1
|
Papadaki MA, Papadaki E, Chatziavraam S, Aggouraki D, Michaelidou K, Fotsitzoudis C, Vassilakopoulou M, Mavroudis D, Agelaki S. Prognostic Value of Fas/Fas Ligand Expression on Circulating Tumor Cells (CTCs) and Immune Cells in the Peripheral Blood of Patients with Metastatic Breast Cancer. Cancers (Basel) 2024; 16:2927. [PMID: 39272785 PMCID: PMC11393959 DOI: 10.3390/cancers16172927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 08/16/2024] [Accepted: 08/20/2024] [Indexed: 09/15/2024] Open
Abstract
The Fas/Fas ligand (FasL) system is a major apoptosis-regulating pathway with a key role in tumor immune surveillance and metastasis. The expression of Fas/FasL on mammary tumor tissues holds prognostic value for breast cancer (BC) patients. We herein assessed Fas/FasL expression on circulating tumor cells (CTCs) and matched peripheral blood mononuclear cells (PBMCs) from 98 patients with metastatic BC receiving first-line treatment. Fas+, FasL+, and Fas+/FasL+ CTCs were identified in 88.5%, 92.3%, and 84.6% of CTC-positive patients, respectively. In addition, Fas+/FasL+, Fas-/FasL+, and Fas-/FasL- PBMCs were identified in 70.3%, 24.2%, and 5.5% of patients, respectively. A reduced progression-free survival (PFS) was revealed among CTC-positive patients (median PFS: 9.5 versus 13.4 months; p = 0.004), and specifically among those harboring Fas+/FasL+ CTCs (median PFS: 9.5 vs. 13.4 months; p = 0.009). On the other hand, an increased overall survival (OS) was demonstrated among patients with Fas+/FasL+ PBMCs rather than those with Fas-/FasL+ and Fas-/FasL- PBMCs (median OS: 35.7 vs. 25.9 vs. 14.4 months, respectively; p = 0.008). These data provide for the first time evidence on Fas/FasL expression on CTCs and PBMCs with significant prognostic value for patients with metastatic BC, thus highlighting the role of the Fas/FasL system in the peripheral immune response and metastatic progression of BC.
Collapse
Affiliation(s)
- Maria A Papadaki
- Laboratory of Translational Oncology, Medical School, University of Crete, Heraklion, 70013 Crete, Greece
| | - Eleni Papadaki
- Laboratory of Translational Oncology, Medical School, University of Crete, Heraklion, 70013 Crete, Greece
| | - Sofia Chatziavraam
- Laboratory of Translational Oncology, Medical School, University of Crete, Heraklion, 70013 Crete, Greece
| | - Despoina Aggouraki
- Laboratory of Translational Oncology, Medical School, University of Crete, Heraklion, 70013 Crete, Greece
| | - Kleita Michaelidou
- Laboratory of Translational Oncology, Medical School, University of Crete, Heraklion, 70013 Crete, Greece
| | - Charalampos Fotsitzoudis
- Laboratory of Translational Oncology, Medical School, University of Crete, Heraklion, 70013 Crete, Greece
- Department of Medical Oncology, University General Hospital of Heraklion, 71500 Heraklion, Greece
| | - Maria Vassilakopoulou
- Laboratory of Translational Oncology, Medical School, University of Crete, Heraklion, 70013 Crete, Greece
| | - Dimitrios Mavroudis
- Laboratory of Translational Oncology, Medical School, University of Crete, Heraklion, 70013 Crete, Greece
- Department of Medical Oncology, University General Hospital of Heraklion, 71500 Heraklion, Greece
| | - Sofia Agelaki
- Laboratory of Translational Oncology, Medical School, University of Crete, Heraklion, 70013 Crete, Greece
- Department of Medical Oncology, University General Hospital of Heraklion, 71500 Heraklion, Greece
| |
Collapse
|
2
|
Huang M, Wang Y, Fang L, Liu C, Feng F, Liu L, Sun C. T cell senescence: a new perspective on immunotherapy in lung cancer. Front Immunol 2024; 15:1338680. [PMID: 38415245 PMCID: PMC10896971 DOI: 10.3389/fimmu.2024.1338680] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 01/30/2024] [Indexed: 02/29/2024] Open
Abstract
T cell senescence is an indication of T cell dysfunction. The ability of senescent T cells to respond to cognate antigens is reduced and they are in the late stage of differentiation and proliferation; therefore, they cannot recognize and eliminate tumor cells in a timely and effective manner, leading to the formation of the suppressive tumor microenvironment. Establishing methods to reverse T cell senescence is particularly important for immunotherapy. Aging exacerbates profound changes in the immune system, leading to increased susceptibility to chronic, infectious, and autoimmune diseases. Patients with malignant lung tumors have impaired immune function with a high risk of recurrence, metastasis, and mortality. Immunotherapy based on PD-1, PD-L1, CTLA-4, and other immune checkpoints is promising for treating lung malignancies. However, T cell senescence can lead to low efficacy or unsuccessful treatment results in some immunotherapies. Efficiently blocking and reversing T cell senescence is a key goal of the enhancement of tumor immunotherapy. This study discusses the characteristics, mechanism, and expression of T cell senescence in malignant lung tumors and the treatment strategies.
Collapse
Affiliation(s)
- Mengge Huang
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yuetong Wang
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Liguang Fang
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Cun Liu
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang, China
| | - Fubin Feng
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China
| | - Lijuan Liu
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China
| | - Changgang Sun
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang, China
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China
| |
Collapse
|
3
|
James A, Akash K, Sharma A, Bhattacharyya S, Sriamornsak P, Nagraik R, Kumar D. Himalayan flora: targeting various molecular pathways in lung cancer. Med Oncol 2023; 40:314. [PMID: 37787816 DOI: 10.1007/s12032-023-02171-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 08/21/2023] [Indexed: 10/04/2023]
Abstract
The fatal amplification of lung cancer across the globe and the limitations of current treatment strategies emphasize the necessity for substitute therapeutics. The incorporation of phyto-derived components in chemo treatment holds promise in addressing those challenges. Despite the significant progressions in lung cancer therapeutics, the complexities of molecular mechanism and pathways underlying this disease remain inadequately understood, necessitating novel biomarker targeting. The Himalayas, abundant in diverse plant varieties with established chemotherapeutic potential, presents a promising avenue for investigating potential cures for lung carcinoma. The vast diversity of phytocompounds herein can be explored for targeting the disease. This review delves into the multifaceted targets of lung cancer and explores the established phytochemicals with their specific molecular targets. It emphasizes comprehending the intricate pathways that govern effective therapeutic interventions for lung cancer. Through this exploration of Himalayan flora, this review seeks to illuminate potential breakthroughs in lung cancer management using natural compounds. The amalgamation of Himalayan plant-derived compounds with cautiously designed combined therapeutic approaches such as nanocarrier-mediated drug delivery and synergistic therapy offers an opportunity to redefine the boundaries of lung cancer treatment by reducing the drug resistance and side effects and enabling an effective targeted delivery of drugs. Furthermore, additional studies are obligatory to understand the possible derivation of natural compounds used in current lung cancer treatment from plant species within the Himalayan region.
Collapse
Affiliation(s)
- Abija James
- Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, Himachal Pradesh, 173229, India
| | - K Akash
- Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, Himachal Pradesh, 173229, India
| | - Avinash Sharma
- Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, Himachal Pradesh, 173229, India
| | - Sanjib Bhattacharyya
- Department of Pharmaceutical Sciences and Chinese Traditional Medicine, Southwest University, Beibei, 400715, Chongqing, People's Republic of China
- Department of Sciences, Nirma University, Ahmedabad, Gujarat, 382481, India
| | | | - Rupak Nagraik
- Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, Himachal Pradesh, 173229, India.
| | - Deepak Kumar
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh, 173229, India.
| |
Collapse
|
4
|
Khodayari Moez E, Warkentin MT, Brhane Y, Lam S, Field JK, Liu G, Zulueta JJ, Valencia K, Mesa-Guzman M, Nialet AP, Atkar-Khattra S, Davies MPA, Grant B, Murison K, Montuenga LM, Amos CI, Robbins HA, Johansson M, Hung RJ. Circulating proteome for pulmonary nodule malignancy. J Natl Cancer Inst 2023; 115:1060-1070. [PMID: 37369027 PMCID: PMC10483334 DOI: 10.1093/jnci/djad122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 05/29/2023] [Accepted: 06/22/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND Although lung cancer screening with low-dose computed tomography is rolling out in many areas of the world, differentiating indeterminate pulmonary nodules remains a major challenge. We conducted one of the first systematic investigations of circulating protein markers to differentiate malignant from benign screen-detected pulmonary nodules. METHODS Based on 4 international low-dose computed tomography screening studies, we assayed 1078 protein markers using prediagnostic blood samples from 1253 participants based on a nested case-control design. Protein markers were measured using proximity extension assays, and data were analyzed using multivariable logistic regression, random forest, and penalized regressions. Protein burden scores (PBSs) for overall nodule malignancy and imminent tumors were estimated. RESULTS We identified 36 potentially informative circulating protein markers differentiating malignant from benign nodules, representing a tightly connected biological network. Ten markers were found to be particularly relevant for imminent lung cancer diagnoses within 1 year. Increases in PBSs for overall nodule malignancy and imminent tumors by 1 standard deviation were associated with odds ratios of 2.29 (95% confidence interval: 1.95 to 2.72) and 2.81 (95% confidence interval: 2.27 to 3.54) for nodule malignancy overall and within 1 year of diagnosis, respectively. Both PBSs for overall nodule malignancy and for imminent tumors were substantially higher for those with malignant nodules than for those with benign nodules, even when limited to Lung Computed Tomography Screening Reporting and Data System (LungRADS) category 4 (P < .001). CONCLUSIONS Circulating protein markers can help differentiate malignant from benign pulmonary nodules. Validation with an independent computed tomographic screening study will be required before clinical implementation.
Collapse
Affiliation(s)
- Elham Khodayari Moez
- Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada
| | - Matthew T Warkentin
- Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Yonathan Brhane
- Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada
| | - Stephen Lam
- Integrative Oncology, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - John K Field
- Molecular & Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - Geoffrey Liu
- Computational Biology and Medicine Program, Princess Margaret Cancer Center, Toronto, ON, Canada
| | - Javier J Zulueta
- Division of Pulmonary, Critical Care and Sleep Medicine, Mount Sinai Morningside Hospital, Icahn School of Medicine, New York, NY, USA
| | - Karmele Valencia
- Center of Applied Medical Research (CIMA) and Schools of Sciences and Medicine, University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer (CIBERONC), Madrid, Spain
| | - Miguel Mesa-Guzman
- Thoracic Surgery Department, Clínica Universidad de Navarra, Pamplona, Spain
| | - Andrea Pasquier Nialet
- Center of Applied Medical Research (CIMA) and Schools of Sciences and Medicine, University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer (CIBERONC), Madrid, Spain
| | | | - Michael P A Davies
- Molecular & Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - Benjamin Grant
- Computational Biology and Medicine Program, Princess Margaret Cancer Center, Toronto, ON, Canada
| | - Kiera Murison
- Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada
| | - Luis M Montuenga
- Center of Applied Medical Research (CIMA) and Schools of Sciences and Medicine, University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer (CIBERONC), Madrid, Spain
| | - Christopher I Amos
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
| | - Hilary A Robbins
- Genomic Epidemiology Branch, International Agency for Research on Cancer, Lyon, France
| | - Mattias Johansson
- Genomic Epidemiology Branch, International Agency for Research on Cancer, Lyon, France
| | - Rayjean J Hung
- Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
5
|
Smok-Kalwat J, Mertowska P, Mertowski S, Smolak K, Kozińska A, Koszałka F, Kwaśniewski W, Grywalska E, Góźdź S. The Importance of the Immune System and Molecular Cell Signaling Pathways in the Pathogenesis and Progression of Lung Cancer. Int J Mol Sci 2023; 24:1506. [PMID: 36675020 PMCID: PMC9861992 DOI: 10.3390/ijms24021506] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/04/2023] [Accepted: 01/08/2023] [Indexed: 01/13/2023] Open
Abstract
Lung cancer is a disease that in recent years has become one of the greatest threats to modern society. Every year there are more and more new cases and the percentage of deaths caused by this type of cancer increases. Despite many studies, scientists are still looking for answers regarding the mechanisms of lung cancer development and progression, with particular emphasis on the role of the immune system. The aim of this literature review was to present the importance of disorders of the immune system and the accompanying changes at the level of cell signaling in the pathogenesis of lung cancer. The collected results showed that in the process of immunopathogenesis of almost all subtypes of lung cancer, changes in the tumor microenvironment, deregulation of immune checkpoints and abnormalities in cell signaling pathways are involved, which contribute to the multistage and multifaceted carcinogenesis of this type of cancer. We, therefore, suggest that in future studies, researchers should focus on a detailed analysis of tumor microenvironmental immune checkpoints, and to validate their validity, perform genetic polymorphism analyses in a wide range of patients and healthy individuals to determine the genetic susceptibility to lung cancer development. In addition, further research related to the analysis of the tumor microenvironment; immune system disorders, with a particular emphasis on immunological checkpoints and genetic differences may contribute to the development of new personalized therapies that improve the prognosis of patients.
