1
|
Sakoguchi A, Arase H. Mechanisms for Host Immune Evasion Mediated by Plasmodium falciparum-Infected Erythrocyte Surface Antigens. Front Immunol 2022; 13:901864. [PMID: 35784341 PMCID: PMC9240312 DOI: 10.3389/fimmu.2022.901864] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 05/10/2022] [Indexed: 12/20/2022] Open
Abstract
Plasmodium falciparum infection causes the most severe form of malaria. It has been hypothesized that P. falciparum directly suppresses host immune responses because sufficient acquired immunity is often not induced even by repeated P. falciparum infections in malaria-endemic areas. It is known that many kinds of P. falciparum-derived proteins are expressed on the surface of P. falciparum-infected erythrocytes (IEs), and these proteins have long been thought to be a key to the elucidation of the host immune evasion mechanisms. Our recent studies have revealed that the P. falciparum-derived erythrocyte surface antigen, RIFIN, the largest multiple gene family protein in the P. falciparum genome, suppresses host immune cell activation through direct interaction with human inhibitory immune receptors. In this review, we will discuss the molecular mechanisms for host immune evasion by P. falciparum-infected erythrocyte surface antigens. In addition, we will discuss the recently identified host immune response to P. falciparum using specialized antibodies that target host-P. falciparum-derived molecule interactions.
Collapse
Affiliation(s)
- Akihito Sakoguchi
- Department of Molecular Protozoology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Hisashi Arase
- Department of Immunochemistry, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- Laboratory of Immunochemistry, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
- Center for Infectious Disease Education and Research, Osaka University, Suita, Japan
- *Correspondence: Hisashi Arase,
| |
Collapse
|
2
|
Abstract
Cancer is a complex disease and a significant cause of mortality worldwide. Over the course of nearly all cancer types, collagen within the tumor microenvironment influences emergence, progression, and metastasis. This review discusses collagen regulation within the tumor microenvironment, pathological involvement of collagen, and predictive values of collagen and related extracellular matrix components in main cancer types. A survey of predictive tests leveraging collagen assays using clinical cohorts is presented. A conclusion is that collagen has high predictive value in monitoring cancer processes and stratifying by outcomes. New approaches should be considered that continue to define molecular facets of collagen related to cancer.
Collapse
|
3
|
Ramos MIP, Tian L, de Ruiter EJ, Song C, Paucarmayta A, Singh A, Elshof E, Vijver SV, Shaik J, Bosiacki J, Cusumano Z, Jensen C, Willumsen N, Karsdal MA, Liu L, Langermann S, Willems S, Flies D, Meyaard L. Cancer immunotherapy by NC410, a LAIR-2 Fc protein blocking human LAIR-collagen interaction. eLife 2021; 10:62927. [PMID: 34121658 PMCID: PMC8225389 DOI: 10.7554/elife.62927] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 06/11/2021] [Indexed: 12/25/2022] Open
Abstract
Collagens are a primary component of the extracellular matrix and are functional ligands for the inhibitory immune receptor leukocyte-associated immunoglobulin-like receptor (LAIR)-1. LAIR-2 is a secreted protein that can act as a decoy receptor by binding collagen with higher affinity than LAIR-1. We propose that collagens promote immune evasion by interacting with LAIR-1 expressed on immune cells, and that LAIR-2 releases LAIR-1-mediated immune suppression. Analysis of public human datasets shows that collagens, LAIR-1 and LAIR-2 have unique and overlapping associations with survival in certain tumors. We designed a dimeric LAIR-2 with a functional IgG1 Fc tail, NC410, and showed that NC410 increases human T cell expansion and effector function in vivo in a mouse xenogeneic-graft versus-host disease model. In humanized mouse tumor models, NC410 reduces tumor growth that is dependent on T cells. Immunohistochemical analysis of human tumors shows that NC410 binds to collagen-rich areas where LAIR-1+ immune cells are localized. Our findings show that NC410 might be a novel strategy for cancer immunotherapy for immune-excluded tumors.
Collapse
Affiliation(s)
- M Ines Pascoal Ramos
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Oncode Institute, Utrecht, Netherlands
| | | | | | | | | | - Akashdip Singh
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Oncode Institute, Utrecht, Netherlands
| | - Eline Elshof
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Oncode Institute, Utrecht, Netherlands
| | - Saskia V Vijver
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Oncode Institute, Utrecht, Netherlands
| | | | | | | | | | | | | | | | | | - Stefan Willems
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | | | - Linde Meyaard
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Oncode Institute, Utrecht, Netherlands
| |
Collapse
|
4
|
Martins Cavaco AC, Dâmaso S, Casimiro S, Costa L. Collagen biology making inroads into prognosis and treatment of cancer progression and metastasis. Cancer Metastasis Rev 2021; 39:603-623. [PMID: 32447477 DOI: 10.1007/s10555-020-09888-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Progression through dissemination to tumor-surrounding tissues and metastasis development is a hallmark of cancer that requires continuous cell-to-cell interactions and tissue remodeling. In fact, metastization can be regarded as a tissue disease orchestrated by cancer cells, leading to neoplastic colonization of new organs. Collagen is a major component of the extracellular matrix (ECM), and increasing evidence suggests that it has an important role in cancer progression and metastasis. Desmoplasia and collagen biomarkers have been associated with relapse and death in cancer patients. Despite the increasing interest in ECM and in the desmoplastic process in tumor microenvironment as prognostic factors and therapeutic targets in cancer, further research is required for a better understanding of these aspects of cancer biology. In this review, published evidence correlating collagen with cancer prognosis is retrieved and analyzed, and the role of collagen and its fragments in cancer pathophysiology is discussed.
Collapse
Affiliation(s)
- Ana C Martins Cavaco
- Luis Costa Lab, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028, Lisboa, Portugal
| | - Sara Dâmaso
- Serviço de Oncologia, Hospital de Santa Maria-CHULN, 1649-028, Lisboa, Portugal
| | - Sandra Casimiro
- Luis Costa Lab, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028, Lisboa, Portugal
| | - Luís Costa
- Luis Costa Lab, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028, Lisboa, Portugal.
- Serviço de Oncologia, Hospital de Santa Maria-CHULN, 1649-028, Lisboa, Portugal.
| |
Collapse
|
5
|
Joseph C, Alsaleem MA, Toss MS, Kariri YA, Althobiti M, Alsaeed S, Aljohani AI, Narasimha PL, Mongan NP, Green AR, Rakha EA. The ITIM-Containing Receptor: Leukocyte-Associated Immunoglobulin-Like Receptor-1 (LAIR-1) Modulates Immune Response and Confers Poor Prognosis in Invasive Breast Carcinoma. Cancers (Basel) 2020; 13:E80. [PMID: 33396670 PMCID: PMC7795350 DOI: 10.3390/cancers13010080] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 12/22/2020] [Accepted: 12/24/2020] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND The leukocyte-associated immunoglobulin-like receptor-1 (LAIR-1) plays a role in immune response homeostasis, extracellular matrix remodelling and it is overexpressed in many high-grade cancers. This study aimed to elucidate the biological and prognostic role of LAIR-1 in invasive breast cancer (BC). METHODS The biological and prognostic effect of LAIR-1 was evaluated at the mRNA and protein levels using well-characterised multiple BC cohorts. Related signalling pathways were evaluated using in silico differential gene expression and siRNA knockdown were used for functional analyses. RESULTS High LAIR-1 expression either in mRNA or protein levels were associated with high tumour grade, poor Nottingham Prognostic Index, hormone receptor negativity, immune cell infiltrates and extracellular matrix remodelling elements. High LAIR-1 protein expression was an independent predictor of shorter BC-specific survival and distant metastasis-free survival in the entire BC cohort and human epidermal growth factor receptor 2 (HER2)+ subtype. Pathway analysis highlights LAIR-1 association with extracellular matrix remodelling-receptor interaction, and cellular proliferation. Depletion of LAIR-1 using siRNA significantly reduced cell proliferation and invasion capability in HER2+ BC cell lines. CONCLUSION High expression of LAIR-1 is associated with poor clinical outcome in BC. Association with immune cells and immune checkpoint markers warrant further studies to assess the underlying mechanistic roles.
Collapse
Affiliation(s)
- Chitra Joseph
- School of Medicine, The University of Nottingham and Nottingham University Hospitals NHS Trust, Nottingham City Hospital, Nottingham NG7 2RD, UK;
| | - Mansour A. Alsaleem
- Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham NG7 2RD, UK; (M.A.A.); (Y.A.K.); (M.A.); (S.A.); (A.I.A.); (P.L.N.); (N.P.M.); (A.R.G.)
- Department of Applied Medical Sciences, Unayzah Community College, Qassim University, Unayzah 56435, Saudi Arabia
| | - Michael S. Toss
- School of Medicine, The University of Nottingham and Nottingham University Hospitals NHS Trust, Nottingham City Hospital, Nottingham NG7 2RD, UK;
- Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham NG7 2RD, UK; (M.A.A.); (Y.A.K.); (M.A.); (S.A.); (A.I.A.); (P.L.N.); (N.P.M.); (A.R.G.)
| | - Yousif A. Kariri
- Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham NG7 2RD, UK; (M.A.A.); (Y.A.K.); (M.A.); (S.A.); (A.I.A.); (P.L.N.); (N.P.M.); (A.R.G.)
- Department of Clinical Laboratory Science, Faculty of Applied Medical Science, Shaqra University 33, Shaqra 11961, Saudi Arabia
| | - Maryam Althobiti
- Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham NG7 2RD, UK; (M.A.A.); (Y.A.K.); (M.A.); (S.A.); (A.I.A.); (P.L.N.); (N.P.M.); (A.R.G.)
- Department of Clinical Laboratory Science, Faculty of Applied Medical Science, Shaqra University 33, Shaqra 11961, Saudi Arabia
| | - Sami Alsaeed
- Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham NG7 2RD, UK; (M.A.A.); (Y.A.K.); (M.A.); (S.A.); (A.I.A.); (P.L.N.); (N.P.M.); (A.R.G.)
| | - Abrar I. Aljohani
- Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham NG7 2RD, UK; (M.A.A.); (Y.A.K.); (M.A.); (S.A.); (A.I.A.); (P.L.N.); (N.P.M.); (A.R.G.)
| | - Pavan L. Narasimha
- Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham NG7 2RD, UK; (M.A.A.); (Y.A.K.); (M.A.); (S.A.); (A.I.A.); (P.L.N.); (N.P.M.); (A.R.G.)
| | - Nigel P. Mongan
- Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham NG7 2RD, UK; (M.A.A.); (Y.A.K.); (M.A.); (S.A.); (A.I.A.); (P.L.N.); (N.P.M.); (A.R.G.)
| | - Andrew R. Green
- Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham NG7 2RD, UK; (M.A.A.); (Y.A.K.); (M.A.); (S.A.); (A.I.A.); (P.L.N.); (N.P.M.); (A.R.G.)
| | - Emad A. Rakha
- School of Medicine, The University of Nottingham and Nottingham University Hospitals NHS Trust, Nottingham City Hospital, Nottingham NG7 2RD, UK;
- Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham NG7 2RD, UK; (M.A.A.); (Y.A.K.); (M.A.); (S.A.); (A.I.A.); (P.L.N.); (N.P.M.); (A.R.G.)
| |
Collapse
|
6
|
Barkovskaya A, Buffone A, Žídek M, Weaver VM. Proteoglycans as Mediators of Cancer Tissue Mechanics. Front Cell Dev Biol 2020; 8:569377. [PMID: 33330449 PMCID: PMC7734320 DOI: 10.3389/fcell.2020.569377] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 11/04/2020] [Indexed: 12/16/2022] Open
Abstract
Proteoglycans are a diverse group of molecules which are characterized by a central protein backbone that is decorated with a variety of linear sulfated glycosaminoglycan side chains. Proteoglycans contribute significantly to the biochemical and mechanical properties of the interstitial extracellular matrix where they modulate cellular behavior by engaging transmembrane receptors. Proteoglycans also comprise a major component of the cellular glycocalyx to influence transmembrane receptor structure/function and mechanosignaling. Through their ability to initiate biochemical and mechanosignaling in cells, proteoglycans elicit profound effects on proliferation, adhesion and migration. Pathologies including cancer and cardiovascular disease are characterized by perturbed expression of proteoglycans where they compromise cell and tissue behavior by stiffening the extracellular matrix and increasing the bulkiness of the glycocalyx. Increasing evidence indicates that a bulky glycocalyx and proteoglycan-enriched extracellular matrix promote malignant transformation, increase cancer aggression and alter anti-tumor therapy response. In this review, we focus on the contribution of proteoglycans to mechanobiology in the context of normal and transformed tissues. We discuss the significance of proteoglycans for therapy response, and the current experimental strategies that target proteoglycans to sensitize cancer cells to treatment.
Collapse
Affiliation(s)
- Anna Barkovskaya
- Center for Bioengineering & Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Alexander Buffone
- Center for Bioengineering & Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Martin Žídek
- Center for Bioengineering & Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Valerie M. Weaver
- Center for Bioengineering & Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
- Department of Radiation Oncology, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, United States
- Department of Bioengineering, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, United States
- Department of Therapeutic Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, United States
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
7
|
Roles of Proteoglycans and Glycosaminoglycans in Cancer Development and Progression. Int J Mol Sci 2020; 21:ijms21175983. [PMID: 32825245 PMCID: PMC7504257 DOI: 10.3390/ijms21175983] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 08/18/2020] [Accepted: 08/18/2020] [Indexed: 12/11/2022] Open
Abstract
The extracellular matrix (ECM) spatiotemporally controls cell fate; however, dysregulation of ECM remodeling can lead to tumorigenesis and cancer development by providing favorable conditions for tumor cells. Proteoglycans (PGs) and glycosaminoglycans (GAGs) are the major macromolecules composing ECM. They influence both cell behavior and matrix properties through direct and indirect interactions with various cytokines, growth factors, cell surface receptors, adhesion molecules, enzymes, and glycoproteins within the ECM. The classical features of PGs/GAGs play well-known roles in cancer angiogenesis, proliferation, invasion, and metastasis. Several lines of evidence suggest that PGs/GAGs critically affect broader aspects in cancer initiation and the progression process, including regulation of cell metabolism, serving as a sensor of ECM's mechanical properties, affecting immune supervision, and participating in therapeutic resistance to various forms of treatment. These functions may be implemented through the characteristics of PGs/GAGs as molecular bridges linking ECM and cells in cell-specific and context-specific manners within the tumor microenvironment (TME). In this review, we intend to present a comprehensive illustration of the ways in which PGs/GAGs participate in and regulate several aspects of tumorigenesis; we put forward a perspective regarding their effects as biomarkers or targets for diagnoses and therapeutic interventions.
Collapse
|
8
|
Exploring the roles of MACIT and multiplexin collagens in stem cells and cancer. Semin Cancer Biol 2019; 62:134-148. [PMID: 31479735 DOI: 10.1016/j.semcancer.2019.08.033] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 08/20/2019] [Accepted: 08/30/2019] [Indexed: 02/07/2023]
Abstract
The extracellular matrix (ECM) is ubiquitously involved in neoplastic transformation, tumour growth and metastatic dissemination, and the interplay between tumour and stromal cells and the ECM is now considered crucial for the formation of a tumour-supporting microenvironment. The 28 different collagens (Col) form a major ECM protein family and display extraordinary functional diversity in tissue homeostasis as well as in pathological conditions, with functions ranging from structural support for tissues to regulatory binding activities and storage of biologically active cryptic domains releasable through ECM proteolysis. Two subfamilies of collagens, namely the plasma membrane-associated collagens with interrupted triple-helices (MACITs, including ColXIII, ColXXIII and ColXXV) and the basement membrane-associated collagens with multiple triple-helix domains with interruptions (multiplexins, including ColXV and ColXVIII), have highly interesting regulatory functions in tissue and organ development, as well as in various diseases, including cancer. An increasing, albeit yet sparse, data suggest that these collagens play crucial roles in conveying regulatory signals from the extracellular space to cells. We summarize here the current knowledge about MACITs and multiplexins as regulators of stemness and oncogenic processes, as well as their roles in influencing cell fate decisions in healthy and cancerous tissues. In addition, we present a bioinformatic analysis of the impacts of MACITs and multiplexins transcript levels on the prognosis of patients representing a wide array of malignant diseases, to aid future diagnostic and therapeutic efforts.
Collapse
|
9
|
Huang Y, Li XM, Chen SG, Deng J, Lei Y, Li WJ, Zhang HZ, Zhang H, Li D, Xie P. Application of antibodies against Borna disease virus phosphoprotein and nucleoprotein on paraffin sections. Mol Med Rep 2018; 17:5416-5422. [PMID: 29363723 DOI: 10.3892/mmr.2018.8467] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Accepted: 10/30/2017] [Indexed: 11/06/2022] Open
Abstract
In order to study the application of antibodies against recombinant proteins for detecting Borna disease virus (BDV) phosphoprotein (p24) and nucleoprotein (p40) (BDV‑p24/p40) on paraffin sections by immunohistochemistry. The purified fusion p24 and p40 proteins were used for the preparation of polyclonal and monoclonal anti‑p24 and anti‑40 antibodies, which were confirmed by ELISA and western blotting. Paraffin sections were made from BDV‑infected Sprague‑Dawley (SD) rats (n=20), PBS‑injected SD rats (n=20), normal SD rats (n=20) and normal C57 mice (n=20). Immunohistochemical staining was performed according to the EnVision™ two‑step protocol. Heat‑mediated antigen retrieval was performed using the retrieval buffer sodium citrate (1 mM; pH 6.0). All the antibodies against recombinant proteins exhibited good sensitivity and specificity. There were significant differences between the BDV‑infected group and the BDV‑uninfected group for poly‑ and monoclonal anti‑p24 and ‑p40 antibodies. These antibodies against recombinant proteins may be used effectively to detect BDV p24 and p40 in paraffin sections.
Collapse
Affiliation(s)
- Ying Huang
- Department of Pathology, Faculty of Basic Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Xiao-Mei Li
- Department of Pathology, Faculty of Basic Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Shi-Gang Chen
- Neuroscience Center, Key Laboratory of Neurobiology of Chongqing, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Jing Deng
- Neuroscience Center, Key Laboratory of Neurobiology of Chongqing, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Yang Lei
- Neuroscience Center, Key Laboratory of Neurobiology of Chongqing, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Wen-Juan Li
- Neuroscience Center, Key Laboratory of Neurobiology of Chongqing, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Hong-Zhi Zhang
- Neuroscience Center, Key Laboratory of Neurobiology of Chongqing, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Hong Zhang
- Neuroscience Center, Key Laboratory of Neurobiology of Chongqing, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Dan Li
- Department of Pathology, Faculty of Basic Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Peng Xie
- Neuroscience Center, Key Laboratory of Neurobiology of Chongqing, Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
10
|
Theocharis AD, Karamanos NK. Proteoglycans remodeling in cancer: Underlying molecular mechanisms. Matrix Biol 2017; 75-76:220-259. [PMID: 29128506 DOI: 10.1016/j.matbio.2017.10.008] [Citation(s) in RCA: 148] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 10/23/2017] [Accepted: 10/24/2017] [Indexed: 02/07/2023]
Abstract
Extracellular matrix is a highly dynamic macromolecular network. Proteoglycans are major components of extracellular matrix playing key roles in its structural organization and cell signaling contributing to the control of numerous normal and pathological processes. As multifunctional molecules, proteoglycans participate in various cell functions during morphogenesis, wound healing, inflammation and tumorigenesis. Their interactions with matrix effectors, cell surface receptors and enzymes enable them with unique properties. In malignancy, extensive remodeling of tumor stroma is associated with marked alterations in proteoglycans' expression and structural variability. Proteoglycans exert diverse functions in tumor stroma in a cell-specific and context-specific manner and they mainly contribute to the formation of a permissive provisional matrix for tumor growth affecting tissue organization, cell-cell and cell-matrix interactions and tumor cell signaling. Proteoglycans also modulate cancer cell phenotype and properties, the development of drug resistance and tumor stroma angiogenesis. This review summarizes the proteoglycans remodeling and their novel biological roles in malignancies with particular emphasis to the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Achilleas D Theocharis
- Biochemistry, Biochemical Analysis & Matrix Pathobiochemistry Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26500 Patras, Greece.
| | - Nikos K Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiochemistry Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26500 Patras, Greece.
| |
Collapse
|
11
|
Heljasvaara R, Aikio M, Ruotsalainen H, Pihlajaniemi T. Collagen XVIII in tissue homeostasis and dysregulation - Lessons learned from model organisms and human patients. Matrix Biol 2016; 57-58:55-75. [PMID: 27746220 DOI: 10.1016/j.matbio.2016.10.002] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 09/12/2016] [Accepted: 10/10/2016] [Indexed: 12/13/2022]
Abstract
Collagen XVIII is a ubiquitous basement membrane (BM) proteoglycan produced in three tissue-specific isoforms that differ in their N-terminal non-collagenous sequences, but share collagenous and C-terminal non-collagenous domains. The collagenous domain provides flexibility to the large collagen XVIII molecules on account of multiple interruptions in collagenous sequences. Each isoform has a complex multi-domain structure that endows it with an ability to perform various biological functions. The long isoform contains a frizzled-like (Fz) domain with Wnt-inhibiting activity and a unique domain of unknown function (DUF959), which is also present in the medium isoform. All three isoforms share an N-terminal laminin-G-like/thrombospondin-1 sequence whose specific functions still remain unconfirmed. The proteoglycan nature of the isoforms further increases the functional diversity of collagen XVIII. An anti-angiogenic domain termed endostatin resides in the C-terminus of collagen XVIII and is proteolytically cleaved from the parental molecule during the BM breakdown for example in the process of tumour progression. Recombinant endostatin can efficiently reduce tumour angiogenesis and growth in experimental models by inhibiting endothelial cell migration and proliferation or by inducing their death, but its efficacy against human cancers is still a subject of debate. Mutations in the COL18A1 gene result in Knobloch syndrome, a genetic disorder characterised mainly by severe eye defects and encephalocele and, occasionally, other symptoms. Studies with gene-modified mice have elucidated some aspects of this rare disease, highlighting in particular the importance of collagen XVIII in the development of the eye. Research with model organisms have also helped in determining other structural and biological functions of collagen XVIII, such as its requirement in the maintenance of BM integrity and its emerging roles in regulating cell survival, stem or progenitor cell maintenance and differentiation and inflammation. In this review, we summarise current knowledge on the properties and endogenous functions of collagen XVIII in normal situations and tissue dysregulation. When data is available, we discuss the functions of the distinct isoforms and their specific domains.
Collapse
Affiliation(s)
- Ritva Heljasvaara
- Oulu Center for Cell-Matrix Research, Biocenter Oulu, Faculty of Biochemistry and Molecular Medicine, University of Oulu, FIN-90014 Oulu, Finland; Centre for Cancer Biomarkers CCBIO, Department of Biomedicine, University of Bergen, N-5009 Bergen, Norway.
| | - Mari Aikio
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Heli Ruotsalainen
- Oulu Center for Cell-Matrix Research, Biocenter Oulu, Faculty of Biochemistry and Molecular Medicine, University of Oulu, FIN-90014 Oulu, Finland
| | - Taina Pihlajaniemi
- Oulu Center for Cell-Matrix Research, Biocenter Oulu, Faculty of Biochemistry and Molecular Medicine, University of Oulu, FIN-90014 Oulu, Finland
| |
Collapse
|
12
|
Song Y, Li A, Zhang L, Duan L. Expression of G protein-coupled receptor 56 is associated with tumor progression in non-small-cell lung carcinoma patients. Onco Targets Ther 2016; 9:4105-12. [PMID: 27462165 PMCID: PMC4939992 DOI: 10.2147/ott.s106907] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background G protein-coupled receptor 56 (GPR56) is an adhesion G protein-coupled receptor with essential functions for cell physiology and survival, and its expression correlates with prognosis in a number of malignancies. The aim of this study is to determine the relationship of GPR56 expression with clinicopathological parameters and prognosis in non-small-cell lung carcinoma (NSCLC). Methods The levels of GPR56 were evaluated by immunohistochemistry in 157 NSCLC tissue samples. The association between GPR56 and clinicopathological parameters was evaluated by χ2 test. Univariate and multivariate analyses were performed to demonstrate the prognosis role of GPR56. The function of GPR56 in NSCLC cell lines was also explored through overexpression and knockdown studies. Results The expression level of GPR56 in tumor tissues was significantly correlated with the TNM stage of NSCLC (P=0.005). Univariate and multivariate analyses revealed that GPR56 can act as an independent prognostic factor for overall survival. Furthermore, through overexpression and knockdown experiments, we confirmed that GPR56 can promote the proliferation and invasion of NSCLC cells. Conclusion GPR56 plays an important role in tumor development and may serve as a promising target for prognostic prediction in NSCLC.
Collapse
Affiliation(s)
- Yanjie Song
- Department of Neurosurgery, Yidu Central Hospital of Weifang, Weifang Medical University, Qingzhou
| | - Aiqin Li
- Department of Neurosurgery, Yidu Central Hospital of Weifang, Weifang Medical University, Qingzhou
| | - Li Zhang
- Department of Neurosurgery, Yidu Central Hospital of Weifang, Weifang Medical University, Qingzhou
| | - Lingling Duan
- Medical Care Department for Personnel, Jinan Central Hospital, Shangdong University, Jinan, Shandong, People's Republic of China
| |
Collapse
|
13
|
A phase II study of Endostatin in combination with paclitaxel, carboplatin, and radiotherapy in patients with unresectable locally advanced non-small cell lung cancer. BMC Cancer 2016; 16:266. [PMID: 27067521 PMCID: PMC4828797 DOI: 10.1186/s12885-016-2234-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 03/01/2016] [Indexed: 12/28/2022] Open
Abstract
Background Endostatin inhibits the pro-angiogenic action of basic fibroblast growth factor and vascular endothelial growth factor in different human cancers. This study assessed the efficacy of endostatin combined with concurrent chemoradiotherapy of non-small cell lung cancer (NSCLC). Methods Nineteen patients with unresectable stage III NSCLC, Eastern Cooperative Oncology Group (ECOG) performance status 0-l, and adequate organ function were treated with 60–66 Gy thoracic radiation therapy over 30–33 fractions concurrent with weekly 7.5 mg/m2 endostatin for 14 days, 50 mg/m2 paclitaxel, and 2 mg/mL/min carboplatin over 30 min. Patients were then treated with 7.5 mg/m2 endostatin for 14 days, 150 mg/m2 paclitaxel, and 5 mg/mL/min carboplatin every 3 weeks for 2 cycles as the consolidation treatment. The objective response rate was recorded according to the Response Evaluation Criteria in Solid Tumors (RECIST) criteria, and the toxicity was evaluated using the National Cancer Institute (NCI) Common Toxicity Criteria. Results Six patients were unable to complete the consolidation treatment (4 pulmonary toxicity, 1 tracheoesophageal fistulae, and 1 progressive disease). Seventeen patients were included for data analysis. Specifically, one (5.9 %) patient had a complete response and 12 (70.6 %) had a partial response, whereas two patients had stable disease and the other two had disease progression. The overall response rate was 76 % (95 % confidence interval [CI], 51 %–97 %). The median progression-free survival was 10 months (95 % CI, 7.6–12.3 months), and the median overall survival was 14 months (95 % CI, 10.7–17.2 months). Early 10 patients who completed the treatment regimen showed that four patients experienced grade III pulmonary toxicity a few months after chemoradiotherapy, leading to the early closure of the trial according to the study design. Conclusions The reslult of concurrent endostatin treatment with chemoradiotherapy in locally advanced unresectable NSCLC did not meet the goal per study design with unacceptable toxicity. The real impact of endostatin as the first-line treatment combined with chemoradiotherapy on the survival of NSCLC patients remains to be determined. (NCT 01158144).
Collapse
|
14
|
Poluzzi C, Iozzo RV, Schaefer L. Endostatin and endorepellin: A common route of action for similar angiostatic cancer avengers. Adv Drug Deliv Rev 2016; 97:156-73. [PMID: 26518982 DOI: 10.1016/j.addr.2015.10.012] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 10/15/2015] [Accepted: 10/16/2015] [Indexed: 12/17/2022]
Abstract
Traditional cancer therapy typically targets the tumor proper. However, newly-formed vasculature exerts a major role in cancer development and progression. Autophagy, as a biological mechanism for clearing damaged proteins and oxidative stress products released in the tumor milieu, could help in tumor resolution by rescuing cells undergoing modifications or inducing autophagic-cell death of tumor blood vessels. Cleaved fragments of extracellular matrix proteoglycans are emerging as key players in the modulation of angiogenesis and endothelial cell autophagy. An essential characteristic of cancer progression is the remodeling of the basement membrane and the release of processed forms of its constituents. Endostatin, generated from collagen XVIII, and endorepellin, the C-terminal segment of the large proteoglycan perlecan, possess a dual activity as modifiers of both angiogenesis and endothelial cell autophagy. Manipulation of these endogenously-processed forms, located in the basement membrane within tumors, could represent new therapeutic approaches for cancer eradication.
