1
|
Li M, Zhou S, Lv H, Cai M, Shui R, Yang W. Neoadjuvant chemotherapy response in androgen receptor-positive triple-negative breast cancer: potential predictive biomarkers and genetic alterations. Breast Cancer Res 2025; 27:41. [PMID: 40114215 PMCID: PMC11927354 DOI: 10.1186/s13058-025-01994-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 03/03/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND The aim of the present study was to investigate whether the androgen receptor (AR) status affects the efficacy of neoadjuvant chemotherapy (NACT) in triple negative breast cancer (TNBC) patients, and to elucidate the predictive biomarkers and mutations associated with pathological complete response (pCR) in AR-positive TNBC patients. METHODS The current retrospective cohort included 226 TNBC patients who underwent NACT. AR and FOXC1 were assessed by immunohistochemistry on pretreatment biopsy specimens of 226 TNBC patients from 2018 to 2022. The clinicopathological features of AR-negative, AR < 10%, and AR ≥ 10% TNBC patients were analyzed to confirm the appropriate threshold. The response was evaluated in terms of pCR and Miller-Payne (MP) grade in the subsequent mastectomy or breast conservation samples. Next-generation sequencing (NGS) was utilized to further investigate the molecular characteristics of 44 AR-positive TNBC patients. RESULTS Among the 226 TNBC patients, compared with AR-negative and AR < 10% tumors (68.58%, 155/226), AR ≥ 10% TNBC patients (31.41%, 71/226) exhibited distinct clinicopathological features, while no significant difference was detected between those with AR-negative tumors and those with AR < 10% tumors. Thus, tumors with AR ≥ 10% expression were defined as having AR positive expression. The pCR rate of AR-positive TNBCs was lower than that of AR-negative TNBC patients (12.68% vs. 34.19%, p < 0.001). In TNBC, multivariate analysis demonstrated that FOXC1 was an independent predictor of pCR (p = 0.042), whereas AR was not. The pCR rate was higher in FOXC1 positive patients than in FOXC1 negative patients (34.44% vs. 3.13%, p < 0.001). In the AR-positive TNBC subgroup, patients with FOXC1 expression had lower AR expression, higher Ki-67 expression, and higher histological grade. Compared with AR-positive TNBC patients who achieved pCR, the non-pCR patients had a greater percentage of mutations in genes involved in the PI3K/AKT/mTOR pathway. CONCLUSIONS The current study indicated that the AR-positive TNBC is correlated with lower rates of pCR after NACT. The expression of FOXC1 in TNBC patients and AR-positive TNBC patients could be utilized as a predictive marker for the efficacy of NACT. The present study provides a rationale for treating these non-pCR AR-positive TNBC tumors with PI3K/AKT/mTOR inhibitors.
Collapse
Affiliation(s)
- Ming Li
- Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dongan Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dongan Road, Shanghai, 200032, China
- Institute of Pathology, Fudan University, 270 Dongan Road, Shanghai, 200032, China
| | - Shuling Zhou
- Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dongan Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dongan Road, Shanghai, 200032, China
- Institute of Pathology, Fudan University, 270 Dongan Road, Shanghai, 200032, China
| | - Hong Lv
- Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dongan Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dongan Road, Shanghai, 200032, China
- Institute of Pathology, Fudan University, 270 Dongan Road, Shanghai, 200032, China
| | - Mengyuan Cai
- Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dongan Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dongan Road, Shanghai, 200032, China
- Institute of Pathology, Fudan University, 270 Dongan Road, Shanghai, 200032, China
| | - Ruohong Shui
- Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dongan Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dongan Road, Shanghai, 200032, China
- Institute of Pathology, Fudan University, 270 Dongan Road, Shanghai, 200032, China
| | - Wentao Yang
- Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dongan Road, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dongan Road, Shanghai, 200032, China.
- Institute of Pathology, Fudan University, 270 Dongan Road, Shanghai, 200032, China.
| |
Collapse
|
2
|
Michail C, Rodrigues Lima F, Viguier M, Deshayes F. Structure and function of the lysine methyltransferase SETD2 in cancer: From histones to cytoskeleton. Neoplasia 2025; 59:101090. [PMID: 39591760 PMCID: PMC11626819 DOI: 10.1016/j.neo.2024.101090] [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: 08/27/2024] [Revised: 11/14/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024]
Abstract
SETD2 is known to be the unique histone methyltransferase responsible for the trimethylation of the lysine 36 of histone H3 thus generating H3K36me3. This epigenetic mark is critical for transcriptional activation and elongation, DNA repair, mRNA splicing, and DNA methylation. Recurrent SETD2-inactivating mutations and altered H3K36me3 levels are found in cancer at high frequency and numerous studies indicate that SETD2 acts as a tumor suppressor. Recently, SETD2 was further shown to methylate non-histone proteins particularly the cytoskeletal proteins tubulin and actin with subsequent impacts on cytoskeleton structure, mitosis and cell migration. Herein, we provide a review of the role of SETD2 in different cancers with special emphasis on the structural basis of the functions of this key lysine methyltransferase. Moreover, beyond the role of this enzyme in epigenetics and H3K36me3-dependent processes, we highlight the putative role of "non-epigenetic/H3K36me3" functions of SETD2 in cancer, particularly those involving the cytoskeleton.
Collapse
Affiliation(s)
- Christina Michail
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, F-75013 Paris, France
| | - Fernando Rodrigues Lima
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, F-75013 Paris, France
| | - Mireille Viguier
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, F-75013 Paris, France.
| | - Frédérique Deshayes
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, F-75013 Paris, France.
| |
Collapse
|
3
|
Takemon Y, Pleasance ED, Gagliardi A, Hughes CS, Csizmok V, Wee K, Trinh DL, Huff RD, Mungall AJ, Moore RA, Chuah E, Mungall KL, Lewis E, Nelson J, Lim HJ, Renouf DJ, Jones SJ, Laskin J, Marra MA. Mapping in silico genetic networks of the KMT2D tumour suppressor gene to uncover novel functional associations and cancer cell vulnerabilities. Genome Med 2024; 16:136. [PMID: 39578878 PMCID: PMC11583415 DOI: 10.1186/s13073-024-01401-9] [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: 08/05/2024] [Accepted: 10/29/2024] [Indexed: 11/24/2024] Open
Abstract
BACKGROUND Loss-of-function (LOF) alterations in tumour suppressor genes cannot be directly targeted. Approaches characterising gene function and vulnerabilities conferred by such mutations are required. METHODS Here, we computationally map genetic networks of KMT2D, a tumour suppressor gene frequently mutated in several cancer types. Using KMT2D loss-of-function (KMT2DLOF) mutations as a model, we illustrate the utility of in silico genetic networks in uncovering novel functional associations and vulnerabilities in cancer cells with LOF alterations affecting tumour suppressor genes. RESULTS We revealed genetic interactors with functions in histone modification, metabolism, and immune response and synthetic lethal (SL) candidates, including some encoding existing therapeutic targets. Notably, we predicted WRN as a novel SL interactor and, using recently available WRN inhibitor (HRO761 and VVD-133214) treatment response data, we observed that KMT2D mutational status significantly distinguishes treatment-sensitive MSI cell lines from treatment-insensitive MSI cell lines. CONCLUSIONS Our study thus illustrates how tumour suppressor gene LOF alterations can be exploited to reveal potentially targetable cancer cell vulnerabilities.
Collapse
Affiliation(s)
- Yuka Takemon
- Genome Science and Technology Graduate Program, University of British Columbia, Vancouver, Canada
- Michael Smith Laboratories, University of British Columbia, Vancouver, Canada
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, Canada
| | - Erin D Pleasance
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, Canada
| | - Alessia Gagliardi
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, Canada
| | | | - Veronika Csizmok
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, Canada
| | - Kathleen Wee
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, Canada
| | - Diane L Trinh
- Michael Smith Laboratories, University of British Columbia, Vancouver, Canada
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, Canada
| | - Ryan D Huff
- Division of Respiratory Medicine, Department of Medicine, Air Pollution Exposure Laboratory, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Andrew J Mungall
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, Canada
| | - Richard A Moore
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, Canada
| | - Eric Chuah
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, Canada
| | - Karen L Mungall
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, Canada
| | - Eleanor Lewis
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, Canada
| | - Jessica Nelson
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, Canada
| | - Howard J Lim
- Department of Medical Oncology, BC Cancer, Vancouver, BC, Canada
| | - Daniel J Renouf
- Department of Medical Oncology, BC Cancer, Vancouver, BC, Canada
- Pancreas Centre BC, Vancouver, BC, Canada
| | - Steven Jm Jones
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, Canada
| | - Janessa Laskin
- Department of Medical Oncology, BC Cancer, Vancouver, BC, Canada
| | - Marco A Marra
- Michael Smith Laboratories, University of British Columbia, Vancouver, Canada.
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, Canada.
- Department of Medical Genetics, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
4
|
Ye C, Jiang S, Zeng T, He S, Cao J, Xiao J. The role of LOXL2 in tumor progression, immune response and cellular senescence: a comprehensive analysis. Discov Oncol 2024; 15:245. [PMID: 38922489 PMCID: PMC11208360 DOI: 10.1007/s12672-024-01107-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 06/15/2024] [Indexed: 06/27/2024] Open
Abstract
LOXL2, an enzyme belonging to the LOX family, facilitates the cross-linking of extracellular matrix (ECM) elements. However, the roles of the LOXL2 gene in mechanisms of oncogenesis and tumor development have not been clearly defined. In this pan-cancer study, we examined the notable disparity in LOXL2 expression at the mRNA and protein levels among various cancer types and elucidated its interconnected roles in tumor progression, mutational profile, immune response, and cellular senescence. Apart from investigating the hyperexpression of LOXL2 being related to poorer prognosis in different types of tumors, this study also unveiled noteworthy connections between LOXL2 and genetic mutations, infiltration of tumor immune cells, and genes in immune checkpoint pathways. Further analysis revealed the participation of LOXL2 in multiple pathways related to cancer extracellular matrix remodeling and cellular senescence. Moreover, our investigation uncovered that the knockdown and inhibition of LOXL2 significantly attenuated the proliferation and migration of PC-9 and HCC-LM3 cells. The knock-down and inhibition of LOXL2 enhanced cellular senescence in lung and liver cancer cells, as confirmed by SA-β-Gal staining and quantitative RT-PCR analyses. This comprehensive analysis offers valuable insights on the functions of LOXL2 in different types of cancer and its role in regulating the senescence of cancer cells.
Collapse
Affiliation(s)
- Chen Ye
- School of Health Science and Technology, University of Shanghai for Science and Technology, 516 Jungong Road, Shanghai, 200093, China
- Spinal Tumor Center, Department of Orthopedic Oncology, Shanghai Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai, 200003, China
| | - Sihan Jiang
- Graduate School, Naval Medical University, 800 Xiangyin Road, Shanghai, 200433, China
| | - Tanlun Zeng
- Graduate School, Naval Medical University, 800 Xiangyin Road, Shanghai, 200433, China
- Clinical Cancer Institute, Center for Translational Medicine, Naval Medical University, 800 Xiangyin Road, Shanghai, 200433, China
| | - Shaohui He
- Spinal Tumor Center, Department of Orthopedic Oncology, Shanghai Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai, 200003, China
| | - Jinjin Cao
- School of Health Science and Technology, University of Shanghai for Science and Technology, 516 Jungong Road, Shanghai, 200093, China.
- Clinical Cancer Institute, Center for Translational Medicine, Naval Medical University, 800 Xiangyin Road, Shanghai, 200433, China.
| | - Jianru Xiao
- School of Health Science and Technology, University of Shanghai for Science and Technology, 516 Jungong Road, Shanghai, 200093, China.
- Spinal Tumor Center, Department of Orthopedic Oncology, Shanghai Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai, 200003, China.
| |
Collapse
|
5
|
Tinsley E, Bredin P, Toomey S, Hennessy BT, Furney SJ. KMT2C and KMT2D aberrations in breast cancer. Trends Cancer 2024; 10:519-530. [PMID: 38453563 DOI: 10.1016/j.trecan.2024.02.003] [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: 11/10/2023] [Revised: 02/09/2024] [Accepted: 02/14/2024] [Indexed: 03/09/2024]
Abstract
KMT2C and KMT2D are histone lysine methyltransferases responsible for the monomethylation of histone 3 lysine 4 (H3K4) residues at gene enhancer sites. KMT2C/D are the most frequently mutated histone methyltransferases (HMTs) in breast cancer, occurring at frequencies of 10-20% collectively. Frequent damaging and truncating somatic mutations indicate a tumour-suppressive role of KMT2C/D in breast oncogenesis. Recent studies using cell lines and mouse models to replicate KMT2C/D loss show that these genes contribute to oestrogen receptor (ER)-driven transcription in ER+ breast cancers through the priming of gene enhancer regions. This review provides an overview of the functions of KMT2C/D and outlines the recent clinical and experimental evidence of the roles of KMT2C and KMT2D in breast cancer development.
