1
|
Espinosa-Sánchez A, Suárez-Martínez E, Sánchez-Díaz L, Carnero A. Therapeutic Targeting of Signaling Pathways Related to Cancer Stemness. Front Oncol 2020; 10:1533. [PMID: 32984007 PMCID: PMC7479251 DOI: 10.3389/fonc.2020.01533] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 07/16/2020] [Indexed: 12/11/2022] Open
Abstract
The theory of cancer stem cells (CSCs) proposes that the different cells within a tumor, as well as metastasis deriving from it, are originated from a single subpopulation of cells with self-renewal and differentiation capacities. These cancer stem cells are supposed to be critical for tumor expansion and metastasis, tumor relapse and resistance to conventional therapies, such as chemo- and radiotherapy. The acquisition of these abilities has been attributed to the activation of alternative pathways, for instance, WNT, NOTCH, SHH, PI3K, Hippo, or NF-κB pathways, that regulate detoxification mechanisms; increase the metabolic rate; induce resistance to apoptotic, autophagic, and senescence pathways; promote the overexpression of drug transporter proteins; and activate specific stem cell transcription factors. The elimination of CSCs is an important goal in cancer therapeutic approaches because it could decrease relapses and metastatic dissemination, which are main causes of mortality in oncology patients. In this work, we discuss the role of these signaling pathways in CSCs along with their therapeutic potential.
Collapse
Affiliation(s)
- Asunción Espinosa-Sánchez
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Seville, Spain
- CIBER de Cancer, Madrid, Spain
| | - Elisa Suárez-Martínez
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Seville, Spain
- CIBER de Cancer, Madrid, Spain
| | - Laura Sánchez-Díaz
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Seville, Spain
- CIBER de Cancer, Madrid, Spain
| | - Amancio Carnero
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Seville, Spain
- CIBER de Cancer, Madrid, Spain
| |
Collapse
|
2
|
Novitskiy SV, Zaynagetdinov R, Vasiukov G, Gutor S, Han W, Serezani A, Matafonov A, Gleaves LA, Sherrill TP, Polosukhin VV, Blackwell TS. Gas6/MerTK signaling is negatively regulated by NF-κB and supports lung carcinogenesis. Oncotarget 2019; 10:7031-7042. [PMID: 31903163 PMCID: PMC6925028 DOI: 10.18632/oncotarget.27345] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 11/07/2019] [Indexed: 01/24/2023] Open
Abstract
Growth arrest-specific 6 (Gas6) has been implicated in carcinogenesis through activation of its receptors, particularly MerTK. To investigate whether Gas6 plays a role in resistance to NF-κB inhibitors, which have not proven to be effective agents for lung cancer therapy, we studied lung cancer models induced by urethane injection or expression of mutant Kras (KrasG12D). We found that Gas6 is primarily produced by macrophages during tumorigenesis and that Gas6 is negatively regulated by NF-κB. Since Gas6 is a vitamin K dependent protein, we used low-dose warfarin to block Gas6 production and showed that this treatment inhibited tumorigenesis in both the urethane and KrasG12D models, most prominently in mice with targeted deletion of IKKβ in myeloid cells (IKKβΔMye mice). In addition, MerTK deficient mice had reduced urethane-induced tumorigenesis. Inhibition of the Gas6-MerTK pathway in all these models reduced macrophages and neutrophils in the lungs of tumor-bearing mice. Analysis of mouse lung tumors revealed MerTK staining on tumor cells and in vitro studies showed that Gas6 increased proliferation of human lung cancer cell lines. To assess the therapeutic potential for combination treatment targeting NF-κB and Gas6-MerTK, we injected Lewis Lung Carcinoma cells subcutaneously and treated mice with Bay 11-70852 (NF-κB inhibitor) and/or Foretinib (MerTK inhibitor). While individual treatments were ineffective, combination therapy markedly reduced tumor growth, blocked tumor cell proliferation, reduced tumor-associated macrophages, and increased CD4+ T cells. Together, our studies unmask a role for Gas6-MerTK signaling in lung carcinogenesis and indicate that up-regulation of Gas6 production in macrophages could be a major mechanism of resistance to NF-κB inhibitors.
