1
|
Yang S, Manna C, Manna PR. Harnessing the Role of ESR1 in Breast Cancer: Correlation with microRNA, lncRNA, and Methylation. Int J Mol Sci 2025; 26:3101. [PMID: 40243758 PMCID: PMC11988918 DOI: 10.3390/ijms26073101] [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: 02/01/2025] [Revised: 03/24/2025] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
Breast cancer (BC) is a multifactorial condition and it primarily expresses the estrogen receptor α (ERα) that is encoded by the gene estrogen receptor 1 (ESR1), which modulates estrogen signaling. ESR1, by facilitating estrogen overproduction, plays an indispensable role in the progression and survival of the majority of BCs. To obtain molecular insights into these phenomena, we analyzed The Cancer Genome Atlas (TCGA) breast invasive carcinoma (BRCA) RNA-Seq datasets for the expression of ESR1 and its correlation to microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), along with its methylation patterns. Regulation of ESR1 was also assessed with a total of 43 cancerous and non-cancerous breast cell lines. Analyses of both TCGA BRCA and breast cell line RNA-Seq data revealed that specific lncRNAs, i.e., MEG3, BIK, MLL, and FAS are negatively correlated with the ESR1, in which PARP1 demonstrates a positive association. Additionally, both miR-30a and miR-145 showed negative correlations with the ESR1 expression. Of the 54 ESR1 methylation loci analyzed, the majority of them exhibited a negative correlation with the ESR1 expression, highlighting a potentially modifiable regulatory mechanism. These findings underscore the complex regulatory events influencing ESR1 expression and its interaction with diverse signaling pathways, demonstrating novel insights into breast pathogenesis and its potential therapeutics.
Collapse
Affiliation(s)
- Shengping Yang
- Pennington Biomedical Research Center, 6400 Perkins Rd., Baton Rouge, LA 70808, USA
| | - Chayan Manna
- Baylor College of Medicine, Ben Taub Research Center, 1 Baylor Plaza, Houston, TX 77030, USA;
| | - Pulak R. Manna
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX 79430, USA
| |
Collapse
|
2
|
Kaur R, Gupta S, Kulshrestha S, Khandelwal V, Pandey S, Kumar A, Sharma G, Kumar U, Parashar D, Das K. Metabolomics-Driven Biomarker Discovery for Breast Cancer Prognosis and Diagnosis. Cells 2024; 14:5. [PMID: 39791706 PMCID: PMC11720085 DOI: 10.3390/cells14010005] [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: 10/21/2024] [Revised: 12/09/2024] [Accepted: 12/13/2024] [Indexed: 01/12/2025] Open
Abstract
Breast cancer is a cancer with global prevalence and a surge in the number of cases with each passing year. With the advancement in science and technology, significant progress has been achieved in the prevention and treatment of breast cancer to make ends meet. The scientific intradisciplinary subject of "metabolomics" examines every metabolite found in a cell, tissue, system, or organism from different sources of samples. In the case of breast cancer, little is known about the regulatory pathways that could be resolved through metabolic reprogramming. Evidence related to the significant changes taking place during the onset and prognosis of breast cancer can be obtained using metabolomics. Innovative metabolomics approaches identify metabolites that lead to the discovery of biomarkers for breast cancer therapy, diagnosis, and early detection. The use of diverse analytical methods and instruments for metabolomics includes Magnetic Resonance Spectroscopy, LC/MS, UPLC/MS, etc., which, along with their high-throughput analysis, give insights into the metabolites and the molecular pathways involved. For instance, metabolome research has led to the discovery of the glutamate-to-glutamate ratio and aerobic glycolysis as biomarkers in breast cancer. The present review comprehends the updates in metabolomic research and its processes that contribute to breast cancer prognosis and metastasis. The metabolome holds a future, and this review is an attempt to amalgamate the present relevant literature that might yield crucial insights for creating innovative therapeutic strategies aimed at addressing metastatic breast cancer.
Collapse
Affiliation(s)
- Rasanpreet Kaur
- Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, Chaumuhan, Mathura 281406, Uttar Pradesh, India; (R.K.); (S.K.); (V.K.); (S.P.)
| | - Saurabh Gupta
- Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, Chaumuhan, Mathura 281406, Uttar Pradesh, India; (R.K.); (S.K.); (V.K.); (S.P.)
| | - Sunanda Kulshrestha
- Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, Chaumuhan, Mathura 281406, Uttar Pradesh, India; (R.K.); (S.K.); (V.K.); (S.P.)
| | - Vishal Khandelwal
- Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, Chaumuhan, Mathura 281406, Uttar Pradesh, India; (R.K.); (S.K.); (V.K.); (S.P.)
| | - Swadha Pandey
- Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, Chaumuhan, Mathura 281406, Uttar Pradesh, India; (R.K.); (S.K.); (V.K.); (S.P.)
- Division of Hematology & Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA;
| | - Anil Kumar
- National Institute of Immunology, New Delhi 110067, India;
| | - Gaurav Sharma
- Cardiovascular and Thoracic Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
- Advanced Imaging Research Center (AIRC), University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Biomedical Engineering, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Umesh Kumar
- Department of Biosciences, Institute of Management Studies Ghaziabad (University Courses Campus), Ghaziabad 201015, Uttar Pradesh, India;
| | - Deepak Parashar
- Division of Hematology & Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA;
| | - Kaushik Das
- Biotechnology Research and Innovation Council-National Institute of Biomedical Genomics, Kalyani 741251, West Bengal, India
| |
Collapse
|
3
|
Zhu R, Ni J, Ren J, Li D, Xu J, Yu X, Ma YJ, Kou L. Transcriptomic era of cancers in females: new epigenetic perspectives and therapeutic prospects. Front Oncol 2024; 14:1464125. [PMID: 39605897 PMCID: PMC11598703 DOI: 10.3389/fonc.2024.1464125] [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: 07/13/2024] [Accepted: 10/16/2024] [Indexed: 11/29/2024] Open
Abstract
In the era of transcriptomics, the role of epigenetics in the study of cancers in females has gained increasing recognition. This article explores the impact of epigenetic modifications, such as DNA methylation, histone modification, and non-coding RNA, on cancers in females, including breast, cervical, and ovarian cancers (1). Our findings suggest that these epigenetic markers not only influence tumor onset, progression, and metastasis but also present novel targets for therapeutic intervention. Detailed analyses of DNA methylation patterns have revealed aberrant events in cancer cells, particularly promoter region hypermethylation, which may lead to silencing of tumor suppressor genes. Furthermore, we examined the complex roles of histone modifications and long non-coding RNAs in regulating the expression of cancer-related genes, thereby providing a scientific basis for developing targeted epigenetic therapies. Our research emphasizes the importance of understanding the functions and mechanisms of epigenetics in cancers in females to develop effective treatment strategies. Future therapeutic approaches may include drugs targeting specific epigenetic markers, which could not only improve therapeutic outcomes but also enhance patient survival and quality of life. Through these efforts, we aim to offer new perspectives and hope for the prevention, diagnosis, and treatment of cancers in females.
Collapse
Affiliation(s)
- Runhe Zhu
- The Traditional Chinese Medicine College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jiawei Ni
- The Traditional Chinese Medicine College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jiayin Ren
- The Traditional Chinese Medicine College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Dongye Li
- The Traditional Chinese Medicine College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jiawei Xu
- The Traditional Chinese Medicine College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xinru Yu
- The Pharmacy College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ying Jie Ma
- The First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Luan Kou
- Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, China
| |
Collapse
|
4
|
Kalra R, Lim B, Ellis MJ, Kavuri SM. The uncharted role of HER2 mutant alleles in breast cancer. Oncotarget 2023; 14:904-907. [PMID: 37921670 PMCID: PMC10624202 DOI: 10.18632/oncotarget.28489] [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: 07/12/2023] [Indexed: 11/04/2023] Open
Abstract
Somatic HER2 mutations are a novel class of therapeutic targets across different cancer types. Treatment with the tyrosine kinase inhibitor (TKI) neratinib as a single agent continues to be evaluated in HER2-mutant metastatic disease. However, responses are heterogeneous, with frequent early progression. Herein, we discuss the under-explored effects of individual HER2 mutant alleles on therapeutic response, a role for HER2 mutation in metastatic propensity, and differences in patient outcomes in ER+ invasive lobular carcinoma (ILC) versus invasive ductal carcinoma (IDC). The preclinical efficacy of additional agents is also discussed, particularly the pan-HER inhibitor poziotinib.
