1
|
Janus P, Kuś P, Jaksik R, Vydra N, Toma-Jonik A, Gramatyka M, Kurpas M, Kimmel M, Widłak W. Transcriptional responses to direct and indirect TGFB1 stimulation in cancerous and noncancerous mammary epithelial cells. Cell Commun Signal 2024; 22:522. [PMID: 39468555 PMCID: PMC11514872 DOI: 10.1186/s12964-024-01821-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 09/07/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Transforming growth factor beta (TGFβ) is important for the morphogenesis and secretory function of the mammary gland. It is one of the main activators of the epithelial-mesenchymal transition (EMT), a process important for tissue remodeling and regeneration. It also provides cells with the plasticity to form metastases during tumor progression. Noncancerous and cancer cells respond differently to TGFβ. However, knowledge of the cellular signaling cascades triggered by TGFβ in various cell types is still limited. METHODS MCF10A (noncancerous, originating from fibrotic breast tissue) and MCF7 (cancer, estrogen receptor-positive) breast epithelial cells were treated with TGFB1 directly or through conditioned media from stimulated cells. Transcriptional changes (via RNA-seq) were assessed in untreated cells and after 1-6 days of treatment. Differentially expressed genes were detected with DESeq2 and the hallmark collection was selected for gene set enrichment analysis. RESULTS TGFB1 induces EMT in both the MCF10A and MCF7 cell lines but via slightly different mechanisms (signaling through SMAD3 is more active in MCF7 cells). Many EMT-related genes are expressed in MCF10A cells at baseline. Both cell lines respond to TGFB1 by decreasing the expression of genes involved in cell proliferation: through the repression of MYC (and the protein targets) in MCF10A cells and the activation of p63-dependent signaling in MCF7 cells (CDKN1A and CDKN2B, which are responsible for the inhibition of cyclin-dependent kinases, are upregulated). In addition, estrogen receptor signaling is inhibited and caspase-dependent cell death is induced only in MCF7 cells. Direct incubation with TGFB1 and treatment of cells with conditioned media similarly affected transcriptional profiles. However, TGFB1-induced protein secretion is more pronounced in MCF10A cells; therefore, the signaling is propagated through conditioned media (bystander effect) more effectively in MCF10A cells than in MCF7 cells. CONCLUSIONS Estrogen receptor-positive breast cancer patients may benefit from high levels of TGFB1 expression due to the repression of estrogen receptor signaling, inhibition of proliferation, and induction of apoptosis in cancer cells. However, some TGFB1-stimulated cells may undergo EMT, which increases the risk of metastasis.
Collapse
Affiliation(s)
- Patryk Janus
- Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej 15, Gliwice, 44-102, Poland
| | - Paweł Kuś
- Department of Systems Biology and Engineering, Silesian University of Technology, Akademicka 16, Gliwice, 44-100, Poland
| | - Roman Jaksik
- Department of Systems Biology and Engineering, Silesian University of Technology, Akademicka 16, Gliwice, 44-100, Poland
| | - Natalia Vydra
- Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej 15, Gliwice, 44-102, Poland
| | - Agnieszka Toma-Jonik
- Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej 15, Gliwice, 44-102, Poland
| | - Michalina Gramatyka
- Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej 15, Gliwice, 44-102, Poland
| | - Monika Kurpas
- Department of Systems Biology and Engineering, Silesian University of Technology, Akademicka 16, Gliwice, 44-100, Poland
| | - Marek Kimmel
- Department of Systems Biology and Engineering, Silesian University of Technology, Akademicka 16, Gliwice, 44-100, Poland.
- Departments of Statistics and Bioengineering, Rice University, Houston, TX, USA.
| | - Wiesława Widłak
- Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej 15, Gliwice, 44-102, Poland.
| |
Collapse
|
2
|
Wang X, Eichhorn PJA, Thiery JP. TGF-β, EMT, and resistance to anti-cancer treatment. Semin Cancer Biol 2023; 97:1-11. [PMID: 37944215 DOI: 10.1016/j.semcancer.2023.10.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 05/08/2023] [Accepted: 10/16/2023] [Indexed: 11/12/2023]
Abstract
Transforming growth factor-β (TGF-β) signaling regulates cell-specific programs involved in embryonic development, wound-healing, and immune homeostasis. Yet, during tumor progression, these TGF-β-mediated programs are altered, leading to epithelial cell plasticity and a reprogramming of epithelial cells into mesenchymal lineages through epithelial-to-mesenchymal transition (EMT), a critical developmental program in morphogenesis and organogenesis. These changes, in turn, lead to enhanced carcinoma cell invasion, metastasis, immune cell differentiation, immune evasion, and chemotherapy resistance. Here, we discuss EMT as one of the critical programs associated with carcinoma cell plasticity and the influence exerted by TGF-β on carcinoma status and function. We further explore the composition of carcinoma and other cell populations within the tumor microenvironment, and consider the relevant outcomes related to the programs associated with cancer treatment resistance.
Collapse
Affiliation(s)
- Xuecong Wang
- Guangzhou National Laboratory, Guangzhou, China; Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Pieter Johan Adam Eichhorn
- Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, GPO Box U1987, Perth, WA 6845, Australia; Curtin Medical School, Curtin University, GPO Box U1987, Perth, WA 6845, Australia; Cancer Science Institute of Singapore, National University of Singapore, 117599 Singapore, Singapore
| | | |
Collapse
|
3
|
Wang CZ, Zhang ZQ, Zhang Y, Zheng LF, Liu Y, Yan AT, Zhang YC, Chang QH, Sha S, Xu ZJ. Comprehensive characterization of TGFB1 across hematological malignancies. Sci Rep 2023; 13:19107. [PMID: 37925591 PMCID: PMC10625629 DOI: 10.1038/s41598-023-46552-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 11/02/2023] [Indexed: 11/05/2023] Open
Abstract
TGFB1, which encodes TGF-β1, a potent cytokine regulating varies cellular processes including immune responses. TGF-β1 plays context-dependent roles in cancers and is increasingly recognized as a therapeutic target to enhance immunotherapy responses. We comprehensively evaluated expression of TGFB1 and its clinical and biological effects across hematological malignancies. TGFB1 expression was first explored using data from the GTEx, CCLE, and TCGA databases. The expression and clinical significances of TGFB1 in hematological malignancies were analyzed using Hemap and our In Silico curated datasets. We also analyzed the relationship between TGFB1 with immune scores and immune cell infiltrations in Hemap. We further assessed the value of TGFB1 in predicting immunotherapy response using TIDE and real-world immunotherapy datasets. TGFB1 showed a hematologic-tissue-specific expression pattern both across normal tissues and cancer types. TGFB1 expression were broadly dysregulated in blood cancers and generally associated with adverse prognosis. TGFB1 expression were associated with distinct TME properties among different blood cancer types. In addition, TGFB1 expression was found to be a useful marker in predicting immunotherapy responses. Our results suggest that TGFB1 is broadly dysregulated in hematological malignancies. TGFB1 might regulate the immune microenvironment in a cancer-type-specific manner, which could be applied in the development of new targeted drugs for immunotherapy.
Collapse
Affiliation(s)
- Cui-Zhu Wang
- Department of Hematology and Oncology, Affiliated Haian Hospital of Nantong University, Nantong, China
| | - Zi-Qi Zhang
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yan Zhang
- Department of Hematology and Oncology, Affiliated Haian Hospital of Nantong University, Nantong, China
| | - Liang-Feng Zheng
- Laboratory Center, Affiliated Haian Hospital of Nantong University, Nantong, China
| | - Yang Liu
- Clinical Nutrition Department, Haian Hospital of Traditional Chinese Medicine, Nantong, China
| | - Ai-Ting Yan
- Department of Hematology and Oncology, Affiliated Haian Hospital of Nantong University, Nantong, China
| | - Yuan-Cui Zhang
- Department of Respiratory Medicine, The Affiliated Zhenjiang Third Hospital of Jiangsu University, 1 Dingmao Bridge, Youth Square, Zhenjiang, 212002, China
| | - Qing-Hua Chang
- Department of Respiratory Medicine, The Affiliated Zhenjiang Third Hospital of Jiangsu University, 1 Dingmao Bridge, Youth Square, Zhenjiang, 212002, China.
| | - Suo Sha
- Surgery of Traditional Chinese Medicine, Haian Hospital of Traditional Chinese Medicine, Nantong, 226600, China.
| | - Zi-Jun Xu
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, 8 Dianli Rd., Zhenjiang, 212002, China.
| |
Collapse
|
4
|
Spira A, Awada A, Isambert N, Lorente D, Penel N, Zhang Y, Ojalvo LS, Hicking C, Rolfe PA, Ihling C, Dussault I, Locke G, Borel C. Identification of HMGA2 as a predictive biomarker of response to bintrafusp alfa in a phase 1 trial in patients with advanced triple-negative breast cancer. Front Oncol 2022; 12:981940. [PMID: 36568239 PMCID: PMC9773992 DOI: 10.3389/fonc.2022.981940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 10/28/2022] [Indexed: 12/13/2022] Open
Abstract
Background We report the clinical activity, safety, and identification of a predictive biomarker for bintrafusp alfa, a first-in-class bifunctional fusion protein composed of the extracellular domain of TGFβRII (a TGF-β "trap") fused to a human IgG1 mAb blocking PD-L1, in patients with advanced triple-negative breast cancer (TNBC). Methods In this expansion cohort of a global phase 1 study, patients with pretreated, advanced TNBC received bintrafusp alfa 1200 mg every 2 weeks intravenously until disease progression, unacceptable toxicity, or withdrawal. The primary objective was confirmed best overall response by RECIST 1.1 assessed per independent review committee (IRC). Results As of May 15, 2020, a total of 33 patients had received bintrafusp alfa, for a median of 6.0 (range, 2.0-48.1) weeks. The objective response rate was 9.1% (95% CI, 1.9%-24.3%) by IRC and investigator assessment. The median progression-free survival per IRC was 1.3 (95% CI, 1.2-1.4) months, and median overall survival was 7.7 (95% CI, 2.1-10.9) months. Twenty-five patients (75.8%) experienced treatment-related adverse events (TRAEs). Grade 3 TRAEs occurred in 5 patients (15.2%); no patients had a grade 4 TRAE. There was 1 treatment-related death (dyspnea, hemolysis, and thrombocytopenia in a patient with extensive disease at trial entry). Responses occurred independently of PD-L1 expression, and tumor RNAseq data identified HMGA2 as a potential biomarker of response. Conclusions Bintrafusp alfa showed clinical activity and manageable safety in patients with heavily pretreated advanced TNBC. HMGA2 was identified as a potential predictive biomarker of response. ClinicalTrialsgov identifier NCT02517398.
Collapse
Affiliation(s)
- Alexander Spira
- Department of Medical Oncology, Virginia Cancer Specialists, Fairfax, VA, United States
- US Oncology Research, The Woodlands, TX, United States
| | - Ahmad Awada
- Medical Oncology Clinic, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Nicolas Isambert
- Department of Medical Oncology, Centre Georges-François Leclerc, Dijon, France
| | - David Lorente
- Department of Medical Oncology, Hospital Universitari I Politècnic La Fe, Valencia, Spain
| | - Nicolas Penel
- Department of Medical Oncology, Centre Oscar Lambret, Lille, France
- Department of Medical Oncology, Université de Lille, Lille, France
| | - Yue Zhang
- EMD Serono Research & Development Institute, Inc, an Affiliate of Merck KGaA, Billerica, MA, United States
| | - Laureen S. Ojalvo
- EMD Serono Research & Development Institute, Inc, an Affiliate of Merck KGaA, Billerica, MA, United States
| | | | - P. Alexander Rolfe
- EMD Serono Research & Development Institute, Inc, an Affiliate of Merck KGaA, Billerica, MA, United States
| | | | - Isabelle Dussault
- EMD Serono Research & Development Institute, Inc, an Affiliate of Merck KGaA, Billerica, MA, United States
| | - George Locke
- EMD Serono Research & Development Institute, Inc, an Affiliate of Merck KGaA, Billerica, MA, United States
| | - Christian Borel
- Department of Medical Oncology, Centre Paul Strauss, Strasbourg, France
| |
Collapse
|
5
|
Shukla N, Naik A, Moryani K, Soni M, Shah J, Dave H. TGF-β at the crossroads of multiple prognosis in breast cancer, and beyond. Life Sci 2022; 310:121011. [PMID: 36179816 DOI: 10.1016/j.lfs.2022.121011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 09/16/2022] [Accepted: 09/25/2022] [Indexed: 10/25/2022]
Abstract
Transforming growth factor β (TGF-β), a pluripotent cytokine and a multifunctional growth factor has a crucial role in varied biological mechanisms like invasion, migration, epithelial-mesenchymal transition, apoptosis, wound healing, and immunosuppression. Moreover, it also has an imperative role both in normal mammary gland development as well as breast carcinogenesis. TGF-β has shown to have a paradoxical role in breast carcinogenesis, by transitioning from a growth inhibitor to a growth promoter with the disease advancement. The inter-communication and crosstalk of TGF-β with different signaling pathways has strengthened the likelihood to explore it as a comprehensive biomarker. In the last two decades, TGF-β has been studied extensively and has been found to be a promising biomarker for early detection, disease monitoring, treatment selection, and tumor progression making it beneficial for disease management. In this review, we focus on the signaling pathways and biological activities of the TGF-β family in breast cancer pathogenesis and its role as a circulatory and independent biomarker for breast cancer progression and metastasis. Moreover, this review highlights TGF-β as a drug target, and the underlying mechanisms through which it is involved in tumorigenesis that will aid in the development of varied therapies targeting the different stages of breast cancer.