Collapse
Affiliation(s)
- Sergey V Novitskiy
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University, Nashville, TN 37212, USA
| | - Rinat Zaynagetdinov
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University, Nashville, TN 37212, USA
| | - Georgii Vasiukov
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University, Nashville, TN 37212, USA
| | - Sergey Gutor
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University, Nashville, TN 37212, USA
| | - Wei Han
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University, Nashville, TN 37212, USA
| | - Ana Serezani
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University, Nashville, TN 37212, USA
| | - Anton Matafonov
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN 37212, USA
| | - Linda A Gleaves
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University, Nashville, TN 37212, USA
| | - Taylor P Sherrill
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University, Nashville, TN 37212, USA
| | - Vasiliy V Polosukhin
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University, Nashville, TN 37212, USA
| | - Timothy S Blackwell
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University, Nashville, TN 37212, USA.,Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN 37212, USA.,Department of Veterans Affairs Medical Center, Nashville, TN 37212, USA
| |
Collapse
|
3
|
Vreka M, Lilis I, Papageorgopoulou M, Giotopoulou GA, Lianou M, Giopanou I, Kanellakis NI, Spella M, Agalioti T, Armenis V, Goldmann T, Marwitz S, Yull FE, Blackwell TS, Pasparakis M, Marazioti A, Stathopoulos GT. IκB Kinase α Is Required for Development and Progression of KRAS-Mutant Lung Adenocarcinoma. Cancer Res 2018; 78:2939-2951. [PMID: 29588349 PMCID: PMC6485619 DOI: 10.1158/0008-5472.can-17-1944] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 10/03/2017] [Accepted: 03/22/2018] [Indexed: 01/02/2023]
Abstract
Although oncogenic activation of NFκB has been identified in various tumors, the NFκB-activating kinases (inhibitor of NFκB kinases, IKK) responsible for this are elusive. In this study, we determined the role of IKKα and IKKβ in KRAS-mutant lung adenocarcinomas induced by the carcinogen urethane and by respiratory epithelial expression of oncogenic KRASG12D Using NFκB reporter mice and conditional deletions of IKKα and IKKβ, we identified two distinct early and late activation phases of NFκB during chemical and genetic lung adenocarcinoma development, which were characterized by nuclear translocation of RelB, IκBβ, and IKKα in tumor-initiated cells. IKKα was a cardinal tumor promoter in chemical and genetic KRAS-mutant lung adenocarcinoma, and respiratory epithelial IKKα-deficient mice were markedly protected from the disease. IKKα specifically cooperated with mutant KRAS for tumor induction in a cell-autonomous fashion, providing mutant cells with a survival advantage in vitro and in vivo IKKα was highly expressed in human lung adenocarcinoma, and a heat shock protein 90 inhibitor that blocks IKK function delivered superior effects against KRAS-mutant lung adenocarcinoma compared with a specific IKKβ inhibitor. These results demonstrate an actionable requirement for IKKα in KRAS-mutant lung adenocarcinoma, marking the kinase as a therapeutic target against this disease.Significance: These findings report a novel requirement for IKKα in mutant KRAS lung tumor formation, with potential therapeutic applications. Cancer Res; 78(11); 2939-51. ©2018 AACR.
Collapse
Affiliation(s)
- Malamati Vreka
- Laboratory for Molecular Respiratory Carcinogenesis, Department of Physiology, Faculty of Medicine, University of Patras, Rio, Greece
- Comprehensive Pneumology Center (CPC) and Institute for Lung Biology and Disease (iLBD), University Hospital, Ludwig-Maximilians University and Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Ioannis Lilis
- Laboratory for Molecular Respiratory Carcinogenesis, Department of Physiology, Faculty of Medicine, University of Patras, Rio, Greece
| | - Maria Papageorgopoulou
- Laboratory for Molecular Respiratory Carcinogenesis, Department of Physiology, Faculty of Medicine, University of Patras, Rio, Greece
- Comprehensive Pneumology Center (CPC) and Institute for Lung Biology and Disease (iLBD), University Hospital, Ludwig-Maximilians University and Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Georgia A Giotopoulou
- Laboratory for Molecular Respiratory Carcinogenesis, Department of Physiology, Faculty of Medicine, University of Patras, Rio, Greece
- Comprehensive Pneumology Center (CPC) and Institute for Lung Biology and Disease (iLBD), University Hospital, Ludwig-Maximilians University and Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Marina Lianou
- Laboratory for Molecular Respiratory Carcinogenesis, Department of Physiology, Faculty of Medicine, University of Patras, Rio, Greece
| | - Ioanna Giopanou
- Laboratory for Molecular Respiratory Carcinogenesis, Department of Physiology, Faculty of Medicine, University of Patras, Rio, Greece
| | - Nikolaos I Kanellakis
- Laboratory for Molecular Respiratory Carcinogenesis, Department of Physiology, Faculty of Medicine, University of Patras, Rio, Greece
| | - Magda Spella
- Laboratory for Molecular Respiratory Carcinogenesis, Department of Physiology, Faculty of Medicine, University of Patras, Rio, Greece
| | - Theodora Agalioti
- Laboratory for Molecular Respiratory Carcinogenesis, Department of Physiology, Faculty of Medicine, University of Patras, Rio, Greece
| | - Vasileios Armenis
- Laboratory for Molecular Respiratory Carcinogenesis, Department of Physiology, Faculty of Medicine, University of Patras, Rio, Greece
| | - Torsten Goldmann
- Clinical and Experimental Pathology, Research Center Borstel, Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Borstel, Germany
| | - Sebastian Marwitz
- Clinical and Experimental Pathology, Research Center Borstel, Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Borstel, Germany
| | - Fiona E Yull
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Timothy S Blackwell
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Manolis Pasparakis
- Mouse Genetics and Inflammation Laboratory, Institute for Genetics, University of Cologne, Cologne, Germany
| | - Antonia Marazioti
- Laboratory for Molecular Respiratory Carcinogenesis, Department of Physiology, Faculty of Medicine, University of Patras, Rio, Greece.
| | - Georgios T Stathopoulos
- Laboratory for Molecular Respiratory Carcinogenesis, Department of Physiology, Faculty of Medicine, University of Patras, Rio, Greece.
- Comprehensive Pneumology Center (CPC) and Institute for Lung Biology and Disease (iLBD), University Hospital, Ludwig-Maximilians University and Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| |
Collapse
|
4
|
Marazioti A, Lilis I, Vreka M, Apostolopoulou H, Kalogeropoulou A, Giopanou I, Giotopoulou GA, Krontira AC, Iliopoulou M, Kanellakis NI, Agalioti T, Giannou AD, Jones-Paris C, Iwakura Y, Kardamakis D, Blackwell TS, Taraviras S, Spella M, Stathopoulos GT. Myeloid-derived interleukin-1β drives oncogenic KRAS-NF-κΒ addiction in malignant pleural effusion. Nat Commun 2018; 9:672. [PMID: 29445180 PMCID: PMC5813197 DOI: 10.1038/s41467-018-03051-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Accepted: 01/15/2018] [Indexed: 12/17/2022] Open
Abstract
Malignant pleural effusion (MPE) is a frequent metastatic manifestation of human cancers. While we previously identified KRAS mutations as molecular culprits of MPE formation, the underlying mechanism remained unknown. Here, we determine that non-canonical IKKα-RelB pathway activation of KRAS-mutant tumor cells mediates MPE development and this is fueled by host-provided interleukin IL-1β. Indeed, IKKα is required for the MPE-competence of KRAS-mutant tumor cells by activating non-canonical NF-κB signaling. IL-1β fuels addiction of mutant KRAS to IKKα resulting in increased CXCL1 secretion that fosters MPE-associated inflammation. Importantly, IL-1β-mediated NF-κB induction in KRAS-mutant tumor cells, as well as their resulting MPE-competence, can only be blocked by co-inhibition of both KRAS and IKKα, a strategy that overcomes drug resistance to individual treatments. Hence we show that mutant KRAS facilitates IKKα-mediated responsiveness of tumor cells to host IL-1β, thereby establishing a host-to-tumor signaling circuit that culminates in inflammatory MPE development and drug resistance.
Collapse
Affiliation(s)
- Antonia Marazioti
- Department of Physiology, Laboratory for Molecular Respiratory Carcinogenesis, Faculty of Medicine, University of Patras, 26504, Rio, Achaia, Greece.
| | - Ioannis Lilis
- Department of Physiology, Laboratory for Molecular Respiratory Carcinogenesis, Faculty of Medicine, University of Patras, 26504, Rio, Achaia, Greece
| | - Malamati Vreka
- Department of Physiology, Laboratory for Molecular Respiratory Carcinogenesis, Faculty of Medicine, University of Patras, 26504, Rio, Achaia, Greece
- Comprehensive Pneumology Center (CPC) and Institute for Lung Biology and Disease (iLBD), University Hospital, Ludwig-Maximilians University and Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), 81377, Munich, Bavaria, Germany
| | - Hara Apostolopoulou
- Department of Physiology, Laboratory for Molecular Respiratory Carcinogenesis, Faculty of Medicine, University of Patras, 26504, Rio, Achaia, Greece
| | - Argyro Kalogeropoulou
- Stem Cell Biology Laboratory, Department of Physiology, Faculty of Medicine, University of Patras, 26504, Rio, Achaia, Greece
| | - Ioanna Giopanou
- Department of Physiology, Laboratory for Molecular Respiratory Carcinogenesis, Faculty of Medicine, University of Patras, 26504, Rio, Achaia, Greece
| | - Georgia A Giotopoulou
- Department of Physiology, Laboratory for Molecular Respiratory Carcinogenesis, Faculty of Medicine, University of Patras, 26504, Rio, Achaia, Greece
| | - Anthi C Krontira
- Department of Physiology, Laboratory for Molecular Respiratory Carcinogenesis, Faculty of Medicine, University of Patras, 26504, Rio, Achaia, Greece
| | - Marianthi Iliopoulou
- Department of Physiology, Laboratory for Molecular Respiratory Carcinogenesis, Faculty of Medicine, University of Patras, 26504, Rio, Achaia, Greece
| | - Nikolaos I Kanellakis
- Department of Physiology, Laboratory for Molecular Respiratory Carcinogenesis, Faculty of Medicine, University of Patras, 26504, Rio, Achaia, Greece
| | - Theodora Agalioti
- Department of Physiology, Laboratory for Molecular Respiratory Carcinogenesis, Faculty of Medicine, University of Patras, 26504, Rio, Achaia, Greece
| | - Anastasios D Giannou
- Department of Physiology, Laboratory for Molecular Respiratory Carcinogenesis, Faculty of Medicine, University of Patras, 26504, Rio, Achaia, Greece
| | - Celestial Jones-Paris
- Division of Allergy, Pulmonary and Critical Care, Department of Internal Medicine, Vanderbilt University School of Medicine, T-1218 MCN, Nashville, TN, 37232-2650, USA
| | - Yoichiro Iwakura
- Research Institute for Biomedical Sciences, Tokyo University of Science, Tokyo, Chiba, 278-0022, Japan
| | - Dimitrios Kardamakis
- Department of Radiation Oncology and Stereotactic Radiotherapy, Faculty of Medicine, University of Patras, 26504, Rio, Achaia, Greece
| | - Timothy S Blackwell
- Division of Allergy, Pulmonary and Critical Care, Department of Internal Medicine, Vanderbilt University School of Medicine, T-1218 MCN, Nashville, TN, 37232-2650, USA
| | - Stavros Taraviras
- Stem Cell Biology Laboratory, Department of Physiology, Faculty of Medicine, University of Patras, 26504, Rio, Achaia, Greece
| | - Magda Spella
- Department of Physiology, Laboratory for Molecular Respiratory Carcinogenesis, Faculty of Medicine, University of Patras, 26504, Rio, Achaia, Greece
| | - Georgios T Stathopoulos
- Department of Physiology, Laboratory for Molecular Respiratory Carcinogenesis, Faculty of Medicine, University of Patras, 26504, Rio, Achaia, Greece.
- Comprehensive Pneumology Center (CPC) and Institute for Lung Biology and Disease (iLBD), University Hospital, Ludwig-Maximilians University and Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), 81377, Munich, Bavaria, Germany.
| |
Collapse
|
5
|
Hsu FT, Liu HS, Ali AAA, Tsai PH, Kao YC, Lu CF, Huang HS, Chen CY. Assessing the selective therapeutic efficacy of superparamagnetic erlotinib nanoparticles in lung cancer by using quantitative magnetic resonance imaging and a nuclear factor kappa-B reporter gene system. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 14:1019-1031. [PMID: 29391212 DOI: 10.1016/j.nano.2018.01.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 01/03/2018] [Accepted: 01/15/2018] [Indexed: 01/11/2023]
Abstract
Non-small-cell lung cancer (NSCLC) is the most common type of lung cancer. Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors are commonly used as the first-line treatment for advanced NSCLC; however, the efficacy of drug delivery remains unknown. Hence, we successfully developed erlotinib-conjugated iron oxide nanoparticles (FeDC-E NPs) as theranostic probe that can potentially provide a new avenue for monitoring drug delivering through noninvasive magnetic resonance imaging. MRI ΔR2* relaxivity measurements offer an opportunity to quantitatively evaluate the uptake of FeDC-E NPs at cellular and tumoral levels. Additionally, NF-κB reporter gene system provides NF-κB activation status monitoring to validate the therapeutic efficiency of FeDC-E NPs. FeDC-E NPs not only inhibit the tumor growth and NF-κB-modulated antiapoptotic mechanism but also trigger extrinsic and intrinsic apoptotic pathways. Taken together, dual functional FeDC-E NPs offer diagnostic and therapeutic benefits against lung cancers, indicating that our presented probe could be applied in clinical.
Collapse
Affiliation(s)
- Fei-Ting Hsu
- Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Medical Imaging, Taipei Medical University Hospital, Taipei, Taiwan; Research Center of Translational Imaging, College of Medicine, Taipei Medical University, Taipei, Taiwan; Translational Laboratory, Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan
| | - Hua-Shan Liu
- School of Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan; International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Ahmed Atef Ahmed Ali
- Research Center of Translational Imaging, College of Medicine, Taipei Medical University, Taipei, Taiwan; Translational Laboratory, Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan; School of Pharmacy, National Defense Medical Center, Taipei, Taiwan
| | - Ping-Huei Tsai
- Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Medical Imaging, Taipei Medical University Hospital, Taipei, Taiwan; Research Center of Translational Imaging, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yu-Chieh Kao
- Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Research Center of Translational Imaging, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chia-Feng Lu
- Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Research Center of Translational Imaging, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hsu-Shan Huang
- School of Pharmacy, National Defense Medical Center, Taipei, Taiwan; Graduate Institute for Cancer Biology & Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taiwan
| | - Cheng-Yu Chen
- Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Medical Imaging, Taipei Medical University Hospital, Taipei, Taiwan; Research Center of Translational Imaging, College of Medicine, Taipei Medical University, Taipei, Taiwan; Translational Laboratory, Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan.
| |
Collapse
|
6
|
Friedman AA, Xia Y, Trippa L, Le LP, Igras V, Frederick DT, Wargo JA, Tanabe KK, Lawrence DP, Neuberg DS, Flaherty KT, Fisher DE. Feasibility of Ultra-High-Throughput Functional Screening of Melanoma Biopsies for Discovery of Novel Cancer Drug Combinations. Clin Cancer Res 2017; 23:4680-4692. [PMID: 28446504 DOI: 10.1158/1078-0432.ccr-16-3029] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 02/22/2017] [Accepted: 04/21/2017] [Indexed: 12/15/2022]
Abstract
Purpose: Successful development of targeted therapy combinations for cancer patients depends on first discovering such combinations in predictive preclinical models. Stable cell lines and mouse xenograft models can have genetic and phenotypic drift and may take too long to generate to be useful as a personalized medicine tool.Experimental Design: To overcome these limitations, we have used a platform of ultra-high-throughput functional screening of primary biopsies preserving both cancer and stroma cell populations from melanoma patients to nominate such novel combinations from a library of thousands of drug combinations in a patient-specific manner within days of biopsy. In parallel, patient-derived xenograft (PDX) mouse models were created and novel combinations tested for their ability to shrink matched PDXs.Results: The screening method identifies specific drug combinations in tumor cells with patterns that are distinct from those obtained from stable cell lines. Screening results were highly specific to individual patients. For patients with matched PDX models, we confirmed that individualized novel targeted therapy combinations could inhibit tumor growth. In particular, a combination of multi-kinase and PI3K/Akt inhibitors was effective in some BRAF-wild-type melanomas, and the addition of cediranib to the BRAF inhibitor PLX4720 was effective in a PDX model with BRAF mutation.Conclusions: This proof-of-concept study demonstrates the feasibility of using primary biopsies directly for combinatorial drug discovery, complementing stable cell lines and xenografts, but with much greater speed and efficiency. This process could potentially be used in a clinical setting to rapidly identify therapeutic strategies for individual patients. Clin Cancer Res; 23(16); 4680-92. ©2017 AACR.
Collapse
Affiliation(s)
- Adam A Friedman
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts.,Dermatology and Cutaneous Biology Research Center, Massachusetts General Hospital, Charlestown, Massachusetts
| | - Yun Xia
- Dermatology and Cutaneous Biology Research Center, Massachusetts General Hospital, Charlestown, Massachusetts.,Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lorenzo Trippa
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Long Phi Le
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Vivien Igras
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts.,Dermatology and Cutaneous Biology Research Center, Massachusetts General Hospital, Charlestown, Massachusetts
| | - Dennie T Frederick
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Jennifer A Wargo
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts.,Division of Surgical Oncology, Massachusetts General Hospital, Charlestown, Massachusetts
| | - Kenneth K Tanabe
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts.,Division of Surgical Oncology, Massachusetts General Hospital, Charlestown, Massachusetts
| | - Donald P Lawrence
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Donna S Neuberg
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Keith T Flaherty
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - David E Fisher
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts. .,Dermatology and Cutaneous Biology Research Center, Massachusetts General Hospital, Charlestown, Massachusetts
| |
Collapse
|
7
|
Krem MM, Yan J. To b(ortezomib) or not to be: the stroma's the thing. J Pathol 2016; 240:123-5. [PMID: 27340009 PMCID: PMC5037003 DOI: 10.1002/path.4763] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 06/13/2016] [Accepted: 06/20/2016] [Indexed: 11/11/2022]
Abstract
The proteasome inhibitor bortezomib has been widely used to treat patients with multiple myeloma (MM). However, some patients show primary or secondary resistance. In recent work published in The Journal of Pathology, Beyar-Katz et al demonstrate that bortezomib treatment stimulates a host inflammatory response, which in turn promotes MM cell migration, viability, and proliferation. These effects appear to be mediated by pro-inflammatory M1-like stromal macrophages partly via secretion of cytokine IL-16. These unexpected findings imply that the binary M1/M2 definition of macrophages may not accurately describe the complexity and heterogeneity of macrophages associated with MM tumour growth and progression, and further suggest that bortezomib treatment stimulates host-driven tumour-promoting activity in addition to its cytotoxic activity, thus leading to potential bortezomib resistance in MM patients. Understanding the underlying mechanisms may identify novel targets to overcome or prevent bortezomib resistance. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Maxwell M Krem
- Division of Blood and Marrow Transplantation, Department of Medicine, James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY, USA
| | - Jun Yan
- Division of Blood and Marrow Transplantation, Department of Medicine, James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY, USA.
