1
|
Jones CE, Sharick JT, Sizemore ST, Cukierman E, Strohecker AM, Leight JL. A miniaturized screening platform to identify novel regulators of extracellular matrix alignment. CANCER RESEARCH COMMUNICATIONS 2022; 2:1471-1486. [PMID: 36530465 PMCID: PMC9757767 DOI: 10.1158/2767-9764.crc-22-0157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 08/03/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022]
Abstract
Extracellular matrix alignment contributes to metastasis in a number of cancers and is a known prognostic stromal factor; however, the mechanisms controlling matrix organization remain unclear. Cancer-associated fibroblasts (CAF) play a critical role in this process, particularly via matrix production and modulation of key signaling pathways controlling cell adhesion and contractility. Stroma normalization, as opposed to elimination, is a highly sought strategy, and screening for drugs that effectively alter extracellular matrix (ECM) alignment is a practical way to identify novel CAF-normalizing targets that modulate ECM organization. To meet this need, we developed a novel high-throughput screening platform in which fibroblast-derived matrices were produced in 384-well plates, imaged with automated confocal microscopy, and analyzed using a customized MATLAB script. This platform is a technical advance because it miniaturizes the assay, eliminates costly and time-consuming experimental steps, and streamlines data acquisition and analysis to enable high-throughput screening applications. As a proof of concept, this platform was used to screen a kinase inhibitor library to identify modulators of matrix alignment. A number of novel potential regulators were identified, including several receptor tyrosine kinases (c-MET, tropomyosin receptor kinase 1 (NTRK1), HER2/ERBB2) and the serine/threonine kinases protein kinase A, C, and G (PKA, PKC, and PKG). The expression of these regulators was analyzed in publicly available patient datasets to examine the association between stromal gene expression and patient outcomes.
Collapse
Affiliation(s)
- Caitlin E. Jones
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio
| | - Joe T. Sharick
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio
- The James Comprehensive Cancer Center, Program in Cancer Biology, The Ohio State University, Columbus, Ohio
| | - Steven T. Sizemore
- The James Comprehensive Cancer Center, Program in Cancer Biology, The Ohio State University, Columbus, Ohio
- Department of Radiation Oncology, The Ohio State University, Columbus, Ohio
| | - Edna Cukierman
- Cancer Signaling and Epigenetics, The Marvin and Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Temple Health, Philadelphia, Pennsylvania
| | - Anne Marie Strohecker
- The James Comprehensive Cancer Center, Program in Cancer Biology, The Ohio State University, Columbus, Ohio
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, Ohio
| | - Jennifer L. Leight
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio
- The James Comprehensive Cancer Center, Program in Cancer Biology, The Ohio State University, Columbus, Ohio
| |
Collapse
|
2
|
Capeloa T, Krzystyniak J, d’Hose D, Canas Rodriguez A, Payen VL, Zampieri LX, Van de Velde JA, Benyahia Z, Pranzini E, Vazeille T, Fransolet M, Bouzin C, Brusa D, Michiels C, Gallez B, Murphy MP, Porporato PE, Sonveaux P. MitoQ Inhibits Human Breast Cancer Cell Migration, Invasion and Clonogenicity. Cancers (Basel) 2022; 14:cancers14061516. [PMID: 35326667 PMCID: PMC8946220 DOI: 10.3390/cancers14061516] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/09/2022] [Accepted: 03/10/2022] [Indexed: 01/31/2023] Open
Abstract
To successfully generate distant metastases, metastatic progenitor cells must simultaneously possess mesenchymal characteristics, resist to anoïkis, migrate and invade directionally, resist to redox and shear stresses in the systemic circulation, and possess stem cell characteristics. These cells primarily originate from metabolically hostile areas of the primary tumor, where oxygen and nutrient deprivation, together with metabolic waste accumulation, exert a strong selection pressure promoting evasion. Here, we followed the hypothesis according to which metastasis as a whole implies the existence of metabolic sensors. Among others, mitochondria are singled out as a major source of superoxide that supports the metastatic phenotype. Molecularly, stressed cancer cells increase mitochondrial superoxide production, which activates the transforming growth factor-β pathway through src directly within mitochondria, ultimately activating focal adhesion kinase Pyk2. The existence of mitochondria-targeted antioxidants constitutes an opportunity to interfere with the metastatic process. Here, using aggressive triple-negative and HER2-positive human breast cancer cell lines as models, we report that MitoQ inhibits all the metastatic traits that we tested in vitro. Compared to other mitochondria-targeted antioxidants, MitoQ already successfully passed Phase I safety clinical trials, which provides an important incentive for future preclinical and clinical evaluations of this drug for the prevention of breast cancer metastasis.