Collapse
Affiliation(s)
- Emily Tinsley
- Genomic Oncology Research Group, Department of Physiology and Medical Physics, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Philip Bredin
- Medical Oncology Group, Department of Molecular Medicine, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Sinead Toomey
- Medical Oncology Group, Department of Molecular Medicine, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Bryan T Hennessy
- Medical Oncology Group, Department of Molecular Medicine, RCSI University of Medicine and Health Sciences, Dublin, Ireland; Department of Medical Oncology, Beaumont Hospital, Dublin, Ireland.
| | - Simon J Furney
- Genomic Oncology Research Group, Department of Physiology and Medical Physics, RCSI University of Medicine and Health Sciences, Dublin, Ireland.
| |
Collapse
|
6
|
Rysenkova KD, Gaboriaud J, Fokin AI, Toubiana R, Bense A, Mirdass C, Jin M, Ho MCN, Glading E, Vacher S, Courtois L, Bièche I, Gautreau AM. PI 3-Kinase and the Histone Methyl-Transferase KMT2D Collaborate to Induce Arp2/3-Dependent Migration of Mammary Epithelial Cells. Cells 2024; 13:876. [PMID: 38786098 PMCID: PMC11119607 DOI: 10.3390/cells13100876] [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: 02/13/2024] [Revised: 05/16/2024] [Accepted: 05/18/2024] [Indexed: 05/25/2024] Open
Abstract
Breast cancer develops upon sequential acquisition of driver mutations in mammary epithelial cells; however, how these mutations collaborate to transform normal cells remains unclear in most cases. We aimed to reconstitute this process in a particular case. To this end, we combined the activated form of the PI 3-kinase harboring the H1047R mutation with the inactivation of the histone lysine methyl-transferase KMT2D in the non-tumorigenic human mammary epithelial cell line MCF10A. We found that PI 3-kinase activation promoted cell-cycle progression, especially when growth signals were limiting, as well as cell migration, both in a collective monolayer and as single cells. Furthermore, we showed that KMT2D inactivation had relatively little influence on these processes, except for single-cell migration, which KMT2D inactivation promoted in synergy with PI 3-kinase activation. The combination of these two genetic alterations induced expression of the ARPC5L gene that encodes a subunit of the Arp2/3 complex. ARPC5L depletion fully abolished the enhanced migration persistence exhibited by double-mutant cells. Our reconstitution approach in MCF10A has thus revealed both the cell function and the single-cell migration, and the underlying Arp2/3-dependent mechanism, which are synergistically regulated when KMT2D inactivation is combined with the activation of the PI 3-kinase.
Collapse
Affiliation(s)
- Karina D. Rysenkova
- Laboratoire de Biologie Structurale de la Cellule, CNRS UMR7654, Ecole Polytechnique, Institut Polytechnique de Paris, 91128 Palaiseau Cedex, France; (K.D.R.); (J.G.); (A.I.F.); (R.T.); (A.B.); (C.M.); (M.J.); (M.C.N.H.); (E.G.)
| | - Julia Gaboriaud
- Laboratoire de Biologie Structurale de la Cellule, CNRS UMR7654, Ecole Polytechnique, Institut Polytechnique de Paris, 91128 Palaiseau Cedex, France; (K.D.R.); (J.G.); (A.I.F.); (R.T.); (A.B.); (C.M.); (M.J.); (M.C.N.H.); (E.G.)
| | - Artem I. Fokin
- Laboratoire de Biologie Structurale de la Cellule, CNRS UMR7654, Ecole Polytechnique, Institut Polytechnique de Paris, 91128 Palaiseau Cedex, France; (K.D.R.); (J.G.); (A.I.F.); (R.T.); (A.B.); (C.M.); (M.J.); (M.C.N.H.); (E.G.)
| | - Raphaëlle Toubiana
- Laboratoire de Biologie Structurale de la Cellule, CNRS UMR7654, Ecole Polytechnique, Institut Polytechnique de Paris, 91128 Palaiseau Cedex, France; (K.D.R.); (J.G.); (A.I.F.); (R.T.); (A.B.); (C.M.); (M.J.); (M.C.N.H.); (E.G.)
| | - Alexandre Bense
- Laboratoire de Biologie Structurale de la Cellule, CNRS UMR7654, Ecole Polytechnique, Institut Polytechnique de Paris, 91128 Palaiseau Cedex, France; (K.D.R.); (J.G.); (A.I.F.); (R.T.); (A.B.); (C.M.); (M.J.); (M.C.N.H.); (E.G.)
| | - Camil Mirdass
- Laboratoire de Biologie Structurale de la Cellule, CNRS UMR7654, Ecole Polytechnique, Institut Polytechnique de Paris, 91128 Palaiseau Cedex, France; (K.D.R.); (J.G.); (A.I.F.); (R.T.); (A.B.); (C.M.); (M.J.); (M.C.N.H.); (E.G.)
| | - Mélissa Jin
- Laboratoire de Biologie Structurale de la Cellule, CNRS UMR7654, Ecole Polytechnique, Institut Polytechnique de Paris, 91128 Palaiseau Cedex, France; (K.D.R.); (J.G.); (A.I.F.); (R.T.); (A.B.); (C.M.); (M.J.); (M.C.N.H.); (E.G.)
| | - Minh Chau N. Ho
- Laboratoire de Biologie Structurale de la Cellule, CNRS UMR7654, Ecole Polytechnique, Institut Polytechnique de Paris, 91128 Palaiseau Cedex, France; (K.D.R.); (J.G.); (A.I.F.); (R.T.); (A.B.); (C.M.); (M.J.); (M.C.N.H.); (E.G.)
| | - Elizabeth Glading
- Laboratoire de Biologie Structurale de la Cellule, CNRS UMR7654, Ecole Polytechnique, Institut Polytechnique de Paris, 91128 Palaiseau Cedex, France; (K.D.R.); (J.G.); (A.I.F.); (R.T.); (A.B.); (C.M.); (M.J.); (M.C.N.H.); (E.G.)
| | - Sophie Vacher
- Pharmacogenomics Unit, Department of Genetics, Institut Curie, Paris Descartes University, 75005 Paris, France; (S.V.); (L.C.); (I.B.)
| | - Laura Courtois
- Pharmacogenomics Unit, Department of Genetics, Institut Curie, Paris Descartes University, 75005 Paris, France; (S.V.); (L.C.); (I.B.)
| | - Ivan Bièche
- Pharmacogenomics Unit, Department of Genetics, Institut Curie, Paris Descartes University, 75005 Paris, France; (S.V.); (L.C.); (I.B.)
| | - Alexis M. Gautreau
- Laboratoire de Biologie Structurale de la Cellule, CNRS UMR7654, Ecole Polytechnique, Institut Polytechnique de Paris, 91128 Palaiseau Cedex, France; (K.D.R.); (J.G.); (A.I.F.); (R.T.); (A.B.); (C.M.); (M.J.); (M.C.N.H.); (E.G.)
