1
|
Matsumoto Y. Recent topics in diagnosis and treatment of malignant spinal tumors. Fukushima J Med Sci 2025; 71:85-95. [PMID: 39909449 PMCID: PMC12079046 DOI: 10.5387/fms.24-00008] [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: 02/15/2024] [Accepted: 11/07/2024] [Indexed: 02/07/2025] Open
Abstract
The diagnosis and treatment of malignant spinal tumors are complex and require an integrated approach known as Jaffe's triangle. This review discusses recent topics in the diagnosis and treatment of primary and metastatic malignant spinal tumors. Integrated diagnostic methods, including the development of a dumbbell scoring system for benign-malignant differentiation and the use of positron emission tomography and magnetic resonance imaging (PET-MRI), have improved diagnostic accuracy. Curative resection techniques such as vertebrectomy, sagittal resection, and posterior resection are crucial for primary malignant tumors. Heavy particle radiation therapy, such as carbon-ion radiotherapy, shows promise against radiation-resistant tumors, whereas novel drug therapies, such as denosumab, are effective for giant cell tumors of the bone arising in the spine. For metastatic spinal tumors, the collaborative efforts of the Bone Metastasis Cancer Board and minimally invasive spine stabilization have expanded surgical indications and improved patient outcomes. The treatment system has shifted towards preventive surgery and outpatient management, aiming to maintain quality of life and continue chemotherapy. Interdisciplinary collaboration is essential for improving treatment outcomes in both primary and metastatic malignant spinal tumors.Primary malignant spinal cord tumors (PMST) and metastatic spinal tumors (MST) are among the most difficult areas of orthopedic surgery. Their diagnosis and treatment require multidisciplinary diagnostic and therapeutic strategies that integrate knowledge and skills in orthopedics, pathology, and diagnostic radiology (the so-called Jaffe triangle), as well as in clinical oncology and tumor biology, which have made remarkable progress in recent years. Here, we review recent topics related to the diagnosis and treatment of PMST and MST.
Collapse
|
2
|
Noguchi R, Osaki J, Ono T, Adachi Y, Iwata S, Yoshimatsu Y, Sasaki K, Kawai A, Kondo T. Pharmacoproteogenomic approach identifies on-target kinase inhibitors for cancer drug repositioning. In Vitro Cell Dev Biol Anim 2024; 60:1200-1214. [PMID: 39422823 DOI: 10.1007/s11626-024-00983-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 09/01/2024] [Indexed: 10/19/2024]
Abstract
Drug repositioning of approved drugs offers advantages over de novo drug development for a rare type of cancer. To efficiently identify on-target drugs from clinically successful kinase inhibitors in cancer drug repositioning, drug screening and molecular profiling of cell lines are essential to exclude off-targets. We developed a pharmacoproteogenomic approach to identify on-target kinase inhibitors, combining molecular profiling of genomic features and kinase activity, and drug screening of patient-derived cell lines. This study examined eight patient-derived giant cell tumor of the bone (GCTB) cell lines, all of which harbored a signature mutation of H3-3A but otherwise without recurrent copy number variants and mutations. Kinase activity profiles of 100 tyrosine kinases with a three-dimensional substrate peptide array revealed that nine kinases were highly activated. Pharmacological screening of 60 clinically used kinase inhibitors found that nine drugs directed at 29 kinases strongly suppressed cell viability. We regarded ABL1, EGFR, and LCK as on-target kinases; among the two corresponding on-target kinase inhibitors, osimertinib and ponatinib emerged as on-target drugs whose target kinases were significantly activated. The remaining 26 kinases and seven kinase inhibitors were excluded as off-targets. Our pharmacoproteomic approach enabled the identification of on-target kinase inhibitors that are useful for drug repositioning.
Collapse
Affiliation(s)
- Rei Noguchi
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Julia Osaki
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Takuya Ono
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Yuki Adachi
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Shuhei Iwata
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Yuki Yoshimatsu
- Department of Patient-Derived Cancer Model, Tochigi Cancer Center, 4-9-13 Yohnan, Utsunomiya, Tochigi, 320-0834, Japan
| | - Kazuki Sasaki
- Department of Oncopeptidomics, Tochigi Cancer Center; 4-9-13 Yohnan, Utsunomiya, Tochigi, 320-0834, Japan
| | - Akira Kawai
- Department of Musculoskeletal Oncology and Rehabilitation Medicine, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Tadashi Kondo
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan.
| |
Collapse
|
3
|
Kimura A, Toda Y, Matsumoto Y, Yamamoto H, Yahiro K, Shimada E, Kanahori M, Oyama R, Fukushima S, Nakagawa M, Setsu N, Endo M, Fujiwara T, Matsunobu T, Oda Y, Nakashima Y. Nuclear β-catenin translocation plays a key role in osteoblast differentiation of giant cell tumor of bone. Sci Rep 2022; 12:13438. [PMID: 35927428 PMCID: PMC9352730 DOI: 10.1038/s41598-022-17728-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 07/29/2022] [Indexed: 12/02/2022] Open
Abstract
Denosumab is a game-changing drug for giant cell tumor of bone (GCTB); however, its clinical biomarker regarding tumor ossification of GCTB has not been elucidated. In this study, we investigated the relationship between Wnt/β-catenin signaling and the ossification of GCTB and evaluated whether endogenous nuclear β-catenin expression predicted denosumab-induced bone formation in GCTB. Genuine patient-derived primary GCTB tumor stromal cells exhibited osteoblastic characteristics. Identified osteoblastic markers and nuclear β-catenin translocation were significantly upregulated via differentiation induction and were inhibited by treating with Wnt signaling inhibitor, GGTI-286, or selective Rac1-LEF inhibitor, NSC23766. Furthermore, we reviewed the endogenous ossification and nuclear β-catenin translocation of 86 GCTB clinical samples and elucidated that intra-tumoral ossification was significantly associated with the nuclear translocation. Three-dimensional quantitative analyses (n = 13) of tumoral CT images have revealed that the nuclear β-catenin translocation of naïve GCTB samples was significantly involved with the denosumab-induced tumor ossification. Our findings suggest a close relationship between the nuclear β-catenin translocation and the osteoblastic differentiation of GCTB. Investigations of the nuclear β-catenin in naïve GCTB samples may provide a promising biomarker for predicting the ossification of GCTB following denosumab treatment.
Collapse
Affiliation(s)
- Atsushi Kimura
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka City, 812-8582, Japan
| | - Yu Toda
- Department of Anatomic Pathology, Pathological Sciences, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshihiro Matsumoto
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka City, 812-8582, Japan.
| | - Hidetaka Yamamoto
- Department of Anatomic Pathology, Pathological Sciences, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kenichiro Yahiro
- Department of Orthopedic Surgery, National Hospital Organization Kyushu Medical Center, Fukuoka, Japan
| | - Eijiro Shimada
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka City, 812-8582, Japan
| | - