + GW280264 30 mg/kg, mean [SD] tumor volume = 68.4 [15.6] mm3, P < .001). Consistently, trastuzumab-treated patients harboring concomitant MEL-18 amplification and low ADAM17 expression showed prolonged relapse-free survival (P = .02 in our cohort, n = 213). CONCLUSION MEL-18 serves to prevent ligand-dependent ErbB heterodimerization and trastuzumab resistance, suggesting MEL-18 amplification as a novel biomarker for HER2+ breast cancer.
Collapse
Affiliation(s)
| | | | | | - Sora Jin
- Department of Pathology, College of Medicine
| | | | | | - Hee Woon An
- Department of Pathology, College of Medicine
| | - Mi Kyung Park
- Hanyang University, Seoul, Republic of Korea; National Cancer Center, Goyang, Republic of Korea
| | - Seung Eun Lee
- Department of Pathology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul, Republic of Korea
| | - Wan-Seop Kim
- Department of Pathology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul, Republic of Korea
| | - Taekwon Son
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | | | - Young-Ha Oh
- Department of Pathology, College of Medicine
| | - Gu Kong
- Department of Pathology, College of Medicine
| |
Collapse
|
27
|
Choi H, Jin S, Cho H, Won H, An HW, Jeong G, Park Y, Kim H, Park MK, Son T, Min K, Jang K, Oh Y, Lee J, Kong G. CDK12 drives breast tumor initiation and trastuzumab resistance via WNT and IRS1-ErbB-PI3K signaling. EMBO Rep 2019; 20:e48058. [PMID: 31468695 PMCID: PMC6776914 DOI: 10.15252/embr.201948058] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 08/06/2019] [Accepted: 08/09/2019] [Indexed: 12/17/2022] Open
Abstract
Cyclin-dependent kinase 12 (CDK12) has emerged as an effective therapeutic target due to its ability to regulate DNA damage repair in human cancers, but little is known about the role of CDK12 in driving tumorigenesis. Here, we demonstrate that CDK12 promotes tumor initiation as a novel regulator of cancer stem cells (CSCs) and induces anti-HER2 therapy resistance in human breast cancer. High CDK12 expression caused by concurrent amplification of CDK12 and HER2 in breast cancer patients is associated with disease recurrence and poor survival. CDK12 induces self-renewal of breast CSCs and in vivo tumor-initiating ability, and also reduces susceptibility to trastuzumab. Furthermore, CDK12 kinase activity inhibition facilitates anticancer efficacy of trastuzumab in HER2+ tumors, and mice bearing trastuzumab-resistant HER2+ tumor show sensitivity to an inhibitor of CDK12. Mechanistically, the catalytic activity of CDK12 is required for the expression of genes involved in the activation of ErbB-PI3K-AKT or WNT-signaling cascades. These results suggest that CDK12 is a major oncogenic driver and an actionable target for HER2+ breast cancer to replace or augment current anti-HER2 therapies.
Collapse
Affiliation(s)
- Hee‐Joo Choi
- Institute for Bioengineering and Biopharmaceutical Research (IBBR)Hanyang UniversitySeoulKorea
- Department of PathologyCollege of MedicineHanyang UniversitySeoulKorea
| | - Sora Jin
- Department of PathologyCollege of MedicineHanyang UniversitySeoulKorea
| | - Hani Cho
- Department of PathologyCollege of MedicineHanyang UniversitySeoulKorea
| | - Hee‐Young Won
- Department of PathologyCollege of MedicineHanyang UniversitySeoulKorea
| | - Hee Woon An
- Department of PathologyCollege of MedicineHanyang UniversitySeoulKorea
| | - Ga‐Young Jeong
- Department of PathologyCollege of MedicineHanyang UniversitySeoulKorea
| | - Young‐Un Park
- Department of PathologyCollege of MedicineHanyang UniversitySeoulKorea
| | - Hyung‐Yong Kim
- Department of PathologyCollege of MedicineHanyang UniversitySeoulKorea
| | | | - Taekwon Son
- College of PharmacySeoul National UniversitySeoulKorea
| | - Kyueng‐Whan Min
- Department of PathologyCollege of MedicineHanyang UniversitySeoulKorea
| | - Ki‐Seok Jang
- Department of PathologyCollege of MedicineHanyang UniversitySeoulKorea
| | - Young‐Ha Oh
- Department of PathologyCollege of MedicineHanyang UniversitySeoulKorea
| | - Jeong‐Yeon Lee
- Department of MedicineCollege of MedicineHanyang UniversitySeoulKorea
| | - Gu Kong
- Institute for Bioengineering and Biopharmaceutical Research (IBBR)Hanyang UniversitySeoulKorea
- Department of PathologyCollege of MedicineHanyang UniversitySeoulKorea
| |
Collapse
|
28
|
Amirfallah A, Arason A, Einarsson H, Gudmundsdottir ET, Freysteinsdottir ES, Olafsdottir KA, Johannsson OT, Agnarsson BA, Barkardottir RB, Reynisdottir I. High expression of the vacuole membrane protein 1 (VMP1) is a potential marker of poor prognosis in HER2 positive breast cancer. PLoS One 2019; 14:e0221413. [PMID: 31442252 PMCID: PMC6707546 DOI: 10.1371/journal.pone.0221413] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 08/06/2019] [Indexed: 02/07/2023] Open
Abstract
Background Fusion genes result from genomic structural changes, which can lead to alterations in gene expression that supports tumor development. The aim of the study was to use fusion genes as a tool to identify new breast cancer (BC) genes with a role in BC progression. Methods Fusion genes from breast tumors and BC cell lines were collected from publications. RNA-Seq data from tumors and cell lines were retrieved from databanks and analyzed for fusions with SOAPfuse or the analysis was purchased. Fusion genes identified in both tumors (n = 1724) and cell lines (n = 45) were confirmed by qRT-PCR and sequencing. Their individual genes were ranked by selection criteria that included correlation of their mRNA level with copy number. The expression of the top ranked gene was measured by qRT-PCR in normal tissue and in breast tumors from an exploratory cohort (n = 141) and a validation cohort (n = 277). Expression levels were correlated with clinical and pathological factors as well as the patients’ survival. The results were followed up in BC cohorts from TCGA (n = 818) and METABRIC (n = 2509). Results Vacuole membrane protein 1 (VMP1) was the most promising candidate based on specific selection criteria. Its expression was higher in breast tumor tissue than normal tissue (p = 1x10-4), and its expression was significantly higher in HER2 positive than HER2 negative breast tumors in all four cohorts analyzed. High expression of VMP1 associated with breast cancer specific survival (BCSS) in cohort 1 (hazard ratio (HR) = 2.31, CI 1.27–4.18) and METABRIC (HR = 1.26, CI 1.02–1.57), and also after adjusting for HER2 expression in cohort 1 (HR = 2.03, CI 1.10–3.72). BCSS was not significant in cohort 2 or TCGA cohort, which may be due to differences in treatment regimens. Conclusions The results suggest that high VMP1 expression is a potential marker of poor prognosis in HER2 positive BC. Further studies are needed to elucidate how VMP1 could affect pathways supportive of tumorigenesis.
Collapse
Affiliation(s)
- Arsalan Amirfallah
- Cell Biology Unit at the Pathology Department, Landspitali–The National University Hospital of Iceland, Reykjavik, Iceland
- The Biomedical Center, University of Iceland, Reykjavik, Iceland
| | - Adalgeir Arason
- The Biomedical Center, University of Iceland, Reykjavik, Iceland
- Molecular Pathology Unit at the Pathology Department, Landspitali–The National University Hospital of Iceland, Reykjavik, Iceland
| | - Hjorleifur Einarsson
- Cell Biology Unit at the Pathology Department, Landspitali–The National University Hospital of Iceland, Reykjavik, Iceland
| | - Eydis Thorunn Gudmundsdottir
- Cell Biology Unit at the Pathology Department, Landspitali–The National University Hospital of Iceland, Reykjavik, Iceland
| | - Edda Sigridur Freysteinsdottir
- Molecular Pathology Unit at the Pathology Department, Landspitali–The National University Hospital of Iceland, Reykjavik, Iceland
| | | | - Oskar Thor Johannsson
- Department of Oncology, Landspitali–The National University Hospital of Iceland, Reykjavik, Iceland
| | - Bjarni Agnar Agnarsson
- Pathology Department, Landspitali–The National University Hospital of Iceland, Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Rosa Bjork Barkardottir
- The Biomedical Center, University of Iceland, Reykjavik, Iceland
- Molecular Pathology Unit at the Pathology Department, Landspitali–The National University Hospital of Iceland, Reykjavik, Iceland
| | - Inga Reynisdottir
- Cell Biology Unit at the Pathology Department, Landspitali–The National University Hospital of Iceland, Reykjavik, Iceland
- The Biomedical Center, University of Iceland, Reykjavik, Iceland
- * E-mail:
| |
Collapse
|
29
|
Walsh N, Zhang H, Hyland PL, Yang Q, Mocci E, Zhang M, Childs EJ, Collins I, Wang Z, Arslan AA, Beane-Freeman L, Bracci PM, Brennan P, Canzian F, Duell EJ, Gallinger S, Giles GG, Goggins M, Goodman GE, Goodman PJ, Hung RJ, Kooperberg C, Kurtz RC, Malats N, LeMarchand L, Neale RE, Olson SH, Scelo G, Shu XO, Van Den Eeden SK, Visvanathan K, White E, Zheng W, Albanes D, Andreotti G, Babic A, Bamlet WR, Berndt SI, Borgida A, Boutron-Ruault MC, Brais L, Brennan P, Bueno-de-Mesquita B, Buring J, Chaffee KG, Chanock S, Cleary S, Cotterchio M, Foretova L, Fuchs C, M Gaziano JM, Giovannucci E, Goggins M, Hackert T, Haiman C, Hartge P, Hasan M, Helzlsouer KJ, Herman J, Holcatova I, Holly EA, Hoover R, Hung RJ, Janout V, Klein EA, Kurtz RC, Laheru D, Lee IM, Lu L, Malats N, Mannisto S, Milne RL, Oberg AL, Orlow I, Patel AV, Peters U, Porta M, Real FX, Rothman N, Sesso HD, Severi G, Silverman D, Strobel O, Sund M, Thornquist MD, Tobias GS, Wactawski-Wende J, Wareham N, Weiderpass E, Wentzensen N, Wheeler W, Yu H, Zeleniuch-Jacquotte A, Kraft P, Li D, Jacobs EJ, Petersen GM, Wolpin BM, Risch HA, Amundadottir LT, Yu K, Klein AP, Stolzenberg-Solomon RZ. Agnostic Pathway/Gene Set Analysis of Genome-Wide Association Data Identifies Associations for Pancreatic Cancer. J Natl Cancer Inst 2019; 111:557-567. [PMID: 30541042 PMCID: PMC6579744 DOI: 10.1093/jnci/djy155] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 06/15/2018] [Accepted: 08/08/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Genome-wide association studies (GWAS) identify associations of individual single-nucleotide polymorphisms (SNPs) with cancer risk but usually only explain a fraction of the inherited variability. Pathway analysis of genetic variants is a powerful tool to identify networks of susceptibility genes. METHODS We conducted a large agnostic pathway-based meta-analysis of GWAS data using the summary-based adaptive rank truncated product method to identify gene sets and pathways associated with pancreatic ductal adenocarcinoma (PDAC) in 9040 cases and 12 496 controls. We performed expression quantitative trait loci (eQTL) analysis and functional annotation of the top SNPs in genes contributing to the top associated pathways and gene sets. All statistical tests were two-sided. RESULTS We identified 14 pathways and gene sets associated with PDAC at a false discovery rate of less than 0.05. After Bonferroni correction (P ≤ 1.3 × 10-5), the strongest associations were detected in five pathways and gene sets, including maturity-onset diabetes of the young, regulation of beta-cell development, role of epidermal growth factor (EGF) receptor transactivation by G protein-coupled receptors in cardiac hypertrophy pathways, and the Nikolsky breast cancer chr17q11-q21 amplicon and Pujana ATM Pearson correlation coefficient (PCC) network gene sets. We identified and validated rs876493 and three correlating SNPs (PGAP3) and rs3124737 (CASP7) from the Pujana ATM PCC gene set as eQTLs in two normal derived pancreas tissue datasets. CONCLUSION Our agnostic pathway and gene set analysis integrated with functional annotation and eQTL analysis provides insight into genes and pathways that may be biologically relevant for risk of PDAC, including those not previously identified.
