1
|
Anobile DP, Salaroglio IC, Tabbò F, La Vecchia S, Akman M, Napoli F, Bungaro M, Benso F, Aldieri E, Bironzo P, Kopecka J, Passiglia F, Righi L, Novello S, Scagliotti GV, Riganti C. Autocrine 17-β-Estradiol/Estrogen Receptor-α Loop Determines the Response to Immune Checkpoint Inhibitors in Non-Small Cell Lung Cancer. Clin Cancer Res 2023; 29:3958-3973. [PMID: 37285115 DOI: 10.1158/1078-0432.ccr-22-3949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 04/17/2023] [Accepted: 06/05/2023] [Indexed: 06/08/2023]
Abstract
PURPOSE The response to immune checkpoint inhibitors (ICI) often differs between genders in non-small cell lung cancer (NSCLC), but metanalyses results are controversial, and no clear mechanisms are defined. We aim at clarifying the molecular circuitries explaining the differential gender-related response to anti-PD-1/anti-PD-L1 agents in NSCLC. EXPERIMENTAL DESIGN We prospectively analyzed a cohort of patients with NSCLC treated with ICI as a first-line approach, and we identified the molecular mechanisms determining the differential efficacy of ICI in 29 NSCLC cell lines of both genders, recapitulating patients' phenotype. We validated new immunotherapy strategies in mice bearing NSCLC patient-derived xenografts and human reconstituted immune system ("immune-PDXs"). RESULTS In patients, we found that estrogen receptor α (ERα) was a predictive factor of response to pembrolizumab, stronger than gender and PD-L1 levels, and was directly correlated with PD-L1 expression, particularly in female patients. ERα transcriptionally upregulated CD274/PD-L1 gene, more in females than in males. This axis was activated by 17-β-estradiol, autocrinely produced by intratumor aromatase, and by the EGFR-downstream effectors Akt and ERK1/2 that activated ERα. The efficacy of pembrolizumab in immune-PDXs was significantly improved by the aromatase inhibitor letrozole, which reduced PD-L1 and increased the percentage of antitumor CD8+T-lymphocytes, NK cells, and Vγ9Vδ2 T-lymphocytes, producing durable control and even tumor regression after continuous administration, with maximal benefit in 17-β-estradiol/ERα highfemale immune-xenografts. CONCLUSIONS Our work unveils that 17-β-estradiol/ERα status predicts the response to pembrolizumab in patients with NSCLC. Second, we propose aromatase inhibitors as new gender-tailored immune-adjuvants in NSCLC. See related commentary by Valencia et al., p. 3832.
Collapse
Affiliation(s)
| | | | - Fabrizio Tabbò
- Thoracic Oncology Unit, Department of Oncology at San Luigi Gonzaga Hospital, University of Torino, Torino, Italy
| | | | - Muhlis Akman
- Department of Oncology, University of Torino, Torino, Italy
| | - Francesca Napoli
- Pathology Unit, Department of Oncology at San Luigi Gonzaga Hospital, University of Torino, Torino, Italy
| | - Maristella Bungaro
- Thoracic Oncology Unit, Department of Oncology at San Luigi Gonzaga Hospital, University of Torino, Torino, Italy
| | - Federica Benso
- Pathology Unit, Department of Oncology at San Luigi Gonzaga Hospital, University of Torino, Torino, Italy
| | | | - Paolo Bironzo
- Thoracic Oncology Unit, Department of Oncology at San Luigi Gonzaga Hospital, University of Torino, Torino, Italy
| | - Joanna Kopecka
- Department of Oncology, University of Torino, Torino, Italy
| | - Francesco Passiglia
- Thoracic Oncology Unit, Department of Oncology at San Luigi Gonzaga Hospital, University of Torino, Torino, Italy
| | - Luisella Righi
- Pathology Unit, Department of Oncology at San Luigi Gonzaga Hospital, University of Torino, Torino, Italy
| | - Silvia Novello
- Thoracic Oncology Unit, Department of Oncology at San Luigi Gonzaga Hospital, University of Torino, Torino, Italy
| | - Giorgio V Scagliotti
- Thoracic Oncology Unit, Department of Oncology at San Luigi Gonzaga Hospital, University of Torino, Torino, Italy
| | - Chiara Riganti
- Department of Oncology, University of Torino, Torino, Italy
- Molecular Biotechnology Center "Guido Tarone", University of Torino, Torino, Italy
| |
Collapse
|
2
|
Chen B, Hu H, Chen X. From Basic Science to Clinical Practice: The Role of Cancerous Inhibitor of Protein Phosphatase 2A (CIP2A)/p90 in Cancer. Front Genet 2023; 14:1110656. [PMID: 36911405 PMCID: PMC9998691 DOI: 10.3389/fgene.2023.1110656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 02/03/2023] [Indexed: 03/14/2023] Open
Abstract
Cancerous inhibitor of protein phosphatase 2A (CIP2A), initially reported as a tumor-associated antigen (known as p90), is highly expressed in most solid and hematological tumors. The interaction of CIP2A/p90, protein phosphatase 2A (PP2A), and c-Myc can hinder the function of PP2A toward c-Myc S62 induction, thus stabilizing c-Myc protein, which represents a potential role of CIP2A/p90 in tumorigeneses such as cell proliferation, invasion, and migration, as well as cancer drug resistance. The signaling pathways and regulation networks of CIP2A/p90 are complex and not yet fully understood. Many previous studies have also demonstrated that CIP2A/p90 can be used as a potential therapeutic cancer target. In addition, the autoantibody against CIP2A/p90 in sera may be used as a promising biomarker in the diagnosis of certain types of cancer. In this Review, we focus on recent advances relating to CIP2A/p90 and their implications for future research.
