1
|
Segatto I, Mattevi MC, Rampioni Vinciguerra GL, Crestan N, Musco L, Favero A, Dall'Acqua A, Di Giustino G, Mungo G, D'Andrea S, Gava C, Ruggiero F, Dugo M, Gerratana L, Puglisi F, Massarut S, Bomben R, Callari M, Perin T, Baldassarre G, Belletti B. A comprehensive luminal breast cancer patient-derived xenografts (PDX) library to capture tumor heterogeneity and explore the mechanisms of resistance to CDK4/6 inhibitors. J Pathol 2024; 264:434-447. [PMID: 39449657 DOI: 10.1002/path.6358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 08/07/2024] [Accepted: 09/04/2024] [Indexed: 10/26/2024]
Abstract
Breast cancer (BC) is marked by significant genetic, morphological and clinical heterogeneity. To capture this heterogeneity and unravel the molecular mechanisms driving tumor progression and drug resistance, we established a comprehensive patient-derived xenograft (PDX) biobank, focusing particularly on luminal (estrogen receptor, ER+) and young premenopausal patients, for whom PDX models are currently scarce. Across all BC subtypes, our efforts resulted in an overall success rate of 17% (26 established PDX lines out of 151 total attempts), specifically 15% in luminal, 12% in human epidermal growth factor receptor 2 positive (HER2+) and 35% in triple negative BC. These PDX mirrored morphologic and genetic features of BC from which they originated, serving as a reliable tool to investigate drug resistance and test therapeutic strategies. We focused on understanding resistance to CDK4/6 inhibitors (CDK4/6i), which are crucial in the treatment of patients with advanced luminal BC. Treating a sensitive luminal BC PDX with the CDK4/6i palbociclib revealed that, despite initial tumor shrinkage, some tumors might eventually regrow under drug treatment. RNA sequencing, followed by gene set enrichment analyses, unveiled that these PDXs have become refractory to CDK4/6i, both at biological and molecular levels, displaying significant enrichment in proliferation pathways, such as MTORC1, E2F and MYC. Using organoids derived from these PDX (PDxO), we observed that acquisition of CDK4/6i resistance conferred cross-resistance to endocrine therapy and that targeting MTORC1 was a successful strategy to overcome CDK4/6i resistance. Considered together, these results indicate that our PDX models may serve as robust tools to elucidate the molecular basis of BC disease progression and, by providing the possibility to simultaneously test different therapies on the same tumor, to surmount treatment resistance. While this approach is of course not feasible in the clinic, its exploitation in PDX may expedite the identification and development of more successful therapies for patients with advanced luminal BC. © 2024 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Ilenia Segatto
- Unit of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy
| | - Maria Chiara Mattevi
- Unit of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy
| | - Gian Luca Rampioni Vinciguerra
- Unit of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy
- Faculty of Medicine and Psychology, Department of Clinical and Molecular Medicine, University of Rome 'Sapienza', Santo Andrea Hospital, Rome, Italy
| | - Nicole Crestan
- Unit of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy
| | - Lorena Musco
- Unit of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy
| | - Andrea Favero
- Unit of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy
| | - Alessandra Dall'Acqua
- Unit of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy
| | - Gabriele Di Giustino
- Unit of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy
| | - Giorgia Mungo
- Unit of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy
| | - Sara D'Andrea
- Unit of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy
| | - Chiara Gava
- Unit of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy
| | - Federica Ruggiero
- Unit of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy
| | | | - Lorenzo Gerratana
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy
| | - Fabio Puglisi
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy
| | - Samuele Massarut
- Unit of Breast Surgery, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy
| | - Riccardo Bomben
- Unit of Clinical and Experimental Onco-Hematology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy
| | | | - Tiziana Perin
- Unit of Pathology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy
| | - Gustavo Baldassarre
- Unit of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy
| | - Barbara Belletti
- Unit of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy
| |
Collapse
|
2
|
Yulak F, Filiz AK, Joha Z, Ergul M. Mechanism of anticancer effect of ETP-45658, a PI3K/AKT/mTOR pathway inhibitor on HT-29 Cells. Med Oncol 2023; 40:341. [PMID: 37891359 DOI: 10.1007/s12032-023-02221-4] [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: 09/06/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023]
Abstract
The PI3K pathway plays a crucial role in tumor cell proliferation across various cancers, including colon cancer, making it a promising treatment target. This study aims to investigate the antiproliferative activity of ETP-45658, a PI3K/AKT/mTOR pathway inhibitor, on colon cancer and elucidate the underlying mechanisms. HT-29 colon cancer cells were treated with varying doses of ETP 45658 and its cytotoxic effect assessed using the XTT cell viability assay.ELISA was also used to measure TAS, TOS, Bax, BCL-2, cleaved caspase 3, cleaved PARP, and 8-oxo-dG levels. Flow cytometry was performed to investigate apoptosis, cell cycle, caspase 3/7 activity, and mitochondrial membrane potential. Additionally, following the administration of DAPI (4,6-diamidino-2-phenylindole) dye, the cells were visualized using an immunofluorescence microscope. It was observed that ETP-45658 exerted a dose-dependent and statistically significant antiproliferative effect on HT-29 colon cancer cells. Further investigations using the IC50 dose showed that ETP-45658 decreased TAS levels and increased TOS levels and revealed that it upregulated apoptotic proteins while downregulating anti-apoptotic proteins. Our findings also showed that it increased Annexin V binding, arrested the cell cycle at G0/G1 phase, induced caspase 3/7 activity, impaired mitochondrial membrane potential, and ultimately triggered apoptosis in HT-29 cells. ETP-45658 shows promise against colon cancer by inducing cell death, and oxidative stress, and arresting the cell cycle. Targeting the PI3K/AKT/mTOR pathway with ETP-45658 offers exciting potential for colon cancer treatment.
Collapse
Affiliation(s)
- Fatih Yulak
- Departments of Physiology, School of Medicine, Cumhuriyet University, Sivas, Turkey
| | - Ahmet Kemal Filiz
- Departments of Physiology, School of Medicine, Cumhuriyet University, Sivas, Turkey
| | - Zıad Joha
- Department of Pharmacology, Faculty of Pharmacy, Sivas Cumhuriyet University, Sivas, Turkey
| | - Mustafa Ergul
- Department of Biochemistry, Faculty of Pharmacy, Sivas Cumhuriyet University, Sivas, Turkey.
| |
Collapse
|
3
|
Jeibouei S, Shams F, Mohebichamkhorami F, Sanooghi D, Faal B, Akbari ME, Zali H. Biological and clinical review of IORT-induced wound fluid in breast cancer patients. Front Oncol 2022; 12:980513. [DOI: 10.3389/fonc.2022.980513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 10/19/2022] [Indexed: 11/22/2022] Open
Abstract
Intraoperative radiotherapy (IORT) has become a growing therapy for early-stage breast cancer (BC). Some studies claim that wound fluid (seroma), a common consequence of surgical excision in the tumor cavity, can reflect the effects of IORT on cancer inhibition. However, further research by our team and other researchers, such as analysis of seroma composition, affected cell lines, and primary tissues in two-dimensional (2D) and three-dimensional (3D) culture systems, clarified that seroma could not address the questions about IORT effectiveness in the surgical site. In this review, we mention the factors involved in tumor recurrence, direct or indirect effects of IORT on BC, and all the studies associated with BC seroma to attain more information about the impact of IORT-induced seroma to make a better decision to remove or remain after surgery and IORT. Finally, we suggest that seroma studies cannot decipher the mechanisms underlying the effectiveness of IORT in BC patients. The question of whether IORT-seroma has a beneficial effect can only be answered in a trial with a clinical endpoint, which is not even ongoing.
Collapse
|
4
|
Harvey J, Mellody KT, Cullum N, Watson REB, Dumville J. Wound fluid sampling methods for proteomic studies: A scoping review. Wound Repair Regen 2022; 30:317-333. [PMID: 35381119 PMCID: PMC9322564 DOI: 10.1111/wrr.13009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 02/01/2022] [Accepted: 03/08/2022] [Indexed: 01/02/2023]
Abstract
Understanding why some wounds are hard to heal is important for improving care and developing more effective treatments. The method of sample collection used is an integral step in the research process and thus may affect the results obtained. The primary objective of this study was to summarise and map the methods currently used to sample wound fluid for protein profiling and analysis. Eligible studies were those that used a sampling method to collect wound fluid from any human wound for analysis of proteins. A search for eligible studies was performed using MEDLINE, Embase and CINAHL Plus in May 2020. All references were screened for eligibility by one reviewer, followed by discussion and consensus with a second reviewer. Quantitative data were mapped and visualised using appropriate software and summarised via a narrative summary. After screening, 280 studies were included in this review. The most commonly used group of wound fluid collection methods were vacuum, drainage or use of other external devices, with surgical wounds being the most common sample source. Other frequently used collection methods were extraction from absorbent materials, collection beneath an occlusive dressing and direct collection of wound fluid. This scoping review highlights the variety of methods used for wound fluid collection. Many studies had small sample sizes and short sample collection periods; these weaknesses have hampered the discovery and validation of novel biomarkers. Future research should aim to assess the reproducibility and feasibility of sampling and analytical methods for use in larger longitudinal studies.
Collapse
Affiliation(s)
- Joe Harvey
- Centre for Dermatology Research, School of Biological SciencesThe University of Manchester & Salford Royal NHS Foundation Trust, Manchester Academic Health Science CentreUK
- NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science CentreManchesterUK
| | - Kieran T. Mellody
- Centre for Dermatology Research, School of Biological SciencesThe University of Manchester & Salford Royal NHS Foundation Trust, Manchester Academic Health Science CentreUK
| | - Nicky Cullum
- NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science CentreManchesterUK
- Division of Nursing, Midwifery & Social WorkSchool of Health Sciences, The University of ManchesterManchesterUK
| | - Rachel E. B. Watson
- Centre for Dermatology Research, School of Biological SciencesThe University of Manchester & Salford Royal NHS Foundation Trust, Manchester Academic Health Science CentreUK
- NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science CentreManchesterUK
- Manchester Institute for Collaborative Research on AgeingThe University of ManchesterManchesterUK
| | - Jo Dumville
- NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science CentreManchesterUK
- Division of Nursing, Midwifery & Social WorkSchool of Health Sciences, The University of ManchesterManchesterUK
| |
Collapse
|
5
|
Xiao Y, Huck BR, Lan R, DeSelm L, Chen X, Qiu H, Neagu C, Johnson T, Mochalkin I, Gardberg A, Jiang X, Tian H, Dutt V, Santos D, Head J, Jackson J, Syed S, Lin J, Wilker E, Ma J, Clark A, Machl A, Bankston D, Jones CCV, Goutopoulos A, Sherer B. Discovery of 4-aminopyrimidine analogs as highly potent dual P70S6K/Akt inhibitors. Bioorg Med Chem Lett 2021; 50:128352. [PMID: 34481987 DOI: 10.1016/j.bmcl.2021.128352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/16/2021] [Accepted: 08/29/2021] [Indexed: 11/15/2022]
Abstract
Activation of the PI3K/Akt/mTOR kinase pathway is associated with human cancers. A dual p70S6K/Akt inhibitor is sufficient to inhibit strong tumor growth and to block negative impact of the compensatory Akt feedback loop activation. A scaffold docking strategy based on an existing quinazoline carboxamide series identified 4-aminopyrimidine analog 6, which showed a single-digit nanomolar and a micromolar potencies in p70S6K and Akt enzymatic assays. SAR optimization improved Akt enzymatic and p70S6K cellular potencies, reduced hERG liability, and ultimately discovered the promising candidate 37, which exhibited with a single digit nanomolar value in both p70S6K and Akt biochemical assays, and hERG activities (IC50 = 17.4 μM). This agent demonstrated dose-dependent efficacy in inhibiting mice breast cancer tumor growth and covered more than 90% pS6 inhibition up to 24 h at a dose of 200 mg/kg po.
Collapse
Affiliation(s)
- Yufang Xiao
- EMD Serono Research and Development Institute, Inc., 45A Middlesex Turnpike, Billerica, MA 01821, USA.
| | - Bayard R Huck
- EMD Serono Research and Development Institute, Inc., 45A Middlesex Turnpike, Billerica, MA 01821, USA.
| | - Ruoxi Lan
- EMD Serono Research and Development Institute, Inc., 45A Middlesex Turnpike, Billerica, MA 01821, USA
| | - Lizbeth DeSelm
- EMD Serono Research and Development Institute, Inc., 45A Middlesex Turnpike, Billerica, MA 01821, USA
| | - Xiaoling Chen
- EMD Serono Research and Development Institute, Inc., 45A Middlesex Turnpike, Billerica, MA 01821, USA
| | - Hui Qiu
- EMD Serono Research and Development Institute, Inc., 45A Middlesex Turnpike, Billerica, MA 01821, USA
| | - Constantin Neagu
- EMD Serono Research and Development Institute, Inc., 45A Middlesex Turnpike, Billerica, MA 01821, USA
| | - Theresa Johnson
- EMD Serono Research and Development Institute, Inc., 45A Middlesex Turnpike, Billerica, MA 01821, USA
| | - Igor Mochalkin
- EMD Serono Research and Development Institute, Inc., 45A Middlesex Turnpike, Billerica, MA 01821, USA
| | - Anna Gardberg
- EMD Serono Research and Development Institute, Inc., 45A Middlesex Turnpike, Billerica, MA 01821, USA
| | - Xuliang Jiang
- EMD Serono Research and Development Institute, Inc., 45A Middlesex Turnpike, Billerica, MA 01821, USA
| | - Hui Tian
- EMD Serono Research and Development Institute, Inc., 45A Middlesex Turnpike, Billerica, MA 01821, USA
| | - Vikram Dutt
- EMD Serono Research and Development Institute, Inc., 45A Middlesex Turnpike, Billerica, MA 01821, USA
| | - Dusica Santos
- EMD Serono Research and Development Institute, Inc., 45A Middlesex Turnpike, Billerica, MA 01821, USA
| | - Jared Head
- EMD Serono Research and Development Institute, Inc., 45A Middlesex Turnpike, Billerica, MA 01821, USA
| | - Jennifer Jackson
- EMD Serono Research and Development Institute, Inc., 45A Middlesex Turnpike, Billerica, MA 01821, USA
| | - Sakeena Syed
- EMD Serono Research and Development Institute, Inc., 45A Middlesex Turnpike, Billerica, MA 01821, USA
| | - Jing Lin
- EMD Serono Research and Development Institute, Inc., 45A Middlesex Turnpike, Billerica, MA 01821, USA
| | - Erik Wilker
- EMD Serono Research and Development Institute, Inc., 45A Middlesex Turnpike, Billerica, MA 01821, USA
| | - Jianguo Ma
- EMD Serono Research and Development Institute, Inc., 45A Middlesex Turnpike, Billerica, MA 01821, USA
| | - Anderson Clark
- EMD Serono Research and Development Institute, Inc., 45A Middlesex Turnpike, Billerica, MA 01821, USA
| | - Andreas Machl
- EMD Serono Research and Development Institute, Inc., 45A Middlesex Turnpike, Billerica, MA 01821, USA
| | - Donald Bankston
- EMD Serono Research and Development Institute, Inc., 45A Middlesex Turnpike, Billerica, MA 01821, USA
| | - Christopher C V Jones
- EMD Serono Research and Development Institute, Inc., 45A Middlesex Turnpike, Billerica, MA 01821, USA
| | - Andreas Goutopoulos
- EMD Serono Research and Development Institute, Inc., 45A Middlesex Turnpike, Billerica, MA 01821, USA
| | - Brian Sherer
- EMD Serono Research and Development Institute, Inc., 45A Middlesex Turnpike, Billerica, MA 01821, USA
| |
Collapse
|
6
|
Spaan I, Timmerman LM, Kimman T, Slomp A, Cuenca M, van Nieuwenhuijzen N, Moesbergen LM, Minnema MC, Raymakers RA, Peperzak V. Direct P70S6K1 inhibition to replace dexamethasone in synergistic combination with MCL-1 inhibition in multiple myeloma. Blood Adv 2021; 5:2593-2607. [PMID: 34152396 PMCID: PMC8270664 DOI: 10.1182/bloodadvances.2020003624] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 03/22/2021] [Indexed: 12/12/2022] Open
Abstract
Novel combination therapies have markedly improved the lifespan of patients with multiple myeloma (MM), but drug resistance and disease relapse remain major clinical problems. Dexamethasone and other glucocorticoids are a cornerstone of conventional and new combination therapies for MM, although their use is accompanied by serious side effects. We aimed to uncover drug combinations that act in synergy and, as such, allow reduced dosing while remaining effective. Dexamethasone and the myeloid cell leukemia 1 (MCL-1) inhibitor S63845 (MCL-1i) proved the most potent combination in our lethality screen and induced apoptosis of human myeloma cell lines (HMCLs) that was 50% higher compared with an additive drug effect. Kinome analysis of dexamethasone-treated HMCLs revealed a reduction in serine/threonine peptide phosphorylation, which was predicted to result from reduced Akt activity. Biochemical techniques showed no dexamethasone-induced effects on FOXO protein or GSK3 but did show a 50% reduction in P70S6K phosphorylation, downstream of the Akt-mTORC1 axis. Replacing dexamethasone by the P70S6K1 isoform-specific inhibitor PF-4708671 (S6K1i) revealed similar and statistically significant synergistic apoptosis of HMCLs in combination with MCL-1i. Interestingly, apoptosis induced by the P70S6K1i and MCL-1i combination was more-than-additive in all 9 primary MM samples tested; this effect was observed for 6 of 9 samples with the dexamethasone and MCL-1i combination. Toxicity on stem and progenitor cell subsets remained minimal. Combined, our results show a strong rationale for combination treatments using the P70S6K inhibitor in MM. Direct and specific inhibition of P70S6K may also provide a solution for patients ineligible or insensitive to dexamethasone or other glucocorticoids.
Collapse
Affiliation(s)
| | | | | | | | | | - Niels van Nieuwenhuijzen
- Center for Translational Immunology and
- Department of Hematology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | | | - Monique C Minnema
- Department of Hematology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Reinier A Raymakers
- Department of Hematology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | | |
Collapse
|
7
|
Vaidya JS, Bulsara M, Baum M, Wenz F, Massarut S, Pigorsch S, Alvarado M, Douek M, Saunders C, Flyger HL, Eiermann W, Brew-Graves C, Williams NR, Potyka I, Roberts N, Bernstein M, Brown D, Sperk E, Laws S, Sütterlin M, Corica T, Lundgren S, Holmes D, Vinante L, Bozza F, Pazos M, Le Blanc-Onfroy M, Gruber G, Polkowski W, Dedes KJ, Niewald M, Blohmer J, McCready D, Hoefer R, Kelemen P, Petralia G, Falzon M, Joseph DJ, Tobias JS. Long term survival and local control outcomes from single dose targeted intraoperative radiotherapy during lumpectomy (TARGIT-IORT) for early breast cancer: TARGIT-A randomised clinical trial. BMJ 2020; 370:m2836. [PMID: 32816842 PMCID: PMC7500441 DOI: 10.1136/bmj.m2836] [Citation(s) in RCA: 161] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
OBJECTIVE To determine whether risk adapted intraoperative radiotherapy, delivered as a single dose during lumpectomy, can effectively replace postoperative whole breast external beam radiotherapy for early breast cancer. DESIGN Prospective, open label, randomised controlled clinical trial. SETTING 32 centres in 10 countries in the United Kingdom, Europe, Australia, the United States, and Canada. PARTICIPANTS 2298 women aged 45 years and older with invasive ductal carcinoma up to 3.5 cm in size, cN0-N1, eligible for breast conservation and randomised before lumpectomy (1:1 ratio, blocks stratified by centre) to either risk adapted targeted intraoperative radiotherapy (TARGIT-IORT) or external beam radiotherapy (EBRT). INTERVENTIONS Random allocation was to the EBRT arm, which consisted of a standard daily fractionated course (three to six weeks) of whole breast radiotherapy, or the TARGIT-IORT arm. TARGIT-IORT was given immediately after lumpectomy under the same anaesthetic and was the only radiotherapy for most patients (around 80%). TARGIT-IORT was supplemented by EBRT when postoperative histopathology found unsuspected higher risk factors (around 20% of patients). MAIN OUTCOME MEASURES Non-inferiority with a margin of 2.5% for the absolute difference between the five year local recurrence rates of the two arms, and long term survival outcomes. RESULTS Between 24 March 2000 and 25 June 2012, 1140 patients were randomised to TARGIT-IORT and 1158 to EBRT. TARGIT-IORT was non-inferior to EBRT: the local recurrence risk at five year complete follow-up was 2.11% for TARGIT-IORT compared with 0.95% for EBRT (difference 1.16%, 90% confidence interval 0.32 to 1.99). In the first five years, 13 additional local recurrences were reported (24/1140 v 11/1158) but 14 fewer deaths (42/1140 v 56/1158) for TARGIT-IORT compared with EBRT. With long term follow-up (median 8.6 years, maximum 18.90 years, interquartile range 7.0-10.6) no statistically significant difference was found for local recurrence-free survival (hazard ratio 1.13, 95% confidence interval 0.91 to 1.41, P=0.28), mastectomy-free survival (0.96, 0.78 to 1.19, P=0.74), distant disease-free survival (0.88, 0.69 to 1.12, P=0.30), overall survival (0.82, 0.63 to 1.05, P=0.13), and breast cancer mortality (1.12, 0.78 to 1.60, P=0.54). Mortality from other causes was significantly lower (0.59, 0.40 to 0.86, P=0.005). CONCLUSION For patients with early breast cancer who met our trial selection criteria, risk adapted immediate single dose TARGIT-IORT during lumpectomy was an effective alternative to EBRT, with comparable long term efficacy for cancer control and lower non-breast cancer mortality. TARGIT-IORT should be discussed with eligible patients when breast conserving surgery is planned. TRIAL REGISTRATION ISRCTN34086741, NCT00983684.
Collapse
Affiliation(s)
- Jayant S Vaidya
- Division of Surgery and Interventional Science, University College London, 43-45 Foley Street, London W1W 7JN, UK
| | - Max Bulsara
- Department of Biostatistics, University of Notre Dame, Fremantle, WA, Australia
| | - Michael Baum
- Division of Surgery and Interventional Science, University College London, 43-45 Foley Street, London W1W 7JN, UK
| | - Frederik Wenz
- Department of Radiation Oncology, University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Samuele Massarut
- Department of Surgery, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Steffi Pigorsch
- Department of Gynaecology and Obstetrics, Red Cross Hospital, Technical University of Munich, Munich, Germany
| | - Michael Alvarado
- Department of Surgery, University of California, San Francisco, CA, USA
| | - Michael Douek
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | | | - Henrik L Flyger
- Department of Breast Surgery, University of Copenhagen, Copenhagen, Denmark
| | - Wolfgang Eiermann
- Department of Gynaecology and Obstetrics, Red Cross Hospital, Technical University of Munich, Munich, Germany
| | - Chris Brew-Graves
- Division of Surgery and Interventional Science, University College London, 43-45 Foley Street, London W1W 7JN, UK
| | - Norman R Williams
- Division of Surgery and Interventional Science, University College London, 43-45 Foley Street, London W1W 7JN, UK
| | - Ingrid Potyka
- Division of Surgery and Interventional Science, University College London, 43-45 Foley Street, London W1W 7JN, UK
| | - Nicholas Roberts
- Division of Surgery and Interventional Science, University College London, 43-45 Foley Street, London W1W 7JN, UK
| | | | - Douglas Brown
- Department of Surgery, Ninewells Hospital, Dundee, UK
| | - Elena Sperk
- Department of Radiation Oncology, University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Siobhan Laws
- Department of Surgery, Royal Hampshire County Hospital, Winchester, UK
| | - Marc Sütterlin
- Department of Gynaecology and Obstetrics, University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Tammy Corica
- Department of Radiation Oncology, Sir Charles Gairdner Hospital, Perth, WA, Australia
| | - Steinar Lundgren
- Department of Oncology, St Olav's University Hospital, Trondheim, Norway
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Dennis Holmes
- University of Southern California, John Wayne Cancer Institute & Helen Rey Breast Cancer Foundation, Los Angeles, CA, USA
| | - Lorenzo Vinante
- Department of Radiation Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | | | - Montserrat Pazos
- Department of Radiation Oncology, University Hospital, The Ludwig Maximilian University of Munich, Munich, Germany
| | | | | | - Wojciech Polkowski
- Department of Surgical Oncology, Medical University of Lublin, Lublin, Poland
| | | | | | - Jens Blohmer
- Sankt Gertrauden Hospital, Charité, Medical University of Berlin, Berlin, Germany
| | - David McCready
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | | | - Pond Kelemen
- Ashikari Breast Center, New York Medical College, New York, NY, USA
| | - Gloria Petralia
- Department of Surgery, University College London Hospitals, London, UK
| | - Mary Falzon
- Department of Pathology, University College London Hospitals, London, UK
| | - David J Joseph
- Department of Radiation Oncology, Sir Charles Gairdner Hospital, Perth, WA, Australia
| | | |
Collapse
|
8
|
Vaidya JS, Bulsara M, Saunders C, Flyger H, Tobias JS, Corica T, Massarut S, Wenz F, Pigorsch S, Alvarado M, Douek M, Eiermann W, Brew-Graves C, Williams N, Potyka I, Roberts N, Bernstein M, Brown D, Sperk E, Laws S, Sütterlin M, Lundgren S, Holmes D, Vinante L, Bozza F, Pazos M, Le Blanc-Onfroy M, Gruber G, Polkowski W, Dedes KJ, Niewald M, Blohmer J, McCready D, Hoefer R, Kelemen P, Petralia G, Falzon M, Baum M, Joseph D. Effect of Delayed Targeted Intraoperative Radiotherapy vs Whole-Breast Radiotherapy on Local Recurrence and Survival: Long-term Results From the TARGIT-A Randomized Clinical Trial in Early Breast Cancer. JAMA Oncol 2020; 6:e200249. [PMID: 32239210 PMCID: PMC7348682 DOI: 10.1001/jamaoncol.2020.0249] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Importance Conventional adjuvant radiotherapy for breast cancer given daily for several weeks is onerous and expensive. Some patients may be obliged to choose a mastectomy instead, and some may forgo radiotherapy altogether. We proposed a clinical trial to test whether radiotherapy could be safely limited to the tumor bed. Objective To determine whether delayed second-procedure targeted intraoperative radiotherapy (TARGIT-IORT) is noninferior to whole-breast external beam radiotherapy (EBRT) in terms of local control. Design, Setting, and Participants In this prospective, randomized (1:1 ratio) noninferiority trial, 1153 patients aged 45 years or older with invasive ductal breast carcinoma smaller than 3.5 cm treated with breast conservation were enrolled from 28 centers in 9 countries. Data were locked in on July 3, 2019. Interventions The TARGIT-A trial was started in March 2000; patients were randomized after needle biopsy to receive TARGIT-IORT immediately after lumpectomy under the same anesthetic vs EBRT and results have been shown to be noninferior. A parallel study, described in this article, was initiated in 2004; patients who had their cancer excised were randomly allocated using separate randomization tables to receive EBRT or delayed TARGIT-IORT given as a second procedure by reopening the lumpectomy wound. Main Outcomes and Measures A noninferiority margin for local recurrence rate of 2.5% at 5 years, and long-term survival outcomes. Results Overall, 581 women (mean [SD] age, 63 [7] years) were randomized to delayed TARGIT-IORT and 572 patients (mean [SD] age, 63 [8] years) were randomized to EBRT. Sixty patients (5%) had tumors larger than 2 cm, or had positive nodes and only 32 (2.7%) were younger than 50 years. Delayed TARGIT-IORT was not noninferior to EBRT. The local recurrence rates at 5-year complete follow-up were: delayed TARGIT-IORT vs EBRT (23/581 [3.96%] vs 6/572 [1.05%], respectively; difference, 2.91%; upper 90% CI, 4.4%). With long-term follow-up (median [IQR], 9.0 [7.5-10.5] years), there was no statistically significant difference in local recurrence-free survival (HR, 0.75; 95% CI, 0.57-1.003; P = .052), mastectomy-free survival (HR, 0.88; 95% CI, 0.65-1.18; P = .38), distant disease-free survival (HR, 1.00; 95% CI, 0.72-1.39; P = .98), or overall survival (HR, 0.96; 95% CI, 0.68-1.35; P = .80). Conclusions and Relevance These long-term data show that despite an increase in the number of local recurrences with delayed TARGIT-IORT, there was no statistically significant decrease in mastectomy-free survival, distant disease-free survival, or overall survival. Trial Registration ISRCTN34086741, ClinicalTrials.gov Identifier: NCT00983684.
Collapse
Affiliation(s)
- Jayant S Vaidya
- Division of Surgery and Interventional Science, University College London, London, United Kingdom
| | - Max Bulsara
- Division of Surgery and Interventional Science, University College London, London, United Kingdom.,Department of Biostatistics, University of Notre Dame, Fremantle, West Australia, Australia
| | - Christobel Saunders
- University of Western Australia School of Surgery, West Australia, Australia
| | - Henrik Flyger
- Department of Breast Surgery, University of Copenhagen, Copenhagen, Denmark
| | - Jeffrey S Tobias
- Department of Clinical Oncology, University College London Hospitals, London, United Kingdom
| | - Tammy Corica
- Department of Radiation Oncology, Sir Charles Gairdner Hospital, Perth, West Australia, Australia
| | - Samuele Massarut
- Department of Surgery, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Frederik Wenz
- University Medical Center Mannheim, Department of Radiation Oncology, Medical Faculty Mannheim, Heidelberg University, Germany
| | - Steffi Pigorsch
- Red Cross Hospital, Department of Gynecology and Obstetrics, Technical University of Munich, Munich, Germany
| | | | - Michael Douek
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Wolfgang Eiermann
- Red Cross Hospital, Department of Gynecology and Obstetrics, Technical University of Munich, Munich, Germany
| | - Chris Brew-Graves
- Division of Surgery and Interventional Science, University College London, London, United Kingdom
| | - Norman Williams
- Division of Surgery and Interventional Science, University College London, London, United Kingdom
| | - Ingrid Potyka
- Division of Surgery and Interventional Science, University College London, London, United Kingdom
| | - Nicholas Roberts
- Division of Surgery and Interventional Science, University College London, London, United Kingdom
| | | | - Douglas Brown
- Department of Surgery, Ninewells Hospital, Dundee, United Kingdom
| | - Elena Sperk
- University Medical Center Mannheim, Department of Radiation Oncology, Medical Faculty Mannheim, Heidelberg University, Germany
| | - Siobhan Laws
- Department of Surgery, Royal Hampshire County Hospital, Winchester, United Kingdom
| | - Marc Sütterlin
- University Medical Center Mannheim, Department of Gynecology and Obstetrics, Medical Faculty Mannheim, Heidelberg University, Germany
| | - Steinar Lundgren
- Department of Oncology, St Olav's University Hospital, Trondheim, Norway.,Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Dennis Holmes
- Helen Rey Breast Cancer Foundation, John Wayne Cancer Institute, University of Southern California, Los Angeles
| | - Lorenzo Vinante
- Department of Radiation Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | | | - Montserrat Pazos
- University Hospital, Department of Radiation Oncology, Ludwig Maximilians Universitat, Munich, Germany
| | | | | | - Wojciech Polkowski
- Department of Surgical Oncology, Medical University of Lublin, Lublin, Poland
| | | | | | - Jens Blohmer
- Sankt Gertrauden-Krankenhaus, and The Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - David McCready
- Princess Margaret Cancer Centre Toronto, Toronto, Ontario, Canada
| | | | - Pond Kelemen
- Ashikari Breast Center, New York Medical College, New York, New York
| | - Gloria Petralia
- Department of Surgery, University College London Hospitals, London, United Kingdom
| | - Mary Falzon
- Department of Pathology, University College London Hospitals, London, United Kingdom
| | - Michael Baum
- Division of Surgery and Interventional Science, University College London, London, United Kingdom
| | - David Joseph
- Department of Radiation Oncology, Sir Charles Gairdner Hospital, Perth, West Australia, Australia
| |
Collapse
|
9
|
Citron F, Segatto I, Vinciguerra GLR, Musco L, Russo F, Mungo G, D'Andrea S, Mattevi MC, Perin T, Schiappacassi M, Massarut S, Marchini C, Amici A, Vecchione A, Baldassarre G, Belletti B. Downregulation of miR-223 Expression Is an Early Event during Mammary Transformation and Confers Resistance to CDK4/6 Inhibitors in Luminal Breast Cancer. Cancer Res 2019; 80:1064-1077. [PMID: 31862778 DOI: 10.1158/0008-5472.can-19-1793] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 11/19/2019] [Accepted: 12/17/2019] [Indexed: 11/16/2022]
Abstract
miR-223 is an anti-inflammatory miRNA that in cancer acts either as an oncosuppressor or oncopromoter, in a context-dependent manner. In breast cancer, we demonstrated that it dampens the activation of the EGF pathway. However, little is known on the role of miR-223 during breast cancer onset and progression. miR-223 expression was decreased in breast cancer of luminal and HER2 subtypes and inversely correlated with patients' prognosis. In normal luminal mammary epithelial cells, miR-223 acted cell autonomously in the control of their growth and morphology in three-dimensional context. In the MMTV-Δ16HER2 transgenic mouse model, oncogene transformation resulted in a timely abrogation of miR-223 expression, likely due to activation of E2F1, a known repressor of miR-223 transcription. Accordingly, treatment with CDK4/6 inhibitors, which eventually results in restraining E2F1 activity, restored miR-223 expression and miR-223 ablation induced luminal breast cancer resistance to CDK4/6 inhibition, both in vitro and in vivo. Notably, miR-223 expression was lost in microdissected ductal carcinoma in situ (DCIS) from patients with luminal and HER2-positive breast cancer. Altogether, these results identify downmodulation of miR-223 as an early step in luminal breast cancer onset and suggest that it could be used to identify aggressive DCIS and predict the response to targeted therapy. SIGNIFICANCE: miR-223 may represent a predictive biomarker of response to CDK4/6 inhibitors and its loss could identify DCIS lesions that are likely to progress into invasive breast cancer.
Collapse
Affiliation(s)
- Francesca Citron
- Molecular Oncology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy
| | - Ilenia Segatto
- Molecular Oncology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy
| | - Gian Luca Rampioni Vinciguerra
- Molecular Oncology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy.,Faculty of Medicine and Psychology, Department of Clinical and Molecular Medicine, University of Rome "Sapienza" Sant'Andrea Hospital, Rome, Italy
| | - Lorena Musco
- Molecular Oncology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy
| | - Francesca Russo
- Molecular Oncology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy
| | - Giorgia Mungo
- Molecular Oncology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy
| | - Sara D'Andrea
- Molecular Oncology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy
| | - Maria Chiara Mattevi
- Molecular Oncology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy
| | - Tiziana Perin
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy
| | - Monica Schiappacassi
- Molecular Oncology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy
| | - Samuele Massarut
- Breast Surgery Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy
| | - Cristina Marchini
- Department of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Augusto Amici
- Department of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Andrea Vecchione
- Faculty of Medicine and Psychology, Department of Clinical and Molecular Medicine, University of Rome "Sapienza" Sant'Andrea Hospital, Rome, Italy
| | - Gustavo Baldassarre
- Molecular Oncology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy.
| | - Barbara Belletti
- Molecular Oncology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy.
| |
Collapse
|
10
|
Zhang J, Li K, Zhang Y, Lu R, Wu S, Tang J, Xia Y, Sun J. Deletion of sorting nexin 27 suppresses proliferation in highly aggressive breast cancer MDA-MB-231 cells in vitro and in vivo. BMC Cancer 2019; 19:555. [PMID: 31182056 PMCID: PMC6558813 DOI: 10.1186/s12885-019-5769-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 05/29/2019] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Sorting Nexin 27 (SNX27) belongs to a family of sortin nexins and possesses a unique binding domain at the C-terminus which mediates protein-protein interaction in intracellular trafficking, membrane remodeling, organelle motility, and tight junctions. However, its role in cancer development, especially in vivo, remains largely unknown. METHODS We have generated a stable SNX27 knockdown clone in a highly aggressive breast cancer cell line MDA-MB-231 using an inducible lentiviral shRNA system. Cell migration and proliferation of SNX27 knockdown (KD) cells were compared with wild-type (WT) cells by MTT and wound healing assay, respectively. The differences in colony formation between SNX27-KD and WT cells were detected by soft agar culture and matrigel 3D culture. Furthermore, tumor growth was examined in a xenograft nude mouse model using SNX27-KD and WT MDA-MB-231 cells. The critical EMT (epithelial-mesenchymal transition) regulators were examined in vitro and in vivo. RESULTS The wound healing assay showed that SNX27 knockdown significantly decreased cell motility and proliferation. Colony formation in soft agar showed that the SNX27 knockdown cells formed significantly fewer and smaller colonies than the parental MDA-MB-231 cells. Western blots and immunostaining showed that knockdown of SNX27 led to increased expression of E-cadherin and β-catenin proteins, which facilitate adhesion formation and reverse EMT. EMT is a cellular program that allows polarized, immotile epithelial cells to convert to motile mesenchymal cells, promoting carcinoma invasion. The expression levels of Vimentin, the transcription factor of EMT, and tight junction protein Claudin-5, were significantly diminished in the SNX27 knockdown cells. The expression of PCNA, the cell proliferation marker, was increased in SNX27-KD cells transfected with E-cadherin siRNA. In a xenograft nude mouse model, we found that knockdown of SNX27 significantly inhibited tumor growth. The tumors from mice with SNX27-KD cells showed less proliferation compared to tumors from mice injected with wildtype cells. The increase in E-cadherin and β-catenin and decrease in Vimentin and Claudin-5 were observed in tumors of mice injected with SNX27-KD cells. CONCLUSIONS Our data have demonstrated that SNX27 plays a crucial role in tumor growth in vitro and in vivo.
Collapse
Affiliation(s)
- Jilei Zhang
- Division of Gastroenterology and Hepatology, Medicine, University of Illinois at Chicago, 840 S Wood Street, Room 704 CSB, MC716, Chicago, IL, 60612, USA
| | - Kendy Li
- Liberal Arts & Human Sciences, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
| | - Yongguo Zhang
- Division of Gastroenterology and Hepatology, Medicine, University of Illinois at Chicago, 840 S Wood Street, Room 704 CSB, MC716, Chicago, IL, 60612, USA
| | - Rong Lu
- Division of Gastroenterology and Hepatology, Medicine, University of Illinois at Chicago, 840 S Wood Street, Room 704 CSB, MC716, Chicago, IL, 60612, USA
| | - Shaoping Wu
- Department of Biochemistry, Rush University, Chicago, IL, USA
| | - Jingrong Tang
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health, Bethesda, MD, USA
| | - Yinglin Xia
- Division of Gastroenterology and Hepatology, Medicine, University of Illinois at Chicago, 840 S Wood Street, Room 704 CSB, MC716, Chicago, IL, 60612, USA
| | - Jun Sun
- Division of Gastroenterology and Hepatology, Medicine, University of Illinois at Chicago, 840 S Wood Street, Room 704 CSB, MC716, Chicago, IL, 60612, USA.
| |
Collapse
|
11
|
Fararjeh AFS, Tu SH, Chen LC, Cheng TC, Liu YR, Chang HL, Chang HW, Huang CC, Wang HCR, Hwang-Verslues WW, Wu CH, Ho YS. Long-term exposure to extremely low-dose of nicotine and 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) induce non-malignant breast epithelial cell transformation through activation of the a9-nicotinic acetylcholine receptor-mediated signaling pathway. ENVIRONMENTAL TOXICOLOGY 2019; 34:73-82. [PMID: 30259641 DOI: 10.1002/tox.22659] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 09/03/2018] [Accepted: 09/06/2018] [Indexed: 06/08/2023]
Abstract
Breast cancer (BC) is the most common cancer affecting women worldwide and has been associated with active tobacco smoking. Low levels of nicotine (Nic) and 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK), have been detected in cases of second-hand smoke (SHS). However, the correlation between SHS and BC risk remains controversial. In this study, we investigated whether the physiological SHS achievable dose of Nic and tobacco specific nitrosamine, NNK act together to induce breast carcinogenesis using an in vitro breast cell carcinogenesis model. Immortalized non-tumorigenic breast epithelial cell line, HBL-100 used for a time-course assay, was exposed to very low levels of either Nic or NNK, or both. The time-course assay consisted of 23 cycles of nitrosamines treatment. In each cycle, HBL-100 cells were exposed to 1pM of Nic and/or 100 femtM of NNK for 48 hours. Cells were passaged every 3 days and harvested after 10, 15, and 23 cycles. Our results demonstrated that the tumorigenicity of HBL-100, defined by soft agar colony forming, proliferation, migration and invasion abilities, was enhanced by co-exposure to physiologically SHS achievable doses of Nic and NNK. In addition, α9-nAChR signaling activation, which plays an important role in cellular proliferation and cell survival, was also observed. Importantly, an increase in stemness properties including the prevalence of CD44+/CD24- cells, increase Nanog expression and mammosphere-forming ability were also observed. Our results indicate that chronic and long term exposure to environmental tobacco smoke, may induce breast cell carcinogenesis even at extremely low doses.