Collapse
Affiliation(s)
- Tania Capeloa
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (T.C.); (J.K.); (A.C.R.); (V.L.P.); (L.X.Z.); (J.A.V.d.V.); (Z.B.); (E.P.); (T.V.); (P.E.P.)
| | - Joanna Krzystyniak
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (T.C.); (J.K.); (A.C.R.); (V.L.P.); (L.X.Z.); (J.A.V.d.V.); (Z.B.); (E.P.); (T.V.); (P.E.P.)
| | - Donatienne d’Hose
- Biomedical Magnetic Resonance Unit, Louvain Drug Research Institute (LDRI), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (D.d.); (B.G.)
| | - Amanda Canas Rodriguez
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (T.C.); (J.K.); (A.C.R.); (V.L.P.); (L.X.Z.); (J.A.V.d.V.); (Z.B.); (E.P.); (T.V.); (P.E.P.)
| | - Valery L. Payen
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (T.C.); (J.K.); (A.C.R.); (V.L.P.); (L.X.Z.); (J.A.V.d.V.); (Z.B.); (E.P.); (T.V.); (P.E.P.)
| | - Luca X. Zampieri
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (T.C.); (J.K.); (A.C.R.); (V.L.P.); (L.X.Z.); (J.A.V.d.V.); (Z.B.); (E.P.); (T.V.); (P.E.P.)
| | - Justine A. Van de Velde
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (T.C.); (J.K.); (A.C.R.); (V.L.P.); (L.X.Z.); (J.A.V.d.V.); (Z.B.); (E.P.); (T.V.); (P.E.P.)
| | - Zohra Benyahia
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (T.C.); (J.K.); (A.C.R.); (V.L.P.); (L.X.Z.); (J.A.V.d.V.); (Z.B.); (E.P.); (T.V.); (P.E.P.)
| | - Erica Pranzini
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (T.C.); (J.K.); (A.C.R.); (V.L.P.); (L.X.Z.); (J.A.V.d.V.); (Z.B.); (E.P.); (T.V.); (P.E.P.)
- Department of Experimental and Clinical Biomedical Sciences Mario Serio, University of Florence, Via le Morgagni 50, 50134 Firenze, Italy
| | - Thibaut Vazeille
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (T.C.); (J.K.); (A.C.R.); (V.L.P.); (L.X.Z.); (J.A.V.d.V.); (Z.B.); (E.P.); (T.V.); (P.E.P.)
| | - Maude Fransolet
- Faculty of Sciences, Bology, Laboratoire de Biochimie et Biologie Cellulaire, University of Namur, Rue de Bruxelles 61, 5000 Namur, Belgium; (M.F.); (C.M.)
| | - Caroline Bouzin
- IREC Imaging Platform (2IP), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium;
| | - Davide Brusa
- IREC Flow Cytometry and Cell Sorting Platform, Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium;
| | - Carine Michiels
- Faculty of Sciences, Bology, Laboratoire de Biochimie et Biologie Cellulaire, University of Namur, Rue de Bruxelles 61, 5000 Namur, Belgium; (M.F.); (C.M.)
| | - Bernard Gallez
- Biomedical Magnetic Resonance Unit, Louvain Drug Research Institute (LDRI), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (D.d.); (B.G.)
| | - Michael P. Murphy
- MRC Mitochondrial Biology Unit, Department of Medicine, University of Cambridge, Hills Road, Cambridge CB2 0XY, UK;
| | - Paolo E. Porporato
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (T.C.); (J.K.); (A.C.R.); (V.L.P.); (L.X.Z.); (J.A.V.d.V.); (Z.B.); (E.P.); (T.V.); (P.E.P.)