Collapse
Affiliation(s)
| | | | | | - Shyam M. Kavuri
- Correspondence to:Shyam M. Kavuri, Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA email
| |
Collapse
|
5
|
Neagu AN, Whitham D, Bruno P, Morrissiey H, Darie CA, Darie CC. Omics-Based Investigations of Breast Cancer. Molecules 2023; 28:4768. [PMID: 37375323 DOI: 10.3390/molecules28124768] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/08/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
Breast cancer (BC) is characterized by an extensive genotypic and phenotypic heterogeneity. In-depth investigations into the molecular bases of BC phenotypes, carcinogenesis, progression, and metastasis are necessary for accurate diagnoses, prognoses, and therapy assessments in predictive, precision, and personalized oncology. This review discusses both classic as well as several novel omics fields that are involved or should be used in modern BC investigations, which may be integrated as a holistic term, onco-breastomics. Rapid and recent advances in molecular profiling strategies and analytical techniques based on high-throughput sequencing and mass spectrometry (MS) development have generated large-scale multi-omics datasets, mainly emerging from the three "big omics", based on the central dogma of molecular biology: genomics, transcriptomics, and proteomics. Metabolomics-based approaches also reflect the dynamic response of BC cells to genetic modifications. Interactomics promotes a holistic view in BC research by constructing and characterizing protein-protein interaction (PPI) networks that provide a novel hypothesis for the pathophysiological processes involved in BC progression and subtyping. The emergence of new omics- and epiomics-based multidimensional approaches provide opportunities to gain insights into BC heterogeneity and its underlying mechanisms. The three main epiomics fields (epigenomics, epitranscriptomics, and epiproteomics) are focused on the epigenetic DNA changes, RNAs modifications, and posttranslational modifications (PTMs) affecting protein functions for an in-depth understanding of cancer cell proliferation, migration, and invasion. Novel omics fields, such as epichaperomics or epimetabolomics, could investigate the modifications in the interactome induced by stressors and provide PPI changes, as well as in metabolites, as drivers of BC-causing phenotypes. Over the last years, several proteomics-derived omics, such as matrisomics, exosomics, secretomics, kinomics, phosphoproteomics, or immunomics, provided valuable data for a deep understanding of dysregulated pathways in BC cells and their tumor microenvironment (TME) or tumor immune microenvironment (TIMW). Most of these omics datasets are still assessed individually using distinct approches and do not generate the desired and expected global-integrative knowledge with applications in clinical diagnostics. However, several hyphenated omics approaches, such as proteo-genomics, proteo-transcriptomics, and phosphoproteomics-exosomics are useful for the identification of putative BC biomarkers and therapeutic targets. To develop non-invasive diagnostic tests and to discover new biomarkers for BC, classic and novel omics-based strategies allow for significant advances in blood/plasma-based omics. Salivaomics, urinomics, and milkomics appear as integrative omics that may develop a high potential for early and non-invasive diagnoses in BC. Thus, the analysis of the tumor circulome is considered a novel frontier in liquid biopsy. Omics-based investigations have applications in BC modeling, as well as accurate BC classification and subtype characterization. The future in omics-based investigations of BC may be also focused on multi-omics single-cell analyses.
Collapse
Affiliation(s)
- Anca-Narcisa Neagu
- Laboratory of Animal Histology, Faculty of Biology, "Alexandru Ioan Cuza" University of Iasi, Carol I Bvd, No. 20A, 700505 Iasi, Romania
| | - Danielle Whitham
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, 8 Clarkson Avenue, Potsdam, NY 13699, USA
| | - Pathea Bruno
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, 8 Clarkson Avenue, Potsdam, NY 13699, USA
| | - Hailey Morrissiey
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, 8 Clarkson Avenue, Potsdam, NY 13699, USA
| | - Celeste A Darie
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, 8 Clarkson Avenue, Potsdam, NY 13699, USA
| | - Costel C Darie
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, 8 Clarkson Avenue, Potsdam, NY 13699, USA
| |
Collapse
|
6
|
Kalra R, Chen CH, Wang J, Salam AB, Dobrolecki LE, Lewis A, Sallas C, Yates CC, Gutierrez C, Karanam B, Anurag M, Lim B, Ellis MJ, Kavuri SM. Poziotinib Inhibits HER2-Mutant-Driven Therapeutic Resistance and Multiorgan Metastasis in Breast Cancer. Cancer Res 2022; 82:2928-2939. [PMID: 35736563 PMCID: PMC9379360 DOI: 10.1158/0008-5472.can-21-3106] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 04/01/2022] [Accepted: 06/14/2022] [Indexed: 01/07/2023]
Abstract
The pan-HER tyrosine kinase inhibitor (TKI) neratinib is therapeutically active against metastatic breast cancers harboring activating HER2 mutations, but responses are variable and often not durable. Here we demonstrate that recurrent HER2 mutations have differential effects on endocrine therapy responsiveness, metastasis, and pan-HER TKI therapeutic sensitivity. The prevalence and prognostic significance may also depend on whether the HER2 mutant has arisen in the context of lobular versus ductal histology. The most highly recurrent HER2 mutant, L755S, was particularly resistant to neratinib but sensitive to the pan-HER TKI poziotinib, alone or in combination with fulvestrant. Poziotinib reduced tumor growth, diminished multiorgan metastasis, and inhibited mTOR activation more effectively than neratinib. Similar therapeutic effects of poziotinib were observed in both an engineered HER2L755S MCF7 model and a patient-derived xenograft harboring a HER2G778_P780dup mutation. Overall, these findings support the need for clinical evaluation of poziotinib for the treatment of HER2-mutant metastatic breast cancer. SIGNIFICANCE Evaluation of the functional impact of HER2 mutations on therapy-induced resistance and metastasis identifies robust antitumor activity of poziotinib and supports the clinical evaluation of poziotinib in ER+ HER2 mutant breast cancer.
Collapse
Affiliation(s)
- Rashi Kalra
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Ching Hui Chen
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Junkai Wang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Ahmad Bin Salam
- Department of Biology and Center for Cancer Research, Tuskegee University, Tuskegee, Alabama
| | - Lacey E. Dobrolecki
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Alaina Lewis
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Christina Sallas
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Clayton C. Yates
- Department of Biology and Center for Cancer Research, Tuskegee University, Tuskegee, Alabama
| | - Carolina Gutierrez
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Balasubramanyam Karanam
- Department of Biology and Center for Cancer Research, Tuskegee University, Tuskegee, Alabama
| | - Meenakshi Anurag
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Bora Lim
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Matthew J. Ellis
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas.,Corresponding Authors: Shyam M. Kavuri, Lester and Sue Smith Breast Center, Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030. Phone: 1314-651-3876; E-mail: ; and Matthew J. Ellis, Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030. E-mail:
| | - Shyam M. Kavuri
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas.,Corresponding Authors: Shyam M. Kavuri, Lester and Sue Smith Breast Center, Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030. Phone: 1314-651-3876; E-mail: ; and Matthew J. Ellis, Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030. E-mail:
| |
Collapse
|
7
|
Ghafouri-Fard S, Asadi M, Sohrabi B, Arsang-Jang S, Mehravaran E, Taheri M, Samsami M. Down-regulation of a panel of immune-related lncRNAs in breast cancer. Pathol Res Pract 2021; 224:153534. [PMID: 34175685 DOI: 10.1016/j.prp.2021.153534] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 06/16/2021] [Accepted: 06/19/2021] [Indexed: 10/21/2022]
Abstract
Breast cancer is a common neoplasm among women. This type of cancer is among malignancies in which role of long non-coding RNAs (lncRNAs) has been extensively explored. Some recently recognized lncRNAs have been less investigated in this neoplastic condition. LncRNAs that regulate tumor immunity are among those contributing in the pathogenesis of cancer. In the present expression assay, we compared expressions of nine immune-related lncRNAs namely lnc-MICAL3-2 (AC016027.1), lnc-DDX31 (AL445645.1), LINC01063, LINC02381, ENST0000615051 (AC083809.1), AC009237.14 (lnc-TRIM43B-1), ENST0000603791, LINC1234 and AC008760.1 between breast cancer samples and their paired non-cancerous samples. Expression levels of lnc-MICAL3-2, lnc-DDX31, LINC01063, LINC02381, ENST0000615051 and lnc-TRIM43B-1 were significantly decreased in breast cancer samples compared with paired control tissues (Posterior mean difference= -2.774, -2.012, -2.012, -2.015, -0.884 and -2.872; P values= 0.019, 0.0001, 0.0001, 0.0001, 0.032 and 0.0001, respectively). Expression levels of these lncRNAs have been associated with a number of clinical characteristics of breast cancer patients. Lnc-TRIM43B-1 had the highest performance in distinguishing between tumoral and non-tumoral tissues (AUC=0.82, Sensitivity=76%, Specificity=73.24%). As these lncRNAs could differentiate tumor samples from control samples, they might be regarded as putative tissue markers for breast cancer.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Asadi
- Department of Biology, Islamic Azad University, Urmia Branch, Urmia, Iran
| | - Behnoush Sohrabi
- Department of Biology, Faculty of Sciences, Arak University, Arak, Iran
| | - Shahram Arsang-Jang
- Cancer Gene therapy Research Center, Zanjan University of Medical Science, Zanjan, Iran
| | - Elham Mehravaran
- Motamed Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Taheri
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Majid Samsami
- Department of Surgery, Imam Hossein Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
8
|
Zhang Y, Tian J, Qu C, Peng Y, Lei J, Sun L, Zong B, Liu S. A look into the link between centrosome amplification and breast cancer. Biomed Pharmacother 2020; 132:110924. [PMID: 33128942 DOI: 10.1016/j.biopha.2020.110924] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/18/2020] [Accepted: 10/20/2020] [Indexed: 02/06/2023] Open
Abstract
Centrosome amplification (CA) is a common feature of human tumors, but it is not clear whether this is a cause or a consequence of cancer. The centrosome amplification observed in tumor cells may be explained by a series of events, such as failure of cell division, dysregulation of centrosome cycle checkpoints, and de novo centriole biogenesis disorder. The formation and progression of breast cancer are characterized by genomic abnormality. The centrosomes in breast cancer cells show characteristic structural aberrations, caused by centrosome amplification, which include: an increase in the number and volume of centrosomes, excessive increase of pericentriolar material (PCM), inappropriate phosphorylation of centrosomal molecular, and centrosome clustering formation induced by the dysregulation of important genes. The mechanism of intracellular centrosome amplification, the impact of which on breast cancer and the latest breast cancer target treatment options for centrosome amplification are exhaustively elaborated in this review.