Collapse
Affiliation(s)
- Beibei Chen
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China.,Zhengzhou Key Laboratory for Precision Therapy of Gastrointestinal Cancer, Zhengzhou, Henan, China
| | - Huihui Hu
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China.,Zhengzhou Key Laboratory for Precision Therapy of Gastrointestinal Cancer, Zhengzhou, Henan, China
| | - Xiaobing Chen
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China.,Zhengzhou Key Laboratory for Precision Therapy of Gastrointestinal Cancer, Zhengzhou, Henan, China
| |
Collapse
|
3
|
Brink H, van Senten JR, De Vries-van Leeuwen IJ, da Costa Pereira D, Piersma SR, Jimenez CR, Centorrino F, Ottmann C, Siderius M, Smit MJ, de Boer AH. Fusicoccin-A Targets Cancerous Inhibitor of Protein Phosphatase 2A by Stabilizing a C-Terminal Interaction with 14-3-3. ACS Chem Biol 2022; 17:2972-2978. [PMID: 36255265 PMCID: PMC9679992 DOI: 10.1021/acschembio.2c00299] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The cancerous inhibitor of protein phosphatase 2A (CIP2A) is an oncoprotein found overexpressed in many types of cancer. CIP2A has been shown to stabilize oncoproteins such as cMYC by shielding them from PP2A-mediated dephosphorylation. Here we report that the penultimate residue Ser904 in the C-terminus of CIP2A can be phosphorylated to create a binding site for the regulatory protein 14-3-3. We demonstrate that 14-3-3 is a new interaction partner of CIP2A. The 14-3-3/CIP2A C-terminal interaction complex can be targeted by the protein-protein interaction (PPI) stabilizer fusicoccin-A (FC-A), resulting in enhanced levels of phosphorylated Ser904. FC-A treatment of TNBC cells leads to the increased association of CIP2A with 14-3-3. We show that the composite interface between 14 and 3-3 and CIP2A's C-terminus can be targeted by the PPI stabilizer FC-A, providing a new interface that could potentially be exploited to modulate CIP2A's activity.
Collapse
Affiliation(s)
- Hendrik
J. Brink
- Amsterdam
Institute for Molecular and Life Sciences (AIMMS), Division of Medicinal
Chemistry, Faculty of Sciences, Vrije Universiteit, De Boelelaan 1108, Amsterdam 1081 HZ, The Netherlands
| | - Jeffrey R. van Senten
- Amsterdam
Institute for Molecular and Life Sciences (AIMMS), Division of Medicinal
Chemistry, Faculty of Sciences, Vrije Universiteit, De Boelelaan 1108, Amsterdam 1081 HZ, The Netherlands
| | - Ingrid J. De Vries-van Leeuwen
- Amsterdam
Institute for Molecular and Life Sciences (AIMMS), Division of Medicinal
Chemistry, Faculty of Sciences, Vrije Universiteit, De Boelelaan 1108, Amsterdam 1081 HZ, The Netherlands
| | - Daniel da Costa Pereira
- Amsterdam
Institute for Molecular and Life Sciences (AIMMS), Division of Medicinal
Chemistry, Faculty of Sciences, Vrije Universiteit, De Boelelaan 1108, Amsterdam 1081 HZ, The Netherlands
| | - Sander R. Piersma
- OncoProteomics
Laboratory, Department of Medical Oncology, Amsterdam University Medical Center (VUmc), 1081 HV Amsterdam, The Netherlands
| | - Connie R. Jimenez
- OncoProteomics
Laboratory, Department of Medical Oncology, Amsterdam University Medical Center (VUmc), 1081 HV Amsterdam, The Netherlands
| | - Federica Centorrino
- Laboratory
of Chemical Biology, Department of Biomedical Engineering and Institute
for Complex Molecular Systems (ICMS), Eindhoven
University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Christian Ottmann
- Laboratory
of Chemical Biology, Department of Biomedical Engineering and Institute
for Complex Molecular Systems (ICMS), Eindhoven
University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Marco Siderius
- Amsterdam
Institute for Molecular and Life Sciences (AIMMS), Division of Medicinal
Chemistry, Faculty of Sciences, Vrije Universiteit, De Boelelaan 1108, Amsterdam 1081 HZ, The Netherlands
| | - Martine J. Smit
- Amsterdam
Institute for Molecular and Life Sciences (AIMMS), Division of Medicinal
Chemistry, Faculty of Sciences, Vrije Universiteit, De Boelelaan 1108, Amsterdam 1081 HZ, The Netherlands
| | - Albertus H. de Boer
- Amsterdam
Institute for Molecular and Life Sciences (AIMMS), Division of Medicinal
Chemistry, Faculty of Sciences, Vrije Universiteit, De Boelelaan 1108, Amsterdam 1081 HZ, The Netherlands,
| |
Collapse
|
4
|
Gong S, Hao X, Bi Y, Yang C, Wang W, Mickael HK, Zhang Y, Chen S, Qian Z, Huang F, Wei D, Yu W. Hepatitis E viral infection regulates estrogen signaling pathways: Inhibition of the cAMPK-PKA-CREB and PI3K-AKT-mTOR signaling pathways. J Med Virol 2021; 93:3769-3778. [PMID: 33128390 DOI: 10.1002/jmv.26641] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 10/17/2020] [Accepted: 10/25/2020] [Indexed: 12/22/2022]
Abstract
Hepatitis E virus (HEV) infection has become a global concern with high mortality rates among pregnant women, especially those in their third trimester of pregnancy. Estrogen plays an important role in mediating the body, regulating physiological and pathological processes. Estrogen is activated by binding to estrogen receptors (ERs) and mediates rapid signaling events by pathways that involve transmembrane ERs. Our previous study had confirmed that high estrogen levels during pregnancy are associated with high HEV titers. However, the association between HEV infection and estrogen signaling pathways remains unclear. In the present study, the regulation of estrogen signaling pathways by HEV infection was evaluated. Results demonstrated that HEV infection significantly inhibits the cAMP-PKA-CREB and PI3K-AKT-mTOR signaling pathways, but is independent of the Ras-Raf-MEK-ERK signaling pathway. In summary, the increasing estrogen levels and highly activated ERα during pregnancy aggravates HEV replication. The exacerbation of HEV replication, in turn, inhibits ERα expression and suppresses both cAMP-PKA-CREB and PI3K-AKT-mTOR signaling pathways.
Collapse
Affiliation(s)
- Shilin Gong
- Medical School, Kunming University of Science and Technology, Kunming, PR China
- Yunnan Provincial Key Laboratory of Clinical Virology, Kunming, PR China
| | - Xianhui Hao
- Medical School, Kunming University of Science and Technology, Kunming, PR China
| | - Yanhong Bi
- Medical School, Kunming University of Science and Technology, Kunming, PR China
| | - Chenchen Yang
- Medical School, Kunming University of Science and Technology, Kunming, PR China
| | - Wenjing Wang
- Medical School, Kunming University of Science and Technology, Kunming, PR China
| | - Houfack K Mickael
- Medical School, Kunming University of Science and Technology, Kunming, PR China
| | - Yike Zhang
- Medical School, Kunming University of Science and Technology, Kunming, PR China
| | - Shuangfeng Chen
- Medical School, Kunming University of Science and Technology, Kunming, PR China
| | - Zhongyao Qian
- Medical School, Kunming University of Science and Technology, Kunming, PR China
| | - Fen Huang
- Medical School, Kunming University of Science and Technology, Kunming, PR China
- Yunnan Provincial Key Laboratory of Clinical Virology, Kunming, PR China
| | - Daqiao Wei
- Medical School, Kunming University of Science and Technology, Kunming, PR China
| | - Wenhai Yu
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, PR China
| |
Collapse
|
5
|
Remmerie M, Janssens V. PP2A: A Promising Biomarker and Therapeutic Target in Endometrial Cancer. Front Oncol 2019; 9:462. [PMID: 31214504 PMCID: PMC6558005 DOI: 10.3389/fonc.2019.00462] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 05/14/2019] [Indexed: 12/14/2022] Open
Abstract
Over the last decade, the use of targeted therapies has immensely increased in the treatment of cancer. However, treatment for endometrial carcinomas (ECs) has lagged behind, although potential molecular markers have been identified. This is particularly problematic for the type II ECs, since these aggressive tumors are usually not responsive toward the current standard therapies. Therefore, type II ECs are responsible for most EC-related deaths, indicating the need for new treatment options. Interestingly, molecular analyses of type II ECs have uncovered frequent genetic alterations (up to 40%) in PPP2R1A, encoding the Aα subunit of the tumor suppressive heterotrimeric protein phosphatase type 2A (PP2A). PPP2R1A mutations were also reported in type I ECs and other common gynecologic cancers, albeit at much lower frequencies (0-7%). Nevertheless, PP2A inactivation in the latter cancer types is common via other mechanisms, in particular by increased expression of Cancerous Inhibitor of PP2A (CIP2A) and PP2A Methylesterase-1 (PME-1) proteins. In this review, we discuss the therapeutic potential of direct and indirect PP2A targeting compounds, possibly in combination with other anti-cancer drugs, in EC. Furthermore, we investigate the potential of the PP2A status as a predictive and/or prognostic marker for type I and II ECs.
Collapse
Affiliation(s)
| | - Veerle Janssens
- Laboratory of Protein Phosphorylation and Proteomics, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
6
|
Hayashi T, Hikichi M, Yukitake J, Wakatsuki T, Nishio E, Utsumi T, Harada N. Forskolin increases the effect of everolimus on aromatase inhibitor-resistant breast cancer cells. Oncotarget 2018; 9:23451-23461. [PMID: 29805747 PMCID: PMC5955115 DOI: 10.18632/oncotarget.25217] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 04/06/2018] [Indexed: 01/31/2023] Open
Abstract
Aromatase inhibitor (AI) resistance is a major obstacle in the treatment of estrogen receptor-positive breast cancer. Everolimus (EVE) ameliorates AI-resistant breast cancer and is therefore used in cancer treatment. However, some patients show resistance to EVE. Here, we used 30 clones of long-term estrogen-deprived (LTED) MCF-7 cells as a model of AI-resistant breast cancer. We examined changes in protein phosphatase type 2A (PP2A) and cancerous inhibitor of PP2A (CIP2A), a negative regulator of PP2A, in LTED cells treated with EVE. In LTED cells with high sensitivity to EVE, CIP2A expression decreased at low EVE concentrations; however, in LTED cells poorly sensitive to EVE, CIP2A and PP2A did not change upon exposure to EVE. Therefore, we hypothesized that there is a relation between expression of CIP2A and sensitivity to EVE. Knockdown of CIP2A increased the sensitivity to EVE in three clones poorly sensitive to EVE. Additionally, we found that treatment with FSK, which activates PP2A, increased the sensitivity of the cells to EVE. Our data point to CIP2A and PP2A as novel therapeutic targets for AI-resistant breast cancer.