Collapse
MESH Headings
- Acetylcholine/metabolism
- Breast Neoplasms/chemically induced
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Carcinogens/toxicity
- Cell Proliferation/drug effects
- Cell Proliferation/genetics
- Cell Transformation, Neoplastic/drug effects
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Cells, Cultured
- Dose-Response Relationship, Drug
- Epithelial Cells/drug effects
- Epithelial Cells/physiology
- Female
- Humans
- Mammary Glands, Human/drug effects
- Mammary Glands, Human/pathology
- Mammary Glands, Human/physiology
- Nicotine/toxicity
- Nitrosamines/toxicity
- Receptors, Nicotinic/genetics
- Receptors, Nicotinic/metabolism
- Signal Transduction/drug effects
- Signal Transduction/genetics
- Time Factors
- Toxicity Tests, Chronic
Collapse
Affiliation(s)
- Abdul-Fattah Salah Fararjeh
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei, Taiwan
| | - Shih-Hsin Tu
- Breast Medical Center, Taipei Medical University Hospital, Taipei, Taiwan
- Taipei Cancer Center, Taipei Medical University, Taipei, Taiwan
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Li-Ching Chen
- Breast Medical Center, Taipei Medical University Hospital, Taipei, Taiwan
- Taipei Cancer Center, Taipei Medical University, Taipei, Taiwan
- TMU Research Center of cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Tzu-Chun Cheng
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yun-Ru Liu
- TMU Research Center of cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
- Joint Biobank, Office of Human Research, Taipei Medical University, Taipei, Taiwan
| | - Hang-Lung Chang
- Department of General Surgery, En Chun Kong Hospital, New Taipei City, Taiwan
| | - Hui-Wen Chang
- Department of Laboratory Medicine, Taipei Medical University Hospital, Taipei, Taiwan
| | - Chi-Cheng Huang
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- School of Medicine, College of Medicine, Fu-Jen Catholic University, New Taipei City, Taiwan
- Department of Surgery, Fu-Jen Catholic University Hospital, New Taipei City, Taiwan
| | - Hwa-Chain Robert Wang
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee, USA
| | | | - Chih-Hsiung Wu
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of General Surgery, En Chun Kong Hospital, New Taipei City, Taiwan
| | - Yuan-Soon Ho
- TMU Research Center of cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Laboratory Medicine, Taipei Medical University Hospital, Taipei, Taiwan
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
12
|
Segatto I, Zompit MDM, Citron F, D'Andrea S, Vinciguerra GLR, Perin T, Berton S, Mungo G, Schiappacassi M, Marchini C, Amici A, Vecchione A, Baldassarre G, Belletti B. Stathmin Is Required for Normal Mouse Mammary Gland Development and Δ16HER2-Driven Tumorigenesis. Cancer Res 2018; 79:397-409. [PMID: 30478213 DOI: 10.1158/0008-5472.can-18-2488] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 10/17/2018] [Accepted: 11/19/2018] [Indexed: 11/16/2022]
Abstract
Postnatal development of the mammary gland relies on the maintenance of oriented cell division and apicobasal polarity, both of which are often deregulated in cancer. The microtubule (MT) network contributes to control these processes; however, very little is known about the impact of altered MT dynamics in the development of a complex organ and on the role played by MT-interacting proteins such as stathmin. In this study, we report that female stathmin knock-out (STM KO) mice are unable to nurse their litters due to frank impairment of mammary gland development. In mouse mammary epithelial cells, loss of stathmin compromised the trafficking of polarized proteins and the achievement of proper apicobasal polarity. In particular, prolactin receptor internalization and localization was altered in STM KO mammary epithelial cells, leading to decreased protein stability and downmodulation of the Prl/PrlR/STAT5 signaling pathway. Absence of stathmin induced alterations in mitotic spindle orientation, accumulation of mitotic defects, and apoptosis, overall contributing to tissue disorganization and further decreasing the expansion of the mammary epithelial compartment. Loss of stathmin in MMTV-Δ16HER2 transgenic mice decreased the incidence and increased the latency of these very aggressive mammary carcinomas. Collectively, these data identify the essential mammary protein stathmin as protumorigenic and suggest it may serve as a potential therapeutic target in breast cancer. SIGNIFICANCE: Stathmin expression is critical to maintain oriented cell division and apicobasal polarity in normal mammary glands and to establish a protumorigenic program that eventually sustains HER2-positive breast cancer formation in mice.
Collapse
Affiliation(s)
- Ilenia Segatto
- Unit of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy
| | - Mara De Marco Zompit
- Unit of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy
| | - Francesca Citron
- Unit of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy
| | - Sara D'Andrea
- Unit of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy
| | - Gian Luca Rampioni Vinciguerra
- Unit of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy.,Faculty of Medicine and Psychology, Department of Clinical and Molecular Medicine, University of Rome "Sapienza" Sant'Andrea Hospital, Rome, Italy
| | - Tiziana Perin
- Unit of Pathology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy
| | - Stefania Berton
- Unit of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy
| | - Giorgia Mungo
- Unit of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy
| | - Monica Schiappacassi
- Unit of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy
| | - Cristina Marchini
- Department of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Augusto Amici
- Department of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Andrea Vecchione
- Faculty of Medicine and Psychology, Department of Clinical and Molecular Medicine, University of Rome "Sapienza" Sant'Andrea Hospital, Rome, Italy
| | - Gustavo Baldassarre
- Unit of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy.
| | - Barbara Belletti
- Unit of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy.
| |
Collapse
|
13
|
STAT3 in Breast Cancer Onset and Progression: A Matter of Time and Context. Int J Mol Sci 2018; 19:ijms19092818. [PMID: 30231553 PMCID: PMC6163512 DOI: 10.3390/ijms19092818] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 09/13/2018] [Accepted: 09/16/2018] [Indexed: 01/05/2023] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) is responsible for mediating the transcriptional programs downstream of several cytokine, growth factor, and oncogenic stimuli. Its expression and activity are consistently linked to cellular transformation, as well as tumor initiation and progression. Due to this central role, STAT3 is widely considered a good target for anti-cancer therapy; however, the success of these approaches has been, so far, very limited. Notably, on one side, STAT3 is aberrantly active in many breast cancers, on the other, at the physiological level, it is the main mediator of epithelial cell death during post-lactation mammary-gland involution, thus strongly suggesting that its biological functions are highly context-specific. One of the most peculiar features of STAT3 is that it can act both in cell-autonomous and non-cell-autonomous manners, simultaneously modulating the phenotypes of the tumor cells and their microenvironment. Here, we focus on the role of STAT3 in breast cancer progression, discussing the potential contrasting roles of STAT3 activation in the establishment of locally recurrent and distant metastatic disease. Based on the most recent literature, depending on the tumor cell type, the local microenvironment status, and the stage of the disease, either STAT3 activation or inactivation can support disease progression. Accordingly, cancer cells dynamically exploit STAT3 activity to carry out transcriptional programs somehow contrasting and complementary, such as supporting survival and growth, dormancy and awakening, stem cell-like features, and inflammation, immune response, and immune evasion. As a consequence, to achieve clinical efficacy, the conception and testing of anti-STAT3 targeted therapies will need a very careful evaluation of these opposing roles and of the most appropriate tumor context, disease stage and patient population to treat.
Collapse
|
14
|
Gomesin inhibits melanoma growth by manipulating key signaling cascades that control cell death and proliferation. Sci Rep 2018; 8:11519. [PMID: 30068931 PMCID: PMC6070509 DOI: 10.1038/s41598-018-29826-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 07/10/2018] [Indexed: 02/03/2023] Open
Abstract
Consistent with their diverse pharmacology, peptides derived from venomous animals have been developed as drugs to treat disorders as diverse as hypertension, diabetes and chronic pain. Melanoma has a poor prognosis due in part to its metastatic capacity, warranting further development of novel targeted therapies. This prompted us to examine the anti-melanoma activity of the spider peptides gomesin (AgGom) and a gomesin-like homolog (HiGom). AgGom and HiGom dose-dependently reduced the viability and proliferation of melanoma cells whereas it had no deleterious effects on non-transformed neonatal foreskin fibroblasts. Concordantly, gomesin-treated melanoma cells showed a reduced G0/G1 cell population. AgGom and HiGom compromised proliferation of melanoma cells via activation of the p53/p21 cell cycle check-point axis and the Hippo signaling cascade, together with attenuation of the MAP kinase pathway. We show that both gomesin peptides exhibit antitumoral activity in melanoma AVATAR-zebrafish xenograft tumors and that HiGom also reduces tumour progression in a melanoma xenograft mouse model. Taken together, our data highlight the potential of gomesin for development as a novel melanoma-targeted therapy.
Collapse
|
15
|
Segatto I, Massarut S, Boyle R, Baldassarre G, Walker D, Belletti B. Preclinical validation of a novel compound targeting p70S6 kinase in breast cancer. Aging (Albany NY) 2017; 8:958-76. [PMID: 27155197 PMCID: PMC4931847 DOI: 10.18632/aging.100954] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 04/20/2016] [Indexed: 11/25/2022]
Abstract
UNLABELLED Breast cancer is a frequent and treatable disease. However, when recurrent, breast cancer often becomes refractory to therapy and progresses into metastatic forms that are typically incurable. Thus, understanding and targeting the critical pathways underlying breast cancer recurrence is urgently needed to eradicate primary disease and achieve better prognosis. Recently, we have demonstrated that the ribosomal protein p70S6K is activated in residual breast cancer cells as a result of post-surgical inflammation and that interfering with its activity in the peri-operative setting strongly suppresses recurrence in a mouse model. In order to develop clinically-exploitable treatments targeting p70S6K, we have tested a newly generated compound, called FS-115. FS-115 potently inhibited p70S6K1 (IC50 35nM) with high selectivity over other AGC kinases or PI3K pathway kinases. In vitro, treatment with FS-115 efficiently blocked p70S6K activity in breast cancer cell lines and impaired colony formation and anchorage independent growth. Pharmacokinetic profiling showed that FS-115 exhibited high oral bioavailability, optimal plasma distribution and high brain penetrance. In nude mice, FS-115 strongly suppressed tumor take-rate and primary tumor growth. Oral dosing with FS-115 in a peri-operative schedule was effective in decreasing local recurrence of breast cancer and a long-term treatment schedule was well tolerated and efficiently suppressed distant metastasis formation. Altogether, we propose that FS-115 might be a good candidate for the treatment of breast cancer patients at high risk to relapse. SUMMARY STATEMENT Our results confirm that inhibition of p70S6K represents a valuable opportunity for restraining loco-regional relapse and metastasis in breast cancer and identify in FS-115 a promising candidate-inhibitor to move from preclinical to clinical treatments.