Collapse
Affiliation(s)
- Jolanta Smok-Kalwat
- Department of Clinical Oncology, Holy Cross Cancer Centre, 3 Artwinskiego Street, 25-734 Kielce, Poland
| | - Paulina Mertowska
- Department of Experimental Immunology, Medical University of Lublin, 4a Chodzki Street, 20-093 Lublin, Poland
| | - Sebastian Mertowski
- Department of Experimental Immunology, Medical University of Lublin, 4a Chodzki Street, 20-093 Lublin, Poland
| | - Konrad Smolak
- Department of Experimental Immunology, Medical University of Lublin, 4a Chodzki Street, 20-093 Lublin, Poland
| | - Aleksandra Kozińska
- Student Research Group of Experimental Immunology, Medical University of Lublin, 4a Chodzki Street, 20-093 Lublin, Poland
| | - Filip Koszałka
- Student Research Group of Experimental Immunology, Medical University of Lublin, 4a Chodzki Street, 20-093 Lublin, Poland
| | - Wojciech Kwaśniewski
- Department of Gynecologic Oncology and Gynecology, Medical University of Lublin, 20-081 Lublin, Poland
| | - Ewelina Grywalska
- Department of Experimental Immunology, Medical University of Lublin, 4a Chodzki Street, 20-093 Lublin, Poland
| | - Stanisław Góźdź
- Department of Clinical Oncology, Holy Cross Cancer Centre, 3 Artwinskiego Street, 25-734 Kielce, Poland
- Institute of Medical Science, Collegium Medicum, Jan Kochanowski University of Kielce, IX Wieków Kielc 19A, 25-317 Kielce, Poland
| |
Collapse
|
6
|
Meijer JJ, Leonetti A, Airò G, Tiseo M, Rolfo C, Giovannetti E, Vahabi M. Small cell lung cancer: Novel treatments beyond immunotherapy. Semin Cancer Biol 2022; 86:376-385. [PMID: 35568295 DOI: 10.1016/j.semcancer.2022.05.004] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 05/03/2022] [Accepted: 05/07/2022] [Indexed: 11/23/2022]
Abstract
Small cell lung cancer (SCLC) arises in peribronchial locations and infiltrates the bronchial submucosa, including about 15% of lung cancer cases. Despite decades of research, the prognosis for SCLC patients remains poor because this tumor is characterized by an exceptionally high proliferative rate, strong tendency for early widespread metastasis and acquired chemoresistance. Omics profiling revealed that SCLC harbor extensive chromosomal rearrangements and a very high mutation burden. This led to the development of immune-checkpoint inhibitors as single agents or in combination with chemotherapy, which however resulted in a prolonged benefit only for a small subset of patients. Thus, the present review discusses the rationale and limitations of immunotherapeutic approaches, presenting the current biological understanding of aberrant signaling pathways that might be exploited with new potential treatments. In particular, new agents targeting DNA damage repair, cell cycle checkpoint, and apoptosis pathways showed several promising results in different preclinical models. Epigenetic alterations, gene amplifications and mutations can act as biomarkers in this context. Future research and improved clinical outcome for SCLC patients will depend on the integration between these omics and pharmacological studies with clinical translational research, in order to identify specific predictive biomarkers that will be hopefully validated using clinical trials with biomarker-selected targeted treatments.
Collapse
Affiliation(s)
- Job-Joris Meijer
- Department of Medical Oncology, Amsterdam UMC, VU University, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Alessandro Leonetti
- Department of Medicine and Surgery, University of Parma, Parma, Italy; Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Giulia Airò
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Marcello Tiseo
- Department of Medicine and Surgery, University of Parma, Parma, Italy; Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Christian Rolfo
- Center for Thoracic Oncology, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, Mount Sinai Health System, New York, NY, USA
| | - Elisa Giovannetti
- Department of Medical Oncology, Amsterdam UMC, VU University, Cancer Center Amsterdam, Amsterdam, Netherlands; Fondazione Pisana per la Scienza, Pisa, Italy.
| | - Mahrou Vahabi
- Department of Medical Oncology, Amsterdam UMC, VU University, Cancer Center Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
7
|
Sulejmani O, Grunewald L, Andersch L, Schwiebert S, Klaus A, Winkler A, Astrahantseff K, Eggert A, Henssen AG, Schulte JH, Anders K, Künkele A. Inhibiting Lysine Demethylase 1A Improves L1CAM-Specific CAR T Cell Therapy by Unleashing Antigen-Independent Killing via the FAS-FASL Axis. Cancers (Basel) 2021; 13:cancers13215489. [PMID: 34771652 PMCID: PMC8583435 DOI: 10.3390/cancers13215489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/18/2021] [Accepted: 10/22/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Solid tumor cells can lose or heterogeneously express antigens to become resistant to chimeric antigen receptor (CAR) T cell therapy. Here, we explore whether epigenetic manipulation to unleash antigen-independent killing mechanisms can overcome this hurdle. KDM1A is overexpressed in many cancers and removes lysine methylation on histones that keeps the DNA firmly packed to selectively activate or repress gene activity, depending on the specific lysine target. KDM1A also regulates the expression of nonhistone proteins. We inhibited KDM1A in the childhood tumor, neuroblastoma, to increase FAS expression on tumor cells. The FAS receptor can be triggered to induce cell death when bound by the FAS ligand on CAR and other activated T cells present in the tumor environment, even if the tumor cells lack the target antigen. FAS upregulation via KDM1A inhibition sensitized neuroblastoma cells to FAS-FASL-mediated killing and augmented CAR T cell therapy against antigen-poor or even antigen-negative neuroblastoma. Abstract Chimeric antigen receptor (CAR) T cell therapy has emerged as a promising treatment strategy, however, therapeutic success against solid tumors such as neuroblastoma remains modest. Recurrence of antigen-poor tumor variants often ultimately results in treatment failure. Using antigen-independent killing mechanisms such as the FAS receptor (FAS)-FAS ligand (FASL) axis through epigenetic manipulation may be a way to counteract the escape achieved by antigen downregulation. Analysis of public RNA-sequencing data from primary neuroblastomas revealed that a particular epigenetic modifier, the histone lysine demethylase 1A (KDM1A), correlated negatively with FAS expression. KDM1A is known to interact with TP53 to repress TP53-mediated transcriptional activation of genes, including FAS. We showed that pharmacologically blocking KDM1A activity in neuroblastoma cells with the small molecule inhibitor, SP-2509, increased FAS cell-surface expression in a strictly TP53-dependent manner. FAS upregulation sensitized neuroblastoma cells to FAS-FASL-dependent killing and augmented L1CAM-directed CAR T cell therapy against antigen-poor or even antigen-negative tumor cells in vitro. The improved therapeutic response was abrogated when the FAS-FASL interaction was abolished with an antagonistic FAS antibody. Our results show that KDM1A inhibition unleashes an antigen-independent killing mechanism via the FAS-FASL axis to make tumor cell variants that partially or totally suppress antigen expression susceptible to CAR T cell therapy.
Collapse
Affiliation(s)
- Ornela Sulejmani
- Department of Pediatric Oncology and Hematology, Berlin Institute of Health, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universiät zu Berlin, 10353 Berlin, Germany; (O.S.); (L.G.); (L.A.); (S.S.); (A.K.); (A.W.); (K.A.); (A.E.); (A.G.H.); (J.H.S.)
| | - Laura Grunewald
- Department of Pediatric Oncology and Hematology, Berlin Institute of Health, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universiät zu Berlin, 10353 Berlin, Germany; (O.S.); (L.G.); (L.A.); (S.S.); (A.K.); (A.W.); (K.A.); (A.E.); (A.G.H.); (J.H.S.)
| | - Lena Andersch
- Department of Pediatric Oncology and Hematology, Berlin Institute of Health, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universiät zu Berlin, 10353 Berlin, Germany; (O.S.); (L.G.); (L.A.); (S.S.); (A.K.); (A.W.); (K.A.); (A.E.); (A.G.H.); (J.H.S.)
| | - Silke Schwiebert
- Department of Pediatric Oncology and Hematology, Berlin Institute of Health, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universiät zu Berlin, 10353 Berlin, Germany; (O.S.); (L.G.); (L.A.); (S.S.); (A.K.); (A.W.); (K.A.); (A.E.); (A.G.H.); (J.H.S.)
| | - Anika Klaus
- Department of Pediatric Oncology and Hematology, Berlin Institute of Health, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universiät zu Berlin, 10353 Berlin, Germany; (O.S.); (L.G.); (L.A.); (S.S.); (A.K.); (A.W.); (K.A.); (A.E.); (A.G.H.); (J.H.S.)
| | - Annika Winkler
- Department of Pediatric Oncology and Hematology, Berlin Institute of Health, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universiät zu Berlin, 10353 Berlin, Germany; (O.S.); (L.G.); (L.A.); (S.S.); (A.K.); (A.W.); (K.A.); (A.E.); (A.G.H.); (J.H.S.)
| | - Kathy Astrahantseff
- Department of Pediatric Oncology and Hematology, Berlin Institute of Health, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universiät zu Berlin, 10353 Berlin, Germany; (O.S.); (L.G.); (L.A.); (S.S.); (A.K.); (A.W.); (K.A.); (A.E.); (A.G.H.); (J.H.S.)
| | - Angelika Eggert
- Department of Pediatric Oncology and Hematology, Berlin Institute of Health, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universiät zu Berlin, 10353 Berlin, Germany; (O.S.); (L.G.); (L.A.); (S.S.); (A.K.); (A.W.); (K.A.); (A.E.); (A.G.H.); (J.H.S.)
- German Cancer Consortium (DKTK), 10117 Berlin, Germany;
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Anton G. Henssen
- Department of Pediatric Oncology and Hematology, Berlin Institute of Health, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universiät zu Berlin, 10353 Berlin, Germany; (O.S.); (L.G.); (L.A.); (S.S.); (A.K.); (A.W.); (K.A.); (A.E.); (A.G.H.); (J.H.S.)
- German Cancer Consortium (DKTK), 10117 Berlin, Germany;
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Experimental and Clinical Research Center, Lindenberger Weg 80, 13125 Berlin, Germany
| | - Johannes H. Schulte
- Department of Pediatric Oncology and Hematology, Berlin Institute of Health, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universiät zu Berlin, 10353 Berlin, Germany; (O.S.); (L.G.); (L.A.); (S.S.); (A.K.); (A.W.); (K.A.); (A.E.); (A.G.H.); (J.H.S.)