| |
Collapse
|
7
|
Kawachi K, Tang X, Kasajima R, Yamanaka T, Shimizu E, Katayama K, Yamaguchi R, Yokoyama K, Yamaguchi K, Furukawa Y, Miyano S, Imoto S, Yoshioka E, Washimi K, Okubo Y, Sato S, Yokose T, Miyagi Y. Genetic analysis of low-grade adenosquamous carcinoma of the breast progressing to high-grade metaplastic carcinoma. Breast Cancer Res Treat 2023; 202:563-573. [PMID: 37650999 PMCID: PMC10564816 DOI: 10.1007/s10549-023-07078-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 08/07/2023] [Indexed: 09/01/2023]
Abstract
PURPOSE Low-grade adenosquamous carcinoma (LGASC) is a rare type of metaplastic carcinoma of the breast (MBC) with an indolent clinical course. A few LGASC cases with high-grade transformation have been reported; however, the genetics underlying malignant progression of LGASC remain unclear. METHODS We performed whole-genome sequencing analysis on five MBCs from four patients, including one case with matching primary LGASC and a lymph node metastatic tumor consisting of high-grade MBC with a predominant metaplastic squamous cell carcinoma component (MSC) that progressed from LGASC and three cases of independent de novo MSC. RESULTS Unlike de novo MSC, LGASC and its associated MSC showed no TP53 mutation and tended to contain fewer structural variants than de novo MSC. Both LGASC and its associated MSC harbored the common GNAS c.C2530T:p.Arg844Cys mutation, which was more frequently detected in the cancer cell fraction of MSC. MSC associated with LGASC showed additional pathogenic deletions of multiple tumor-suppressor genes, such as KMT2D and BTG1. Copy number analysis revealed potential 18q loss of heterozygosity in both LGASC and associated MSC. The frequency of SMAD4::DCC fusion due to deletions increased with progression to MSC; however, chimeric proteins were not detected. SMAD4 protein expression was already decreased at the LGASC stage due to unknown mechanisms. CONCLUSION Not only LGASC but also its associated high-grade MBC may be genetically different from de novo high-grade MBC. Progression from LGASC to high-grade MBC may involve the concentration of driver mutations caused by clonal selection and inactivation of tumor-suppressor genes.
Collapse
Affiliation(s)
- Kae Kawachi
- Department of Pathology, Kanagawa Cancer Center, 2-3-2 Nakao, Aasahi-ku, Yokohama, Japan
- Department of Pathology, The Jikei University School of Medicine, 3-25-8 Nishishinbashi, Minato-ku, Tokyo, Japan
| | - Xiaoyan Tang
- Department of Pathology, Nihon University Hospital, 1-6 Kandasurugadai, Chiyoda-ku, Tokyo, Japan
| | - Rika Kasajima
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Aasahi-ku, Yokohama, Japan
- Division of Health Medical Intelligence, Human Genome Center, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, Japan
| | - Takashi Yamanaka
- Department of Breast and Endocrine Surgery, Kanagawa Cancer Center, 2-3-2 Nakao, Aasahi-ku, Yokohama, Japan
| | - Eigo Shimizu
- Division of Health Medical Intelligence, Human Genome Center, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, Japan
| | - Kotoe Katayama
- Division of Health Medical Intelligence, Human Genome Center, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, Japan
| | - Rui Yamaguchi
- Division of Health Medical Intelligence, Human Genome Center, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, Japan
- Division of Cancer Systems Biology, Aichi Cancer Center Research Institute, 1-1 Kanokoden, Chikusa-ku, Nagoya, Japan
- Division of Cancer Informatics, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-ku, Nagoya, Japan
| | - Kazuaki Yokoyama
- Department of Hematology/Oncology, Research Hospital, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Kiyoshi Yamaguchi
- Division of Clinical Genome Research, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, Japan
| | - Yoichi Furukawa
- Division of Clinical Genome Research, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, Japan
| | - Satoru Miyano
- Division of Health Medical Intelligence, Human Genome Center, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, Japan
- Department of Integrated Data Science, Medical and Dental Data Science Center, Tokyo Medical and Dental University, 2-3-10 Kandasurugadai, Chiyoda-ku, Tokyo, Japan
| | - Seiya Imoto
- Division of Health Medical Intelligence, Human Genome Center, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, Japan
| | - Emi Yoshioka
- Department of Pathology, Kanagawa Cancer Center, 2-3-2 Nakao, Aasahi-ku, Yokohama, Japan
| | - Kota Washimi
- Department of Pathology, Kanagawa Cancer Center, 2-3-2 Nakao, Aasahi-ku, Yokohama, Japan
| | - Yoichiro Okubo
- Department of Pathology, Kanagawa Cancer Center, 2-3-2 Nakao, Aasahi-ku, Yokohama, Japan
| | - Shinya Sato
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Aasahi-ku, Yokohama, Japan
| | - Tomoyuki Yokose
- Department of Pathology, Kanagawa Cancer Center, 2-3-2 Nakao, Aasahi-ku, Yokohama, Japan
| | - Yohei Miyagi
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Aasahi-ku, Yokohama, Japan.
| |
Collapse
|
8
|
Park AY, Han MR, Seo BK, Ju HY, Son GS, Lee HY, Chang YW, Choi J, Cho KR, Song SE, Woo OH, Park HS. MRI-based breast cancer radiogenomics using RNA profiling: association with subtypes in a single-center prospective study. Breast Cancer Res 2023; 25:79. [PMID: 37391754 PMCID: PMC10311893 DOI: 10.1186/s13058-023-01668-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 05/31/2023] [Indexed: 07/02/2023] Open
Abstract
BACKGROUND There are few prospective studies on the correlations between MRI features and whole RNA-sequencing data in breast cancer according to molecular subtypes. The purpose of our study was to explore the association between genetic profiles and MRI phenotypes of breast cancer and to identify imaging markers that influences the prognosis and treatment according to subtypes. METHODS From June 2017 to August 2018, MRIs of 95 women with invasive breast cancer were prospectively analyzed, using the breast imaging-reporting and data system and texture analysis. Whole RNA obtained from surgical specimens was analyzed using next-generation sequencing. The association between MRI features and gene expression profiles was analyzed in the entire tumor and subtypes. Gene networks, enriched functions, and canonical pathways were analyzed using Ingenuity Pathway Analysis. The P value for differential expression was obtained using a parametric F test comparing nested linear models and adjusted for multiple testing by reporting Q value. RESULTS In 95 participants (mean age, 53 years ± 11 [standard deviation]), mass lesion type was associated with upregulation of CCL3L1 (sevenfold) and irregular mass shape was associated with downregulation of MIR421 (sixfold). In estrogen receptor-positive cancer with mass lesion type, CCL3L1 (21-fold), SNHG12 (11-fold), and MIR206 (sevenfold) were upregulated, and MIR597 (265-fold), MIR126 (12-fold), and SOX17 (fivefold) were downregulated. In triple-negative breast cancer with increased standard deviation of texture analysis on precontrast T1-weighted imaging, CLEC3A (23-fold), SRGN (13-fold), HSPG2 (sevenfold), KMT2D (fivefold), and VMP1 (fivefold) were upregulated, and IGLC2 (73-fold) and PRDX4 (sevenfold) were downregulated (all, P < 0.05 and Q < 0.1). Gene network and functional analysis showed that mass type estrogen receptor-positive cancers were associated with cell growth, anti-estrogen resistance, and poor survival. CONCLUSION MRI characteristics are associated with the different expressions of genes related to metastasis, anti-drug resistance, and prognosis, depending on the molecular subtypes of breast cancer.