- Department of Molecular Biotechnology and Health Science, Molecular Biotechnology Center, University of Turin, Via Nizza 52, 10126 Turin, Italy
| | - Pierre Sonveaux
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (T.C.); (J.K.); (A.C.R.); (V.L.P.); (L.X.Z.); (J.A.V.d.V.); (Z.B.); (E.P.); (T.V.); (P.E.P.)
- Correspondence:
| |
Collapse
|
3
|
Capeloa T, Krzystyniak J, Rodriguez AC, Payen VL, Zampieri LX, Pranzini E, Derouane F, Vazeille T, Bouzin C, Duhoux FP, Murphy MP, Porporato PE, Sonveaux P. MitoQ Prevents Human Breast Cancer Recurrence and Lung Metastasis in Mice. Cancers (Basel) 2022; 14:cancers14061488. [PMID: 35326639 PMCID: PMC8946761 DOI: 10.3390/cancers14061488] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 03/08/2022] [Indexed: 02/05/2023] Open
Abstract
Simple Summary Entry in the metastatic phase is often devastating for cancer patients. Metastases originate from metastatic progenitor cells that are selected in the primary tumor and which simultaneously possess several phenotypic capabilities, including migration, invasion, and clonogenicity. We previously provided in vitro evidence that these features are collectively enforced by mitochondrial superoxide in a paradigm where mitochondria act as metabolic sensors of the tumor microenvironment and produce subcytotoxic levels of superoxide to prime metastatic progenitor cells. We also showed that these metastatic traits can be collectively countered by MitoQ, a mitochondria-targeted antioxidant that selectively deactivates mitochondrial superoxide. Here, we further establish that MitoQ prevents primary tumor recurrence after surgery, tumor take and metastasis as a whole, notably in a model of human breast cancer in mice. Since MitoQ already successfully passed Phase I clinical trials, our findings support the development of this drug as a preventive treatment against breast cancer metastasis. Abstract In oncology, the occurrence of distant metastases often marks the transition from curative to palliative care. Such outcome is highly predictable for breast cancer patients, even if tumors are detected early, and there is no specific treatment to prevent metastasis. Previous observations indicated that cancer cell mitochondria are bioenergetic sensors of the tumor microenvironment that produce superoxide to promote evasion. Here, we tested whether mitochondria-targeted antioxidant MitoQ is capable to prevent metastasis in the MDA-MB-231 model of triple-negative human breast cancer in mice and in the MMTV-PyMT model of spontaneously metastatic mouse breast cancer. At clinically relevant doses, we report that MitoQ not only prevented metastatic take and dissemination, but also local recurrence after surgery. We further provide in vitro evidence that MitoQ does not interfere with conventional chemotherapies used to treat breast cancer patients. Since MitoQ already successfully passed Phase I safety clinical trials, our preclinical data collectively provide a strong incentive to test this drug for the prevention of cancer dissemination and relapse in clinical trials with breast cancer patients.
Collapse
Affiliation(s)
- Tania Capeloa
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (T.C.); (J.K.); (A.C.R.); (V.L.P.); (L.X.Z.); (T.V.)
| | - Joanna Krzystyniak
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (T.C.); (J.K.); (A.C.R.); (V.L.P.); (L.X.Z.); (T.V.)
| | - Amanda Canas Rodriguez
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (T.C.); (J.K.); (A.C.R.); (V.L.P.); (L.X.Z.); (T.V.)
| | - Valéry L. Payen
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (T.C.); (J.K.); (A.C.R.); (V.L.P.); (L.X.Z.); (T.V.)
| | - Luca X. Zampieri
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (T.C.); (J.K.); (A.C.R.); (V.L.P.); (L.X.Z.); (T.V.)
| | - Erica Pranzini
- Department of Experimental and Clinical Biomedical Sciences Mario Serio, University of Florence, 50134 Firenze, Italy;
| | - Françoise Derouane
- Pole of Medical Imaging, Radiotherapy and Oncology, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (F.D.); (F.P.D.)
