1
|
Kim JY, Lee J, Koh JS, Park MJ, Chang UK. Establishment and characterization of a chordoma cell line from the tissue of a patient with dedifferentiated-type chordoma. J Neurosurg Spine 2016; 25:626-635. [DOI: 10.3171/2016.3.spine151077] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE
Chordoma is a rare bone tumor of the axial skeleton believed to originate from the remnants of the embryonic notochord. The available tumor cells are characteristically physaliferous and express brachyury, a transcription factor critical for mesoderm specification. Although chordomas are histologically not malignant, treatments remain challenging because they are resistant to radiation therapy and because wide resection is impossible in most cases. Therefore, a better understanding of the biology of chordomas using established cell lines may lead to the advancement of effective treatment strategies. The authors undertook a study to obtain this insight.
METHODS
Chordoma cells were isolated from the tissue of a patient with dedifferentiated-type chordoma (DTC) that had recurred. Cells were cultured with DMEM/F12 containing 10% fetal bovine serum and antibiotics (penicillin and streptomycin). Cell proliferation rate was measured by MTS assay. Cell-cycle distribution and cell surface expression of proteins were analyzed by fluorescence-activated cell sorting (FACS) analysis. Expression of proteins was analyzed by Western blot and immunocytochemistry. Radiation resistance was measured by clonogenic survival assay. Tumor formation was examined by injection of chordoma cells at hindlimb of nude mice.
RESULTS
The putative (DTC) cells were polygonal and did not have the conventional physaliferous characteristic seen in the U-CH1 cell line. The DTC cells exhibited similar growth rate and cell-cycle distribution, but they exhibited higher clonogenic activity in soft agar than U-CH1 cells. The DTC cells expressed high levels of platelet-derived growth factor receptor–β and a low level of brachyury and cytokeratins; they showed higher expression of stemness-related and epithelial to mesenchymal transition–related proteins than the U-CH1 cells. Intriguingly, FACS analysis revealed that DTC cells exhibited marginal surface expression of CD24 and CD44 and high surface expression of CXCR4 in comparison to U-CH1 cells. In addition, blockade of CXCR4 with its antagonist AMD3100 effectively suppressed the growth of both cell lines. The DTC cells were more resistant to paclitaxel, cisplatin, etoposide, and ionizing radiation than the U-CH1 cells. Injection of DTC cells into the hindlimb region of nude mice resulted in the efficient formation of tumors, and the histology of xenograft tumors was very similar to that of the original patient tumor.
CONCLUSIONS
The use of the established DTC cells along with preestablished cell lines of chordoma may help bring about greater understanding of the mechanisms underlying the chordoma that will lead to therapeutic strategies targeting chordomas.
Collapse
Affiliation(s)
- Jeong-Yub Kim
- 1Division of Radiation Cancer Research, Research Center for Radio-Senescence,
- 4Department of Pathology, College of Medicine, Korea University, Seoul, South Korea
| | - Jongsun Lee
- 1Division of Radiation Cancer Research, Research Center for Radio-Senescence,
| | | | - Myung-Jin Park
- 1Division of Radiation Cancer Research, Research Center for Radio-Senescence,
| | - Ung-Kyu Chang
- 3Department of Neurosurgery, Korea Institute of Radiological and Medical Sciences; and
| |
Collapse
|
2
|
Fujita N, Suzuki S, Watanabe K, Ishii K, Watanabe R, Shimoda M, Takubo K, Tsuji T, Toyama Y, Miyamoto T, Horiuchi K, Nakamura M, Matsumoto M. Chordoma-derived cell line U-CH1-N recapitulates the biological properties of notochordal nucleus pulposus cells. J Orthop Res 2016; 34:1341-50. [PMID: 27248133 PMCID: PMC5108487 DOI: 10.1002/jor.23320] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 05/25/2016] [Indexed: 02/04/2023]
Abstract
Intervertebral disc degeneration proceeds with age and is one of the major causes of lumbar pain and degenerative lumbar spine diseases. However, studies in the field of intervertebral disc biology have been hampered by the lack of reliable cell lines that can be used for in vitro assays. In this study, we show that a chordoma-derived cell line U-CH1-N cells highly express the nucleus pulposus (NP) marker genes, including T (encodes T brachyury transcription factor), KRT19, and CD24. These observations were further confirmed by immunocytochemistry and flow cytometry. Reporter analyses showed that transcriptional activity of T was enhanced in U-CH1-N cells. Chondrogenic capacity of U-CH1-N cells was verified by evaluating the expression of extracellular matrix (ECM) genes and Alcian blue staining. Of note, we found that proliferation and synthesis of chondrogenic ECM proteins were largely dependent on T in U-CH1-N cells. In accordance, knockdown of the T transcripts suppressed the expression of PCNA, a gene essential for DNA replication, and SOX5 and SOX6, the master regulators of chondrogenesis. On the other hand, the CD24-silenced cells showed no reduction in the mRNA expression level of the chondrogenic ECM genes. These results suggest that U-CH1-N shares important biological properties with notochordal NP cells and that T plays crucial roles in maintaining the notochordal NP cell-like phenotype in this cell line. Taken together, our data indicate that U-CH1-N may serve as a useful tool in studying the biology of intervertebral disc. © 2016 The Authors. Journal of Orthopaedic Research Published by Wiley Periodicals, Inc. on behalf of Orthopaedic Research Society. J Orthop Res 34:1341-1350, 2016.