Collapse
Affiliation(s)
- Ah Young Park
- Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Mi-Ryung Han
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea
| | - Bo Kyoung Seo
- Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, 123 Jeokgeum-ro, Danwon-gu, Ansan City, Gyeonggi-do, 15355, Republic of Korea.
| | - Hye-Yeon Ju
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea
| | - Gil Soo Son
- Division of Breast and Endocrine Surgery, Department of Surgery, Korea University Ansan Hospital, Korea University College of Medicine, Ansan City, Gyeonggi-do, Republic of Korea
| | - Hye Yoon Lee
- Division of Breast and Endocrine Surgery, Department of Surgery, Korea University Ansan Hospital, Korea University College of Medicine, Ansan City, Gyeonggi-do, Republic of Korea
| | - Young Woo Chang
- Division of Breast and Endocrine Surgery, Department of Surgery, Korea University Ansan Hospital, Korea University College of Medicine, Ansan City, Gyeonggi-do, Republic of Korea
| | - Jungyoon Choi
- Division of Hematology/Oncology, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Ansan City, Gyeonggi-do, Republic of Korea
| | - Kyu Ran Cho
- Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Sung Eun Song
- Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Ok Hee Woo
- Department of Radiology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Hyun Soo Park
- Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, 123 Jeokgeum-ro, Danwon-gu, Ansan City, Gyeonggi-do, 15355, Republic of Korea
| |
Collapse
|
9
|
He J, Xu T, Zhao F, Guo J, Hu Q. SETD2-H3K36ME3: an important bridge between the environment and tumors. Front Genet 2023; 14:1204463. [PMID: 37359376 PMCID: PMC10288198 DOI: 10.3389/fgene.2023.1204463] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 06/01/2023] [Indexed: 06/28/2023] Open
Abstract
Epigenetic regulation plays an important role in the occurrence, development and treatment of tumors. The histone methyltransferase SET-domain-containing 2 (SETD2) plays a key role in mammalian epigenetic regulation by catalyzing histone methylation and interacting with RNA polymerase II to mediate transcription elongation and mismatch repair. As an important bridge between the environment and tumors, SETD2-H3K36me3 plays an important role in the occurrence and development of tumors. Many tumors, including renal cancer, gastric cancer, lung cancer, are closely related to SETD2 gene mutations. As a key component of common tumor suppressor mechanisms, SETD2-H3K36me3is an important target for clinical disease diagnosis and treatment. Here, we reviewed the structure and function of the SETD2 and how SETD2-H3K36me3 functions as a bridge between the environment and tumors to provide an in-depth understanding of its role in the occurrence and development of various tumors, which is of great significance for future disease diagnosis and treatment.
Collapse
Affiliation(s)
- Jiahui He
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Tangpeng Xu
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Fangrui Zhao
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jin Guo
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Qinyong Hu
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
10
|
Dauch C, Shim S, Cole MW, Pollock NC, Beer AJ, Ramroop J, Klee V, Allain DC, Shakya R, Knoblaugh SE, Kulewsky J, Toland AE. KMT2D loss drives aggressive tumor phenotypes in cutaneous squamous cell carcinoma. Am J Cancer Res 2022; 12:1309-1322. [PMID: 35411237 PMCID: PMC8984905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 02/16/2022] [Indexed: 06/14/2023] Open
Abstract
Cutaneous squamous cell carcinoma (cSCC) is the second most lethal skin cancer. Due to ultraviolet light-induced damage, cSCCs have a high mutation rate, but some genes are more frequently mutated in aggressive cSCCs. Lysine-specific histone methyltransferase 2D (KMT2D) has a two-fold higher mutation frequency in metastatic cSCCs relative to primary non-metastatic associated cSCCs. The role of KMT2D in more aggressive phenotypes in cSCC is uncharacterized. Studies of other tumor types suggest that KMT2D acts to suppress tumor development. To determine whether KMT2D loss has an impact on tumor characteristics, we disrupted KMT2D in a cSCC cell line using CRISPR-cas9 and performed phenotypic analyses. KMT2D loss modestly increased cell proliferation and colony formation (1.4- and 1.6-fold respectively). Cells lacking KMT2D showed increased rates of migration and faster cell cycle progression. In xenograft models, tumors with KMT2D loss showed slight increases in mitotic indices. Collectively, these findings suggest that KMT2D loss-of-function mutations may promote more aggressive and invasive behaviors in cSCC, suggesting that KMT2D-related pathways could be targets for cancer therapies. Future studies to determine the downstream genes and mechanism of phenotypic effect are needed.
Collapse
Affiliation(s)
- Cara Dauch
- Department of Cancer Biology and Genetics, The Ohio State University College of MedicineColumbus, OH 43210, USA
| | - Sharon Shim
- Central Michigan University College of MedicineMount Pleasant, MI 48858, USA
| | - Matthew Wyatt Cole
- Department of Cancer Biology and Genetics, The Ohio State University College of MedicineColumbus, OH 43210, USA
- Department of Radiation Oncology, The Ohio State UniversityColumbus, OH 43210, USA
| | - Nijole C Pollock
- Department of Cancer Biology and Genetics, The Ohio State University College of MedicineColumbus, OH 43210, USA
| | - Abigail J Beer
- Department of Cancer Biology and Genetics, The Ohio State University College of MedicineColumbus, OH 43210, USA
| | - Johnny Ramroop
- Department of Cancer Biology and Genetics, The Ohio State University College of MedicineColumbus, OH 43210, USA
| | - Victoria Klee
- Department of Internal Medicine, Division of Human Genetics, The Ohio State UniversityColumbus, OH 43210, USA
| | - Dawn C Allain
- Department of Internal Medicine, Division of Human Genetics, The Ohio State UniversityColumbus, OH 43210, USA
| | - Reena Shakya
- Comprehensive Cancer Center, The Ohio State UniversityColumbus, OH 43210, USA
| | - Sue E Knoblaugh
- Department of Veterinary Biosciences, The Ohio State UniversityColumbus, OH 43210, USA
| | - Jesse Kulewsky
- Department of Pathology, The Ohio State University Wexner Medical CenterColumbus, OH 43210, USA
| | - Amanda Ewart Toland
- Department of Cancer Biology and Genetics, The Ohio State University College of MedicineColumbus, OH 43210, USA
- Department of Internal Medicine, Division of Human Genetics, The Ohio State UniversityColumbus, OH 43210, USA
- Comprehensive Cancer Center, The Ohio State UniversityColumbus, OH 43210, USA
| |
Collapse
|
11
|
Sabatier R, Vicier C, Garnier S, Guille A, Carbuccia N, Isambert N, Dalenc F, Robert M, Levy C, Pakradouni J, Adelaïde J, Chaffanet M, Sfumato P, Mamessier E, Bertucci F, Goncalves A. Circulating tumor DNA predicts efficacy of a dual AKT/p70S6K inhibitor (LY2780301) plus paclitaxel in metastatic breast cancer: plasma analysis of the TAKTIC phase IB/II study. Mol Oncol 2022; 16:2057-2070. [PMID: 35122700 PMCID: PMC9120890 DOI: 10.1002/1878-0261.13188] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/07/2022] [Accepted: 02/03/2022] [Indexed: 11/10/2022] Open
Abstract
The phosphatidylinositol-3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) pathway is frequently activated in HER2-negative breast cancer and may play a role in taxane resistance. The phase IB/II TAKTIC trial (NCT01980277) has shown that combining a dual AKT and p70 ribosomal protein S6 kinase (p70S6K) inhibitor (LY2780301) taken orally with weekly paclitaxel in HER2-negative advanced breast cancer is feasible, with preliminary evidence of efficacy. We wanted to explore whether circulating tumor DNA (ctDNA) may be a surrogate marker of treatment efficacy in this setting. Serial plasma samples were collected and cell-free DNA was sequenced using low-coverage whole-genome sequencing, and analysis was completed with droplet digital PCR for some patients with driver mutations. Baseline tumor fraction (TF) and TF after 7 weeks on treatment were compared to progression-free survival (PFS) and overall response rate. We also explored circulating copy number alterations associated with treatment failure. Of the 51 patients enrolled in the TAKTIC trial, at least one plasma sample was available for 44 cases (96 time points). All patients with tumor TP53, PI3KCA or AKT1 mutations harbored at least one of these alterations in plasma. TF at inclusion was correlated to PFS (6m-PFS was 92% for ctDNAneg patients vs 68% for ctDNApos cases; HR=3.45, 95%CI [1.34-8.90], p=0.007). ctDNA status at week 7 was not correlated to prognosis. Even though most circulating copy number alterations were conserved at disease progression, some genomic regions of interest were altered in post-progression samples. In conclusions, ctDNA detection at baseline was associated with shorter PFS in patients included in the TAKTIC trial. Plasma-based copy number analysis may help to identify alterations involved in resistance to treatment.
Collapse
Affiliation(s)
- Renaud Sabatier
- Aix-Marseille Univ, Inserm, CNRS, Institut Paoli-Calmettes, CRCM-Predictive Oncology laboratory, Marseille, France.,Aix-Marseille Univ, Inserm, CNRS, Institut Paoli-Calmettes, Department of Medical Oncology, Marseille, France
| | - Cécile Vicier
- Aix-Marseille Univ, Inserm, CNRS, Institut Paoli-Calmettes, Department of Medical Oncology, Marseille, France
| | - Séverine Garnier
- Aix-Marseille Univ, Inserm, CNRS, Institut Paoli-Calmettes, CRCM-Predictive Oncology laboratory, Marseille, France
| | - Arnaud Guille
- Aix-Marseille Univ, Inserm, CNRS, Institut Paoli-Calmettes, CRCM-Predictive Oncology laboratory, Marseille, France
| | - Nadine Carbuccia
- Aix-Marseille Univ, Inserm, CNRS, Institut Paoli-Calmettes, CRCM-Predictive Oncology laboratory, Marseille, France
| | - Nicolas Isambert
- Drug Development Department, Centre Georges François Leclerc, Dijon, France
| | - Florence Dalenc
- Department of Medical Oncology, Institut Claudius Regaud, IUCT-Oncopole, CRCT, Inserm, Toulouse, France
| | - Marie Robert
- Institut de Cancérologie de l'Ouest-René Gauducheau, Saint-Herblain, France
| | - Christelle Levy
- Centre François Baclesse, Department of Medical Oncology, Caen, France
| | - Jihane Pakradouni
- Depatment of Clinical Research and Innovation, Institut Paoli-Calmettes, Marseille, France
| | - José Adelaïde
- Aix-Marseille Univ, Inserm, CNRS, Institut Paoli-Calmettes, CRCM-Predictive Oncology laboratory, Marseille, France
| | - Max Chaffanet
- Aix-Marseille Univ, Inserm, CNRS, Institut Paoli-Calmettes, CRCM-Predictive Oncology laboratory, Marseille, France
| | - Patrick Sfumato
- Depatment of Clinical Research and Innovation, Institut Paoli-Calmettes, Marseille, France
| | - Emilie Mamessier
- Aix-Marseille Univ, Inserm, CNRS, Institut Paoli-Calmettes, Department of Medical Oncology, Marseille, France
| | - François Bertucci
- Aix-Marseille Univ, Inserm, CNRS, Institut Paoli-Calmettes, CRCM-Predictive Oncology laboratory, Marseille, France.,Aix-Marseille Univ, Inserm, CNRS, Institut Paoli-Calmettes, Department of Medical Oncology, Marseille, France
| | - Anthony Goncalves
- Aix-Marseille Univ, Inserm, CNRS, Institut Paoli-Calmettes, CRCM-Predictive Oncology laboratory, Marseille, France.,Aix-Marseille Univ, Inserm, CNRS, Institut Paoli-Calmettes, Department of Medical Oncology, Marseille, France
| |
Collapse
|
12
|
Wang C, Feng G, Zhu J, Wei K, Huang C, Wu Z, Yu Y, Qin G. Developing an immune signature for triple-negative breast cancer to predict prognosis and immune checkpoint inhibitor response. Future Oncol 2022; 18:1055-1066. [PMID: 35105171 DOI: 10.2217/fon-2021-0600] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Aim: We aimed to develop a new signature based on immune-related genes to predict prognosis and response to immune checkpoint inhibitors in patients with triple-negative breast cancer (TNBC). Materials & methods: Single-sample gene set enrichment was used to develop an immune-based prognostic signature (IPRS) for TNBC patients. We conducted multivariate Cox analysis to evaluate the prognosis value of the IPRS. Result: An IPRS based on 66 prognostic genes was developed. Multivariate Cox analysis indicated that the IPRS was an independent factor for prognosis. PD-1, PD-L1, PD-L2 and CTLA4 gene expression was higher in the low-risk group, suggesting IPRS could predict the response to immune checkpoint inhibitors. Conclusion: The IPRS might be a reliable signature to predict TNBC patients' prognosis and response to immune checkpoint inhibitors, but needs prospective validation.