Collapse
Affiliation(s)
- Takanori Hayashi
- Department of Biochemistry, School of Medicine, Fujita Health University, Aichi, Japan
| | - Masahiro Hikichi
- Department of Breast Surgery, School of Medicine, Fujita Health University, Aichi, Japan
| | - Jun Yukitake
- Department of Clinical Immunology, School of Health Sciences, Fujita Health University, Aichi, Japan
| | - Toru Wakatsuki
- Department of Health Science, School of Medicine, Fujita Health University, Aichi, Japan
| | - Eiji Nishio
- Department of Obstetrics and Gynecology, School of Medicine, Fujita Health University, Aichi, Japan
| | - Toshiaki Utsumi
- Department of Breast Surgery, School of Medicine, Fujita Health University, Aichi, Japan
| | - Nobuhiro Harada
- Department of Biochemistry, School of Medicine, Fujita Health University, Aichi, Japan
| |
Collapse
|
7
|
Zhao H, Li D, Zhang B, Qi Y, Diao Y, Zhen Y, Shu X. PP2A as the Main Node of Therapeutic Strategies and Resistance Reversal in Triple-Negative Breast Cancer. Molecules 2017; 22:molecules22122277. [PMID: 29261144 PMCID: PMC6149800 DOI: 10.3390/molecules22122277] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 12/07/2017] [Accepted: 12/19/2017] [Indexed: 12/31/2022] Open
Abstract
Triple negative breast cancer (TNBC), is defined as a type of tumor lacking the expression of estrogen receptor (ER), progesterone receptor (PR) and human epidermal growth factor receptor 2 (HER2). The ER, PR and HER2 are usually the molecular therapeutic targets for breast cancers, but they are ineffective for TNBC because of their negative expressions, so chemotherapy is currently the main treatment strategy in TNBC. However, drug resistance remains a major impediment to TNBC chemotherapeutic treatment. Recently, the protein phosphatase 2A (PP2A) has been found to regulate the phosphorylation of some substrates involved in the relevant target of TNBC, such as cell cycle control, DNA damage responses, epidermal growth factor receptor, immune modulation and cell death resistance, which may be the effective therapeutic strategies or influence drug sensitivity to TNBCs. Furthermore, PP2A has also been found that could induce ER re-expression in ER-negative breast cancer cells, and which suggests PP2A could promote the sensitivity of tamoxifen to TNBCs as a resistance reversal agent. In this review, we will summarize the potential therapeutic value of PP2A as the main node in developing targeting agents, disrupting resistance or restoring drug sensitivity in TNBC.
Collapse
Affiliation(s)
- Henan Zhao
- Department of Pathophysiology, Dalian Medical University, Dalian 116044, China.
| | - Duojiao Li
- Kamp Pharmaceutical Co. Ltd., Changsha 410008, China.
| | - Baojing Zhang
- College of Pharmacy, Dalian Medical University, Dalian 116044, China.
| | - Yan Qi
- College of Pharmacy, Dalian Medical University, Dalian 116044, China.
| | - Yunpeng Diao
- College of Pharmacy, Dalian Medical University, Dalian 116044, China.
| | - Yuhong Zhen
- College of Pharmacy, Dalian Medical University, Dalian 116044, China.
| | - Xiaohong Shu
- College of Pharmacy, Dalian Medical University, Dalian 116044, China.
| |
Collapse
|
8
|
Baldacchino S, Wastall LM, Saliba C, Hughes TA, Scerri C, Berwick A, Speirs V, Hanby AM, Grech G. CIP2A expression predicts recurrences of tamoxifen-treated breast cancer. Tumour Biol 2017; 39:1010428317722064. [PMID: 29034804 DOI: 10.1177/1010428317722064] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
CIP2A is emerging as an oncoprotein overexpressed commonly across many tumours and generally correlated with higher tumour grade and therapeutic resistance. CIP2A drives an oncogenic potential through inhibiting protein phosphatase 2A, stabilizing MYC, and promoting epithelial-to-mesenchymal transition, although further biological mechanisms for CIP2A are yet to be defined. CIP2A protein expression was studied by immunohistochemistry in oestrogen receptor-positive primary breast cancers (n = 250) obtained from the Leeds Tissue Bank. In total, 51 cases presented with a relapse or metastasis during adjuvant treatment with tamoxifen and were regarded as tamoxifen resistant. CIP2A expression was scored separately for cytoplasmic, nuclear, or membranous staining, and scores were tested for statistically significant relationships with clinicopathological features. Membranous CIP2A was preferentially expressed in cases who experienced a recurrence during tamoxifen treatment thus predicting a worse overall survival (log rank = 8.357, p = 0.004) and disease-free survival (log rank = 21.766, p < 0.001). Cox multivariate analysis indicates that it is an independent prognostic indicator for overall survival (hazard ratio = 4.310, p = 0.013) and disease-free survival (hazard ratio = 5.449, p = 0.002). In this study, we propose the assessment of membranous CIP2A expression as a potential novel prognostic and predictive indicator for tamoxifen resistance and recurrence within oestrogen receptor-positive breast cancer.