- German Cancer Consortium (DKTK), 10117 Berlin, Germany;
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Kathleen Anders
- German Cancer Consortium (DKTK), 10117 Berlin, Germany;
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Annette Künkele
- Department of Pediatric Oncology and Hematology, Berlin Institute of Health, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universiät zu Berlin, 10353 Berlin, Germany; (O.S.); (L.G.); (L.A.); (S.S.); (A.K.); (A.W.); (K.A.); (A.E.); (A.G.H.); (J.H.S.)
- German Cancer Consortium (DKTK), 10117 Berlin, Germany;
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Correspondence: ; Tel.: +49-(0)30-450-616178
| |
Collapse
|
8
|
Apoptosis-Inducing TNF Superfamily Ligands for Cancer Therapy. Cancers (Basel) 2021; 13:cancers13071543. [PMID: 33801589 PMCID: PMC8036978 DOI: 10.3390/cancers13071543] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/21/2021] [Accepted: 03/25/2021] [Indexed: 02/07/2023] Open
Abstract
Cancer is a complex disease with apoptosis evasion as one of its hallmarks; therefore, apoptosis induction in transformed cells seems a promising approach as a cancer treatment. TNF apoptosis-inducing ligands, which are naturally present in the body and possess tumoricidal activity, are attractive candidates. The most studied proteins are TNF-α, FasL, and TNF-related apoptosis-inducing ligand (TRAIL). Over the years, different recombinant TNF family-derived apoptosis-inducing ligands and agonists have been designed. Their stability, specificity, and half-life have been improved because most of the TNF ligands have the disadvantages of having a short half-life and affinity to more than one receptor. Here, we review the outlook on apoptosis-inducing ligands as cancer treatments in diverse preclinical and clinical stages and summarize strategies of overcoming their natural limitations to improve their effectiveness.
Collapse
|
9
|
Vacchelli E, Galluzzi L, Rousseau V, Rigoni A, Tesniere A, Delahaye N, Schlemmer FDR, Menger L, Sukkurwala AQ, Adjemian S, Martins I, Michaud M, Dunant A, Kepp O, Brambilla E, Soria JC, Zitvogel L, Kroemer G. Loss-of-function alleles of P2RX7 and TLR4 fail to affect the response to chemotherapy in non-small cell lung cancer. Oncoimmunology 2021; 1:271-278. [PMID: 22737602 PMCID: PMC3382853 DOI: 10.4161/onci.18684] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The success of anticancer chemotherapy relies at least in part on the induction of an immune response against tumor cells. Thus, tumors growing on mice that lack the pattern recognition receptor TLR4 or the purinergic receptor P2RX7 fail to respond to chemotherapy with anthracyclins or oxaliplatin in conditions in which the same neoplasms growing on immunocompetent mice would do so. Similarly, the therapeutic efficacy (measured as progression-free survival) of adjuvant chemotherapy with anthracyclins is reduced in breast cancer patients bearing loss-of-function alleles of TLR4 or P2RX7. TLR4 loss-of-function alleles also have a negative impact on the therapeutic outcome of oxaliplatin in colorectal cancer patients. Here, we report that loss-of-function TLR4 and P2RX7 alleles do not affect overall survival in non-small cell lung cancer (NSCLC) patients, irrespective of the administration and type of chemotherapy. The intrinsic characteristics of NSCLC (which near-to-always is chemoresistant and associated with poor prognosis) and/or the type of therapy that is employed to treat this malignancy (which near-to-always is based on cisplatin) may explain why two genes that affect the immune response to dying cells fail to influence the clinical progression of NSCLC patients.
Collapse
Affiliation(s)
- Erika Vacchelli
- INSERM; U848; Villejuif, France ; Institut Gustave Roussy; Villejuif, France ; Université Paris Sud-XI; Faculté de Médecine; Le Kremlin Bicêtre, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Harnessing the Immune System to Tackle Small Cell Lung Cancer. ACTA ACUST UNITED AC 2020; 26:502-506. [PMID: 33298721 DOI: 10.1097/ppo.0000000000000490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Immunotherapy has improved first-line therapy for small cell lung cancer and has activity in the relapsed setting as well. The immunobiology of small cell lung cancer poses challenges for immunotherapy, and efforts are underway to unlock to the potential of immunotherapy through the identification of meaningful disease subsets and the development of novel combination therapies.
Collapse
|
11
|
miR-196b-5p-mediated downregulation of FAS promotes NSCLC progression by activating IL6-STAT3 signaling. Cell Death Dis 2020; 11:785. [PMID: 32963220 PMCID: PMC7508872 DOI: 10.1038/s41419-020-02997-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 08/31/2020] [Accepted: 09/04/2020] [Indexed: 02/07/2023]
Abstract
Our recent study demonstrated that the QKI-5 regulated miRNA, miR-196b-5p, and it functions as an onco-microRNA in non-small cell lung cancer (NSCLC) by directly targeting GATA6 and TSPAN12. However, the role of miR-196b-5p in NSCLC progression and metastasis still remains unclear. We found that miR-196b-5p promotes lung cancer cell proliferation and colony formation by directly targeting tumor suppressor, FAS. The expression of FAS was significantly downregulated in NSCLC tissue samples and was negatively correlated with the miR-196b-5p expression. Knocking down FAS activates NFkB signaling and subsequent IL6 secretion, resulting in phosphorylation of signal transducer and activator of transcription 3 (STAT3) to promote lung cancer cell growth. Our findings indicated that miR-196b-5p might exhibit novel oncogenic function by FAS-mediated STAT3 activation in NSCLC, and suggested that targeting the miR-196b-5p/FAS/NFkB/IL6/STAT3 pathway might be a promising therapeutic strategy in treating NSCLC.
Collapse
|
12
|
Lin YM, Chen ML, Chen CL, Yeh CM, Sung WW. Overexpression of EIF5A2 Predicts Poor Prognosis in Patients with Oral Squamous Cell Carcinoma. Diagnostics (Basel) 2020; 10:436. [PMID: 32605067 PMCID: PMC7400414 DOI: 10.3390/diagnostics10070436] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/17/2020] [Accepted: 06/25/2020] [Indexed: 12/20/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) is the most common epithelial malignancy affecting the oral cavity, and it is especially significant in Asian countries. Patients diagnosed with OSCC have an unfavorable prognosis and additional prognostic markers would help improve therapeutic strategies. We sought to investigate the association between eukaryotic translation initiation factor 5A2 (EIF5A2) and epithelial-mesenchymal transition (EMT) markers as well as the prognostic significance of EIF5A2 in OSCC. The expression of EIF5A2 and EMT markers was measured through the immunohistochemical staining of specimens from 272 patients with OSCC. In addition, the correlation between different clinicopathological factors and EIF5A2 expression was analyzed. The prognostic role of EIF5A2 was then analyzed via Kaplan-Meier analysis and Cox proportional hazard models. Among the 272 patients, high EIF5A2 expression was significantly associated with an advanced N value (p = 0.008). High tumor expression of EIF5A2 was prone to the expression of low E-cadherin and high beta-catenin (p = 0.046 and p = 0.020, respectively). Patients with high EIF5A2 expression had unfavorable five-year survival rates as compared with those with low expression (49.7% and 67.3%, respectively). The prognostic role of EIF5A2 was further confirmed through multivariate analysis (hazard ratio = 1.714, 95% confidence interval: 1.134-2.590, p = 0.011). High EIF5A2 expression is associated with an advanced N value and EMT markers and may serve as a marker for an unfavorable prognosis in patients with OSCC.
Collapse
Affiliation(s)
- Yueh-Min Lin
- School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan; (Y.-M.L.); (M.-L.C.); (C.-L.C.)
- Department of Surgical Pathology, Changhua Christian Hospital, Changhua 50006, Taiwan;
| | - Mei-Ling Chen
- School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan; (Y.-M.L.); (M.-L.C.); (C.-L.C.)
- Department of Surgical Pathology, Changhua Christian Hospital, Changhua 50006, Taiwan;
| | - Chia-Lo Chen
- School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan; (Y.-M.L.); (M.-L.C.); (C.-L.C.)
| | - Chung-Min Yeh
- Department of Surgical Pathology, Changhua Christian Hospital, Changhua 50006, Taiwan;
- Department of Medical Technology, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli 35664, Taiwan
| | - Wen-Wei Sung
- School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan; (Y.-M.L.); (M.-L.C.); (C.-L.C.)
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
- Department of Urology, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| |
Collapse
|
13
|
Wan Mohd Tajuddin WNB, Lajis NH, Abas F, Othman I, Naidu R. Mechanistic Understanding of Curcumin's Therapeutic Effects in Lung Cancer. Nutrients 2019; 11:E2989. [PMID: 31817718 PMCID: PMC6950067 DOI: 10.3390/nu11122989] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 11/22/2019] [Accepted: 11/30/2019] [Indexed: 12/24/2022] Open
Abstract
Lung cancer is among the most common cancers with a high mortality rate worldwide. Despite the significant advances in diagnostic and therapeutic approaches, lung cancer prognoses and survival rates remain poor due to late diagnosis, drug resistance, and adverse effects. Therefore, new intervention therapies, such as the use of natural compounds with decreased toxicities, have been considered in lung cancer therapy. Curcumin, a natural occurring polyphenol derived from turmeric (Curcuma longa) has been studied extensively in recent years for its therapeutic effects. It has been shown that curcumin demonstrates anti-cancer effects in lung cancer through various mechanisms, including inhibition of cell proliferation, invasion, and metastasis, induction of apoptosis, epigenetic alterations, and regulation of microRNA expression. Several in vitro and in vivo studies have shown that these mechanisms are modulated by multiple molecular targets such as STAT3, EGFR, FOXO3a, TGF-β, eIF2α, COX-2, Bcl-2, PI3KAkt/mTOR, ROS, Fas/FasL, Cdc42, E-cadherin, MMPs, and adiponectin. In addition, limitations, strategies to overcome curcumin bioavailability, and potential side effects as well as clinical trials were also reviewed.
Collapse
Affiliation(s)
- Wan Nur Baitty Wan Mohd Tajuddin
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Selangor Darul Ehsan 47500, Malaysia; (W.N.B.W.M.T.); (I.O.)
| | - Nordin H. Lajis
- Laboratory of Natural Products, Faculty of Science, Universiti Putra Malaysia, UPM, Serdang 43400, Malaysia; (N.H.L.); (F.A.)
| | - Faridah Abas
- Laboratory of Natural Products, Faculty of Science, Universiti Putra Malaysia, UPM, Serdang 43400, Malaysia; (N.H.L.); (F.A.)
