1
|
Qiu C, Sun N, Zeng S, Chen L, Gong F, Tian J, Xiong Y, Peng L, He H, Ming Y. Unveiling the therapeutic promise of EphA2 in glioblastoma: a comprehensive review. Discov Oncol 2024; 15:501. [PMID: 39331302 PMCID: PMC11436538 DOI: 10.1007/s12672-024-01380-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 09/20/2024] [Indexed: 09/28/2024] Open
Abstract
Glioblastoma (GBM), a primary brain tumor, exhibits remarkable invasiveness and is characterized by its intricate location, infiltrative behavior, the presence of both the blood-brain barrier (BBB) and the blood-brain tumor barrier (BBTB), phenotypic diversity, an immunosuppressive microenvironment with limited development yet rich vascularity, as well as the resistant nature of glioblastoma stem cells (GSCs) towards traditional chemotherapy and radiotherapy. These formidable factors present substantial obstacles in the quest for effective GBM treatments. Following extensive research spanning three decades, the hepatocellular receptor A2 (EphA2) receptor tyrosine kinase has emerged as a promising molecular target with translational potential in the realm of cancer therapy. Numerous compounds aimed at targeting EphA2 have undergone rigorous evaluation and clinical investigation. This article provides a comprehensive account of the distinctive roles played by canonical and non-canonical EphA2 signaling in various contexts, while also exploring the involvement of the EphA2-ephrin A1 signaling axis in GBM pathogenesis. Additionally, the review offers an overview of completed clinical trials targeting EphA2 for GBM treatment, shedding light on both the prospects and challenges associated with EphA2-directed interventions in the domain of cancer therapeutics.
Collapse
Affiliation(s)
- Caohang Qiu
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
- Sichuan Clinical Research Center of Neurosurgery, Luzhou, 646000, People's Republic of China
- Academician (Expert) Workstation of Sichuan Province, Luzhou, 646000, People's Republic of China
- Neurological Diseases and Brain Function Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
| | - Ning Sun
- Department of Pediatric Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Shan Zeng
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
- Sichuan Clinical Research Center of Neurosurgery, Luzhou, 646000, People's Republic of China
- Academician (Expert) Workstation of Sichuan Province, Luzhou, 646000, People's Republic of China
- Neurological Diseases and Brain Function Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
| | - Ligang Chen
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
- Sichuan Clinical Research Center of Neurosurgery, Luzhou, 646000, People's Republic of China
- Academician (Expert) Workstation of Sichuan Province, Luzhou, 646000, People's Republic of China
- Neurological Diseases and Brain Function Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
| | - Feilong Gong
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
- Sichuan Clinical Research Center of Neurosurgery, Luzhou, 646000, People's Republic of China
- Academician (Expert) Workstation of Sichuan Province, Luzhou, 646000, People's Republic of China
- Neurological Diseases and Brain Function Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
| | - Junjie Tian
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
- Sichuan Clinical Research Center of Neurosurgery, Luzhou, 646000, People's Republic of China
- Academician (Expert) Workstation of Sichuan Province, Luzhou, 646000, People's Republic of China
- Neurological Diseases and Brain Function Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
| | - Yu Xiong
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
- Sichuan Clinical Research Center of Neurosurgery, Luzhou, 646000, People's Republic of China
- Academician (Expert) Workstation of Sichuan Province, Luzhou, 646000, People's Republic of China
- Neurological Diseases and Brain Function Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
| | - Lilei Peng
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
- Sichuan Clinical Research Center of Neurosurgery, Luzhou, 646000, People's Republic of China
- Academician (Expert) Workstation of Sichuan Province, Luzhou, 646000, People's Republic of China
- Neurological Diseases and Brain Function Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
| | - Haiping He
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
- Sichuan Clinical Research Center of Neurosurgery, Luzhou, 646000, People's Republic of China
- Academician (Expert) Workstation of Sichuan Province, Luzhou, 646000, People's Republic of China
- Neurological Diseases and Brain Function Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
| | - Yang Ming
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China.
- Sichuan Clinical Research Center of Neurosurgery, Luzhou, 646000, People's Republic of China.
- Academician (Expert) Workstation of Sichuan Province, Luzhou, 646000, People's Republic of China.