Collapse
Affiliation(s)
- Shawn Baldacchino
- 1 Laboratory of Molecular Pathology, Department of Pathology, Faculty of Medicine & Surgery, University of Malta, Msida, Malta
| | - Laura M Wastall
- 2 School of Medicine, University of Leeds, Leeds, UK.,3 Department of Cellular Pathology, St James's University Hospital, Leeds, UK
| | - Christian Saliba
- 4 Centre for Molecular Medicine and Biobanking, University of Malta, Msida, Malta
| | | | - Christian Scerri
- 5 Department of Physiology & Biochemistry, Faculty of Medicine & Surgery, University of Malta, Msida, Malta
| | - Angelene Berwick
- 6 Leeds Institute of Cancer & Pathology, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Valerie Speirs
- 6 Leeds Institute of Cancer & Pathology, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Andrew M Hanby
- 3 Department of Cellular Pathology, St James's University Hospital, Leeds, UK.,6 Leeds Institute of Cancer & Pathology, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Godfrey Grech
- 1 Laboratory of Molecular Pathology, Department of Pathology, Faculty of Medicine & Surgery, University of Malta, Msida, Malta
| |
Collapse
|
9
|
Cristóbal I, Torrejón B, Martínez-Useros J, Madoz-Gurpide J, Rojo F, García-Foncillas J. PP2A regulates signaling through hormonal receptors in breast cancer with important therapeutic implications. Biochim Biophys Acta Rev Cancer 2017; 1868:435-438. [PMID: 28916342 DOI: 10.1016/j.bbcan.2017.08.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 08/10/2017] [Accepted: 08/31/2017] [Indexed: 11/27/2022]
Abstract
The functional inhibition of protein phosphatase 2A (PP2A) has emerged in the last years as a common alteration in breast cancer that determines poor outcome and contributes to disease progression and aggressiveness. Furthermore, expression of estrogen receptor (ER) is a high relevant molecular event with key therapeutic implications in breast cancer, and androgen receptor (AR) signaling is involved in the pathogenesis of breast cancer and represents a novel target with crescent importance in this disease. In this review, we summarize the role of the tumor suppressor PP2A in modulating ER and AR signaling in breast cancer, the molecular mechanisms involved, and its biological and therapeutic impact.
Collapse
Affiliation(s)
- Ion Cristóbal
- Translational Oncology Division, Oncohealth Institute, IIS-Fundacion Jimenez Diaz, UAM, University Hospital "Fundacion Jimenez Diaz", Madrid, Spain.
| | - Blanca Torrejón
- Translational Oncology Division, Oncohealth Institute, IIS-Fundacion Jimenez Diaz, UAM, University Hospital "Fundacion Jimenez Diaz", Madrid, Spain
| | - Javier Martínez-Useros
- Translational Oncology Division, Oncohealth Institute, IIS-Fundacion Jimenez Diaz, UAM, University Hospital "Fundacion Jimenez Diaz", Madrid, Spain
| | | | - Federico Rojo
- Pathology Department, IIS "Fundación Jiménez Diaz", Madrid, Spain.
| | - Jesús García-Foncillas
- Translational Oncology Division, Oncohealth Institute, IIS-Fundacion Jimenez Diaz, UAM, University Hospital "Fundacion Jimenez Diaz", Madrid, Spain.
| |
Collapse
|
10
|
Ma LM, Liang ZR, Zhou KR, Zhou H, Qu LH. 27-Hydroxycholesterol increases Myc protein stability via suppressing PP2A, SCP1 and FBW7 transcription in MCF-7 breast cancer cells. Biochem Biophys Res Commun 2016; 480:328-333. [PMID: 27751849 DOI: 10.1016/j.bbrc.2016.10.038] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 10/14/2016] [Indexed: 12/11/2022]
Abstract
27-hydroxycholesterol (27-HC), the most abundant metabolite of cholesterol, is a risk factor for breast cancer. It can increase the proliferation of breast cancer cells and promote the metastasis of breast tumours in mouse models. Myc is a critical oncoprotein overexpressed in breast cancer. However, whether 27-HC affects Myc expression has not been reported. In the current study, we aimed to investigate the effects of 27-HC on Myc and the underlying mechanisms in MCF-7 breast cancer cells. Our data demonstrated that 27-HC activated Myc via increasing its protein stability. Three key negative modulators of Myc protein stability, PP2A, SCP1 and FBW7, were suppressed by 27-HC at the transcriptional level. We performed a data-mining analysis of the chromatin immunoprecipitation with next-generation DNA sequencing (ChIP-Seq) data in the ChIPBase, and discovered that a number of putative transcription factors (TFs), including Myc itself, were involved in the transcriptional regulation of PP2A, SCP1 and FBW7. Our results provide a novel mechanistic insight into the activation of Myc by 27-HC via transcriptional repression of PP2A, SCP1 and FBW7 to increase Myc protein stability in breast cancer cells.