Collapse
Affiliation(s)
- Yingzi Zhang
- Department of Endocrine Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, 1 Yixueyuan Road, Yuanjiagang, Yuzhong District, Chongqing, 400016, China.
| | - Jiao Tian
- Department of Endocrine Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, 1 Yixueyuan Road, Yuanjiagang, Yuzhong District, Chongqing, 400016, China.
| | - Chi Qu
- Department of Endocrine Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, 1 Yixueyuan Road, Yuanjiagang, Yuzhong District, Chongqing, 400016, China.
| | - Yang Peng
- Department of Endocrine Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, 1 Yixueyuan Road, Yuanjiagang, Yuzhong District, Chongqing, 400016, China.
| | - Jinwei Lei
- Department of Endocrine Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, 1 Yixueyuan Road, Yuanjiagang, Yuzhong District, Chongqing, 400016, China.
| | - Lu Sun
- Department of Endocrine Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, 1 Yixueyuan Road, Yuanjiagang, Yuzhong District, Chongqing, 400016, China.
| | - Beige Zong
- Department of Endocrine Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, 1 Yixueyuan Road, Yuanjiagang, Yuzhong District, Chongqing, 400016, China.
| | - Shengchun Liu
- Department of Endocrine Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, 1 Yixueyuan Road, Yuanjiagang, Yuzhong District, Chongqing, 400016, China.
| |
Collapse
|
9
|
Buhagiar A, Borg J, Ayers D. Overview of current microRNA biomarker signatures as potential diagnostic tools for leukaemic conditions. Noncoding RNA Res 2020; 5:22-26. [PMID: 32110743 PMCID: PMC7033436 DOI: 10.1016/j.ncrna.2020.02.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 02/07/2020] [Accepted: 02/08/2020] [Indexed: 02/06/2023] Open
Abstract
Haematological malignancies encompass all variations of leukaemia at both the chronic and acute level, together with the specific cell type induced into tumourigenesis. Current diagnostic protocols for leukaemic conditions rely heavily on cytomorphology and other histological examinations from bone marrow aspirates, with the latter being a highly invasive surgical procedure for the patient. The discovery of microRNAs as one of the key gene regulatory networks in the past two decades has enabled researchers to investigate the possibility of exploiting the identification of dysregulated expression profiles for specific microRNAs present in the leukaemic patient's bloodstream as novel liquid biopsy diagnostic tools. This review article serves to consolidate recent global research efforts aiming to achieve such scopes.
Collapse
Affiliation(s)
- Alfred Buhagiar
- Faculty of Medicine and Surgery, University of Malta, Msida, MSD 2080, Malta
| | - Joseph Borg
- Faculty of Health Sciences, University of Malta, Msida, MSD 2080, Malta
| | - Duncan Ayers
- Centre for Molecular Medicine and Biobanking, University of Malta, Msida, MSD2080, Malta
- Faculty of Biology, Medicine and Health Sciences, The University of Manchester, Manchester, M13 9PL, United Kingdom
| |
Collapse
|
10
|
Genetic and Genomic Advances in Breast Cancer Diagnosis and Treatment. Nurs Womens Health 2019; 23:518-525. [PMID: 31669147 DOI: 10.1016/j.nwh.2019.09.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 06/18/2019] [Accepted: 09/01/2019] [Indexed: 02/06/2023]
Abstract
Advances in genetic testing for people at high risk for cancer and in targeted gene therapy for breast cancer are rapidly emerging, including newly developed key hormone receptor-targeted therapies and individualized molecular fusion identification and treatment options. These advances are contributing to a new era in cancer treatment modalities and care delivery. As more innovative and advanced treatment options emerge, women's health outcomes and survival rates may improve. Nursing professionals in primary care and women's health specialties must be aware of the latest options for testing, referrals, and treatment modalities.
Collapse
|
11
|
Cao L, Basudan A, Sikora MJ, Bahreini A, Tasdemir N, Levine KM, Jankowitz RC, McAuliffe PF, Dabbs D, Haupt S, Haupt Y, Lucas PC, Lee AV, Oesterreich S, Atkinson JM. Frequent amplifications of ESR1, ERBB2 and MDM4 in primary invasive lobular breast carcinoma. Cancer Lett 2019; 461:21-30. [PMID: 31229512 PMCID: PMC6682463 DOI: 10.1016/j.canlet.2019.06.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 06/02/2019] [Accepted: 06/17/2019] [Indexed: 01/09/2023]
Abstract
Invasive lobular carcinoma (ILC) is the second most common histological subtype of breast cancer following invasive ductal carcinoma (IDC). To identify potential genetic drivers of ILC progression, we used NanoString nCounter technology to investigate the DNA copy number (CN) in 70 well-curated primary ILC samples. We confirmed prior observations of frequent amplification of CCND1 (33%), and MYC (17%) in ILC, but additionally identified a substantial subset of ILCs with ESR1 and ERBB2 (19%) amplifications. Of interest, tumors with ESR1 CN gains (14%) and amplification (10%) were more likely to recur compared to those with normal CN. Finally, we observed that MDM4 (MDMX) was amplified in 17% of ILC samples. MDM4 knockdown in TP53 wild-type ILC cell lines caused increased apoptosis, decreased proliferation associated with cell cycle arrest, and concomitant activation of TP53 target genes. Similar effects were seen in TP53 mutant cells, indicting a TP53-independent role for MDM4 in ILC. To conclude, amplification of ESR1 and MDM4 are potential genetic drivers of ILC. These amplifications may represent actionable, targetable tumor dependencies, and thus have potential clinical implications and warrant further study.
Collapse
MESH Headings
- Apoptosis
- Biomarkers, Tumor
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Carcinoma, Ductal, Breast/genetics
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Lobular/genetics
- Carcinoma, Lobular/metabolism
- Carcinoma, Lobular/pathology
- Cell Cycle Checkpoints
- Cell Cycle Proteins/genetics
- Cell Cycle Proteins/metabolism
- Cell Proliferation
- DNA Copy Number Variations
- Estrogen Receptor alpha/genetics
- Female
- Follow-Up Studies
- Gene Amplification
- Gene Expression Regulation, Neoplastic
- Humans
- Middle Aged
- Neoplasm Invasiveness
- Neoplasm Recurrence, Local/genetics
- Neoplasm Recurrence, Local/metabolism
- Neoplasm Recurrence, Local/pathology
- Prognosis
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/metabolism
- Receptor, ErbB-2/genetics
- Retrospective Studies
- Survival Rate
- Tumor Cells, Cultured
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/metabolism
Collapse
Affiliation(s)
- Lan Cao
- Women’s Cancer Research Center, Magee-Women Research Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Obstetrics and Gynecology, the Third Xiangya Hospital, Central South University, Changsha, China
| | - Ahmed Basudan
- Women’s Cancer Research Center, Magee-Women Research Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Clinical Laboratory Sciences, King Saud University, Saudi Arabia
| | - Matthew J Sikora
- Women’s Cancer Research Center, Magee-Women Research Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Amir Bahreini
- Women’s Cancer Research Center, Magee-Women Research Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Genetics and Molecular Biology; School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nilgun Tasdemir
- Women’s Cancer Research Center, Magee-Women Research Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology; University of Pittsburgh, Pittsburgh, PA, USA
| | - Kevin M Levine
- Women’s Cancer Research Center, Magee-Women Research Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Rachel C. Jankowitz
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Medicine, Division of Hematology Oncology; University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Priscilla F McAuliffe
- Women’s Cancer Research Center, Magee-Women Research Institute, University of Pittsburgh, Pittsburgh, PA, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Division of Surgical Oncology, Department of Surgery, Pittsburgh, PA
| | - David Dabbs
- Division of Breast and Gynecologic Pathology, Department of Pathology, Pittsburgh, PA
| | - Sue Haupt
- Peter MacCallum Cancer Center, Melbourne, Australia
| | - Ygal Haupt
- Peter MacCallum Cancer Center, Melbourne, Australia
| | - Peter C. Lucas
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Adrian V Lee
- Women’s Cancer Research Center, Magee-Women Research Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology; University of Pittsburgh, Pittsburgh, PA, USA
| | - Steffi Oesterreich
- Women’s Cancer Research Center, Magee-Women Research Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology; University of Pittsburgh, Pittsburgh, PA, USA
| | - Jennifer M Atkinson
- Women’s Cancer Research Center, Magee-Women Research Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology; University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
12
|
Wang W, Wu J, Fei X, Chen W, Li Y, Shen K, Zhu L. CHD1L promotes cell cycle progression and cell motility by up-regulating MDM2 in breast cancer. Am J Transl Res 2019; 11:1581-1592. [PMID: 30972184 PMCID: PMC6456556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 01/14/2019] [Indexed: 06/09/2023]
Abstract
Chromodomain helicase/ATPase DNA-binding protein 1-like (CHD1L) gene is a novel oncogene amplified in many solid tumors. We investigated its role in breast cancer. CHD1L was over-expressed in 49.1% (57/116) breast cancer patients. Overexpression of CHD1L was significantly associated with younger age at diagnosis (P = 0.016), lymph node involvement (P = 0.040), higher tumor grade (P = 0.027), higher proliferation rate (P = 0.007) and shorter disease-free survival rate (77.2% vs. 91.5%, P = 0.037). A cDNA microarray analysis identified MDM2 as an important downstream target of CHD1L. And MDM2/p53 signaling pathway was showed to be significantly modulated by CHD1L. Further in vitro study showed that overexpression of CHD1L can promote tumorigenesis, metastasis, invasion and cell cycle progress. In vivo study confirmed the tumorigenesis ability of CHD1L. shRNA-mediated CHD1L silencing could abolishes the tumor-promotion effect of CHD1L in vitro and in vivo. In conclusion, CHD1L may promote the progress of breast cancer cells via the MDM2/p53 signaling pathway. This study identified CHD1L as a prognostic factor for breast cancer and MDM2 might be used as a potential target for therapeutic intervention in CHD1L overexpression breast cancer.