Collapse
Affiliation(s)
- Naomi Walsh
- National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin, Ireland
| | - Han Zhang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Paula L Hyland
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD
- Division of Applied Regulatory Science, Office of Translational Science, Center for Drug Evaluation & Research, U.S. Food and Drug Administration, Silver Spring, MD
| | - Qi Yang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Evelina Mocci
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD
| | - Mingfeng Zhang
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD
- Division of Epidemiology II, Office of Surveillance and Epidemiology, Center for Drug Evaluation & Research, U.S. Food and Drug Administration, Silver Spring, MD
| | - Erica J Childs
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD
| | - Irene Collins
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Zhaoming Wang
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD
- Department of Computational Biology, St Jude Children’s Research Hospital, Memphis, Tennessee
| | - Alan A Arslan
- Department of Obstetrics and Gynecology, New York University School of Medicine, New York, NY
- Department of Environmental Medicine, New York University School of Medicine, New York, NY
- Department of Population Health, New York University School of Medicine, New York, NY
| | - Laura Beane-Freeman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Paige M Bracci
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA
| | - Paul Brennan
- International Agency for Research on Cancer (IARC), Lyon, France
| | - Federico Canzian
- Genomic Epidemiology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Eric J Duell
- Unit of Nutrition and Cancer, Cancer Epidemiology Research Program, Bellvitge Biomedical Research Institute (IDIBELL), Catalan Institute of Oncology (ICO), Barcelona, Spain
| | - Steven Gallinger
- Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Graham G Giles
- Cancer Epidemiology and Intelligence Division, Cancer Council Victoria, Melbourne, Victoria, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Victoria, Australia
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Michael Goggins
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins School of Medicine, Baltimore, MD
| | - Gary E Goodman
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Phyllis J Goodman
- SWOG Statistical Center, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Rayjean J Hung
- Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Robert C Kurtz
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Núria Malats
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center (CNIO), Madrid, Spain
- CIBERONC, Madrid, Spain
| | - Loic LeMarchand
- Cancer Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI
| | - Rachel E Neale
- Population Health Department, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Sara H Olson
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ghislaine Scelo
- International Agency for Research on Cancer (IARC), Lyon, France
| | - Xiao O Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN
| | | | - Kala Visvanathan
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Emily White
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Epidemiology, University of Washington, Seattle, WA
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN
| | | | - Demetrius Albanes
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Gabriella Andreotti
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Ana Babic
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - William R Bamlet
- Department of Health Sciences Research, Mayo Clinic College of Medicine, Rochester, MN
| | - Sonja I Berndt
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Ayelet Borgida
- Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Marie-Christine Boutron-Ruault
- Centre de Recherche en Épidémiologie et Santé des Populations (CESP, Inserm U1018), Facultés de Medicine, Université Paris-Saclay, UPS, UVSQ, Gustave Roussy, Villejuif, France
| | - Lauren Brais
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Paul Brennan
- International Agency for Research on Cancer (IARC), Lyon, France
| | - Bas Bueno-de-Mesquita
- Department for Determinants of Chronic Diseases (DCD), National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- Department of Social & Preventive Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Julie Buring
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Kari G Chaffee
- Department of Health Sciences Research, Mayo Clinic College of Medicine, Rochester, MN
| | - Stephen Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Sean Cleary
- Division of Hepatobiliary and Pancreas Surgery, Mayo Clinic, Rochester, MN
| | - Michelle Cotterchio
- Cancer Care Ontario, University of Toronto, Toronto, ON, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Lenka Foretova
- Department of Cancer Epidemiology and Genetics, Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | | | - J Michael M Gaziano
- Division of Aging, Brigham and Women's Hospital, Boston, MA
- Boston VA Healthcare System, Boston, MA
| | - Edward Giovannucci
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Michael Goggins
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins School of Medicine, Baltimore, MD
| | - Thilo Hackert
- Department of General Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Christopher Haiman
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Patricia Hartge
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Manal Hasan
- Department of Epidemiology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Kathy J Helzlsouer
- Division of Cancer Control and Population Sciences, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Joseph Herman
- Department of Radiation Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD
| | - Ivana Holcatova
- Institute of Public Health and Preventive Medicine, Charles University, 2nd Faculty of Medicine, Prague, Czech Republic
| | - Elizabeth A Holly
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA
| | - Robert Hoover
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Rayjean J Hung
- Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Vladimir Janout
- Department of Epidemiology and Public Health, Faculty of Medicine, University of Ostrava, Czech Republic
- Faculty of Medicine, University of Olomouc, Olomouc, Czech Republic
| | - Eric A Klein
- Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH
| | - Robert C Kurtz
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Daniel Laheru
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD
| | - I-Min Lee
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Lingeng Lu
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, CT
| | - Núria Malats
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center (CNIO), Madrid, Spain
- CIBERONC, Madrid, Spain
| | - Satu Mannisto
- Department of Public Health Solutions, National Institute for Health and Welfare, Helsinki, Finland
| | - Roger L Milne
- Cancer Epidemiology and Intelligence Division, Cancer Council Victoria, Melbourne, Victoria, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Victoria, Australia
| | - Ann L Oberg
- Department of Health Sciences Research, Mayo Clinic College of Medicine, Rochester, MN
| | - Irene Orlow
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Alpa V Patel
- Epidemiology Research Program, American Cancer Society, Atlanta, GA
| | - Ulrike Peters
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Miquel Porta
- CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
- Hospital del Mar Institute of Medical Research (IMIM), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Francisco X Real
- CIBERONC, Madrid, Spain
- Epithelial Carcinogenesis Group, Spanish National Cancer Research Centre-CNIO, Madrid, Spain
- Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Spain
| | - Nathaniel Rothman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Howard D Sesso
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Gianluca Severi
- Cancer Epidemiology and Intelligence Division, Cancer Council Victoria, Melbourne, Victoria, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Victoria, Australia
- Centre de Recherche en Épidémiologie et Santé des Populations (CESP, Inserm U1018), Facultés de Medicine, Université Paris-Saclay, UPS, UVSQ, Gustave Roussy, Villejuif, France
| | - Debra Silverman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Oliver Strobel
- Department of General Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Malin Sund
- Department of Surgical and Perioperative Sciences, Umeå University, Umeå, Sweden
| | - Mark D Thornquist
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Geoffrey S Tobias
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Jean Wactawski-Wende
- Department of Epidemiology and Environmental Health, University at Buffalo, Buffalo, NY
| | - Nick Wareham
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Elisabete Weiderpass
- Department of Research, Cancer Registry of Norway, Institute of Population-Based Cancer Research, Oslo, Norway
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Genetic Epidemiology Group, Folkhälsan Research Center and Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Community Medicine, Faculty of Health Sciences, University of Tromsø, The Arctic University of Norway, Tromsø, Norway
| | - Nicolas Wentzensen
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | | | - Herbert Yu
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY
| | - Anne Zeleniuch-Jacquotte
- Department of Population Health, New York University School of Medicine, New York, NY
- Department of Biostatistics, Harvard School of Public Health, Boston, MA
| | - Peter Kraft
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Donghui Li
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Eric J Jacobs
- Epidemiology Research Program, American Cancer Society, Atlanta, GA
| | - Gloria M Petersen
- Department of Health Sciences Research, Mayo Clinic College of Medicine, Rochester, MN
| | - Brian M Wolpin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Harvey A Risch
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, CT
| | - Laufey T Amundadottir
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Kai Yu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Alison P Klein
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins School of Medicine, Baltimore, MD
| | | |
Collapse
|
30
|
Chen J, Wu F, Shi Y, Yang D, Xu M, Lai Y, Liu Y. Identification of key candidate genes involved in melanoma metastasis. Mol Med Rep 2019; 20:903-914. [PMID: 31173190 PMCID: PMC6625188 DOI: 10.3892/mmr.2019.10314] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 01/18/2019] [Indexed: 12/16/2022] Open
Abstract
Metastasis is the most lethal stage of cancer progression. The present study aimed to investigate the underlying molecular mechanisms of melanoma metastasis using bioinformatics. Using the microarray dataset GSE8401 from the Gene Expression Omnibus database, which included 52 biopsy specimens from patients with melanoma metastasis and 31 biopsy specimens from patients with primary melanoma, differentially expressed genes (DEGs) were identified, subsequent to data preprocessing with the affy package, followed by Gene Ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses. A protein-protein interaction (PPI) network was constructed. Mutated genes were analyzed with 80 mutated cases with melanoma from The Cancer Genome Atlas. The overall survival of key candidate DEGs, which were within a filtering of degree >30 criteria in the PPI network and involved three or more KEGG signaling pathways, and genes with a high mutation frequency were delineated. The expression analysis of key candidate DEGs, mutant genes and their associated genes were performed on UALCAN. Of the 1,187 DEGs obtained, 505 were upregulated and 682 were downregulated. ‘Extracellular exosome’ processes, the ‘amoebiasis’ pathway, the ‘ECM-receptor interaction’ pathway and the ‘focal adhesion’ signaling pathway were significantly enriched and identified as important processes or signaling pathways. The overall survival analysis of phosphoinositide-3-kinase regulator subunit 3 (PIK3R3), centromere protein M (CENPM), aurora kinase A (AURKA), laminin subunit α 1 (LAMA1), proliferating cell nuclear antigen (PCNA), adenylate cyclase 1 (ADCY1), BUB1 mitotic checkpoint serine/threonine kinase (BUB1), NDC80 kinetochore complex component (NDC80) and protein kinase C α (PRKCA) in DEGs was statistically significant. Mutation gene analysis identified that BRCA1-associated protein 1 (BAP1) had a higher mutation frequency and survival analysis, and its associated genes in the BAP1-associated PPI network, including ASXL transcriptional regulator 1 (ASXL1), proteasome 26S subunit, non-ATPase 3 (PSMD3), proteasome 26S subunit, non ATPase 11 (PSMD11) and ubiquitin C (UBC), were statistically significantly associated with the overall survival of patients with melanoma. The expression levels of PRKCA, BUB1, BAP1 and ASXL1 were significantly different between primary melanoma and metastatic melanoma. Based on the present study, ‘extracellular exosome’ processes, ‘amoebiasis’ pathways, ‘ECM-receptor interaction’ pathways and ‘focal adhesion’ signaling pathways may be important in the formation of metastases from melanoma. The involved genes, including PIK3R3, CENPM, AURKA, LAMA1, PCNA, ADCY1, BUB1, NDC80 and PRKCA, and mutation associated genes, including BAP1, ASXL1, PSMD3, PSMD11 and UBC, may serve important roles in metastases of melanoma.