Collapse
Affiliation(s)
- Ce Wang
- Department of Biostatistics, School of Public Health, & The Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, China.,Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University & Capital Medical University, Beijing 100083, China
| | - Guoshuang Feng
- Big Data & Engineering Research Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China.,Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University & Capital Medical University, Beijing 100083, China
| | - Jingjing Zhu
- Department of Biostatistics, School of Public Health, & The Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, China
| | - Kecheng Wei
- Department of Biostatistics, School of Public Health, & The Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, China
| | - Chen Huang
- Department of Biostatistics, School of Public Health, & The Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, China
| | - Zhenyu Wu
- Department of Biostatistics, School of Public Health, & The Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, China
| | - Yongfu Yu
- Department of Biostatistics, School of Public Health, & The Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, China
| | - Guoyou Qin
- Department of Biostatistics, School of Public Health, & The Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, China.,Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University & Capital Medical University, Beijing 100083, China
| |
Collapse
|
13
|
Role of histone demethylases and histone methyltransferases in triple-negative breast cancer: Epigenetic mnemonics. Life Sci 2022; 292:120321. [PMID: 35031259 DOI: 10.1016/j.lfs.2022.120321] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/29/2021] [Accepted: 01/06/2022] [Indexed: 12/30/2022]
Abstract
Triple-negative breast cancer (TNBC) is a particularly lethal subtype of breast cancer owing to its heterogeneity, high drug resistance, poor prognosis and lack of therapeutic targets. Recent insights into the complexity of TNBC have been explained by epigenetic regulation and its ability to modulate certain oncogenes and tumour suppressor genes. This has opened an emerging area in anti-cancer therapy using epigenetic modulating drugs, highlighting the epigenetic reprogramming during tumorigenesis and tumour development. Histone methylation and demethylation are such dynamic epigenetic mechanisms mediated by histone methyltransferases (HMTs) and histone demethylases (HDMs), respectively. The interplay between HMTs and HDMs in histone methylation extrapolates their viability as druggable epigenetic targets in TNBC. In this review, we aim to summarize recent progress in the field of epigenetics focusing on HMTs and HDMs in TNBC development and their potential use in targeted therapy for TNBC management.
Collapse
|
14
|
The MLL3/4 H3K4 methyltransferase complex in establishing an active enhancer landscape. Biochem Soc Trans 2021; 49:1041-1054. [PMID: 34156443 PMCID: PMC8286814 DOI: 10.1042/bst20191164] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/21/2021] [Accepted: 05/24/2021] [Indexed: 12/23/2022]
Abstract
Enhancers are cis-regulatory elements that play essential roles in tissue-specific gene expression during development. Enhancer function in the expression of developmental genes requires precise regulation, while deregulation of enhancer function could be the main cause of tissue-specific cancer development. MLL3/KMT2C and MLL4/KMT2D are two paralogous histone modifiers that belong to the SET1/MLL (also named COMPASS) family of lysine methyltransferases and play critical roles in enhancer-regulated gene activation. Importantly, large-scale DNA sequencing studies have revealed that they are amongst the most frequently mutated genes associated with human cancers. MLL3 and MLL4 form identical multi-protein complexes for modifying mono-methylation of histone H3 lysine 4 (H3K4) at enhancers, which together with the p300/CBP-mediated H3K27 acetylation can generate an active enhancer landscape for long-range target gene activation. Recent studies have provided a better understanding of the possible mechanisms underlying the roles of MLL3/MLL4 complexes in enhancer regulation. Moreover, accumulating studies offer new insights into our knowledge of the potential role of MLL3/MLL4 in cancer development. In this review, we summarize recent evidence on the molecular mechanisms of MLL3/MLL4 in the regulation of active enhancer landscape and long-range gene expression, and discuss their clinical implications in human cancers.
Collapse
|
15
|
Gao C, Li H, Liu C, Xu X, Zhuang J, Zhou C, Liu L, Feng F, Sun C. Tumor Mutation Burden and Immune Invasion Characteristics in Triple Negative Breast Cancer: Genome High-Throughput Data Analysis. Front Immunol 2021; 12:650491. [PMID: 33968045 PMCID: PMC8097167 DOI: 10.3389/fimmu.2021.650491] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 04/01/2021] [Indexed: 12/14/2022] Open
Abstract
In recent years, the emergence of immunotherapy has provided a new perspective for the treatment and management of triple-negative breast cancer (TNBC). However, the relationship between tumor mutation burden (TMB) and immune infiltration and the prognosis of TNBC remains unclear. In this study, to explore the immunogenicity of TNBC, we divided patients with TNBC into high and low TMB groups based on the somatic mutation data of TNBC in The Cancer Genome Atlas (TCGA), and screened out genes with mutation rate ≥10. Then, Kaplan-Meier survival analysis revealed that the 5-year survival rate of the high TMB group was much higher than that of the low TMB group and the two groups also showed differences in immune cell infiltration. Further exploration found that the FAT3 gene, which displays significant difference and a higher mutation rate between the two groups, is not only significantly related to the prognosis of TNBC patients but also exhibits difference in immune cell infiltration between the wild group and the mutant group of the FAT3 gene. The results of gene set enrichment analysis and drug sensitivity analysis further support the importance of the FAT3 gene in TNBC. This study reveals the characteristics of TMB and immune cell infiltration in triple-negative breast cancer and their relationship with prognosis, to provide new biomarkers and potential treatment options for the future treatment of TNBC. The FAT3 gene, as a risk predictor gene of TNBC, is considered a potential biological target and may provide new insight for the treatment of TNBC.