Collapse
Affiliation(s)
- Nirali Shukla
- Institute of Science, Nirma University, Ahmedabad, Gujarat 382481, India
| | - Ankit Naik
- Ahmedabad University, Ahmedabad, Gujarat 390009, India
| | - Kamlesh Moryani
- Institute of Science, Nirma University, Ahmedabad, Gujarat 382481, India
| | - Molisha Soni
- Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat 382481, India
| | - Jigna Shah
- Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat 382481, India
| | - Heena Dave
- Institute of Science, Nirma University, Ahmedabad, Gujarat 382481, India.
| |
Collapse
|
6
|
Corvino D, Kumar A, Bald T. Plasticity of NK cells in Cancer. Front Immunol 2022; 13:888313. [PMID: 35619715 PMCID: PMC9127295 DOI: 10.3389/fimmu.2022.888313] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/12/2022] [Indexed: 12/19/2022] Open
Abstract
Natural killer (NK) cells are crucial to various facets of human immunity and function through direct cytotoxicity or via orchestration of the broader immune response. NK cells exist across a wide range of functional and phenotypic identities. Murine and human studies have revealed that NK cells possess substantial plasticity and can alter their function and phenotype in response to external signals. NK cells also play a critical role in tumor immunity and form the basis for many emerging immunotherapeutic approaches. NK cells can directly target and lyse malignant cells with their inherent cytotoxic capabilities. In addition to direct targeting of malignant cells, certain subsets of NK cells can mediate antibody-dependent cellular cytotoxicity (ADCC) which is integral to some forms of immune checkpoint-blockade immunotherapy. Another important feature of various NK cell subsets is to co-ordinate anti-tumor immune responses by recruiting adaptive and innate leukocytes. However, given the diverse range of NK cell identities it is unsurprising that both pro-tumoral and anti-tumoral NK cell subsets have been described. Here, NK cell subsets have been shown to promote angiogenesis, drive inflammation and immune evasion in the tumor microenvironment. To date, the signals that drive tumor-infiltrating NK cells towards the acquisition of a pro- or anti-tumoral function are poorly understood. The notion of tumor microenvironment-driven NK cell plasticity has substantial implications for the development of NK-based immunotherapeutics. This review will highlight the current knowledge of NK cell plasticity pertaining to the tumor microenvironment. Additionally, this review will pose critical and relevant questions that need to be addressed by the field in coming years.
Collapse
Affiliation(s)
- Dillon Corvino
- Tumor-Immunobiology, Institute for Experimental Oncology, University Hospital Bonn, Bonn, Germany
| | - Ananthi Kumar
- Tumor-Immunobiology, Institute for Experimental Oncology, University Hospital Bonn, Bonn, Germany
| | - Tobias Bald
- Tumor-Immunobiology, Institute for Experimental Oncology, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
7
|
Abstract
Transforming growth factor-β (TGFβ) signalling controls multiple cell fate decisions during development and tissue homeostasis; hence, dysregulation of this pathway can drive several diseases, including cancer. Here we discuss the influence that TGFβ exerts on the composition and behaviour of different cell populations present in the tumour immune microenvironment, and the context-dependent functions of this cytokine in suppressing or promoting cancer. During homeostasis, TGFβ controls inflammatory responses triggered by exposure to the outside milieu in barrier tissues. Lack of TGFβ exacerbates inflammation, leading to tissue damage and cellular transformation. In contrast, as tumours progress, they leverage TGFβ to drive an unrestrained wound-healing programme in cancer-associated fibroblasts, as well as to suppress the adaptive immune system and the innate immune system. In consonance with this key role in reprogramming the tumour microenvironment, emerging data demonstrate that TGFβ-inhibitory therapies can restore cancer immunity. Indeed, this approach can synergize with other immunotherapies - including immune checkpoint blockade - to unleash robust antitumour immune responses in preclinical cancer models. Despite initial challenges in clinical translation, these findings have sparked the development of multiple therapeutic strategies that inhibit the TGFβ pathway, many of which are currently in clinical evaluation.
Collapse
Affiliation(s)
- Daniele V F Tauriello
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Elena Sancho
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Barcelona, Spain
| | - Eduard Batlle
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Barcelona, Spain.
- Institucio Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain.
| |
Collapse
|
8
|
Liu Q, Chen G, Moore J, Guix I, Placantonokis D, Barcellos-Hoff MH. Exploiting Canonical TGFβ Signaling in Cancer Treatment. Mol Cancer Ther 2021; 21:16-24. [PMID: 34670783 PMCID: PMC8742762 DOI: 10.1158/1535-7163.mct-20-0891] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/15/2021] [Accepted: 10/18/2021] [Indexed: 11/16/2022]
Abstract
Transforming growth factor β (TGFβ) is a pleiotropic cytokine that plays critical roles to define cancer cell phenotypes, construct the tumor microenvironment, and suppress anti-tumor immune responses. As such, TGFβ is a lynchpin for integrating cancer cell intrinsic pathways and communication among host cells in the tumor and beyond that together affect responses to genotoxic, targeted, and immune therapy. Despite decades of preclinical and clinical studies, evidence of clinical benefit from targeting TGFβ in cancer remains elusive. Here, we review the mechanisms by which TGFβ acts to oppose successful cancer therapy, the reported prognostic and predictive value of TGFβ biomarkers, and the potential impact of inhibiting TGFβ in precision oncology. Paradoxically, the diverse mechanisms by which TGFβ impedes therapeutic response are a principal barrier to implementing TGFβ inhibitors because it is unclear which TGFβ mechanism is functional in which patient. Companion diagnostic tools and specific biomarkers of TGFβ targeted biology will be the key to exploiting TGFβ biology for patient benefit.
Collapse
Affiliation(s)
- Qi Liu
- Shenzhen Bay Laboratory, Institute for Biomedical Engineering
| | - Genwen Chen
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University
| | - Jade Moore
- Department of Radiation Oncology, University of California, San Francicsco
| | - Ines Guix
- Department of Radiation Oncology, University of California, San Francicsco
| | | | | |
Collapse
|
9
|
Lee S, Byeon S, Ko J, Hyung S, Lee I, Jeon NL, Hong JY, Kim ST, Park SH, Lee J. Reducing tumor invasiveness by ramucirumab and TGF-β receptor kinase inhibitor in a diffuse-type gastric cancer patient-derived cell model. Cancer Med 2021; 10:7253-7262. [PMID: 34542244 PMCID: PMC8525100 DOI: 10.1002/cam4.4259] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 07/19/2021] [Accepted: 08/10/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Diffuse-type gastric cancer (GC) is known to be more aggressive and relatively resistant to conventional chemotherapy. Hence, more optimized treatment strategy is urgently needed in diffuse-type GC. METHODS Using a panel of 10 GC cell lines and 3 GC patient-derived cells (PDCs), we identified cell lines with high EMTness which is a distinct feature for diffuse-type GC. We treated GC cells with high EMTness with ramucirumab alone, TGF-β receptor kinase inhibitor (TEW-7197) alone, or in combination to investigate the drug's effects on invasiveness, spheroid formation, EMT marker expression, and tumor-induced angiogenesis using a spheroid-on-a-chip model. RESULTS Both TEW-7197 and ramucirumab treatments profoundly decreased invasiveness of EMT-high cell lines and PDCs. With a 3D tumor spheroid-on-a-chip, we identified versatile influence of co-treatment on cancer cell-induced blood vessel formation as well as on EMT progression in tumor spheroids. The 3D tumor spheroid-on-a-chip demonstrated that TEW-7197 + ramucirumab combination significantly decreased PDC-induced vessel formation. CONCLUSIONS In this study, we showed TEW-7197 and ramucirumab considerably decreased invasiveness, thus EMTness in a panel of diffuse-type GC cell lines including GC PDCs. Taken together, we confirmed that combination of TEW-7197 and ramucirumab reduced tumor spheroid and GC PDC-induced blood vessel formation concomitantly in the spheroid-on-a-chip model.
Collapse
Affiliation(s)
- Song‐Yi Lee
- Division of Hematology‐OncologyDepartment of MedicineSamsung Medical CenterSungkyunkwan UniversitySeoulKorea
| | - Seonggyu Byeon
- Department of Internal MedicineChungbuk National University HospitalChungbuk National University College of MedicineCheongjuKorea
| | - Jihoon Ko
- Division of Hematology‐OncologyDepartment of MedicineSamsung Medical CenterSungkyunkwan UniversitySeoulKorea
- Department of Mechanical EngineeringSeoul National UniversitySeoulKorea
| | - Sujin Hyung
- Division of Hematology‐OncologyDepartment of MedicineSamsung Medical CenterSungkyunkwan UniversitySeoulKorea
| | - In‐Kyoung Lee
- Division of Hematology‐OncologyDepartment of MedicineSamsung Medical CenterSungkyunkwan UniversitySeoulKorea
| | - Noo Li Jeon
- Department of Mechanical EngineeringSeoul National UniversitySeoulKorea
| | - Jung Yong Hong
- Division of Hematology‐OncologyDepartment of MedicineSamsung Medical CenterSungkyunkwan UniversitySeoulKorea
| | - Seung Tae Kim
- Division of Hematology‐OncologyDepartment of MedicineSamsung Medical CenterSungkyunkwan UniversitySeoulKorea
| | - Se Hoon Park
- Division of Hematology‐OncologyDepartment of MedicineSamsung Medical CenterSungkyunkwan UniversitySeoulKorea
| | - Jeeyun Lee
- Division of Hematology‐OncologyDepartment of MedicineSamsung Medical CenterSungkyunkwan UniversitySeoulKorea
- Department of Intelligent Precision Healthcare ConvergenceSungkyunkwan UniversitySuwonKorea
| |
Collapse
|
10
|
Tarannum N, Hendrickson OD, Khatoon S, Zherdev AV, Dzantiev BB. Molecularly imprinted polymers as receptors for assays of antibiotics. Crit Rev Anal Chem 2019; 50:291-310. [DOI: 10.1080/10408347.2019.1626697] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Nazia Tarannum
- Department of Chemistry, Chaudhary Charan Singh University, Meerut, India
| | - Olga D. Hendrickson
- A.N. Bach Institute of Biochemistry, Research Centre of Biotechnology of the Russian Academy of Sciences, Moscow, Russia
| | - Shahjadi Khatoon
- Department of Chemistry, Chaudhary Charan Singh University, Meerut, India
| | - Anatoly V. Zherdev
- A.N. Bach Institute of Biochemistry, Research Centre of Biotechnology of the Russian Academy of Sciences, Moscow, Russia
| | - Boris B. Dzantiev
- A.N. Bach Institute of Biochemistry, Research Centre of Biotechnology of the Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
11
|
Cai F, Cai L, Zhou Z, Pan X, Wang M, Chen S, Luis MAF, Cen C, Biskup E. Prognostic role of Tif1γ expression and circulating tumor cells in patients with breast cancer. Mol Med Rep 2019; 19:3685-3695. [PMID: 30896800 PMCID: PMC6470918 DOI: 10.3892/mmr.2019.10033] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 03/06/2019] [Indexed: 01/06/2023] Open
Abstract
Transcription intermediary factor 1γ (Tif1γ), a ubiquitous nuclear protein, is a regulator of transforming growth factor-β (TGF-β)/Smad signaling. Tif1γ can function as an oncogene and as a tumor suppressor. In the present study, Tif1γ levels were measured in the plasma of patients with breast cancer in order to investigate the association of Tif1γ with overall survival (OS). The results indicated that Tif1γ is an independent prognostic and predictive factor in breast cancer, and thus, a promising target protein for use in diagnostics and patient follow-up. Plasma levels of Tif1γ were measured in samples obtained from 110 patients with operable breast cancer and in 110 healthy volunteers at the Breast Cancer Department of Yangpu Hospital between 2008 and 2016. The association between Tif1γ levels and clinicopathologic parameters, and the OS in a follow-up period of 98 months was evaluated. The prognostic significance was assessed using the Kaplan-Meier method. The levels of Tif1γ were significantly lower in patients with breast cancer compared with healthy controls. The average concentration of 18.40 ng/ml was used to discriminate between Tif1γ-positive (52) and Tif1γ-negative patients (58). Tif1γ-positive patients had a significantly improved OS compared with Tif1γ-negative patients. In the multivariate analysis, Tif1γ was an independent predictor of a favorable OS in a prospective follow-up setting; thus, Tif1γ plasma levels are an independent prognostic factor for patients with breast cancer. These findings support the potential of using measurements of Tif1γ plasma levels to guide breast cancer therapy and monitoring. Further studies are required to validate Tif1γ as an easily detectable, non-invasive prognostic biomarker for breast cancer.
Collapse
Affiliation(s)
- Fengfeng Cai
- Department of Breast Surgery, Yangpu Hospital, Tongji University School of Medicine, Shanghai 200090, P.R. China
| | - Lu Cai
- Department of Breast Surgery, Yangpu Hospital, Tongji University School of Medicine, Shanghai 200090, P.R. China
| | - Zhuchao Zhou
- Department of General Surgery, Huashan Hospital, Fudan University, School of Medicine, Shanghai 200041, P.R. China
| | - Xin Pan
- Department of Central Laboratory, Yangpu Hospital, Tongji University School of Medicine, Shanghai 200090, P.R. China
| | - Minghong Wang
- Department of Cardiology, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, P.R. China
| | - Su Chen
- Department of Molecular and Cellular Biology, School of Forensic Sciences, Xi'an Jiao Tong University Health Science Center, Xi'an, Shanxi 710061, P.R. China
| | - Manuel Antonio Falar Luis
- Department of Breast Surgery, Yangpu Hospital, Tongji University School of Medicine, Shanghai 200090, P.R. China
| | - Chunmei Cen
- Department of Breast Surgery, Yangpu Hospital, Tongji University School of Medicine, Shanghai 200090, P.R. China
| | - Ewelina Biskup
- Department of Basic Medical Sciences, Shanghai University of Medicine and Health Sciences, Shanghai 201318, P.R. China
| |
Collapse
|
12
|
Abstract
The interaction between hair and malignancy is complicated. Various hair abnormalities can manifest in oncology patients as a clinical manifestation, the result of cancer therapy, or due to a paraneoplastic condition. The mechanisms of these changes remain unclear. Alopecia is one of the common clinical presentations occurring in oncology patients that affects their quality of life. The condition can concomitantly develop during the course of malignancy or when patients undergo cancer treatment. It is important for physicians to understand alopecia in association with malignancy as it may be an important associated finding or provide the clues to aid diagnosis. The aim of this review is to summarize the clinical characteristics of alopecia that occur in cancer patients and their relationship with the type of malignancy and its treatment.
Collapse
|
13
|
Eser PÖ, Jänne PA. TGFβ pathway inhibition in the treatment of non-small cell lung cancer. Pharmacol Ther 2018; 184:112-130. [DOI: 10.1016/j.pharmthera.2017.11.004] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
14
|
Flanders KC, Yang YA, Herrmann M, Chen J, Mendoza N, Mirza AM, Wakefield LM. Quantitation of TGF-β proteins in mouse tissues shows reciprocal changes in TGF-β1 and TGF-β3 in normal vs neoplastic mammary epithelium. Oncotarget 2018; 7:38164-38179. [PMID: 27203217 PMCID: PMC5122380 DOI: 10.18632/oncotarget.9416] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 04/26/2016] [Indexed: 12/14/2022] Open
Abstract
Transforming growth factor-βs (TGF-βs) regulate tissue homeostasis, and their expression is perturbed in many diseases. The three isoforms (TGF-β1, -β2, and -β3) have similar bioactivities in vitro but show distinct activities in vivo. Little quantitative information exists for expression of TGF-β isoform proteins in physiology or disease. We developed an optimized method to quantitate protein levels of the three isoforms, using a Luminex® xMAP®-based multianalyte assay following acid-ethanol extraction of tissues. Analysis of multiple tissues and plasma from four strains of adult mice showed that TGF-β1 is the predominant isoform with TGF-β2 being ~10-fold lower. There were no sex-specific differences in isoform expression, but some tissues showed inter-strain variation, particularly for TGF-β2. The only adult tissue expressing appreciable TGF-β3 was the mammary gland, where its levels were comparable to TGF-β1. In situ hybridization showed the luminal epithelium as the major source of all TGF-β isoforms in the normal mammary gland. TGF-β1 protein was 3-8-fold higher in three murine mammary tumor models than in normal mammary gland, while TGF-β3 protein was 2-3-fold lower in tumors than normal tissue, suggesting reciprocal regulation of these isoforms in mammary tumorigenesis.
Collapse
Affiliation(s)
- Kathleen C Flanders
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Yu-An Yang
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Michelle Herrmann
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, Bethesda, Maryland, United States of America
| | - JinQiu Chen
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Nerissa Mendoza
- XOMA Corporation, Berkeley, California, United States of America
| | - Amer M Mirza
- XOMA Corporation, Berkeley, California, United States of America
| | - Lalage M Wakefield
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, Bethesda, Maryland, United States of America
| |
Collapse
|
15
|
Tian J, Al-Odaini AA, Wang Y, Korah J, Dai M, Xiao L, Ali S, Lebrun JJ. KiSS1 gene as a novel mediator of TGFβ-mediated cell invasion in triple negative breast cancer. Cell Signal 2017; 42:1-10. [PMID: 28988968 DOI: 10.1016/j.cellsig.2017.10.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 09/28/2017] [Accepted: 10/05/2017] [Indexed: 12/31/2022]
Abstract
The invasive and metastatic phenotypes of breast cancer correlate with high recurrence rates and poor survival outcomes. Transforming growth factor-β (TGFβ) promotes tumor progression and metastasis in aggressive breast cancer. Here, we identified the kisspeptin KiSS1 as a downstream target of canonical TGFβ/Smad2 pathway in triple negative breast cancer cells. We also found KiSS1 expression to be required for TGFβ-induced cancer cell invasion. Indeed, knockdown expression of KiSS1 blocked TGFβ-mediated cancer cell invasion as well as metalloproteinase (MMP9) expression and activity. Interestingly, Kisspeptin-10 (KP-10), the smallest active form of kisspeptin also stimulates cancer cell invasive behavior through activation of MAPK/Erk pathway. We described a positive feedback loop between KiSS1 and p21 downstream of TGFβ, further contributing to TGFβ-induced cancer cell invasion. Lastly, we explored both the clinical utility of KiSS1 as a lymph node involvement predictive tool and its potential as a therapeutic target. We found KiSS1 high expression to correlate with lymph node positive status. Furthermore, blocking KiSS1 using a specific small peptide antagonist (p234) impaired TGFβ-mediated cell invasion and MMP9 induction. Together, our results define an essential role of KiSS1 in regulating TGFβ pro-invasive effects and define KiSS1 as a therapeutic new target for triple negative breast cancer.
Collapse
Affiliation(s)
- Jun Tian
- Department of Medicine, McGill University Health Center, Cancer Research Program, Montreal, QC, H4A 3J1, Canada
| | - Amal A Al-Odaini
- Department of Medicine, McGill University Health Center, Cancer Research Program, Montreal, QC, H4A 3J1, Canada
| | - Yun Wang
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing 400038, China
| | - Juliana Korah
- Department of Medicine, McGill University Health Center, Cancer Research Program, Montreal, QC, H4A 3J1, Canada
| | - Meiou Dai
- Department of Medicine, McGill University Health Center, Cancer Research Program, Montreal, QC, H4A 3J1, Canada
| | - Lan Xiao
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing 400038, China
| | - Suhad Ali
- Department of Medicine, McGill University Health Center, Cancer Research Program, Montreal, QC, H4A 3J1, Canada
| | - Jean-Jacques Lebrun
- Department of Medicine, McGill University Health Center, Cancer Research Program, Montreal, QC, H4A 3J1, Canada.
| |
Collapse
|
16
|
Zhang Y, Yang WB. Down-regulation of tripartite motif protein 59 inhibits proliferation, migration and invasion in breast cancer cells. Biomed Pharmacother 2017; 89:462-467. [PMID: 28249247 DOI: 10.1016/j.biopha.2017.02.039] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 02/10/2017] [Accepted: 02/13/2017] [Indexed: 01/18/2023] Open
Abstract
Tripartite motif protein 59 (TRIM59), also known as mouse ring finger protein 1 (MRF1), is a surface molecule and belongs to the TRIM family. Recently, TRIM59 has been described in multiple cancers such as gastric, prostatic and lung cancer. However, there have been no reports on its functions in breast cancer. In this study, we elucidated the biological roles of TRIM59 in breast cancer. We found that TRIM59 was up-regulated in breast cancer cells. Down-regulation of TRIM59 inhibited breast cancer cell proliferation, migration and invasion in vitro as well as tumor growth in vivo. In addition, TRIM59 down-regulation reduced the protein expression level of p-Smad2 and thus inhibited the activity of transforming growth factor-β (TGF-β) signaling. Taken together, our study results provided new evidence showing that TRIM59 may be considered as a promising therapeutic target for breast cancer.
Collapse
Affiliation(s)
- Yan Zhang
- Department of General Surgery, The Second Affiliated Hospital of Medical School, Xi'an Jiaotong University, Xi'an 710004, China
| | - Wen-Bin Yang
- Department of General Surgery, The Second Affiliated Hospital of Medical School, Xi'an Jiaotong University, Xi'an 710004, China.
| |
Collapse
|
17
|
Hejna M, Wöll E, Tschandl P, Raderer M. Cutaneous paraneoplastic disorders in stomach cancer: Collaboration between oncologically active dermatologists and clinical oncologists. Crit Rev Oncol Hematol 2016; 103:78-85. [PMID: 27247117 DOI: 10.1016/j.critrevonc.2016.04.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 03/14/2016] [Accepted: 04/26/2016] [Indexed: 01/25/2023] Open
Abstract
To our knowledge this is the first systemic review that provides an overview of the cutaneous paraneoplastic syndromes (CPS) (i.e., clinical manifestations, pathomechanisms, and treatment modalities) occurring in stomach cancer. CPS are caused by substances produced by stomach cancer and may precede, coincide with, or follow the diagnosis of this malignancy. More than 20 possible CPS in association with stomach cancer have been identified. CPS mostly compromises the patient's quality of life by skin impairment plus discomfort and are often associated with a dismal prognosis on survival. Studies of these CPS not only in stomach cancer have partially contributed to the understanding of pathomechanism and since CPS may be the presenting sign of an occult cancer, cognizance of their features and clinical implications are of considerable importance. Patients with these syndromes should have an appropriate work-up for a possibly occult malignancy with consecutive successful early treatment.