Collapse
Affiliation(s)
- Jia Chen
- Department of Dermatopathology, Tongji University Affiliated Shanghai Skin Disease Hospital, Shanghai 200443, P.R. China
| | - Fei Wu
- Department of Dermatopathology, Tongji University Affiliated Shanghai Skin Disease Hospital, Shanghai 200443, P.R. China
| | - Yu Shi
- Department of Medical Cosmetology, Tongji University Affiliated Shanghai Skin Disease Hospital, Shanghai 200443, P.R. China
| | - Degang Yang
- Department of Treatment, Tongji University Affiliated Shanghai Skin Disease Hospital, Shanghai 200443, P.R. China
| | - Mingyuan Xu
- Department of Dermatopathology, Tongji University Affiliated Shanghai Skin Disease Hospital, Shanghai 200443, P.R. China
| | - Yongxian Lai
- Department of Dermatologic Surgery, Tongji University Affiliated Shanghai Skin Disease Hospital, Shanghai 200443, P.R. China
| | - Yeqiang Liu
- Department of Dermatopathology, Tongji University Affiliated Shanghai Skin Disease Hospital, Shanghai 200443, P.R. China
| |
Collapse
|
31
|
Chu PY, Tai YL, Shen TL. Grb7, a Critical Mediator of EGFR/ErbB Signaling, in Cancer Development and as a Potential Therapeutic Target. Cells 2019; 8:cells8050435. [PMID: 31083325 PMCID: PMC6562560 DOI: 10.3390/cells8050435] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 05/02/2019] [Accepted: 05/09/2019] [Indexed: 12/20/2022] Open
Abstract
The partner of activated epidermal growth factor receptor (EGFR), growth factor receptor bound protein-7 (Grb7), a functionally multidomain adaptor protein, has been demonstrated to be a pivotal regulator for varied physiological and pathological processes by interacting with phospho-tyrosine-related signaling molecules to affect the transmission through a number of signaling pathways. In particular, critical roles of Grb7 in erythroblastic leukemia viral oncogene homolog (ERBB) family-mediated cancer development and malignancy have been intensively evaluated. The overexpression of Grb7 or the coamplification/cooverexpression of Grb7 and members of the ERBB family play essential roles in advanced human cancers and are associated with decreased survival and recurrence of cancers, emphasizing Grb7's value as a prognostic marker and a therapeutic target. Peptide inhibitors of Grb7 are being tested in preclinical trials for their possible therapeutic effects. Here, we review the molecular, functional, and clinical aspects of Grb7 in ERBB family-mediated cancer development and malignancy with the aim to reveal alternative and effective therapeutic strategies.
Collapse
Affiliation(s)
- Pei-Yu Chu
- Department of Plant Pathology and Microbiology, National Taiwan University, Taipei 10617, Taiwan.
| | - Yu-Ling Tai
- Department of Plant Pathology and Microbiology, National Taiwan University, Taipei 10617, Taiwan.
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Tang-Long Shen
- Department of Plant Pathology and Microbiology, National Taiwan University, Taipei 10617, Taiwan.
- Center for Biotechnology, National Taiwan University, Taipei 10617, Taiwan.
| |
Collapse
|
32
|
Molina-Crespo Á, Cadete A, Sarrio D, Gámez-Chiachio M, Martinez L, Chao K, Olivera A, Gonella A, Díaz E, Palacios J, Dhal PK, Besev M, Rodríguez-Serrano M, García Bermejo ML, Triviño JC, Cano A, García-Fuentes M, Herzberg O, Torres D, Alonso MJ, Moreno-Bueno G. Intracellular Delivery of an Antibody Targeting Gasdermin-B Reduces HER2 Breast Cancer Aggressiveness. Clin Cancer Res 2019; 25:4846-4858. [PMID: 31064780 DOI: 10.1158/1078-0432.ccr-18-2381] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 03/21/2019] [Accepted: 05/02/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Gasdermin B (GSDMB) overexpression/amplification occurs in about 60% of HER2 breast cancers, where it promotes cell migration, resistance to anti-HER2 therapies, and poor clinical outcome. Thus, we tackle GSDMB cytoplasmic overexpression as a new therapeutic target in HER2 breast cancers. EXPERIMENTAL DESIGN We have developed a new targeted nanomedicine based on hyaluronic acid-biocompatible nanocapsules, which allow the intracellular delivery of a specific anti-GSDMB antibody into HER2 breast cancer cells both in vitro and in vivo. RESULTS Using different models of HER2 breast cancer cells, we show that anti-GSDMB antibody loaded to nanocapsules has significant and specific effects on GSDMB-overexpressing cancer cells' behavior in ways such as (i) lowering the in vitro cell migration induced by GSDMB; (ii) enhancing the sensitivity to trastuzumab; (iii) reducing tumor growth by increasing apoptotic rate in orthotopic breast cancer xenografts; and (iv) diminishing lung metastasis in MDA-MB-231-HER2 cells in vivo. Moreover, at a mechanistic level, we have shown that AbGB increases GSDMB binding to sulfatides and consequently decreases migratory cell behavior and may upregulate the potential intrinsic procell death activity of GSDMB. CONCLUSIONS Our findings portray the first evidence of the effectiveness and specificity of an antibody-based nanomedicine that targets an intracellular oncoprotein. We have proved that intracellular-delivered anti-GSDMB reduces diverse protumor GSDMB functions (migration, metastasis, and resistance to therapy) in an efficient and specific way, thus providing a new targeted therapeutic strategy in aggressive HER2 cancers with poor prognosis.
Collapse
Affiliation(s)
- Ángela Molina-Crespo
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), IdiPaz, Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Monforte de Lemos, Madrid, Spain
| | - Ana Cadete
- Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CiMUS), Campus Vida, Universidad de Santiago de Compostela, Avenida de Barcelona s/n, Santiago de Compostela, Spain
- Departamento de Farmacia y Tecnología Farmacéutica, Campus Vida, Universidad de Santiago de Compostela, Avenida de Barcelona s/n, Santiago de Compostela, Spain
| | - David Sarrio
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), IdiPaz, Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Monforte de Lemos, Madrid, Spain
| | - Manuel Gámez-Chiachio
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), IdiPaz, Madrid, Spain
| | - Lidia Martinez
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), IdiPaz, Madrid, Spain
| | - Kinlin Chao
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, Maryland
| | - Ana Olivera
- Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CiMUS), Campus Vida, Universidad de Santiago de Compostela, Avenida de Barcelona s/n, Santiago de Compostela, Spain
- Departamento de Farmacia y Tecnología Farmacéutica, Campus Vida, Universidad de Santiago de Compostela, Avenida de Barcelona s/n, Santiago de Compostela, Spain
| | - Andrea Gonella
- Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CiMUS), Campus Vida, Universidad de Santiago de Compostela, Avenida de Barcelona s/n, Santiago de Compostela, Spain
- Departamento de Farmacia y Tecnología Farmacéutica, Campus Vida, Universidad de Santiago de Compostela, Avenida de Barcelona s/n, Santiago de Compostela, Spain
| | - Eva Díaz
- Fundación MD Anderson Internacional, C/Gómez Hemans, Madrid, Spain
| | - José Palacios
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Monforte de Lemos, Madrid, Spain
- Departamento de Patología, Hospital Ramón y Cajal, IRYCIS, Ctra De Colmenar, Madrid, Spain
| | | | | | | | | | | | - Amparo Cano
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), IdiPaz, Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Monforte de Lemos, Madrid, Spain
| | - Marcos García-Fuentes
- Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CiMUS), Campus Vida, Universidad de Santiago de Compostela, Avenida de Barcelona s/n, Santiago de Compostela, Spain
- Departamento de Farmacia y Tecnología Farmacéutica, Campus Vida, Universidad de Santiago de Compostela, Avenida de Barcelona s/n, Santiago de Compostela, Spain
| | - Osnat Herzberg
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, Maryland
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland
| | - Dolores Torres
- Departamento de Farmacia y Tecnología Farmacéutica, Campus Vida, Universidad de Santiago de Compostela, Avenida de Barcelona s/n, Santiago de Compostela, Spain
| | - Maria José Alonso
- Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CiMUS), Campus Vida, Universidad de Santiago de Compostela, Avenida de Barcelona s/n, Santiago de Compostela, Spain.