Collapse
Affiliation(s)
- Li-Ming Ma
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, PR China.
| | - Zi-Rui Liang
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, PR China.
| | - Ke-Ren Zhou
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, PR China.
| | - Hui Zhou
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, PR China.
| | - Liang-Hu Qu
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, PR China.
| |
Collapse
|
11
|
Zhao Q, Zhao M, Parris AB, Xing Y, Yang X. Genistein targets the cancerous inhibitor of PP2A to induce growth inhibition and apoptosis in breast cancer cells. Int J Oncol 2016; 49:1203-10. [PMID: 27574003 PMCID: PMC4948957 DOI: 10.3892/ijo.2016.3588] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 05/30/2016] [Indexed: 12/17/2022] Open
Abstract
Genistein is a soy isoflavone with phytoestrogen and tyrosine kinase inhibitory properties. High intake of soy/genistein has been associated with reduced breast cancer risk. Despite the advances in genistein-mediated antitumor studies, the underlying mechanisms remain unclear. In the present study, we investigated genistein-induced regulation of the cancerous inhibitor of protein phosphatase 2A (CIP2A), a novel oncogene frequently overexpressed in breast cancer, and its functional impact on genistein-induced growth inhibition and apoptosis. We demonstrated that genistein induced downregulation of CIP2A in MCF-7-C3 and T47D breast cancer cells, which was correlated with its growth inhibition and apoptotic activities. Overexpression of CIP2A attenuated, whereas CIP2A knockdown sensitized, genistein-induced growth inhibition and apoptosis. We further showed that genistein-induced downregulation of CIP2A involved both transcriptional suppression and proteasomal degradation. In particular, genistein at higher concentrations induced concurrent downregulation of E2F1 and CIP2A. Overexpression of E2F1 attenuated genistein-induced downregulation of CIP2A mRNA, indicating the role of E2F1 in genistein-induced transcriptional suppression of CIP2A. Taken together, our results identified CIP2A as a functional target of genistein and demonstrated that modulation of E2F1-mediated transcriptional regulation of CIP2A contributes to its downregulation. These data advance our understanding of genistein-induced growth inhibition and apoptosis, and support further investigation on CIP2A as a therapeutic target of relevant anticancer agents.
Collapse
Affiliation(s)
- Qingxia Zhao
- Basic Medical College of Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Ming Zhao
- Julius L. Chambers Biomedical/Biotechnology Research Institute and Department of Biology, North Carolina Central University, Kannapolis, NC 28081, USA
| | - Amanda B Parris
- Julius L. Chambers Biomedical/Biotechnology Research Institute and Department of Biology, North Carolina Central University, Kannapolis, NC 28081, USA
| | - Ying Xing
- Basic Medical College of Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Xiaohe Yang
- Julius L. Chambers Biomedical/Biotechnology Research Institute and Department of Biology, North Carolina Central University, Kannapolis, NC 28081, USA
| |
Collapse
|
12
|
Prizant H, Taya M, Lerman I, Light A, Sen A, Mitra S, Foster TH, Hammes SR. Estrogen maintains myometrial tumors in a lymphangioleiomyomatosis model. Endocr Relat Cancer 2016; 23:265-80. [PMID: 26880751 PMCID: PMC4992946 DOI: 10.1530/erc-15-0505] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 02/15/2016] [Indexed: 12/17/2022]
Abstract
Lymphangioleiomyomatosis (LAM) is a rare disease in women. Patients with LAM develop metastatic smooth-muscle cell adenomas within the lungs, resulting in reduced pulmonary function. LAM cells contain mutations in tuberous sclerosis genes (TSC1 or TSC2), leading to up-regulation of mTORC1 activity and elevated proliferation. The origin of LAM cells remains unknown; however, inactivation of Tsc2 gene in the mouse uterus resulted in myometrial tumors exhibiting LAM features, and approximately 50% of animals developed metastatic myometrial lung tumors. This suggests that LAM tumors might originate from the uterine myometrium, possibly explaining the overwhelming prevalence of LAM in female. Here, we demonstrate that mouse Tsc2-null myometrial tumors exhibit nearly all the features of LAM, including mTORC1/S6K activation, as well as expression of melanocytic markers and matrix metalloproteinases (MMPs). Estrogen ablation reduces S6K signaling and results in Tsc2-null myometrial tumor regression. Thus, even without TSC2, estradiol is required to maintain tumors and mTORC1/S6K signaling. Additionally, we find that MMP-2 and -9, as well as neutrophil elastase (NE), are overexpressed in Tsc2-null myometrial tumors in an estrogen-dependent fashion. In vivo fluorescent imaging using MMP- or NE-sensitive optical biomarkers confirms that protease activity is specific to myometrial tumors. Similar to LAM cells, uterine Tsc2-null myometrial cells also overexpress melanocytic markers in an estrogen-dependent fashion. Finally, we identify glycoprotein NMB (GPNMB) as a melanocytic marker up-regulated in Tsc2-null mouse uteri and human LAM samples. Our data highlight the potential importance of estradiol in LAM cells, suggesting that anti-estrogen therapy may be a treatment modality. Furthermore, proteases and GPNMB might be useful LAM biomarkers.