Collapse
Affiliation(s)
- Nobuyuki Fujita
- Departments of Orthopaedic SurgeryKeio University School of Medicine35 ShinanomachiShinjuku‐kuTokyo160‐8582Japan
| | - Satoshi Suzuki
- Departments of Orthopaedic SurgeryKeio University School of Medicine35 ShinanomachiShinjuku‐kuTokyo160‐8582Japan
| | - Kota Watanabe
- Departments of Orthopaedic SurgeryKeio University School of Medicine35 ShinanomachiShinjuku‐kuTokyo160‐8582Japan
| | - Ken Ishii
- Departments of Orthopaedic SurgeryKeio University School of Medicine35 ShinanomachiShinjuku‐kuTokyo160‐8582Japan
| | - Ryuichi Watanabe
- Departments of Orthopaedic SurgeryKeio University School of Medicine35 ShinanomachiShinjuku‐kuTokyo160‐8582Japan
| | - Masayuki Shimoda
- Departments of PathologyKeio University School of Medicine35 ShinanomachiShinjuku‐kuTokyo160‐8582Japan
| | - Keiyo Takubo
- Department of Stem Cell BiologyResearch Institute, National Center for Global Health and Medicine1‐21‐1 ToyamaShinjuku‐kuTokyo160‐8582Japan
| | - Takashi Tsuji
- Kitasato Institute Hospital5‐9‐1 ShiroganeMinato‐kuTokyo108‐8642Japan
| | - Yoshiaki Toyama
- Departments of Orthopaedic SurgeryKeio University School of Medicine35 ShinanomachiShinjuku‐kuTokyo160‐8582Japan
| | - Takeshi Miyamoto
- Departments of Orthopaedic SurgeryKeio University School of Medicine35 ShinanomachiShinjuku‐kuTokyo160‐8582Japan
| | - Keisuke Horiuchi
- Departments of Orthopaedic SurgeryKeio University School of Medicine35 ShinanomachiShinjuku‐kuTokyo160‐8582Japan
| | - Masaya Nakamura
- Departments of Orthopaedic SurgeryKeio University School of Medicine35 ShinanomachiShinjuku‐kuTokyo160‐8582Japan
| | - Morio Matsumoto
- Departments of Orthopaedic SurgeryKeio University School of Medicine35 ShinanomachiShinjuku‐kuTokyo160‐8582Japan
| |
Collapse
|
3
|
Di Maio S, Yip S, Al Zhrani GA, Alotaibi FE, Al Turki A, Kong E, Rostomily RC. Novel targeted therapies in chordoma: an update. Ther Clin Risk Manag 2015; 11:873-83. [PMID: 26097380 PMCID: PMC4451853 DOI: 10.2147/tcrm.s50526] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Chordomas are rare, locally aggressive skull base neoplasms known for local recurrence and not-infrequent treatment failure. Current evidence supports the role of maximal safe surgical resection. In addition to open skull-base approaches, the endoscopic endonasal approach to clival chordomas has been reported with favorable albeit early results. Adjuvant radiation is prescribed following complete resection, alternatively for gross residual disease or at the time of recurrence. The modalities of adjuvant radiation therapy reported vary widely and include proton-beam, carbon-ion, fractionated photon radiotherapy, and photon and gamma-knife radiosurgery. As of now, no direct comparison is available, and high-level evidence demonstrating superiority of one modality over another is lacking. While systemic therapies have yet to form part of any first-line therapy for chordomas, a number of targeted agents have been evaluated to date that inhibit specific molecules and their respective pathways known to be implicated in chordomas. These include EGFR (erlotinib, gefitinib, lapatinib), PDGFR (imatinib), mTOR (rapamycin), and VEGF (bevacizumab). This article provides an update of the current multimodality treatment of cranial base chordomas, with an emphasis on how current understanding of molecular pathogenesis provides a framework for the development of novel targeted approaches.