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, UPM, Serdang 43400, Malaysia
| | - Iekhsan Othman
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Selangor Darul Ehsan 47500, Malaysia; (W.N.B.W.M.T.); (I.O.)
| | - Rakesh Naidu
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Selangor Darul Ehsan 47500, Malaysia; (W.N.B.W.M.T.); (I.O.)
| |
Collapse
|
14
|
Yadegari M, Sellami M, Riahy S, Mirdar S, Hamidian G, Saeidi A, Abderrahman AB, Hackney AC, Zouhal H. Supplementation of Adiantum capillus-veneris Modulates Alveolar Apoptosis under Hypoxia Condition in Wistar Rats Exposed to Exercise. ACTA ACUST UNITED AC 2019; 55:medicina55070401. [PMID: 31340610 PMCID: PMC6681305 DOI: 10.3390/medicina55070401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 07/13/2019] [Accepted: 07/18/2019] [Indexed: 11/16/2022]
Abstract
Background and Objectives: Several studies have reported that some conditions such as exercise and hypoxia induce DNA damage and dysfunction and apoptosis. Some plant foods contain numerous bioactive compounds and anti-inflammatory properties that can help fight DNA damage. Therefore, the current study evaluated the effect of supplementation of Adiantum capillus-veneris (ACV) extract on Bax/B-cell lymphoma 2 (Bcl-2) ratio apoptotic index and remodeling of pulmonary alveolar epithelial cells in lung tissue of healthy Wistar rats during stressful conditions (hypoxia). Materials and Methods: Twenty-seven Wistar male rats (four-week old, 72 ± 9 g) were randomly assigned into three groups: normoxic, sedentary, and not-supplemented (NG, n = 9); exercise and hypoxia and not-supplemented (HE, n = 9); and exercise and hypoxia and supplemented group (HS, n = 9). The NG remained sedentary in the normoxia environment for nine weeks. The HE group participated in a high-intensity (IT) program for six weeks, then remained sedentary in the hypoxia environment for three weeks. The low-pressure chamber simulated a ~2800 M altitude 24 h/d. HS participated in IT, then entered and remained sedentary in the hypoxia environment for three weeks, and they consumed 500 mg per kg of body weight ACV extract. Results: The Bax/Bcl-2 ratio of the HE group increased significantly (+50.27%, p ≤ 0.05), the average number of type I pneumocytes was reduced significantly (−18.85%, p ≤ 0.05), and the average number of type II pneumocytes was increased significantly (+14.69%, p ≤ 0.05). Also, after three weeks of consuming the ACV extract, the HS group in comparison with the HE group had their Bax/Bcl-2 ratio reduced significantly (−24.27%, p ≤ 0.05), the average number of type I pneumocytes increased significantly (+10.15%, p ≤ 0.05), and the average number of type II pneumocytes reduced significantly (−7.18%, p ≤ 0.05). Conclusion: The findings show that after three weeks of hypoxia following six weeks of high-intensity interval training in Wistar rats, the Bax/Bcl-2 ratio and the number of type II pneumocytes were increased and the number of type I pneumocytes was reduced significantly. These results strongly suggest that an apoptosis state was induced in the lung parenchyma, and consuming ACV extract modulated this state.
Collapse
Affiliation(s)
- Mehdi Yadegari
- Department of Exercise Physiology, Faculty of Physical Education and Sport Sciences,University of Mazandaran, Babolsar 4741613534, Iran
| | - Maha Sellami
- Sport Science Program (SSP), College of Arts and Sciences (CAS), Qatar University, Doha 2713, Qatar
| | - Simin Riahy
- Faculty of Aerospace Medicine and Subsurface, Army Medical University, Tehran 611/14185, Iran
| | - Shadmehr Mirdar
- Department of Exercise Physiology, Faculty of Physical Education and Sport Sciences,University of Mazandaran, Babolsar 4741613534, Iran
| | - Gholamreza Hamidian
- Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz 5166616471, Iran
| | - Ayoub Saeidi
- Department of Biological Sciences in Sport, Faculty of Sports Sciences and Health, Shahid Beheshti University, Tehran 1983969411, Iran
| | | | - Anthony C Hackney
- Department of Exercise & Sport Science, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Hassane Zouhal
- Laboratoire M2S, University of Rennes, EA 1274, F-35000 Rennes, France.
| |
Collapse
|
15
|
Tian Y, Zhai X, Han A, Zhu H, Yu J. Potential immune escape mechanisms underlying the distinct clinical outcome of immune checkpoint blockades in small cell lung cancer. J Hematol Oncol 2019; 12:67. [PMID: 31253167 PMCID: PMC6599302 DOI: 10.1186/s13045-019-0753-2] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 06/14/2019] [Indexed: 12/26/2022] Open
Abstract
Small cell lung cancer (SCLC) is one of the deadliest cancer types in the world. Despite the high response rate to frontline platinum-containing doublets, relapse is inevitable for the majority of patients and the prognosis is poor. Topotecan, which has limited efficacy, has remained the standard second-line therapy for approximately three decades. Although SCLC has a high mutation burden, the clinical efficacy of immune checkpoint blockades (ICBs) in SCLC is far less pronounced than that in non-small cell lung cancer (NSCLC). Only atezolizumab in combination with chemotherapy improved overall survival over chemotherapy alone in the phase III CheckMate 133 trial and has recently received FDA approval as first-line therapy. Most studies concerning ICBs in SCLC are limited to early-phase studies and found that ICBs were not superior to traditional chemotherapy. Why is there such a large difference between SCLC and NSCLC? In this review, comparative analyses of previous studies indicate that SCLC is even more immunodeficient than NSCLC and the potential immune escape mechanisms in SCLC may involve the low expression of PD-L1 and the downregulation of major histocompability complex (MHC) molecules and regulatory chemokines. In consideration of these immune dysfunctions, we speculate that chemotherapy and radiotherapy prior to immunotherapy, the combination of ICBs with antiangiogenic treatment, and selecting tumor mutation burden in combination with PD-L1 expression as biomarkers could be promising strategies to improve the clinical efficacy of immunotherapy for SCLC.
Collapse
Affiliation(s)
- Yaru Tian
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong University, Jinan, China
| | - Xiaoyang Zhai
- Department of Radiation Oncology, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, 250117, China
| | - Anqin Han
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, 250117, China
| | - Hui Zhu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong University, Jinan, China.
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, 250117, China.
| | - Jinming Yu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong University, Jinan, China.
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, 250117, China.
| |
Collapse
|
16
|
Farmer T, O'Neill KL, Naslavsky N, Luo X, Caplan S. Retromer facilitates the localization of Bcl-xL to the mitochondrial outer membrane. Mol Biol Cell 2019; 30:1138-1146. [PMID: 30840537 PMCID: PMC6724524 DOI: 10.1091/mbc.e19-01-0044] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The anti-apoptotic Bcl-2 family protein Bcl-xL plays a critical role in cell survival by protecting the integrity of the mitochondrial outer membrane (MOM). The mechanism through which Bcl-xL is recruited to the MOM has not been fully discerned. The retromer is a conserved endosomal scaffold complex involved in membrane trafficking. Here we identify VPS35 and VPS26, two core components of the retromer, as novel regulators of Bcl-xL. We observed interactions and colocalization between Bcl-xL, VPS35, VPS26, and MICAL-L1, a protein involved in recycling endosome biogenesis that also interacts with the retromer. We also found that upon VPS35 depletion, levels of nonmitochondrial Bcl-xL were increased. In addition, retromer-depleted cells displayed more rapid Bax activation and apoptosis. These results suggest that the retromer regulates apoptosis by facilitating Bcl-xL's transport to the MOM. Importantly, our studies suggest a previously uncharacterized relationship between the machineries of cell death/survival and endosomal trafficking.
Collapse
Affiliation(s)
- Trey Farmer
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870
| | - Katelyn L O'Neill
- Eppley Institute for Research in Cancer and Allied Diseases, The Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198-5870
| | - Naava Naslavsky
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870
| | - Xu Luo
- Eppley Institute for Research in Cancer and Allied Diseases, The Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198-5870
| | - Steve Caplan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870.,Eppley Institute for Research in Cancer and Allied Diseases, The Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198-5870
| |
Collapse
|
17
|
Gravett AM, Dalgleish AG, Copier J. In vitro culture with gemcitabine augments death receptor and NKG2D ligand expression on tumour cells. Sci Rep 2019; 9:1544. [PMID: 30733494 PMCID: PMC6367314 DOI: 10.1038/s41598-018-38190-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 12/06/2018] [Indexed: 01/11/2023] Open
Abstract
Much effort has been made to try to understand the relationship between chemotherapeutic treatment of cancer and the immune system. Whereas much of that focus has been on the direct effect of chemotherapy drugs on immune cells and the release of antigens and danger signals by malignant cells killed by chemotherapy, the effect of chemotherapy on cells surviving treatment has often been overlooked. In the present study, tumour cell lines: A549 (lung), HCT116 (colon) and MCF-7 (breast), were treated with various concentrations of the chemotherapeutic drugs cyclophosphamide, gemcitabine (GEM) and oxaliplatin (OXP) for 24 hours in vitro. In line with other reports, GEM and OXP upregulated expression of the death receptor CD95 (fas) on live cells even at sub-cytotoxic concentrations. Further investigation revealed that the increase in CD95 in response to GEM sensitised the cells to fas ligand treatment, was associated with increased phosphorylation of stress activated protein kinase/c-Jun N-terminal kinase and that other death receptors and activatory immune receptors were co-ordinately upregulated with CD95 in certain cell lines. The upregulation of death receptors and NKG2D ligands together on cells after chemotherapy suggest that although the cells have survived preliminary treatment with chemotherapy they may now be more susceptible to immune cell-mediated challenge. This re-enforces the idea that chemotherapy-immunotherapy combinations may be useful clinically and has implications for the make-up and scheduling of such treatments.
Collapse
Affiliation(s)
- Andrew M Gravett
- Oncology Group, Institute for Infection and Immunity, St. George's, University of London, London, UK.
| | - Angus G Dalgleish
- Oncology Group, Institute for Infection and Immunity, St. George's, University of London, London, UK
| | - John Copier
- Oncology Group, Institute for Infection and Immunity, St. George's, University of London, London, UK
| |
Collapse
|
18
|
Nishihira M, Nakazato Y, Maeda S, Inoue T, Araki O, Karube Y, Chida M. Impact of tumor infiltrating lymphocytes and lymphoid follicle formation on patient survival following surgery for lung squamous cell carcinoma. Thorac Cancer 2018; 10:219-225. [PMID: 30561902 PMCID: PMC6360201 DOI: 10.1111/1759-7714.12935] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 11/15/2018] [Accepted: 11/16/2018] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Tumor infiltrating lymphocytes (TILs) are known to correlate with the prognosis of patients affected by a variety of cancer types. We evaluated TILs in patients who underwent surgery for lung squamous cell carcinoma (SCC). METHODS Specimens obtained from patients during resection of lung SCC were examined for TIL density, lymphoid follicle formation, PD-L1 expression, and the appearance of regulatory T cells (Tregs). RESULTS We enrolled 72 patients who underwent surgery for SCC (TIL grades 0, 1, and 2: 29, 18, and 25, respectively). Lymphoid follicles were observed in 13 (18.1%) patients and 8 were positive for Tregs, which were always observed in association with lymphoid follicles (P < 0.001). Multivariate analysis revealed that lymphoid follicle formation, the appearance of Tregs, pathological stage, and pleural invasion were independent prognostic factors related to overall survival, whereas TIL density and PD-L1 expression were not. CONCLUSION SCC patients with lymphoid follicle formation accompanied by Tregs show poor survival following lung resection surgery.
Collapse
Affiliation(s)
- Morimichi Nishihira
- Department of General Thoracic Surgery, Dokkyo Medical University School of Medicine, Tochigi, Japan
| | - Yoshimasa Nakazato
- Department of Diagnostic Pathology, Dokkyo Medical University School of Medicine, Tochigi, Japan
| | - Sumiko Maeda
- Department of General Thoracic Surgery, Dokkyo Medical University School of Medicine, Tochigi, Japan
| | - Takashi Inoue
- Department of General Thoracic Surgery, Dokkyo Medical University School of Medicine, Tochigi, Japan
| | - Osamu Araki
- Department of General Thoracic Surgery, Dokkyo Medical University School of Medicine, Tochigi, Japan
| | - Yoko Karube
- Department of General Thoracic Surgery, Dokkyo Medical University School of Medicine, Tochigi, Japan
| | - Masayuki Chida
- Department of General Thoracic Surgery, Dokkyo Medical University School of Medicine, Tochigi, Japan
| |
Collapse
|
19
|
Ezzeldin N, Farouk H, Kandil DM, Darwish A, El-Bastawisy A. Impact of cell death pathway genes Fas 21377AA and FasL 2844CC polymorphisms on the risk of developing non-small cell lung cancer. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2018. [DOI: 10.1016/j.ejmhg.2017.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
20
|
Merchant N, Nagaraju GP, Rajitha B, Lammata S, Jella KK, Buchwald ZS, Lakka SS, Ali AN. Matrix metalloproteinases: their functional role in lung cancer. Carcinogenesis 2017. [DOI: 10.1093/carcin/bgx063] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
21
|
FAIM2, as a novel diagnostic maker and a potential therapeutic target for small-cell lung cancer and atypical carcinoid. Sci Rep 2016; 6:34022. [PMID: 27677402 PMCID: PMC5039724 DOI: 10.1038/srep34022] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 09/06/2016] [Indexed: 12/23/2022] Open
Abstract
Lung neuroendocrine (NE) tumors are a heterogeneous group of tumors arising from neuroendocrine cells that includes typical carcinoid, atypical carcinoid, small cell lung cancer (SCLC), and large cell NE cancer. The subtyping of NE tumors is based on the number of mitoses per high powered field and the presences of necrosis. However, the best diagnostic criteria to differentiate various subtypes of lung NE tumors remains controversial and few diagnostic markers distinguish typical and atypical carcinoid. In this study, we show that FAIM2, an inhibitory molecule in the Fas-apoptosis pathway, is significantly overexpressed in SCLC compared to non-small cell lung cancer. In addition, FAIM2 expression is significantly higher in atypical carcinoid than typical carcinoid. As atypical carcinoid has been shown to have worse clinical outcomes than typical carcinoid, our data suggests that FAIM2 may be a useful diagnostic marker for atypical carcinoid. Knockdown of FAIM2 expression increases Fas-induced apoptotic cell death in SCLC cells. Etoposide treatment combined with FAIM2 inhibition also shows modest but significant reduction of viable SCLC cells. Taken together, our results suggest that FAIM2 is a potential NE tumor marker with higher expression in atypical carcinoid and SCLC, and could be a new therapeutic target for SCLC.
Collapse
|
22
|
Sharma G, Rani I, Bhatnagar A, Agnihotri N. Apoptosis-Mediated Chemoprevention by Different Ratios of Fish Oil in Experimental Colon Carcinogenesis. Cancer Invest 2016; 34:220-30. [PMID: 27191482 DOI: 10.1080/07357907.2016.1183023] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Apoptosis plays an important role in prevention of colon cancer. In the present study, different ratios of fish oil and corn oil increased Fas expression in both phases and a decrease in FasL expression only in post initiation phase. Treatment with fish oil activated the intrinsic apoptotic pathway by increasing Bax expression and Cyt c release and decreasing Bcl-2 levels in both phases. This suggests that intrinsic pathway is upregulated by fish oil; however, Fas-FasL activity may be involved in inhibition of reversal of immune surveillance in tumor cells.
Collapse
Affiliation(s)
- Gayatri Sharma
- a Department of Biochemistry , Panjab University , Chandigarh , India
| | - Isha Rani
- a Department of Biochemistry , Panjab University , Chandigarh , India
| | - Archana Bhatnagar
- a Department of Biochemistry , Panjab University , Chandigarh , India
| | - Navneet Agnihotri
- a Department of Biochemistry , Panjab University , Chandigarh , India
| |
Collapse
|
23
|
Yu X, Li Y, Yu Y, Lei J, Wan G, Cao F. Associations between FAS rs2234767 and FASL rs763110 polymorphisms and the risk of lung cancer: a meta-analysis of 39,736 subjects. Onco Targets Ther 2016; 9:2049-56. [PMID: 27103831 PMCID: PMC4827906 DOI: 10.2147/ott.s102723] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Background Previous studies have investigated the associations between the common polymorphisms in FAS/FASL genes and lung cancer risk; however, the results remain inconsistent and inconclusive. Hence, we performed a meta-analysis to reassess the relationships between FAS rs2234767 and FASL rs763110 polymorphisms and the risk of lung cancer. Methods Eligible studies retrieved by an electronic search were pooled to calculate the strength of the associations using the odds ratio (OR) and 95% confidence interval (95% CI). Results A total of 13 case–control studies involving 39,736 subjects (9,237 cases and 10,838 controls on FAS rs2234767 and 8,957 cases and 10,704 controls on FASL rs763110) were included in the meta-analysis. The results showed a significant association between FAS rs2234767 polymorphism and increased risk of lung cancer (A vs G: OR =1.07, 95% CI =1.01–1.13; AA vs GG: OR =1.23, 95% CI =1.06–1.43; AA vs GA + GG: OR =1.24, 95% CI =1.08–1.43). Similar association was also observed in Asian population (AA vs GA + GG: OR =1.30, 95% CI =1.01–1.67) and in the studies with large sample size (A vs G: OR =1.07, 95% CI =1.00–1.14; AA vs GG: OR =1.30, 95% CI =1.07–1.58). However, no significant association between FASL rs763110 polymorphism and lung cancer risk was found other than in the Asian population (CC vs TC + TT: OR =1.35, 95% CI =1.01–1.80). Conclusion The meta-analysis indicated that FAS rs2234767 polymorphism was significantly associated with an increased risk of lung cancer and FASL rs763110 polymorphism may not contribute to susceptibility to lung cancer other than in Asian population.
Collapse
Affiliation(s)
- Xiongjie Yu
- Department of Oncology, Hubei University of Medicine, Shiyan, Hubei, People's Republic of China
| | - Yanli Li
- Department of Endocrinology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, People's Republic of China
| | - Yuandong Yu
- Department of Oncology, Hubei University of Medicine, Shiyan, Hubei, People's Republic of China
| | - Jinhua Lei
- Department of Oncology, Hubei University of Medicine, Shiyan, Hubei, People's Republic of China
| | - Guoxing Wan
- Department of Oncology, Hubei University of Medicine, Shiyan, Hubei, People's Republic of China
| | - Fengjun Cao
- Department of Oncology, Hubei University of Medicine, Shiyan, Hubei, People's Republic of China
| |
Collapse
|
24
|
Huang CC, Wu DW, Lin PL, Lee H. Paxillin promotes colorectal tumor invasion and poor patient outcomes via ERK-mediated stabilization of Bcl-2 protein by phosphorylation at Serine 87. Oncotarget 2016; 6:8698-708. [PMID: 25826088 PMCID: PMC4496177 DOI: 10.18632/oncotarget.3537] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 02/10/2015] [Indexed: 01/13/2023] Open
Abstract
Stabilization of Bcl-2 protein by paxillin (PXN)-mediated ERK activation was recently reported to cause an unfavorable response to 5-Fluorouracil-based chemotherapy. Here, we present evidence from cell and animal models to demonstrate that stabilization of Bcl-2 protein by phosphorylation at Serine 87 (pBcl-2-S87) via PXN-mediated ERK activation is responsible for cancer cell invasiveness and occurs via upregulation of MMP2 expression. Immunostainings of 190 tumors resected from colorectal cancer patients indicated that PXN expression was positively correlated with Bcl-2, pBcl-2-S87, and MMP2 expression. A positive correlation of pBcl-2-S87 with Bcl-2 and MMP2 was also observed in this study population. Patients with high PXN, Bcl-2, pBcl-2-S87, and MMP2 had poor overall survival (OS) and shorter relapse free survival (RFS). In conclusion, PXN promotes Bcl-2 phosphorylation at Serine 87 via PXN-mediated ERK activation, and its stabilization associated with increased tumor formation efficacy in mice and poor patient outcome in colorectal cancer patients.
Collapse
Affiliation(s)
- Chi-Chou Huang
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Surgery, Division of Colon and Rectum, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - De-Wei Wu
- Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan
| | - Po-Lin Lin
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Huei Lee
- Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
25
|
Azzimonti B, Zavattaro E, Provasi M, Vidali M, Conca A, Catalano E, Rimondini L, Colombo E, Valente G. Intense Foxp3+CD25+regulatory T-cell infiltration is associated with high-grade cutaneous squamous cell carcinoma and counterbalanced by CD8+/Foxp3+CD25+ratio. Br J Dermatol 2014; 172:64-73. [DOI: 10.1111/bjd.13172] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/28/2014] [Indexed: 02/01/2023]
Affiliation(s)
- B. Azzimonti
- Department of Health Sciences; Medical School; University of Piemonte Orientale ‘A. Avogadro’; 28100 Novara Italy
| | - E. Zavattaro
- Dermatology; Azienda Ospedaliero-Universitaria ‘Maggiore della Carità’; Università del Piemonte Orientale ‘A. Avogadro’; 28100 Novara Italy
| | - M. Provasi
- Dermatology; Azienda Ospedaliero-Universitaria ‘Maggiore della Carità’; Università del Piemonte Orientale ‘A. Avogadro’; 28100 Novara Italy
| | - M. Vidali
- Clinical Chemistry; Azienda Ospedaliero-Universitaria ‘Maggiore della Carità’; Università del Piemonte Orientale ‘A. Avogadro’; 28100 Novara Italy
| | - A. Conca
- Pathology Units; Azienda Ospedaliero-Universitaria ‘Maggiore della Carità’; Università del Piemonte Orientale ‘A. Avogadro’; 28100 Novara Italy
| | - E. Catalano
- Department of Health Sciences; Medical School; University of Piemonte Orientale ‘A. Avogadro’; 28100 Novara Italy
| | - L. Rimondini
- Department of Health Sciences; Medical School; University of Piemonte Orientale ‘A. Avogadro’; 28100 Novara Italy
| | - E. Colombo
- Dermatology; Azienda Ospedaliero-Universitaria ‘Maggiore della Carità’; Università del Piemonte Orientale ‘A. Avogadro’; 28100 Novara Italy
- Department of Translational Medicine; Medical School; University of Piemonte Orientale ‘A. Avogadro’; 28100 Novara Italy
| | - G. Valente
- Pathology Units; Azienda Ospedaliero-Universitaria ‘Maggiore della Carità’; Università del Piemonte Orientale ‘A. Avogadro’; 28100 Novara Italy
- Department of Translational Medicine; Medical School; University of Piemonte Orientale ‘A. Avogadro’; 28100 Novara Italy
| |
Collapse
|
26
|
Li L, Yao YC, Fang SH, Ma CQ, Cen Y, Xu ZM, Dai ZY, Li C, Li S, Zhang T, Hong HH, Qi WW, Zhou T, Li CY, Yang X, Gao GQ. Pigment epithelial-derived factor (PEDF)-triggered lung cancer cell apoptosis relies on p53 protein-driven Fas ligand (Fas-L) up-regulation and Fas protein cell surface translocation. J Biol Chem 2014; 289:30785-30799. [PMID: 25225287 DOI: 10.1074/jbc.m114.590000] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Pigment epithelium-derived factor (PEDF), a potent antiangiogenesis agent, has recently attracted attention for targeting tumor cells in several types of tumors. However, less is known about the apoptosis-inducing effect of PEDF on human lung cancer cells and the underlying molecular events. Here we report that PEDF has a growth-suppressive and proapoptotic effect on lung cancer xenografts. Accordingly, in vitro, PEDF apparently induced apoptosis in A549 and Calu-3 cells, predominantly via the Fas-L/Fas death signaling pathway. Interestingly, A549 and Calu-3 cells are insensitive to the Fas-L/Fas apoptosis pathway because of the low level of cell surface Fas. Our results revealed that, in addition to the enhancement of Fas-L expression, PEDF increased the sensitivity of A549 and Calu-3 cells to Fas-L-mediated apoptosis by triggering the translocation of Fas protein to the plasma membrane in a p53- and FAP-1-dependent manner. Similarly, the up-regulation of Fas-L by PEDF was also mediated by p53. Furthermore, peroxisome proliferator-activated receptor γ was determined to be the upstream regulator of p53. Together, these findings uncover a novel mechanism of tumor cell apoptosis induced by PEDF and provide a potential therapeutic strategy for tumors that are insensitive to Fas-L/Fas-dependent apoptosis because of a low level of cell surface Fas.