Collapse
Affiliation(s)
- Michael Hejna
- Comprehensive Cancer Center-GET, Medical University of Vienna, Vienna, Austria.
| | - Ewald Wöll
- St.Vinzenz Krankenhaus Betriebs GmbH, Zams, Austria
| | - Philipp Tschandl
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Markus Raderer
- Department of Internal Medicine I, Division of Oncology Medical University of Vienna, Vienna, Austria
| |
Collapse
|
18
|
Parallel Aspects of the Microenvironment in Cancer and Autoimmune Disease. Mediators Inflamm 2016; 2016:4375120. [PMID: 26997761 PMCID: PMC4779817 DOI: 10.1155/2016/4375120] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 01/13/2016] [Indexed: 02/07/2023] Open
Abstract
Cancer and autoimmune diseases are fundamentally different pathological conditions. In cancer, the immune response is suppressed and unable to eradicate the transformed self-cells, while in autoimmune diseases it is hyperactivated against a self-antigen, leading to tissue injury. Yet, mechanistically, similarities in the triggering of the immune responses can be observed. In this review, we highlight some parallel aspects of the microenvironment in cancer and autoimmune diseases, especially hypoxia, and the role of macrophages, neutrophils, and their interaction. Macrophages, owing to their plastic mode of activation, can generate a pro- or antitumoral microenvironment. Similarly, in autoimmune diseases, macrophages tip the Th1/Th2 balance via various effector cytokines. The contribution of neutrophils, an additional plastic innate immune cell population, to the microenvironment and disease progression is recently gaining more prominence in both cancer and autoimmune diseases, as they can secrete cytokines, chemokines, and reactive oxygen species (ROS), as well as acquire an enhanced ability to produce neutrophil extracellular traps (NETs) that are now considered important initiators of autoimmune diseases. Understanding the contribution of macrophages and neutrophils to the cancerous or autoimmune microenvironment, as well as the role their interaction and cooperation play, may help identify new targets and improve therapeutic strategies.
Collapse
|
19
|
Kassem L, Deygas M, Fattet L, Lopez J, Goulvent T, Lavergne E, Chabaud S, Carrabin N, Chopin N, Bachelot T, Gillet G, Treilleux I, Rimokh R. TIF1γ interferes with TGFβ1/SMAD4 signaling to promote poor outcome in operable breast cancer patients. BMC Cancer 2015; 15:453. [PMID: 26040677 PMCID: PMC4453047 DOI: 10.1186/s12885-015-1471-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2014] [Accepted: 05/26/2015] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND The Transforming growth factor β (TGFβ) signaling has a paradoxical role in cancer development and outcome. Besides, the prognostic significance of the TGFβ1, SMAD4 in breast cancer patients is an area of many contradictions. The transcriptional intermediary factor 1γ (TIF1γ) is thought to interact with the TGFβ/SMAD signaling through different mechanisms. Our study aims to define the prognostic significance of TGFβ1, SMAD4 and TIF1γ expression in breast cancer patients and to detect possible interactions among those markers that might affect the outcome. METHODS Immunohistochemistry was performed on tissue microarray (TMA) blocks prepared from samples of 248 operable breast cancer patients who presented at Centre Léon Bérard (CLB) between 1998 and 2001. The intensity and the percentage of stained tumor cells were integrated into a single score (0-6) and a cutoff was defined for high or low expression for each marker. Correlation was done between TGFβ1, SMAD4 and TIF1γ expression with the clinico-pathologic parameters using Pearson's chi-square test. Kaplan-Meier method was used to estimate distant metastasis free survival (DMFS), disease free survival (DFS) and overall survival (OS) and the difference between the groups was evaluated with log-rank test. RESULTS 223 cases were assessable for TIF1γ, 204 for TGFβ1 and 173 for SMAD4. Median age at diagnosis was 55.8 years (range: 27 to 89 years). Tumors were larger than 20 mm in 49.2% and 45.2% had axillary lymph node (LN) metastasis (N1a to N3). 19.4% of the patients had SBR grade I tumors, 46.8% grade II tumors and 33.9% grade III tumors. ER was positive in 85.4%, PR in 75.5% and Her2-neu was over-expressed in 10% of the cases. Nuclear TIF1γ, cytoplasmic TGFβ1, nuclear and cytoplasmic SMAD4 stainings were high in 35.9%, 30.4%, 27.7% and 52.6% respectively. TIF1γ expression was associated with younger age (p=0.006), higher SBR grade (p<0.001), more ER negativity (p=0.035), and tumors larger than 2 cm (p=0.081), while TGFβ1 was not associated with any of the traditional prognostic factors. TGFβ1 expression in tumor cells was a marker of poor prognosis regarding DMFS (HR=2.28; 95% CI: 1.4 to 3.8; p=0.002), DFS (HR=2.00; 95% CI: 1.25 to 3.5; p=0.005) and OS (HR=1.89; 95 % CI: 1.04 to 3.43; p=0.037). TIF1γ expression carried a tendency towards poorer DMFS (p=0.091), DFS (p=0.143) and OS (p=0.091). In the multivariate analysis TGFβ1 remained an independent predictor of shorter DMFS, DFS and OS. Moreover, the prognostic significance of TGFβ1 was more obvious in the TIF1γ high patient subgroup than in the patients with TIF1γ low expression. The subgroup expressing both markers had the worst DMFS (HR=3.2; 95% CI: 1.7 to 5.9; p<0.0001), DFS (HR=3.02; 95 % CI: 1.6 to 5.6; p<0.0001) and OS (HR=2.7; 95 % CI: 1.4 to 5.4; p=0.005). CONCLUSION There is a crosstalk between the TIF1γ and the TGFβ1/SMAD4 signaling that deteriorates the outcome of operable breast cancer patients and when combined together they can serve as an effective prognostic tool for those patients.
Collapse
Affiliation(s)
- Loay Kassem
- Clinical Oncology Department, Faculty of medicine, Cairo University, Cairo, Egypt.
| | - Mathieu Deygas
- U1052 Inserm, UMR CNRS 5286. Université de Lyon, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Lyon, France.
| | - Laurent Fattet
- U1052 Inserm, UMR CNRS 5286. Université de Lyon, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Lyon, France.
| | - Jonathan Lopez
- U1052 Inserm, UMR CNRS 5286. Université de Lyon, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Lyon, France.
| | - Thibaut Goulvent
- U1052 Inserm, UMR CNRS 5286. Université de Lyon, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Lyon, France.
| | | | | | | | - Nicolas Chopin
- Department of surgery, Centre Léon Bérard, Lyon, France.
| | - Thomas Bachelot
- Department of medical oncology, Centre Léon Bérard, Lyon, France.
| | - Germain Gillet
- U1052 Inserm, UMR CNRS 5286. Université de Lyon, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Lyon, France.
| | | | - Ruth Rimokh
- U1052 Inserm, UMR CNRS 5286. Université de Lyon, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Lyon, France.
| |
Collapse
|
20
|
Pellicciotta I, Marciscano AE, Hardee ME, Francis D, Formenti S, Barcellos-Hoff MH. Development of a novel multiplexed assay for quantification of transforming growth factor-β (TGF-β). Growth Factors 2015; 33:79-91. [PMID: 25586866 DOI: 10.3109/08977194.2014.999367] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Changes in activity or levels of transforming growth factor-β (TGF-β) are associated with a variety of diseases; however, measurement of TGF-β in biological fluids is highly variable. TGF-β is biologically inert when associated with its latency-associated peptide (LAP). Most available immunoassays require exogenous activation by acid/heat to release TGF-β from the latent complex. We developed a novel electrochemiluminescence-based multiplexed assay on the MesoScale Discovery® platform that eliminates artificial activation, simultaneously measures both active TGF-β1 and LAP1 and includes an internal control for platelet-derived TGF-β contamination in blood specimens. We optimized this assay to evaluate plasma levels as a function of activation type and clinical specimen preparation. We determined that breast cancer patients' plasma have higher levels of circulating latent TGF-β (LTGF-β) as measured by LAP1 than healthy volunteers (p < 0.0001). This assay provides a robust tool for correlative studies of LTGF-β levels with disease, treatment outcomes and toxicity with a broad clinical applicability.
Collapse
|
21
|
Bahhnassy A, Mohanad M, Shaarawy S, Ismail MF, El-Bastawisy A, Ashmawy AM, Zekri AR. Transforming growth factor-β, insulin-like growth factor I/insulin-like growth factor I receptor and vascular endothelial growth factor-A: prognostic and predictive markers in triple-negative and non-triple-negative breast cancer. Mol Med Rep 2015; 12:851-64. [PMID: 25824321 PMCID: PMC4438878 DOI: 10.3892/mmr.2015.3560] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 01/15/2015] [Indexed: 12/20/2022] Open
Abstract
In the current study, the prognostic and predictive values of serum transforming growth factor-β1 (TGF-β1), insulin-like growth factor I (IGF-I)/IGF-I receptor (IGF-IR) and vascular endothelial growth factor-A (VEGF-A) were evaluated in triple-negative and non-triple-negative breast cancer (TNBC and non-TNBC). The aim was to identify a group of serological biomarkers and to identify possible candidates for targeted therapy in patients with TNBC and non-TNBC. Protein levels of TGF-β1, IGF-I/IGF-IR and VEGF-A in the serum were measured in 43 TNBC, 53 non-TNBC and 20 normal control participants using quantitative ELISA assays. Results were correlated against standard prognostic factors, response to treatment and survival. TNBC was identified to be associated with poor prognosis and serum levels of VEGF-A and IGF/IGF-IR were significantly higher in the TNBC group compared with the non-TNBC group. IGF-IR and VEGF-A overexpression was observed to be correlated with TGF-β1 expression and all of the markers investigated were associated with metastasis and disease progression. In the multivariate analysis, VEGF-A, IGF-I and IGF-IR were observed to be independent predictors for overall survival, whereas TGF-β1 and lymph node status were identified as independent predictors for disease-free survival. The overall response rate was significantly lower in patients with TNBC and those with high levels of TGF-β1, IGF-I/IGF-IR and VEGF-A. In view of the present results, it was concluded that TGF-β1, IGF-I/IGF-IR and VEGF-A overexpression is associated with the presence of aggressive tumors, which exhibit an increased probability of metastasis, a poor response to treatment and reduced survival rate. This indicates that VEGF-A, IGF-IR and IGF-I have the potential to be used as surrogate biomarkers and are promising candidates for targeted therapy, particularly in patients with TNBC.
Collapse
Affiliation(s)
- Abeer Bahhnassy
- Molecular Pathology Unit, Pathology Department, National Cancer Institute, Cairo University, Cairo 11796, Egypt
| | - Marwa Mohanad
- Department of Biochemistry, Faculty of Pharmacy, Misr University for Science and Technology, Cairo 11796, Egypt
| | - Sabry Shaarawy
- Department of Cancer Biology, National Cancer Institute, Cairo University, Cairo 11796, Egypt
| | - Manal F Ismail
- Faculty of Pharmacy, Cairo University, Cairo 11796, Egypt
| | - Ahmed El-Bastawisy
- Department of Medical Oncology, National Cancer Institute, Cairo University, Cairo 11796, Egypt
| | - Abeer M Ashmawy
- Department of Cancer Biology, National Cancer Institute, Cairo University, Cairo 11796, Egypt
| | - Abdel-Rahman Zekri
- Department of Cancer Biology, National Cancer Institute, Cairo University, Cairo 11796, Egypt
| |
Collapse
|
22
|
Abstract
Identifying novel mechanisms, which are at the core of breast cancer biology, is of critical importance. Such mechanisms may explain response to treatment, reveal novel targets or drive detection assays. To uncover such novel mechanisms, we used survival analysis on gene expression datasets encompassing 1363 patients. By iterating over the compendia of genes, we screened for their significance as prognosis biomarkers and identified SUMO-specific protease 5 (SENP5) to significantly stratify patients into two survival groups across five unrelated tested datasets. According to these findings, low expression of SENP5 is associated with good prognosis among breast cancer patients. Following these findings, we analyzed SENP5 silencing and show it is followed by inhibition of anchorage-independence growth, proliferation, migration and invasion in breast cancer cell lines. We further show that these changes are conducted via regulation of TGFβRI levels. These data relate to recent reports about the SUMOylation of TGFβRI. Following TGFβRI changes in expression, we show that one of its target genes, MMP9, which plays a key role in degrading the extracellular matrix and contributes to TGFβ-induced invasion, is dramatically down regulated upon SENP5 silencing. This is the first report represents SENP5-TGFβ-MMP9 cascade and its mechanistic involvement in breast cancer.