- Division of Hematology/Oncology, Department of Medicine, James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY, USA.
| |
Collapse
|
8
|
McLoed AG, Sherrill TP, Cheng DS, Han W, Saxon JA, Gleaves LA, Wu P, Polosukhin VV, Karin M, Yull FE, Stathopoulos GT, Georgoulias V, Zaynagetdinov R, Blackwell TS. Neutrophil-Derived IL-1β Impairs the Efficacy of NF-κB Inhibitors against Lung Cancer. Cell Rep 2016; 16:120-132. [PMID: 27320908 DOI: 10.1016/j.celrep.2016.05.085] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 04/26/2016] [Accepted: 05/19/2016] [Indexed: 01/03/2023] Open
Abstract
Although epithelial NF-κB signaling is important for lung carcinogenesis, NF-κB inhibitors are ineffective for cancer treatment. To explain this paradox, we studied mice with genetic deletion of IKKβ in myeloid cells and found enhanced tumorigenesis in Kras(G12D) and urethane models of lung cancer. Myeloid-specific inhibition of NF-κB augmented pro-IL-1β processing by cathepsin G in neutrophils, leading to increased IL-1β and enhanced epithelial cell proliferation. Combined treatment with bortezomib, a proteasome inhibitor that blocks NF-κB activation, and IL-1 receptor antagonist reduced tumor formation and growth in vivo. In lung cancer patients, plasma IL-1β levels correlated with poor prognosis, and IL-1β increased following bortezomib treatment. Together, our studies elucidate an important role for neutrophils and IL-1β in lung carcinogenesis and resistance to NF-κB inhibitors.
Collapse
Affiliation(s)
- Allyson G McLoed
- Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Taylor P Sherrill
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University, Nashville, TN 37232, USA
| | - Dong-Sheng Cheng
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University, Nashville, TN 37232, USA
| | - Wei Han
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University, Nashville, TN 37232, USA
| | - Jamie A Saxon
- Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Linda A Gleaves
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University, Nashville, TN 37232, USA
| | - Pingsheng Wu
- Department of Biostatistics, Vanderbilt University, Nashville, TN 37232, USA
| | - Vasiliy V Polosukhin
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University, Nashville, TN 37232, USA
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Fiona E Yull
- Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt-Ingram Cancer Center, 691 Preston Building, 2220 Pierce Ave., Nashville, TN 37232, USA
| | - Georgios T Stathopoulos
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University, Nashville, TN 37232, USA; Laboratory for Molecular Respiratory Carcinogenesis, Department of Physiology, University of Patras, Rio, 26504 Patras, Greece; Comprehensive Pneumology Center (CPC), University Hospital, Ludwig-Maximilians University and Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich 81377, Germany
| | - Vassilis Georgoulias
- Department of Medical Oncology, University General Hospital of Heraklion, Heraklion, Crete 71110, Greece
| | - Rinat Zaynagetdinov
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University, Nashville, TN 37232, USA.
| | - Timothy S Blackwell
- Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA; Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt-Ingram Cancer Center, 691 Preston Building, 2220 Pierce Ave., Nashville, TN 37232, USA; U.S. Department of Veterans Affairs, Washington, DC 20420, USA; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232 USA
| |
Collapse
|
9
|
Alexander CM, Xiong KN, Velmurugan K, Xiong J, Osgood RS, Bauer AK. Differential innate immune cell signatures and effects regulated by toll-like receptor 4 during murine lung tumor promotion. Exp Lung Res 2016; 42:154-73. [PMID: 27093379 PMCID: PMC5506691 DOI: 10.3109/01902148.2016.1164263] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Tumor promotion is an early and critical stage during lung adenocarcinoma (ADC). We previously demonstrated that Tlr4 mutant mice were more susceptible to butylated hydroxytoluene (BHT)-induced pulmonary inflammation and tumor promotion in comparison to Tlr4-sufficient mice. Our study objective was to elucidate the underlying differences in Tlr4 mutant mice in innate immune cell populations, their functional responses, and the influence of these cellular differences on ADC progenitor (type II) cells following BHT-treatment. BALB (Tlr4-sufficient) and C.C3-Tlr4(Lps-d)/J (BALB(Lpsd); Tlr4 mutant) mice were treated with BHT (promoter) followed by bronchoalveolar lavage (BAL) and flow cytometry processing on the lungs. ELISAs, Club cell enrichment, macrophage function, and RNA isolation were also performed. Bone marrow-derived macrophages (BMDM) co-cultured with a type II cell line were used for wound healing assays. Innate immune cells significantly increased in whole lung in BHT-treated BALB(Lpsd) mice compared to BALB mice. BHT-treated BALB(Lpsd) mice demonstrated enhanced macrophage functionality, increased epithelial wound closure via BMDMs, and increased Club cell number in BALB(Lpsd) mice, all compared to BALB BHT-treated mice. Cytokine/chemokine (Kc, Mcp1) and growth factor (Igf1) levels also significantly differed among the strains and within macrophages, gene expression, and cell surface markers collectively demonstrated a more plastic phenotype in BALB(Lpsd) mice. Therefore, these correlative studies suggest that distinct innate immune cell populations are associated with the differences observed in the Tlr4-mutant model. Future studies will investigate the macrophage origins and the utility of the pathways identified herein as indicators of immune system deficiencies and lung tumorigenesis.