- Departamento de Farmacia y Tecnología Farmacéutica, Campus Vida, Universidad de Santiago de Compostela, Avenida de Barcelona s/n, Santiago de Compostela, Spain
| | - Gema Moreno-Bueno
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), IdiPaz, Madrid, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Monforte de Lemos, Madrid, Spain
- Fundación MD Anderson Internacional, C/Gómez Hemans, Madrid, Spain
| |
Collapse
|
33
|
Tang Y, Yang S, Wang M, Liu D, Liu Y, Zhang Y, Zhang Q. Epigenetically altered miR‑193a‑3p promotes HER2 positive breast cancer aggressiveness by targeting GRB7. Int J Mol Med 2019; 43:2352-2360. [PMID: 31017268 PMCID: PMC6488183 DOI: 10.3892/ijmm.2019.4167] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 03/12/2019] [Indexed: 12/19/2022] Open
Abstract
Emerging evidence has demonstrated that microRNAs (miRNAs/miRs) have various biological functions in the development of human epidermal growth factor receptor 2 (HER2) positive breast cancer. The aim of the present study is to reveal the mechanism of miR‑193a‑3p inhibiting the progress of HER2 positive breast cancer. The expression of miR‑193a‑3p was evaluated by quantitative polymerase chain reaction (PCR). The methylation status of miR‑193a‑3p was evaluated by PCR and pyrosequencing analysis. Overexpression of miR‑193a‑3p and growth factor receptor bound protein 7 (GRB7) combined with in vitro tumorigenic assays were conducted to determine the carcinostatic capacities of miR‑193a‑3p in HER2 positive breast cancer cells. The association between miR‑193a‑3p and GRB7 was determined by luciferase reporter assay. Protein level was evaluated using western blot analysis. miR‑193a‑3p was downregulated in HER2 positive breast cancer cells and clinical tissues. Methylation‑mediated silencing led to decreased expression of miR‑193a‑3p in HER2 positive breast cancer. Overexpression of miR‑193a‑3p could inhibit proliferation, migration and invasion of breast cancer cells. Overexpression of GRB7 could abolish this effect. miR‑193a‑3p could directly target the 3' untranslated region of GRB7. miR‑193a‑3p could directly or indirectly target extracellular signal‑regulated kinase 1/2 (ERK1/2) and forkhead box M1 (FOXM1) signaling. In conclusion, it was identified that silencing of miR‑193a‑3p through hypermethylation can promote HER2 positive breast cancer progress by targeting GRB7, ERK1/2 and FOXM1 signaling. The function of miR‑193a‑3p in HER2 positive breast cancer implicates its potential application in therapy.
Collapse
Affiliation(s)
- Yiyin Tang
- First Department of Mammary Surgery, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming, Yunnan 650118, P.R. China
| | - Siyuan Yang
- First Department of Mammary Surgery, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming, Yunnan 650118, P.R. China
| | - Maohua Wang
- First Department of Mammary Surgery, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming, Yunnan 650118, P.R. China
| | - Dequan Liu
- First Department of Mammary Surgery, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming, Yunnan 650118, P.R. China
| | - Yang Liu
- First Department of Mammary Surgery, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming, Yunnan 650118, P.R. China
| | - Ying Zhang
- First Department of Mammary Surgery, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming, Yunnan 650118, P.R. China
| | - Qian Zhang
- First Department of Mammary Surgery, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming, Yunnan 650118, P.R. China
| |
Collapse
|
34
|
Fararjeh AS, Chen LC, Ho YS, Cheng TC, Liu YR, Chang HL, Chang HW, Wu CH, Tu SH. Proteasome 26S Subunit, non-ATPase 3 (PSMD3) Regulates Breast Cancer by Stabilizing HER2 from Degradation. Cancers (Basel) 2019; 11:cancers11040527. [PMID: 31013812 PMCID: PMC6549480 DOI: 10.3390/cancers11040527] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 03/28/2019] [Accepted: 04/09/2019] [Indexed: 12/24/2022] Open
Abstract
It is well-known that human epidermal growth factor receptor 2 (HER2) is critical for breast cancer (BC) development and progression. Several studies have revealed the role of the ubiquitin/proteasome system (UPS) in cancer. In this study, we investigated the expression level of Proteasome 26S subunit, non-ATPase 3 (PSMD3) in BC using BC cell lines, human BC tissue samples, Oncomine, and TCGA databases and studied the PSMD3-HER2 protein interaction. PSMD3 was upregulated in BC, particularly in the HER2+ subtype. PSMD3 immunostaining was detected in the cytoplasm and nucleus of BC tumor tissues. Strong interaction between PSMD3 and HER2 at the protein level was observed. Knockdown of PSMD3 significantly impaired the stability of HER2, inhibited BC cell proliferation and colony formation, and induced cell apoptosis. Ubiquitination process was strongly enhanced after knockdown of PSMD3 in association with decreased HER2 level. Accumulation and Localization of LAMP-1 in the cell membrane with decreased HER2 immunostaining was observed after knockdown of PSMD3. High expression level of PSMD3 was associated with HER2 expression (p < 0.001), tumor size (p < 0.001), and clinical stage (p = 0.036). High expression level of PSMD3 predicted a short overall survival (OS), particularly for HER2+. Overall, we provide a novel function for PSMD3 in stabilizing HER2 from degradation in HER2+ BC, which suggests that PSMD3 is a novel target for HER2+ BC.
Collapse
Affiliation(s)
- Abdulfattah Salah Fararjeh
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 110, Taiwan.
| | - Li-Ching Chen
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 110, Taiwan.
- Division of Breast Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei 110, Taiwan.
- Taipei Cancer Center, Taipei Medical University, Taipei 110, Taiwan.
| | - Yuan-Soon Ho
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 110, Taiwan.
- Taipei Cancer Center, Taipei Medical University, Taipei 110, Taiwan.
- Department of Medical Laboratory, Taipei Medical University Hospital, Taipei 110, Taiwan.
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan.
| | - Tzu-Chun Cheng
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan.
| | - Yun-Ru Liu
- Joint Biobank, Office of Human Research, Taipei Medical University, Taipei 110, Taiwan.
| | - Hang-Lung Chang
- Department of Surgery, EnChu Kong Hospital, New Taipei City237, Taiwan.
| | - Hui-Wen Chang
- Department of Medical Laboratory, Taipei Medical University Hospital, Taipei 110, Taiwan.
| | - Chih-Hsiung Wu
- Department of Surgery, EnChu Kong Hospital, New Taipei City237, Taiwan.
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
| | - Shih-Hsin Tu
- Division of Breast Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei 110, Taiwan.
- Taipei Cancer Center, Taipei Medical University, Taipei 110, Taiwan.
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
| |
Collapse
|
35
|
Basudan A, Priedigkeit N, Hartmaier RJ, Sokol ES, Bahreini A, Watters RJ, Boisen MM, Bhargava R, Weiss KR, Karsten MM, Denkert C, Blohmer JU, Leone JP, Hamilton RL, Brufsky AM, Elishaev E, Lucas PC, Lee AV, Oesterreich S. Frequent ESR1 and CDK Pathway Copy-Number Alterations in Metastatic Breast Cancer. Mol Cancer Res 2019; 17:457-468. [PMID: 30355675 PMCID: PMC6359977 DOI: 10.1158/1541-7786.mcr-18-0946] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 10/04/2018] [Accepted: 10/11/2018] [Indexed: 12/30/2022]
Abstract
DNA sequencing has identified a limited number of driver mutations in metastatic breast cancer beyond single base-pair mutations in the estrogen receptor (ESR1). However, our previous studies and others have observed that structural variants, such as ESR1 fusions, may also play a role. Therefore, we expanded upon these observations by performing a comprehensive and highly sensitive characterization of copy-number (CN) alterations in a large clinical cohort of metastatic specimens. NanoString DNA hybridization was utilized to measure CN gains, amplifications, and deletions of 67 genes in 108 breast cancer metastases, and in 26 cases, the patient-matched primary tumor. For ESR1, a copyshift algorithm was applied to identify CN imbalances at exon-specific resolution and queried large data sets (>15,000 tumors) that had previously undergone next-generation sequencing (NGS). Interestingly, a subset of ER+ tumors showed increased ESR1 CN (11/82, 13%); three had CN amplifications (4%) and eight had gains (10%). Increased ESR1 CN was enriched in metastatic specimens versus primary tumors, and this was orthogonally confirmed in a large NGS data set. ESR1-amplified tumors showed a site-specific enrichment for bone metastases and worse outcomes than nonamplified tumors. No ESR1 CN amplifications and only one gain was identified in ER- tumors. ESR1 copyshift was present in 5 of the 11 ESR1-amplified tumors. Other frequent amplifications included ERBB2, GRB7, and cell-cycle pathway members CCND1 and CDK4/6, which showed mutually exclusivity with deletions of CDKN2A, CDKN2B, and CDKN1B. IMPLICATIONS: Copy-number alterations of ESR1 and key CDK pathway genes are frequent in metastatic breast cancers, and their clinical relevance should be tested further.
Collapse
Affiliation(s)
- Ahmed Basudan
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania
- Women's Cancer Research Center, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Clinical Lab Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Nolan Priedigkeit
- Women's Cancer Research Center, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania
- School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ryan J Hartmaier
- Women's Cancer Research Center, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Amir Bahreini
- Women's Cancer Research Center, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Rebecca J Watters
- Department of Orthopedic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Michelle M Boisen
- Women's Cancer Research Center, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Obstetrics and Gynecology, University of Pittsburgh, Pittsburgh, Pennsylvania
- Magee-Women Hospital, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Rohit Bhargava
- Women's Cancer Research Center, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Kurt R Weiss
- Department of Orthopedic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Surgical Oncology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | | | | | - Jose P Leone
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ronald L Hamilton
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Adam M Brufsky
- Women's Cancer Research Center, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Esther Elishaev
- Women's Cancer Research Center, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania
- Magee-Women Hospital, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Peter C Lucas
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Adrian V Lee
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania
- Women's Cancer Research Center, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Steffi Oesterreich
- Women's Cancer Research Center, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania.