- Department of Medical Oncology, Institut Roi Albert II, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Thibaut Vazeille
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (T.C.); (J.K.); (A.C.R.); (V.L.P.); (L.X.Z.); (T.V.)
| | - Caroline Bouzin
- IREC Imaging Platform (2IP), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium;
| | - François P. Duhoux
- Pole of Medical Imaging, Radiotherapy and Oncology, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (F.D.); (F.P.D.)
- Department of Medical Oncology, Institut Roi Albert II, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Michael P. Murphy
- MRC Mitochondrial Biology Unit, Department of Medicine, University of Cambridge, Cambridge CB2 0XY, UK;
| | - Paolo E. Porporato
- Department of Molecular Biotechnology and Health Science, Molecular Biotechnology Center, University of Turin, 10126 Turin, Italy;
| | - Pierre Sonveaux
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (T.C.); (J.K.); (A.C.R.); (V.L.P.); (L.X.Z.); (T.V.)
- Correspondence:
| |
Collapse
|
4
|
Purrahman D, Mahmoudian-Sani MR, Saki N, Wojdasiewicz P, Kurkowska-Jastrzębska I, Poniatowski ŁA. Involvement of progranulin (PGRN) in the pathogenesis and prognosis of breast cancer. Cytokine 2022; 151:155803. [DOI: 10.1016/j.cyto.2022.155803] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 12/26/2021] [Accepted: 01/09/2022] [Indexed: 12/19/2022]
|
5
|
Kannan A, Philley JV, Hertweck KL, Ndetan H, Singh KP, Sivakumar S, Wells RB, Vadlamudi RK, Dasgupta S. Cancer Testis Antigen Promotes Triple Negative Breast Cancer Metastasis and is Traceable in the Circulating Extracellular Vesicles. Sci Rep 2019; 9:11632. [PMID: 31406142 PMCID: PMC6690992 DOI: 10.1038/s41598-019-48064-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 07/16/2019] [Indexed: 12/15/2022] Open
Abstract
Triple negative breast cancer (TNBC) has poor survival, exhibits rapid metastases, lacks targeted therapies and reliable prognostic markers. Here, we examined metastasis promoting role of cancer testis antigen SPANXB1 in TNBC and its utility as a therapeutic target and prognostic biomarker. Expression pattern of SPANXB1 was determined using matched primary cancer, lymph node metastatic tissues and circulating small extracellular vesicles (sEVs). cDNA microarray analysis of TNBC cells stably integrated with a metastasis suppressor SH3GL2 identified SPANXB1 as a potential target gene. TNBC cells overexpressing SH3GL2 exhibited decreased levels of both SPANXB1 mRNA and protein. Silencing of SPANXB1 reduced migration, invasion and reactive oxygen species production of TNBC cells. SPANXB1 depletion augmented SH3GL2 expression and decreased RAC-1, FAK, A-Actinin and Vinculin expression. Phenotypic and molecular changes were reversed upon SPANXB1 re-expression. SPANXB1 overexpressing breast cancer cells with an enhanced SPANXB1:SH3GL2 ratio achieved pulmonary metastasis within 5 weeks, whereas controls cells failed to do so. Altered expression of SPANXB1 was detected in the sEVs of SPANXB1 transduced cells. Exclusive expression of SPANXB1 was traceable in circulating sEVs, which was associated with TNBC progression. SPANXB1 represents a novel and ideal therapeutic target for blocking TNBC metastases due to its unique expression pattern and may function as an EV based prognostic marker to improve TNBC survival. Uniquely restricted expression of SPANXB1 in TNBCs, makes it an ideal candidate for targeted therapeutics and prognostication.