Collapse
Affiliation(s)
- Salvatore Di Maio
- Division of Neurosurgery, Jewish General Hospital, McGill University, Montreal, QC, Canada
| | - Stephen Yip
- Department of Pathology and Laboratory Medicine, Vancouver General Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Gmaan A Al Zhrani
- National Neuroscience Institute, Department of Neurosurgery, King Fahad Medical City, Riyadh, Saudi Arabia ; Department of Neurology and Neurosurgery, The Montreal Neurological Institute and Hospital, McGill University Health Centre, Montreal, QC, Canada
| | - Fahad E Alotaibi
- National Neuroscience Institute, Department of Neurosurgery, King Fahad Medical City, Riyadh, Saudi Arabia ; Department of Neurology and Neurosurgery, The Montreal Neurological Institute and Hospital, McGill University Health Centre, Montreal, QC, Canada
| | - Abdulrahman Al Turki
- National Neuroscience Institute, Department of Neurosurgery, King Fahad Medical City, Riyadh, Saudi Arabia ; Department of Neurology and Neurosurgery, The Montreal Neurological Institute and Hospital, McGill University Health Centre, Montreal, QC, Canada
| | - Esther Kong
- Department of Pathology and Laboratory Medicine, Vancouver General Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Robert C Rostomily
- Department of Neurological Surgery, University of Washington, University of Washington Medical Center, Seattle, WA, USA
| |
Collapse
|
4
|
Patel P, Brooks C, Seneviratne A, Hess DA, Séguin CA. Investigating microenvironmental regulation of human chordoma cell behaviour. PLoS One 2014; 9:e115909. [PMID: 25541962 PMCID: PMC4277432 DOI: 10.1371/journal.pone.0115909] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 12/01/2014] [Indexed: 12/30/2022] Open
Abstract
The tumour microenvironment is complex and composed of many different constituents, including matricellular proteins such as connective tissue growth factor (CCN2), and is characterized by gradients in oxygen levels. In various cancers, hypoxia and CCN2 promote stem and progenitor cell properties, and regulate the proliferation, migration and phenotype of cancer cells. Our study was aimed at investigating the effects of hypoxia and CCN2 on chordoma cells, using the human U-CH1 cell line. We demonstrate that under basal conditions, U-CH1 cells express multiple CCN family members including CCN1, CCN2, CCN3 and CCN5. Culture of U-CH1 cells in either hypoxia or in the presence of recombinant CCN2 peptide promoted progenitor cell-like characteristics specific to the notochordal tissue of origin. Specifically, hypoxia induced the most robust increase in progenitor-like characteristics in U-CH1 cells, including increased expression of the notochord-associated markers T, CD24, FOXA1, ACAN and CA12, increased cell growth and tumour-sphere formation, and a decrease in the percentage of vacuolated cells present in the heterogeneous population. Interestingly, the effects of recombinant CCN2 peptide on U-CH1 cells were more pronounced under normoxia than hypoxia, promoting increased expression of CCN1, CCN2, CCN3 and CCN5, the notochord-associated markers SOX5, SOX6, T, CD24, and FOXA1 as well as increased tumour-sphere formation. Overall, this study highlights the importance of multiple factors within the tumour microenvironment and how hypoxia and CCN2 may regulate human chordoma cell behaviour.
Collapse
Affiliation(s)
- Priya Patel
- Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Courtney Brooks
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Ayesh Seneviratne
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada
- Robarts Research Institute, The University of Western Ontario, London, Ontario, Canada
| | - David A. Hess
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada
- Robarts Research Institute, The University of Western Ontario, London, Ontario, Canada
| | - Cheryle A. Séguin
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
5
|
Chemotherapy of skull base chordoma tailored on responsiveness of patient-derived tumor cells to rapamycin. Neoplasia 2014; 15:773-82. [PMID: 23814489 DOI: 10.1593/neo.13150] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Revised: 04/05/2013] [Accepted: 04/11/2013] [Indexed: 01/05/2023] Open
Abstract
Skull base chordomas are challenging tumors due to their deep surgical location and resistance to conventional radiotherapy. Chemotherapy plays a marginal role in the treatment of chordoma resulting from lack of preclinical models due to the difficulty in establishing tumor cell lines and valuable in vivo models. Here, we established a cell line from a recurrent clival chordoma. Cells were cultured for more than 30 passages and the expression of the chordoma cell marker brachyury was monitored using both immunohistochemistry and Western blot. Sensitivity of chordoma cells to the inhibition of specific signaling pathways was assessed through testing of a commercially available small molecule kinase inhibitor library. In vivo tumorigenicity was evaluated by grafting chordoma cells onto immunocompromised mice and established tumor xenografts were treated with rapamycin. Rapamycin was administered to the donor patient and its efficacy was assessed on follow-up neuroimaging. Chordoma cells maintained brachyury expression at late passages and generated xenografts closely mimicking the histology and phenotype of the parental tumor. Rapamycin was identified as an inhibitor of chordoma cell proliferation. Molecular analyses on tumor cells showed activation of the mammalian target of rapamycin signaling pathway and mutation of KRAS gene. Rapamycin was also effective in reducing the growth of chordoma xenografts. On the basis of these results, our patient received rapamycin therapy with about six-fold reduction of the tumor growth rate upon 10-month follow-up neuroimaging. This is the first case of chordoma in whom chemotherapy was tailored on the basis of the sensitivity of patient-derived tumor cells.
Collapse
|
6
|
Liu R, Wang K, Yuan K, Wei Y, Huang C. Integrative oncoproteomics strategies for anticancer drug discovery. Expert Rev Proteomics 2014; 7:411-29. [DOI: 10.1586/epr.10.14] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
7
|
Davies JM, Robinson AE, Cowdrey C, Mummaneni PV, Ducker GS, Shokat KM, Bollen A, Hann B, Phillips JJ. Generation of a patient-derived chordoma xenograft and characterization of the phosphoproteome in a recurrent chordoma. J Neurosurg 2013; 120:331-6. [PMID: 24286145 DOI: 10.3171/2013.10.jns13598] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
OBJECT The management of patients with locally recurrent or metastatic chordoma is a challenge. Preclinical disease models would greatly accelerate the development of novel therapeutic options for chordoma. The authors sought to establish and characterize a primary xenograft model for chordoma that faithfully recapitulates the molecular features of human chordoma. METHODS Chordoma tissue from a recurrent clival tumor was obtained at the time of surgery and implanted subcutaneously into NOD-SCID interleukin-2 receptor gamma (IL-2Rγ) null (NSG) mouse hosts. Successful xenografts were established and passaged in the NSG mice. The recurrent chordoma and the derived human chordoma xenograft were compared by histology, immunohistochemistry, and phospho-specific immunohistochemistry. Based on these results, mice harboring subcutaneous chordoma xenografts were treated with the mTOR inhibitor MLN0128, and tumors were subjected to phosphoproteome profiling using Luminex technology and immunohistochemistry. RESULTS SF8894 is a novel chordoma xenograft established from a recurrent clival chordoma that faithfully recapitulates the histopathological, immunohistological, and phosphoproteomic features of the human tumor. The PI3K/Akt/mTOR pathway was activated, as evidenced by diffuse immunopositivity for phospho-epitopes, in the recurrent chordoma and in the established xenograft. Treatment of mice harboring chordoma xenografts with MLN0128 resulted in decreased activity of the PI3K/Akt/mTOR signaling pathway as indicated by decreased phospho-mTOR levels (p = 0.019, n = 3 tumors per group). CONCLUSIONS The authors report the establishment of SF8894, a recurrent clival chordoma xenograft that mimics many of the features of the original tumor and that should be a useful preclinical model for recurrent chordoma.
Collapse
|
8
|
Di Maio S, Kong E, Yip S, Rostomily R. Converging paths to progress for skull base chordoma: Review of current therapy and future molecular targets. Surg Neurol Int 2013; 4:72. [PMID: 23776758 PMCID: PMC3683175 DOI: 10.4103/2152-7806.112822] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Accepted: 04/12/2013] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Chordomas of the skull base are rare locally aggressive neoplasms with a predilection for encapsulating critical neurovascular structures, bony destruction and irregular growth patterns, and from which patients succumb to recurrence and treatment failures. METHODS A review of the medical literature is performed, using standard search engines and identifying articles related to skull base chordomas, surgery, radiation therapy, chemotherapy, molecular genetics, and prospective trials. RESULTS A synthesis of the literature is presented, including sections on pathology, treatment, molecular genetics, challenges, and future directions. CONCLUSION Beyond an understanding of the current treatment paradigms for skull base chordomas, the reader gains insight into the collaborative approach applied to orphan diseases, of which chordomas is a prime exemplar.
Collapse
Affiliation(s)
- Salvatore Di Maio
- Division of Neurosurgery, McGill University, Jewish General Hospital, Montreal, QC, Canada
| | | | | | | |
Collapse
|
9
|
Liu X, Nielsen GP, Rosenberg AE, Waterman PR, Yang W, Choy E, Sassi S, Yang S, Harmon DC, Yang C, Schwab JH, Kobayashi E, Mankin HJ, Xavier R, Weissleder R, Duan Z, Hornicek FJ. Establishment and characterization of a novel chordoma cell line: CH22. J Orthop Res 2012; 30:1666-73. [PMID: 22504929 DOI: 10.1002/jor.22113] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Accepted: 03/08/2012] [Indexed: 02/04/2023]
Abstract
Chordoma is a rare primary malignant bone tumor and there exist only a few established human chordoma cell lines. The scarcity of robust chordoma cell lines has limited the ability to study this tumor. In this report, we describe the establishment of a novel chordoma cell line and characterize its in vitro and in vivo behaviors. The tumor tissue was isolated from a patient with recurrent chordoma of the sacrum. After 6 months in culture, the chordoma cell line, referred here as CH22, was established. Microscopic analysis of two-dimensional culture confirmed that the CH22 cells exhibited a typical vacuolated cytoplasm similar to the well-established chordoma cell line U-CH1. Electron microscopy showed cohesive cells with numerous surface filopodia, pockets of glycogen and aggregates of intermediate tonofilaments in cytoplasm. Three-dimensional culture revealed that the CH22 cells could grow and form clusters by day 8. The MTT assays demonstrated that, compared with sensitive osteosarcoma cell lines, CH22 cells were relatively resistant to conventional chemotherapeutic drugs. Western blotting and immunofluorescence analysis confirmed that the CH22 cells expressed brachyury, vimentin, and cytokeratin. Finally, histological analysis of CH22 xenograft tumor tissues demonstrated the appearance of physaliphorous cells and positive staining of brachyury, cytokeratin, and S100. By CT and MRI, imaging xenografts showed the typical appearances seen in human chordomas. These findings suggest that the established novel human chordoma cell line CH22 and its tumorigenecity in SCID nude mice may serve as an important model for studying chordoma cell biology and the development of new therapeutic modalities.
Collapse
Affiliation(s)
- Xianzhe Liu
- Department of Orthopaedic Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Bydon M, Papadimitriou K, Witham T, Wolinsky JP, Bydon A, Sciubba D, Gokaslan Z. Novel therapeutic targets in chordoma. Expert Opin Ther Targets 2012; 16:1139-43. [PMID: 22860993 DOI: 10.1517/14728222.2012.714772] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Chordomas are malignant bone tumors arising from notochordal remnants. They most commonly occur at the sacrum, skull base, and spine. The gold standard treatment for these tumors is a combination of en-bloc resection and radiation therapy. AREAS COVERED Recent genomic studies have identified duplication of the gene brachyury as a major susceptibility mutation in familial chordomas. Studies on sporadic chordomas have identified several tumor markers, using microRNAs and Comparative Genome Hybridization. In this article, we highlight current advances in research on the molecular characterization of chordomas. EXPERT OPINION Scientific advances have allowed for the identification of numerous tumor markers involved in chordoma pathogenesis. In the future, chordoma cell lines will be produced that silence or over-express these tumor markers. As we increase our understanding of the mechanism of chordoma tumor proliferation, we can expect the development of targeted drug therapies.