Collapse
Affiliation(s)
- Carla-Maria Alexander
- a Department of Environmental and Occupational Health , Colorado School of Public Health , University of Colorado at Denver-Anschutz Medical Campus , Aurora , Colorado , USA
| | - Ka-Na Xiong
- a Department of Environmental and Occupational Health , Colorado School of Public Health , University of Colorado at Denver-Anschutz Medical Campus , Aurora , Colorado , USA
| | - Kalpana Velmurugan
- a Department of Environmental and Occupational Health , Colorado School of Public Health , University of Colorado at Denver-Anschutz Medical Campus , Aurora , Colorado , USA
| | - Julie Xiong
- a Department of Environmental and Occupational Health , Colorado School of Public Health , University of Colorado at Denver-Anschutz Medical Campus , Aurora , Colorado , USA
| | - Ross S Osgood
- b Department of Pharmaceutical Sciences , School of Pharmacy , University of Colorado Denver , Aurora , Colorado , USA
| | - Alison K Bauer
- a Department of Environmental and Occupational Health , Colorado School of Public Health , University of Colorado at Denver-Anschutz Medical Campus , Aurora , Colorado , USA
| |
Collapse
|
10
|
Bauer AK, Velmurugan K, Xiong KN, Alexander CM, Xiong J, Brooks R. Epiregulin is required for lung tumor promotion in a murine two-stage carcinogenesis model. Mol Carcinog 2016; 56:94-105. [PMID: 26894620 DOI: 10.1002/mc.22475] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 01/27/2016] [Accepted: 02/06/2016] [Indexed: 01/08/2023]
Abstract
Adenocarcinoma accounts for ∼40% of lung cancer, equating to ∼88 500 new patients in 2015, most of who will succumb to this disease, thus, the public health burden is evident. Unfortunately, few early biomarkers as well as effective therapies exist, hence the need for novel targets in lung cancer treatment. We previously identified epiregulin (Ereg), an EGF-like ligand, as a biomarker in several mouse lung cancer models. In the present investigation we used a primary two-stage initiation/promotion model to test our hypothesis that Ereg deficiency would reduce lung tumor promotion in mice. We used 3-methylcholanthrene (initiator) or oil vehicle followed by multiple weekly exposures to butylated hydroxytoluene (BHT; promoter) in mice lacking Ereg (Ereg-/- ) and wildtype controls (BALB/ByJ; Ereg+/+ ) and examined multiple time points and endpoints (bronchoalveolar lavage analysis, tumor analysis, mRNA expression, ELISA, wound assay) during tumor promotion. At the early time points (4 and 12 wk), we observed significantly reduced amounts of inflammation (macrophages, PMNs) in the Ereg-/- mice compared to controls (Ereg+/+ ). At 20 wk, tumor multiplicity was also significantly decreased in the Ereg-/- mice versus controls (Ereg+/+ ). IL10 expression, an anti-inflammatory mediator, and downstream signaling events (Stat3) were significantly increased in the Ereg-/- mice in response to BHT, supporting both reduced inflammation and tumorigenesis. Lastly, wound healing was significantly increased with recombinant Ereg in both human and mouse lung epithelial cell lines. These results indicate that Ereg has proliferative potential and may be utilized as an early cancer biomarker as well as a novel potential therapeutic target. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Alison K Bauer
- Department of Environmental and Occupational Health, Colorado School of Public Health, University of Colorado Denver, Aurora, Colorado
| | - Kalpana Velmurugan
- Department of Environmental and Occupational Health, Colorado School of Public Health, University of Colorado Denver, Aurora, Colorado
| | - Ka-Na Xiong
- Department of Environmental and Occupational Health, Colorado School of Public Health, University of Colorado Denver, Aurora, Colorado
| | - Carla-Maria Alexander
- Department of Environmental and Occupational Health, Colorado School of Public Health, University of Colorado Denver, Aurora, Colorado
| | - Julie Xiong
- Department of Environmental and Occupational Health, Colorado School of Public Health, University of Colorado Denver, Aurora, Colorado
| | - Rana Brooks
- Department of Environmental and Occupational Health, Colorado School of Public Health, University of Colorado Denver, Aurora, Colorado
| |
Collapse
|
11
|
Wilson AJ, Saskowski J, Barham W, Khabele D, Yull F. Microenvironmental effects limit efficacy of thymoquinone treatment in a mouse model of ovarian cancer. Mol Cancer 2015; 14:192. [PMID: 26552746 PMCID: PMC4640396 DOI: 10.1186/s12943-015-0463-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 10/23/2015] [Indexed: 01/01/2023] Open
Abstract