Collapse
Affiliation(s)
- Anbarasu Kannan
- Departments of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas, USA
| | - Julie V Philley
- Departments of Medicine, The University of Texas Health Science Center at Tyler, Tyler, Texas, USA
| | - Kate L Hertweck
- Departments of Biology, The University of Texas at Tyler, Tyler, Texas, USA.,Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Harrison Ndetan
- Departments of Epidemiology and Biostatistics, The University of Texas Health Science Center at Tyler, Tyler, Texas, USA
| | - Karan P Singh
- Departments of Epidemiology and Biostatistics, The University of Texas Health Science Center at Tyler, Tyler, Texas, USA
| | - Subramaniam Sivakumar
- Departments of Biochemistry, Sri Sankara Arts and Science College, Kanchipuram, India
| | - Robert B Wells
- Departments of Pathology, The University of Texas Health Science Center at Tyler, Tyler, Texas, USA
| | - Ratna K Vadlamudi
- Departments of Obstetrics and Gynecology, CDP program, Mays Cancer Center, University of Texas Health at San Antonio, San Antonio, Texas, USA
| | - Santanu Dasgupta
- Departments of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas, USA. .,Departments of Medicine, The University of Texas Health Science Center at Tyler, Tyler, Texas, USA.
| |
Collapse
|
6
|
Camidge DR, Lee EQ, Lin NU, Margolin K, Ahluwalia MS, Bendszus M, Chang SM, Dancey J, de Vries EGE, Harris GJ, Hodi FS, Lassman AB, Macdonald DR, Peereboom DM, Schiff D, Soffietti R, van den Bent MJ, Wefel JS, Wen PY. Clinical trial design for systemic agents in patients with brain metastases from solid tumours: a guideline by the Response Assessment in Neuro-Oncology Brain Metastases working group. Lancet Oncol 2018; 19:e20-e32. [PMID: 29304358 DOI: 10.1016/s1470-2045(17)30693-9] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 07/24/2017] [Accepted: 08/22/2017] [Indexed: 12/30/2022]
Abstract
Patients with active CNS disease are often excluded from clinical trials, and data regarding the CNS efficacy of systemic agents are usually obtained late in the drug development process or not at all. In this guideline from the Response Assessment in Neuro-Oncology Brain Metastases (RANO-BM) working group, we provide detailed recommendations on when patients with brain metastases from solid tumours should be included or excluded in clinical trials of systemic agents. We also discuss the limitations of retrospective studies in determining the CNS efficacy of systemic drugs. Inclusion of patients with brain metastases early on in the clinical development of a drug or a regimen is needed to generate appropriate CNS efficacy or non-efficacy signals. We consider how to optimally incorporate or exclude such patients in systemic therapy trials depending on the likelihood of CNS activity of the agent by considering three scenarios: drugs that are considered very unlikely to have CNS antitumour activity or efficacy; drugs that are considered very likely to have CNS activity or efficacy; and drugs with minimal baseline information on CNS activity or efficacy. We also address trial design issues unique to patients with brain metastases, including the selection of appropriate CNS endpoints in systemic therapy trials.
Collapse
Affiliation(s)
- D Ross Camidge
- Anschutz Medical Campus, University of Colorado, Aurora, CO, USA.
| | - Eudocia Q Lee
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Nancy U Lin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Kim Margolin
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, CA, USA
| | - Manmeet S Ahluwalia
- Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Solid Tumor Oncology, Cleveland Clinic, Cleveland, OH, USA
| | - Martin Bendszus
- Department of Neuroradiology, University of Heidelberg, Heidelberg, Germany
| | - Susan M Chang
- Department of Neurosurgery, University of California, San Francisco, San Francisco, CA, USA
| | - Janet Dancey
- Department of Oncology, Queen's University, Kingston, ON, Canada
| | - Elisabeth G E de Vries
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Netherlands
| | - Gordon J Harris
- Department of Radiology, 3D Imaging Lab, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - F Stephen Hodi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Department of Medicine, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Andrew B Lassman
- Department of Neurology and Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, Columbia University, NY, USA
| | - David R Macdonald
- Department of Oncology and Department of Clinical Neurological Sciences, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - David M Peereboom
- Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Solid Tumor Oncology, Cleveland Clinic, Cleveland, OH, USA
| | - David Schiff
- Division of Neuro-Oncology, University of Virginia, Charlottesville, VA, USA
| | - Ricardo Soffietti
- Department of Neurology/Neuro-Oncology, University of Turin, Turin, Italy
| | | | - Jeffrey S Wefel
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
7
|
O’Sullivan CC, Davarpanah NN, Abraham J, Bates SE. Current challenges in the management of breast cancer brain metastases. Semin Oncol 2017; 44:85-100. [DOI: 10.1053/j.seminoncol.2017.06.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 06/29/2017] [Indexed: 02/06/2023]
|
8
|
Hu Y, Yu X, Xu G, Liu S. Metastasis: an early event in cancer progression. J Cancer Res Clin Oncol 2016; 143:745-757. [DOI: 10.1007/s00432-016-2279-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 09/22/2016] [Indexed: 01/15/2023]
|
9
|
Winer A, Janosky M, Harrison B, Zhong J, Moussai D, Siyah P, Schatz-Siemers N, Zeng J, Adams S, Mignatti P. Inhibition of Breast Cancer Metastasis by Presurgical Treatment with an Oral Matrix Metalloproteinase Inhibitor: A Preclinical Proof-of-Principle Study. Mol Cancer Ther 2016; 15:2370-2377. [PMID: 27466357 DOI: 10.1158/1535-7163.mct-16-0194] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 07/19/2016] [Indexed: 11/16/2022]
Abstract
Breast cancer has the second highest death toll in women worldwide, despite significant progress in early diagnosis and treatments. The main cause of death is metastatic disease. Matrix metalloproteinases (MMP) are required for the initial steps of metastasis, and have therefore been considered as ideal pharmacologic targets for antimetastatic therapy. However, clinical trials of MMP inhibitors were unsuccessful. These trials were conducted in patients with advanced disease, beyond the stage when these compounds could have been effective. We hypothesized that early treatment with a selective MMP inhibitor between the time of diagnosis and definitive surgery, the so-called "window-of-opportunity," can inhibit metastasis and thereby improve survival. To investigate our hypothesis, we used the 4T1 mouse model of aggressive mammary carcinoma. We treated the animals with SD-7300, an oral inhibitor of MMP-2, -9, and -13, starting after the initial detection of the primary tumor. Seven days later, the primary tumors were excised and analyzed for MMP activity, and the SD-7300 treatment was discontinued. After 4 weeks, the animals were sacrificed and their lungs analyzed histologically for number of metastases and metastatic burden (metastases' area/lung section area). SD-7300 treatment inhibited 70% to 80% of tumor-associated MMP activity (P = 0.0003), reduced metastasis number and metastatic burden by 50% to 60% (P = 0.002 and P = 0.0082, respectively), and increased survival (92% vs. 66.7%; P = 0.0409), relative to control vehicle. These results show that treatment of early invasive breast cancer with selective MMP inhibitors can lower the risk of recurrence and increase long-term disease-free survival. Mol Cancer Ther; 15(10); 2370-7. ©2016 AACR.