Collapse
Affiliation(s)
- Mohamad Bydon
- The Johns Hopkins Hospital, Department of Neurological Surgery, 600 N Wolfe St, Meyer 7-109, Baltimore, MD 21287, USA.
| | | | | | | | | | | | | |
Collapse
|
11
|
Siu IM, Salmasi V, Orr BA, Zhao Q, Binder ZA, Tran C, Ishii M, Riggins GJ, Hann CL, Gallia GL. Establishment and characterization of a primary human chordoma xenograft model. J Neurosurg 2012; 116:801-9. [PMID: 22283186 DOI: 10.3171/2011.12.jns111123] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT Chordomas are rare tumors arising from remnants of the notochord. Because of the challenges in achieving a complete resection, the radioresistant nature of these tumors, and the lack of effective chemotherapeutics, the median survival for patients with chordomas is approximately 6 years. Reproducible preclinical model systems that closely mimic the original patient's tumor are essential for the development and evaluation of effective therapeutics. Currently, there are only a few established chordoma cell lines and no primary xenograft model. In this study, the authors aimed to develop a primary chordoma xenograft model. METHODS The authors implanted independent tumor samples from 2 patients into athymic nude mice. The resulting xenograft line was characterized by histopathological analysis and immunohistochemical staining. The patient's tumor and serial passages of the xenograft were genomically analyzed using a 660,000 single-nucleotide polymorphism array. RESULTS A serially transplantable xenograft was established from one of the 2 patient samples. Histopathological analysis and immunohistochemical staining for S100 protein, epithelial membrane antigen, and cytokeratin AE1/AE3 of the primary patient sample and the xenografts confirmed that the xenografts were identical to the original chordoma obtained from the patient. Immunohistochemical staining and western blot analysis confirmed the presence of brachyury, a recently described marker of chordomas, in the tumor from the patient and each of the xenografts. Genome-wide variation was assessed between the patient's tumor and the xenografts and was found to be more than 99.9% concordant. CONCLUSIONS To the best of their knowledge, the authors have established the first primary chordoma xenograft that will provide a useful preclinical model for this disease and a platform for therapeutic development.
Collapse
Affiliation(s)
- I-Mei Siu
- Department of Neurosurgery, The Johns Hopkins University, Baltimore, MD, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Hsu W, Mohyeldin A, Shah SR, ap Rhys CM, Johnson LF, Sedora-Roman NI, Kosztowski TA, Awad OA, McCarthy EF, Loeb DM, Wolinsky JP, Gokaslan ZL, Quiñones-Hinojosa A. Generation of chordoma cell line JHC7 and the identification of Brachyury as a novel molecular target. J Neurosurg 2011; 115:760-9. [PMID: 21699479 PMCID: PMC4273567 DOI: 10.3171/2011.5.jns11185] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
OBJECT Chordoma is a malignant bone neoplasm hypothesized to arise from notochordal remnants along the length of the neuraxis. Recent genomic investigation of chordomas has identified T (Brachyury) gene duplication as a major susceptibility mutation in familial chordomas. Brachyury plays a vital role during embryonic development of the notochord and has recently been shown to regulate epithelial-to-mesenchymal transition in epithelial-derived cancers. However, current understanding of the role of this transcription factor in chordoma is limited due to the lack of availability of a fully characterized chordoma cell line expressing Brachyury. Thus, the objective of this study was to establish the first fully characterized primary chordoma cell line expressing gain of the T gene locus that readily recapitulates the original parental tumor phenotype in vitro and in vivo. METHODS Using an intraoperatively obtained tumor sample from a 61-year-old woman with primary sacral chordoma, a chordoma cell line (JHC7, or Johns Hopkins Chordoma Line 7) was established. Molecular characterization of the primary tumor and cell line was conducted using standard immunostaining and Western blotting. Chromosomal aberrations and genomic amplification of the T gene in this cell line were determined. Using this cell line, a xenograft model was established and the histopathological analysis of the tumor was performed. Silencing of Brachyury and changes in gene expression were assessed. RESULTS The authors report, for the first time, the successful establishment of a chordoma cell line (JHC7) from a patient with pathologically confirmed sacral chordoma. This cell line readily forms tumors in immunodeficient mice that recapitulate the parental tumor phenotype with conserved histological features consistent with the parental tumor. Furthermore, it is demonstrated for the first time that silencing of Brachyury using short hairpin RNA renders the morphology of chordoma cells to a more differentiated-like state and leads to complete growth arrest and senescence with an inability to be passaged serially in vitro. CONCLUSIONS This report represents the first xenograft model of a sacral chordoma line described in the literature and the first cell line established with stable Brachyury expression. The authors propose that Brachyury is an attractive therapeutic target in chordoma and that JHC7 will serve as a clinically relevant model for the study of this disease.