Collapse
|
23
|
Alqumber MA, Dar SA, Haque S, Wahid M, Singh R, Akhter N. No Association of the TGF-β1 29T/C Polymorphism with Breast Cancer Risk in Caucasian and Asian Populations: Evidence from a Meta-Analysis Involving 55, 841 Subjects. Asian Pac J Cancer Prev 2014; 15:8725-34. [DOI: 10.7314/apjcp.2014.15.20.8725] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
24
|
Zou A, Lambert D, Yeh H, Yasukawa K, Behbod F, Fan F, Cheng N. Elevated CXCL1 expression in breast cancer stroma predicts poor prognosis and is inversely associated with expression of TGF-β signaling proteins. BMC Cancer 2014; 14:781. [PMID: 25344051 PMCID: PMC4221705 DOI: 10.1186/1471-2407-14-781] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 10/14/2014] [Indexed: 12/30/2022] Open
Abstract
Background CXCL1 is a chemotactic cytokine shown to regulate breast cancer progression and chemo-resistance. However, the prognostic significance of CXCL1 expression in breast cancer has not been fully characterized. Fibroblasts are important cellular components of the breast tumor microenvironment, and recent studies indicate that this cell type is a potential source of CXCL1 expression in breast tumors. The goal of this study was to further characterize the expression patterns of CXCL1 in breast cancer stroma, determine the prognostic significance of stromal CXCL1 expression, and identify factors affecting stromal CXCL1 expression. Methods Stromal CXCL1 protein expression was analyzed in 54 normal and 83 breast carcinomas by immunohistochemistry staining. RNA expression of CXCL1 in breast cancer stroma was analyzed through data mining in http://www.Oncomine.org. The relationships between CXCL1 expression and prognostic factors were analyzed by univariate analysis. Co-immunofluorescence staining for CXCL1, α-Smooth Muscle Actin (α-SMA) and Fibroblast Specific Protein 1 (FSP1) expression was performed to analyze expression of CXCL1 in fibroblasts. By candidate profiling, the TGF-β signaling pathway was identified as a regulator of CXCL1 expression in fibroblasts. Expression of TGF-β and SMAD gene products were analyzed by immunohistochemistry and data mining analysis. The relationships between stromal CXCL1 and TGF-β signaling components were analyzed by univariate analysis. Carcinoma associated fibroblasts isolated from MMTV-PyVmT mammary tumors were treated with recombinant TGF-β and analyzed for CXCL1 promoter activity by luciferase assay, and protein secretion by ELISA. Results Elevated CXCL1 expression in breast cancer stroma correlated with tumor grade, disease recurrence and decreased patient survival. By co-immunofluorescence staining, CXCL1 expression overlapped with expression of α-SMA and FSP1 proteins. Expression of stromal CXCL1 protein expression inversely correlated with expression of TGF-β signaling components. Treatment of fibroblasts with TGF-β suppressed CXCL1 secretion and promoter activity. Conclusions Increased CXCL1 expression in breast cancer stroma correlates with poor patient prognosis. Furthermore, CXCL1 expression is localized to α-SMA and FSP1 positive fibroblasts, and is negatively regulated by TGF-β signaling. These studies indicate that decreased TGF-β signaling in carcinoma associated fibroblasts enhances CXCL1 expression in fibroblasts, which could contribute to breast cancer progression. Electronic supplementary material The online version of this article (doi:10.1186/1471-2407-14-781) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Nikki Cheng
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| |
Collapse
|
25
|
Addison CL, Pond GR, Zhao H, Mazzarello S, Vandermeer L, Goldstein R, Amir E, Clemons M. Effects of de-escalated bisphosphonate therapy on bone turnover biomarkers in breast cancer patients with bone metastases. SPRINGERPLUS 2014; 3:577. [PMID: 25332877 PMCID: PMC4194305 DOI: 10.1186/2193-1801-3-577] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 09/09/2014] [Indexed: 01/10/2023]
Abstract
While de-escalation of bisphosphonates from 4 to 12-weekly dosing has been shown to be clinically non-inferior to standard dosing, there is evidence the de-escalation is associated with increased bone turnover biomarkers. Here we evaluated the effect of de-escalated dosing on a panel of biomarkers and determined their association with incidence of skeletal related events (SREs) in breast cancer patients with ‘low risk’ bone metastases. As part of a pilot randomized trial, women with baseline C-telopeptide levels <600 ng/L after >3 months of 3–4 weekly pamidronate were randomized to continue pamidronate every 4 weeks or de-escalation to 12-weekly treatment. Serum was analysed for bone biomarkers (C-telopeptide, N-telopeptide, bone-specific alkaline phosphatase, transforming growth factor-β, procollagen type 1 N-propeptide, activinA and bone sialoprotein) using ELISA. The associations between changes in biomarkers, pain scores and SREs were assessed by univariable logistic regression. Numerical increases in all biomarkers were observed between baseline and 12 weeks but were of higher magnitude in the de-escalated arm. Pain scores in the de-escalated treatment arm showed a greater magnitude of pain reduction from baseline to 12 weeks. Neither baseline levels nor changes in biomarkers from baseline to 12 weeks on treatment were associated with on study SREs. Baseline pain as measured by the FACT-BP was associated with increased risk of SRE. In conclusion, biomarkers of bone activity do not appear to predict for SREs in ‘low risk’ cohorts. However, baseline bone pain appears to be associated with SRE occurrence, a finding which warrants evaluation in larger cohorts.
Collapse
Affiliation(s)
- Christina L Addison
- Program for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON Canada ; Department of Medicine, University of Ottawa, Ottawa, ON Canada ; Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON Canada
| | - Gregory R Pond
- Department of Oncology, McMaster University, Hamilton, Canada
| | - Huijun Zhao
- Program for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON Canada
| | - Sasha Mazzarello
- Program for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON Canada
| | - Lisa Vandermeer
- Program for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON Canada
| | | | - Eitan Amir
- Princess Margaret Hospital, Toronto, ON Canada
| | - Mark Clemons
- Program for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON Canada ; Department of Medicine, University of Ottawa, Ottawa, ON Canada
| |
Collapse
|
26
|
Wild-type p53 inhibits pro-invasive properties of TGF-β3 in breast cancer, in part through regulation of EPHB2, a new TGF-β target gene. Breast Cancer Res Treat 2014; 148:7-18. [DOI: 10.1007/s10549-014-3147-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 09/19/2014] [Indexed: 01/08/2023]
|
27
|
Ciftci R, Tas F, Yasasever CT, Aksit E, Karabulut S, Sen F, Keskin S, Kilic L, Yildiz I, Bozbey HU, Duranyildiz D, Vatansever S. High serum transforming growth factor beta 1 (TGFB1) level predicts better survival in breast cancer. Tumour Biol 2014; 35:6941-8. [PMID: 24740564 DOI: 10.1007/s13277-014-1932-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 04/03/2014] [Indexed: 12/24/2022] Open
Abstract
The transforming growth factor beta 1 (TGFB1) is a regulatory cytokine with both tumor suppressor and tumor-promoting effects in breast cancer (BC) cell lines and tissue. Data about level of circulating TGFB1 and its prognostic significance in BC patients is conflicting. The objective of this study is to determine the clinical significance of the serum TGFB1 levels in BC patients. We enrolled 96 female patients with histopathologically diagnosed BC who did not receive chemotherapy (CT) or radiotherapy. Serum TGFB1 levels were measured by ELISA method and compared with 30 healthy controls. The mean serum TGFB1 level of BC patients was significantly higher than controls (0.08 vs. 0.04 ng/ml, p < 0.001). There was no significant difference according to known disease-related clinicopathological or laboratory parameters. Serum TGFB1 level had a significant impact on overall survival in both univariate (p = 0.01) and multivariate analysis (p = 0.013). Serum TGFB1 level is elevated in BC patients and has a favorable prognostic value. However, it has no predictive role on CT response.
Collapse
Affiliation(s)
- Rumeysa Ciftci
- Medical Oncology Department, Institute of Oncology, Istanbul University, Capa, Istanbul, Turkey,
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
A phase II, multicentre trial evaluating the efficacy of de-escalated bisphosphonate therapy in metastatic breast cancer patients at low-risk of skeletal-related events. Breast Cancer Res Treat 2014; 144:615-24. [PMID: 24638849 PMCID: PMC3962742 DOI: 10.1007/s10549-014-2906-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 03/03/2014] [Indexed: 11/27/2022]
Abstract
The optimal frequency of intravenous (IV) bisphosphonate administration is unclear. We thus performed a study evaluating the effects of switching from 3–4 to 12 weekly therapy in patients with biochemically defined low-risk bone metastases. Patients with serum C-telopeptide (CTx) levels ≤600 ng/L after ≥3 months of 3–4 weekly IV pamidronate were switched to 12 weekly therapy for 48 weeks. Primary endpoint was the proportion of patients maintaining CTx levels in the lower-risk range. All endpoints (serum CTx and bone-specific alkaline phosphatase (BSAP), skeletal-related events (SREs) and self-reported pain) were measured at baseline, 6, 12, 24, 36 and 48 weeks. Treatment failure was defined as biochemical failure (CTx > 600 ng/L) or a SRE. Exploratory biomarkers including; serum TGF-β, activin-A, bone sialoprotein (BSP), procollagen type 1 N-terminal propeptide and urinary N-telopeptide (NTx) were assessed at baseline as predictors for failure to complete treatment. Seventy-one patients accrued and 43 (61 %) completed 48 weeks of de-escalated therapy. Reasons for failure to complete treatment included; biochemical failure (CTx > 600 ng/L) (n = 10, 14.1 %), on-study SRE (n = 9, 12.7 %), disease progression (n = 7, 9.9 % including death from disease [n = 1, 1.4 %]) or patient choice (n = 2, 2.8 %). Elevated baseline levels of CTx, BSAP, NTx and BSP were associated with treatment failure. The majority of patients in this biochemically defined low-risk population could switch from 3–4 weekly to 12 weekly bisphosphonate therapy with no effect on CTx levels or SREs during the 48 week study. Larger trials are required to assess the roles of biomarkers as predictors of adequacy of de-escalated therapy.
Collapse
|
29
|
Loss of Dab2 expression in breast cancer cells impairs their ability to deplete TGF-β and induce Tregs development via TGF-β. PLoS One 2014; 9:e91709. [PMID: 24638085 PMCID: PMC3956763 DOI: 10.1371/journal.pone.0091709] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Accepted: 02/14/2014] [Indexed: 11/19/2022] Open
Abstract
Dab2 is a multifunctional adapter protein which is frequently under-expressed in a variety of cancers. It is implicated in many critical functions, including several signaling pathways, cell arrangement, differentiation of stem cells, and receptor endocytosis. Transforming growth factor-β (TGF-β) is a secreted multifunctional protein that controls several developmental processes and pathogenesis of many diseases. It has been documented that Dab2 played an important role in TGF-β receptors endocytosis. Here, we present evidence that re-expression of Dab2 in SK-BR-3 cell partially restored its ability to deplete TGF-β in surrounding medium by normalizing the trafficking of TGF-β receptors. We also demonstrate that the difference in TGF-β depletions produced by Dab2 expression was sufficient to impact on the conversion of naive CD4+ T cells to regulatory T cells (Tregs), and thus inhibited the proliferation of T cells. This work revealed a critical result that breast cancer cell was deficient in Dab2 expression and related receptor endocytosis-mediated TGF-β depletion, which may contribute to the accumulation of TGF-β in tumor microenvironment and the induction of immune tolerance.
Collapse
|
30
|
Slattery ML, Lundgreen A, Stern MC, Hines L, Wolff RK, Giuliano AR, Baumgartner KB, John EM. The influence of genetic ancestry and ethnicity on breast cancer survival associated with genetic variation in the TGF-β-signaling pathway: The Breast Cancer Health Disparities Study. Cancer Causes Control 2013; 25:293-307. [PMID: 24337772 DOI: 10.1007/s10552-013-0331-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Accepted: 12/05/2013] [Indexed: 10/25/2022]
Abstract
The TGF-β signaling pathway regulates cellular proliferation and differentiation. We evaluated genetic variation in this pathway, its association with breast cancer survival, and survival differences by genetic ancestry and self-reported ethnicity. The Breast Cancer Health Disparities Study includes participants from the 4-Corners Breast Cancer Study (n = 1,391 cases) and the San Francisco Bay Area Breast Cancer Study (n = 946 cases) who have been followed for survival. We evaluated 28 genes in the TGF-β signaling pathway using a tagSNP approach. Adaptive rank truncated product (ARTP) was used to test the gene and pathway significance by Native American (NA) ancestry and by self-reported ethnicity (non-Hispanic white (NHW) and Hispanic/NA). Genetic variation in the TGF-β signaling pathway was associated with overall breast cancer survival (P ARTP = 0.05), especially for women with low NA ancestry (P ARTP = 0.007) and NHW women (P ARTP = 0.006). BMP2, BMP4, RUNX1, and TGFBR3 were significantly associated with breast cancer survival overall (P ARTP = 0.04, 0.02, 0.002, and 0.04, respectively). Among women with low NA, ancestry associations were as follows: BMP4 (P ARTP = 0.007), BMP6 (P ARTP = 0.001), GDF10 (P ARTP = 0.05), RUNX1 (P ARTP = 0.002), SMAD1 (P ARTP = 0.05), and TGFBR2 (P ARTP = 0.02). A polygenic risk model showed that women with low NA ancestry and high numbers of at-risk alleles had twice the risk of dying from breast cancer as did women with high NA ancestry. Our data suggest that genetic variation in the TGF-β signaling pathway influences breast cancer survival. Associations were similar when the analyses were stratified by genetic ancestry or by self-reported ethnicity.