Collapse
Affiliation(s)
- Ilenia Segatto
- Division of Molecular Oncology, CRO, National Cancer Institute, Aviano 33081, Italy
| | - Samuele Massarut
- Breast Surgery Unit, CRO, National Cancer Institute, Aviano 33081, Italy
| | - Robert Boyle
- Sentinel Oncology Limited, Cambridge, United Kingdom
| | - Gustavo Baldassarre
- Division of Molecular Oncology, CRO, National Cancer Institute, Aviano 33081, Italy
| | - David Walker
- Sentinel Oncology Limited, Cambridge, United Kingdom
| | - Barbara Belletti
- Division of Molecular Oncology, CRO, National Cancer Institute, Aviano 33081, Italy
| |
Collapse
|
16
|
Kaverina N, Borovjagin AV, Kadagidze Z, Baryshnikov A, Baryshnikova M, Malin D, Ghosh D, Shah N, Welch DR, Gabikian P, Karseladze A, Cobbs C, Ulasov IV. Astrocytes promote progression of breast cancer metastases to the brain via a KISS1-mediated autophagy. Autophagy 2017; 13:1905-1923. [PMID: 28981380 PMCID: PMC5788498 DOI: 10.1080/15548627.2017.1360466] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 07/07/2017] [Accepted: 07/24/2017] [Indexed: 12/14/2022] Open
Abstract
Formation of metastases, also known as cancer dissemination, is an important stage of breast cancer (BrCa) development. KISS1 expression is associated with inhibition of metastases development. Recently we have demonstrated that BrCa metastases to the brain exhibit low levels of KISS1 expression at both mRNA and protein levels. By using multicolor immunofluorescence and coculture techniques here we show that normal adult astrocytes in the brain are capable of promoting metastatic transformation of circulating breast cancer cells localized to the brain through secretion of chemokine CXCL12. The latter was found in this study to downregulate KISS1 expression at the post-transcriptional level via induction of microRNA-345 (MIR345). Furthermore, we demonstrated that ectopic expression of KISS1 downregulates ATG5 and ATG7, 2 key modulators of autophagy, and works concurrently with autophagy inhibitors, thereby implicating autophagy in the mechanism of KISS1-mediated BrCa metastatic transformation. We also found that expression of KISS1 in human breast tumor specimens inversely correlates with that of MMP9 and IL8, implicated in the mechanism of metastatic invasion, thereby supporting the role of KISS1 as a potential regulator of BrCa metastatic invasion in the brain. This conclusion is further supported by the ability of KISS1, ectopically overexpressed from an adenoviral vector in MDA-MB-231Br cells with silenced expression of the endogenous gene, to revert invasive phenotype of those cells. Taken together, our results strongly suggest that human adult astrocytes can promote brain invasion of the brain-localized circulating breast cancer cells by upregulating autophagy signaling pathways via the CXCL12-MIR345- KISS1 axis.
Collapse
Affiliation(s)
- Natalya Kaverina
- Department of Tumor Immunology, Institute of Experimental Diagnostics and Therapy of Tumors, N.N. Blokhin Cancer Research Center, Moscow, Russia
| | - Anton V. Borovjagin
- University of Alabama at Birmingham School of Dentistry, Institute of Oral Health Research, Birmingham, AL, USA
| | - Zaira Kadagidze
- Department of Tumor Immunology, Institute of Experimental Diagnostics and Therapy of Tumors, N.N. Blokhin Cancer Research Center, Moscow, Russia
| | - Anatoly Baryshnikov
- Institute of Experimental Diagnostics and Therapy of Tumors, N.N. Blokhin Cancer Research Center, Moscow, Russia
| | - Maria Baryshnikova
- Institute of Experimental Diagnostics and Therapy of Tumors, N.N. Blokhin Cancer Research Center, Moscow, Russia
| | - Dmitry Malin
- Department of Endocrinology, University of Wisconsin-Madison, Madison, WI, USA
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Dhimankrishhna Ghosh
- Center for Advanced Brain Tumor Treatment, Swedish Neuroscience Institute, Seattle, WA, USA
| | - Nameeta Shah
- Center for Advanced Brain Tumor Treatment, Swedish Neuroscience Institute, Seattle, WA, USA
| | - Danny R. Welch
- Department of Cancer Biology, Kansas University Medical Center (KUMC), Kansas City, KS, USA
| | - Patrik Gabikian
- Department of Neurosurgery, Kaiser Permanente Los Angeles Medical Center, Los Angeles, CA, USA
| | - Apollon Karseladze
- Pathology, Institute of Experimental Diagnostics and Therapy of Tumors, N.N. Blokhin Cancer Research Center, Moscow, Russia
| | - Charles Cobbs
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Ilya V. Ulasov
- Center for Advanced Brain Tumor Treatment, Swedish Neuroscience Institute, Seattle, WA, USA
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
17
|
Yang F, Gao JY, Chen H, Du ZH, Zhang XQ, Gao W. Targeted inhibition of the phosphoinositide 3-kinase impairs cell proliferation, survival, and invasion in colon cancer. Onco Targets Ther 2017; 10:4413-4422. [PMID: 28979133 PMCID: PMC5602681 DOI: 10.2147/ott.s145601] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Background Colon cancer is the third most common cancer in the world, and its metastasis and drug resistance are challenging for its effective treatment. The PI3K/Akt/mTOR pathway plays a crucial role in the pathogenesis of colon cancer. The aim of this study was to investigate the targeting of PI3K in colon cancer cells HT-29 and HCT-116 in vitro. Methods In HT-29 and HCT-116 cells, BEZ235, a dual inhibitor of PI3K/mTOR, and shRNAtarget to PI3KCA were used to inhibit PI3K/Akt/mTOR pathway. The inhibition efficiency of PI3K/Akt/mTOR pathway was detected by RT-PCR and Western blot. Cell proliferation, migration, invasion, and apoptosis were evaluated by Cell Counting Kit-8, Transwell, and flow cytometry assays. The expression of apoptosis-related proteins (cleavage caspase 3, Bcl-2, Bax, and Bim) were also detected. Results We found that in HT-29 and HCT-116 cells, the treatment of BEZ235 (1 μM) and PI3KCA knockdown inhibited the activation of PI3K/Akt/mTOR pathway and significantly suppressed cell proliferation, migration, and invasion of HT-29 and HCT-116 cells. In addition, we confirmed that knockdown of BEZ235 and PI3KCA induced cell apoptosis through the upregulated levels of cleavage caspase 3 and Bax and downregulated expression of Bcl-2 and Bim. Conclusion Our results indicated that targeted inhibition of the PI3K/Akt/mTOR pathway impaired cell proliferation, survival, and invasion in human colon cancer.
Collapse
Affiliation(s)
- Fei Yang
- Department of Pathology, Jinan Central Hospital Affiliated to Shandong University, Jinan
| | - Jun-Yi Gao
- Department of Clinical Medicine, Weifang Medical College, Weifang
| | - Hua Chen
- Department of Pathology, Jinan Central Hospital Affiliated to Shandong University, Jinan
| | - Zhen-Hua Du
- Department of Pathology, Jinan Central Hospital Affiliated to Shandong University, Jinan
| | | | - Wei Gao
- Department of Oncology, Jinan Central Hospital Affiliated to Shandong University, Jinan, People's Republic of China
| |
Collapse
|
18
|
Dana P, Kariya R, Vaeteewoottacharn K, Sawanyawisuth K, Seubwai W, Matsuda K, Okada S, Wongkham S. Upregulation of CD147 Promotes Metastasis of Cholangiocarcinoma by Modulating the Epithelial-to-Mesenchymal Transitional Process. Oncol Res 2017; 25:1047-1059. [PMID: 28244853 PMCID: PMC7841082 DOI: 10.3727/096504016x14813899000565] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
CD147 is a transmembrane protein that can induce the expression and activity of matrix metalloproteinases (MMPs). Expression of CD147 has been shown to potentiate cell migration, invasion, and metastasis of cancer. In this study, the critical role of CD147 in metastasis was elucidated using CD147-overexpressing cholangiocarcinoma (CCA) cells in vitro and in vivo. The molecular mechanism, demonstrated herein, supported the hypothesis that metastasis increased in CD147-overexpressing cells. Five CD147-overexpressing clones (Ex-CD147) were established from a low CD147-expressing CCA cell line, KKU-055, using lentivirus containing pReceiver-Lenti-CD147. The metastatic capability was determined using the tail vein injection mouse model and an in vitro 3D invasion assay. Liver colonization was assessed using anti-HLA class I immunohistochemistry. Adhesion abilities, cytoskeletal arrangements, MMP activities, the expressions of adhesion molecules, and epithelial-mesenchymal transitional markers were analyzed. All Ex-CD147 clones exhibited a high CD147 expression and high liver colonization in the tail vein-injected mouse model, whereas parental cells lacked this ability. Ex-CD147 clones exhibited metastatic phenotypes (i.e., an increase in F-actin rearrangement) and cell invasion and a decrease in cell adhesion. The molecular mechanisms were shown to be via the induction of MMP-2 activity and enhancement of epithelial-mesenchymal transitions. An increase in mesenchymal markers Slug, vimentin, and N-cadherin, and a decrease in epithelial markers E-cadherin and claudin-1, together with suppression of the adhesion molecule ICAM-1, were observed in the Ex-CD147 clones. Moreover, suppression of CD147 expression using siCD147 in two CCA cell lines with high CD147 expression significantly decreased cell migration and invasion of these CCA cells. These findings emphasize the essential role of CD147 in CCA metastasis and suggest CD147 as a promising target for the effective treatment of CCA.
Collapse
Affiliation(s)
- Paweena Dana
- *Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- †Liver Fluke and Cholangiocarcinoma Research Center, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- ‡Division of Hematopoiesis, Center for AIDS Research, Kumamoto University, Kumamoto, Japan
| | - Ryusho Kariya
- ‡Division of Hematopoiesis, Center for AIDS Research, Kumamoto University, Kumamoto, Japan
| | - Kulthida Vaeteewoottacharn
- *Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- †Liver Fluke and Cholangiocarcinoma Research Center, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Kanlayanee Sawanyawisuth
- *Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- †Liver Fluke and Cholangiocarcinoma Research Center, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Wunchana Seubwai
- †Liver Fluke and Cholangiocarcinoma Research Center, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- §Department of Forensic Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Kouki Matsuda
- ‡Division of Hematopoiesis, Center for AIDS Research, Kumamoto University, Kumamoto, Japan
| | - Seiji Okada
- ‡Division of Hematopoiesis, Center for AIDS Research, Kumamoto University, Kumamoto, Japan
| | - Sopit Wongkham
- *Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- †Liver Fluke and Cholangiocarcinoma Research Center, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
19
|
Zaleska K, Przybyła A, Kulcenty K, Wichtowski M, Mackiewicz A, Suchorska W, Murawa D. Wound fluids affect miR-21, miR-155 and miR-221 expression in breast cancer cell lines, and this effect is partially abrogated by intraoperative radiation therapy treatment. Oncol Lett 2017; 14:4029-4036. [PMID: 28943910 PMCID: PMC5592850 DOI: 10.3892/ol.2017.6718] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 07/13/2017] [Indexed: 12/31/2022] Open
Abstract
Breast cancer is the most common malignant disease occurring in women. Conservative breast cancer surgery followed by radiation therapy is currently the standard treatment for this type of cancer. The majority of metastases occur within the scar, which initiated a series of studies. As a result, clinical trials aimed to assess whether localized radiotherapy, as intraoperative radiotherapy (IORT), may more effective in inhibiting the formation of local recurrence compared with the standard postoperative whole breast radiotherapy. The present study determined the role of postoperative wound fluids (WFs) from patients diagnosed with breast cancer subsequent to breast conserving surgery or breast conserving surgery followed by IORT on the expression of three microRNAs (miRNAs), consisting of miR-21, miR-155 and miR-221, in distinct breast cancer cell lines that represent the general subtypes of breast cancer. It was determined that the miRNAs responsible for breast cancer progression, induction of tumorigenesis and enrichment of the cancer stem cell phenotype, which is responsible for resistance to tumor therapy, were highly upregulated in the human epidermal growth factor receptor 2-positive breast cancer SK-BR-3 cell line following stimulation with WFs. It is worth emphasizing, that those changes were more significant in WFs collected from patients after surgery alone. The BT-549 cell line showed altered expression only of miR-155 following incubation with WFs. Notably, this change was not associated with IORT. Additionally, it was indicated that both WFs and RT-WF strongly downregulated the expression of miR-21, miR-155 and miR-221 in basal/epithelial and luminal subtypes of breast cancer. It was concluded that the present study contributes to an increased understanding of the role of surgical WFs and IORT treatment in the regulation of miRNA expression. This may enable the development of the current knowledge of breast cancer biology subsequent to IORT treatment and substantially to improve the therapy in the future.
Collapse
Affiliation(s)
- Karolina Zaleska
- Radiobiology Laboratory, Greater Poland Cancer Centre, 61-866 Poznań, Poland
| | - Anna Przybyła
- Department of Cancer Immunology, Chair of Medical Biotechnology, Poznań University of Medical Sciences, 61-701 Poznań, Poland
| | - Katarzyna Kulcenty
- Radiobiology Laboratory, Greater Poland Cancer Centre, 61-866 Poznań, Poland.,Department of Electoradiology, Poznań University of Medical Sciences, 61-701 Poznań, Poland
| | - Mateusz Wichtowski
- First Clinic of Surgical Oncology and General Surgery, Greater Poland Cancer Centre, 61-866 Poznań, Poland
| | - Andrzej Mackiewicz
- Department of Cancer Immunology, Chair of Medical Biotechnology, Poznań University of Medical Sciences, 61-701 Poznań, Poland.,Department of Cancer Diagnostics and Immunology, Greater Poland Cancer Centre, 61-866 Poznań, Poland
| | - Wiktoria Suchorska
- Radiobiology Laboratory, Greater Poland Cancer Centre, 61-866 Poznań, Poland.,Department of Electoradiology, Poznań University of Medical Sciences, 61-701 Poznań, Poland
| | - Dawid Murawa
- First Clinic of Surgical Oncology and General Surgery, Greater Poland Cancer Centre, 61-866 Poznań, Poland.,Research and Development Centre, Regional Specialist Hospital in Wrocław, 51-124 Wrocław, Poland
| |
Collapse
|
20
|
Piotrowski I, Kulcenty K, Wichtowski M, Murawa D, Suchorska W. Intraoperative Radiotherapy of Breast Cancer and Its Biological Effects. Breast Care (Basel) 2017; 12:109-113. [PMID: 28559768 DOI: 10.1159/000454673] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Conservative breast cancer surgery followed by radiation therapy is the standard treatment for this type of cancer. Numerous studies demonstrate that 90% of local recurrences after traditional surgery occur in the same quadrant as the primary cancer. The published data suggest that the wound healing process after surgery alters the area surrounding the original tumor and the modified microenvironment is more favorable for the tumor to recur. The majority of metastases within scar initiated much research, and the consequences of these studies led to clinical trials aimed at assessing whether localized radiotherapy, such as intraoperative radiotherapy (IORT), would be more effective in inhibiting formation of local recurrence than the standard postoperative whole breast radiotherapy. IORT involves irradiation of diseased tissue directly during surgery. The rationale for this approach is the fact that the increase in the radiation dose increases local tumor control, which is the primary goal of radiation therapy. The biological basis of this process are still not thoroughly understood. Gaining new knowledge about the recurrence formation at the molecular level could serve as a starting point for further analysis and to create an opportunity to identify new targets of therapy, and possibly new therapeutic agents.
Collapse
Affiliation(s)
- Igor Piotrowski
- Radiobiology Laboratory, Department of Medical Physics, Greater Poland Cancer Centre, Pozna'n, Poland.,Department of Electroradiology, University of Medical Sciences, Pozna'n, Poland
| | - Katarzyna Kulcenty
- Radiobiology Laboratory, Department of Medical Physics, Greater Poland Cancer Centre, Pozna'n, Poland.,Department of Electroradiology, University of Medical Sciences, Pozna'n, Poland
| | - Mateusz Wichtowski
- Oncological and General Surgery Department I, Greater Poland Cancer Centre, Pozna'n, Poland
| | - Dawid Murawa
- Oncological and General Surgery Department I, Greater Poland Cancer Centre, Pozna'n, Poland
| | - Wiktoria Suchorska
- Radiobiology Laboratory, Department of Medical Physics, Greater Poland Cancer Centre, Pozna'n, Poland.,Department of Electroradiology, University of Medical Sciences, Pozna'n, Poland
| |
Collapse
|
21
|
Abstract
Breast cancer is the most common cancer among women worldwide. Great scientific, economical, and organizational efforts are in place to understand the causes of onset, identify the critical molecular players of progression, and define new lines of intervention providing more benefits and less toxicity. These efforts have certainly not been vain, since overall survival, especially in specific subsets of breast cancer, has greatly improved during the last decades. At present, breast cancer patients’ treatment and care have reached a high standard of quality, and currently one of the most urgent needs resides in the necessity to better distinguish the tumors that need to be more aggressively treated and identify the best therapeutic option tailored to each patient. This objective will be achievable only if the information clarifying the biology of breast cancer can be successfully transferred to the clinic. A common effort by scientists and clinicians toward this integration and toward the use of multidisciplinary approaches will be necessary to reach this important goal.
Collapse
Affiliation(s)
- Gustavo Baldassarre
- Department of Experimental Oncology, Department of Translational Research, CRO Aviano IRCCS, National Cancer Institute, Italy, Aviano, Italy
| | - Barbara Belletti
- Department of Experimental Oncology, Department of Translational Research, CRO Aviano IRCCS, National Cancer Institute, Italy, Aviano, Italy
| |
Collapse
|
22
|
Radiotherapy-induced miR-223 prevents relapse of breast cancer by targeting the EGF pathway. Oncogene 2016; 35:4914-26. [PMID: 26876200 PMCID: PMC5028421 DOI: 10.1038/onc.2016.23] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 11/04/2015] [Accepted: 12/18/2015] [Indexed: 12/17/2022]
Abstract
In breast cancer (BC) patients, local recurrences often arise in proximity of the surgical scar, suggesting that response to surgery may have a causative role. Radiotherapy (RT) after lumpectomy significantly reduces the risk of recurrence. We investigated the direct effects of surgery and of RT delivered intraoperatively (IORT), by collecting irradiated and non-irradiated breast tissues from BC patients, after tumor removal. These breast tissue specimens have been profiled for their microRNA (miR) expression, in search of differentially expressed miR among patients treated or not with IORT. Our results demonstrate that IORT elicits effects that go beyond the direct killing of residual tumor cells. IORT altered the wound response, inducing the expression of miR-223 in the peri-tumoral breast tissue. miR-223 downregulated the local expression of epidermal growth factor (EGF), leading to decreased activation of EGF receptor (EGFR) on target cells and, eventually, dampening a positive EGF-EGFR autocrine/paracrine stimulation loop induced by the post-surgical wound-healing response. Accordingly, both RT-induced miR-223 and peri-operative inhibition of EGFR efficiently prevented BC cell growth and reduced recurrence formation in mouse models of BC. Our study uncovers unknown effects of RT delivered on a wounded tissue and prompts to the use of anti-EGFR treatments, in a peri-operative treatment schedule, aimed to timely treat BC patients and restrain recurrence formation.
Collapse
|
23
|
Segatto I, Berton S, Sonego M, Massarut S, Perin T, Piccoli E, Colombatti A, Vecchione A, Baldassarre G, Belletti B. Surgery-induced wound response promotes stem-like and tumor-initiating features of breast cancer cells, via STAT3 signaling. Oncotarget 2015; 5:6267-79. [PMID: 25026286 PMCID: PMC4171628 DOI: 10.18632/oncotarget.2195] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Inflammation is clinically linked to cancer but the mechanisms are not fully understood. Surgery itself elicits a range of inflammatory responses, suggesting that it could represent a perturbing factor in the process of local recurrence and/or metastasis formation. Post-surgery wound fluids (WF), drained from breast cancer patients, are rich in cytokines and growth factors, stimulate the in vitro growth of breast cancer cells and are potent activators of the STAT transcription factors. We wondered whether STAT signaling was functionally involved in the response of breast cancer cells to post-surgical inflammation. We discovered that WF induced the enrichment of breast cancer cells with stem-like phenotypes, via activation of STAT3. In vitro, WF highly stimulated mammosphere formation and self-renewal of breast cancer cells. In vivo, STAT3 signaling was critical for breast cancer cell tumorigenicity and for the formation of local relapse after surgery. Overall, we demonstrate here that surgery-induced inflammation promotes stem-like phenotypes and tumor-initiating abilities of breast cancer cells. Interfering with STAT3 signaling with a peri-surgical treatment is sufficient to strongly suppress this process. The understanding of the crosstalk between breast tumor-initiating cells and their microenvironment may open the way to successful targeting of these cells in their initial stages of growth and be eventually curative.
Collapse
Affiliation(s)
- Ilenia Segatto
- Division of Experimental Oncology 2, CRO, National Cancer Institute, Aviano 33081, Italy
| | - Stefania Berton
- Division of Experimental Oncology 2, CRO, National Cancer Institute, Aviano 33081, Italy
| | - Maura Sonego
- Division of Experimental Oncology 2, CRO, National Cancer Institute, Aviano 33081, Italy
| | - Samuele Massarut
- Breast Surgery Unit, CRO, National Cancer Institute, Aviano 33081, Italy
| | - Tiziana Perin
- Pathology Unit, CRO, National Cancer Institute, Aviano 33081, Italy
| | - Erica Piccoli
- Breast Surgery Unit, CRO, National Cancer Institute, Aviano 33081, Italy
| | - Alfonso Colombatti
- Division of Experimental Oncology 2, CRO, National Cancer Institute, Aviano 33081, Italy; Department of Scienze Biologiche e Mediche, MATI Center of Excellence, University of Udine, 33100 Udine, Italy
| | - Andrea Vecchione
- Division of Pathology, II University of Rome "La Sapienza", Santo Andrea Hospital, Rome 00189, Italy
| | - Gustavo Baldassarre
- Division of Experimental Oncology 2, CRO, National Cancer Institute, Aviano 33081, Italy
| | - Barbara Belletti
- Division of Experimental Oncology 2, CRO, National Cancer Institute, Aviano 33081, Italy
| |
Collapse
|
24
|
Propofol promotes cell apoptosis via inhibiting HOTAIR mediated mTOR pathway in cervical cancer. Biochem Biophys Res Commun 2015; 468:561-7. [PMID: 26523512 DOI: 10.1016/j.bbrc.2015.10.129] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 10/24/2015] [Indexed: 12/26/2022]
Abstract
OBJECTIVES Cervical cancer is one of the most common gynecologic malignant tumors. Propofol has been proposed to play a role of antitumor in various cancers. However, the functions and mechanisms of Propofol in cervical cancer is still not clear. METHODS In vitro, the different concentrations of propofol were co-incubated with cervical cancer cell lines, including Hela, Caski and C-33A cells respectively. The pcDNA-HOTAIR plasmid was transfected into cells after the treatment of 10 μg/ml propofol. The cell viability and apoptosis were detected by MTT assay and TUNEL method. In vivo, propofol was injected into mice of transplantation tumor with Caski cells or with pcDNA-HOTAIR treated Caski cells. RESULTS Propofol significantly decreased the cell viability and increased the cell apoptosis in Hela, Caski and C-33A cells, while HOTAIR overexpression promoted cell viability and inhibits cell apoptosis. mTOR/p70S6K protein expression levels were also markedly reduced by propofol but the effects were reversed with pcDNA-HOTAIR. In vivo, propofol inhibited the tumor size but had no inhibition effect in HOTAIR overexpression group. CONCLUSION Propofol inhibited tumor size, cell viability and promoted cell apoptosis via inhibiting mTOR/p70S6K pathway mediated by HOTAIR in cervical cancer.
Collapse
|
25
|
Baldassarre G, Segatto I, Belletti B. Time-tuning cancer therapy. Aging (Albany NY) 2015; 7:531-2. [PMID: 26298846 PMCID: PMC4586098 DOI: 10.18632/aging.100793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Affiliation(s)
- Gustavo Baldassarre
- Division of Experimental Oncology 2, Department of Translational Research, C.R.O. Aviano IRCCS, National Cancer Institute, 33081 Aviano, Italy
| | - Ilenia Segatto
- Division of Experimental Oncology 2, Department of Translational Research, C.R.O. Aviano IRCCS, National Cancer Institute, 33081 Aviano, Italy
| | - Barbara Belletti
- Division of Experimental Oncology 2, Department of Translational Research, C.R.O. Aviano IRCCS, National Cancer Institute, 33081 Aviano, Italy
| |
Collapse
|
26
|
Halacli SO, Dogan AL. FOXP1 regulation via the PI3K/Akt/p70S6K signaling pathway in breast cancer cells. Oncol Lett 2015; 9:1482-1488. [PMID: 25663935 PMCID: PMC4315073 DOI: 10.3892/ol.2015.2885] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 01/07/2015] [Indexed: 02/01/2023] Open
Abstract
Loss of Forkhead box P1 (FOXP1) protein expression confers a poor prognosis in sporadic and familial breast cancer patients, and the FOXP1 gene maps to a tumor suppressor locus at chromosome 3p14. Although correlation studies have indicated that FOXP1 has a role in tumor suppression, determination of the regulatory mechanism of FOXP1 is required to establish its function in breast cancer. It has previously been identified that FOXP1 is regulated by estrogen in breast cancer and that treatment with bisphenol A is effective for regulating the transformation of the normal human breast epithelial cell line, MCF-10F. In addition, FOXO-regulated activation of FOXP1 inhibits the apoptosis of MCF-10F cells following tamoxifen and Akt inhibitor VIII administration. The present study indicates that FOXP1 regulation occurs via a PI3K/Akt/p70S6 kinase (p70S6K) signaling pathway. Following treatment with wortmannin, an inhibitor of phosphatidylinositol 3-kinase (PI3K)/Akt, MCF7 and MDA-MB-231 breast cancer cells demonstrated decreased FOXP1 protein expression levels; this result was also observed in the small interfering (si)RNA silencing of Akt. By contrast, overexpression of Akt resulted in increased FOXP1 protein expression levels in the MDA-MB-231 cells compared with the control cell lysates. Furthermore, treatment with rapamycin, a specific inhibitor of the mammalian target of rapamycin/p70S6K cascade, resulted in decreased FOXP1 expression in the MCF7 cells, but not in the MDA-MB-231 cells, which were resistant to rapamycin-induced inhibition. In addition, silencing of p70S6K using siRNA produced a marked decrease in FOXP1 expression. These data indicate that FOXP1 protein expression is regulated by a PI3K/Akt/p70S6K signaling cascade in breast cancer.
Collapse
Affiliation(s)
- Sevil Oskay Halacli
- Pediatric Immunology Unit, Institute of Children's Health, Hacettepe University, Ankara 06100, Turkey
| | - Ayse Lale Dogan
- Department of Basic Oncology, Institute of Oncology, Hacettepe University, Ankara 06100, Turkey
| |
Collapse
|
27
|
Perspectives of brachytherapy: Patterns of care, new technologies, and “new biology”. Cancer Radiother 2014; 18:434-6. [DOI: 10.1016/j.canrad.2014.07.143] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 07/10/2014] [Accepted: 07/14/2014] [Indexed: 01/20/2023]
|
28
|
Segatto I, Berton S, Sonego M, Massarut S, Fabris L, Armenia J, Mileto M, Colombatti A, Vecchione A, Baldassarre G, Belletti B. p70S6 kinase mediates breast cancer cell survival in response to surgical wound fluid stimulation. Mol Oncol 2014; 8:766-80. [PMID: 24661902 PMCID: PMC5528623 DOI: 10.1016/j.molonc.2014.02.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 02/15/2014] [Indexed: 01/25/2023] Open
Abstract
In early breast cancer, local relapses represent a determinant and not simply an indicator of risk for distant relapse and death. Notably, 90% of local recurrences occur at or close to the same quadrant of the primary cancer. Relevance of PI3K/mTOR/p70S6K signaling in breast tumorigenesis is very well documented. However, the pathway/s involved in the process of breast cancer local relapse are not well understood. The ribosomal protein p70S6K has been implicated in breast cancer cell response to post‐surgical inflammation, supporting the hypothesis that it may be crucial also for breast cancer recurrence. Here, we show that p70S6K activity is required for the survival of breast cancer cells challenged in “hostile” microenvironments. We found that impairment of p70S6K activity in breast cancer cells strongly decreased their tumor take rate in nude mice. In line with this observation, if cells were challenged to grow in anchorage independence or in clonogenic assay, growth of colonies was strongly dependent on an intact p70S6K signaling. This in vitro finding was particularly evident when breast cancer cells were grown in the presence of wound fluids harvested following surgery from breast cancer patients, suggesting that the stimuli present in the post‐surgical setting at least partially relied on activity of p70S6K to stimulate breast cancer relapse. From a mechanistic point of view, our results indicated that p70S6K signaling was able to activate Gli1 and up‐regulate the anti‐apoptotic protein Bcl2, thereby activating a survival response in breast cancer cells challenged in hostile settings. Our work highlights a previously poorly recognized function of p70S6K in preserving breast cancer cell survival, which could eventually be responsible for local relapse and opens the way to the design of new and more specific therapies aiming to restrain the deleterious effects of wound response. p70S6K is activated in breast cancer cells exposed to post‐surgical wound fluids. p70S6K activity is necessary to prevent cell death in “hostile” environments. Inhibition of p70S6K1 leads to different outcomes respect to inhibition of mTOR. The p70S6K/Gli1/Bcl2 signaling axis is necessary to elicit a survival response. p70S6K represents a promising target to prevent breast cancer local recurrence.
Collapse
Affiliation(s)
- Ilenia Segatto
- Division of Experimental Oncology 2, CRO, National Cancer Institute, Aviano 33081, Italy
| | - Stefania Berton
- Division of Experimental Oncology 2, CRO, National Cancer Institute, Aviano 33081, Italy
| | - Maura Sonego
- Division of Experimental Oncology 2, CRO, National Cancer Institute, Aviano 33081, Italy
| | - Samuele Massarut
- Breast Surgery Unit, CRO, National Cancer Institute, Aviano 33081, Italy
| | - Linda Fabris
- Division of Experimental Oncology 2, CRO, National Cancer Institute, Aviano 33081, Italy
| | - Joshua Armenia
- Division of Experimental Oncology 2, CRO, National Cancer Institute, Aviano 33081, Italy
| | - Mario Mileto
- Breast Surgery Unit, CRO, National Cancer Institute, Aviano 33081, Italy
| | - Alfonso Colombatti
- Division of Experimental Oncology 2, CRO, National Cancer Institute, Aviano 33081, Italy; Department of Scienze Biologiche e Mediche, MATI Center of Excellence, University of Udine, 33100 Udine, Italy
| | - Andrea Vecchione
- Division of Pathology, University of Rome "La Sapienza", Sant' Andrea Hospital, Rome 00189, Italy
| | - Gustavo Baldassarre
- Division of Experimental Oncology 2, CRO, National Cancer Institute, Aviano 33081, Italy
| | - Barbara Belletti
- Division of Experimental Oncology 2, CRO, National Cancer Institute, Aviano 33081, Italy.
| |
Collapse
|