Collapse
Affiliation(s)
- Wei Wang
- Comprehensive Breast Health Center, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University197 Ruijin Second Road, Shanghai 200025, P. R. China
| | - Jiayi Wu
- Comprehensive Breast Health Center, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University197 Ruijin Second Road, Shanghai 200025, P. R. China
| | - Xiaochun Fei
- Pathology Department, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University197 Ruijin Second Road, Shanghai 200025, P. R. China
| | - Weiguo Chen
- Comprehensive Breast Health Center, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University197 Ruijin Second Road, Shanghai 200025, P. R. China
| | - Yafen Li
- Comprehensive Breast Health Center, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University197 Ruijin Second Road, Shanghai 200025, P. R. China
| | - Kunwei Shen
- Comprehensive Breast Health Center, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University197 Ruijin Second Road, Shanghai 200025, P. R. China
| | - Li Zhu
- Comprehensive Breast Health Center, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University197 Ruijin Second Road, Shanghai 200025, P. R. China
| |
Collapse
|
13
|
Cheasley D, Li N, Rowley SM, Elder K, Mann GB, Loi S, Savas P, Goode DL, Kader T, Zethoven M, Semple T, Fox SB, Pang JM, Byrne D, Devereux L, Nickson C, Procopio P, Lee G, Hughes S, Saunders H, Fujihara KM, Kuykhoven K, Connaughton J, James PA, Gorringe KL, Campbell IG. Molecular comparison of interval and screen-detected breast cancers. J Pathol 2019; 248:243-252. [PMID: 30746706 DOI: 10.1002/path.5251] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 01/15/2019] [Accepted: 01/30/2019] [Indexed: 01/22/2023]
Abstract
Breast cancer (BC) diagnosed after a negative mammogram but prior to the next screening episode is termed an 'interval BC' (IBC). Understanding the molecular differences between IBC and screen-detected BCs (SDBC) could improve mammographic screening and management options. Therefore, we assessed both germline and somatic genomic aberrations in a prospective cohort. Utilising the Lifepool cohort of >54 000 women attending mammographic screening programs, 930 BC cases with screening status were identified (726 SDBC and 204 IBC). Clinico-pathological and family history information were recorded. Germline and tumour DNA were collected where available and sequenced for BC predisposition and driver gene mutations. Compared to SDBC, IBCs were significantly associated with a younger age at diagnosis and tumour characteristics associated with worse prognosis. Germline DNA assessment of BC cases that developed post-enrolment (276 SDBCs and 77 IBCs) for pathogenic mutations in 12 hereditary BC predisposition genes identified 8 carriers (2.27%). The germline mutation frequency was higher in IBC versus SDBC, although not statistically significant (3.90% versus 1.81%, p = 0.174). Comparing somatic genetic features of IBC and SDBC matched for grade, histological subtype and hormone receptor revealed no significant differences, with the exception of higher homologous recombination deficiency scores in IBC, and copy number changes on chromosome Xq in triple negative SDBCs. Our data demonstrates that while IBCs are clinically more aggressive than SDBC, when matched for confounding clinico-pathological features they do not represent a unique molecular class of invasive BC, but could be a consequence of timing of tumour initiation and mammographic screening. Copyright © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Dane Cheasley
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Na Li
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Simone M Rowley
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Kenneth Elder
- Department of Surgery, University of Melbourne, Melbourne, Victoria, Australia.,The Royal Melbourne and Royal Women's Hospitals, Parkville, Victoria, Australia.,The Edinburgh Breast Unit, Western General Hospital, Edinburgh, UK
| | - G Bruce Mann
- Department of Surgery, University of Melbourne, Melbourne, Victoria, Australia.,The Royal Melbourne and Royal Women's Hospitals, Parkville, Victoria, Australia
| | - Sherene Loi
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia.,Division of Clinical Medicine and Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Peter Savas
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia.,Division of Clinical Medicine and Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - David L Goode
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Tanjina Kader
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Magnus Zethoven
- Bioinformatics Consulting Core, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Tim Semple
- Genomics Core, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Stephen B Fox
- Department of Pathology, Peter MacCallum Cancer Centre, and University of Melbourne, Melbourne, Victoria, Australia
| | - Jia-Min Pang
- Department of Pathology, Peter MacCallum Cancer Centre, and University of Melbourne, Melbourne, Victoria, Australia
| | - David Byrne
- Department of Pathology, Peter MacCallum Cancer Centre, and University of Melbourne, Melbourne, Victoria, Australia
| | - Lisa Devereux
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Lifepool, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Carolyn Nickson
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Pietro Procopio
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Grant Lee
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Siobhan Hughes
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Hugo Saunders
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Kenji M Fujihara
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Keilly Kuykhoven
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Jacquie Connaughton
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Paul A James
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia.,Parkville Familial Cancer Centre, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Kylie L Gorringe
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia.,Cancer Genetics and Genomics Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Ian G Campbell
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
14
|
Inhibition of mTOR downregulates expression of DNA repair proteins and is highly efficient against BRCA2-mutated breast cancer in combination to PARP inhibition. Oncotarget 2018; 9:29587-29600. [PMID: 30038706 PMCID: PMC6049870 DOI: 10.18632/oncotarget.25640] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 06/01/2018] [Indexed: 11/25/2022] Open
Abstract
Breast cancer is a complex disease in which each patient could present several genetic alterations that are therapeutically relevant in cancers. Here we explored the therapeutic benefit of combining PARP and mTOR inhibitors in a context of DNA repair deficiency and PI3K pathway activation. The combination of everolimus and olaparib was tested in BRCA2-mutated patient-derived xenografts (PDX) carrying alterations in the PI3K/AKT/mTOR pathway. An RPPA analysis of different signalling pathways was performed in untreated and treated xenografts. Everolimus and olaparib showed marked anti-tumor activities in the monotherapy setting and high efficacy when given in combination with 100% of mice showing tumor regressions. The fraction of P-H2AX positive cells was increased in both monotherapy arms and strongly increased in the combination setting. Everolimus given as monotherapy resulted in downregulation of different proteins involved in DNA damage repair, including FANCD2, RAD50 and SUV39H1. In the combination setting, expression of these proteins was almost completely abolished, suggesting convergence of PARP and mTOR in downregulation of DNA damage repair components. In conclusion, our results suggest that combining mTOR and DNA repair inhibition could be a successful strategy to treat a subset of breast cancer with BRCA2 mutation and alterations in the PI3K/AKT/mTOR pathway.
Collapse
|
15
|
Ogden A, Rida PCG, Aneja R. Centrosome amplification: a suspect in breast cancer and racial disparities. Endocr Relat Cancer 2017; 24:T47-T64. [PMID: 28515047 PMCID: PMC5837860 DOI: 10.1530/erc-17-0072] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 05/17/2017] [Indexed: 12/31/2022]
Abstract
The multifaceted involvement of centrosome amplification (CA) in tumorigenesis is coming into focus following years of meticulous experimentation, which have elucidated the powerful abilities of CA to promote cellular invasion, disrupt stem cell division, drive chromosomal instability (CIN) and perturb tissue architecture, activities that can accelerate tumor progression. Integration of the extant in vitro, in vivo and clinical data suggests that in some tissues CA may be a tumor-initiating event, in others a consequential 'hit' in multistep tumorigenesis, and in some others, non-tumorigenic. However, in vivo data are limited and primarily focus on PLK4 (which has CA-independent mechanisms by which it promotes aggressive cellular phenotypes). In vitro breast cancer models suggest that CA can promote tumorigenesis in breast cancer cells in the setting of p53 loss or mutation, which can both trigger CA and promote cellular tolerance to its tendency to slow proliferation and induce aneuploidy. It is thus our perspective that CA is likely an early hit in multistep breast tumorigenesis that may sometimes be lost to preserve aggressive karyotypes acquired through centrosome clustering-mediated CIN, both numerical and structural. We also envision that the robust link between p53 and CA may underlie, to a considerable degree, racial health disparity in breast cancer outcomes. This question is clinically significant because, if it is true, then analysis of centrosomal profiles and administration of centrosome declustering drugs could prove highly efficacious in risk stratifying breast cancers and treating African American (AA) women with breast cancer.