Background Ovarian cancer is the most lethal gynecologic malignancy, with limited treatment options for chemoresistant disease. An important link between inflammation and peritoneal spread of ovarian cancer is NF-κB signaling. Thymoquinone (TQ) exerts multiple anti-tumorigenic cellular effects, including NF-κB inhibition. We aimed to investigate the therapeutic potential of TQ in an established murine syngeneic model of ovarian cancer. Methods ID8-NGL mouse ovarian cancer cells stably expressing an NF-κB reporter transgene were injected intra-peritoneally into C57BL/6 mice, and mice were treated with TQ or vehicle for 10 or 30 days. TQ was combined with the macrophage depleting drug, liposomal clodronate, in selected experiments. Effects on peritoneal tumor burden were measured by volume of ascites, number of peritoneal implants and mesenteric tumor mass. NF-κB reporter activity and markers of proliferation and apoptosis were measured in tumors and in confirmatory in vitro experiments. Protein or mRNA expression of M1 (anti-tumor) and M2 (pro-tumor) macrophage markers, and soluble cytokine profiles, were examined from harvested ascites fluid, peritoneal lavages and/or tumor sections. 2-tailed Mann–Whitney tests were used for measuring differences between groups in in vivo experiments. Results Consistent with its effects in vitro, TQ reduced proliferation and increased apoptosis in ID8-NGL tumors after 10 and 30 day treatment. Prolonged TQ treatment did not significantly alter tumor number or mass compared to vehicle, but rather exerted an overall deleterious effect by stimulating ascites formation. Increased ascites was accompanied by elevated NF-κB activity in tumors and macrophages, increased pro-tumor M2 macrophages and expression of pro-tumorigenic soluble factors such as VEGF in ascites fluid, and increased tumor infiltration of M2 macrophages. In contrast, a 10 day exposure to TQ produced no ascites, and reduced tumor NF-κB activity, M2 macrophages and soluble VEGF levels. Peritoneal macrophage depletion by clodronate significantly reduced tumor burden. However, TQ-stimulated ascites was further enhanced by co-treatment with clodronate, with macrophages present overwhelmingly of the M2 phenotype. Conclusions Our findings show that pro-tumorigenic microenvironmental effects limited the efficacy of TQ in a syngeneic mouse model of ovarian cancer, and provide caution regarding its potential use in clinical trials in ovarian cancer patients. Electronic supplementary material The online version of this article (doi:10.1186/s12943-015-0463-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Andrew J Wilson
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Vanderbilt University School of Medicine, Nashville, TN, USA. .,Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, B1100 Medical Center North, Nashville, TN, 37232, USA.
| | - Jeanette Saskowski
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Whitney Barham
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Dineo Khabele
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Vanderbilt University School of Medicine, Nashville, TN, USA.,Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Fiona Yull
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN, USA.,Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
12
|
Thymoquinone enhances cisplatin-response through direct tumor effects in a syngeneic mouse model of ovarian cancer. J Ovarian Res 2015. [PMID: 26215403 PMCID: PMC4517540 DOI: 10.1186/s13048-015-0177-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Ovarian cancer is the most lethal gynecologic malignancy characterized by the frequent development of resistance to platinum chemotherapy. Finding new drug combinations to overcome platinum resistance is a key clinical challenge. Thymoquinone (TQ) is a component of black seed oil that exerts multiple anti-tumorigenic effects on cells, including inhibition of NF-κB and promotion of DNA damage. We aimed to determine whether TQ enhances cisplatin cytotoxicity in cultured ovarian cancer cells and in an established murine syngeneic model of ovarian cancer. METHODS Ovarian cancer cell viability in vitro was measured by sulforhodamine B (SRB) assays, and drug interactions tested for synergism by isobologram analysis. ID8-NGL mouse ovarian cancer cells stably expressing an NF-κB reporter transgene were injected intra-peritoneally into C57BL/6 mice. After 30 day TQ and/or cisplatin treatment, we measured the following indices: tumor burden (ascites volume, number of peritoneal implants and mesenteric tumor mass); NF-κB reporter activity (luciferase assay); protein expression of the double-strand DNA break marker, pH2AX(ser139), the proliferation markers, Ki67/mib-1 and PCNA, and the apoptosis markers, cleaved caspase-3, cleaved PARP and Bax; and mRNA expression of NF-κB targets, TNF-α and IL-1β. Two-tailed Mann-Whitney tests were used for measuring differences between groups in mouse experiments. RESULTS In SRB assays, TQ and cisplatin synergized in ID8-NGL cells. In mice, cisplatin significantly reduced cell proliferation and increased apoptosis in tumors, resulting in decreased overall tumor burden. Combining TQ with cisplatin further decreased these indices, indicating co-operative effects between the drugs. TQ treatment promoted cisplatin-induced pH2AX expression in cultured cells and in tumors. While NF-κB inhibition by TQ induced anti-tumor effects in vitro, we made the unexpected observation that TQ alone increased both tumor NF-κB activity and formation of ascites in vivo. CONCLUSIONS TQ enhanced cisplatin-mediated cytoxicity in ovarian cancer cells in vitro and in a mouse syngeneic model, effects associated with increased DNA damage. However, our results strongly caution that TQ treatment alone may have an overall deleterious effect in the immunocompetent host through stimulation of ascites. Since TQ is a potential candidate for future clinical trials in ovarian cancer patients, this finding has considerable potential relevance to the clinic.