- Neurological Diseases and Brain Function Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China.
| |
Collapse
|
2
|
Absent in melanoma 2 regulates tumor cell proliferation in glioblastoma multiforme. J Neurooncol 2019; 144:265-273. [DOI: 10.1007/s11060-019-03230-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 06/24/2019] [Indexed: 10/26/2022]
|
3
|
Vo TM, Jain S, Burchett R, Monckton EA, Godbout R. A positive feedback loop involving nuclear factor IB and calpain 1 suppresses glioblastoma cell migration. J Biol Chem 2019; 294:12638-12654. [PMID: 31262726 DOI: 10.1074/jbc.ra119.008291] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 06/27/2019] [Indexed: 12/15/2022] Open
Abstract
Glioblastoma (GBM) is a brain tumor that remains largely incurable because of its highly-infiltrative properties. Nuclear factor I (NFI)-type transcription factors regulate genes associated with GBM cell migration and infiltration. We have previously shown that NFI activity depends on the NFI phosphorylation state and that calcineurin phosphatase dephosphorylates and activates NFI. Calcineurin is cleaved and activated by calpain proteases whose activity is, in turn, regulated by an endogenous inhibitor, calpastatin (CAST). The CAST gene is a target of NFI in GBM cells, with differentially phosphorylated NFIs regulating the levels of CAST transcript variants. Here, we uncovered an NFIB-calpain 1-positive feedback loop mediated through CAST and calcineurin. In NFI-hyperphosphorylated GBM cells, NFIB expression decreased the CAST-to-calpain 1 ratio in the cytoplasm. This reduced ratio increased autolysis and activity of cytoplasmic calpain 1. Conversely, in NFI-hypophosphorylated cells, NFIB expression induced differential subcellular compartmentalization of CAST and calpain 1, with CAST localizing primarily to the cytoplasm and calpain 1 to the nucleus. Overall, this altered compartmentalization increased nuclear calpain 1 activity. We also show that nuclear calpain 1, by cleaving and activating calcineurin, induces NFIB dephosphorylation. Of note, knockdown of calpain 1, NFIB, or both increased GBM cell migration and up-regulated the pro-migratory factors fatty acid-binding protein 7 (FABP7) and Ras homolog family member A (RHOA). In summary, our findings reveal bidirectional cross-talk between NFIB and calpain 1 in GBM cells. A physiological consequence of this positive feedback loop appears to be decreased GBM cell migration.
Collapse
Affiliation(s)
- The Minh Vo
- Cross Cancer Institute, Department of Oncology, University of Alberta, Edmonton, Alberta T6G 1Z2, Canada
| | - Saket Jain
- Cross Cancer Institute, Department of Oncology, University of Alberta, Edmonton, Alberta T6G 1Z2, Canada
| | - Rebecca Burchett
- Cross Cancer Institute, Department of Oncology, University of Alberta, Edmonton, Alberta T6G 1Z2, Canada
| | - Elizabeth A Monckton
- Cross Cancer Institute, Department of Oncology, University of Alberta, Edmonton, Alberta T6G 1Z2, Canada
| | - Roseline Godbout
- Cross Cancer Institute, Department of Oncology, University of Alberta, Edmonton, Alberta T6G 1Z2, Canada
| |
Collapse
|
4
|
Dettling S, Stamova S, Warta R, Schnölzer M, Rapp C, Rathinasamy A, Reuss D, Pocha K, Roesch S, Jungk C, Warnken U, Eckstein V, Grabe N, Schramm C, Weigand MA, von Deimling A, Unterberg A, Beckhove P, Herold-Mende C. Identification of CRKII, CFL1, CNTN1, NME2, and TKT as Novel and Frequent T-Cell Targets in Human IDH-Mutant Glioma. Clin Cancer Res 2018; 24:2951-2962. [PMID: 29563135 DOI: 10.1158/1078-0432.ccr-17-1839] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 11/23/2017] [Accepted: 03/15/2018] [Indexed: 11/16/2022]
Abstract
Purpose: Successful immunotherapies for IDHmut gliomas require better knowledge of T-cell target antigens. Here, we elucidated their antigen repertoire recognized by spontaneous T-cell responses using an unbiased proteomic approach.Experimental Design: Protein fractionations of tissue lysates from IDHmut gliomas (n = 4) were performed. Fractions were tested by IFNγ ELISpot assay for recognition through patients' T cells. Proteins of immunogenic fractions were identified by mass spectrometry and validated by in silico-predicted synthetic long peptides in patients of origin, additional IDHmut glioma patients (n = 16), and healthy donors (n = 13). mRNA and protein expression of immunogenic antigens was analyzed in tumor tissues and IDHmut glioma stem-like cells (GSC). HLA-A*02-restricted T-cell epitopes were functionally determined by short peptides and numbers of antigen-specific T cells by HLA-peptide tetramer analysis.Results: A total of 2,897 proteins were identified in immunogenic tumor fractions. Based on a thorough filter process, 79 proteins were selected as potential T-cell antigens. Twenty-six of these were recognized by the patients' T cells, and five of them (CRKII, CFL1, CNTN1, NME2, and TKT) in up to 56% unrelated IDHmut glioma patients. Most immunogenic tumor-associated antigens (TAA) were expressed in IDHmut gliomas and GSCs, while being almost absent in normal brain tissues. Finally, we identified HLA-A*02-restricted epitopes for CRKII, NME2, and TKT that were recognized by up to 2.82% of antigen-specific peripheral cytotoxic T cells in IDHmut glioma patients.Conclusions: By analyzing the repertoire of T-cell target antigens in IDHmut glioma patients, we identified five novel immunogenic TAAs and confirmed their expression on IDHmut tumors and GSCs. Clin Cancer Res; 24(12); 2951-62. ©2018 AACR.
Collapse
Affiliation(s)
- Steffen Dettling
- Division of Experimental Neurosurgery, Department of Neurosurgery, University Hospital of Heidelberg, Heidelberg, Germany
| | - Slava Stamova
- Regensburg Center for Interventional Immunology (RCI) and University Medical Center of Regensburg, Regensburg, Germany
| | - Rolf Warta
- Division of Experimental Neurosurgery, Department of Neurosurgery, University Hospital of Heidelberg, Heidelberg, Germany
| | - Martina Schnölzer
- Functional Proteome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Carmen Rapp
- Division of Experimental Neurosurgery, Department of Neurosurgery, University Hospital of Heidelberg, Heidelberg, Germany
| | - Anchana Rathinasamy
- Regensburg Center for Interventional Immunology (RCI) and University Medical Center of Regensburg, Regensburg, Germany
| | - David Reuss
- Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany.,German Cancer Consortium (DKTK), CCU Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kolja Pocha
- Division of Experimental Neurosurgery, Department of Neurosurgery, University Hospital of Heidelberg, Heidelberg, Germany
| | - Saskia Roesch
- Division of Experimental Neurosurgery, Department of Neurosurgery, University Hospital of Heidelberg, Heidelberg, Germany
| | - Christine Jungk
- Division of Experimental Neurosurgery, Department of Neurosurgery, University Hospital of Heidelberg, Heidelberg, Germany
| | - Uwe Warnken
- Functional Proteome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Volker Eckstein
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Niels Grabe
- Hamamatsu Tissue Imaging and Analysis Center (TIGA), BIOQUANT, University of Heidelberg, Heidelberg, Germany
| | - Christoph Schramm
- Department of Anesthesiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Markus A Weigand
- Department of Anesthesiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Andreas von Deimling
- Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany.,German Cancer Consortium (DKTK), CCU Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Andreas Unterberg
- Division of Experimental Neurosurgery, Department of Neurosurgery, University Hospital of Heidelberg, Heidelberg, Germany
| | - Philipp Beckhove
- Regensburg Center for Interventional Immunology (RCI) and University Medical Center of Regensburg, Regensburg, Germany
| | - Christel Herold-Mende
- Division of Experimental Neurosurgery, Department of Neurosurgery, University Hospital of Heidelberg, Heidelberg, Germany.
| |
Collapse
|
5
|
Chimeric antigen receptor T-cell therapy for glioblastoma. Transl Res 2017; 187:93-102. [PMID: 28755873 DOI: 10.1016/j.trsl.2017.07.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 06/25/2017] [Accepted: 07/11/2017] [Indexed: 02/06/2023]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has shown great promise in the treatment of hematological disease, and its utility for treatment of solid tumors is beginning to unfold. Glioblastoma continues to portend a grim prognosis and immunotherapeutic approaches are being explored as a potential treatment strategy. Identification of appropriate glioma-associated antigens, barriers to cell delivery, and presence of an immunosuppressive microenvironment are factors that make CAR T-cell therapy for glioblastoma particularly challenging. However, insights gained from preclinical studies and ongoing clinical trials indicate that CAR T-cell therapy will continue to evolve and likely become integrated with current therapeutic strategies for malignant glioma.