Collapse
Affiliation(s)
- Hen Prizant
- Division of Endocrinology and MetabolismDepartment of Medicine, University of Rochester Medical Center, Rochester, New York, USA
| | - Manisha Taya
- Division of Endocrinology and MetabolismDepartment of Medicine, University of Rochester Medical Center, Rochester, New York, USA
| | - Irina Lerman
- Division of Endocrinology and MetabolismDepartment of Medicine, University of Rochester Medical Center, Rochester, New York, USA
| | - Allison Light
- Division of Endocrinology and MetabolismDepartment of Medicine, University of Rochester Medical Center, Rochester, New York, USA
| | - Aritro Sen
- Division of Endocrinology and MetabolismDepartment of Medicine, University of Rochester Medical Center, Rochester, New York, USA
| | - Soumya Mitra
- Department of Imaging SciencesUniversity of Rochester Medical Center, Rochester, New York, USA
| | - Thomas H Foster
- Department of Imaging SciencesUniversity of Rochester Medical Center, Rochester, New York, USA
| | - Stephen R Hammes
- Division of Endocrinology and MetabolismDepartment of Medicine, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
13
|
Michaut M, Chin SF, Majewski I, Severson TM, Bismeijer T, de Koning L, Peeters JK, Schouten PC, Rueda OM, Bosma AJ, Tarrant F, Fan Y, He B, Xue Z, Mittempergher L, Kluin RJ, Heijmans J, Snel M, Pereira B, Schlicker A, Provenzano E, Ali HR, Gaber A, O’Hurley G, Lehn S, Muris JJ, Wesseling J, Kay E, Sammut SJ, Bardwell HA, Barbet AS, Bard F, Lecerf C, O’Connor DP, Vis DJ, Benes CH, McDermott U, Garnett MJ, Simon IM, Jirström K, Dubois T, Linn SC, Gallagher WM, Wessels LF, Caldas C, Bernards R. Integration of genomic, transcriptomic and proteomic data identifies two biologically distinct subtypes of invasive lobular breast cancer. Sci Rep 2016; 6:18517. [PMID: 26729235 PMCID: PMC4700448 DOI: 10.1038/srep18517] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 11/19/2015] [Indexed: 12/23/2022] Open
Abstract
Invasive lobular carcinoma (ILC) is the second most frequently occurring histological breast cancer subtype after invasive ductal carcinoma (IDC), accounting for around 10% of all breast cancers. The molecular processes that drive the development of ILC are still largely unknown. We have performed a comprehensive genomic, transcriptomic and proteomic analysis of a large ILC patient cohort and present here an integrated molecular portrait of ILC. Mutations in CDH1 and in the PI3K pathway are the most frequent molecular alterations in ILC. We identified two main subtypes of ILCs: (i) an immune related subtype with mRNA up-regulation of PD-L1, PD-1 and CTLA-4 and greater sensitivity to DNA-damaging agents in representative cell line models; (ii) a hormone related subtype, associated with Epithelial to Mesenchymal Transition (EMT), and gain of chromosomes 1q and 8q and loss of chromosome 11q. Using the somatic mutation rate and eIF4B protein level, we identified three groups with different clinical outcomes, including a group with extremely good prognosis. We provide a comprehensive overview of the molecular alterations driving ILC and have explored links with therapy response. This molecular characterization may help to tailor treatment of ILC through the application of specific targeted, chemo- and/or immune-therapies.
Collapse
Affiliation(s)
- Magali Michaut
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Suet-Feung Chin
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| | - Ian Majewski
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Tesa M. Severson
- Division of Molecular Pathology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Tycho Bismeijer
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Leanne de Koning
- Translational Research Department, Institut Curie, 26 rue d’Ulm, 75248 Paris cedex 05, France
| | | | - Philip C. Schouten
- Division of Molecular Pathology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Oscar M. Rueda
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| | - Astrid J. Bosma
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Finbarr Tarrant
- Cancer Biology and Therapeutics Laboratory, UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
- OncoMark Limited, NovaUCD, Belfield Innovation Park, Dublin 4, Ireland
| | - Yue Fan
- Cancer Biology and Therapeutics Laboratory, UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Beilei He
- Translational Research Department, Institut Curie, 26 rue d’Ulm, 75248 Paris cedex 05, France
| | - Zheng Xue
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Lorenza Mittempergher
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Roelof J.C. Kluin
- Genomic Core Facility, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Jeroen Heijmans
- Agendia NV, Science Park 406, 1098 XH Amsterdam, The Netherlands
| | - Mireille Snel
- Agendia NV, Science Park 406, 1098 XH Amsterdam, The Netherlands
| | - Bernard Pereira
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| | - Andreas Schlicker
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Elena Provenzano
- Cambridge Experimental Cancer Medicine Centre (ECMR) and NIHR Cambridge Biomedical Research Centre, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK
- Cambridge Breast Unit and Cambridge University Hospitals, NHS Foundation Trust, Hills Road, Cambridge CB2 0QQ, UK
| | - Hamid Raza Ali
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
| | - Alexander Gaber
- Department of Clinical Sciences Lund, Division of Oncology and Pathology, Lund University, SE-221 85 Lund, Sweden
| | - Gillian O’Hurley
- OncoMark Limited, NovaUCD, Belfield Innovation Park, Dublin 4, Ireland
| | - Sophie Lehn
- Department of Clinical Sciences Lund, Division of Oncology and Pathology, Lund University, SE-221 85 Lund, Sweden
| | - Jettie J.F. Muris
- Division of Molecular Pathology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Jelle Wesseling
- Division of Molecular Pathology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Elaine Kay
- Department of Pathology, RCSI ERC, Beaumont Hospital, Dublin 9, Ireland
| | - Stephen John Sammut
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| | - Helen A. Bardwell
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| | - Aurélie S. Barbet
- Translational Research Department, Institut Curie, 26 rue d’Ulm, 75248 Paris cedex 05, France
| | - Floriane Bard
- Translational Research Department, Institut Curie, 26 rue d’Ulm, 75248 Paris cedex 05, France
| | - Caroline Lecerf
- Translational Research Department, Institut Curie, 26 rue d’Ulm, 75248 Paris cedex 05, France
| | - Darran P. O’Connor
- Cancer Biology and Therapeutics Laboratory, UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Daniël J. Vis
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Cyril H. Benes
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | - Ultan McDermott
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton CB10 1SA, UK
| | - Mathew J. Garnett
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton CB10 1SA, UK
| | - Iris M. Simon
- Agendia NV, Science Park 406, 1098 XH Amsterdam, The Netherlands
| | - Karin Jirström
- Department of Clinical Sciences Lund, Division of Oncology and Pathology, Lund University, SE-221 85 Lund, Sweden
| | - Thierry Dubois
- Translational Research Department, Institut Curie, 26 rue d’Ulm, 75248 Paris cedex 05, France
| | - Sabine C. Linn
- Division of Molecular Pathology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
- Division of Medical Oncology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
- Department of Pathology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - William M. Gallagher
- Cancer Biology and Therapeutics Laboratory, UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
- OncoMark Limited, NovaUCD, Belfield Innovation Park, Dublin 4, Ireland
| | - Lodewyk F.A. Wessels
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
- Department of EEMCS, Delft University of Technology, Delft, The Netherlands
| | - Carlos Caldas
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
- Cambridge Experimental Cancer Medicine Centre (ECMR) and NIHR Cambridge Biomedical Research Centre, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK
- Cambridge Breast Unit and Cambridge University Hospitals, NHS Foundation Trust, Hills Road, Cambridge CB2 0QQ, UK
- Department of Oncology, University of Cambridge, Addenbrooke’s Hospital, Hills Road, Cambridge, CB2 0QQ, UK
| | - Rene Bernards
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
- Agendia NV, Science Park 406, 1098 XH Amsterdam, The Netherlands
- Cancer Genomics Netherlands, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| |
Collapse
|
14
|
Rincón R, Cristóbal I, Zazo S, Arpí O, Menéndez S, Manso R, Lluch A, Eroles P, Rovira A, Albanell J, García-Foncillas J, Madoz-Gúrpide J, Rojo F. PP2A inhibition determines poor outcome and doxorubicin resistance in early breast cancer and its activation shows promising therapeutic effects. Oncotarget 2015; 6:4299-314. [PMID: 25726524 PMCID: PMC4414191 DOI: 10.18632/oncotarget.3012] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 12/31/2014] [Indexed: 11/26/2022] Open
Abstract
The protein phosphatase 2A (PP2A) is a key tumor suppressor which has emerged as a novel molecular target in some human cancers. Here, we show that PP2A inhibition is a common event in breast cancer and identified PP2A phosphorylation and deregulation SET and CIP2A as molecular contributing mechanisms to inactivate PP2A. Interestingly, restoration of PP2A activity after FTY720 treatment reduced cell growth, induced apoptosis and decreased AKT and ERK activation. Moreover, FTY720 led to PP2A activation then enhancing doxorubicin-induced antitumor effects both in vitro and in vivo. PP2A inhibition (CPscore: PP2A phosphorylation and/or CIP2A overexpression) was detected in 27% of cases (62/230), and associated with grade (p = 0.017), relapse (p < 0.001), negative estrogen (p < 0.001) and progesterone receptor expression (p < 0.001), HER2-positive tumors (p = 0.049), Ki-67 expression (p < 0.001), and higher AKT (p < 0.001) and ERK (p < 0.001) phosphorylation. Moreover, PP2A inhibition determined shorter overall (p = 0.006) and event-free survival (p = 0.003), and multivariate analysis confirmed its independent prognostic impact. Altogether, our results indicate that PP2A is frequently inactivated in breast cancer and determines worse outcome, and its restoration using PP2A activators represents an alternative therapeutic strategy in this disease.
Collapse
Affiliation(s)
- Raúl Rincón
- Translational Oncology Division, Oncohealth Institute, Health Research Institute FJD-UAM, University Hospital "Fundación Jiménez Diaz", Madrid, Spain
| | - Ion Cristóbal
- Translational Oncology Division, Oncohealth Institute, Health Research Institute FJD-UAM, University Hospital "Fundación Jiménez Diaz", Madrid, Spain
| | - Sandra Zazo
- Pathology Department, IIS "Fundación Jiménez Diaz", Madrid, Spain
| | - Oriol Arpí
- Medical Oncology Department, Hospital del Mar, Barcelona, Spain
| | - Silvia Menéndez
- Medical Oncology Department, Hospital del Mar, Barcelona, Spain
| | - Rebeca Manso
- Pathology Department, IIS "Fundación Jiménez Diaz", Madrid, Spain
| | - Ana Lluch
- Institute of Health Research INCLIVA, Valencia, Spain
| | - Pilar Eroles
- Institute of Health Research INCLIVA, Valencia, Spain
| | - Ana Rovira
- Medical Oncology Department, Hospital del Mar, Barcelona, Spain
| | - Joan Albanell
- Medical Oncology Department, Hospital del Mar, Barcelona, Spain
| | - Jesús García-Foncillas
- Translational Oncology Division, Oncohealth Institute, Health Research Institute FJD-UAM, University Hospital "Fundación Jiménez Diaz", Madrid, Spain
| | | | - Federico Rojo
- Pathology Department, IIS "Fundación Jiménez Diaz", Madrid, Spain.,Medical Oncology Department, Hospital del Mar, Barcelona, Spain
| |
Collapse
|