Collapse
Affiliation(s)
- Chiara Poluzzi
- Pharmazentrum Frankfurt/ZAFES, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany
| | - Renato V Iozzo
- Department of Pathology, Anatomy and Cell Biology, and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Liliana Schaefer
- Pharmazentrum Frankfurt/ZAFES, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany.
| |
Collapse
|
15
|
Zhang Y, Yu LK, Lu GJ, Xia N, Xie HY, Hu W, Hao KK, Xu CH, Qian Q. Prognostic values of VEGF and endostatin with malignant pleural effusions in patients with lung cancer. Asian Pac J Cancer Prev 2015; 15:8435-40. [PMID: 25339042 DOI: 10.7314/apjcp.2014.15.19.8435] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
AIMS Angiogenesis is important in malignant pleural effusion (MPE) formation and it is regulated by a number of pro- and anti-angiogenic cytokines. The purpose of this study was to evaluate the prognostic value of angiogenic factor vascular endothelial growth factor (VEGF) and angiogenesis inhibitor endostatin in lung cancer patients with MPE, and investigate the relationship between these two kinds of agent. METHODS Using enzyme-linked immunoadsorbent assay, the concentrations of VEGF and endostatin were measured in pleural effusions (PE) and serum from a total of 70 lung cancer patients with MPE and 20 patients with tuberculosis. RESULTS Compared to patients with tuberculosis, the levels of VEGF and endostatin in both PE and serum were significantly higher in patients with lung cancer. There were statistically significant correlations between VEGF levels in PE and serum (r=0.696, <0.001), endostatin levels in PE and serum (r=0.310, p=0.022), and VEGF and endostatin levels in PE (r=0.287, p=0.019). Cox multivariate analysis revealed that elevated pleural VEGF and endostatin levels and serum endostatin level were independent predictors of shorter overall survival. CONCLUSION Both pro- and anti-angiogenic factors are likely contributors to PE formation. Our results suggest that the levels of VEGF and endostatin in PE, together with endostatin in serum, may be potential prognostic parameters for lung cancer patients with MPE.
Collapse
Affiliation(s)
- Yu Zhang
- First Department of Respiratory Medicine, Nanjing Chest Hospital, Nanjing, Jiangsu Province, China E-mail :
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Vachani A, Pass HI, Rom WN, Midthun DE, Edell ES, Laviolette M, Li XJ, Fong PY, Hunsucker SW, Hayward C, Mazzone PJ, Madtes DK, Miller YE, Walker MG, Shi J, Kearney P, Fang KC, Massion PP. Validation of a multiprotein plasma classifier to identify benign lung nodules. J Thorac Oncol 2015; 10:629-37. [PMID: 25590604 PMCID: PMC4382127 DOI: 10.1097/jto.0000000000000447] [Citation(s) in RCA: 51] [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] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Indeterminate pulmonary nodules (IPNs) lack clinical or radiographic features of benign etiologies and often undergo invasive procedures unnecessarily, suggesting potential roles for diagnostic adjuncts using molecular biomarkers. The primary objective was to validate a multivariate classifier that identifies likely benign lung nodules by assaying plasma protein expression levels, yielding a range of probability estimates based on high negative predictive values (NPVs) for patients with 8 to 30 mm IPNs. METHODS A retrospective, multicenter, case-control study was performed using multiple reaction monitoring mass spectrometry, a classifier comprising five diagnostic and six normalization proteins, and blinded analysis of an independent validation set of plasma samples. RESULTS The classifier achieved validation on 141 lung nodule-associated plasma samples based on predefined statistical goals to optimize sensitivity. Using a population based nonsmall-cell lung cancer prevalence estimate of 23% for 8 to 30 mm IPNs, the classifier identified likely benign lung nodules with 90% negative predictive value and 26% positive predictive value, as shown in our prior work, at 92% sensitivity and 20% specificity, with the lower bound of the classifier's performance at 70% sensitivity and 48% specificity. Classifier scores for the overall cohort were statistically independent of patient age, tobacco use, nodule size, and chronic obstructive pulmonary disease diagnosis. The classifier also demonstrated incremental diagnostic performance in combination with a four-parameter clinical model. CONCLUSIONS This proteomic classifier provides a range of probability estimates for the likelihood of a benign etiology that may serve as a noninvasive, diagnostic adjunct for clinical assessments of patients with IPNs.
Collapse
Affiliation(s)
- Anil Vachani
- Division of Pulmonary, Allergy, and Critical Care Medicine, Penn Lung Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Cardiothoracic Surgery, New York University Langone Medical Center, New York, New York; Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine and Department of Environmental Medicine, New York University School of Medicine, New York University Langone Medical Center, New York, New York; Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minnesota; Unité de Recherche en Pneumologie, Centre de Recherche de l’Hôpital Laval, Institut Universitaire de Cardiologie et de Pneumologie de l’Université Laval; Québec, Canada; Integrated Diagnostics, Seattle, Washington; Respiratory Institute, Cleveland Clinic, Cleveland, Ohio; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, Washington; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Veterans Administration Eastern Colorado Healthcare System, University of Colorado Denver School of Medicine, Denver, Colorado; Statistics Consultant, Carlsbad, California; Thoracic Program, Vanderbilt-Ingram Comprehensive Cancer Center, Nashville, Tennessee; Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville Campus, Nashville, Tennessee
| | - Harvey I. Pass
- Division of Pulmonary, Allergy, and Critical Care Medicine, Penn Lung Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Cardiothoracic Surgery, New York University Langone Medical Center, New York, New York; Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine and Department of Environmental Medicine, New York University School of Medicine, New York University Langone Medical Center, New York, New York; Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minnesota; Unité de Recherche en Pneumologie, Centre de Recherche de l’Hôpital Laval, Institut Universitaire de Cardiologie et de Pneumologie de l’Université Laval; Québec, Canada; Integrated Diagnostics, Seattle, Washington; Respiratory Institute, Cleveland Clinic, Cleveland, Ohio; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, Washington; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Veterans Administration Eastern Colorado Healthcare System, University of Colorado Denver School of Medicine, Denver, Colorado; Statistics Consultant, Carlsbad, California; Thoracic Program, Vanderbilt-Ingram Comprehensive Cancer Center, Nashville, Tennessee; Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville Campus, Nashville, Tennessee
| | - William N. Rom
- Division of Pulmonary, Allergy, and Critical Care Medicine, Penn Lung Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Cardiothoracic Surgery, New York University Langone Medical Center, New York, New York; Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine and Department of Environmental Medicine, New York University School of Medicine, New York University Langone Medical Center, New York, New York; Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minnesota; Unité de Recherche en Pneumologie, Centre de Recherche de l’Hôpital Laval, Institut Universitaire de Cardiologie et de Pneumologie de l’Université Laval; Québec, Canada; Integrated Diagnostics, Seattle, Washington; Respiratory Institute, Cleveland Clinic, Cleveland, Ohio; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, Washington; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Veterans Administration Eastern Colorado Healthcare System, University of Colorado Denver School of Medicine, Denver, Colorado; Statistics Consultant, Carlsbad, California; Thoracic Program, Vanderbilt-Ingram Comprehensive Cancer Center, Nashville, Tennessee; Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville Campus, Nashville, Tennessee
| | - David E. Midthun
- Division of Pulmonary, Allergy, and Critical Care Medicine, Penn Lung Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Cardiothoracic Surgery, New York University Langone Medical Center, New York, New York; Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine and Department of Environmental Medicine, New York University School of Medicine, New York University Langone Medical Center, New York, New York; Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minnesota; Unité de Recherche en Pneumologie, Centre de Recherche de l’Hôpital Laval, Institut Universitaire de Cardiologie et de Pneumologie de l’Université Laval; Québec, Canada; Integrated Diagnostics, Seattle, Washington; Respiratory Institute, Cleveland Clinic, Cleveland, Ohio; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, Washington; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Veterans Administration Eastern Colorado Healthcare System, University of Colorado Denver School of Medicine, Denver, Colorado; Statistics Consultant, Carlsbad, California; Thoracic Program, Vanderbilt-Ingram Comprehensive Cancer Center, Nashville, Tennessee; Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville Campus, Nashville, Tennessee
| | - Eric S. Edell
- Division of Pulmonary, Allergy, and Critical Care Medicine, Penn Lung Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Cardiothoracic Surgery, New York University Langone Medical Center, New York, New York; Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine and Department of Environmental Medicine, New York University School of Medicine, New York University Langone Medical Center, New York, New York; Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minnesota; Unité de Recherche en Pneumologie, Centre de Recherche de l’Hôpital Laval, Institut Universitaire de Cardiologie et de Pneumologie de l’Université Laval; Québec, Canada; Integrated Diagnostics, Seattle, Washington; Respiratory Institute, Cleveland Clinic, Cleveland, Ohio; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, Washington; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Veterans Administration Eastern Colorado Healthcare System, University of Colorado Denver School of Medicine, Denver, Colorado; Statistics Consultant, Carlsbad, California; Thoracic Program, Vanderbilt-Ingram Comprehensive Cancer Center, Nashville, Tennessee; Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville Campus, Nashville, Tennessee
| | - Michel Laviolette
- Division of Pulmonary, Allergy, and Critical Care Medicine, Penn Lung Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Cardiothoracic Surgery, New York University Langone Medical Center, New York, New York; Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine and Department of Environmental Medicine, New York University School of Medicine, New York University Langone Medical Center, New York, New York; Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minnesota; Unité de Recherche en Pneumologie, Centre de Recherche de l’Hôpital Laval, Institut Universitaire de Cardiologie et de Pneumologie de l’Université Laval; Québec, Canada; Integrated Diagnostics, Seattle, Washington; Respiratory Institute, Cleveland Clinic, Cleveland, Ohio; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, Washington; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Veterans Administration Eastern Colorado Healthcare System, University of Colorado Denver School of Medicine, Denver, Colorado; Statistics Consultant, Carlsbad, California; Thoracic Program, Vanderbilt-Ingram Comprehensive Cancer Center, Nashville, Tennessee; Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville Campus, Nashville, Tennessee
| | - Xiao-Jun Li
- Division of Pulmonary, Allergy, and Critical Care Medicine, Penn Lung Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Cardiothoracic Surgery, New York University Langone Medical Center, New York, New York; Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine and Department of Environmental Medicine, New York University School of Medicine, New York University Langone Medical Center, New York, New York; Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minnesota; Unité de Recherche en Pneumologie, Centre de Recherche de l’Hôpital Laval, Institut Universitaire de Cardiologie et de Pneumologie de l’Université Laval; Québec, Canada; Integrated Diagnostics, Seattle, Washington; Respiratory Institute, Cleveland Clinic, Cleveland, Ohio; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, Washington; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Veterans Administration Eastern Colorado Healthcare System, University of Colorado Denver School of Medicine, Denver, Colorado; Statistics Consultant, Carlsbad, California; Thoracic Program, Vanderbilt-Ingram Comprehensive Cancer Center, Nashville, Tennessee; Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville Campus, Nashville, Tennessee
| | - Pui-Yee Fong
- Division of Pulmonary, Allergy, and Critical Care Medicine, Penn Lung Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Cardiothoracic Surgery, New York University Langone Medical Center, New York, New York; Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine and Department of Environmental Medicine, New York University School of Medicine, New York University Langone Medical Center, New York, New York; Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minnesota; Unité de Recherche en Pneumologie, Centre de Recherche de l’Hôpital Laval, Institut Universitaire de Cardiologie et de Pneumologie de l’Université Laval; Québec, Canada; Integrated Diagnostics, Seattle, Washington; Respiratory Institute, Cleveland Clinic, Cleveland, Ohio; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, Washington; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Veterans Administration Eastern Colorado Healthcare System, University of Colorado Denver School of Medicine, Denver, Colorado; Statistics Consultant, Carlsbad, California; Thoracic Program, Vanderbilt-Ingram Comprehensive Cancer Center, Nashville, Tennessee; Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville Campus, Nashville, Tennessee
| | - Stephen W. Hunsucker
- Division of Pulmonary, Allergy, and Critical Care Medicine, Penn Lung Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Cardiothoracic Surgery, New York University Langone Medical Center, New York, New York; Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine and Department of Environmental Medicine, New York University School of Medicine, New York University Langone Medical Center, New York, New York; Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minnesota; Unité de Recherche en Pneumologie, Centre de Recherche de l’Hôpital Laval, Institut Universitaire de Cardiologie et de Pneumologie de l’Université Laval; Québec, Canada; Integrated Diagnostics, Seattle, Washington; Respiratory Institute, Cleveland Clinic, Cleveland, Ohio; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, Washington; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Veterans Administration Eastern Colorado Healthcare System, University of Colorado Denver School of Medicine, Denver, Colorado; Statistics Consultant, Carlsbad, California; Thoracic Program, Vanderbilt-Ingram Comprehensive Cancer Center, Nashville, Tennessee; Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville Campus, Nashville, Tennessee
| | - Clive Hayward
- Division of Pulmonary, Allergy, and Critical Care Medicine, Penn Lung Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Cardiothoracic Surgery, New York University Langone Medical Center, New York, New York; Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine and Department of Environmental Medicine, New York University School of Medicine, New York University Langone Medical Center, New York, New York; Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minnesota; Unité de Recherche en Pneumologie, Centre de Recherche de l’Hôpital Laval, Institut Universitaire de Cardiologie et de Pneumologie de l’Université Laval; Québec, Canada; Integrated Diagnostics, Seattle, Washington; Respiratory Institute, Cleveland Clinic, Cleveland, Ohio; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, Washington; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Veterans Administration Eastern Colorado Healthcare System, University of Colorado Denver School of Medicine, Denver, Colorado; Statistics Consultant, Carlsbad, California; Thoracic Program, Vanderbilt-Ingram Comprehensive Cancer Center, Nashville, Tennessee; Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville Campus, Nashville, Tennessee
| | - Peter J. Mazzone
- Division of Pulmonary, Allergy, and Critical Care Medicine, Penn Lung Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Cardiothoracic Surgery, New York University Langone Medical Center, New York, New York; Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine and Department of Environmental Medicine, New York University School of Medicine, New York University Langone Medical Center, New York, New York; Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minnesota; Unité de Recherche en Pneumologie, Centre de Recherche de l’Hôpital Laval, Institut Universitaire de Cardiologie et de Pneumologie de l’Université Laval; Québec, Canada; Integrated Diagnostics, Seattle, Washington; Respiratory Institute, Cleveland Clinic, Cleveland, Ohio; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, Washington; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Veterans Administration Eastern Colorado Healthcare System, University of Colorado Denver School of Medicine, Denver, Colorado; Statistics Consultant, Carlsbad, California; Thoracic Program, Vanderbilt-Ingram Comprehensive Cancer Center, Nashville, Tennessee; Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville Campus, Nashville, Tennessee
| | - David K. Madtes
- Division of Pulmonary, Allergy, and Critical Care Medicine, Penn Lung Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Cardiothoracic Surgery, New York University Langone Medical Center, New York, New York; Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine and Department of Environmental Medicine, New York University School of Medicine, New York University Langone Medical Center, New York, New York; Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minnesota; Unité de Recherche en Pneumologie, Centre de Recherche de l’Hôpital Laval, Institut Universitaire de Cardiologie et de Pneumologie de l’Université Laval; Québec, Canada; Integrated Diagnostics, Seattle, Washington; Respiratory Institute, Cleveland Clinic, Cleveland, Ohio; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, Washington; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Veterans Administration Eastern Colorado Healthcare System, University of Colorado Denver School of Medicine, Denver, Colorado; Statistics Consultant, Carlsbad, California; Thoracic Program, Vanderbilt-Ingram Comprehensive Cancer Center, Nashville, Tennessee; Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville Campus, Nashville, Tennessee
| | - York E. Miller
- Division of Pulmonary, Allergy, and Critical Care Medicine, Penn Lung Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Cardiothoracic Surgery, New York University Langone Medical Center, New York, New York; Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine and Department of Environmental Medicine, New York University School of Medicine, New York University Langone Medical Center, New York, New York; Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minnesota; Unité de Recherche en Pneumologie, Centre de Recherche de l’Hôpital Laval, Institut Universitaire de Cardiologie et de Pneumologie de l’Université Laval; Québec, Canada; Integrated Diagnostics, Seattle, Washington; Respiratory Institute, Cleveland Clinic, Cleveland, Ohio; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, Washington; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Veterans Administration Eastern Colorado Healthcare System, University of Colorado Denver School of Medicine, Denver, Colorado; Statistics Consultant, Carlsbad, California; Thoracic Program, Vanderbilt-Ingram Comprehensive Cancer Center, Nashville, Tennessee; Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville Campus, Nashville, Tennessee
| | - Michael G. Walker
- Division of Pulmonary, Allergy, and Critical Care Medicine, Penn Lung Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Cardiothoracic Surgery, New York University Langone Medical Center, New York, New York; Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine and Department of Environmental Medicine, New York University School of Medicine, New York University Langone Medical Center, New York, New York; Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minnesota; Unité de Recherche en Pneumologie, Centre de Recherche de l’Hôpital Laval, Institut Universitaire de Cardiologie et de Pneumologie de l’Université Laval; Québec, Canada; Integrated Diagnostics, Seattle, Washington; Respiratory Institute, Cleveland Clinic, Cleveland, Ohio; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, Washington; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Veterans Administration Eastern Colorado Healthcare System, University of Colorado Denver School of Medicine, Denver, Colorado; Statistics Consultant, Carlsbad, California; Thoracic Program, Vanderbilt-Ingram Comprehensive Cancer Center, Nashville, Tennessee; Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville Campus, Nashville, Tennessee
| | - Jing Shi
- Division of Pulmonary, Allergy, and Critical Care Medicine, Penn Lung Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Cardiothoracic Surgery, New York University Langone Medical Center, New York, New York; Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine and Department of Environmental Medicine, New York University School of Medicine, New York University Langone Medical Center, New York, New York; Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minnesota; Unité de Recherche en Pneumologie, Centre de Recherche de l’Hôpital Laval, Institut Universitaire de Cardiologie et de Pneumologie de l’Université Laval; Québec, Canada; Integrated Diagnostics, Seattle, Washington; Respiratory Institute, Cleveland Clinic, Cleveland, Ohio; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, Washington; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Veterans Administration Eastern Colorado Healthcare System, University of Colorado Denver School of Medicine, Denver, Colorado; Statistics Consultant, Carlsbad, California; Thoracic Program, Vanderbilt-Ingram Comprehensive Cancer Center, Nashville, Tennessee; Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville Campus, Nashville, Tennessee
| | - Paul Kearney
- Division of Pulmonary, Allergy, and Critical Care Medicine, Penn Lung Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Cardiothoracic Surgery, New York University Langone Medical Center, New York, New York; Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine and Department of Environmental Medicine, New York University School of Medicine, New York University Langone Medical Center, New York, New York; Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minnesota; Unité de Recherche en Pneumologie, Centre de Recherche de l’Hôpital Laval, Institut Universitaire de Cardiologie et de Pneumologie de l’Université Laval; Québec, Canada; Integrated Diagnostics, Seattle, Washington; Respiratory Institute, Cleveland Clinic, Cleveland, Ohio; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, Washington; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Veterans Administration Eastern Colorado Healthcare System, University of Colorado Denver School of Medicine, Denver, Colorado; Statistics Consultant, Carlsbad, California; Thoracic Program, Vanderbilt-Ingram Comprehensive Cancer Center, Nashville, Tennessee; Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville Campus, Nashville, Tennessee
| | - Kenneth C. Fang
- Division of Pulmonary, Allergy, and Critical Care Medicine, Penn Lung Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Cardiothoracic Surgery, New York University Langone Medical Center, New York, New York; Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine and Department of Environmental Medicine, New York University School of Medicine, New York University Langone Medical Center, New York, New York; Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minnesota; Unité de Recherche en Pneumologie, Centre de Recherche de l’Hôpital Laval, Institut Universitaire de Cardiologie et de Pneumologie de l’Université Laval; Québec, Canada; Integrated Diagnostics, Seattle, Washington; Respiratory Institute, Cleveland Clinic, Cleveland, Ohio; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, Washington; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Veterans Administration Eastern Colorado Healthcare System, University of Colorado Denver School of Medicine, Denver, Colorado; Statistics Consultant, Carlsbad, California; Thoracic Program, Vanderbilt-Ingram Comprehensive Cancer Center, Nashville, Tennessee; Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville Campus, Nashville, Tennessee
| | - Pierre P. Massion
- Division of Pulmonary, Allergy, and Critical Care Medicine, Penn Lung Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Cardiothoracic Surgery, New York University Langone Medical Center, New York, New York; Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine and Department of Environmental Medicine, New York University School of Medicine, New York University Langone Medical Center, New York, New York; Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minnesota; Unité de Recherche en Pneumologie, Centre de Recherche de l’Hôpital Laval, Institut Universitaire de Cardiologie et de Pneumologie de l’Université Laval; Québec, Canada; Integrated Diagnostics, Seattle, Washington; Respiratory Institute, Cleveland Clinic, Cleveland, Ohio; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, Washington; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Veterans Administration Eastern Colorado Healthcare System, University of Colorado Denver School of Medicine, Denver, Colorado; Statistics Consultant, Carlsbad, California; Thoracic Program, Vanderbilt-Ingram Comprehensive Cancer Center, Nashville, Tennessee; Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville Campus, Nashville, Tennessee
| |
Collapse
|
17
|
Hu MM, Hu Y, Zhang HQ, Jia WY, Qian Z, Yang Y, Li BL. Clinical significance of serum and tumor tissue endostatin evaluation in operable non-small cell lung cancer. Biomed Rep 2014; 2:898-904. [PMID: 25279166 DOI: 10.3892/br.2014.319] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2014] [Accepted: 07/15/2014] [Indexed: 12/31/2022] Open
Abstract
Endostatin, as the most potential antiangiogenic factor, is a naturally occurring fragment of collagen XVIII in bloodstream capable of inhibiting tumor growth and metastasis. This study was conducted to explore the clinical value of endostatin in serum and tumor tissue in patients with operable non-small cell lung cancer (NSCLC). ELISA and immunohistochemistry were applied to detect the expression of endostatin in serum and tumor tissue in 105 patient-matched operable NSCLC patients. The serum level of endostatin was significantly higher in NSCLC patients than healthy individuals (P=0.0018). Cases with poorer differentiation showed a higher endostatin serum level (P=0.008). There was no significant correlation between tumor tissue expression and clinical parameters, such as TNM stage, differentiation degree, histological type and lymph node invasion status. A stronger expression of endostain in tumor tissue was associated with a higher serum level (r=0.223). The univariate and multivariate analyses with Cox proportional hazards model for overall survival showed that tumor stage and node status were independent prognostic factors, whereas neither endostatin levels in serum nor in tumor tissue showed potential in predicting the long-term survival of operable NSCLC patients. In conclusion, the results observed in the present study did not support the prediction of overall survival in operable NSCLC based on the expression levels of endostatin in serum and tumor tissue.
Collapse
Affiliation(s)
- Ming-Ming Hu
- Department of General Medicine, Beijing Tuberculosis and Thoracic Tumor Research Institute/Beijing Chest Hospital, Capital Medical University, Beijing 101149, P.R. China
| | - Ying Hu
- Department of General Medicine, Beijing Tuberculosis and Thoracic Tumor Research Institute/Beijing Chest Hospital, Capital Medical University, Beijing 101149, P.R. China
| | - Hai-Qing Zhang
- Department of Pathology, Beijing Tuberculosis and Thoracic Tumor Research Institute/Beijing Chest Hospital, Capital Medical University, Beijing 101149, P.R. China
| | - Wen-Yun Jia
- Department of General Medicine, Beijing Tuberculosis and Thoracic Tumor Research Institute/Beijing Chest Hospital, Capital Medical University, Beijing 101149, P.R. China
| | - Zhe Qian
- Department of General Medicine, Beijing Tuberculosis and Thoracic Tumor Research Institute/Beijing Chest Hospital, Capital Medical University, Beijing 101149, P.R. China
| | - Yuan Yang
- Department of General Medicine, Beijing Tuberculosis and Thoracic Tumor Research Institute/Beijing Chest Hospital, Capital Medical University, Beijing 101149, P.R. China
| | - Bao-Lan Li
- Department of General Medicine, Beijing Tuberculosis and Thoracic Tumor Research Institute/Beijing Chest Hospital, Capital Medical University, Beijing 101149, P.R. China
| |
Collapse
|
18
|
Li XJ, Hayward C, Fong PY, Dominguez M, Hunsucker SW, Lee LW, McLean M, Law S, Butler H, Schirm M, Gingras O, Lamontagne J, Allard R, Chelsky D, Price ND, Lam S, Massion PP, Pass H, Rom WN, Vachani A, Fang KC, Hood L, Kearney P. A blood-based proteomic classifier for the molecular characterization of pulmonary nodules. Sci Transl Med 2014; 5:207ra142. [PMID: 24132637 DOI: 10.1126/scitranslmed.3007013] [Citation(s) in RCA: 147] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Each year, millions of pulmonary nodules are discovered by computed tomography and subsequently biopsied. Because most of these nodules are benign, many patients undergo unnecessary and costly invasive procedures. We present a 13-protein blood-based classifier that differentiates malignant and benign nodules with high confidence, thereby providing a diagnostic tool to avoid invasive biopsy on benign nodules. Using a systems biology strategy, we identified 371 protein candidates and developed a multiple reaction monitoring (MRM) assay for each. The MRM assays were applied in a three-site discovery study (n = 143) on plasma samples from patients with benign and stage IA lung cancer matched for nodule size, age, gender, and clinical site, producing a 13-protein classifier. The classifier was validated on an independent set of plasma samples (n = 104), exhibiting a negative predictive value (NPV) of 90%. Validation performance on samples from a nondiscovery clinical site showed an NPV of 94%, indicating the general effectiveness of the classifier. A pathway analysis demonstrated that the classifier proteins are likely modulated by a few transcription regulators (NF2L2, AHR, MYC, and FOS) that are associated with lung cancer, lung inflammation, and oxidative stress networks. The classifier score was independent of patient nodule size, smoking history, and age, which are risk factors used for clinical management of pulmonary nodules. Thus, this molecular test provides a potential complementary tool to help physicians in lung cancer diagnosis.
Collapse
Affiliation(s)
- Xiao-jun Li
- Integrated Diagnostics, Seattle, WA 98109, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Hu Y, Hu MM, Shi GL, Han Y, Li BL. Imbalance between vascular endothelial growth factor and endostatin correlates with the prognosis of operable non-small cell lung cancer. Eur J Surg Oncol 2014; 40:1136-42. [PMID: 24998915 DOI: 10.1016/j.ejso.2014.05.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Revised: 05/16/2014] [Accepted: 05/22/2014] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Angiogenesis is regulated by a balance of pro-angiogenic and anti-angiogenic factors. Vascular endothelial growth factor (VEGF) and endostatin respectively represents a frequent component of inducers and inhibitors in the process of angiogenesis. The ratio of VEGF/endostatin may reflect the balance of angiogenic switch. This study aimed to determine whether an imbalance between VEGF/endostatin exists in operable non-small cell lung cancer (NSCLC) patients and to assess the correlation, if any, between the imbalance and the prognosis. METHODS Preoperative serum levels of VEGF and endostatin were simultaneously determined by quantitiative enzyme-linked immunosorbent assay (ELISA) and the ratio of them was calculated among 98 NSCLC patients and 51 healthy controls. The relationship between these factors and clinicopathological features, including prognosis, was examined. RESULTS The ratio of VEGF/endostatin levels was significantly higher in operable NSCLC patients [median, 10.4; interquartile range (IQR), 5.9-19.8] than in normal controls [median, 5.1; IQR, 3.3-9.7] (P = 0.002). While the ratio in patients who were still alive for more than 60 months was 8.3 (IQR, 4.3-17.9), the ratio in those who died was 12.9 (IQR, 8.0-22.1) (p = 0.017). In subgroup analysis of patients with pathological stage N0, there was a statistically significant increase of the survival time in the group with a lower ratio than in the group with a higher ratio (p = 0.032). Multivariate analysis confirmed that the VEGF/endostatin ratio was an independent prognostic factor (p = 0.018). CONCLUSION There was an imbalance between VEGF and endostatin in serum of operable NSCLC patients. The imbalance correlated with the prognosis of operable NSCLC.