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
36
|
Paroni G, Bolis M, Zanetti A, Ubezio P, Helin K, Staller P, Gerlach LO, Fratelli M, Neve RM, Terao M, Garattini E. HER2-positive breast-cancer cell lines are sensitive to KDM5 inhibition: definition of a gene-expression model for the selection of sensitive cases. Oncogene 2018; 38:2675-2689. [DOI: 10.1038/s41388-018-0620-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 11/09/2018] [Accepted: 11/19/2018] [Indexed: 12/31/2022]
|
37
|
Yang J, Wei X, Tufan T, Kuscu C, Unlu H, Farooq S, Demirtas E, Paschal BM, Adli M. Recurrent mutations at estrogen receptor binding sites alter chromatin topology and distal gene expression in breast cancer. Genome Biol 2018; 19:190. [PMID: 30404658 PMCID: PMC6223090 DOI: 10.1186/s13059-018-1572-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 10/22/2018] [Indexed: 02/16/2023] Open
Abstract
BACKGROUND The mutational processes underlying non-coding cancer mutations and their biological significance in tumor evolution are poorly understood. To get better insights into the biological mechanisms of mutational processes in breast cancer, we integrate whole-genome level somatic mutations from breast cancer patients with chromatin states and transcription factor binding events. RESULTS We discover that a large fraction of non-coding somatic mutations in estrogen receptor (ER)-positive breast cancers are confined to ER binding sites. Notably, the highly mutated estrogen receptor binding sites are associated with more frequent chromatin loop contacts and the associated distal genes are expressed at higher level. To elucidate the functional significance of these non-coding mutations, we focus on two of the recurrently mutated estrogen receptor binding sites. Our bioinformatics and biochemical analysis suggest loss of DNA-protein interactions due to the recurrent mutations. Through CRISPR interference, we find that the recurrently mutated regulatory element at the LRRC3C-GSDMA locus impacts the expression of multiple distal genes. Using a CRISPR base editor, we show that the recurrent C→T conversion at the ZNF143 locus results in decreased TF binding, increased chromatin loop formation, and increased expression of multiple distal genes. This single point mutation mediates reduced response to estradiol-induced cell proliferation but increased resistance to tamoxifen-induced growth inhibition. CONCLUSIONS Our data suggest that ER binding is associated with localized accumulation of somatic mutations, some of which affect chromatin architecture, distal gene expression, and cellular phenotypes in ER-positive breast cancer.
Collapse
Affiliation(s)
- Jiekun Yang
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, 1340 Jefferson Park Ave, Pinn Hall, Room: 6228, Charlottesville, VA, 22903, USA
| | - Xiaolong Wei
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, 1340 Jefferson Park Ave, Pinn Hall, Room: 6228, Charlottesville, VA, 22903, USA
| | - Turan Tufan
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, 1340 Jefferson Park Ave, Pinn Hall, Room: 6228, Charlottesville, VA, 22903, USA
| | - Cem Kuscu
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, 1340 Jefferson Park Ave, Pinn Hall, Room: 6228, Charlottesville, VA, 22903, USA
| | - Hayrunnisa Unlu
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, 1340 Jefferson Park Ave, Pinn Hall, Room: 6228, Charlottesville, VA, 22903, USA
| | - Saadia Farooq
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, 1340 Jefferson Park Ave, Pinn Hall, Room: 6228, Charlottesville, VA, 22903, USA
| | - Elif Demirtas
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, 1340 Jefferson Park Ave, Pinn Hall, Room: 6228, Charlottesville, VA, 22903, USA
| | - Bryce M Paschal
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, 1340 Jefferson Park Ave, Pinn Hall, Room: 6228, Charlottesville, VA, 22903, USA
- Center for Cell Signalling, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Mazhar Adli
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, 1340 Jefferson Park Ave, Pinn Hall, Room: 6228, Charlottesville, VA, 22903, USA.
| |
Collapse
|
38
|
Characterizing functional consequences of DNA copy number alterations in breast and ovarian tumors by spaceMap. J Genet Genomics 2018; 45:361-371. [PMID: 30057342 DOI: 10.1016/j.jgg.2018.07.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 07/09/2018] [Accepted: 07/09/2018] [Indexed: 01/18/2023]
Abstract
We propose a novel conditional graphical model - spaceMap - to construct gene regulatory networks from multiple types of high dimensional omic profiles. A motivating application is to characterize the perturbation of DNA copy number alterations (CNAs) on downstream protein levels in tumors. Through a penalized multivariate regression framework, spaceMap jointly models high dimensional protein levels as responses and high dimensional CNAs as predictors. In this setup, spaceMap infers an undirected network among proteins together with a directed network encoding how CNAs perturb the protein network. spaceMap can be applied to learn other types of regulatory relationships from high dimensional molecular profiles, especially those exhibiting hub structures. Simulation studies show spaceMap has greater power in detecting regulatory relationships over competing methods. Additionally, spaceMap includes a network analysis toolkit for biological interpretation of inferred networks. We applies spaceMap to the CNAs, gene expression and proteomics data sets from CPTAC-TCGA breast (n=77) and ovarian (n=174) cancer studies. Each cancer exhibits disruption of 'ion transmembrane transport' and 'regulation from RNA polymerase II promoter' by CNA events unique to each cancer. Moreover, using protein levels as a response yields a more functionally-enriched network than using RNA expressions in both cancer types. The network results also help to pinpoint crucial cancer genes and provide insights on the functional consequences of important CNA in breast and ovarian cancers. The R package spaceMap - including vignettes and documentation - is hosted on https://topherconley.github.io/spacemap.
Collapse
|
39
|
Lacher MD, Bauer G, Fury B, Graeve S, Fledderman EL, Petrie TD, Coleal-Bergum DP, Hackett T, Perotti NH, Kong YY, Kwok WW, Wagner JP, Wiseman CL, Williams WV. SV-BR-1-GM, a Clinically Effective GM-CSF-Secreting Breast Cancer Cell Line, Expresses an Immune Signature and Directly Activates CD4 + T Lymphocytes. Front Immunol 2018; 9:776. [PMID: 29867922 PMCID: PMC5962696 DOI: 10.3389/fimmu.2018.00776] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 03/28/2018] [Indexed: 12/18/2022] Open
Abstract
Targeted cancer immunotherapy with irradiated, granulocyte–macrophage colony-stimulating factor (GM-CSF)-secreting, allogeneic cancer cell lines has been an effective approach to reduce tumor burden in several patients. It is generally assumed that to be effective, these cell lines need to express immunogenic antigens coexpressed in patient tumor cells, and antigen-presenting cells need to take up such antigens then present them to patient T cells. We have previously reported that, in a phase I pilot study (ClinicalTrials.gov NCT00095862), a subject with stage IV breast cancer experienced substantial regression of breast, lung, and brain lesions following inoculation with clinical formulations of SV-BR-1-GM, a GM-CSF-secreting breast tumor cell line. To identify diagnostic features permitting the prospective identification of patients likely to benefit from SV-BR-1-GM, we conducted a molecular analysis of the SV-BR-1-GM cell line and of patient-derived blood, as well as a tumor specimen. Compared to normal human breast cells, SV-BR-1-GM cells overexpress genes encoding tumor-associated antigens (TAAs) such as PRAME, a cancer/testis antigen. Curiously, despite its presumptive breast epithelial origin, the cell line expresses major histocompatibility complex (MHC) class II genes (HLA-DRA, HLA-DRB3, HLA-DMA, HLA-DMB), in addition to several other factors known to play immunostimulatory roles. These factors include MHC class I components (B2M, HLA-A, HLA-B), ADA (encoding adenosine deaminase), ADGRE5 (CD97), CD58 (LFA3), CD74 (encoding invariant chain and CLIP), CD83, CXCL8 (IL8), CXCL16, HLA-F, IL6, IL18, and KITLG. Moreover, both SV-BR-1-GM cells and the responding study subject carried an HLA-DRB3*02:02 allele, raising the question of whether SV-BR-1-GM cells can directly present endogenous antigens to T cells, thereby inducing a tumor-directed immune response. In support of this, SV-BR-1-GM cells (which also carry the HLA-DRB3*01:01 allele) treated with yellow fever virus (YFV) envelope (Env) 43–59 peptides reactivated YFV-DRB3*01:01-specific CD4+ T cells. Thus, the partial HLA allele match between SV-BR-1-GM and the clinical responder might have enabled patient T lymphocytes to directly recognize SV-BR-1-GM TAAs as presented on SV-BR-1-GM MHCs. Taken together, our findings are consistent with a potentially unique mechanism of action by which SV-BR-1-GM cells can act as APCs for previously primed CD4+ T cells.