Collapse
Affiliation(s)
- Angela Ogden
- Department of BiologyGeorgia State University, Atlanta, Georgia, USA
| | | | - Ritu Aneja
- Department of BiologyGeorgia State University, Atlanta, Georgia, USA
| |
Collapse
|
16
|
Wu HC, Southey MC, Hibshoosh H, Santella RM, Terry MB. DNA Methylation in Breast Tumor from High-risk Women in the Breast Cancer Family Registry. Anticancer Res 2017; 37:659-664. [PMID: 28179314 DOI: 10.21873/anticanres.11361] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 01/13/2017] [Accepted: 01/17/2017] [Indexed: 01/22/2023]
Abstract
To examine DNA methylation profiles in breast tumors of women with a strong breast cancer family history, we measured methylation by bisulfite sequencing in 40 genes in 40 breast tumor tissues from women in the Breast Cancer Family Registry. We selected candidate genes from analysis of the Cancer Genome Atlas project (TCGA) breast data. Compared to TCGA breast cancer, BCFR cases are younger and more likely to be ER-negative. Overall, we found that many of the methylation differences between BCFR tumor and normal adjacent tissues were smaller than that in TCGA samples. We found only 32% of tested genes were hypermethylated in BCFR; the largest difference was 36.1% for SEPW1, and the smallest difference was 10% for RYR2. These data suggest the importance of examining breast cancer cases including familial cases enriched with early-onset cancers to identify methylation markers that can be examined in blood as biomarkers for early detection.
Collapse
Affiliation(s)
- Hui-Chen Wu
- Department of Environmental Health Sciences, Mailman School of Public Health of Columbia University, New York, NY, U.S.A.
| | - Melissa C Southey
- Department of Pathology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Hanina Hibshoosh
- Department of Pathology and Cell Biology, College of Physicians & Surgeons of Columbia University, New York, NY, U.S.A
| | - Regina M Santella
- Department of Environmental Health Sciences, Mailman School of Public Health of Columbia University, New York, NY, U.S.A.,Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY, U.S.A
| | - Mary Beth Terry
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY, U.S.A.,Department of Epidemiology, Mailman School of Public Health of Columbia University, New York, NY, U.S.A
| |
Collapse
|
17
|
Silwal-Pandit L, Langerød A, Børresen-Dale AL. TP53 Mutations in Breast and Ovarian Cancer. Cold Spring Harb Perspect Med 2017; 7:cshperspect.a026252. [PMID: 27815305 DOI: 10.1101/cshperspect.a026252] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Breast and ovarian cancers are the second and fifth leading causes of cancer deaths among women. Both breast and ovarian cancers are highly heterogeneous and are presented with diverse morphology, natural history, and response to therapy. In recent years, international efforts have led to extensive molecular characterization of both breast and ovarian tumors and identified biologically and clinically relevant subtypes of the diseases based on these molecular features. The role of TP53 in tumor initiation and progression is context dependent, and abrogation of the TP53 pathway seems to be essential for the development of basal-like breast cancers and high-grade serous ovarian cancers. These subtypes of breast and ovarian cancer show several genomic similarities including high frequency of TP53 mutation, which seems to be an early, initiating, and driving alteration in these cancer subtypes.
Collapse
Affiliation(s)
- Laxmi Silwal-Pandit
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital The Norwegian Radium Hospital, Montebello, 0310 Oslo, Norway
| | - Anita Langerød
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital The Norwegian Radium Hospital, Montebello, 0310 Oslo, Norway
| | - Anne-Lise Børresen-Dale
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital The Norwegian Radium Hospital, Montebello, 0310 Oslo, Norway
| |
Collapse
|
18
|
Couto JP, Bentires-Alj M. Mouse Models of Breast Cancer: Deceptions that Reveal the Truth. Breast Cancer 2017. [DOI: 10.1007/978-3-319-48848-6_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
19
|
Desmedt C, Zoppoli G, Gundem G, Pruneri G, Larsimont D, Fornili M, Fumagalli D, Brown D, Rothé F, Vincent D, Kheddoumi N, Rouas G, Majjaj S, Brohée S, Van Loo P, Maisonneuve P, Salgado R, Van Brussel T, Lambrechts D, Bose R, Metzger O, Galant C, Bertucci F, Piccart-Gebhart M, Viale G, Biganzoli E, Campbell PJ, Sotiriou C. Genomic Characterization of Primary Invasive Lobular Breast Cancer. J Clin Oncol 2016; 34:1872-81. [DOI: 10.1200/jco.2015.64.0334] [Citation(s) in RCA: 194] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Purpose Invasive lobular breast cancer (ILBC) is the second most common histologic subtype after invasive ductal breast cancer (IDBC). Despite clinical and pathologic differences, ILBC is still treated as IDBC. We aimed to identify genomic alterations in ILBC with potential clinical implications. Methods From an initial 630 ILBC primary tumors, we interrogated oncogenic substitutions and insertions and deletions of 360 cancer genes and genome-wide copy number aberrations in 413 and 170 ILBC samples, respectively, and correlated those findings with clinicopathologic and outcome features. Results Besides the high mutation frequency of CDH1 in 65% of tumors, alterations in one of the three key genes of the phosphatidylinositol 3-kinase pathway, PIK3CA, PTEN, and AKT1, were present in more than one-half of the cases. HER2 and HER3 were mutated in 5.1% and 3.6% of the tumors, with most of these mutations having a proven role in activating the human epidermal growth factor receptor/ERBB pathway. Mutations in FOXA1 and ESR1 copy number gains were detected in 9% and 25% of the samples. All these alterations were more frequent in ILBC than in IDBC. The histologic diversity of ILBC was associated with specific alterations, such as enrichment for HER2 mutations in the mixed, nonclassic, and ESR1 gains in the solid subtype. Survival analyses revealed that chromosome 1q and 11p gains showed independent prognostic value in ILBC and that HER2 and AKT1 mutations were associated with increased risk of early relapse. Conclusion This study demonstrates that we can now begin to individualize the treatment of ILBC, with HER2, HER3, and AKT1 mutations representing high-prevalence therapeutic targets and FOXA1 mutations and ESR1 gains deserving urgent dedicated clinical investigation, especially in the context of endocrine treatment.