Collapse
Affiliation(s)
- Lei Li
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China,; Department of Reproductive Medicine Center, Key Laboratory for Reproductive Medicine of Guangdong Province, Third Affiliated Hospital of Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China
| | - Ya-Chao Yao
- Laboratory Center of Guangdong No. 2 Provincial People's Hospital, Guangzhou 510317, Guangdong Province, China
| | - Shu-Huan Fang
- DME Center, Clinical Pharmacology Institute, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Cai-Qi Ma
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Yi Cen
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Zu-Min Xu
- Cancer Center, Affiliated Hospital of Guangdong Medical College, Zhanjiang 524000, China
| | - Zhi-Yu Dai
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Cen Li
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Shuai Li
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Ting Zhang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Hong-Hai Hong
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Wei-Wei Qi
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Ti Zhou
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Chao-Yang Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China,.
| | - Xia Yang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China,; China Key Laboratory of Tropical Disease Control, Sun Yat-sen University, Ministry of Education, Guangzhou 510080, China, and.
| | - Guo-Quan Gao
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China,; Key Laboratory of Functional Molecules from Marine Microorganisms, Sun Yat-sen University, Department of Education of Guangdong Province, Guangdong 510080, China.
| |
Collapse
|
27
|
Yuan HP, Liu QD, Li GQ, Cong YQ. Fas −670A/G (rs1800682) Polymorphism and Digestive Cancer Risk in Asians: A Meta-Analysis. Genet Test Mol Biomarkers 2014; 18:482-8. [PMID: 24800976 DOI: 10.1089/gtmb.2014.0032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Hai-Peng Yuan
- Department of Gastroenterology, Tai'an City Central Hospital, Tai'an, China
| | - Qing-Dong Liu
- Department of Emergency, Tai'an City Hospital of Traditional Chinese Medicine, Tai'an, China
| | - Gai-Qin Li
- Department of Gastroenterology, Tai'an City Central Hospital, Tai'an, China
| | - Yan-Qun Cong
- Department of Gastroenterology, Zhejiang Hospital, Hangzhou, China
| |
Collapse
|
28
|
Zhu J, Lu L, Cheng X, Xie R, Chen Z, Li Y, Lin G, Liu J, Yang Y. Association between CD95L polymorphism and cervical cancer risk: evidence from a meta-analysis. Tumour Biol 2014; 35:5137-42. [PMID: 24619598 DOI: 10.1007/s13277-014-1652-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2013] [Accepted: 01/12/2014] [Indexed: 01/17/2023] Open
Abstract
Several studies have assessed the association of CD95L polymorphism with cervical cancer risk, but the data lack the power to provide compelling evidence. In this study, we aimed to clarify the association through a meta-analysis. A comprehensive search was conducted in PubMed, Embase, and Web of Science. The fixed-effects model was used to calculate odds ratio (OR) with 95 % confidence intervals (CIs). A total of five papers with six case-control studies were derived and finally included in this meta-analysis. The overall estimate did not reveal any significant association between CD95L -844C/T polymorphism and cervical cancer risk. Subgroup analysis in Asian population indicated nonsignificant nevertheless potentially increased risk in CC genotype carriers in comparison with the carriers of CT+TT genotypes (ORCC vs. CT+TT=1.16, 95 % CI=0.99-1.36, P for heterogeneity=0.231). Based on current epidemiological studies, this meta-analysis suggests that CD95L polymorphism may not be a risk factor contributing to cervical cancer development.
Collapse
Affiliation(s)
- Jing Zhu
- Department of Gynaecology and Obstetrics, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Wang X, Xing GH, Fan CC. Association between the FAS rs2234767G/A polymorphism and cancer risk: a systematic review and meta-analysis. DNA Cell Biol 2014; 33:320-7. [PMID: 24568648 DOI: 10.1089/dna.2013.2273] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Abnormal regulation of apoptosis can lead to carcinogenesis. Single nucleotide polymorphisms in apoptotic genes have been associated with cancer risk, such as the FAS rs2234767G/A polymorphism, which alters transcription of the FAS promoter. Downregulation of FAS, with resultant cellular resistance to death signals, has been found in many cancers. However, the association between the FAS rs2234767G/A polymorphism and cancer risk is still controversial. Here, we performed a meta-analysis including 41 articles (44 case-control studies, 17,814 cases and 24,307 controls) identified from PubMed and Chinese language (CNKI and WanFang) databases related to cancer susceptibility and the FAS rs2234767G/A polymorphism. We used odds ratios (ORs) and 95% confidence intervals (CIs) to assess the strength of the associations. We found that the rs2234767 G-allele was a protective factor for cancer risk (GG vs. AA: OR=0.88, 95% CI=0.79-0.98; GG+GA vs. AA: OR=0.87, 95% CI=0.79-0.96). Similar associations were detected in the "source of control", ethnicity, and cancer type subgroups. Further studies on a larger sample size and considering gene-environment interactions should be conducted to confirm the role of FAS polymorphisms, especially rs2234767G/A, in cancer risk.
Collapse
Affiliation(s)
- Xin Wang
- 1 Department of Respiration, General Hospital of Jinan Military Command , Jinan, People's Republic of China
| | | | | |
Collapse
|
30
|
FasL gene -844T/C mutation of esophageal cancer in South China and its clinical significance. Sci Rep 2014; 4:3866. [PMID: 24473454 PMCID: PMC5379236 DOI: 10.1038/srep03866] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 01/07/2014] [Indexed: 01/22/2023] Open
Abstract
In this study, we investigated the association between the FasL -844T/C polymorphism and the risk of developing esophageal squamous cell carcinoma (ESCC) in South China. For the investigation, we randomly selected 248 patients suffering from ESCC from Southern China along with 297 healthy individuals as the control group. The relationship between the FasL gene -844T/C SNP and ESCC was studied using PCR-RFLP and immunohistochemistry. The Fas -1377G/A SNP was also selected for investigation to detect whether it interferes with the functional effect of the FasL -844C/T polymorphism in ESCC development. A significant difference in the FasL -844T/C genotypes between the patients and the control group was observed (P<0.05), with those expressing the C allele having a significantly reduced risk of developing ESCC, however younger patients (<60 years) exhibited a more
malignant pathological T grade if they were homozygous for the C allele. FasL -844 CC combined with the Fas -1377 G allele is a protective factor against ESCC. Having said this, even though the C allele has a protective effect prior to development of ESCC, once the host does develop the condition the tumour will develop faster and have a higher degree of malignancy than T carriers.
Collapse
|
31
|
FAS−1377 A/G polymorphism in breast cancer: a meta-analysis. Tumour Biol 2013; 35:2575-81. [DOI: 10.1007/s13277-013-1339-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 10/16/2013] [Indexed: 01/04/2023] Open
|
32
|
Zhong-Xing Z, Yuan-Yuan M, Hai Zhen M, Jian-Gang Z, Li-Feng Z. FAS-1377 G/A (rs2234767) polymorphism and cancer susceptibility: a meta-analysis of 17,858 cases and 24,311 controls. PLoS One 2013; 8:e73700. [PMID: 24014103 PMCID: PMC3754923 DOI: 10.1371/journal.pone.0073700] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Accepted: 07/19/2013] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Disruption of apoptosis has been implicated in carcinogenesis. Specifically, various single-nucleotide polymorphisms (SNPs) in apoptotic genes, such as FAS-1377 G/A SNP, have been associated with cancer risk. FAS-1377 G/A SNP has been shown to alter FAS gene promoter transcriptional activity. Down-regulation of FAS and cell death resistance is key to many cancers, but an association between FAS-1377 G/A SNP and cancer risk is uncertain. Therefore, we conducted a meta-analysis of the current literature to clarify this relationship. METHODOLOGY/PRINCIPAL FINDINGS From PubMed and Chinese language (CNKI and WanFang) databases, we located articles published up to March 5, 2013, obtaining 44 case-control studies from 41 different articles containing 17,858 cases and 24,311 controls based on search criteria for cancer susceptibility related to the FAS gene -1377 G/A SNP. Odds ratios (ORs) and 95% confidence intervals (CI) revealed association strengths. Data show that the -1377 G allele was protective against cancer risk. Similar associations were detected in "source of control," ethnicity and cancer type subgroups. Lower cancer risk was found in both smokers with a GG+GA genotype and in non-smokers with the GG+GA genotype, when compared to smokers and nonsmokers with the AA genotype. Males carrying the -1377G allele (GG+GA) had lower cancer incidence than those with the AA genotype. Individuals who carried both FAS-1377(GG+GA)/FASL-844(TT+TC) genotypes appeared to have lower risk of cancer than those who carried both FAS-1377 AA/FASL-844 CC genotypes. CONCLUSIONS/SIGNIFICANCE The FAS-1377 G/A SNP may decrease cancer risk. Studies with larger samples to study gene-environment interactions are warranted to understand the role of FAS gene polymorphisms, especially -1377 G/A SNP, in cancer risk.
Collapse
Affiliation(s)
- Zhou Zhong-Xing
- Department of Urology, The Affiliated Changzhou No 2. Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Mi Yuan-Yuan
- Department of Urology, The Third Affiliated Hospital of Nantong University, Wuxi, Jiangsu, China
| | - Ma Hai Zhen
- Department of Operating Room, The Third Affiliated Hospital of Nantong University, Wuxi, Jiangsu, China
| | - Zou Jian-Gang
- Department of Urology, The Affiliated Changzhou No 2. Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Zhang Li-Feng
- Department of Urology, The Affiliated Changzhou No 2. Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| |
Collapse
|
33
|
Functional Investigation of Fas Ligand Expressions in Human Non-Small Cell Lung Cancer Cells and Its Clinical Implications. Ann Thorac Surg 2013; 95:412-8. [DOI: 10.1016/j.athoracsur.2012.08.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Revised: 07/31/2012] [Accepted: 08/02/2012] [Indexed: 01/06/2023]
|
34
|
Chen CJ, Sung WW, Su TC, Chen MK, Wu PR, Yeh KT, Ko JL, Lee H. High expression of interleukin 10 might predict poor prognosis in early stage oral squamous cell carcinoma patients. Clin Chim Acta 2013; 415:25-30. [PMID: 22981868 DOI: 10.1016/j.cca.2012.09.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2012] [Revised: 08/30/2012] [Accepted: 09/06/2012] [Indexed: 11/20/2022]
Abstract
BACKGROUND Interleukin 10 (IL10) plays an important role in immunosuppression and suppression of antitumor immunity. This study examined the IL10 expression of tumor cells and assessed its significance in patients with oral squamous cell carcinoma (OSCC). METHODS Tumor tissues and adjacent normal tissues were obtained from 325 patients with OSCC and were arranged in a tissue microarray. We examined 325 surgical specimens for associations between IL10 expression in tumor cells and clinical parameters of oral cancer. RESULTS High IL10 expression in OSCC patients was significantly associated with male gender (P<0.001), smoking (P=0.015), alcohol consumption (P=0.018), betel quid chewing (P=0.003), poor relapse free survival (P=0.012), and poor overall survival (P=0.001). Patients with high IL10 expression, and particularly early stage OSCC patients, had significantly worse overall survival as defined by the log-rank test (P=0.014 for all cases; P=0.004 for early stage patients). In early stage patients, high IL10 expression in tumor cells was associated with poor prognosis (P=0.018) and a 1.99-fold higher death risk, as determined by Cox regression. CONCLUSION High IL10 expression is significantly associated with aggressive clinical manifestations and might be an independent survival predictor, particularly in early stage OSCC patients.