Collapse
Affiliation(s)
- Chundi Gao
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Huayao Li
- College of Basic Medical, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Cun Liu
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiaowei Xu
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jing Zhuang
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China
| | - Chao Zhou
- College of Basic Medical, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lijuan Liu
- College of Basic Medical, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Fubin Feng
- College of Basic Medical, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Changgang Sun
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China.,Qingdao Academy of Chinese Medical Sciences, Shandong University of Traditional Chinese Medicine, Qingdao, China
| |
Collapse
|
16
|
Reactive oxygen species (ROS): Critical roles in breast tumor microenvironment. Crit Rev Oncol Hematol 2021; 160:103285. [DOI: 10.1016/j.critrevonc.2021.103285] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 01/18/2021] [Accepted: 02/27/2021] [Indexed: 02/06/2023] Open
|
17
|
Zolota V, Tzelepi V, Piperigkou Z, Kourea H, Papakonstantinou E, Argentou MI, Karamanos NK. Epigenetic Alterations in Triple-Negative Breast Cancer-The Critical Role of Extracellular Matrix. Cancers (Basel) 2021; 13:cancers13040713. [PMID: 33572395 PMCID: PMC7916242 DOI: 10.3390/cancers13040713] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/04/2021] [Accepted: 02/05/2021] [Indexed: 12/11/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subgroup of breast cancer characterized by genomic complexity and therapeutic options limited to only standard chemotherapy. Although it has been suggested that stratifying TNBC patients by pathway-specific molecular alterations may predict benefit from specific therapeutic agents, application in routine clinical practice has not yet been established. There is a growing body of the literature supporting that epigenetic modifications comprised by DNA methylation, chromatin remodeling and non-coding RNAs play a fundamental role in TNBC pathogenesis. Extracellular matrix (ECM) is a highly dynamic 3D network of macromolecules with structural and cellular regulatory roles. Alterations in the expression of ECM components result in uncontrolled matrix remodeling, thus affecting its ability to regulate vital functions of cancer cells, including proliferation, migration, adhesion, invasion and epithelial-to-mesenchymal transition (EMT). Recent molecular data highlight the major role of tumor microenvironment and ECM alterations in TNBC and approaches for targeting tumor microenvironment have recently been recognized as potential therapeutic strategies. Notably, many of the ECM/EMT modifications in cancer are largely driven by epigenetic events, highlighting the pleiotropic effects of the epigenetic network in TNBC. This article presents and critically discusses the current knowledge on the epigenetic alterations correlated with TNBC pathogenesis, with emphasis on those associated with ECM/EMT modifications, their prognostic and predictive value and their use as therapeutic targets.
Collapse
Affiliation(s)
- Vasiliki Zolota
- Department of Pathology, School of Medicine, University of Patras, 26504 Rion, Greece; (V.T.); (H.K.)
- Correspondence: ; Tel.: +30-0693613366
| | - Vasiliki Tzelepi
- Department of Pathology, School of Medicine, University of Patras, 26504 Rion, Greece; (V.T.); (H.K.)
| | - Zoi Piperigkou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26110 Patras, Greece; (Z.P.); (N.K.K.)
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), 26110 Patras, Greece
| | - Helen Kourea
- Department of Pathology, School of Medicine, University of Patras, 26504 Rion, Greece; (V.T.); (H.K.)
| | - Efthymia Papakonstantinou
- Department of Gynecology and Obstetrics School of Medicine, University of Patras, 26504 Rion, Greece;
| | - Maria-Ioanna Argentou
- Department of Surgery, School of Medicine, University of Patras, 26504 Rion, Greece;
| | - Nikos K. Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26110 Patras, Greece; (Z.P.); (N.K.K.)
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), 26110 Patras, Greece
| |
Collapse
|
18
|
Chen R, Zhao WQ, Fang C, Yang X, Ji M. Histone methyltransferase SETD2: a potential tumor suppressor in solid cancers. J Cancer 2020; 11:3349-3356. [PMID: 32231741 PMCID: PMC7097956 DOI: 10.7150/jca.38391] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Accepted: 02/09/2020] [Indexed: 12/24/2022] Open
Abstract
Epigenetic regulation plays an important role in the occurrence, development and treatment of malignant tumors; and a great deal of attention has been paid to the histone methylation level in recent years. As a 230-kD epigenetic regulator, the histone H3 lysine 36 histone (H3K36) methyltransferase SETD2 is a key enzyme of the nuclear receptor SET domain-containing (NSD) family, which is associated with a specific hyperphosphorylated domain, a large subunit of RNA polymerase II (RNAPII), named RNAPII subunit B1 (RPB1), and SETD2 which methylates the ly-36 position of dimethylated histone H3 (H3K36me2) to generate trimethylated H3K36 (H3K36me3). SETD2 is involved in various cellular processes, including transcriptional regulation, DNA damage repair, non-histone protein-related functions and some other processes. Great efforts of high-throughput sequencing have revealed that SETD2 is mutated or its function is lost in a range of solid cancers, including renal cancer, gastrointestinal cancer, lung cancer, pancreatic cancer, osteosarcoma, and so on. Mutation, or functional loss, of the SETD2 gene produces dysfunction in corresponding tumor tissue proteins, leading to tumorigenesis, progression, chemotherapy resistance, and unfavorable prognosis, suggesting that SETD2 possibly acts as a tumor suppressor. However, its underlying mechanism remains largely unexplored. In the present study, we summarized the latest advances of effects of SETD2 expression at the mRNA and protein levels in solid cancers, and its potential molecular and cellular functions as well as clinical applications were also reviewed.
Collapse
Affiliation(s)
- Rui Chen
- Department of Oncology, the Third Affiliated Hospital of Soochow University, The First People's Hospital of Changzhou, No. 185 Juqian Road, Tianning District, Changzhou 213003, China
| | - Wei-Qing Zhao
- Department of Oncology, the Third Affiliated Hospital of Soochow University, The First People's Hospital of Changzhou, No. 185 Juqian Road, Tianning District, Changzhou 213003, China
| | - Cheng Fang
- Department of Oncology, the Third Affiliated Hospital of Soochow University, The First People's Hospital of Changzhou, No. 185 Juqian Road, Tianning District, Changzhou 213003, China
| | - Xin Yang
- Department of Oncology, the Third Affiliated Hospital of Soochow University, The First People's Hospital of Changzhou, No. 185 Juqian Road, Tianning District, Changzhou 213003, China
| | - Mei Ji
- Department of Oncology, the Third Affiliated Hospital of Soochow University, The First People's Hospital of Changzhou, No. 185 Juqian Road, Tianning District, Changzhou 213003, China
| |
Collapse
|
19
|
The Rac3 GTPase in Neuronal Development, Neurodevelopmental Disorders, and Cancer. Cells 2019; 8:cells8091063. [PMID: 31514269 PMCID: PMC6770886 DOI: 10.3390/cells8091063] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 09/06/2019] [Accepted: 09/08/2019] [Indexed: 12/23/2022] Open
Abstract
Rho family small guanosine triphosphatases (GTPases) are important regulators of the cytoskeleton, and are critical in many aspects of cellular and developmental biology, as well as in pathological processes such as intellectual disability and cancer. Of the three members of the family, Rac3 has a more restricted expression in normal tissues compared to the ubiquitous member of the family, Rac1. The Rac3 polypeptide is highly similar to Rac1, and orthologues of the gene for Rac3 have been found only in vertebrates, indicating the late appearance of this gene during evolution. Increasing evidence over the past few years indicates that Rac3 plays an important role in neuronal development and in tumor progression, with specificities that distinguish the functions of Rac3 from the established functions of Rac1 in these processes. Here, results highlighting the importance of Rac3 in distinct aspects of neuronal development and tumor cell biology are presented, in support of the non-redundant role of different members of the two Rac GTPases in physiological and pathological processes.
Collapse
|