Collapse
|
6
|
Wang L, Shen YF, Shi ZM, Shang XJ, Jin DL, Xi F. Overexpression miR-211-5p hinders the proliferation, migration, and invasion of thyroid tumor cells by downregulating SOX11. J Clin Lab Anal 2017; 32. [PMID: 28703321 DOI: 10.1002/jcla.22293] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Accepted: 06/08/2017] [Indexed: 12/25/2022] Open
Abstract
PURPOSE This study was aimed to investigate the relationship between miR-211-5p and SOX11, and the effects of their interaction on the proliferation, viability, and invasion of human thyroid cancer (TC) cells. METHODS We used quantitative real-time PCR (qRT-PCR) to determine the expression of miR-211-5p and SOX11mRNA in the thyroid tumorous and the adjacent tissues. The target relationship between miR-211-5p and SOX11 was confirmed using dual luciferase reporter gene assay. Flow cytometry, colony formation assay, Transwell assay, and MTT assay were performed to determine the cell-cycle progression, cell apoptosis, proliferation and invasion, respectively. In addition, the tumor formation assay in nude mice was done to assess the effect of miR-211-5p on TC development in vivo. RESULTS MiR-211-5p was underexpressed, whereas SOX11 was overexpressed in TC. The overexpression of miR-211-5p inhibited the expression of SOX11. The cell cycle was arrested and the proliferation as well as invasiveness was suppressed by exogenous miR-211-5p in TC cell line. The antitumor role of miR-211-5p was proved by the animal experiment. CONCLUSION MiR-211-5p affected the viability, proliferation and invasion of TC by negatively regulating SOX11 expression.
Collapse
Affiliation(s)
- Lei Wang
- Department of Pathology, Medical College of Hebei University of Engineering, Handan, Hebei Province, China
| | - Yan-Feng Shen
- Department of Oncology, Affiliated Hospital of Hebei University of Engineering, Handan, Hebei Province, China
| | - Zhi-Min Shi
- Department of Pathology, Medical College of Hebei University of Engineering, Handan, Hebei Province, China
| | - Xiao-Juan Shang
- Microscope Room, Medicine College of Hebei University of Engineering, Handan, Hebei Province, China
| | - Dong-Ling Jin
- Department of Pathology, Medical College of Hebei University of Engineering, Handan, Hebei Province, China
| | - Feng Xi
- Department of Pathology, Medical College of Hebei University of Engineering, Handan, Hebei Province, China
| |
Collapse
|
7
|
Long X, Hu H, Li S, Chen M, Cai J, Song B. Hippocampal YKL-40 expression in rats after status epilepticus. Epilepsy Res 2016; 125:52-7. [DOI: 10.1016/j.eplepsyres.2016.05.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 04/27/2016] [Accepted: 05/30/2016] [Indexed: 10/21/2022]
|
8
|
Ge LS, Hoa NT, Lambrecht N, Dacosta-Iyer M, Ouyang Y, Abolhoda A, Jadus MR. Changes in tumor-antigen expression profile as human small-cell lung cancers progress. Cancer Biol Med 2015; 12:96-105. [PMID: 26175925 PMCID: PMC4493377 DOI: 10.7497/j.issn.2095-3941.2015.0027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 05/13/2015] [Indexed: 12/03/2022] Open
Abstract
Objective Our group has previously observed that in patients with small-cell lung cancers (SCLCs), the expression of a tumor antigen, glioma big potassium (gBK) ion channel, is higher at the time of death than when the cancer is first treated by surgical resection. This study aimed to determine whether this dichotomy was common in other potential lung tumor antigens by examining the same patient samples using our more extensive profile analysis of tumor-antigen precursor protein (TAPP). We then tested the hypothesis that therapeutic intervention may inadvertently cause this increased gBK production. Methods SCLC samples (eight surgical resections and three autopsy samples) and three control lungs were examined by quantitative real-time polymerase chain reaction for 42 potential TAPPs that represent potential T-cell-mediated immunological targets. Results Twenty-two TAPP mRNAs displayed the same profile as gBK, i.e., more mRNAs were expressed at autopsy than in their surgical counterparts. B-cyclin and mouse double minute 2, human homolog of P53-binding protein were elevated in both autopsy and surgical specimens above the normal-lung controls. When HTB119 cells were incubated with doxorubicin, gBK was strongly induced, as confirmed by intracellular flow cytometry with a gBK-specific antibody. Conclusion Our findings suggested that more immunological targets became available as the tumor responded to chemotherapy and proceeded toward its terminal stages.