Collapse
Affiliation(s)
- Arthur Winer
- Department of Medicine, New York University School of Medicine, New York, New York
| | - Maxwell Janosky
- Department of Medicine, New York University School of Medicine, New York, New York
| | - Beth Harrison
- Department of Pathology, New York University School of Medicine, New York, New York
| | - Judy Zhong
- Department of Population Health, New York University School of Medicine, New York, New York
| | - Dariush Moussai
- Department of Medicine, New York University School of Medicine, New York, New York
| | - Pinar Siyah
- Department of Medicine, New York University School of Medicine, New York, New York
| | - Nina Schatz-Siemers
- Department of Pathology, New York University School of Medicine, New York, New York
| | - Jennifer Zeng
- Department of Pathology, New York University School of Medicine, New York, New York
| | - Sylvia Adams
- Department of Medicine, New York University School of Medicine, New York, New York. Laura and Isaac Perlmutter Cancer Center, New York University School of Medicine, New York, New York
| | - Paolo Mignatti
- Department of Medicine, New York University School of Medicine, New York, New York. Laura and Isaac Perlmutter Cancer Center, New York University School of Medicine, New York, New York. Department of Cell Biology, New York University School of Medicine, New York, New York.
| |
Collapse
|
10
|
Tanaka T, Ikegami Y, Nakazawa H, Kuriyama N, Oki M, Hanai JI, Sukhatme VP, Kaneki M. Low-Dose Farnesyltransferase Inhibitor Suppresses HIF-1α and Snail Expression in Triple-Negative Breast Cancer MDA-MB-231 Cells In Vitro. J Cell Physiol 2016; 232:192-201. [PMID: 27137755 DOI: 10.1002/jcp.25411] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 04/28/2016] [Indexed: 12/13/2022]
Abstract
The aggressiveness of triple-negative breast cancer (TNBC), which lacks estrogen receptor, progesterone receptor and epidermal growth factor receptor 2 (HER2), represents a major challenge in breast cancer. Migratory and self-renewal capabilities are integral components of invasion, metastasis and recurrence of TNBC. Elevated hypoxia-inducible factor-1α (HIF-1α) expression is associated with aggressiveness of cancer. Nonetheless, how HIF-1α expression is regulated and how HIF-1α induces aggressive phenotype are not completely understood in TNBC. The cytotoxic effects of farnesyltransferase (FTase) inhibitors (FTIs) have been studied in cancer and leukemia cells. In contrast, the effect of FTIs on HIF-1α expression has not yet been studied. Here, we show that clinically relevant low-dose FTI, tipifarnib (300 nM), decreased HIF-1α expression, migration and tumorsphere formation in human MDA-MB-231 TNBC cells under a normoxic condition. In contrast, the low-dose FTIs did not inhibit cell growth and activity of the Ras pathway in MDA-MB 231 cells. Tipifarnib-induced decrease in HIF-1α expression was associated with amelioration of the Warburg effect, hypermetabolic state, increases in Snail expression and ATP release, and suppressed E-cadherin expression, major contributors to invasion, metastasis and recurrence of TBNC. These data suggest that FTIs may be capable of ameliorating the aggressive phenotype of TNBC by suppressing the HIF-1α-Snail pathway. J. Cell. Physiol. 232: 192-201, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Tomokazu Tanaka
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts
| | - Yuichi Ikegami
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts
| | - Harumasa Nakazawa
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts.,Shriners Hospitals for Children, Boston, Massachusetts
| | - Naohide Kuriyama
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts.,Shriners Hospitals for Children, Boston, Massachusetts
| | - Miwa Oki
- Division of Nephrology, Division of Interdisciplinary Medicine and Biotechnology, Department of Medicine, Beth Israel Deaconess Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Jun-Ichi Hanai
- Division of Nephrology, Division of Interdisciplinary Medicine and Biotechnology, Department of Medicine, Beth Israel Deaconess Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Vikas P Sukhatme
- Division of Nephrology, Division of Interdisciplinary Medicine and Biotechnology, Department of Medicine, Beth Israel Deaconess Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Masao Kaneki
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts. .,Shriners Hospitals for Children, Boston, Massachusetts.