Collapse
Affiliation(s)
- Martha L Slattery
- Department of Medicine, University of Utah, 383 Colorow, Salt Lake City, UT, 84108, USA,
| | | | | | | | | | | | | | | |
Collapse
|
31
|
YKL-40 expression could be a poor prognostic marker in the breast cancer tissue. Tumour Biol 2013; 35:277-86. [PMID: 23918300 DOI: 10.1007/s13277-013-1036-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Accepted: 07/18/2013] [Indexed: 02/01/2023] Open
Abstract
YKL-40 is a glycoprotein involved in cellular growth, migration, and the inflammatory process. Elevation in serum levels of YKL-40 has been associated with worse prognosis in various cancers, including breast cancer. Given that the clinical significance of YKL-40 expression in breast cancer tissue is unclear, we aimed to determine the prognostic value of YKL-40 expression in breast cancer tissue using immunohistochemistry. We performed tissue microarray (TMA) analysis of 425 breast cancer tissues collected during operation. Immunohistochemical staining was performed to measure expression of YKL-40 and several breast cancer biomarkers, such as aldehyde dehyadrogenase1, TGF-beta, and Gli-1 as well as hormonal receptor and Her-2/neu status. Statistical analysis of the relationship of YKL-40 expression with clinicopathological characteristics was performed for 390 TMA samples. YKL-40 was expressed to varying degrees in 84.9% of breast cancer tissues. YKL-40 expression was correlated with estrogen receptor and progesterone receptor negativity and was positively correlated with TGF-beta and Gli-1 expression. Strong YKL-40 expression was associated with a larger proportion of Her-2/neu-enriched and basal-like tumors. The results of this study demonstrate that YKL-40 expression in breast cancer tissues is associated with hormone receptor negativity and Her-2/neu-enriched molecular subtypes of breast cancer, and therefore could be considered a poor prognostic predictor.
Collapse
|
32
|
Naber HPH, Drabsch Y, Snaar-Jagalska BE, ten Dijke P, van Laar T. Snail and Slug, key regulators of TGF-β-induced EMT, are sufficient for the induction of single-cell invasion. Biochem Biophys Res Commun 2013; 435:58-63. [PMID: 23618854 DOI: 10.1016/j.bbrc.2013.04.037] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Accepted: 04/04/2013] [Indexed: 11/24/2022]
Abstract
TGF-β plays a dual role in cancer; in early stages it inhibits tumor growth, whereas later it promotes invasion and metastasis. TGF-β is thought to be pro-invasive by inducing epithelial-to-mesenchymal transition (EMT) via induction of transcriptional repressors, including Slug and Snail. In this study, we investigated the role of Snail and Slug in TGF-β-induced invasion in an in vitro invasion assay and in an embryonic zebrafish xenograft model. Ectopic expression of Slug or Snail promoted invasion of single, rounded amoeboid cells in vitro. In an embryonic zebrafish xenograft model, forced expression of Slug and Snail promoted single cell invasion and metastasis. Slug and Snail are sufficient for the induction of single-cell invasion in an in vitro invasion assay and in an embryonic zebrafish xenograft model.
Collapse
Affiliation(s)
- Hildegonda P H Naber
- Department of Molecular Cell Biology and Centre for Biomedical Genetics, Leiden University Medical Centre, Leiden, The Netherlands
| | | | | | | | | |
Collapse
|
33
|
de Kruijf EM, Dekker TJA, Hawinkels LJAC, Putter H, Smit VTHBM, Kroep JR, Kuppen PJK, van de Velde CJH, Ten Dijke P, Tollenaar RAEM, Mesker WE. The prognostic role of TGF-β signaling pathway in breast cancer patients. Ann Oncol 2013; 24:384-390. [PMID: 23022998 DOI: 10.1093/annonc/mds333] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/07/2023] Open
Abstract
BACKGROUND The transforming growth factor-β (TGF-β) pathway has dual effects on tumor growth. Seemingly, discordant results have been published on the relation between TGF-β signaling markers and prognosis in breast cancer. Improved prognostic information for breast cancer patients might be obtained by assessing interactions among TGF-β signaling biomarkers. PATIENTS AND METHODS The expression of nuclear Smad4, nuclear phosphorylated-Smad2 (p-Smad2), and the membranous expression of TGF-β receptors I and II (TβRI and TβRII) was determined on a tissue microarray of 574 breast carcinomas. Tumors were stratified according to the Smad4 expression in combination with p-Smad2 expression or Smad4 in combination with the expression of both TGF-β receptors. RESULTS Tumors with high expression of TβRII, TβRI and TβRII, and p-Smad2 (P = 0.018, 0.005, and 0.022, respectively), and low expression of Smad4 (P = 0.005) had an unfavorable prognosis concerning progression-free survival. Low Smad4 expression combined with high p-Smad2 expression or low expression of Smad4 combined with high expression of both TGF-β receptors displayed an increased hazard ratio of 3.04 [95% confidence interval (CI) 1.390-6.658] and 2.20 (95% CI 1.464-3.307), respectively, for disease relapse. CONCLUSIONS Combining TGF-β biomarkers provides prognostic information for patients with stage I-III breast cancer. This can identify patients at increased risk for disease recurrence that might therefore be candidates for additional treatment.
Collapse
Affiliation(s)
| | - T J A Dekker
- Departments of Surgery; Departments of Medical Oncology
| | | | | | - V T H B M Smit
- Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | - P Ten Dijke
- Molecular Cell Biology and Centre for Biomedical Genetics; Ludwig Institute for Cancer Research, Uppsala University, Uppsala, Sweden
| | | | | |
Collapse
|
34
|
Khan SA, Joyce J, Tsuda T. Quantification of active and total transforming growth factor-β levels in serum and solid organ tissues by bioassay. BMC Res Notes 2012; 5:636. [PMID: 23151377 PMCID: PMC3556312 DOI: 10.1186/1756-0500-5-636] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Accepted: 10/29/2012] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Transforming growth factor-β (TGF-β) is a multi-factorial peptide growth factor that has a vital role in the regulation of cell growth, differentiation, inflammation, and tissue repair. Quantification of biologically active TGF-β levels in tissues is crucial to illustrate mechanisms involved in various physiological and pathological processes, but direct measurement of bioactive TGF-β level in the tissue has been hampered by lack of reliable methods. Here, we introduced mink lung epithelial cell bioassay to quantify both active and total TGF-β levels in serum and protein lysates from solid organs in the mouse model. FINDINGS Mink lung epithelial cells were stably transfected with plasminogen activator inhibitor-1 promoter/luciferase construct, in which bioactive TGF-β level was represented by luciferase activity. Serum total TGF-β levels were comparable between the bioassay and enzyme-linked immunosorbent assay (ELISA), but active TGF-β levels measured by ELISA were significantly lower than those obtained by the bioassay. Active and total TGF-β levels in the solid organs including heart, liver, and kidney were also measured. Total TGF-β levels were relatively comparable among these organs, but active TGF-β levels were slightly higher in hearts and kidneys than in livers. Positive luciferase activities in the bioassay were almost completely inhibited by adding pan-TGF-β neutralizing antibodies, suggesting its high specificity to bioactive TGF-β. We also measured myocardial TGF-β levels after myocardial infarction and sham control by the bioassay, and compared the values with those obtained by ELISA. The bioassay demonstrated that both active and total tissue TGF-β levels were significantly higher in post-myocardial infarction than in sham myocardium. ELISA was markedly less sensitive in detecting both active and total TGF-β levels than our bioassay and failed to show any statistically significant difference in TGF-β levels between myocardial infarction and sham myocardium. CONCLUSIONS Our data suggested that the bioassay was significantly more sensitive than ELISA in detecting active TGF-β in serum and both active and total TGF-β in solid organ tissues. The bioassay will be useful in investigating TGF-β profile in various solid organs in physiological and pathological conditions.
Collapse
Affiliation(s)
- Shaukat A Khan
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children, 1600 Rockland Rd, Wilmington, DE, 19803, USA
| | - Jennifer Joyce
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children, 1600 Rockland Rd, Wilmington, DE, 19803, USA
| | - Takeshi Tsuda
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children, 1600 Rockland Rd, Wilmington, DE, 19803, USA
- Nemours Cardiac Center, Alfred I. duPont Hospital for Children, 1600 Rockland Rd, Wilmington, DE, 19803, USA
| |
Collapse
|
35
|
Kubiczkova L, Sedlarikova L, Hajek R, Sevcikova S. TGF-β - an excellent servant but a bad master. J Transl Med 2012; 10:183. [PMID: 22943793 PMCID: PMC3494542 DOI: 10.1186/1479-5876-10-183] [Citation(s) in RCA: 387] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Accepted: 08/28/2012] [Indexed: 12/13/2022] Open
Abstract
The transforming growth factor (TGF-β) family of growth factors controls an immense number of cellular responses and figures prominently in development and homeostasis of most human tissues. Work over the past decades has revealed significant insight into the TGF-β signal transduction network, such as activation of serine/threonine receptors through ligand binding, activation of SMAD proteins through phosphorylation, regulation of target genes expression in association with DNA-binding partners and regulation of SMAD activity and degradation. Disruption of the TGF-β pathway has been implicated in many human diseases, including solid and hematopoietic tumors. As a potent inhibitor of cell proliferation, TGF-β acts as a tumor suppressor; however in tumor cells, TGF-β looses anti-proliferative response and become an oncogenic factor. This article reviews current understanding of TGF-β signaling and different mechanisms that lead to its impairment in various solid tumors and hematological malignancies.
Collapse
Affiliation(s)
- Lenka Kubiczkova
- Babak Myeloma Group, Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Brno, 625 00, Czech Republic
| | | | | | | |
Collapse
|
36
|
Khajah MA, Al Saleh S, Mathew PM, Luqmani YA. Differential effect of growth factors on invasion and proliferation of endocrine resistant breast cancer cells. PLoS One 2012; 7:e41847. [PMID: 22860018 PMCID: PMC3408495 DOI: 10.1371/journal.pone.0041847] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Accepted: 06/29/2012] [Indexed: 01/27/2023] Open
Abstract
We have established several breast cancer cell lines that exhibit a permanent ER-depleted phenotype, induced by shRNA transfection of MCF-7 cells, which afford a useful model for studying acquired endocrine resistance. Previously we showed that MDA-231 as well as ER-silenced cells could invade through simulated extracellular matrix components. However, the contribution of individual serum components responsible for cell invasion was not determined. In the present study, an under-agarose gel assay was used to quantitatively assess the invasive movement of two ER-silenced cell lines (pII and YS2.5) in comparison to the parental MCF-7, the ER negative MDA-231, and normal HBL100 cells, as well as a line that was ER-shRNA transfected but failed to exhibit ER down-regulation (YS1.2). We also examined the effect of the growth factors EGF, IGF-1, TGFβ, PDGFC and RANTES on pII cell invasion and proliferation. All breast cancer cell lines which had reduced ER expression exhibited a serum-dependent invasive ability related to the degree of induced ER loss. TGFβ treatment inhibited pII cell proliferation and enhanced their invasive ability but at a relatively high dose. IGF-1 and EGF enhanced pII cell proliferation, with the latter playing the major role in promoting cell invasion. PDGFC did not affect either process although it is highly expressed in pII cells. Differential effects were observed on activation of Akt and ERK1/2 suggesting their involvement as intracellular mediators of EGF induced invasion, in part through the regulation of matrix metalloproteinase activity. Targeting EGF receptor tyrosine kinase activity by erlotinib resulted in significant inhibition of both pII cell proliferation and directional invasion towards EGF suggesting that this drug has potential therapeutic usefulness for preventing spread of particularly endocrine resistant breast cancer.