Collapse
Affiliation(s)
- Chih-Jung Chen
- Institute of Medicine, Chung Shan Medical University, Taichuang, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Tian J, Pan F, Li J, Ma Y, Cen H, Pan HF, Pan YY, Ye DQ. Association between the FAS/FASL polymorphisms and gastric cancer risk: a meta-analysis. Asian Pac J Cancer Prev 2012; 13:945-51. [PMID: 22631677 DOI: 10.7314/apjcp.2012.13.3.945] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE FAS/FASL gene promoter polymorphisms have been repeatedly associated with gastric cancer risk, but findings are inconclusive across studies. To address a more precise estimation of the relationship, a meta-analysis was performed. METHODS Data were collected from the Pubmed, Medline and EMBASE databases, with the last report up to 1 December, 2011. Crude ORs with 95% CIs were used to assess the strength of the association by (1) the additive, (2) the codominant, (3) the dominant, and (4) the recessive models. RESULTS A total of seven studies, including six studies on FAS -1377G>A polymorphism, five studies on FAS -670A>G polymorphism, and six studies on FASL -844T>C polymorphism, were identified in the current meta-analysis. Overall, an association of FAS -1377G>A (AA versus GG: OR = 1.313, 95% CI = 1.045-1.650, Ph = 0.347, I2 = 10.8) and FASL -844T>C (CC versus TT: OR = 1.352, 95% CI = 1.043-1.752, Ph = 0.461, I2 = 0.0) polymorphisms with gastric cancer was found in the codominant model. However, we did not detect any association between gastric cancer and the FAS -670A>G polymorphism. In the subgroup analysis by ethnicity, similar elevated risks were also observed in Asian population for FAS -1377G>A (AA versus GG: OR = 1.309, 95% CI = 1.041- 1.646, Ph = 0.240, I2 = 27.3) and FASL -844T>C (CC versus TT: OR = 1.420, 95% CI = 1.081-1.865, Ph = 0.524, I2 = 0.0) polymorphisms. CONCLUSIONS This meta-analysis indicated that FAS -1377G>A and FASL -844T>C polymorphisms might be associated with gastric cancer risk.
Collapse
Affiliation(s)
- Jing Tian
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Hammam O, Mahmoud O, Zahran M, Aly S, Hosny K, Helmy A, Anas A. The role of fas/fas ligand system in the pathogenesis of liver cirrhosis and hepatocellular carcinoma. HEPATITIS MONTHLY 2012; 12:e6132. [PMID: 23300494 PMCID: PMC3539063 DOI: 10.5812/hepatmon.6132] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Revised: 08/01/2012] [Accepted: 08/15/2012] [Indexed: 12/11/2022]
Abstract
BACKGROUND The Fas receptor/ligand system including soluble forms is the most important apoptotic initiator in the liver. Dysregulation of this pathway may contribute to abnormal cell proliferation and cell death and is regarded as one of the mechanisms preventing the immune system from rejecting the tumor cells. OBJECTIVES To analyze the role of Fas system Fas/ Fas ligand (Fas/ FasL) in the multi-step process of hepatic fibrosis/carcinogenesis, and to use of the serum markers as possible candidate biomarkers for early detection of hepatocellular carcinoma (HCC). PATIENTS AND METHODS NINETY PATIENTS WERE ENROLLED: 30 cases of chronic hepatitis C (CHC) without cirrhosis, 30 cases of CHC with liver cirrhosis, and 30 cases of HCC and hepatitis V virus (HCV) infection. Ten wedge liver biopsies, taken during laparoscopic cholecystectomy, were served as normal controls. Serum soluble Fas (sFas) levels were measured using ELISA technique; Fas and FasL proteins were detected in hepatic tissue by indirect Immuno-histochemical technique (IHC); electron microscopic (EM) and immune electron microscopic examinations were performed for detection of Fas expression on lymphocytes. RESULTS Hepatic expression of both Fas and FasL as well as expression of Fas on separated lymphocytes were significantly increased in the diseased groups (P < 0. 01) compared to the control specimens. The highest expression was noticed in CHC specimens, particularly with the necro-inflammatory activity and advancement of the fibrosis. The sFas in cirrhotic patients and HCC were significantly higher than that in normal controls and CHC without cirrhosis group (P < 0.01). CONCLUSIONS Apoptosis and the Fas system were significantly involved in the process of converting liver cirrhosis into hepatocellular carcinoma. Down-regulation of Fas expression, up regulation of FasL expression in hepatocytes, and elevation of serum sFas levels were important in tumor evasion from immune surveillance, and in hepatic carcinogenesis.
Collapse
Affiliation(s)
- Olfat Hammam
- Departments of Pathology, Theodor Bilharz Research Institute, Cairo, Egypt
| | - Ola Mahmoud
- Departments of Hematology, Theodor Bilharz Research Institute, Cairo, Egypt
- Corresponding author: Ola Mahmoud, Departments of Hematology, Theodor Bilharz Research Institute, No. 1 Sharara Building, Hassan Elmamoun St., Nasr City, Cairo, Egypt. Tel.: +20-224723116, Fax: +20-1224590806, E-mail:
| | - Manal Zahran
- Departments of Hematology, Theodor Bilharz Research Institute, Cairo, Egypt
| | - Sohair Aly
- Malaysia and Medicinal Chemistry Department, Advanced Dental and Medical institute, IPPT, USM, NRC, Cairo, Egypt
| | - Karim Hosny
- Surgical Department, Kasr El Aini Hospital, Cairo, Egypt
| | - Amira Helmy
- Departments of Electron Microscopy, Theodor Bilharz Research Institute, Cairo, Egypt
| | - Amgad Anas
- Departments of Gastroenterology, Theodor Bilharz Research Institute, Cairo, Egypt
| |
Collapse
|
37
|
Abstract
Since the initial description of apoptosis, a number of different forms of cell death have been described. In this review we will focus on classic caspase-dependent apoptosis and its variations that contribute to diseases. Over fifty years of research have clarified molecular mechanisms involved in apoptotic signaling as well and shown that alterations of these pathways lead to human diseases. Indeed both reduced and increased apoptosis can result in pathology. More recently these findings have led to the development of therapeutic approaches based on regulation of apoptosis, some of which are in clinical trials or have entered medical practice.
Collapse
Affiliation(s)
- Bartolo Favaloro
- Dipartimento di Scienze Biomediche, Universita' "G. d'Annunzio" Chieti-Pescara, Italy
| | | | | | | | | |
Collapse
|
38
|
Wu DW, Tsai LH, Chen PM, Lee MC, Wang L, Chen CY, Cheng YW, Lee H. Loss of TIMP-3 promotes tumor invasion via elevated IL-6 production and predicts poor survival and relapse in HPV-infected non-small cell lung cancer. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 181:1796-806. [PMID: 22982189 DOI: 10.1016/j.ajpath.2012.07.032] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Revised: 06/19/2012] [Accepted: 07/20/2012] [Indexed: 12/21/2022]
Abstract
Human papillomavirus (HPV) 16/18 E6 oncoprotein is expressed in lung tumors and is associated with p53 inactivation. The tissue inhibitor of metalloproteinase 3 (TIMP-3) is essential for limiting inflammation; therefore, we expected that TIMP-3 loss might induce chronic inflammation, thereby promoting tumor malignancy as well as poor survival and relapse in patients with HPV-infected non-small cell lung cancer. In this study, the loss of TIMP-3 by loss of heterozygosity and/or promoter hypermethylation was more frequent in HPV16/18 E6-positive tumors than in E6-negative tumors. To explore the possible underlying mechanism, E6-negative TL4 and CL1-0 cells were transfected with an E6 cDNA plasmid. A marked decrease in TIMP-3 expression was caused by promoter hypermethylation via increased DNA (cytosine-5-)-methyltransferase 1 (DNMT1) expression. Mechanistic studies indicated that TIMP-3 loss promoted interleukin-6 (IL-6) production, which led to cell invasion and anchorage-independent growth on soft agar plates. Kaplan-Meier and Cox regression models showed that patients with low-TIMP-3/high-IL-6 tumors had shorter overall survival and relapse-free survival periods when compared with patients with high-TIMP-3/low-IL-6 tumors. In summary, loss of TIMP-3 may increase IL-6 production via the tumor necrosis factor α/nuclear factor κB axis, thereby promoting tumor malignancy and subsequent relapse and poor survival in patients with HPV-infected non-small cell lung cancer.
Collapse
Affiliation(s)
- De-Wei Wu
- Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Kayser G, Schulte-Uentrop L, Sienel W, Werner M, Fisch P, Passlick B, Zur Hausen A, Stremmel C. Stromal CD4/CD25 positive T-cells are a strong and independent prognostic factor in non-small cell lung cancer patients, especially with adenocarcinomas. Lung Cancer 2012; 76:445-51. [PMID: 22300751 DOI: 10.1016/j.lungcan.2012.01.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2011] [Revised: 11/16/2011] [Accepted: 01/07/2012] [Indexed: 12/20/2022]
Abstract
Within the concert of immune reactions against tumour cells cytotoxic and regulatory T-cells are of utmost importance. Several studies revealed contradictory results on this issue. We therefore focused on functional expression patterns and localization of tumour-infiltrating T-lymphocytes in non-small cell lung cancer (NSCLC) and their impact on patient's survival. 232 curatively operated NSCLC patients were included. After histological reevaluation and construction of tissue-multi-arrays immunohistochemical doublestains for CD3/CD8 and CD4/CD25 were performed to evaluate the total number of T-cells and their subsets of cytotoxic and activated T-cells. Additionally, the localization of the lymphocytes was included in the analysis. Hereby, T-cells within the tumour stroma were regarded as stromal, those among cancer cells as intraepithelial. The number of lymphocytes differed significantly between the histological subtypes being most prominent in large cell carcinomas. Survival analysis showed that high numbers of stromal T-lymphocytes are of beneficial prognostic influence in NSCLC patients. This also proved to be an independent prognostic factor in adenocarcinomas. Thus, in a large and well characterized cohort of NSCLC this is the first study to determine the prognostic value of stromal T-lymphocytes, as these are an independent prognosticator in NSCLC especially in adenocarcinomas whereas intraepithelial T-cells are not.
Collapse
Affiliation(s)
- Gian Kayser
- Institute of Pathology, University Hospital Freiburg, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Sung WW, Wang YC, Cheng YW, Lee MC, Yeh KT, Wang L, Wang J, Chen CY, Lee H. A polymorphic -844T/C in FasL promoter predicts survival and relapse in non-small cell lung cancer. Clin Cancer Res 2011; 17:5991-5999. [PMID: 21807637 DOI: 10.1158/1078-0432.ccr-11-0227] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Fas ligand (FasL) -844T/C polymorphism (rs763110) has a demonstrated association with lung cancer risk. FasL -844CC with higher FasL expression has been suggested to contribute to tumor progression via immune escape. However, the impact of FasL -844T/C polymorphism on the clinical outcome of non-small cell lung cancer (NSCLC) remains to be identified. EXPERIMENTAL DESIGN A total of 385 adjacent normal lung tissues from patients with NSCLC were collected to determine FasL -844T/C polymorphism by PCR-based restriction fragment length polymorphism. FasL mRNA and protein expression in lung tumors were evaluated by real-time PCR and immunohistochemistry. The prognostic value of FasL -844T/C polymorphism on survival and relapse was determined by Kaplan-Meier analysis and Cox proportional hazards models. RESULTS The FasL -844CC genotype had higher prevalence in those with advanced tumors than in those with early tumors (P = 0.008). In addition, patients with the FasL -844CC genotype were more prone to tumor relapse than those with the FasL -844TT+TC genotype (62.1% vs. 37.9%, P = 0.001). Multivariate Cox regression analysis showed that patients with the FasL -844CC genotype had poorer survival in terms of overall survival (OS) and relapse-free survival (RFS) than those with the FasL -844TT+TC genotype (24.1 vs. 42.8 months for OS, HR = 1.455, P = 0.004; 15.4 vs. 31.4 months for RFS, HR = 1.710, P < 0.001). CONCLUSIONS FasL -844T/C polymorphism may predict survival and relapse in NSCLC. We suggest that FasL may be a molecular target for immunotherapeutic interventions to improve the clinical outcome of patients with NSCLC. This finding should be validated by another investigative group.