Collapse
Affiliation(s)
| | - Gerhard Bauer
- GMP Facility, Institute for Regenerative Cures, University of California, Davis (UCD), Sacramento, CA, United States
| | - Brian Fury
- GMP Facility, Institute for Regenerative Cures, University of California, Davis (UCD), Sacramento, CA, United States
| | - Sanne Graeve
- BriaCell Therapeutics Corp., Berkeley, CA, United States
| | - Emily L Fledderman
- GMP Facility, Institute for Regenerative Cures, University of California, Davis (UCD), Sacramento, CA, United States
| | - Tye D Petrie
- GMP Facility, Institute for Regenerative Cures, University of California, Davis (UCD), Sacramento, CA, United States
| | - Dane P Coleal-Bergum
- GMP Facility, Institute for Regenerative Cures, University of California, Davis (UCD), Sacramento, CA, United States
| | - Tia Hackett
- GMP Facility, Institute for Regenerative Cures, University of California, Davis (UCD), Sacramento, CA, United States
| | - Nicholas H Perotti
- GMP Facility, Institute for Regenerative Cures, University of California, Davis (UCD), Sacramento, CA, United States
| | - Ying Y Kong
- Benaroya Research Institute at Virginia Mason, Seattle, WA, United States
| | - William W Kwok
- Benaroya Research Institute at Virginia Mason, Seattle, WA, United States
| | | | | | | |
Collapse
|
40
|
Agersborg S, Mixon C, Nguyen T, Aithal S, Sudarsanam S, Blocker F, Weiss L, Gasparini R, Jiang S, Chen W, Hess G, Albitar M. Immunohistochemistry and alternative FISH testing in breast cancer with HER2 equivocal amplification. Breast Cancer Res Treat 2018; 170:321-328. [PMID: 29564742 PMCID: PMC5999182 DOI: 10.1007/s10549-018-4755-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 03/13/2018] [Indexed: 02/04/2023]
Abstract
Purpose While HER2 testing is well established in directing appropriate treatment for breast cancer, a small percentage of cases show equivocal results by immunohistochemistry (IHC) and fluorescence in situ hybridization (FISH). Alternative probes may be used in equivocal cases. We present a single community-based institution’s experience in further evaluating these cases. Patients and methods Between 2014 and 2016, 4255 samples were submitted for HER2 amplification testing by alternative probes, TP53, RAI1, and RARA. Of the patients tested by FISH, 505/3908 (12.9%) also had IHC data. Results Most (73.9%) FISH equivocal cases remained equivocal after IHC testing. However, 50.5% of equivocal cases were classified as HER2 amplified by alternative probes. Most cases were positive by more than one probe: 78% of positive cases by RAI1 and 73.9% by TP53. There was a significant difference between IHC and FISH alternative testing (p < 0.0001) among the equivocal cases by conventional FISH testing, 44% of IHC negative cases became positive while 36% of the positive IHC cases became negative by alternative FISH testing. Available data showed that 41% of patients were treated with palbociclib and were positive by alternative FISH. Conclusion The prevalence of double HER2 equivocal cases and the discrepancy between IHC and alternative FISH testing suggest that FISH alternative testing using both RAI1 and TP53 probes is necessary for conclusive classification. Because almost half of FISH equivocal cases converted to HER2 amplified upon alternative testing, clinical studies to determine the benefit of anti-HER2 therapy in these patients are urgently needed.
Collapse
Affiliation(s)
- Sally Agersborg
- NeoGenomics Laboratories, Research and Development, 31 Columbia, Aliso Viejo, CA, 92656, USA
| | - Christopher Mixon
- NeoGenomics Laboratories, Research and Development, 31 Columbia, Aliso Viejo, CA, 92656, USA
| | - Thanh Nguyen
- NeoGenomics Laboratories, Research and Development, 31 Columbia, Aliso Viejo, CA, 92656, USA
| | - Sramila Aithal
- Cancer Treatment Centers of America, Philadelphia, PA, USA
| | - Sucha Sudarsanam
- NeoGenomics Laboratories, Research and Development, 31 Columbia, Aliso Viejo, CA, 92656, USA
| | - Forrest Blocker
- NeoGenomics Laboratories, Research and Development, 31 Columbia, Aliso Viejo, CA, 92656, USA
| | - Lawrence Weiss
- NeoGenomics Laboratories, Research and Development, 31 Columbia, Aliso Viejo, CA, 92656, USA
| | - Robert Gasparini
- NeoGenomics Laboratories, Research and Development, 31 Columbia, Aliso Viejo, CA, 92656, USA
| | - Shiping Jiang
- NeoGenomics Laboratories, Research and Development, 31 Columbia, Aliso Viejo, CA, 92656, USA
| | - Wayne Chen
- NeoGenomics Laboratories, Research and Development, 31 Columbia, Aliso Viejo, CA, 92656, USA
| | | | - Maher Albitar
- NeoGenomics Laboratories, Research and Development, 31 Columbia, Aliso Viejo, CA, 92656, USA.
| |
Collapse
|
41
|
Daemen A, Manning G. HER2 is not a cancer subtype but rather a pan-cancer event and is highly enriched in AR-driven breast tumors. Breast Cancer Res 2018; 20:8. [PMID: 29382369 PMCID: PMC5791377 DOI: 10.1186/s13058-018-0933-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 01/09/2018] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Approximately one in five breast cancers are driven by amplification and overexpression of the human epidermal growth factor receptor 2 (HER2) receptor kinase, and HER2-enriched (HER2E) is one of four major transcriptional subtypes of breast cancer. We set out to understand the genomics of HER2 amplification independent of subtype, and the underlying drivers and biology of HER2E tumors. METHODS We investigated published genomic data from 3155 breast tumors and 5391 non-breast tumors. RESULTS HER2 amplification is a distinct driver event seen in all breast cancer subtypes, rather than a subtype marker, with major characteristics restricted to amplification and overexpression of HER2 and neighboring genes. The HER2E subtype has a distinctive transcriptional landscape independent of HER2A that reflects androgen receptor signaling as replacement for estrogen receptor (ER)-driven tumorigenesis. HER2 amplification is also an event in 1.8% of non-breast tumors. CONCLUSIONS These discoveries reveal therapeutic opportunities for combining anti-HER2 therapy with anti-androgen agents in breast cancer, and highlight the potential for broader therapeutic use of HER2 inhibitors.
Collapse
Affiliation(s)
- Anneleen Daemen
- Bioinformatics & Computational Biology, Genentech, Inc, 1 DNA Way, MS444a, South San Francisco, CA, 94080, USA.
| | - Gerard Manning
- Bioinformatics & Computational Biology, Genentech, Inc, 1 DNA Way, MS444a, South San Francisco, CA, 94080, USA.
| |
Collapse
|
42
|
Hergueta-Redondo M, Sarrio D, Molina-Crespo Á, Vicario R, Bernadó-Morales C, Martínez L, Rojo-Sebastián A, Serra-Musach J, Mota A, Martínez-Ramírez Á, Castilla MÁ, González-Martin A, Pernas S, Cano A, Cortes J, Nuciforo PG, Peg V, Palacios J, Pujana MÁ, Arribas J, Moreno-Bueno G. Gasdermin B expression predicts poor clinical outcome in HER2-positive breast cancer. Oncotarget 2018; 7:56295-56308. [PMID: 27462779 PMCID: PMC5302915 DOI: 10.18632/oncotarget.10787] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 07/06/2016] [Indexed: 01/03/2023] Open
Abstract
Around, 30–40% of HER2-positive breast cancers do not show substantial clinical benefit from the targeted therapy and, thus, the mechanisms underlying resistance remain partially unknown. Interestingly, ERBB2 is frequently co-amplified and co-expressed with neighbour genes that may play a relevant role in this cancer subtype. Here, using an in silico analysis of data from 2,096 breast tumours, we reveal a significant correlation between Gasdermin B (GSDMB) gene (located 175 kilo bases distal from ERBB2) expression and the pathological and clinical parameters of poor prognosis in HER2-positive breast cancer. Next, the analysis of three independent cohorts (totalizing 286 tumours) showed that approximately 65% of the HER2-positive cases have GSDMB gene amplification and protein over-expression. Moreover, GSDMB expression was also linked to poor therapeutic responses in terms of lower relapse free survival and pathologic complete response as well as positive lymph node status and the development of distant metastasis under neoadjuvant and adjuvant treatment settings, respectively. Importantly, GSDMB expression promotes survival to trastuzumab in different HER2-positive breast carcinoma cells, and is associated with trastuzumab resistance phenotype in vivo in Patient Derived Xenografts. In summary, our data identifies the ERBB2 co-amplified and co-expressed gene GSDMB as a critical determinant of poor prognosis and therapeutic response in HER2-positive breast cancer.
Collapse
Affiliation(s)
- Marta Hergueta-Redondo
- Biochemistry Department, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), IdiPAZ, Madrid, Spain
| | - David Sarrio
- Biochemistry Department, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), IdiPAZ, Madrid, Spain
| | - Ángela Molina-Crespo
- Biochemistry Department, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), IdiPAZ, Madrid, Spain
| | - Rocío Vicario
- Preclinical Oncology Program, Vall d'Hebron Institute of Oncology (VHIO), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Cristina Bernadó-Morales
- Preclinical Oncology Program, Vall d'Hebron Institute of Oncology (VHIO), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Lidia Martínez
- Biochemistry Department, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), IdiPAZ, Madrid, Spain
| | | | - Jordi Serra-Musach
- Breast Cancer and Systems Biology Unit, ProCURE, Catalan Institute of Oncology, IDIBELL, L'Hospitalet del Llobregat, Barcelona, Spain
| | - Alba Mota
- Biochemistry Department, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), IdiPAZ, Madrid, Spain.,Translational Research Laboratory, MD Anderson Internacional Foundation, Madrid, Spain
| | | | - Mª Ángeles Castilla
- Pathology Department, Hospital Universitario Virgen del Rocío, Sevilla, Spain
| | | | - Sonia Pernas
- Breast Cancer and Systems Biology Unit, ProCURE, Catalan Institute of Oncology, IDIBELL, L'Hospitalet del Llobregat, Barcelona, Spain
| | - Amparo Cano
- Biochemistry Department, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), IdiPAZ, Madrid, Spain
| | - Javier Cortes
- Clinical Oncology Program, Vall d'Hebron Institute of Oncology (VHIO), Universitat Autonoma de Barcelona, Barcelona, Spain.,Oncology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Paolo G Nuciforo
- Molecular Oncology Program, Vall d'Hebron Institute of Oncology (VHIO), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Vicente Peg
- Pathology Department, Hospital Vall d'Hebron University, Barcelona, Spain
| | - José Palacios
- Pathology Department, Hospital Universitario Virgen del Rocío, Sevilla, Spain.,Pathology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Miguel Ángel Pujana
- Breast Cancer and Systems Biology Unit, ProCURE, Catalan Institute of Oncology, IDIBELL, L'Hospitalet del Llobregat, Barcelona, Spain
| | - Joaquín Arribas
- Preclinical Oncology Program, Vall d'Hebron Institute of Oncology (VHIO), Universitat Autònoma de Barcelona, Barcelona, Spain.,Clinical Oncology Program, Vall d'Hebron Institute of Oncology (VHIO), Universitat Autonoma de Barcelona, Barcelona, Spain.,Molecular Oncology Program, Vall d'Hebron Institute of Oncology (VHIO), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Gema Moreno-Bueno
- Biochemistry Department, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), IdiPAZ, Madrid, Spain.,Translational Research Laboratory, MD Anderson Internacional Foundation, Madrid, Spain
| |
Collapse
|
43
|
GRB7 Expression and Correlation With HER2 Amplification in Invasive Breast Carcinoma. Appl Immunohistochem Mol Morphol 2017; 25:553-558. [DOI: 10.1097/pai.0000000000000349] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
44
|
Davidson MA, Shanks EJ. 3q26-29 Amplification in head and neck squamous cell carcinoma: a review of established and prospective oncogenes. FEBS J 2017; 284:2705-2731. [PMID: 28317270 DOI: 10.1111/febs.14061] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 02/23/2017] [Accepted: 03/15/2017] [Indexed: 12/22/2022]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is significantly underrepresented in worldwide cancer research, yet survival rates for the disease have remained static for over 50 years. Distant metastasis is often present at the time of diagnosis, and is the primary cause of death in cancer patients. In the absence of routine effective targeted therapies, the standard of care treatment remains chemoradiation in combination with (often disfiguring) surgery. A defining characteristic of HNSCC is the amplification of a region of chromosome 3 (3q26-29), which is consistently associated with poorer patient outcome. This review provides an overview of the role the 3q26-29 region plays in HNSCC, in terms of both known and as yet undiscovered processes, which may have potential clinical relevance.
Collapse
|
45
|
Zhao HB, Zhang XF, Jia XL, Wang HB. Grb7 is over-expressed in cervical cancer and facilitate invasion and inhibit apoptosis in cervical cancer cells. Pathol Res Pract 2017; 213:1180-1184. [DOI: 10.1016/j.prp.2017.05.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Revised: 04/21/2017] [Accepted: 05/25/2017] [Indexed: 12/18/2022]
|
46
|
Birkeland AC, Yanik M, Tillman BN, Scott MV, Foltin SK, Mann JE, Michmerhuizen NL, Ludwig ML, Sandelski MM, Komarck CM, Carey TE, Prince MEP, Bradford CR, McHugh JB, Spector ME, Brenner JC. Identification of Targetable ERBB2 Aberrations in Head and Neck Squamous Cell Carcinoma. JAMA Otolaryngol Head Neck Surg 2017; 142:559-67. [PMID: 27077364 DOI: 10.1001/jamaoto.2016.0335] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
IMPORTANCE ERBB2 (formerly HER2) is an important drug target in breast cancer, where anti-ERBB2 therapy has been shown to lead to improvements in disease recurrence and overall survival. ERBB2 status in head and neck squamous cell carcinoma (HNSCC) has not been well studied. Identification of ERBB2-positive tumors and characterization of response to ERBB2 therapy could lead to targeted treatment options in HNSCC. OBJECTIVE To identify ERBB2 aberrations in HNSCCs and investigate the potential for ERBB2-targeted therapy in HNSCCs. DESIGN, SETTING, AND PARTICIPANTS A retrospective case series of patients with laryngeal (42 tumor specimens) and oral cavity (94 tumor specimens) SCC enrolled in the University of Michigan Head and Neck Specialized Program of Research Excellence was conducted. Publicly available sequencing data (The Cancer Genome Atlas), as well as data from other studies, were reviewed to identify additional mutations and overexpression in ERBB2 in HNSCC. Established HNSCC cell lines were used for follow-up in vitro analysis. The study was conducted from October 1, 2014, to August 30, 2015. INTERVENTIONS With the use of targeted, amplicon-based sequencing with the Oncomine Cancer Panel, the copy number and mutation status of commonly altered genes in HNSCCs were assessed. Immunohistochemical staining was performed on tissue microarrays of HNSCCs to assess the expression of ERBB2. Western blotting for HNSCC cell line ERBB2 expression and cell survival assays after treatment with ERBB2 inhibitors were performed. MAIN OUTCOMES AND MEASURES The prevalence of ERBB2 genetic aberrations and ERBB2 overexpression in laryngeal and oral cavity SCCs, prevalence of ERBB2 aberrations in HNSCC in The Cancer Genome Atlas, ERBB2 protein expression in HNSCC cell lines, and response of HNSCC cell lines to targeted ERBB2 inhibitors. RESULTS Of the 42 laryngeal SCC samples screened by targeted sequencing, 4 (10%) were positive for ERBB2 amplification. Two of these samples showed ERBB2 overexpression on immunohistochemistry. Two of the 94 oral cavity SCC samples (2%) were positive for ERBB2 on immunohistochemistry. Analysis of 288 patients from publicly available HNSCC sequencing data revealed 9 amplifications (3%) in ERBB2. Protein expression was variable across HNSCC cell lines, and a subset of these cell lines showed responsiveness to anti-ERBB2 therapy. CONCLUSIONS AND RELEVANCE ERBB2 aberrations were identified in a subset of HNSCCs. These tumors may be responsive to targeted therapy against ERBB2. Screening for ERBB2 aberrations and applying targeted therapy in ERBB2-positive patients may be useful in personalized therapy trials, particularly in patients who are refractory to current treatment paradigms.
Collapse
Affiliation(s)
- Andrew C Birkeland
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan Medical School, Ann Arbor
| | - Megan Yanik
- medical student, University of Michigan Medical School, Ann Arbor
| | - Brittny N Tillman
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan Medical School, Ann Arbor
| | - Megan V Scott
- medical student, University of Michigan Medical School, Ann Arbor
| | - Susan K Foltin
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan Medical School, Ann Arbor
| | - Jacqueline E Mann
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan Medical School, Ann Arbor
| | - Nicole L Michmerhuizen
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan Medical School, Ann Arbor
| | - Megan L Ludwig
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan Medical School, Ann Arbor
| | | | - Christine M Komarck
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan Medical School, Ann Arbor
| | - Thomas E Carey
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan Medical School, Ann Arbor3Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor
| | - Mark E P Prince
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan Medical School, Ann Arbor3Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor
| | - Carol R Bradford
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan Medical School, Ann Arbor3Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor
| | - Jonathan B McHugh
- Department of Pathology, University of Michigan Medical School, Ann Arbor
| | - Matthew E Spector
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan Medical School, Ann Arbor3Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor
| | - J Chad Brenner
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan Medical School, Ann Arbor3Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor
| |
Collapse
|
47
|
Aure MR, Vitelli V, Jernström S, Kumar S, Krohn M, Due EU, Haukaas TH, Leivonen SK, Vollan HKM, Lüders T, Rødland E, Vaske CJ, Zhao W, Møller EK, Nord S, Giskeødegård GF, Bathen TF, Caldas C, Tramm T, Alsner J, Overgaard J, Geisler J, Bukholm IRK, Naume B, Schlichting E, Sauer T, Mills GB, Kåresen R, Mælandsmo GM, Lingjærde OC, Frigessi A, Kristensen VN, Børresen-Dale AL, Sahlberg KK. Integrative clustering reveals a novel split in the luminal A subtype of breast cancer with impact on outcome. Breast Cancer Res 2017; 19:44. [PMID: 28356166 PMCID: PMC5372339 DOI: 10.1186/s13058-017-0812-y] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 02/05/2017] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Breast cancer is a heterogeneous disease at the clinical and molecular level. In this study we integrate classifications extracted from five different molecular levels in order to identify integrated subtypes. METHODS Tumor tissue from 425 patients with primary breast cancer from the Oslo2 study was cut and blended, and divided into fractions for DNA, RNA and protein isolation and metabolomics, allowing the acquisition of representative and comparable molecular data. Patients were stratified into groups based on their tumor characteristics from five different molecular levels, using various clustering methods. Finally, all previously identified and newly determined subgroups were combined in a multilevel classification using a "cluster-of-clusters" approach with consensus clustering. RESULTS Based on DNA copy number data, tumors were categorized into three groups according to the complex arm aberration index. mRNA expression profiles divided tumors into five molecular subgroups according to PAM50 subtyping, and clustering based on microRNA expression revealed four subgroups. Reverse-phase protein array data divided tumors into five subgroups. Hierarchical clustering of tumor metabolic profiles revealed three clusters. Combining DNA copy number and mRNA expression classified tumors into seven clusters based on pathway activity levels, and tumors were classified into ten subtypes using integrative clustering. The final consensus clustering that incorporated all aforementioned subtypes revealed six major groups. Five corresponded well with the mRNA subtypes, while a sixth group resulted from a split of the luminal A subtype; these tumors belonged to distinct microRNA clusters. Gain-of-function studies using MCF-7 cells showed that microRNAs differentially expressed between the luminal A clusters were important for cancer cell survival. These microRNAs were used to validate the split in luminal A tumors in four independent breast cancer cohorts. In two cohorts the microRNAs divided tumors into subgroups with significantly different outcomes, and in another a trend was observed. CONCLUSIONS The six integrated subtypes identified confirm the heterogeneity of breast cancer and show that finer subdivisions of subtypes are evident. Increasing knowledge of the heterogeneity of the luminal A subtype may add pivotal information to guide therapeutic choices, evidently bringing us closer to improved treatment for this largest subgroup of breast cancer.