Collapse
Affiliation(s)
- Christine Desmedt
- Christine Desmedt, Gabriele Zoppoli, Denis Larsimont, Debora Fumagalli, David Brown, Françoise Rothé, Delphine Vincent, Naima Kheddoumi, Ghizlane Rouas, Samira Majjaj, Sylvain Brohée, Roberto Salgado, Martine Piccart-Gebhart, and Christos Sotiriou, Institut Jules Bordet; Christine Galant, Cliniques Universitaires Saint Luc, Brussels; Peter Van Loo, University of Leuven; Thomas Van Brussel and Diether Lambrechts, VIB Vesalius Research Center, Leuven, Belgium; Gabriele Zoppoli, University of Genoa and
| | - Gabriele Zoppoli
- Christine Desmedt, Gabriele Zoppoli, Denis Larsimont, Debora Fumagalli, David Brown, Françoise Rothé, Delphine Vincent, Naima Kheddoumi, Ghizlane Rouas, Samira Majjaj, Sylvain Brohée, Roberto Salgado, Martine Piccart-Gebhart, and Christos Sotiriou, Institut Jules Bordet; Christine Galant, Cliniques Universitaires Saint Luc, Brussels; Peter Van Loo, University of Leuven; Thomas Van Brussel and Diether Lambrechts, VIB Vesalius Research Center, Leuven, Belgium; Gabriele Zoppoli, University of Genoa and
| | - Gunes Gundem
- Christine Desmedt, Gabriele Zoppoli, Denis Larsimont, Debora Fumagalli, David Brown, Françoise Rothé, Delphine Vincent, Naima Kheddoumi, Ghizlane Rouas, Samira Majjaj, Sylvain Brohée, Roberto Salgado, Martine Piccart-Gebhart, and Christos Sotiriou, Institut Jules Bordet; Christine Galant, Cliniques Universitaires Saint Luc, Brussels; Peter Van Loo, University of Leuven; Thomas Van Brussel and Diether Lambrechts, VIB Vesalius Research Center, Leuven, Belgium; Gabriele Zoppoli, University of Genoa and
| | - Giancarlo Pruneri
- Christine Desmedt, Gabriele Zoppoli, Denis Larsimont, Debora Fumagalli, David Brown, Françoise Rothé, Delphine Vincent, Naima Kheddoumi, Ghizlane Rouas, Samira Majjaj, Sylvain Brohée, Roberto Salgado, Martine Piccart-Gebhart, and Christos Sotiriou, Institut Jules Bordet; Christine Galant, Cliniques Universitaires Saint Luc, Brussels; Peter Van Loo, University of Leuven; Thomas Van Brussel and Diether Lambrechts, VIB Vesalius Research Center, Leuven, Belgium; Gabriele Zoppoli, University of Genoa and
| | - Denis Larsimont
- Christine Desmedt, Gabriele Zoppoli, Denis Larsimont, Debora Fumagalli, David Brown, Françoise Rothé, Delphine Vincent, Naima Kheddoumi, Ghizlane Rouas, Samira Majjaj, Sylvain Brohée, Roberto Salgado, Martine Piccart-Gebhart, and Christos Sotiriou, Institut Jules Bordet; Christine Galant, Cliniques Universitaires Saint Luc, Brussels; Peter Van Loo, University of Leuven; Thomas Van Brussel and Diether Lambrechts, VIB Vesalius Research Center, Leuven, Belgium; Gabriele Zoppoli, University of Genoa and
| | - Marco Fornili
- Christine Desmedt, Gabriele Zoppoli, Denis Larsimont, Debora Fumagalli, David Brown, Françoise Rothé, Delphine Vincent, Naima Kheddoumi, Ghizlane Rouas, Samira Majjaj, Sylvain Brohée, Roberto Salgado, Martine Piccart-Gebhart, and Christos Sotiriou, Institut Jules Bordet; Christine Galant, Cliniques Universitaires Saint Luc, Brussels; Peter Van Loo, University of Leuven; Thomas Van Brussel and Diether Lambrechts, VIB Vesalius Research Center, Leuven, Belgium; Gabriele Zoppoli, University of Genoa and
| | - Debora Fumagalli
- Christine Desmedt, Gabriele Zoppoli, Denis Larsimont, Debora Fumagalli, David Brown, Françoise Rothé, Delphine Vincent, Naima Kheddoumi, Ghizlane Rouas, Samira Majjaj, Sylvain Brohée, Roberto Salgado, Martine Piccart-Gebhart, and Christos Sotiriou, Institut Jules Bordet; Christine Galant, Cliniques Universitaires Saint Luc, Brussels; Peter Van Loo, University of Leuven; Thomas Van Brussel and Diether Lambrechts, VIB Vesalius Research Center, Leuven, Belgium; Gabriele Zoppoli, University of Genoa and
| | - David Brown
- Christine Desmedt, Gabriele Zoppoli, Denis Larsimont, Debora Fumagalli, David Brown, Françoise Rothé, Delphine Vincent, Naima Kheddoumi, Ghizlane Rouas, Samira Majjaj, Sylvain Brohée, Roberto Salgado, Martine Piccart-Gebhart, and Christos Sotiriou, Institut Jules Bordet; Christine Galant, Cliniques Universitaires Saint Luc, Brussels; Peter Van Loo, University of Leuven; Thomas Van Brussel and Diether Lambrechts, VIB Vesalius Research Center, Leuven, Belgium; Gabriele Zoppoli, University of Genoa and
| | - Françoise Rothé
- Christine Desmedt, Gabriele Zoppoli, Denis Larsimont, Debora Fumagalli, David Brown, Françoise Rothé, Delphine Vincent, Naima Kheddoumi, Ghizlane Rouas, Samira Majjaj, Sylvain Brohée, Roberto Salgado, Martine Piccart-Gebhart, and Christos Sotiriou, Institut Jules Bordet; Christine Galant, Cliniques Universitaires Saint Luc, Brussels; Peter Van Loo, University of Leuven; Thomas Van Brussel and Diether Lambrechts, VIB Vesalius Research Center, Leuven, Belgium; Gabriele Zoppoli, University of Genoa and
| | - Delphine Vincent
- Christine Desmedt, Gabriele Zoppoli, Denis Larsimont, Debora Fumagalli, David Brown, Françoise Rothé, Delphine Vincent, Naima Kheddoumi, Ghizlane Rouas, Samira Majjaj, Sylvain Brohée, Roberto Salgado, Martine Piccart-Gebhart, and Christos Sotiriou, Institut Jules Bordet; Christine Galant, Cliniques Universitaires Saint Luc, Brussels; Peter Van Loo, University of Leuven; Thomas Van Brussel and Diether Lambrechts, VIB Vesalius Research Center, Leuven, Belgium; Gabriele Zoppoli, University of Genoa and
| | - Naima Kheddoumi
- Christine Desmedt, Gabriele Zoppoli, Denis Larsimont, Debora Fumagalli, David Brown, Françoise Rothé, Delphine Vincent, Naima Kheddoumi, Ghizlane Rouas, Samira Majjaj, Sylvain Brohée, Roberto Salgado, Martine Piccart-Gebhart, and Christos Sotiriou, Institut Jules Bordet; Christine Galant, Cliniques Universitaires Saint Luc, Brussels; Peter Van Loo, University of Leuven; Thomas Van Brussel and Diether Lambrechts, VIB Vesalius Research Center, Leuven, Belgium; Gabriele Zoppoli, University of Genoa and
| | - Ghizlane Rouas
- Christine Desmedt, Gabriele Zoppoli, Denis Larsimont, Debora Fumagalli, David Brown, Françoise Rothé, Delphine Vincent, Naima Kheddoumi, Ghizlane Rouas, Samira Majjaj, Sylvain Brohée, Roberto Salgado, Martine Piccart-Gebhart, and Christos Sotiriou, Institut Jules Bordet; Christine Galant, Cliniques Universitaires Saint Luc, Brussels; Peter Van Loo, University of Leuven; Thomas Van Brussel and Diether Lambrechts, VIB Vesalius Research Center, Leuven, Belgium; Gabriele Zoppoli, University of Genoa and
| | - Samira Majjaj
- Christine Desmedt, Gabriele Zoppoli, Denis Larsimont, Debora Fumagalli, David Brown, Françoise Rothé, Delphine Vincent, Naima Kheddoumi, Ghizlane Rouas, Samira Majjaj, Sylvain Brohée, Roberto Salgado, Martine Piccart-Gebhart, and Christos Sotiriou, Institut Jules Bordet; Christine Galant, Cliniques Universitaires Saint Luc, Brussels; Peter Van Loo, University of Leuven; Thomas Van Brussel and Diether Lambrechts, VIB Vesalius Research Center, Leuven, Belgium; Gabriele Zoppoli, University of Genoa and
| | - Sylvain Brohée
- Christine Desmedt, Gabriele Zoppoli, Denis Larsimont, Debora Fumagalli, David Brown, Françoise Rothé, Delphine Vincent, Naima Kheddoumi, Ghizlane Rouas, Samira Majjaj, Sylvain Brohée, Roberto Salgado, Martine Piccart-Gebhart, and Christos Sotiriou, Institut Jules Bordet; Christine Galant, Cliniques Universitaires Saint Luc, Brussels; Peter Van Loo, University of Leuven; Thomas Van Brussel and Diether Lambrechts, VIB Vesalius Research Center, Leuven, Belgium; Gabriele Zoppoli, University of Genoa and
| | - Peter Van Loo
- Christine Desmedt, Gabriele Zoppoli, Denis Larsimont, Debora Fumagalli, David Brown, Françoise Rothé, Delphine Vincent, Naima Kheddoumi, Ghizlane Rouas, Samira Majjaj, Sylvain Brohée, Roberto Salgado, Martine Piccart-Gebhart, and Christos Sotiriou, Institut Jules Bordet; Christine Galant, Cliniques Universitaires Saint Luc, Brussels; Peter Van Loo, University of Leuven; Thomas Van Brussel and Diether Lambrechts, VIB Vesalius Research Center, Leuven, Belgium; Gabriele Zoppoli, University of Genoa and
| | - Patrick Maisonneuve
- Christine Desmedt, Gabriele Zoppoli, Denis Larsimont, Debora Fumagalli, David Brown, Françoise Rothé, Delphine Vincent, Naima Kheddoumi, Ghizlane Rouas, Samira Majjaj, Sylvain Brohée, Roberto Salgado, Martine Piccart-Gebhart, and Christos Sotiriou, Institut Jules Bordet; Christine Galant, Cliniques Universitaires Saint Luc, Brussels; Peter Van Loo, University of Leuven; Thomas Van Brussel and Diether Lambrechts, VIB Vesalius Research Center, Leuven, Belgium; Gabriele Zoppoli, University of Genoa and