Collapse
Affiliation(s)
- Y Hu
- Department of General Medicine, Beijing Chest Hospital, Capital Medical University & Beijing Tuberculosis and Thoracic Tumor Research Institute, 101149, China
| | - M-m Hu
- Department of General Medicine, Beijing Chest Hospital, Capital Medical University & Beijing Tuberculosis and Thoracic Tumor Research Institute, 101149, China
| | - G-L Shi
- Department of Clinical Laboratory, Beijing Chest Hospital, Capital Medical University & Beijing Tuberculosis and Thoracic Tumor Research Institute, China
| | - Y Han
- Department of Thoracic Surgery, Beijing Chest Hospital, Capital Medical University & Beijing Tuberculosis and Thoracic Tumor Research Institute, China
| | - B-L Li
- Department of General Medicine, Beijing Chest Hospital, Capital Medical University & Beijing Tuberculosis and Thoracic Tumor Research Institute, 101149, China.
| |
Collapse
|
20
|
Chen P, Zhu J, Liu DY, Li HY, Xu N, Hou M. Over-expression of survivin and VEGF in small-cell lung cancer may predict the poorer prognosis. Med Oncol 2013; 31:775. [PMID: 24338338 DOI: 10.1007/s12032-013-0775-5] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2013] [Accepted: 11/13/2013] [Indexed: 01/14/2023]
Abstract
The expression of survivin, an inhibitor of apoptosis can be seen in most tumors and is correlated with the angiogenic factor vascular endothelial growth factor (VEGF). But little is known about their contribution in small-cell lung cancer (SCLC). This study was designed to investigate the expression of survivin and VEGF in SCLC, and to explore their correlation with clinical-pathological feature and prognosis. Forty-five patients with pathological histology of SCLC were entered into this study. Forty-five cases of matched adjacent non-tumor samples and 10 samples of operated patients with benign lung tumor were also included as control. The expression of survivin and VEGF was detected by immunohistochemistry (IHC, SP). These two sets of data were processed and tested for correlation with major patients' characteristics, and overall survival. The correlations between survivin and VEGF expressions and the clinical-pathological features were evaluated by chi-square test. The correlation between survivin and VEGF expressions was analyzed by Spearman's rank correlation test; the overall survival was analyzed by the Kaplan-Meier method; and the relationship between clinical and pathological features and overall survival was analyzed by the Cox proportional hazard models. Positive expression rate of survivin and VEGF was significantly higher in SCLC than those of adjacent non-tumor tissues and benign lung tumor tissues (73.3 vs. 15.6 vs. 0 %, P < 0.05) and (75.6 vs. 20 vs. 0 %, P < 0.05), respectively. Survivin and VEGF expressions were significantly associated with lymph node metastasis (P = 0.003, 0.011) and clinical stage (P = 0.006, 0.021). The expression of survivin was significantly coincident with the expression of VEGF (r = 0.644, P = 0.000). The median overall survival in survivin positive group and VEGF positive group was significantly shorter than those in survivin negative and VEGF negative group, respectively (log-rank P = 0.000). Moreover, multivariate analysis showed that survivin expression (HR 0.224; 95 % CI 0.074-0.675; P = 0.008) and VEGF expression (HR 0.172; 95 % CI 0.054-0.559; P = 0.003) were statistically independent predictive factors of poorer prognosis for SCLC patients. Our results indicated that survivin and VEGF were over-expressed in small-cell lung cancer, each of them may be an independent poor prognostic factor.
Collapse
Affiliation(s)
- Ping Chen
- Department of Oncology, Chengdu Seventh People's Hospital, Chengdu Cancer Hospital, Chengdu, 610041, China,
| | | | | | | | | | | |
Collapse
|
21
|
Wang L, Stueckle TA, Mishra A, Derk R, Meighan T, Castranova V, Rojanasakul Y. Neoplastic-like transformation effect of single-walled and multi-walled carbon nanotubes compared to asbestos on human lung small airway epithelial cells. Nanotoxicology 2013; 8:485-507. [PMID: 23634900 DOI: 10.3109/17435390.2013.801089] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Accumulating evidence indicates that carbon nanotubes (CNTs) are biopersistent and can cause lung damage. With similar fibrous morphology and mode of exposure to asbestos, a known human carcinogen, growing concern has arisen for elevated risk of CNT-induced lung carcinogenesis; however, relatively little is known about the long-term carcinogenic effect of CNT. Neoplastic transformation is a key early event leading to carcinogenesis. We studied the ability of single- and multi-walled CNTs to induce neoplastic transformation of human lung epithelial cells compared to asbestos. Long-term (6-month) exposure of the cells to occupationally relevant concentrations of CNT in culture caused a neoplastic-like transformation phenotype as demonstrated by increased cell proliferation, anchorage-independent growth, invasion and angiogenesis. Whole-genome expression signature and protein expression analyses showed that single- and multi-walled CNTs shared similar signaling signatures which were distinct from asbestos. These results provide novel toxicogenomic information and suggest distinct particle-associated mechanisms of neoplasia promotion induced by CNTs and asbestos.
Collapse
Affiliation(s)
- Liying Wang
- HELD/PPRB, National Institute for Occupational Safety and Health , Morgantown, WV 26505 , USA
| | | | | | | | | | | | | |
Collapse
|
22
|
Mehan MR, Ayers D, Thirstrup D, Xiong W, Ostroff RM, Brody EN, Walker JJ, Gold L, Jarvis TC, Janjic N, Baird GS, Wilcox SK. Protein signature of lung cancer tissues. PLoS One 2012; 7:e35157. [PMID: 22509397 PMCID: PMC3324437 DOI: 10.1371/journal.pone.0035157] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2011] [Accepted: 03/09/2012] [Indexed: 12/13/2022] Open
Abstract
Lung cancer remains the most common cause of cancer-related mortality. We applied a highly multiplexed proteomic technology (SOMAscan) to compare protein expression signatures of non small-cell lung cancer (NSCLC) tissues with healthy adjacent and distant tissues from surgical resections. In this first report of SOMAscan applied to tissues, we highlight 36 proteins that exhibit the largest expression differences between matched tumor and non-tumor tissues. The concentrations of twenty proteins increased and sixteen decreased in tumor tissue, thirteen of which are novel for NSCLC. NSCLC tissue biomarkers identified here overlap with a core set identified in a large serum-based NSCLC study with SOMAscan. We show that large-scale comparative analysis of protein expression can be used to develop novel histochemical probes. As expected, relative differences in protein expression are greater in tissues than in serum. The combined results from tissue and serum present the most extensive view to date of the complex changes in NSCLC protein expression and provide important implications for diagnosis and treatment.
Collapse
Affiliation(s)
| | - Deborah Ayers
- SomaLogic, Inc., Boulder, Colorado, United States of America
| | - Derek Thirstrup
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, United States of America
| | - Wei Xiong
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, United States of America
| | | | - Edward N. Brody
- SomaLogic, Inc., Boulder, Colorado, United States of America
| | | | - Larry Gold
- SomaLogic, Inc., Boulder, Colorado, United States of America
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Colorado, United States of America
| | - Thale C. Jarvis
- SomaLogic, Inc., Boulder, Colorado, United States of America
| | - Nebojsa Janjic
- SomaLogic, Inc., Boulder, Colorado, United States of America
| | - Geoffrey S. Baird
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, United States of America
| | - Sheri K. Wilcox
- SomaLogic, Inc., Boulder, Colorado, United States of America
- * E-mail:
| |
Collapse
|
23
|
Smit EF, Socinski MA, Mullaney BP, Myrand SP, Scagliotti GV, Lorigan P, Reck M, Ciuleanu T, von Pawel J, Karaseva NA, Szczesna A, Ohannesian D, Powell E, Hozak RR, Hong S, Guba SC, Thatcher N. Biomarker analysis in a phase III study of pemetrexed-carboplatin versus etoposide-carboplatin in chemonaive patients with extensive-stage small-cell lung cancer. Ann Oncol 2011; 23:1723-9. [PMID: 22186609 DOI: 10.1093/annonc/mdr563] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Clinical results of a randomized phase III trial comparing pemetrexed-carboplatin (PC) with etoposide-carboplatin (EC) in chemonaive patients with extensive-stage disease small-cell lung cancer (ED-SCLC) resulted in trial closure for futility; biomarker analyses using immunohistochemistry (IHC) and single-nucleotide polymorphisms (SNPs) are described herein. PATIENTS AND METHODS Thymidylate synthase (TS), excision repair cross complementing-1 (ERCC1), glycinamide ribonucleotide formyltransferase (GARFT), and folylpolyglutamate synthetase (FPGS) were investigated using IHC (n=395). SNPs were genotyped for TS, FPGS, γ-glutamyl hydrolase (GGH), methylenetetrahydrofolate reductase (MTHFR), folate receptor-α FR-α, and solute carrier 19A1 (SLC19A1; n=611). RESULTS None of the IHC biomarkers (folate pathway or ERCC1) were found to be predictive or prognostic in this setting. rs2838952 (adjacent to SLC19A1) had significant treatment-independent association with overall survival (OS; hazard ratio 0.590, P=0.01). Nine GGH-associated SNPs interacted with rs3788205 (SLC19A1) for OS on the PC arm. rs12379987 (FPGS) interacted with treatment for OS (interaction P=0.036). CONCLUSION Potential ERCC1 and folate pathway IHC biomarkers failed to predict outcome in either study arm in ED-SCLC. SNPs in regions including FPGS and SLC19A1 and interacting SNPs in GGH and SLC19A1 were associated with differences in OS; however, none of these SNPs predicted for greater survival with PC over EC.
Collapse
Affiliation(s)
- E F Smit
- Department of Pulmonary Diseases, Vrije University Medical Center, Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Boosani CS, Sudhakar YA. Proteolytically Derived Endogenous Angioinhibitors Originating from the Extracellular Matrix. Pharmaceuticals (Basel) 2011; 4:1551-1577. [PMID: 22267953 PMCID: PMC3260939 DOI: 10.3390/ph4121551] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Angiogenesis, a neovascularization process induced from the existing parent blood vessels, is a prerequisite for many physiological and pathological conditions. Under physiological conditions it is regulated by a balance between endogenous angioinhibitors and angioactivators, and an imbalance between them would lead to pathological conditions such as cancer, age-related macular degeneration (AMD), diabetic retinopathy, cardiovascular diseases, etc. Several proteolytically generated endogenous molecules have been identified which exhibit angioinhibition and/or antitumor activities. These angioinhibitors interact with endothelial and tumor cells by binding to distinct integrins and initiate many of their intracellular signaling mechanisms regulating the cell survival and or apoptotic pathways. The present review will focus on the extracellular matrix derived angioinhibitors, and their mechanisms of actions that point to the clinical significance and therapeutic implications.
Collapse
Affiliation(s)
- Chandra Shekhar Boosani
- Cell Signaling, Retinal and Tumor Angiogenesis Laboratory, Department of Genetics, Boys Town National Research Hospital, Omaha, NE 68131, USA; E-Mail:
| | - Yakkanti A. Sudhakar
- Cell Signaling, Retinal and Tumor Angiogenesis Laboratory, Department of Genetics, Boys Town National Research Hospital, Omaha, NE 68131, USA; E-Mail:
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-402-498-6681; Fax: +1-402-498-6331
| |
Collapse
|
25
|
Szarvas T, László V, Vom Dorp F, Reis H, Szendröi A, Romics I, Tilki D, Rübben H, Ergün S. Serum endostatin levels correlate with enhanced extracellular matrix degradation and poor patients' prognosis in bladder cancer. Int J Cancer 2011; 130:2922-9. [PMID: 21815140 DOI: 10.1002/ijc.26343] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2011] [Accepted: 07/20/2011] [Indexed: 01/13/2023]
Abstract
Endostatin, the proteolytic fragment of collagen XVIII, is an inhibitor of angiogenesis and tumor growth. Interestingly, elevated circulating endostatin levels have been found to correlate with poor patients' prognosis in several cancers. The aim of this study was to assess the prognostic value of endostatin in bladder cancer (BC) and to gain insight into the mechanisms involved in its production. This retrospective study included a total of 337 patients with BC and 103 controls. Collagen XVIII gene expression was analyzed using real-time PCR (n = 82). Endostatin tissue localization was assessed by immunohistochemistry (n = 27). Endostatin serum (n = 87) and urine (n = 153) levels were determined by ELISA. In 12 cases, both serum and paraffinized tissue samples from the same patients were available. We found decreased collagen XVIII tissue expression and increased endostatin urine and serum concentration in samples of patients with BC compared to controls. High serum endostatin levels correlated with the presence of lymph node metastases and MMP-7 concentrations and were independently associated with poor metastasis-free and disease-specific survival. Immunohistochemical analysis revealed a strong endostatin staining in the wall of tumor associated blood vessels in superficial but not in muscle-invasive BCs. Based on these, we concluded that elevated endostatin levels in patients with BC are the consequence of enhanced extracellular matrix degradation and are independent from collagen XVIII expression. Furthermore, serum endostatin levels may provide prognostic information independent from histopathological parameters and may therefore help to optimize therapy decisions. Loss of endostatin expression in tumor associated blood vessels might represent an important step supporting tumor-induced angiogenesis.
Collapse
Affiliation(s)
- Tibor Szarvas
- Department of Urology, University of Duisburg-Essen, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Salgia R. Prognostic significance of angiogenesis and angiogenic growth factors in nonsmall cell lung cancer. Cancer 2011; 117:3889-99. [PMID: 21858799 PMCID: PMC3160199 DOI: 10.1002/cncr.25935] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2010] [Revised: 11/30/2010] [Accepted: 12/16/2010] [Indexed: 01/22/2023]
Abstract
Currently, nonsmall-cell lung cancer (NSCLC) is the leading cause of cancer-related death in the United States. Angiogenesis, the formation of new vasculature, is a complex and tightly regulated process that promotes metastasis and disease progression in lung cancer and other malignancies. Developmental antiangiogenic agents have shown activity in NSCLC, and bevacizumab, an antiangiogenic monoclonal antibody, is approved for the treatment of patients with advanced disease. However, predictive biomarkers are needed to guide the administration of antiangiogenic agents. It is possible that angiogenic molecules could accurately predict patient response to targeted antiangiogenic therapies, which would allow individualized and perhaps more effective treatment. Angiogenic signaling molecules may also have value as prognostic indicators, which may be useful for the management of NSCLC. Here the author provides an overview of angiogenic molecules currently being investigated as prognostic biomarkers in NSCLC and discusses their potential to guide treatment choices.