Collapse
|
37
|
TGF-β1 modulates the homeostasis between MMPs and MMP inhibitors through p38 MAPK and ERK1/2 in highly invasive breast cancer cells. BMC Cancer 2012; 12:26. [PMID: 22260435 PMCID: PMC3277461 DOI: 10.1186/1471-2407-12-26] [Citation(s) in RCA: 151] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Accepted: 01/19/2012] [Indexed: 11/21/2022] Open
Abstract
Background Metastasis is the main factor responsible for death in breast cancer patients. Matrix metalloproteinases (MMPs) and their inhibitors, known as tissue inhibitors of MMPs (TIMPs), and the membrane-associated MMP inhibitor (RECK), are essential for the metastatic process. We have previously shown a positive correlation between MMPs and their inhibitors expression during breast cancer progression; however, the molecular mechanisms underlying this coordinate regulation remain unknown. In this report, we investigated whether TGF-β1 could be a common regulator for MMPs, TIMPs and RECK in human breast cancer cell models. Methods The mRNA expression levels of TGF-β isoforms and their receptors were analyzed by qRT-PCR in a panel of five human breast cancer cell lines displaying different degrees of invasiveness and metastatic potential. The highly invasive MDA-MB-231 cell line was treated with different concentrations of recombinant TGF-β1 and also with pharmacological inhibitors of p38 MAPK and ERK1/2. The migratory and invasive potential of these treated cells were examined in vitro by transwell assays. Results In general, TGF-β2, TβRI and TβRII are over-expressed in more aggressive cells, except for TβRI, which was also highly expressed in ZR-75-1 cells. In addition, TGF-β1-treated MDA-MB-231 cells presented significantly increased mRNA expression of MMP-2, MMP-9, MMP-14, TIMP-2 and RECK. TGF-β1 also increased TIMP-2, MMP-2 and MMP-9 protein levels but downregulated RECK expression. Furthermore, we analyzed the involvement of p38 MAPK and ERK1/2, representing two well established Smad-independent pathways, in the proposed mechanism. Inhibition of p38MAPK blocked TGF-β1-increased mRNA expression of all MMPs and MMP inhibitors analyzed, and prevented TGF-β1 upregulation of TIMP-2 and MMP-2 proteins. Moreover, ERK1/2 inhibition increased RECK and prevented the TGF-β1 induction of pro-MMP-9 and TIMP-2 proteins. TGF-β1-enhanced migration and invasion capacities were blocked by p38MAPK, ERK1/2 and MMP inhibitors. Conclusion Altogether, our results support that TGF-β1 modulates the mRNA and protein levels of MMPs (MMP-2 and MMP-9) as much as their inhibitors (TIMP-2 and RECK). Therefore, this cytokine plays a crucial role in breast cancer progression by modulating key elements of ECM homeostasis control. Thus, although the complexity of this signaling network, TGF-β1 still remains a promising target for breast cancer treatment.
Collapse
|
38
|
Todorović-Raković N, Milovanović J, Nikolić-Vukosavljević D. TGF-β and its coreceptors in cancerogenesis: an overview. Biomark Med 2011; 5:855-863. [PMID: 22103622 DOI: 10.2217/bmm.11.59] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Besides signaling serine/threonine kinases, such as TGF-β receptors I and II, the TGF-β pathway involves several auxiliary receptors or coreceptors. Recent studies show that these coreceptors, particulary endoglin and β-glycan, have greater significance than previously thought. They regulate the availability of ligands to the key receptors, as well as their interaction and response, which could be variable and context-dependent. Understanding their true mechanism of action is important for delineating the complexity of the entire TGF-β signaling pathway. This is especially important in the context of cancerogenesis, because of therapeutic possibilities to manipulate the TGF-β system.
Collapse
Affiliation(s)
- Nataša Todorović-Raković
- Department of Experimental Oncology, Institute for Oncology & Radiology of Serbia, Pasterova 14, Belgrade, Serbia
| | | | | |
Collapse
|
39
|
BMP-7 inhibits TGF-β-induced invasion of breast cancer cells through inhibition of integrin β(3) expression. Cell Oncol (Dordr) 2011; 35:19-28. [PMID: 21935711 PMCID: PMC3268977 DOI: 10.1007/s13402-011-0058-0] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2011] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND The transforming growth factor (TGF)-β superfamily comprises cytokines such as TGF-β and Bone Morphogenetic Proteins (BMPs), which have a critical role in a multitude of biological processes. In breast cancer, high levels of TGF-β are associated with poor outcome, whereas inhibition of TGF-β-signaling reduces metastasis. In contrast, BMP-7 inhibits bone metastasis of breast cancer cells. METHODS In this study, we investigated the effect of BMP-7 on TGF-β-induced invasion in a 3 dimensional invasion assay. RESULTS BMP-7 inhibited TGF-β-induced invasion of the metastatic breast cancer cell line MCF10CA1a, but not of its premalignant precursor MCF10AT in a spheroid invasion model. The inhibitory effect appears to be specific for BMP-7, as its closest homolog, BMP-6, did not alter the invasion of MCF10CA1a spheroids. To elucidate the mechanism by which BMP-7 inhibits TGF-β-induced invasion, we analyzed invasion-related genes. BMP-7 inhibited TGF-β-induced expression of integrin α(v)β(3) in the spheroids. Moreover, targeting of integrins by a chemical inhibitor or knockdown of integrin β(3) negatively affected TGF-β-induced invasion. On the other hand, overexpression of integrin β(3) counteracted the inhibitory effect of BMP7 on TGF-β-induced invasion. CONCLUSION Thus, BMP-7 may exert anti-invasive actions by inhibiting TGF-β-induced expression of integrin β(3).
Collapse
|
40
|
Transforming growth factor β signaling pathway associated gene polymorphisms may explain lower breast cancer risk in western Indian women. PLoS One 2011; 6:e21866. [PMID: 21829601 PMCID: PMC3150347 DOI: 10.1371/journal.pone.0021866] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2011] [Accepted: 06/08/2011] [Indexed: 11/19/2022] Open
Abstract
Transforming growth factor β1 (TGFB1) T29C and TGF β receptor type 1 (TGFBR1) 6A/9A polymorphisms have been implicated in the modulation of risk for breast cancer in Caucasian women. We analyzed these polymorphisms and combinations of their genotypes, in pre menopausal breast cancer patients (N = 182) and healthy women (N = 236) from western India as well as in breast cancer patients and healthy women from the Parsi community (N = 48 & 171, respectively). Western Indian women were characterized by a higher frequency of TGFB1*C allele of the TGF β T29C polymorphism (0.48 vs 0.44) and a significantly lower frequency of TGFBR1*6A allele of the TGFBR1 6A/9A polymorphism (0.02 vs 0.068, p<0.01) as compared to healthy Parsi women. A strong protective effect of TGFB1*29C allele was seen in younger western Indian women (<40 yrs; OR = 0.45, 95% CI 0.25-0.81). Compared to healthy women, the strikingly higher frequencies of low or intermediate TGF β signalers in patients suggested a strong influence of the combination of these genotypes on the risk for breast cancer in Parsi women (for intermediate signalers, OR = 4.47 95%CI 1.01-19.69). The frequency of low signalers in Parsi healthy women, while comparable to that reported in Europeans and Americans, was three times higher than that in healthy women from western India (10.6% vs 3.3%, p<0.01). These observations, in conjunction with the low incidence rate of breast cancer in Indian women compared to White women, raise a possibility that the higher frequency of TGFB1*29C allele and lower frequency of TGFBR1*6A allele may represent important genetic determinants that together contribute to a lower risk of breast cancer in western Indian women.
Collapse
|
41
|
Abstract
Transforming growth factor-β (TGF-β) is a multifunctional cytokine, with important roles in maintaining tissue homeostasis. TGF-β signals via transmembrane serine/threonine kinase receptors and intracellular Smad transcriptional regulators. Perturbed TGF-β signaling has been implicated in a large variety of pathological conditions. Increased TGF-β levels have been found in patients with cancer, fibrosis, and systemic sclerosis, and were correlated with disease severity. In cancer, TGF-β mediates tumor invasion and metastasis by affecting both tumor cells and the tumor microenvironment including fibroblast activation and immune suppression. Furthermore, TGF-β is a strong stimulator of extracellular matrix deposition. On the basis of these observations, small molecule inhibitors of the TGF-β receptor kinases, neutralizing antibodies that interfere with ligand?receptor interactions, antisense oligonucleotides reducing TGF-β expression, and soluble receptor ectodomains that sequester TGF-β have been developed to intervene with excessive TGF-β signaling activity in the aforementioned disorders. Here, we review the current state of anti-TGF-β therapy in clinical trials.
Collapse
Affiliation(s)
- Lukas J A C Hawinkels
- Department of Molecular Cell Biology and Centre for Biomedical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | | |
Collapse
|
42
|
Capietto AH, Martinet L, Fournié JJ. Stimulated γδ T cells increase the in vivo efficacy of trastuzumab in HER-2+ breast cancer. THE JOURNAL OF IMMUNOLOGY 2011; 187:1031-8. [PMID: 21670311 DOI: 10.4049/jimmunol.1100681] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
One fourth of women with HER-2(+) metastatic breast carcinoma are treated with a combination regimen with trastuzumab, but the frequent resistance to this Ab requires definition of new means to improve its bioactivity. The mechanisms of action of trastuzumab involve several pathways including Ab-dependent cellular cytotoxicity. Because human γδ T lymphocytes mediate Ab-dependent cellular cytotoxicity and can be activated further by phosphoantigens, these cells are prone to improve the efficacy of Abs, as recently demonstrated for CD20(+) B cell lymphomas. Whether this concept applies as well with carcinomas remained to be demonstrated in vivo, however. In this study, we asked whether a combination of trastuzumab and phosphoantigen-stimulated γδ lymphocytes increases the efficacy of trastuzumab against HER-2(+) breast carcinoma cell lines in vivo. We report that repeated infusions of this combination had a better efficacy than that of trastuzumab alone against HER-2(+) mammary carcinoma xenografts in mice. In these models, reduction of tumor growth was observed together with trastuzumab opsonization of HER-2(+) cells and tumor infiltration by γδ lymphocytes. In addition in humans, the mammary carcinomas of 27 of 30 patients showed significant γδ T cell infiltrates. Altogether, these findings indicate that combination of trastuzumab and stimulated γδ cells represents a new strategy to improve the efficacy of Herceptin (trastuzumab) in HER-2(+) breast cancer.
Collapse
Affiliation(s)
- Aude-Hélène Capietto
- INSERM, Unité Mixte de Recherche 1037, Cancer Research Center of Toulouse, 31024 Toulouse Cedex, France
| | | | | |
Collapse
|
43
|
Babyshkina N, Malinovskaya E, Stakheyeva M, Volkomorov V, Slonimskaya E, Maximov V, Cherdyntseva N. Association of functional -509C>T polymorphism in the TGF-β1 gene with infiltrating ductal breast carcinoma risk in a Russian Western Siberian population. Cancer Epidemiol 2011; 35:560-3. [PMID: 21470928 DOI: 10.1016/j.canep.2011.02.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2010] [Revised: 01/31/2011] [Accepted: 02/04/2011] [Indexed: 10/18/2022]
Abstract
BACKGROUND Transforming growth factor β1 (TGF-β1) is a multifunctional cytokine that plays an important role in human mammary carcinogenesis. The purpose of this study was to investigate the association between -509C>T single nucleotide polymorphism (SNP) of the TGF-β1 gene and infiltrating ductal breast carcinoma risk in Russian patients of Western Siberian region. MATERIALS AND METHODS Blood samples collected from 218 women with histologically confirmed infiltrating ductal breast carcinoma and 290 healthy female controls were analyzed through polymerase chain reaction-restriction fragment length polymorphism methods. RESULTS The -509TT genotype was significantly associated with a decreased risk for ductal breast carcinoma (OR=0.47, CI: 0.26-0.82, P=0.004). Similarly, the -509T was significantly less in ductal breast cancer patients (34.4%) than in control individuals (41.6%; OR=0.74, CI: 0.57-0.96, P=0.02). With the exception of association between the -509TT genotype and large tumor size (P=0.01), there was no significant association between the studied polymorphism and clinicopathological characteristics. CONCLUSION The results of this study suggest that polymorphism of TGF-β1 -509C>T gene may modify individual susceptibility to infiltrating ductal breast carcinoma in Russian women of Western Siberian region.