| | - Roberto Salgado
- Christine Desmedt, Gabriele Zoppoli, Denis Larsimont, Debora Fumagalli, David Brown, Françoise Rothé, Delphine Vincent, Naima Kheddoumi, Ghizlane Rouas, Samira Majjaj, Sylvain Brohée, Roberto Salgado, Martine Piccart-Gebhart, and Christos Sotiriou, Institut Jules Bordet; Christine Galant, Cliniques Universitaires Saint Luc, Brussels; Peter Van Loo, University of Leuven; Thomas Van Brussel and Diether Lambrechts, VIB Vesalius Research Center, Leuven, Belgium; Gabriele Zoppoli, University of Genoa and
| | - Thomas Van Brussel
- Christine Desmedt, Gabriele Zoppoli, Denis Larsimont, Debora Fumagalli, David Brown, Françoise Rothé, Delphine Vincent, Naima Kheddoumi, Ghizlane Rouas, Samira Majjaj, Sylvain Brohée, Roberto Salgado, Martine Piccart-Gebhart, and Christos Sotiriou, Institut Jules Bordet; Christine Galant, Cliniques Universitaires Saint Luc, Brussels; Peter Van Loo, University of Leuven; Thomas Van Brussel and Diether Lambrechts, VIB Vesalius Research Center, Leuven, Belgium; Gabriele Zoppoli, University of Genoa and
| | - Diether Lambrechts
- Christine Desmedt, Gabriele Zoppoli, Denis Larsimont, Debora Fumagalli, David Brown, Françoise Rothé, Delphine Vincent, Naima Kheddoumi, Ghizlane Rouas, Samira Majjaj, Sylvain Brohée, Roberto Salgado, Martine Piccart-Gebhart, and Christos Sotiriou, Institut Jules Bordet; Christine Galant, Cliniques Universitaires Saint Luc, Brussels; Peter Van Loo, University of Leuven; Thomas Van Brussel and Diether Lambrechts, VIB Vesalius Research Center, Leuven, Belgium; Gabriele Zoppoli, University of Genoa and
| | - Ron Bose
- Christine Desmedt, Gabriele Zoppoli, Denis Larsimont, Debora Fumagalli, David Brown, Françoise Rothé, Delphine Vincent, Naima Kheddoumi, Ghizlane Rouas, Samira Majjaj, Sylvain Brohée, Roberto Salgado, Martine Piccart-Gebhart, and Christos Sotiriou, Institut Jules Bordet; Christine Galant, Cliniques Universitaires Saint Luc, Brussels; Peter Van Loo, University of Leuven; Thomas Van Brussel and Diether Lambrechts, VIB Vesalius Research Center, Leuven, Belgium; Gabriele Zoppoli, University of Genoa and
| | - Otto Metzger
- Christine Desmedt, Gabriele Zoppoli, Denis Larsimont, Debora Fumagalli, David Brown, Françoise Rothé, Delphine Vincent, Naima Kheddoumi, Ghizlane Rouas, Samira Majjaj, Sylvain Brohée, Roberto Salgado, Martine Piccart-Gebhart, and Christos Sotiriou, Institut Jules Bordet; Christine Galant, Cliniques Universitaires Saint Luc, Brussels; Peter Van Loo, University of Leuven; Thomas Van Brussel and Diether Lambrechts, VIB Vesalius Research Center, Leuven, Belgium; Gabriele Zoppoli, University of Genoa and
| | - Christine Galant
- Christine Desmedt, Gabriele Zoppoli, Denis Larsimont, Debora Fumagalli, David Brown, Françoise Rothé, Delphine Vincent, Naima Kheddoumi, Ghizlane Rouas, Samira Majjaj, Sylvain Brohée, Roberto Salgado, Martine Piccart-Gebhart, and Christos Sotiriou, Institut Jules Bordet; Christine Galant, Cliniques Universitaires Saint Luc, Brussels; Peter Van Loo, University of Leuven; Thomas Van Brussel and Diether Lambrechts, VIB Vesalius Research Center, Leuven, Belgium; Gabriele Zoppoli, University of Genoa and
| | - François Bertucci
- Christine Desmedt, Gabriele Zoppoli, Denis Larsimont, Debora Fumagalli, David Brown, Françoise Rothé, Delphine Vincent, Naima Kheddoumi, Ghizlane Rouas, Samira Majjaj, Sylvain Brohée, Roberto Salgado, Martine Piccart-Gebhart, and Christos Sotiriou, Institut Jules Bordet; Christine Galant, Cliniques Universitaires Saint Luc, Brussels; Peter Van Loo, University of Leuven; Thomas Van Brussel and Diether Lambrechts, VIB Vesalius Research Center, Leuven, Belgium; Gabriele Zoppoli, University of Genoa and
| | - Martine Piccart-Gebhart
- Christine Desmedt, Gabriele Zoppoli, Denis Larsimont, Debora Fumagalli, David Brown, Françoise Rothé, Delphine Vincent, Naima Kheddoumi, Ghizlane Rouas, Samira Majjaj, Sylvain Brohée, Roberto Salgado, Martine Piccart-Gebhart, and Christos Sotiriou, Institut Jules Bordet; Christine Galant, Cliniques Universitaires Saint Luc, Brussels; Peter Van Loo, University of Leuven; Thomas Van Brussel and Diether Lambrechts, VIB Vesalius Research Center, Leuven, Belgium; Gabriele Zoppoli, University of Genoa and
| | - Giuseppe Viale
- Christine Desmedt, Gabriele Zoppoli, Denis Larsimont, Debora Fumagalli, David Brown, Françoise Rothé, Delphine Vincent, Naima Kheddoumi, Ghizlane Rouas, Samira Majjaj, Sylvain Brohée, Roberto Salgado, Martine Piccart-Gebhart, and Christos Sotiriou, Institut Jules Bordet; Christine Galant, Cliniques Universitaires Saint Luc, Brussels; Peter Van Loo, University of Leuven; Thomas Van Brussel and Diether Lambrechts, VIB Vesalius Research Center, Leuven, Belgium; Gabriele Zoppoli, University of Genoa and
| | - Elia Biganzoli
- Christine Desmedt, Gabriele Zoppoli, Denis Larsimont, Debora Fumagalli, David Brown, Françoise Rothé, Delphine Vincent, Naima Kheddoumi, Ghizlane Rouas, Samira Majjaj, Sylvain Brohée, Roberto Salgado, Martine Piccart-Gebhart, and Christos Sotiriou, Institut Jules Bordet; Christine Galant, Cliniques Universitaires Saint Luc, Brussels; Peter Van Loo, University of Leuven; Thomas Van Brussel and Diether Lambrechts, VIB Vesalius Research Center, Leuven, Belgium; Gabriele Zoppoli, University of Genoa and
| | - Peter J. Campbell
- Christine Desmedt, Gabriele Zoppoli, Denis Larsimont, Debora Fumagalli, David Brown, Françoise Rothé, Delphine Vincent, Naima Kheddoumi, Ghizlane Rouas, Samira Majjaj, Sylvain Brohée, Roberto Salgado, Martine Piccart-Gebhart, and Christos Sotiriou, Institut Jules Bordet; Christine Galant, Cliniques Universitaires Saint Luc, Brussels; Peter Van Loo, University of Leuven; Thomas Van Brussel and Diether Lambrechts, VIB Vesalius Research Center, Leuven, Belgium; Gabriele Zoppoli, University of Genoa and
| | - Christos Sotiriou
- Christine Desmedt, Gabriele Zoppoli, Denis Larsimont, Debora Fumagalli, David Brown, Françoise Rothé, Delphine Vincent, Naima Kheddoumi, Ghizlane Rouas, Samira Majjaj, Sylvain Brohée, Roberto Salgado, Martine Piccart-Gebhart, and Christos Sotiriou, Institut Jules Bordet; Christine Galant, Cliniques Universitaires Saint Luc, Brussels; Peter Van Loo, University of Leuven; Thomas Van Brussel and Diether Lambrechts, VIB Vesalius Research Center, Leuven, Belgium; Gabriele Zoppoli, University of Genoa and
| |
Collapse
|
20
|
Choudhary V, Gupta S. Comprehensive Gene Mutation Profiling of Breast Tumors: Is It Ready for Prime Time Use? CURRENT BREAST CANCER REPORTS 2016. [DOI: 10.1007/s12609-016-0213-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
21
|
Griseri P, Garrone O, Lo Sardo A, Monteverde M, Rusmini M, Tonissi F, Merlano M, Bruzzi P, Lo Nigro C, Ceccherini I. Genetic and epigenetic factors affect RET gene expression in breast cancer cell lines and influence survival in patients. Oncotarget 2016; 7:26465-79. [PMID: 27034161 PMCID: PMC5041993 DOI: 10.18632/oncotarget.8417] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Accepted: 03/04/2016] [Indexed: 12/14/2022] Open
Abstract
Germline and somatic mutations play a crucial role in breast cancer (BC), driving the initiation, progression, response to therapy and outcome of the disease. Hormonal therapy is limited to patients with tumors expressing steroid hormone receptors, such as estrogen receptor (ER), nevertheless resistance often limits its success.The RET gene is known to be involved in neurocristopathies such as Hirschsprung disease and Multiple Endocrine Neoplasia type 2, in the presence of loss-of-function and gain-of-function mutations, respectively. More recently, RET over-expression has emerged as a new player in ER-positive (ER+) BC, and as a potential target to enhance sensitivity and avoid resistance to tamoxifen therapy.Therefore, targeting the RET pathway may lead to new therapies in ER+ BC. To this end, we have investigated the molecular mechanisms which underlie RET overexpression and its possible modulation in two BC cell lines, MCF7 and T47D, showing different RET expression levels. Moreover, we have carried out a pilot association study in 93 ER+ BC patients. Consistent with the adverse role of RET over-expression in BC, increased overall survival was observed in carriers of the variant allele of SNP rs2435357, a RET polymorphism already known to be associated with reduced RET expression.
Collapse
Affiliation(s)
- Paola Griseri
- UOC Medical Genetics, IRCCS Giannina Gaslini Institute, Genoa, Italy
| | - Ornella Garrone
- Unit of Medical Oncology, Department of Oncology, S. Croce & Carle Teaching Hospital, Cuneo, Italy
| | | | - Martino Monteverde
- Laboratory of Cancer Genetics and Translational Oncology, Department of Oncology, S. Croce & Carle Teaching Hospital, Cuneo, Italy
| | - Marta Rusmini
- UOC Medical Genetics, IRCCS Giannina Gaslini Institute, Genoa, Italy
| | - Federica Tonissi
- Laboratory of Cancer Genetics and Translational Oncology, Department of Oncology, S. Croce & Carle Teaching Hospital, Cuneo, Italy
| | - Marco Merlano
- Unit of Medical Oncology, Department of Oncology, S. Croce & Carle Teaching Hospital, Cuneo, Italy
| | - Paolo Bruzzi
- Clinical Epidemiology, IRCCS AUO San Martino IST, Genoa, Italy
| | - Cristiana Lo Nigro
- Laboratory of Cancer Genetics and Translational Oncology, Department of Oncology, S. Croce & Carle Teaching Hospital, Cuneo, Italy
| | | |
Collapse
|
22
|
Ali S, Mondal N, Choudhry H, Rasool M, Pushparaj PN, Khan MA, Mahfooz M, Sami GA, Jarullah J, Ali A, Jamal MS. Current Management Strategies in Breast Cancer by Targeting Key Altered Molecular Players. Front Oncol 2016; 6:45. [PMID: 26973813 PMCID: PMC4771739 DOI: 10.3389/fonc.2016.00045] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Accepted: 02/12/2016] [Indexed: 01/08/2023] Open
Abstract
Breast cancer is the second largest disease affecting women worldwide. It remains the most frequently reported and leading cause of death among women in both developed and developing countries. Tamoxifen and raloxifene are commonly used selective estrogen receptor modulators for treatment of breast cancer in women with high risk, although resistance occurs by tamoxifen after 5 years of therapy and both drugs cause uterine cancer and thromboembolic events. Aromatase inhibitors (AIs) are one of the optional modes used for breast cancer treatment. The combination of AIs along with tamoxifen can also be beneficial. Various therapeutic agents from different sources are being studied, which further need to be improved for potential outcome. For this, clinical trials based on large number of patients with optimal dose and lesser side effects have to be more in practice. Despite the clinical trials going on, there is need of better molecular models, which can identify high risk population, new agents with better benefit having less side effects, and improved biomarkers for treating breast cancer.
Collapse
Affiliation(s)
- Shazia Ali
- School of Life Science, Jawaharlal Nehru University , New Delhi , India
| | - Neelima Mondal
- School of Life Science, Jawaharlal Nehru University , New Delhi , India
| | - Hani Choudhry
- Department of Biochemistry, Faculty of Science, Center of Innovation in Personalized Medicine, King Fahd Center for Medical Research, King Abdulaziz University , Jeddah , Saudi Arabia
| | - Mahmood Rasool
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University , Jeddah , Saudi Arabia
| | - Peter N Pushparaj
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University , Jeddah , Saudi Arabia
| | | | - Maryam Mahfooz
- Department of Computer Science, Jamia Millia Islamia , New Delhi , India
| | - Ghufrana A Sami
- Department of Biotechnology, Jamia Millia Islamia , New Delhi , India
| | - Jummanah Jarullah
- King Fahd Medical Research Center, King Abdulaziz University , Jeddah , Saudi Arabia
| | - Ashraf Ali
- King Fahd Medical Research Center, King Abdulaziz University , Jeddah , Saudi Arabia
| | - Mohammad S Jamal
- King Fahd Medical Research Center, King Abdulaziz University , Jeddah , Saudi Arabia
| |
Collapse
|
23
|
Changing molecular profile of brain metastases compared with matched breast primary cancers and impact on clinical outcomes. Br J Cancer 2016; 114:793-800. [PMID: 26908328 PMCID: PMC4984859 DOI: 10.1038/bjc.2016.34] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 01/12/2016] [Accepted: 01/22/2016] [Indexed: 12/21/2022] Open
Abstract
Background: Breast cancer commonly metastasises to the brain, but little is known about changes in the molecular profile of the brain secondaries and impact on clinical outcomes. Methods: Patients with samples from brain metastases and matched breast cancers were included. Immunohistochemical analysis for oestrogen receptor, progesterone receptor, p27kip1, cyclin D1, epidermal growth factor receptor, insulin like growth factor 1, insulin like growth factor 1 receptor, vascular endothelial growth factor A, transforming growth factor-β and HER2 receptor was performed. Borderline HER2 results were analysed by fluorescent in situ hybridisation. Levels of expression were compared, with review of effect on clinical outcomes. Results: A total of 41 patients were included. Of the patients, 20% had a change in oestrogen receptor or HER2 in their brain metastasis that could affect therapeutic decisions. There were statistically significant rises in brain metastases for p27kip1 (P=0.023) and cyclin D1 (P=0.030) and a fall in vascular endothelial growth factor A (P=0.012). Overall survival from the time of metastasis increased significantly with oestrogen receptor-positive (P=0.005) and progesterone receptor-positive (P=0.013) brain lesions and with a longer duration from diagnosis of the breast primary (P<0.001). Conclusions: In this cohort there were phenotypic differences in metastatic brain tumours compared with matched primary breast tumours. These could be relevant for aetiology, and have an impact on prognostication, current and future therapies.
Collapse
|
24
|
Martin SD, Coukos G, Holt RA, Nelson BH. Targeting the undruggable: immunotherapy meets personalized oncology in the genomic era. Ann Oncol 2015; 26:2367-74. [PMID: 26371284 PMCID: PMC4658541 DOI: 10.1093/annonc/mdv382] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 08/14/2015] [Accepted: 08/17/2015] [Indexed: 12/22/2022] Open
Abstract
Owing to recent advances in genomic technologies, personalized oncology is poised to fundamentally alter cancer therapy. In this paradigm, the mutational and transcriptional profiles of tumors are assessed, and personalized treatments are designed based on the specific molecular abnormalities relevant to each patient's cancer. To date, such approaches have yielded impressive clinical responses in some patients. However, a major limitation of this strategy has also been revealed: the vast majority of tumor mutations are not targetable by current pharmacological approaches. Immunotherapy offers a promising alternative to exploit tumor mutations as targets for clinical intervention. Mutated proteins can give rise to novel antigens (called neoantigens) that are recognized with high specificity by patient T cells. Indeed, neoantigen-specific T cells have been shown to underlie clinical responses to many standard treatments and immunotherapeutic interventions. Moreover, studies in mouse models targeting neoantigens, and early results from clinical trials, have established proof of concept for personalized immunotherapies targeting next-generation sequencing identified neoantigens. Here, we review basic immunological principles related to T-cell recognition of neoantigens, and we examine recent studies that use genomic data to design personalized immunotherapies. We discuss the opportunities and challenges that lie ahead on the road to improving patient outcomes by incorporating immunotherapy into the paradigm of personalized oncology.
Collapse
Affiliation(s)
- S D Martin
- Trev and Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria Interdisciplinary Oncology Program, University of British Columbia, Vancouver Michael Smith's Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, Canada
| | - G Coukos
- Ludwig Center for Cancer Research, University of Lausanne, Lausanne Hospital of the University of Lausanne (CHUV), Lausanne, Switzerland
| | - R A Holt
- Michael Smith's Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, Canada Molecular Biology and Biochemistry, Simon Fraser University, Vancouver Department of Medical Genetics, University of British Columbia, Vancouver
| | - B H Nelson
- Trev and Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria Department of Medical Genetics, University of British Columbia, Vancouver Department of Microbiology and Biochemistry, University of Victoria, Victoria, Canada
| |
Collapse
|
25
|
Cirqueira MB, Moreira MAR, Soares LR, Cysneiros MADPC, Vilela MHT, Freitas-Junior R. Effect of Ki-67 on Immunohistochemical Classification of Luminal A to Luminal B Subtypes of Breast Carcinoma. Breast J 2015; 21:465-72. [DOI: 10.1111/tbj.12441] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Magno Belém Cirqueira
- Biomedical of Pathology Department; Teaching Hospital; Federal University of Goiás; Goiânia Goiás Brazil
| | | | | | | | | | - Ruffo Freitas-Junior
- Gynecology and Breast Service of Hospital Araújo Jorge of the Associação de Combate ao Câncer de Goiás (ACCG); Gynecology and Obstetrics Department; Federal University of Goiás; Goiânia Goiás Brazil
| |
Collapse
|