Collapse
Affiliation(s)
- Ravi Salgia
- Department of Medicine, Section of Hematology/Oncology, The University of Chicago, Chicago, Illinois, USA.
| |
Collapse
|
27
|
Mahajan VB, Olney AH, Garrett P, Chary A, Dragan E, Lerner G, Murray J, Bassuk AG. Collagen XVIII mutation in Knobloch syndrome with acute lymphoblastic leukemia. Am J Med Genet A 2011; 152A:2875-9. [PMID: 20799329 DOI: 10.1002/ajmg.a.33621] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Knobloch syndrome (KNO) is caused by mutations in the collagen XVIII gene (COL18A1) and patients develop encephalocele and vitreoretinal degeneration. Here, we report an El Salvadorian family where two sisters showed features of KNO. One of the siblings also developed acute lymphoblastic leukemia. DNA sequencing of COL18A1 revealed a homozygous, 2-bp deletion (c3514-3515delCT) in exon 41, which leads to abnormal collagen XVIII and deficiency of its proteolytic cleavage product endostatin. KNO patients with mutations in COL18A1 may be at risk for endostatin-related conditions including malignancy.
Collapse
Affiliation(s)
- Vinit B Mahajan
- Department of Ophthalmology and Visual Sciences, The University of Iowa Hospitals & Clinics, Iowa City, Iowa, USA
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Ostroff RM, Bigbee WL, Franklin W, Gold L, Mehan M, Miller YE, Pass HI, Rom WN, Siegfried JM, Stewart A, Walker JJ, Weissfeld JL, Williams S, Zichi D, Brody EN. Unlocking biomarker discovery: large scale application of aptamer proteomic technology for early detection of lung cancer. PLoS One 2010; 5:e15003. [PMID: 21170350 PMCID: PMC2999620 DOI: 10.1371/journal.pone.0015003] [Citation(s) in RCA: 166] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2010] [Accepted: 10/07/2010] [Indexed: 02/08/2023] Open
Abstract
Background Lung cancer is the leading cause of cancer deaths worldwide. New diagnostics are needed to detect early stage lung cancer because it may be cured with surgery. However, most cases are diagnosed too late for curative surgery. Here we present a comprehensive clinical biomarker study of lung cancer and the first large-scale clinical application of a new aptamer-based proteomic technology to discover blood protein biomarkers in disease. Methodology/Principal Findings We conducted a multi-center case-control study in archived serum samples from 1,326 subjects from four independent studies of non-small cell lung cancer (NSCLC) in long-term tobacco-exposed populations. Sera were collected and processed under uniform protocols. Case sera were collected from 291 patients within 8 weeks of the first biopsy-proven lung cancer and prior to tumor removal by surgery. Control sera were collected from 1,035 asymptomatic study participants with ≥10 pack-years of cigarette smoking. We measured 813 proteins in each sample with a new aptamer-based proteomic technology, identified 44 candidate biomarkers, and developed a 12-protein panel (cadherin-1, CD30 ligand, endostatin, HSP90α, LRIG3, MIP-4, pleiotrophin, PRKCI, RGM-C, SCF-sR, sL-selectin, and YES) that discriminates NSCLC from controls with 91% sensitivity and 84% specificity in cross-validated training and 89% sensitivity and 83% specificity in a separate verification set, with similar performance for early and late stage NSCLC. Conclusions/Significance This study is a significant advance in clinical proteomics in an area of high unmet clinical need. Our analysis exceeds the breadth and dynamic range of proteome interrogated of previously published clinical studies of broad serum proteome profiling platforms including mass spectrometry, antibody arrays, and autoantibody arrays. The sensitivity and specificity of our 12-biomarker panel improves upon published protein and gene expression panels. Separate verification of classifier performance provides evidence against over-fitting and is encouraging for the next development phase, independent validation. This careful study provides a solid foundation to develop tests sorely needed to identify early stage lung cancer.
Collapse
Affiliation(s)
- Rachel M. Ostroff
- SomaLogic, Boulder, Colorado, United States of America
- * E-mail: (RMO); (JJW)
| | - William L. Bigbee
- Department of Pathology, University of Pittsburgh School of Medicine, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, United States of America
| | - Wilbur Franklin
- University of Colorado Cancer Center, University of Colorado at Denver, Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Larry Gold
- SomaLogic, Boulder, Colorado, United States of America
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Colorado, United States of America
| | - Mike Mehan
- SomaLogic, Boulder, Colorado, United States of America
| | - York E. Miller
- University of Colorado Cancer Center, University of Colorado at Denver, Anschutz Medical Campus, Aurora, Colorado, United States of America
- Denver Veterans Affairs Medical Center, Denver, Colorado, United States of America
| | - Harvey I. Pass
- Langone Medical Center and Cancer Center, New York University School of Medicine, New York, New York, United States of America
| | - William N. Rom
- Division of Pulmonary, and Critical Care, and Sleep Medicine, New York University School of Medicine, New York, New York, United States of America
| | - Jill M. Siegfried
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, United States of America
| | - Alex Stewart
- SomaLogic, Boulder, Colorado, United States of America
| | - Jeffrey J. Walker
- SomaLogic, Boulder, Colorado, United States of America
- * E-mail: (RMO); (JJW)
| | - Joel L. Weissfeld
- Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, United States of America
| | | | - Dom Zichi
- SomaLogic, Boulder, Colorado, United States of America
| | | |
Collapse
|
29
|
Abstract
Endogenous inhibitors of angiogenesis are proteins or fragments of proteins that are formed in the body, which can inhibit the angiogenic process. These molecules can be found both in the circulation and sequestered in the extracellular matrix (ECM) surrounding cells. Many matrix-derived inhibitors of angiogenesis, such as endostatin, tumstatin, canstatin and arresten, are bioactive fragments of larger ECM molecules. These substances become released upon proteolysis of the ECM and the vascular basement membrane (VBM) by enzymes of the tumor microenvironment. Although the role of matrix-derived angiogenesis inhibitors is well studied in animal models of cancer, their role in human cancers is less established. In this review we discuss the current knowledge about these molecules and their potential use as cancer therapeutics and biomarkers.
Collapse
|
30
|
Spivey KA, Banyard J, Solis LM, Wistuba II, Barletta JA, Gandhi L, Feldman HA, Rodig SJ, Chirieac LR, Zetter BR. Collagen XXIII: a potential biomarker for the detection of primary and recurrent non-small cell lung cancer. Cancer Epidemiol Biomarkers Prev 2010; 19:1362-72. [PMID: 20447926 DOI: 10.1158/1055-9965.epi-09-1095] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Collagen XXIII is a transmembrane collagen previously shown to be upregulated in metastatic prostate cancer. The purpose of this study was to determine the protein expression of collagen XXIII in tumor tissues from a variety of cancers and to assess the utility of collagen XXIII as a biomarker for non-small cell lung cancer (NSCLC). METHODS A multicancer tissue microarray was used for the immunohistochemical examination of collagen XXIII protein expression in a variety of cancers. Subsequently, collagen XXIII expression was analyzed in three separate cohorts using tissue microarrays with representative tumor and control lung tissues from NSCLC patients. In addition, NSCLC patient urine samples were analyzed for the presence of collagen XXIII through Western blot. RESULTS Collagen XXIII was present in tissue samples from a variety of cancers. Within lung cancer tissues, collagen XXIII staining was enriched in NSCLC subtypes. Collagen XXIII was present in 294 of 333 (88%) lung adenocarcinomas and 97 of 133 (73%) squamous cell carcinomas. In urine, collagen XXIII was present in 23 of 29 (79%) NSCLC patient samples but only in 15 of 54 (28%) control samples. High collagen XXIII staining intensity correlated with shorter recurrence-free survival in NSCLC patients. CONCLUSIONS We show the capability of collagen XXIII as a tissue and urinary biomarker for NSCLC, in which positivity in tissue or urine significantly correlates with the presence of NSCLC and high staining intensity is a significant recurrence predictor. IMPACT Inclusion of collagen XXIII in a tissue- or urine-based cancer biomarker panel could inform NSCLC patient treatment decisions.
Collapse
Affiliation(s)
- Kristin A Spivey
- Department of Surgery, Children's Hospital, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Lee KS, Park GS, Hong SH, Byun JH, Woo IS, Jeon HM, Hong YS. Prognostic relevance of collagen XVIII expression in metastatic gastric carcinoma. Tumour Biol 2010; 31:165-70. [PMID: 20361288 DOI: 10.1007/s13277-010-0022-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2009] [Accepted: 02/10/2010] [Indexed: 01/17/2023] Open
Abstract
Collagen XVIII is a component of vascular and epithelial basement membranes. The C-terminal fragment of the protein is termed endostatin, and is a potent inhibitor of angiogenesis. No reports on the clinical implications of collagen XVIII expression in human gastric cancer are currently available. Here, we investigate the clinical significance of collagen XVIII expression in gastric cancer. Seven gastric cancer cell lines were subjected to Western blotting. Collagen XVIII expression was examined in 118 gastric carcinoma tissues via immunohistochemistry. Western blotting revealed the presence of the 22-kDa collagen XVIII protein in four of seven gastric cancer cell lines. Immunohistochemistry detected collagen XVIII expression in the tumor cytoplasm in 115 of 118 gastric carcinoma patients (97%). No correlation was evident between collagen XVIII expression score and clinicopathologic findings when all patients were considered together. However, on subgroup analysis, 42 of 70 patients with distant metastasis were classified into low or moderate collagen XVIII expression groups, whereas the remaining 28 patients were grouped as showing high collagen XVIII expression. The prognosis for patients with high collagen XVIII-expressing gastric carcinoma was significantly worse than that for patients displaying low or moderate collagen XVIII expression (median survival time, 7.8 months vs. 18.3 months [log-rank, p = 0.01]; median time to progression, 3 months vs. 8 months [log-rank, p = 0.01]). High expression of collagen XVIII is associated with poor prognosis in patients with metastatic gastric carcinoma. Further studies on larger patient populations are warranted to validate the utility of collagen XVIII as a prognostic biomarker in gastric carcinoma.
Collapse
Affiliation(s)
- Kyu Sang Lee
- Catholic Research Institutes of Medical Science, Catholic University, Seoul, South Korea
| | | | | | | | | | | | | |
Collapse
|
32
|
Luo YQ, Li-JuanYao, Zhao L, Sun AY, Dong H, Du JP, Wu SZ, Hu W. Development of an ELISA for quantification of tumstatin in serum samples and tissue extracts of patients with lung carcinoma. Clin Chim Acta 2010; 411:510-5. [DOI: 10.1016/j.cca.2010.01.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2009] [Revised: 12/31/2009] [Accepted: 01/04/2010] [Indexed: 10/20/2022]
|
33
|
Li M, Xiao T, Zhang Y, Feng L, Lin D, Liu Y, Mao Y, Guo S, Han N, Di X, Zhang K, Cheng S, Gao Y. Prognostic significance of matrix metalloproteinase-1 levels in peripheral plasma and tumour tissues of lung cancer patients. Lung Cancer 2010; 69:341-7. [PMID: 20060194 DOI: 10.1016/j.lungcan.2009.12.007] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2009] [Revised: 12/09/2009] [Accepted: 12/11/2009] [Indexed: 11/18/2022]
Abstract
Matrix metalloproteinase-1 (MMP-1) participates in a variety of physiological and pathological processes. We previously found that MMP-1 was one of the lung cancer-related proteins detectable in peripheral blood. To validate our preliminary observations and explore the clinical significance of MMP-1 for lung cancer further, we carried out the present study. The concentrations of MMP-1 in circulating plasma specimens of 170 lung cancer patients and 70 healthy individuals were measured by an enzyme-linked immunosorbance assay. The expression status of the MMP-1 in archival tissue samples from 122 lung cancer patients was examined by immunohistochemical analysis. The correlation between the MMP-1 levels and prognosis of the lung cancer patients was then assessed statistically. Protein levels of MMP-1 were considerably raised in the plasma from lung cancer patients relative to those in healthy controls. The high plasma MMP-1 levels were associated with advanced-stage of the disease and significantly lower overall survival rate of the patients. Coincidently, MMP-1 protein extraordinarily overexpressed in the tumour tissues of lung cancer; and the up-regulated MMP-1 was associated with the progression (including tumour size, staging and lymphatic invasion), especially with decreased survival rate of the patients. Statistic analysis revealed that MMP-1 protein levels had an independent influence on survival. MMP-1 levels were elevated in both tumour tissue and blood; the latter may serve as an independent predictor for survival of lung cancer patients. MMP-1 protein levels in plasma/serum thus represent a potential and clinically relevant biomarker for the prognosis of patients with lung cancers.
Collapse
Affiliation(s)
- Min Li
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, Cancer Institute Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100021, PR China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Hwang KT, Chung JK, Jung IM, Heo SC, Ahn YJ, Ahn HS, Chang MS, Kim JA, Han W, Noh DY. COL18A1 as the Candidate Gene for the Prognostic Marker of Breast Cancer According to the Analysis of the DNA Copy Number Variation by Array CGH. J Breast Cancer 2010. [DOI: 10.4048/jbc.2010.13.1.37] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Affiliation(s)
- Ki-Tae Hwang
- Department of Surgery, Seoul National University Boramae Hospital, Seoul, Korea
| | - Jung Kee Chung
- Department of Surgery, Seoul National University Boramae Hospital, Seoul, Korea
| | - In Mok Jung
- Department of Surgery, Seoul National University Boramae Hospital, Seoul, Korea
| | - Seung Chul Heo
- Department of Surgery, Seoul National University Boramae Hospital, Seoul, Korea
| | - Young Joon Ahn
- Department of Surgery, Seoul National University Boramae Hospital, Seoul, Korea
| | - Hye Seong Ahn
- Department of Surgery, Seoul National University Boramae Hospital, Seoul, Korea
| | - Mee Soo Chang
- Department of Pathology, Seoul National University Boramae Hospital, Seoul, Korea
| | - Jeong-Ah Kim
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul, Korea
| | - Wonshik Han
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul, Korea
- Department of Surgery, College of Medicine, Seoul National University, Seoul, Korea
| | - Dong-Young Noh
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul, Korea
- Department of Surgery, College of Medicine, Seoul National University, Seoul, Korea
| |
Collapse
|
35
|
Kague E, Bessling SL, Lee J, Hu G, Passos-Bueno MR, Fisher S. Functionally conserved cis-regulatory elements of COL18A1 identified through zebrafish transgenesis. Dev Biol 2009; 337:496-505. [PMID: 19895802 DOI: 10.1016/j.ydbio.2009.10.028] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2009] [Revised: 10/13/2009] [Accepted: 10/21/2009] [Indexed: 11/26/2022]
Abstract
Type XVIII collagen is a component of basement membranes, and expressed prominently in the eye, blood vessels, liver, and the central nervous system. Homozygous mutations in COL18A1 lead to Knobloch Syndrome, characterized by ocular defects and occipital encephalocele. However, relatively little has been described on the role of type XVIII collagen in development, and nothing is known about the regulation of its tissue-specific expression pattern. We have used zebrafish transgenesis to identify and characterize cis-regulatory sequences controlling expression of the human gene. Candidate enhancers were selected from non-coding sequence associated with COL18A1 based on sequence conservation among mammals. Although these displayed no overt conservation with orthologous zebrafish sequences, four regions nonetheless acted as tissue-specific transcriptional enhancers in the zebrafish embryo, and together recapitulated the major aspects of col18a1 expression. Additional post-hoc computational analysis on positive enhancer sequences revealed alignments between mammalian and teleost sequences, which we hypothesize predict the corresponding zebrafish enhancers; for one of these, we demonstrate functional overlap with the orthologous human enhancer sequence. Our results provide important insight into the biological function and regulation of COL18A1, and point to additional sequences that may contribute to complex diseases involving COL18A1. More generally, we show that combining functional data with targeted analyses for phylogenetic conservation can reveal conserved cis-regulatory elements in the large number of cases where computational alignment alone falls short.