Collapse
|
13
|
Giopanou I, Lilis I, Papaleonidopoulos V, Marazioti A, Spella M, Vreka M, Papadaki H, Stathopoulos GT. Comprehensive Evaluation of Nuclear Factor-κΒ Expression Patterns in Non-Small Cell Lung Cancer. PLoS One 2015; 10:e0132527. [PMID: 26147201 PMCID: PMC4493092 DOI: 10.1371/journal.pone.0132527] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 06/15/2015] [Indexed: 12/22/2022] Open
Abstract
Nuclear factor (NF)-κB signalling is required for lung adenocarcinoma development in mice, and both of its subunits RelA and RelB were independently reported to be highly expressed in human non-small cell lung cancer (NSCLC). To comprehensively examine NF-κB expression in NSCLC, we analyzed serial sections of primary tumor samples from 77 well-documented patients (36 adenocarcinomas, 40 squamous cell carcinomas and 3 large cell carcinomas) for immunoreactivity of RelA, RelB, P50, and P52/P100. Tumor and intratumoral stroma areas were discriminated based on proliferating cell nuclear antigen immunoreactivity and inflammatory infiltration was assessed in intratumoral stroma areas. NF-κB immunoreactivity was quantified by intensity, extent, and nuclear localization and was cross-examined with tumor cell proliferation, inflammatory infiltration, and clinical-pathologic data. We found that the expression of the different NF-κB subunits was not concordant, warranting our integral approach. Overall, RelA, RelB, and P50 were expressed at higher levels compared with P52/P100. However, RelA and P50 were predominantly expressed in intratumoral stroma, but RelB in tumor cells. Importantly, tumor area RelA expression was correlated with the intensity of inflammatory infiltration, whereas RelB expression was identified in proliferating tumor cells. Using multiple logistic regression, we identified that tumor RelB expression was an independent predictor of lymph node metastasis, and tumor P50 was an independent predictor of TNM6 stage IIB or higher, whereas tumor RelA was an independent predictor of inflammatory infiltration. We conclude that pathologic studies of NF-κB expression in cancer should include multiple pathway components. Utilizing such an approach, we identified intriguing associations between distinct NF-κB subunits and clinical and pathologic features of NSCLC.
Collapse
Affiliation(s)
- Ioanna Giopanou
- Laboratory for Molecular Respiratory Carcinogenesis, Department of Physiology, Faculty of Medicine, University of Patras, Rio, Achaia, Greece
| | - Ioannis Lilis
- Laboratory for Molecular Respiratory Carcinogenesis, Department of Physiology, Faculty of Medicine, University of Patras, Rio, Achaia, Greece
| | - Vassilios Papaleonidopoulos
- Laboratory for Molecular Respiratory Carcinogenesis, Department of Physiology, Faculty of Medicine, University of Patras, Rio, Achaia, Greece
| | - Antonia Marazioti
- Laboratory for Molecular Respiratory Carcinogenesis, Department of Physiology, Faculty of Medicine, University of Patras, Rio, Achaia, Greece
| | - Magda Spella
- Laboratory for Molecular Respiratory Carcinogenesis, Department of Physiology, Faculty of Medicine, University of Patras, Rio, Achaia, Greece
| | - Malamati Vreka
- Laboratory for Molecular Respiratory Carcinogenesis, Department of Physiology, Faculty of Medicine, University of Patras, Rio, Achaia, Greece
| | - Helen Papadaki
- Department of Anatomy, Faculty of Medicine, University of Patras, Rio, Achaia, Greece
| | - Georgios T. Stathopoulos
- Laboratory for Molecular Respiratory Carcinogenesis, Department of Physiology, Faculty of Medicine, University of Patras, Rio, Achaia, Greece
- Comprehensive Pneumology Center (CPC), University Hospital, Ludwig-Maximilians University and Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
- * E-mail:
| |
Collapse
|
14
|
Feng F, Wu Y, Zhang S, Liu Y, Qin L, Wu Y, Yan Z, Wu W. Macrophages facilitate coal tar pitch extract-induced tumorigenic transformation of human bronchial epithelial cells mediated by NF-κB. PLoS One 2012; 7:e51690. [PMID: 23227270 PMCID: PMC3515562 DOI: 10.1371/journal.pone.0051690] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Accepted: 11/05/2012] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE Chronic respiratory inflammation has been associated with lung cancer. Tumor-associated macrophages (TAMs) play a critical role in the formation of inflammation microenvironment. We sought to characterize the role of TAMs in coal tar pitch extract (CTPE)-induced tumorigenic transformation of human bronchial epithelial cells and the underlying mechanisms. METHODS The expression of TAMs-specific CD68 in lung cancer tissues and paired adjacent tissues from cancer patients was determined using immunostaining. Co-culture of human bronchial epithelial cells (BEAS-2B) and macrophage-like THP-1 cells were conducted to evaluate the promotive effect of macrophages on CTPE-induced tumorigenic transformation of BEAS-2B cells. BEAS-2B cells were first treated with 2.4 µg/mL CTPE for 72 hours. After removal of CTPE, the cells were continuously cultured either with or without THP-1 cells and passaged using trypsin-EDTA. Alterations of cell cycle, karyotype, colony formation in soft agar and tumor xenograft growth in nude mice of BEAS-2B cells at passages 10, 20 and 30, indicative of tumorigenecity, were determined, respectively. In addition, mRNA and protein levels of NF-κB in BEAS-2B cells were measured with RT-PCR and western blot, respectively. B(a)P was used as the positive control. RESULTS The over-expression of TAMs-specific CD68 around lung tumor tissues was detected and associated with lung cancer progression. The tumorigenic alterations of BEAS-2B cells including increase in cell growth rate, number of cells with aneuploidy, clonogenicity in soft agar, and tumor size in nude mice in vivo occurred at passage 10, becoming significant at passages 20 and 30 of the co-culture following CTPE removal in compared to BEAS-2B cells alone. In addition, the expression levels of NF-κB in BEAS-2B cells were positively correlated to the malignancy of BEAS-2B cells under different conditions of treatment. CONCLUSION The presence of macrophages facilitated CTPE-induced tumorigenic transformation of BEAS-2B cells, which may be mediated by NF-κB.
Collapse
Affiliation(s)
- Feifei Feng
- Department of Occupational and Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China.
| | | | | | | | | | | | | | | |
Collapse
|