Collapse
Affiliation(s)
- Nataliya Babyshkina
- Department of Experimental Oncology, Cancer Research Institute of Siberian Branch of Russian Academy of Medical Sciences, Tomsk 634001, Russian Federation.
| | | | | | | | | | | | | |
Collapse
|
44
|
Disis ML. Immunologic biomarkers as correlates of clinical response to cancer immunotherapy. Cancer Immunol Immunother 2011; 60:433-42. [PMID: 21221967 PMCID: PMC11028861 DOI: 10.1007/s00262-010-0960-8] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2010] [Accepted: 12/10/2010] [Indexed: 01/08/2023]
Abstract
Over the last few years, several newly developed immune-based cancer therapies have been shown to induce clinical responses in significant numbers of patients. As a result, there is a need to identify immune biomarkers capable of predicting clinical response. If there were laboratory parameters that could define patients with improved disease outcomes after immunomodulation, product development would accelerate, optimization of existing immune-based treatments would be facilitated and patient selection for specific interventions might be optimized. Although there are no validated cancer immunologic biomarkers that are predictive of clinical response currently in widespread use, there is much published literature that has informed investigators as to which markers may be the most promising. Population-based studies of endogenous tumor immune infiltrates and gene expression analyses have identified specific cell populations and phenotypes of immune cells that are most likely to mediate anti-tumor immunity. Further, clinical trials of cancer vaccines and other cancer directed immunotherapy have identified candidate immunologic biomarkers that are statistically associated with beneficial clinical outcomes after immune-based cancer therapies. Biomarkers that measure the magnitude of the Type I immune response generated with immune therapy, epitope spreading, and autoimmunity are readily detected in the peripheral blood and, in clinical trials of cancer immunotherapy, have been associated with response to treatment.
Collapse
Affiliation(s)
- Mary L Disis
- Tumor Vaccine Group, Center for Translational Medicine in Women's Health, University of Washington, 815 Mercer Street, 2nd Floor, Box 358050, Seattle, WA 98195-8050, USA.
| |
Collapse
|
45
|
Butterfield LH, Disis ML, Khleif SN, Balwit JM, Marincola FM. Immuno-oncology biomarkers 2010 and beyond: perspectives from the iSBTc/SITC biomarker task force. J Transl Med 2010; 8:130. [PMID: 21138581 PMCID: PMC3014892 DOI: 10.1186/1479-5876-8-130] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2010] [Accepted: 12/07/2010] [Indexed: 11/22/2022] Open
Abstract
The International Society for Biological Therapy of Cancer (iSBTc, recently renamed the Society for Immunotherapy of Cancer, SITC) hosted a one-day symposium at the National Institutes of Health on September 30, 2010 to address development and application of biomarkers in cancer immunotherapy. The symposium, titled Immuno-Oncology Biomarkers 2010 and Beyond: Perspectives from the iSBTc/SITC Biomarker Task Force, gathered approximately 230 investigators equally from academia, industry and governmental/regulatory agencies from around the globe for panel discussions and presentations on the following topics: 1) immunologic monitoring: standardization and validation of assays; 2) correlation of immunity to biologic activity, clinical response and potency assays; 3) novel methodologies for assessing the immune landscape: clinical utility of novel technologies; and 4) recommendations on incorporation of biomarkers into the clinical arena. The presentations are summarized in this report; additional program information and slides are available online at the iSBTc/SITC website.
Collapse
Affiliation(s)
- Lisa H Butterfield
- Departments of Medicine, Surgery and Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | | | | |
Collapse
|
46
|
Dave B, Wynne R, Su Y, Korourian S, Chang JC, Simmen RCM. Enhanced mammary progesterone receptor-A isoform activity in the promotion of mammary tumor progression by dietary soy in rats. Nutr Cancer 2010; 62:774-82. [PMID: 20661826 DOI: 10.1080/01635581.2010.494334] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Dietary contribution to breast cancer risk, recurrence, and progression remains incompletely understood. Increased consumption of soy and soy isoflavones is associated with reduced mammary cancer susceptibility in women and in rodent models of carcinogenesis. In rats treated with N-methyl-N-nitrosourea, dietary intake of soy protein isolate (SPI) reduced mammary tumor occurrence but increased incidence of more invasive tumors in tumored rats, relative to the control diet casein. Here we evaluated whether mammary tumor progression in tumor-bearing rats lifetime exposed to SPI is associated with deregulated progesterone receptor (PR) isoform expression. In histologically normal mammary glands of rats with invasive ductal carcinoma lesions, PR-A protein levels were higher for SPI- than casein-fed rats, whereas PR-B was undetectable for both groups. Increased mammary PR-A expression was associated with higher transforming growth factor-beta1, stanniocalcin-1, and CD44 transcript levels; lower E-cadherin and estrogen receptor-alpha expression; and reduced apoptotic status in ductal epithelium. Serum progesterone (ng/ml) (CAS: 25.94 +/- 3.81; SPI: 13.19 +/- 2.32) and estradiol (pg/ml) (CAS: 27.9 +/- 4.49; SPI: 68.48 +/- 23.87) levels differed with diet. However, sera from rats of both diet groups displayed comparable mammosphere-forming efficiency in human MCF-7 cells. Thus, soy-rich diets may influence the development of more aggressive tumors by enhancing PR-A-dependent signaling in premalignant breast tissues.
Collapse
Affiliation(s)
- Bhuvanesh Dave
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, and Arkansas Children's Nutrition Center, Little Rock, AR 72202, USA
| | | | | | | | | | | |
Collapse
|
47
|
Okimura A, Terada N, Hata M, Kawahara K, Iwasaki T, Oota M, Hirano H. Expression of adhesion molecules and transforming growth factor-β in pleomorphic carcinomas of the lung. Oncol Lett 2010; 1:959-965. [PMID: 22870095 DOI: 10.3892/ol.2010.166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2010] [Accepted: 08/09/2010] [Indexed: 11/05/2022] Open
Abstract
Pleomorphic carcinoma (PC) of the lung consists of an epithelial component showing the histology of poorly differentiated non-small cell carcinoma of the lung and a sarcomatous component, that is more aggressive compared to non-small cell carcinoma of the lung. To determine the differences between an epithelial component of PC and poorly differentiated non-small cell carcinoma, the expression of adhesion molecules (E-cadherin, β-catenin and N-cadherin) and transforming growth factor-β (TGF-β) was compared immunohistochemically among 14 poorly differentiated adenocarcinomas of the lung (PDAs) and 14 PCs of the lung, with an epithelial component, showing the histology of PDA. Expression levels of E-cadherin and β-catenin were significantly lower in epithelial or sarcomatous components of PCs than in PDAs while that of TGF-β was significantly higher in epithelial components of PCs than in PDAs. No significant difference was found in incidences of the expression of these molecules between epithelial and sarcomatous components of PCs. No significant difference was noted in the expression level of N-cadherin among PDAs and epithelial and sarcomatous components of PCs. The present results showed that E-cadherin and β-catenin expression is reduced and TGF-β expression is increased in epithelial components of PCs with the same histology as PDA when compared to PDAs, suggesting that an epithelial component of PC is distinct from non-small cell carcinoma with the same histology.
Collapse
Affiliation(s)
- Akira Okimura
- Department of Pathology, Osaka Prefectural Medical Center for Respiratory and Allergic Diseases, Habikino, Osaka 583-8588
| | | | | | | | | | | | | |
Collapse
|
48
|
Mazouni C, Bonnier P, Goubar A, Romain S, Martin PM. Is quantitative oestrogen receptor expression useful in the evaluation of the clinical prognosis? Analysis of a homogeneous series of 797 patients with prospective determination of the ER status using simultaneous EIA and IHC. Eur J Cancer 2010; 46:2716-25. [DOI: 10.1016/j.ejca.2010.05.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2010] [Revised: 05/15/2010] [Accepted: 05/20/2010] [Indexed: 11/26/2022]
|
49
|
The TGF-β/Smad pathway induces breast cancer cell invasion through the up-regulation of matrix metalloproteinase 2 and 9 in a spheroid invasion model system. Breast Cancer Res Treat 2010; 128:657-66. [PMID: 20821046 DOI: 10.1007/s10549-010-1147-x] [Citation(s) in RCA: 152] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2010] [Accepted: 08/20/2010] [Indexed: 02/06/2023]
Abstract
Transforming growth factor-β (TGF-β) has opposing roles in breast cancer progression by acting as a tumor suppressor in the initial phase, but stimulating invasion and metastasis at later stages. In contrast to the mechanisms by which TGF-β induces growth arrest, the pathways that mediate tumor invasion are not well understood. Here, we describe a TGF-β-dependent invasion assay system consisting of spheroids of MCF10A1 normal breast epithelial cells (M1) and RAS-transformed (pre-)malignant derivatives (M2 and M4) embedded in collagen gels. Both basal and TGF-β-induced invasion of these cell lines was found to correlate with their tumorigenic potential; M4 showing the most aggressive behavior and M1 showing the least. Basal invasion was strongly inhibited by the TGF-β receptor kinase inhibitor SB-431542, indicating the involvement of autocrine TGF-β or TGF-β-like activity. TGF-β-induced invasion in premalignant M2 and highly malignant M4 cells was also inhibited upon specific knockdown of Smad3 or Smad4. Interestingly, both a broad spectrum matrix metalloproteinase (MMP) inhibitor and a selective MMP2 and MMP9 inhibitor mitigated TGF-β-induced invasion of M4 cells, while leaving basal invasion intact. In line with this, TGF-β was found to strongly induce MMP2 and MMP9 expression in a Smad3- and Smad4-dependent manner. This collagen-embedded spheroid system therefore offers a valuable screening model for TGF-β/Smad- and MMP2- and MMP9-dependent breast cancer invasion.
Collapse
|
50
|
Paiva CE, Drigo SA, Rosa FE, Moraes Neto FA, Caldeira JRF, Soares FA, Domingues MAC, Rogatto SR. Absence of transforming growth factor-beta type II receptor is associated with poorer prognosis in HER2-negative breast tumours. Ann Oncol 2010; 21:734-740. [PMID: 19914962 DOI: 10.1093/annonc/mdp518] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND The clinical relevance of transforming growth factor-beta (TGF-beta)-signalling pathway in breast carcinomas (BCs) remained elusive. This study aimed to evaluate the prognostic value of TGF-beta1 and transforming growth factor-beta type II receptor (TGF-betaRII) expression levels in tumour cells and their association with the established biomarkers in BC. PATIENTS AND METHODS In 324 BC from patients with long-term follow-up, the TGF-beta1 and TGF-betaRII transcript and protein expression levels were assessed. RESULTS TGF-beta1 and TGF-betaRII down-expression was significantly associated with BC. Negative TGF-beta1 and TGF-betaRII protein status was associated with the development of distant metastasis (P = 0.003 and P = 0.029, respectively). In multivariate analysis, TGF-beta1-positive tumours were associated with increased disease-free survival (DFS) [hazard ratio (HR) = 0.489, P = 0.003]. TGF-betaRII positivity was an independent prognostic factor for DFS (HR = 0.439, P = 0.001) and overall survival (OS) (HR = 0.409, P = 0.003) in human epidermal growth factor receptor-2 (HER2)-negative patients. Absence of TGF-beta1 and TGF-betaRII proteins in breast tumour cells was significantly associated with metastasis development. CONCLUSIONS To the best of our knowledge, this is the first report indicating the relevance of HER2 status in discriminating TGF-betaRII as a prognostic marker for DFS and OS in human BC. These data indicate that TGF-betaRII protein analysis in tumour cells could be introduced in clinical practice as additional prognostic biomarker in HER2-negative BC.
Collapse
Affiliation(s)
- C E Paiva
- Oncological and Hemato-oncological Center, São Paulo State University, Botucatu
| | - S A Drigo
- NeoGene Laboratory, Department of Urology, São Paulo State University, Botucatu and A. C. Camargo Cancer Treatment and Research Center
| | - F E Rosa
- NeoGene Laboratory, Department of Urology, São Paulo State University, Botucatu and A. C. Camargo Cancer Treatment and Research Center
| | | | | | - F A Soares
- Department of Pathology, A. C. Camargo Cancer Treatment and Research Center
| | - M A C Domingues
- Department of Pathology, São Paulo State University, Botucatu, São Paulo, Brazil
| | - S R Rogatto
- NeoGene Laboratory, Department of Urology, São Paulo State University, Botucatu and A. C. Camargo Cancer Treatment and Research Center.
| |
Collapse
|