Collapse
Affiliation(s)
- Erika Kague
- Centro de Estudo do Genoma Humano, Department Genetica e Biologia Evolutiva, Instituto de Biociencias/Universidade de São Paulo, Brazil
| | | | | | | | | | | |
Collapse
|
36
|
Sen E, Ulger F, Kaya A, Akar N, Gonullu U. Serum Endothelial Monocyte-Activating Polypeptide–II: A Novel Biomarker in Patients with Non–Small-Cell Lung Cancer. Clin Lung Cancer 2008; 9:166-70. [DOI: 10.3816/clc.2008.n.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
37
|
Eren B, Sar M, Oz B, Dincbas FHO. MMP-2, TIMP-2 and CD44v6 Expression in Non-small-cell Lung Carcinomas. ANNALS OF THE ACADEMY OF MEDICINE, SINGAPORE 2008. [DOI: 10.47102/annals-acadmedsg.v37n1p32] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Introduction: Factors that emerge as crucial participants in tumour invasion and metastases are matrix metalloproteinases (MMPs), tissue inhibitor of metalloproteinase (TIMP) inhibitors and cellular adhesion molecules (CD44 and similar molecules). They play important roles in tumour invasion and metastasis in non-small-cell lung carcinomas (NSCLCs).
Materials and Methods: The study was performed using the data of 33 patients. MMP-2 from the metalloproteinase family, TIMP-2 from the metalloproteinase inhibitor family and the adhesion molecule CD44v6 expression were investigated immunohistochemically to search their role in the metastasis and the clinical outcome of the patients with NSCLCs.
Results: Twenty-three tumours (70%) were squamous cell carcinoma (SCC), 9 (27%) were adenocarcinoma (AC), and 1 (3%) was large cell carcinoma (LCC). MMP-2 and TIMP-2 were expressed in high rates in NSCLC but CD44v6 expression was about 50%. Lymphatic invasion was less frequent in TIMP-2-positive patients and this difference was statistically significant (P = 0.005). There was a statistically significant difference between SCCs and ACs with respect to CD44v6 tumoral expression (P = 0.004). Also, there was a negative correlation between lymphatic invasion and the extent of CD44v6; lymphatic invasion was significantly less in CD44v6-positive cases (P = 0.013).
Conclusion: We found that TIMP-2 and CD44v6 can decrease the lymphatic invasion in NSCLCs. Also there was observed histiotype-related pattern of CD44v6 variant expression in SCCs.
Key words: Immunohistochemistry, Metastasis, Pulmonary neoplasms
Collapse
Affiliation(s)
- Bulent Eren
- Uludag University Medical Faculty, Forensic Medicine Department, Bursa, Turkey
| | - Mehmet Sar
- Istanbul University Cerrahpasa Medical Faculty, Pathology Department, Istanbul, Turkey
| | - Buge Oz
- Istanbul University Cerrahpasa Medical Faculty, Pathology Department, Istanbul, Turkey
| | - Fazilet H Oner Dincbas
- Istanbul University Cerrahpasa Medical Faculty, Radiation Oncology Department, Istanbul, Turkey
| |
Collapse
|
38
|
To K, Zhao Y, Jiang H, Hu K, Wang M, Wu J, Lee C, Yokom DW, Stratford AL, Klinge U, Mertens PR, Chen CS, Bally M, Yapp D, Dunn SE. The phosphoinositide-dependent kinase-1 inhibitor 2-amino-N-[4-[5-(2-phenanthrenyl)-3-(trifluoromethyl)-1H-pyrazol-1-yl]phenyl]-acetamide (OSU-03012) prevents Y-box binding protein-1 from inducing epidermal growth factor receptor. Mol Pharmacol 2007; 72:641-52. [PMID: 17595327 DOI: 10.1124/mol.107.036111] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The epidermal growth factor receptor (EGFR) is integral to basal-like and human epidermal growth factor receptor-2 (Her-2)-overexpressing breast cancers. Such tumors are associated with poor prognosis, the majority of which express high levels of EGFR. We reported that EGFR expression is induced by the oncogenic transcription factor Y-box binding protein-1 (YB-1) that occurs in a manner dependent on phosphorylation by Akt. Herein, we questioned whether blocking Akt with 2-amino-N-[4-[5-(2-phenanthrenyl)-3-(trifluoromethyl)-1H-pyrazol-1-yl]phenyl]-acetamide (OSU-03012), a phosphoinositide-dependent protein kinase-1 (PDK-1) small-molecule inhibitor, could prevent YB-1 from binding to the EGFR promoter. MDA-MB-468 and SUM 149 are basal-like breast cancer (BLBC) cells that were used for our studies because they express high levels of activated PDK-1, YB-1, and EGFR compared with the immortalized breast epithelial cell line 184htrt. In these cell lines, YB-1 preferentially bound to the -1 kilobase of the EGFR promoter, whereas this did not occur in the 184htrt cells based on chromatin immunoprecipitation. When the cells were exposed to OSU-03012 for 6 h, YB-1/EGFR promoter binding was significantly attenuated. To further confirm this observation, gel-shift assays showed that the drug inhibits YB-1/EGFR promoter binding. The inhibitory effect of OSU-03012 on EGFR was also observed at the mRNA and protein levels. OSU-03012 ultimately inhibited the growth of BLBC in monolayer and soft agar coordinate with the induction of apoptosis using an Array-Scan VTI high-content screening system. Furthermore, OSU-03012 inhibited the expression of EGFR by 48% in tumor xenografts derived from MDA-MB-435/Her-2 cells. This correlated with loss of YB-1 binding to the EGFR promoter. Hence, we find that OSU-03012 inhibits YB-1 resulting in a loss of EGFR expression in vitro and in vivo.
Collapse
Affiliation(s)
- K To
- Laboratory for Oncogenomic Research, Department of Pediatrics, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Papay J, Krenacs T, Moldvay J, Stelkovics E, Furak J, Molnar B, Kopper L. Immunophenotypic profiling of nonsmall cell lung cancer progression using the tissue microarray approach. Appl Immunohistochem Mol Morphol 2007; 15:19-30. [PMID: 17536303 DOI: 10.1097/01.pai.0000213143.32030.f5] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
This study, using tissue microarrays, aimed at the immunomorphologic profiling of nonsmall cell lung cancer (NSCLC) cases to reveal clinically relevant disease groups and biomarkers associated with patients' survival and tumor progression including brain metastatic potential. Donor tissue blocks were form 59 patients, including 33 primary tumors without distant metastasis and 26 brain metastatic primary tumors as well as the brain metastases. Sections were immunostained for 29 markers targeting molecules of cell adhesion, cell growth, cell cycle, and apoptosis regulation. beta-Catenin expression was the only independent prognostic marker associated with better outcome. Elevated expression of collagen XVII, CD44v6, and caspase-9, and the reduced production of beta-catenin and cellular apoptosis susceptibility protein were significantly associated with the metastatic potential of primary NSCLC. Expression of positive cell cycle regulators cyclin D1 and cyclin D3 was also increased in metastatic primary tumors. Metastatic tumor progression into the brain was accompanied by prominent p16, syndecan-1, p53 (DO7), and caspase-3 protein levels. Hierarchical clustering of complex immunoprofiles based on the differentially expressed markers grouped NSCLCs of the poorest outcome with high correlation including 2/3 of brain metastases of mixed histology. The brain metastatic potential of NSCLCs may be linked to the elevated levels of cyclinD1, cyclinD3, p16, p53, caspase-3, caspase-9, CD44v6, and collagen XVII and the down-regulation of beta-catenin and cellular apoptosis susceptibility protein. Unsupervised immunoprofiles based on differentially expressed biomarkers may help selecting lung cancers with aggressive behavior.
Collapse
Affiliation(s)
- Judit Papay
- Department of Pathology, Semmelweis University, Budapest, Hungary
| | | | | | | | | | | | | |
Collapse
|
40
|
Yoo J, Jung JH, Lee MA, Seo KJ, Shim BY, Kim SH, Cho DG, Ahn MI, Kim CH, Cho KD, Kang SJ, Kim HK. Immunohistochemical analysis of non-small cell lung cancer: correlation with clinical parameters and prognosis. J Korean Med Sci 2007; 22:318-25. [PMID: 17449943 PMCID: PMC2693601 DOI: 10.3346/jkms.2007.22.2.318] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Non-small cell lung cancers (NSCLC) vary in their biologic behavior. Recurrence and tumor-related mortality may be attributable to molecular abnormalities in primary tumors. This study evaluated such immunophenotypes with regard to cell cycle regulation and proliferation, apoptosis, and angiogenesis, to determine their significance for patient outcome. Core biopsies from 219 patients with NSCLC were assembled on tissue microarrays, and the expressions of p16, p21, p27, cyclin B1, cyclin E, Ki-67, caspase-3, survivin, bcl-2, VEGF, and endostatin were evaluated by immunohistochemistry. Despite previously described prognostic relevance of some of the investigated molecules, many of those markers were not directly associated with recurrence or survival. However, there was a trend for p16 immunoreactivity to be associated with a good prognosis (57% vs. 42% in 5-yr survival) (p=0.071). bcl-2 expression was strongly correlated with a better outcome (65% vs. 45% in 5-yr survival) (p=0.029), and the hazard of death for bcl-2 positive patients was 0.42 times of that for bcl-2 negative patients (p=0.047). A multivariate analysis with Cox proportional hazards model confirmed that the lymph node status (p=0.043) and stage (p=0.003) were other independent prognostic factors. Our results suggest that p16 and bcl-2 provide prognostic information independent of the TNM stage in NSCLC.
Collapse
Affiliation(s)
- Jinyoung Yoo
- Department of Pathology, St. Vincent's Hospital, The Catholic University of Korea, Suwon, Korea
| | - Ji Han Jung
- Department of Pathology, St. Vincent's Hospital, The Catholic University of Korea, Suwon, Korea
| | - Myung A Lee
- Department of Internal Medicine, St. Vincent's Hospital, The Catholic University of Korea, Suwon, Korea
| | - Kyung Jin Seo
- Department of Pathology, St. Vincent's Hospital, The Catholic University of Korea, Suwon, Korea
| | - Byoung Yong Shim
- Department of Internal Medicine, St. Vincent's Hospital, The Catholic University of Korea, Suwon, Korea
| | - Sung Hwan Kim
- Department of Radiation Oncology, St. Vincent's Hospital, The Catholic University of Korea, Suwon, Korea
| | - Deog Gon Cho
- Department of Thoracic Surgery, St. Vincent's Hospital, The Catholic University of Korea, Suwon, Korea
| | - Myeong Im Ahn
- Department of Diagnostic Radiology, St. Vincent's Hospital, The Catholic University of Korea, Suwon, Korea
| | - Chi Hong Kim
- Department of Internal Medicine, St. Vincent's Hospital, The Catholic University of Korea, Suwon, Korea
| | - Kyu Do Cho
- Department of Thoracic Surgery, St. Vincent's Hospital, The Catholic University of Korea, Suwon, Korea
| | - Seok Jin Kang
- Department of Pathology, St. Vincent's Hospital, The Catholic University of Korea, Suwon, Korea
| | - Hoon Kyo Kim
- Department of Internal Medicine, St. Vincent's Hospital, The Catholic University of Korea, Suwon, Korea
| |
Collapse
|
41
|
Lebbink RJ, Meyaard L. Non-MHC ligands for inhibitory immune receptors: novel insights and implications for immune regulation. Mol Immunol 2006; 44:2153-64. [PMID: 17188357 DOI: 10.1016/j.molimm.2006.11.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2006] [Accepted: 11/12/2006] [Indexed: 02/08/2023]
Abstract
Regulation of cellular responses by inhibitory receptors is crucial for proper function of the immune system. The prototype inhibitory immune receptors are major histocompatibility complex (MHC) class I binding killer-Ig like receptors (KIRs) present on effector cells such as natural killer (NK) cells and effector T cells. However, the recent identification of non-MHC class I ligands for inhibitory immune receptors, such as KLRG1, KLRB1 and LAIR-1, indicates that also MHC class I-independent inhibitory immune receptors play crucial roles in inducing peripheral tolerance. The presence of these receptors on many other immune cell types besides effector cells suggests that tight regulation of cell activation is necessary in all facets of the immune response in both normal and diseased tissue. Here, we review novel insights and implications of non-MHC class I ligand binding to inhibitory immune receptors. We give an overview of the known ligand-receptor pairs by grouping the ligands according to their properties and discuss implications of these interactions for the maintenance of immune balance and for the defense against tumors and pathogens.