Collapse
Affiliation(s)
- Li-Sheng Ge
- 1 Research Service, 2 Pathology and Laboratory Medicine Service, VA Long Beach Healthcare System, Long Beach, CA 90822, USA ; 3 Pathology and Laboratory Medicine, University of California, Irvine, CA 92697, USA ; 4 Surgical Health Care Group, Veterans Affairs Medical Center, Long Beach, CA 90822, USA ; 5 Chao Family Comprehensive Cancer Center, UC, Irvine School of Medicine, University of California, Irvine, Orange, CA 92868, USA
| | - Neil T Hoa
- 1 Research Service, 2 Pathology and Laboratory Medicine Service, VA Long Beach Healthcare System, Long Beach, CA 90822, USA ; 3 Pathology and Laboratory Medicine, University of California, Irvine, CA 92697, USA ; 4 Surgical Health Care Group, Veterans Affairs Medical Center, Long Beach, CA 90822, USA ; 5 Chao Family Comprehensive Cancer Center, UC, Irvine School of Medicine, University of California, Irvine, Orange, CA 92868, USA
| | - Nils Lambrecht
- 1 Research Service, 2 Pathology and Laboratory Medicine Service, VA Long Beach Healthcare System, Long Beach, CA 90822, USA ; 3 Pathology and Laboratory Medicine, University of California, Irvine, CA 92697, USA ; 4 Surgical Health Care Group, Veterans Affairs Medical Center, Long Beach, CA 90822, USA ; 5 Chao Family Comprehensive Cancer Center, UC, Irvine School of Medicine, University of California, Irvine, Orange, CA 92868, USA
| | - Maria Dacosta-Iyer
- 1 Research Service, 2 Pathology and Laboratory Medicine Service, VA Long Beach Healthcare System, Long Beach, CA 90822, USA ; 3 Pathology and Laboratory Medicine, University of California, Irvine, CA 92697, USA ; 4 Surgical Health Care Group, Veterans Affairs Medical Center, Long Beach, CA 90822, USA ; 5 Chao Family Comprehensive Cancer Center, UC, Irvine School of Medicine, University of California, Irvine, Orange, CA 92868, USA
| | - Yi Ouyang
- 1 Research Service, 2 Pathology and Laboratory Medicine Service, VA Long Beach Healthcare System, Long Beach, CA 90822, USA ; 3 Pathology and Laboratory Medicine, University of California, Irvine, CA 92697, USA ; 4 Surgical Health Care Group, Veterans Affairs Medical Center, Long Beach, CA 90822, USA ; 5 Chao Family Comprehensive Cancer Center, UC, Irvine School of Medicine, University of California, Irvine, Orange, CA 92868, USA
| | - Amir Abolhoda
- 1 Research Service, 2 Pathology and Laboratory Medicine Service, VA Long Beach Healthcare System, Long Beach, CA 90822, USA ; 3 Pathology and Laboratory Medicine, University of California, Irvine, CA 92697, USA ; 4 Surgical Health Care Group, Veterans Affairs Medical Center, Long Beach, CA 90822, USA ; 5 Chao Family Comprehensive Cancer Center, UC, Irvine School of Medicine, University of California, Irvine, Orange, CA 92868, USA
| | - Martin R Jadus
- 1 Research Service, 2 Pathology and Laboratory Medicine Service, VA Long Beach Healthcare System, Long Beach, CA 90822, USA ; 3 Pathology and Laboratory Medicine, University of California, Irvine, CA 92697, USA ; 4 Surgical Health Care Group, Veterans Affairs Medical Center, Long Beach, CA 90822, USA ; 5 Chao Family Comprehensive Cancer Center, UC, Irvine School of Medicine, University of California, Irvine, Orange, CA 92868, USA
| |
Collapse
|
9
|
Aref AM, Hoa NT, Ge L, Agrawal A, Dacosta-Iyer M, Lambrecht N, Ouyang Y, Cornforth AN, Jadus MR. HCA519/TPX2: a potential T-cell tumor-associated antigen for human hepatocellular carcinoma. Onco Targets Ther 2014; 7:1061-70. [PMID: 24966688 PMCID: PMC4063820 DOI: 10.2147/ott.s61442] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Background Immunotherapy for human hepatocellular cancer (HCC) is slowly making progress towards treating these fatal cancers. The identification of new antigens can improve this approach. We describe a possible new antigen, hepatocellular carcinoma‐associated antigen‐519/targeting protein for Xklp‐2 (HCA519/TPX2), for HCC that might be beneficial for T‐cell specific HCC immunotherapy. Methods HCC was