Collapse
Affiliation(s)
- Wesley Hsu
- Department of Neurosurgery and Oncology, Brain Tumor Stem Cell Laboratory, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ahmed Mohyeldin
- Department of Neurosurgery and Oncology, Brain Tumor Stem Cell Laboratory, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Sagar R. Shah
- Department of Neurosurgery and Oncology, Brain Tumor Stem Cell Laboratory, The Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Colette M. ap Rhys
- Department of Neurosurgery and Oncology, Brain Tumor Stem Cell Laboratory, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Lakesha F. Johnson
- Department of Neurosurgery and Oncology, Brain Tumor Stem Cell Laboratory, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Neda I. Sedora-Roman
- Department of Neurosurgery and Oncology, Brain Tumor Stem Cell Laboratory, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Thomas A. Kosztowski
- Department of Neurosurgery and Oncology, Brain Tumor Stem Cell Laboratory, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ola A. Awad
- Department of Oncology and Pediatrics, Musculoskeletal Tumor Program, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Edward F. McCarthy
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - David M. Loeb
- Department of Oncology and Pediatrics, Musculoskeletal Tumor Program, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jean-Paul Wolinsky
- Department of Neurosurgery and Oncology, Brain Tumor Stem Cell Laboratory, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ziya L. Gokaslan
- Department of Neurosurgery and Oncology, Brain Tumor Stem Cell Laboratory, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Alfredo Quiñones-Hinojosa
- Department of Neurosurgery and Oncology, Brain Tumor Stem Cell Laboratory, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
13
|
Diaz RJ, Cusimano MD. The biological basis for modern treatment of chordoma. J Neurooncol 2011; 104:411-22. [PMID: 21384217 DOI: 10.1007/s11060-011-0559-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2011] [Accepted: 02/21/2011] [Indexed: 01/03/2023]
Abstract
Chordomas are rare malignant tumors arising in bone of the spheno-occiput, sacrum, and vertebral column which can cause neurological deficit. Current management of chordoma involves safe resection followed by radiation therapy. However, surgical resection is often subtotal and chordoma often recurs despite optimal therapy. Despite years of effort, effective adjuvant therapy for denovo, recurrent and metastatic chordoma are absent and 5-year survival is at best 65%. While no chemotherapeutic agent has been demonstrated to be effective against chordoma in vivo, a greater understanding of the genetics and molecular biology of chordoma is opening up avenues of investigation towards the rational development of targeted therapies. Although enthusiasm for the use of already established or new investigational agents will increase with greater understanding of chordoma biology, laboratory studies of these agents are important prior to incorporation into clinical human trials. The authors review the current state of knowledge regarding chordoma and offer insight into potential new therapies for this rare and challenging tumor.
Collapse
Affiliation(s)
- Roberto Jose Diaz
- Arthur and Sonia Labatt Brain Tumor Research Centre, The Hospital for Sick Children, Toronto, ON, Canada.
| | | |
Collapse
|
14
|
Molecular characterization of putative chordoma cell lines. Sarcoma 2010; 2010:630129. [PMID: 21253487 PMCID: PMC3022207 DOI: 10.1155/2010/630129] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2010] [Revised: 10/28/2010] [Accepted: 11/22/2010] [Indexed: 01/15/2023] Open
Abstract
Immortal tumor cell lines are an important model system for cancer research, however, misidentification and cross-contamination of cell lines are a common problem. Seven chordoma cell lines are reported in the literature, but none has been characterized in detail. We analyzed gene expression patterns and genomic copy number variations in five putative chordoma cell lines (U-CH1, CCL3, CCL4, GB60, and CM319). We also created a new chordoma cell line, U-CH2, and provided genotypes for cell lines for identity confirmation. Our analyses revealed that CCL3, CCL4, and GB60 are not chordoma cell lines, and that CM319 is a cancer cell line possibly derived from chordoma, but lacking expression of key chordoma biomarkers. U-CH1 and U-CH2 both have gene expression profiles, copy number aberrations, and morphology consistent with chordoma tumors. These cell lines also harbor genetic changes, such as loss of p16, MTAP, or PTEN, that make them potentially useful models for studying mechanisms of chordoma pathogenesis and for evaluating targeted therapies.