| |
Collapse
|
11
|
Pinedo-Carpio E, Davidson D, Martinez Marignac VL, Panasci J, Aloyz R. Adaptive metabolic rewiring to chronic SFK inhibition. Oncotarget 2016; 8:66758-66768. [PMID: 28977994 PMCID: PMC5620134 DOI: 10.18632/oncotarget.8146] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 03/02/2016] [Indexed: 12/21/2022] Open
Abstract
Src family kinases (SFK) are key regulators of cellular proliferation, differentiation, survival, motility and angiogenesis. As such, SFK inhibitors are being tested in clinical trials to prevent metastasis as an alternative to current treatment regimens for a variety of cancers including breast cancer. To contribute to the development of molecular tools improving SFK-targeted therapies, we used the SFK inhibitor dasatinib and a well characterized triple negative breast cancer cell line (BT20). Comparison of the response of BT20 cells with acquired resistance to dasatinib and its’ parental counterpart suggest that chronic exposure to SFK inhibition results in increased dependency on TGFβ signaling for proliferation, both in the absence or the presence of dasatinib. In addition, we found that acquired (but not de novo) resistance to dasatinib was reduced by non-cytotoxic concentrations compounds hindering on PI3K, mTORC1 signaling, endoplasmic reticulum stress or autophagy.
Collapse
Affiliation(s)
- Edgar Pinedo-Carpio
- Jewish General Hospital, Lady Davis Institute & McGill University, Faculty of Medicine, Division of Experimental Medicine & Department of Oncology, Montréal, Québec H3T 1E2, Canada
| | - David Davidson
- Jewish General Hospital, Lady Davis Institute & McGill University, Faculty of Medicine, Division of Experimental Medicine & Department of Oncology, Montréal, Québec H3T 1E2, Canada
| | | | - Justin Panasci
- Jewish General Hospital, Lady Davis Institute & McGill University, Faculty of Medicine, Division of Experimental Medicine & Department of Oncology, Montréal, Québec H3T 1E2, Canada
| | - Raquel Aloyz
- Jewish General Hospital, Lady Davis Institute & McGill University, Faculty of Medicine, Division of Experimental Medicine & Department of Oncology, Montréal, Québec H3T 1E2, Canada
| |
Collapse
|
12
|
Entschladen F, Thyssen DA, Drell DW. Re-Use of Established Drugs for Anti-Metastatic Indications. Cells 2016; 5:cells5010002. [PMID: 26771645 PMCID: PMC4810087 DOI: 10.3390/cells5010002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Revised: 01/04/2016] [Accepted: 01/08/2016] [Indexed: 12/28/2022] Open
Abstract
Most patients that die from cancer do not die due to the primary tumor but due to the development of metastases. However, there is currently still no drug on the market that specifically addresses and inhibits metastasis formation. This lack was, in the past, largely due to the lack of appropriate screening models, but recent developments have established such models and have provided evidence that tumor cell migration works as a surrogate for metastasis formation. Herein we deliver on several examples a rationale for not only testing novel cancer drugs by use of these screening assays, but also reconsider established drugs even of other fields of indication.
Collapse
Affiliation(s)
- Frank Entschladen
- MetaVì Labs Inc., 16238 Ranch Road 620 North, Suite F-347, Austin, TX 78717, USA.
- Faculty of Health-School of Medicine, Witten/Herdecke University, Alfred-Herrhausen-Straße 50, 58448 Witten, Germany.
| | - Dane A Thyssen
- MetaVì Labs Inc., 16238 Ranch Road 620 North, Suite F-347, Austin, TX 78717, USA.
| | - David W Drell
- MetaVì Labs Inc., 16238 Ranch Road 620 North, Suite F-347, Austin, TX 78717, USA.
| |
Collapse
|
13
|
Chambers AF, Werb Z. Invasion and metastasis--recent advances and future challenges. J Mol Med (Berl) 2015; 93:361-8. [PMID: 25772709 DOI: 10.1007/s00109-015-1269-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 02/19/2015] [Indexed: 12/12/2022]
Affiliation(s)
- Ann F Chambers
- London Regional Cancer Program, 790 Commissioners Road East, London, Ontario, N6A 4L6, Canada,
| | | |
Collapse
|