Collapse
Affiliation(s)
- Robert Jan Lebbink
- Department of Immunology, University Medical Center Utrecht, Rm KC02.085.2, Lundlaan 6, 3584 EA Utrecht, The Netherlands
| | | |
Collapse
|
42
|
Ohta Y, Waseda R, Minato H, Endo N, Shimizu Y, Matsumoto I, Watanabe G. Surgical Results in T2N0M0 Nonsmall Cell Lung Cancer Patients With Large Tumors 5 cm or Greater in Diameter: What Regulates Outcome? Ann Thorac Surg 2006; 82:1180-4. [PMID: 16996904 DOI: 10.1016/j.athoracsur.2006.04.034] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2006] [Revised: 04/05/2006] [Accepted: 04/07/2006] [Indexed: 11/25/2022]
Abstract
BACKGROUND We assessed the surgical results along with the clinical and biological features of nonsmall-cell lung cancer (NSCLC) patients with localized large tumors. METHODS The study population consisted of 86 NSCLC patients who underwent complete resection of tumors 5 cm or larger in diameter in stage IB (T2N0M0). We immunohistochemically assessed the expression of angiostatin and endostatin. RESULTS The median tumor size was 6.0 cm (range, 5 to 14 cm). The operative procedures used were lobectomy in 71 cases, bilobectomy in 8 cases, and pneumonectomy in 11 cases. Fifty patients (58.1%) relapsed during the mean follow-up period of 33.6 +/- 4.5 months. The median disease-free interval was 9 months. Of 44 recurrent patients whose disease-free interval could be identified, 25 patients (56.8%) relapsed within 12 months after the operation. The overall 5- and 10-year survival rates were 42.0% and 24.2%, respectively. Multivariate analysis showed that the degree of pleural involvement and angiostatin expression within the tumor were independent prognostic indicators. The endostatin expression within tumors also had a weaker relationship with outcome. CONCLUSIONS Long-term surgical results were poor and early relapse was common in this cohort. In addition to pleural involvement, the tumor-induced expression of angiostatin and endostatin merit further investigation to gain possible insights into selection of patients who will benefit from surgery as the first line treatment.
Collapse
Affiliation(s)
- Yasuhiko Ohta
- Department of General and Cardiothoracic Surgery and Pathology, Kanazawa University School of Medicine, Kanazawa, Japan.
| | | | | | | | | | | | | |
Collapse
|
43
|
Alba E, Llombart A, Ribelles N, Ramos M, Fernández R, Mayordomo JI, Tusquets I, Gil M, Barnadas A, Carabante F, Ruiz M, Vera R, Palomero I, Soriano V, González J, Colomer R. Serum endostatin and bFGF as predictive factors in advanced breast cancer patients treated with letrozole. Clin Transl Oncol 2006; 8:193-9. [PMID: 16648119 DOI: 10.1007/s12094-006-0010-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
INTRODUCTION To investigate the value of baseline serum levels of VEGF, bFGF, endostatin and their ratio as predictive factors of response to endocrine therapy in patients with metastatic breast cancer (MBC) and positive ER treated with letrozole after tamoxifen failure. MATERIALS AND METHOD The serum levels of endostatin, VEGF and bFGF were determined in postmenopausal patients with progressing MBC from serum samples obtained before initiation of letrozole. The relation between serum angiogenic factor levels and TTP was investigated. RESULTS Seventy-six patients (45.2%) presented a high endostatin level (> 24.6 ng/ml), 40% low bFGF levels (0 pg/ml) and 50.4% low VEGF (=/< 187 ng/ml). With a median follow-up of 22 months, the median TTP was 12.3 months. Median TTP was worse in patients with high endostatin concentration as well as in the low bFGF group, but was not affected when VEGF was considered. When the two factors were combined, the median TTP of patients with endostatin > 24.6 ng/ml and bFGF equal 0 pg/ml was 9.5 months versus 19.5 months in patients with endostatin =/< 24.6 ng/ml and bFGF > 0 pg/ml. CONCLUSIONS The baseline levels of bFGF and endostatin are predictive factors of efficacy in patients with MBC treated with letrozole and can select groups with different TTP.
Collapse
Affiliation(s)
- Emilio Alba
- Hospital Clínico Universitario, Málaga, Spain.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Lebbink RJ, de Ruiter T, Adelmeijer J, Brenkman AB, van Helvoort JM, Koch M, Farndale RW, Lisman T, Sonnenberg A, Lenting PJ, Meyaard L. Collagens are functional, high affinity ligands for the inhibitory immune receptor LAIR-1. ACTA ACUST UNITED AC 2006; 203:1419-25. [PMID: 16754721 PMCID: PMC2118306 DOI: 10.1084/jem.20052554] [Citation(s) in RCA: 247] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Collagens are the most abundant proteins in the human body, important in maintenance of tissue structure and hemostasis. Here we report that collagens are high affinity ligands for the broadly expressed inhibitory leukocyte-associated immunoglobulin-like receptor-1 (LAIR-1). The interaction is dependent on the conserved Gly-Pro-Hyp collagen repeats. Antibody cross-linking of LAIR-1 is known to inhibit immune cell function in vitro. We now show that collagens are functional ligands for LAIR-1 and directly inhibit immune cell activation in vitro. Thus far, all documented ligands for immune inhibitory receptors are membrane molecules, implying a regulatory role in cell–cell interaction. Our data reveal a novel mechanism of peripheral immune regulation by inhibitory immune receptors binding to extracellular matrix collagens.
Collapse
Affiliation(s)
- Robert Jan Lebbink
- Department of Immunology, University Medical Center, 3584 EA Utrecht, Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Woo IS, Kim KA, Jeon HM, Hong SH, Rho SY, Koh SJ, Lee MA, Byun JH, Kang JH, Hong YS, Lee KS, Cho CS, Choi MG, Chung IS. Pretreatment serum endostatin as a prognostic indicator in metastatic gastric carcinoma. Int J Cancer 2006; 119:2901-6. [PMID: 16998835 DOI: 10.1002/ijc.22216] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Endostatin is the C-terminal antiangiogenic fragment of the extracellular matrix protein collagen XVIII, and is generated by tumor-derived proteases. The presence of serum endostatin in patients with gastric cancer has not been reported. The authors assessed the serum levels of endostatin in patients with gastric carcinoma and evaluated their association with the levels of vascular endothelial growth factor (VEGF) and the clinical outcome. A total of 107 patients with gastric cancer were included in the study. Pretherapeutic serum levels of endostatin and VEGF were measured using an ELISA, and compared with those in 23 healthy controls. The serum levels of endostatin and VEGF were higher in gastric cancer patients than in healthy controls (endostatin, 70.1 +/- 16.6 vs. 52.2 +/- 6.2 ng/mL [p < 0.001]; VEGF, 55.1 +/- 7.6 vs. 32.1 +/- 2.4 ng/mL [p < 0.001]; mean +/- SD). Serum endostatin levels were significantly associated with the presence of distant metastases (r = 0.556, p < 0.001) and VEGF levels (r = 0.335, p < 0.001), but not with the depth of tumor invasion, differentiation, or regional lymph node status. A serum endostatin level above the 75th percentile of the distribution for the patients (79.2 ng/mL) was associated with a poor outcome (last follow-up at 42 months; median survival time, 9 vs. 20 months [log-rank, p = 0.017]; median time to progression, 5 vs. 10 months [log-rank, p = 0.022]) in the patients with metastatic gastric cancer. The results suggest for the first time that an elevated serum level of endostatin at the diagnosis of metastatic gastric cancer could be predictive of a poor outcome.
Collapse
Affiliation(s)
- In Sook Woo
- Division of Medical Oncology, Department of Internal Medicine, College of Medicine, Catholic University, Seoul, Korea.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Clamp AR, Jayson GC. The clinical potential of antiangiogenic fragments of extracellular matrix proteins. Br J Cancer 2005; 93:967-72. [PMID: 16234821 PMCID: PMC2361682 DOI: 10.1038/sj.bjc.6602820] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2005] [Revised: 09/19/2005] [Accepted: 09/22/2005] [Indexed: 01/09/2023] Open
Abstract
Neovasculature development is a crucial step in the natural history of a cancer. While much emphasis has been placed on proangiogenic growth factors such as VEGF, it is clear that endogenous angiogenesis inhibitors also have critical roles in the regulation of this process. Recent research has identified several cryptic fragments of extracellular matrix/vascular basement membrane proteins that have potent antiangiogenic properties in vivo. It has become apparent that many of these fragments signal via interactions with endothelial integrins, although multiple downstream effector pathways have been implicated and endostatin, the first non-collagenous domain of collagen XVIII, influences an intricate signalling network. The activity of these molecules in animal models suggests that they may have significant clinical activity; however, results of phase I/II trials with endostatin were disappointing. Many possible reasons can be found for the failure of these studies. Weaknesses in trial design, endostatin administration regimen and patient selection are identifiable, and importantly the lack of a clearly defined antiangiogenic mechanism for endostatin hindered assessment of biologically effective dose. Additionally, in vivo immunological and proteolytic function-neutralising mechanisms may have negated endostatin's actions. Lessons learned from these studies will aid the future clinical development of other antiangiogenic extracellular matrix protein fragments.
Collapse
Affiliation(s)
- A R Clamp
- Cancer Research UK Department of Medical Oncology, Christie Hospital, Wilmslow Road, Manchester M20 4BX, UK.
| | | |
Collapse
|
47
|
Heljasvaara R, Nyberg P, Luostarinen J, Parikka M, Heikkilä P, Rehn M, Sorsa T, Salo T, Pihlajaniemi T. Generation of biologically active endostatin fragments from human collagen XVIII by distinct matrix metalloproteases. Exp Cell Res 2005; 307:292-304. [PMID: 15950618 DOI: 10.1016/j.yexcr.2005.03.021] [Citation(s) in RCA: 143] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2005] [Revised: 03/04/2005] [Accepted: 03/14/2005] [Indexed: 11/29/2022]
Abstract
Endostatin, a potent inhibitor of endothelial cell proliferation, migration, angiogenesis and tumor growth, is proteolytically cleaved from the C-terminal noncollagenous NC1 domain of type XVIII collagen. We investigated the endostatin formation from human collagen XVIII by several MMPs in vitro. The generation of endostatin fragments differing in molecular size (24-30 kDa) and in N-terminal sequences was identified in the cases of MMP-3, -7, -9, -13 and -20. The cleavage sites were located in the protease-sensitive hinge region between the trimerization and endostatin domains of NC1. MMP-1, -2, -8 and -12 did not show any significant activity against the C-terminus of collagen XVIII. The anti-proliferative effect of the 20-kDa endostatin, three longer endostatin-containing fragments generated in vitro by distinct MMPs and the entire NC1 domain, on bFGF-stimulated human umbilical vein endothelial cells was established. The anti-migratory potential of some of these fragments was also studied. In addition, production of endostatin fragments between 24-30 kDa by human hepatoblastoma cells was shown to be due to MMP action on type XVIII collagen. Our results indicate that certain, especially cancer-related, MMP family members can generate biologically active endostatin-containing polypeptides from collagen XVIII and thus, by releasing endostatin fragments, may participate in the inhibition of endothelial cell proliferation, migration and angiogenesis.
Collapse
Affiliation(s)
- Ritva Heljasvaara
- Collagen Research Unit, Biocenter Oulu and Department of Medical Biochemistry and Molecular Biology, University of Oulu, PO Box 5000, FIN-90014 Oulu, Finland
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Singhal S, Vachani A, Antin-Ozerkis D, Kaiser LR, Albelda SM. Prognostic implications of cell cycle, apoptosis, and angiogenesis biomarkers in non-small cell lung cancer: a review. Clin Cancer Res 2005; 11:3974-86. [PMID: 15930332 DOI: 10.1158/1078-0432.ccr-04-2661] [Citation(s) in RCA: 242] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Lung cancer is the leading cause of cancer death in the U.S. with survival restricted to a subset of those patients able to undergo surgical resection. However, even with surgery, recurrence rates range from 30% to 60%, depending on the pathologic stage. With the advent of partially effective, but potentially toxic adjuvant chemotherapy, it has become increasingly important to discover biomarkers that will identify those patients who have the highest likelihood of recurrence and who thus might benefit most from adjuvant chemotherapy. Hundreds of papers have appeared over the past several decades proposing a variety of molecular markers or proteins that may have prognostic significance in non-small cell lung cancer. This review analyzes the largest and most rigorous of these studies with the aim of compiling the most important prognostic markers in early stage non-small cell lung cancer. In this review, we focused on biomarkers primarily involved in one of three major pathways: cell cycle regulation, apoptosis, and angiogenesis. Although no single marker has yet been shown to be perfect in predicting patient outcome, a profile based on the best of these markers may prove useful in directing patient therapy. The markers with the strongest evidence as independent predictors of patient outcome include cyclin E, cyclin B1, p21, p27, p16, survivin, collagen XVIII, and vascular endothelial cell growth factor.
Collapse
Affiliation(s)
- Sunil Singhal
- Section of Thoracic Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | |
Collapse
|
49
|
Gruszka A, Kunert-Radek J, Pawlikowski M, Stepien H. Serum endostatin levels are elevated and correlate with serum vascular endothelial growth factor levels in patients with pituitary adenomas. Pituitary 2005; 8:163-8. [PMID: 16379029 DOI: 10.1007/s11102-005-5258-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Endostatin, a cleaved fragment of collagen XVIII, is a potent endogenous angiogenesis inhibitor. Elevated serum endostatin levels have been recently reported in patients with various types of neoplasms. The purpose of our study was to evaluate serum concentrations of endostatin in patients harbouring various pituitary adenoma types and to examine the relationship of serum endostatin levels to circulating vascular endothelial growth factor (VEGF) levels. Preoperative serum endostatin and VEGF concentrations were measured using competitive enzyme immunoassays in 71 patients with pituitary adenomas (20 somatotropinomas, 3 corticotropinomas, 6 prolactinomas and 42 clinically nonfunctioning pituitary adenomas - CNFPAs) and compared with levels from age-matched controls. In 35 patients postoperative immunohistochemical investigations were performed. Serum endostatin concentrations were significantly higher in all pituitary adenoma types, except for prolactinomas (somatotropinomas: 124 +/- 16; p < 0.02, corticotropinomas: 157 +/- 42; p < 0.02, prolactinomas: 141 +/- 37; p > 0.05, CNFPAs: 169 +/- 11 ng/ml; p < 0.000005 vs 73 +/- 10 ng/ml in controls). There was a significant positive correlation between endostatin and VEGF serum levels in patients with pituitary adenomas (r = +0.322; p = 0.006). In the control group a significant negative correlation xbetween circulating endostatin and VEGF was found (r = -0.653; p = 0.00975). The simultaneous elevation of endostatin and VEGF may attenuate the pro-angiogenic action of VEGF and be responsible for rather weak neovascularization of pituitary adenomas. Prospective studies are required to assess the usefulness of circulating endostatin and VEGF as markers of progression or recurrence of pituitary tumors.
Collapse
Affiliation(s)
- Anna Gruszka
- Dept. of Neuroendocrinology, Chair of Endocrinology, Medical University of Lodz, Sterling str. 1/3, 91-425, Lodz, Poland.
| | | | | | | |
Collapse
|