Collapse
Affiliation(s)
- Wen-Wei Sung
- Institute of Medical & Molecular Toxicology, School of Medicine, Institute of Medicine, and School of Public Health, Chung Shan Medical University, Changhua, Taiwan, ROC
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Immunotherapy for lung cancers. J Biomed Biotechnol 2011; 2011:250860. [PMID: 21318107 PMCID: PMC3035001 DOI: 10.1155/2011/250860] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2010] [Revised: 11/15/2010] [Accepted: 12/23/2010] [Indexed: 11/21/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths worldwide. Although treatment methods in surgery, irradiation, and chemotherapy have improved, prognosis remains unsatisfactory and developing new therapeutic strategies is still an urgent demand. Immunotherapy is a novel therapeutic approach wherein activated immune cells can specifically kill tumor cells by recognition of tumor-associated antigens without damage to normal cells. Several lung cancer vaccines have demonstrated prolonged survival time in phase II and phase III trials, and several clinical trials are under investigation. However, many clinical trials involving cancer vaccination with defined tumor antigens work in only a small number of patients. Cancer immunotherapy is not completely effective in eradicating tumor cells because tumor cells escape from host immune scrutiny. Understanding of the mechanism of immune evasion regulated by tumor cells is required for the development of more effective immunotherapeutic approaches against lung cancer. This paper discusses the identification of tumor antigens in lung cancer, tumor immune escape mechanisms, and clinical vaccine trials in lung cancer.
Collapse
|
42
|
Cao Y, Miao XP, Huang MY, Deng L, Lin DX, Zeng YX, Shao JY. Polymorphisms of death pathway genes FAS and FASL and risk of nasopharyngeal carcinoma. Mol Carcinog 2010; 49:944-50. [PMID: 20842669 DOI: 10.1002/mc.20676] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The FAS receptor/ligand system is a key regulator of apoptotic cell death and corruption of this signaling pathway has been shown to participate in carcinogenesis. Functional polymorphisms in the FAS (FAS -1377G/A) and FASL (FASL -844T/C) genes alter their transcriptional activity. Therefore, we examined the association between these polymorphisms and the risk of developing nasopharyngeal carcinoma (NPC). FAS -1377G/A and FASL -844T/C genotypes were determined by PCR-based RFLP analysis in 582 patients with NPC and 613 frequency-matched controls. We observed a significantly increased risk of NPC associated with the FAS -1377AA genotype [odds ratio (OR) = 1.69, 95% confidence interval (CI) = 1.21-2.35] compared with the FAS -1377 GG genotype. In addition, elevated NPC risk was also found among subjects carrying both FAS -1377AA and FASL -844CC genotypes compared with both FAS -1377GG and FASL -844CT or -844TT, the OR was 2.39 (95% CI = 1.50-3.79). After stratification by smoking status, heavy smokers (≥15 pack-years) carrying FAS -1377AA genotype had an increased risk of NPC compared with FAS -1377GG genotype (OR = 3.48, 95% CI = 1.66-7.30). Furthermore, we observed a statistically significant interaction between the two polymorphisms and heavy smoking status (OR = 5.92, 95% CI = 1.91-18.3). Our study provides the first evidence that functional FAS -1377 G/A and FASL -844 T/C polymorphisms are associated with the risk of NPC, and this association is especially noteworthy in tobacco smokers.
Collapse
Affiliation(s)
- Yun Cao
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | | | | | | | | | | | | |
Collapse
|
43
|
Shunmei E, Zhao Y, Huang Y, Lai K, Chen C, Zeng J, Zou J. Heat shock factor 1 is a transcription factor of Fas gene. Mol Cells 2010; 29:527-31. [PMID: 20396959 DOI: 10.1007/s10059-010-0065-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2009] [Revised: 01/28/2010] [Accepted: 01/29/2010] [Indexed: 01/17/2023] Open
Abstract
In mammalian cells, stress-induced expression of heat shock protein is controlled by heat shock factor 1 (HSF1). However, HSF1 functions as a regulator of additional genes. In this study, we observed that heat treatment effectively induced expression of Fas. Using bioinformatics, a high affinity and functional HSF1-binding element within the -1996/-1985 oligonucleotide of the 5'-flanking region of the Fas gene was found, and was determined by electrophoretic mobility shift assay and chromatin immunoprecipitation assay. Exogenous expression of a constitutively activative HSF1, induced Fas gene transcription and protein synthesis in the absence of heat stress. Moreover, RNA interference-mediated HSF1 gene-silencing attenuated Fas expression in a heat-induced model. Our results suggested that HSF1 is an important transcription factor of Fas gene.
Collapse
Affiliation(s)
- E Shunmei
- The Tradition Chinese Hospital of Guangdong Province, Guangzhou, 510120, China
| | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
Small-cell lung carcinomas account for about 15-20% of lung cancer and are characterized by an intrinsic resistance to apoptosis. Increasing evidence suggests that alteration in apoptosis/antiapoptosis balance could lead to fundamental resistance of small-cell lung cancer to chemotherapy and radiation. These molecular alterations include alteration of mitochondrial pathways (BCL2 and BCLXL overexpression, activation of stress protein such as HSP 90 and HSP70, activation of PI3K/AKT/mTOR pathway). Others abnormalities could inhibit activation of extrinsic pathway such as caspase-8 and FAS underexpression as well as C-FLIP overexpression. New therapies targeting some of these abnormalities are under clinical evaluation and predictive factors of response are needed to personalize these therapies.
Collapse
|
45
|
Zhu Q, Wang T, Ren J, Hu K, Liu W, Wu G. FAS-670A/G polymorphism: A biomarker for the metastasis of nasopharyngeal carcinoma in a Chinese population. Clin Chim Acta 2010; 411:179-83. [DOI: 10.1016/j.cca.2009.10.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2009] [Revised: 10/25/2009] [Accepted: 10/29/2009] [Indexed: 01/10/2023]
|
46
|
Lung cancer-associated tumor antigens and the present status of immunotherapy against non-small-cell lung cancer. Gen Thorac Cardiovasc Surg 2009; 57:449-57. [DOI: 10.1007/s11748-008-0433-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2008] [Indexed: 01/25/2023]
|
47
|
Abstract
Lung cancer is the major cancer killer worldwide, and 5-yr survival is extremely poor (
Collapse
Affiliation(s)
- E Brambilla
- Dept of Pathology, Institut Albert Bonniot, INSERM U823, University Joseph Fourier, CHRU Grenoble Hôpital Michallon, Grenoble, France.
| | | |
Collapse
|
48
|
Park JY, Lee WK, Jung DK, Choi JE, Park TI, Lee EB, Cho S, Park JY, Cha SI, Kim CH, Kam S, Jung TH, Jheon S. Polymorphisms in theFASandFASLGenes and Survival of Early Stage Non–small Cell Lung Cancer. Clin Cancer Res 2009; 15:1794-800. [DOI: 10.1158/1078-0432.ccr-08-1770] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
49
|
Ramakrishnan R, Antonia S, Gabrilovich DI. Combined modality immunotherapy and chemotherapy: a new perspective. Cancer Immunol Immunother 2008; 57:1523-9. [PMID: 18488219 PMCID: PMC11030293 DOI: 10.1007/s00262-008-0531-4] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2008] [Accepted: 04/26/2008] [Indexed: 12/19/2022]
Abstract
The results of recent clinical trials have demonstrated that cancer vaccines continue to struggle to achieve tangible clinical benefits as monotherapy. Tumor-induced abnormalities in the immune system hamper anti-tumor T cell responses limiting the effectiveness of cancer immunotherapy. Recently, evidence has been mounting to suggest that immunotherapy has the possibility of achieving better success when used in combination with conventional chemotherapy. In clinical trials, immune responses elicited by cancer vaccines appear to augment the effectiveness of subsequent conventional cancer therapies.
Collapse
Affiliation(s)
- Rupal Ramakrishnan
- H. Lee Moffitt Cancer Center and Research Institute, MRC 2067, 12902 Magnolia Dr., Tampa, FL 33612 USA
| | - Scott Antonia
- H. Lee Moffitt Cancer Center and Research Institute, MRC 2067, 12902 Magnolia Dr., Tampa, FL 33612 USA
| | - Dmitry I. Gabrilovich
- H. Lee Moffitt Cancer Center and Research Institute, MRC 2067, 12902 Magnolia Dr., Tampa, FL 33612 USA
| |
Collapse
|
50
|
Prado-Garcia H, Aguilar-Cazares D, Meneses-Flores M, Morales-Fuentes J, Lopez-Gonzalez JS. Lung carcinomas do not induce T-cell apoptosis via the Fas/Fas ligand pathway but down-regulate CD3 epsilon expression. Cancer Immunol Immunother 2008; 57:325-36. [PMID: 17668204 PMCID: PMC11030893 DOI: 10.1007/s00262-007-0372-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2007] [Accepted: 07/11/2007] [Indexed: 10/23/2022]
Abstract
BACKGROUND Non-small cell lung carcinoma (NSCLC) patients have impaired cellular immune responses. It has been hypothesized that tumor cells expressing Fas Ligand (FasL) induce in T lymphocytes: (a) apoptosis (tumor counterattack) and (b) down-regulation of CD3zeta expression. However, the hypothesis of tumor counterattack is still controversial. METHODS We analyzed FasL expression on NSCLC cell lines and on tumor cells from lung adenocarcinoma patients by flow cytometry and immunocytochemistry. FasL mRNA expression was detected in NSCLC cell lines using RT-PCR, and functional FasL was evaluated on Fas-expressing Jurkat T-cells by annexin-V-FITC staining and by SubG(1) peak detection. Also, the proapoptotic effect of microvesicles released from NSCLC cell lines in Jurkat T-cells was studied. Alterations in the expression levels of CD3zeta, CD3epsilon, and CD28 [measured as mean fluorescence intensity (MFI)] were determined in Jurkat T-cells after co-culture with NSCLC cell lines or tumor-derived microvesicles. Furthermore, the expression levels of CD3zeta and CD3epsilon in CD4+T and CD8+T lymphocytes from lung adenocarcinoma patients was studied. RESULTS Our results indicate that NSCLC cells neither FasL expressed nor induced apoptosis in Jurkat T-cells. Tumor-derived microvesicles did not induce apoptosis in Jurkat T-cells. In contrast, NSCLC cell lines down-regulated CD3epsilon but not CD3zeta chain expression in Jurkat T-cells; this effect was induced by soluble factors but not by microvesicles. In lung adenocarcinoma patients, significant decreases of MFI values for CD3epsilon, but not CD3zeta, were found in CD4+T and CD8+T cells from pleural effusion compared to peripheral blood and in peripheral blood of patients compared to healthy donors. CONCLUSIONS Our data do not support the tumor counterattack hypothesis for NSCLC. Nonetheless, down-regulation of CD3epsilon in T-cells induced by NSCLC cells might lead to T-cell dysfunction.
Collapse
Affiliation(s)
- Heriberto Prado-Garcia
- Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Tlalpan 4502, Col. Seccion XVI, CP 14080 Mexico City, Mexico
| | - Dolores Aguilar-Cazares
- Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Tlalpan 4502, Col. Seccion XVI, CP 14080 Mexico City, Mexico
| | - Manuel Meneses-Flores
- Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Tlalpan 4502, Col. Seccion XVI, CP 14080 Mexico City, Mexico
| | - Jorge Morales-Fuentes
- Servicio Clinico 3, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Tlalpan 4502, Col. Seccion XVI, CP 14080 Mexico City, Mexico
| | - Jose Sullivan Lopez-Gonzalez
- Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Tlalpan 4502, Col. Seccion XVI, CP 14080 Mexico City, Mexico
| |
Collapse
|