Collapse
Affiliation(s)
- Miriam Ragle Aure
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
- K.G. Jebsen Centre for Breast Cancer Research, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Valeria Vitelli
- Oslo Center for Biostatistics and Epidemiology, Institute of Basic Medical Science, University of Oslo, Oslo, Norway
| | - Sandra Jernström
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
- K.G. Jebsen Centre for Breast Cancer Research, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Surendra Kumar
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
- K.G. Jebsen Centre for Breast Cancer Research, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Clinical Molecular Biology (EpiGen), Division of Medicine, Akershus University Hospital, Lørenskog, Norway
| | - Marit Krohn
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
- K.G. Jebsen Centre for Breast Cancer Research, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Eldri U. Due
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
- K.G. Jebsen Centre for Breast Cancer Research, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Tonje Husby Haukaas
- K.G. Jebsen Centre for Breast Cancer Research, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Suvi-Katri Leivonen
- Genome-Scale Biology Research Program, University of Helsinki, Helsinki, Finland
| | - Hans Kristian Moen Vollan
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
- K.G. Jebsen Centre for Breast Cancer Research, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Torben Lüders
- K.G. Jebsen Centre for Breast Cancer Research, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Clinical Molecular Biology (EpiGen), Division of Medicine, Akershus University Hospital, Lørenskog, Norway
| | - Einar Rødland
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | | | - Wei Zhao
- Department of Systems Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX USA
| | - Elen K. Møller
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
- K.G. Jebsen Centre for Breast Cancer Research, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Silje Nord
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
- K.G. Jebsen Centre for Breast Cancer Research, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Guro F. Giskeødegård
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Tone Frost Bathen
- K.G. Jebsen Centre for Breast Cancer Research, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Carlos Caldas
- Cambridge University Hospitals Trust, Addenbrookes Hospital, Cambridge, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Trine Tramm
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Jan Alsner
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Jens Overgaard
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Jürgen Geisler
- Department of Oncology, Akershus University Hospital, Lørenskog, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Ida R. K. Bukholm
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Surgery, Akershus University Hospital, Lørenskog, Norway
| | - Bjørn Naume
- K.G. Jebsen Centre for Breast Cancer Research, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Oncology, Division of Cancer Medicine, Oslo University Hospital, Oslo, Norway
| | - Ellen Schlichting
- K.G. Jebsen Centre for Breast Cancer Research, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Breast and Endocrine Surgery, Oslo University Hospital, Oslo, Norway
| | - Torill Sauer
- K.G. Jebsen Centre for Breast Cancer Research, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Pathology, Akershus University Hospital, Lørenskog, Norway
| | - Gordon B. Mills
- Department of Systems Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX USA
| | - Rolf Kåresen
- K.G. Jebsen Centre for Breast Cancer Research, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Breast and Endocrine Surgery, Oslo University Hospital, Oslo, Norway
| | - Gunhild M. Mælandsmo
- K.G. Jebsen Centre for Breast Cancer Research, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | - Ole Christian Lingjærde
- K.G. Jebsen Centre for Breast Cancer Research, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
- Centre for Cancer Biomedicine, University of Oslo, Oslo, Norway
- Department of Computer Science, University of Oslo, Oslo, Norway
| | - Arnoldo Frigessi
- Oslo Center for Biostatistics and Epidemiology, Institute of Basic Medical Science, University of Oslo, Oslo, Norway
- Oslo Center for Biostatistics and Epidemiology, Oslo University Hospital, Oslo, Norway
| | - Vessela N. Kristensen
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
- K.G. Jebsen Centre for Breast Cancer Research, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Clinical Molecular Biology (EpiGen), Division of Medicine, Akershus University Hospital, Lørenskog, Norway
| | - Anne-Lise Børresen-Dale
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
- K.G. Jebsen Centre for Breast Cancer Research, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Kristine K. Sahlberg
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
- K.G. Jebsen Centre for Breast Cancer Research, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Research, Vestre Viken Hospital Trust, Drammen, Norway
| |
Collapse
|
48
|
Jernström S, Hongisto V, Leivonen SK, Due EU, Tadele DS, Edgren H, Kallioniemi O, Perälä M, Mælandsmo GM, Sahlberg KK. Drug-screening and genomic analyses of HER2-positive breast cancer cell lines reveal predictors for treatment response. BREAST CANCER-TARGETS AND THERAPY 2017; 9:185-198. [PMID: 28356768 PMCID: PMC5367762 DOI: 10.2147/bctt.s115600] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background Approximately 15%–20% of all diagnosed breast cancers are characterized by amplified and overexpressed HER2 (= ErbB2). These breast cancers are aggressive and have a poor prognosis. Although improvements in treatment have been achieved after the introduction of trastuzumab and lapatinib, many patients do not benefit from these drugs. Therefore, in-depth understanding of the mechanisms behind the treatment responses is essential to find alternative therapeutic strategies. Materials and methods Thirteen HER2 positive breast cancer cell lines were screened with 22 commercially available compounds, mainly targeting proteins in the ErbB2-signaling pathway, and molecular mechanisms related to treatment sensitivity were sought. Cell viability was measured, and treatment responses between the cell lines were compared. To search for response predictors and genomic and transcriptomic profiling, PIK3CA mutations and PTEN status were explored and molecular features associated with drug sensitivity sought. Results The cell lines were divided into three groups according to the growth-retarding effect induced by trastuzumab and lapatinib. Interestingly, two cell lines insensitive to trastuzumab (KPL4 and SUM190PT) showed sensitivity to an Akt1/2 kinase inhibitor. These cell lines had mutation in PIK3CA and loss of PTEN, suggesting an activated and druggable Akt-signaling pathway. Expression levels of five genes (CDC42, MAPK8, PLCG1, PTK6, and PAK6) were suggested as predictors for the Akt1/2 kinase-inhibitor response. Conclusion Targeting the Akt-signaling pathway shows promise in cell lines that do not respond to trastuzumab. In addition, our results indicate that several molecular features determine the growth-retarding effects induced by the drugs, suggesting that parameters other than HER2 amplification/expression should be included as markers for therapy decisions.
Collapse
Affiliation(s)
- Sandra Jernström
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital; KG Jebsen Centre for Breast Cancer Research, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | | | - Suvi-Katri Leivonen
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital; KG Jebsen Centre for Breast Cancer Research, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Eldri Undlien Due
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital
| | - Dagim Shiferaw Tadele
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital
| | - Henrik Edgren
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki; Medisapiens, Helsinki, Finland
| | - Olli Kallioniemi
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki
| | - Merja Perälä
- VTT Technical Research Centre of Finland, Turku, Finland
| | - Gunhild Mari Mælandsmo
- KG Jebsen Centre for Breast Cancer Research, Institute for Clinical Medicine, University of Oslo, Oslo, Norway; Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway; Institute of Pharmacy, Faculty of Health Sciences, University of Tromsø, Tromsø
| | - Kristine Kleivi Sahlberg
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital; Department of Research, Vestre Viken Hospital Trust, Drammen, Norway
| |
Collapse
|
49
|
Grasso S, Chapelle J, Salemme V, Aramu S, Russo I, Vitale N, Verdun di Cantogno L, Dallaglio K, Castellano I, Amici A, Centonze G, Sharma N, Lunardi S, Cabodi S, Cavallo F, Lamolinara A, Stramucci L, Moiso E, Provero P, Albini A, Sapino A, Staaf J, Di Fiore PP, Bertalot G, Pece S, Tosoni D, Confalonieri S, Iezzi M, Di Stefano P, Turco E, Defilippi P. The scaffold protein p140Cap limits ERBB2-mediated breast cancer progression interfering with Rac GTPase-controlled circuitries. Nat Commun 2017; 8:14797. [PMID: 28300085 PMCID: PMC5357316 DOI: 10.1038/ncomms14797] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 01/27/2017] [Indexed: 12/29/2022] Open
Abstract
The docking protein p140Cap negatively regulates tumour cell features. Its relevance on breast cancer patient survival, as well as its ability to counteract relevant cancer signalling pathways, are not fully understood. Here we report that in patients with ERBB2-amplified breast cancer, a p140Cap-positive status associates with a significantly lower probability of developing a distant event, and a clear difference in survival. p140Cap dampens ERBB2-positive tumour cell progression, impairing tumour onset and growth in the NeuT mouse model, and counteracting epithelial mesenchymal transition, resulting in decreased metastasis formation. One major mechanism is the ability of p140Cap to interfere with ERBB2-dependent activation of Rac GTPase-controlled circuitries. Our findings point to a specific role of p140Cap in curbing the aggressiveness of ERBB2-amplified breast cancers and suggest that, due to its ability to impinge on specific molecular pathways, p140Cap may represent a predictive biomarker of response to targeted anti-ERBB2 therapies. p140Cap adaptor proteins interfere with adhesion and growth factor-dependent signalling in cancer cells but the mechanisms are unclear. Here the authors show that p140Cap interferes with ERBB2-dependent activation of Rac GTPase-controlled circuitries reducing metastasis and cancer progression.
Collapse
Affiliation(s)
- Silvia Grasso
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| | - Jennifer Chapelle
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| | - Vincenzo Salemme
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| | - Simona Aramu
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| | - Isabella Russo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| | - Nicoletta Vitale
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| | | | - Katiuscia Dallaglio
- Research Infrastructure, IRCCS Arcispedale Santa Maria Nuova, 42100 Reggio Emilia, Italy
| | | | - Augusto Amici
- Department of Bioscience and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy
| | - Giorgia Centonze
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| | - Nanaocha Sharma
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| | - Serena Lunardi
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| | - Sara Cabodi
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| | - Alessia Lamolinara
- Department of Medicine and Aging Science, Center of Excellence on Aging and Translational Medicine (CeSi-Met), G. D'Annunzio University, Chieti-Pescara, 66100 Chieti, Italy
| | - Lorenzo Stramucci
- Department of Medicine and Aging Science, Center of Excellence on Aging and Translational Medicine (CeSi-Met), G. D'Annunzio University, Chieti-Pescara, 66100 Chieti, Italy
| | - Enrico Moiso
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| | - Paolo Provero
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| | - Adriana Albini
- Scientific and Technology Pole, IRCCS MultiMedica, 20100 Milan, Italy
| | - Anna Sapino
- Department of Medical Sciences, University of Torino, 10126 Torino, Italy
| | - Johan Staaf
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund 22100, Sweden
| | - Pier Paolo Di Fiore
- Molecular Medicine Program, European Institute of Oncology, 20100 Milan, Italy.,IFOM, The FIRC Institute for Molecular Oncology Foundation, 20100 Milan, Italy.,Department of Oncology and Hemato-oncology, University of Milan, 20100 Milan, Italy
| | - Giovanni Bertalot
- Molecular Medicine Program, European Institute of Oncology, 20100 Milan, Italy
| | - Salvatore Pece
- Molecular Medicine Program, European Institute of Oncology, 20100 Milan, Italy.,Department of Oncology and Hemato-oncology, University of Milan, 20100 Milan, Italy
| | - Daniela Tosoni
- Molecular Medicine Program, European Institute of Oncology, 20100 Milan, Italy
| | - Stefano Confalonieri
- Molecular Medicine Program, European Institute of Oncology, 20100 Milan, Italy.,IFOM, The FIRC Institute for Molecular Oncology Foundation, 20100 Milan, Italy
| | - Manuela Iezzi
- Department of Medicine and Aging Science, Center of Excellence on Aging and Translational Medicine (CeSi-Met), G. D'Annunzio University, Chieti-Pescara, 66100 Chieti, Italy
| | - Paola Di Stefano
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| | - Emilia Turco
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| | - Paola Defilippi
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| |
Collapse
|
50
|
Pourteimoor V, Mohammadi-Yeganeh S, Paryan M. Breast cancer classification and prognostication through diverse systems along with recent emerging findings in this respect; the dawn of new perspectives in the clinical applications. Tumour Biol 2016; 37:14479-14499. [DOI: 10.1007/s13277-016-5349-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 09/06/2016] [Indexed: 01/10/2023] Open
|