studied for the expression for 15 tumor‐associated antigens considered useful for immunotherapy within three HCC cell lines (HepG2, Hep3B, and PLC/PRF/5), lymphocytes, non‐cancerous livers, and clinical HCC. The expression of tumor antigenic precursor proteins (TAPPs) messenger RNA was first screened by reverse transcriptase quantitative real‐time polymerase chain reaction. Results Four antigens (alpha fetoprotein, aspartyl/asparaginyl βhydroxylase, glypican3 and HCA519/TPX2) proved to be the best expressed TAPPs within the HCC specimens by molecular analyses. HCA519/TPX2 was detected by intracellular cell flow cytometry within HCC cell lines by using a specific antibody towards this TAPP. This antibody also detected the protein within primary HCCs. We synthesized two HCA519/TPX2 peptides (HCA519464–472 and HCA519351–359) which can bind to human leukocyte antigen (HLA)‐A*0201. Dendritic cells pulsed with these peptides stimulated cytolytic T lymphocytes (CTLs). These killer T‐cells lysed HLA‐A*0201+ T2 cells exogenously loaded with the correct specific peptide. The CTLs killed HepG2 (HLA‐A2+ and HCA519+), but not the Hep3B and PLC/PRF/5 cell lines, which are HCA519+ but HLA‐A2‐negative. In silico analysis reveals that HCA519/TPX2 has the inherent ability to bind to a very wide variety of HLA antigens. Conclusion HCA519/TPX2 is a viable immunotarget that should be further investigated within HCC patients.
Collapse
Affiliation(s)
- Ahmed M Aref
- Biological Science Department, Modern Sciences and Arts University, Faculty of Dentistry, Cairo, Egypt ; Southern California Institute for Research and Education, Veterans Affairs Medical Center, Long Beach, CA, USA ; Research Health Care Group, Veterans Affairs Medical Center Long Beach, CA, USA
| | - Neil T Hoa
- Research Health Care Group, Veterans Affairs Medical Center Long Beach, CA, USA
| | - Lisheng Ge
- Research Health Care Group, Veterans Affairs Medical Center Long Beach, CA, USA
| | - Anshu Agrawal
- Department of Medicine, Division of Basic and Clinical Immunology, University of California, Irvine, CA, USA
| | - Maria Dacosta-Iyer
- Pathology and Laboratory Medicine Department, Veterans Affairs Medical Center Long Beach, CA, USA ; Department of Pathology and Laboratory Medicine, University of California, Irvine, CA, USA
| | - Nils Lambrecht
- Pathology and Laboratory Medicine Department, Veterans Affairs Medical Center Long Beach, CA, USA ; Department of Pathology and Laboratory Medicine, University of California, Irvine, CA, USA
| | - Yi Ouyang
- Pathology and Laboratory Medicine Department, Veterans Affairs Medical Center Long Beach, CA, USA ; Department of Pathology and Laboratory Medicine, University of California, Irvine, CA, USA
| | | | - Martin R Jadus
- Pathology and Laboratory Medicine Department, Veterans Affairs Medical Center Long Beach, CA, USA ; Department of Pathology and Laboratory Medicine, University of California, Irvine, CA, USA ; Neuro-Oncology Program, Chao Comprehensive Cancer Center, University of California, Irvine, CA, USA
| |
Collapse
|
10
|
Hoa NT, Ge L, Tajhya RB, Beeton C, Cornforth AN, Abolhoda A, Lambrecht N, DaCosta-Iyer M, Ouyang Y, Mai AP, Hong E, Shon J, Hickey MJ, Erickson KL, Kruse CA, Jadus MR. Small cell lung cancer cells express the late stage gBK tumor antigen: a possible immunotarget for the terminal disease. Am J Transl Res 2014; 6:188-205. [PMID: 24936214 PMCID: PMC4058303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 03/24/2014] [Indexed: 06/03/2023]
Abstract