Collapse
|
15
|
Slowed diffusion of single nanoparticles in the extracellular microenvironment of living cells revealed by darkfield microscopy. Anal Bioanal Chem 2010; 399:353-9. [DOI: 10.1007/s00216-010-4340-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2010] [Revised: 10/08/2010] [Accepted: 10/12/2010] [Indexed: 12/14/2022]
|
16
|
Abstract
STUDY DESIGN An experimental study to investigate the characterization of 3 chordoma cell lines. OBJECTIVE To characterize chordoma cell lines and generate hypothesis for further chordoma studies. SUMMARY OF BACKGROUND DATA Three cultured human chordoma cell lines have been successfully generated; however, their characterization is incomplete. Complete characterization of chordoma cell lines is necessary for these reagents to be a useful preclinical model. METHODS Three chordoma cell lines, CH 8, U-CH1, and GP 60, were cultured in different commercially available tissue culture media. They were also cultured in different environments, which included collagen substrate, various concentrations of glucose, and various levels of hypoxic conditions. The rate of cell proliferation was assessed by either MTT or numeration assay. A 3-dimensional (3D) cell culture model of these chordoma cell lines was also studied, and the expression of vimentin and cytokeratin was measured by immunofluorescence and Western blot. Additionally, the sensitivity of the 3 chordoma cell lines to 6 chemotherapeutic drugs was analyzed. RESULTS CH 8, GP 60, and U-CH1 cells proliferate more actively in Iscove Modified Dulbecco Medium or Dulbecco modified Eagle Medium and less actively in RPMI medium. All 3 chordoma cell lines universally grow better in collagen substrate and survive in hypoxic conditions, whereas glucose concentration has no significant influence on their growth properties. Chordoma cell lines grew well in 3D culture systems and formed acini-like spheroids and retained the expression of vimentin and cytokeratin. MTT analysis indicates that all 3 chordoma cell lines are sensitive to doxorubicin, yondelis, zalypsis, and cisplatin. CONCLUSION We characterized 3 chordoma cell lines for differential growth properties in a variety of media and response to chemotherapeutic agents.
Collapse
|
17
|
Schwab JH, Boland PJ, Agaram NP, Socci ND, Guo T, O'Toole GC, Wang X, Ostroumov E, Hunter CJ, Block JA, Doty S, Ferrone S, Healey JH, Antonescu CR. Chordoma and chondrosarcoma gene profile: implications for immunotherapy. Cancer Immunol Immunother 2008; 58:339-49. [PMID: 18641983 DOI: 10.1007/s00262-008-0557-7] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2007] [Accepted: 06/26/2008] [Indexed: 01/10/2023]
Abstract
Chordoma and chondrosarcoma are malignant bone tumors characterized by the abundant production of extracellular matrix. The resistance of these tumors to conventional therapeutic modalities has prompted us to delineate the gene expression profile of these two tumor types, with the expectation to identify potential molecular therapeutic targets. Furthermore the transcriptional profile of chordomas and chrondrosarcomas was compared to a wide variety of sarcomas as well as to that of normal tissues of similar lineage, to determine whether they express unique gene signatures among other tumors of mesenchymal origin, and to identify changes associated with malignant transformation. A HG-U133A Affymetrix Chip platform was used to determine the gene expression signature in 6 chordoma and 14 chondrosarcoma lesions. Validation of selected genes was performed by qPCR and immunohistochemistry (IHC) on an extended subset of tumors. By unsupervised clustering, chordoma and chondrosarcoma tumors grouped together in a genomic cluster distinct from that of other sarcoma types. They shared overexpression of many extracellular matrix genes including aggrecan, type II & X collagen, fibronectin, matrillin 3, high molecular weight-melanoma associated antigen (HMW-MAA), matrix metalloproteinase MMP-9, and MMP-19. In contrast, T Brachyury and CD24 were selectively expressed in chordomas, as were Keratin 8,13,15,18 and 19. Chondrosarcomas are distinguished by high expression of type IX and XI collagen. Because of its potential usefulness as a target for immunotherapy, the expression of HMW-MAA was analyzed by IHC and was detected in 62% of chordomas and 48% of chondrosarcomas, respectively. Furthermore, western blotting analysis showed that HMW-MAA synthesized by chordoma cell lines has a structure similar to that of the antigen synthesized by melanoma cells. In conclusion, chordomas and chondrosarcomas share a similar gene expression profile of up-regulated extracellular matrix genes. HMW-MAA represents a potential useful target to apply immunotherapy to these tumors.
Collapse
Affiliation(s)
- Joseph H Schwab
- Department of Surgery, Orthopedic Service, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|