Big Potassium (BK) ion channels have several splice variants. One splice variant initially described within human glioma cells is called the glioma BK channel (gBK). Using a gBK-specific antibody, we detected gBK within three human small cell lung cancer (SCLC) lines. Electrophysiology revealed that functional membrane channels were found on the SCLC cells. Prolonged exposure to BK channel activators caused the SCLC cells to swell within 20 minutes and resulted in their death within five hours. Transduction of BK-negative HEK cells with gBK produced functional gBK channels. Quantitative RT-PCR analysis using primers specific for gBK, but not with a lung-specific marker, Sox11, confirmed that advanced, late-stage human SCLC tissues strongly expressed gBK mRNA. Normal human lung tissue and early, lower stage SCLC resected tissues very weakly expressed this transcript. Immunofluorescence using the anti-gBK antibody confirmed that SCLC cells taken at the time of the autopsy intensely displayed this protein. gBK may represent a late-stage marker for SCLC. HLA-A*0201 restricted human CTL were generated in vitro using gBK peptide pulsed dendritic cells. The exposure of SCLC cells to interferon-γ (IFN-γ) increased the expression of HLA; these treated cells were killed by the CTL better than non-IFN-γ treated cells even though the IFN-γ treated SCLC cells displayed diminished gBK protein expression. Prolonged incubation with recombinant IFN-γ slowed the in vitro growth and prevented transmigration of the SCLC cells, suggesting IFN-γ might inhibit tumor growth in vivo. Immunotherapy targeting gBK might impede advancement to the terminal stage of SCLC via two pathways.
Collapse
Affiliation(s)
- Neil T Hoa
- Research Health Care Group, Veterans Affairs Medical CenterLong Beach, CA 90822, USA
| | - Lisheng Ge
- Research Health Care Group, Veterans Affairs Medical CenterLong Beach, CA 90822, USA
| | - Rajeev B Tajhya
- Department of Molecular Physiology and Biophysics, Baylor College of MedicineHouston, Texas 77030, USA
| | - Christine Beeton
- Department of Molecular Physiology and Biophysics, Baylor College of MedicineHouston, Texas 77030, USA
| | | | - Amir Abolhoda
- Surgical Health Care Group, Veterans Affairs Medical CenterLong Beach, CA 90822, USA
| | - Nils Lambrecht
- Pathology and Laboratory Medicine Service, Veterans Affairs Medical CenterLong Beach, CA 90822, USA
- Department of Pathology and Laboratory Medicine, University of CaliforniaIrvine, Orange, CA 92868, USA
| | - Maria DaCosta-Iyer
- Pathology and Laboratory Medicine Service, Veterans Affairs Medical CenterLong Beach, CA 90822, USA
- Department of Pathology and Laboratory Medicine, University of CaliforniaIrvine, Orange, CA 92868, USA
| | - Yi Ouyang
- Pathology and Laboratory Medicine Service, Veterans Affairs Medical CenterLong Beach, CA 90822, USA
- Department of Pathology and Laboratory Medicine, University of CaliforniaIrvine, Orange, CA 92868, USA
| | - Anthony P Mai
- Research Health Care Group, Veterans Affairs Medical CenterLong Beach, CA 90822, USA
| | - Erin Hong
- Research Health Care Group, Veterans Affairs Medical CenterLong Beach, CA 90822, USA
| | - Judy Shon
- Research Health Care Group, Veterans Affairs Medical CenterLong Beach, CA 90822, USA
| | - Michelle J Hickey
- Department of Neurosurgery, David Geffen School of Medicine, University of CaliforniaLos Angeles, Los Angeles, CA 90095, USA
| | - Kate L Erickson
- Department of Neurosurgery, David Geffen School of Medicine, University of CaliforniaLos Angeles, Los Angeles, CA 90095, USA
| | - Carol A Kruse
- Department of Neurosurgery, David Geffen School of Medicine, University of CaliforniaLos Angeles, Los Angeles, CA 90095, USA
| | - Martin R Jadus
- Research Health Care Group, Veterans Affairs Medical CenterLong Beach, CA 90822, USA
- Pathology and Laboratory Medicine Service, Veterans Affairs Medical CenterLong Beach, CA 90822, USA
- Department of Pathology and Laboratory Medicine, University of CaliforniaIrvine, Orange, CA 92868, USA
- Neuro-Oncology Program, Chao Comprehensive Cancer Center, University of CaliforniaIrvine, Orange, CA 92868, USA
| |
Collapse
|
11
|
Farkas SA, Milutin-Gašperov N, Grce M, Nilsson TK. Genome-wide DNA methylation assay reveals novel candidate biomarker genes in cervical cancer. Epigenetics 2013; 8:1213-25. [PMID: 24030264 DOI: 10.4161/epi.26346] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The oncogenic human papilloma viruses (HPVs) are associated with precancerous cervical lesions and development of cervical cancer. The DNA methylation signatures of the host genome in normal, precancerous and cervical cancer tissue may indicate tissue-specific perturbation in carcinogenesis. The aim of this study was to identify new candidate genes that are differentially methylated in squamous cell carcinoma compared with DNA samples from cervical intraepithelial neoplasia grade 3 (CIN3) and normal cervical scrapes. The Illumina Infinium HumanMethylation450 BeadChip method identifies genome-wide DNA methylation changes in CpG islands, CpG shores and shelves. Our findings showed an extensive differential methylation signature in cervical cancer compared with the CIN3 or normal cervical tissues. The identified candidate biomarker genes for cervical cancer represent several types of mechanisms in the cellular machinery that are epigenetically deregulated by hypermethylation, such as membrane receptors, intracellular signaling and gene transcription. The results also confirm extensive hypomethylation of genes in the immune system in cancer tissues. These insights into the functional role of DNA methylome alterations in cervical cancer could be clinically applicable in diagnostics and prognostics, and may guide the development of new epigenetic therapies.
Collapse
Affiliation(s)
- Sanja A Farkas
- Department of Laboratory Medicine; Örebro University Hospital; Örebro, Sweden
| | | | - Magdalena Grce
- Department of Molecular Medicine; Rudjer Boskovic Institute; Zagreb, Croatia
| | - Torbjörn K Nilsson
- Department of Laboratory Medicine; Örebro University Hospital; Örebro, Sweden; School of Health and Medical Sciences; Örebro University; Örebro, Sweden
| |
Collapse
|
12
|
Ge L, Hoa NT, Cornforth AN, Bota DA, Mai A, Kim DI, Chiou SK, Hickey MJ, Kruse CA, Jadus MR. Glioma big potassium channel expression in human cancers and possible T cell epitopes for their immunotherapy. THE JOURNAL OF IMMUNOLOGY 2012; 189:2625-34. [PMID: 22844111 DOI: 10.4049/jimmunol.1102965] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Big potassium (BK) ion channels have several spliced variants. One spliced variant initially described within human glioma cells is the glioma BK (gBK) channel. This isoform consists of 34 aa inserted into the intracellular region of the basic BK ion channel. PCR primers specific for this inserted region confirmed that human glioma cell lines and freshly resected surgical tissues from glioblastoma multiforme patients strongly expressed gBK mRNA. Normal human brain tissue very weakly expressed this transcript. An Ab specific for this gBK isoform confirmed that human glioma cells displayed this protein in the cell membrane, mitochondria, Golgi, and endoplasmic reticulum. Within the gBK region, two putative epitopes (gBK1 and gBK2) are predicted to bind to the HLA-A*0201 molecule. HLA-A*0201-restricted human CTLs were generated in vitro using gBK peptide-pulsed dendritic cells. Both gBK1 and gBK2 peptide-specific CTLs killed HLA-A2⁺/gBK⁺ gliomas, but they failed to kill non-HLA-A2-expressing but gBK⁺ target cells in cytolytic assays. T2 cells loaded with exogenous gBK peptides, but not with the influenza M1 control peptide, were only killed by their respective CTLs. The gBK-specific CTLs also killed a variety of other HLA-A*0201⁺ cancer cells that possess gBK, as well as HLA-A2⁺ HEK cells transfected with the gBK gene. Of clinical relevance, we found that T cells derived from glioblastoma multiforme patients that were sensitized to the gBK peptide could also kill target cells expressing gBK. This study shows that peptides derived from cancer-associated ion channels maybe useful targets for T cell-mediated immunotherapy.
Collapse
Affiliation(s)
- Lisheng Ge
- Diagnostic and Molecular Health Care Group, Long Beach, CA 90822, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|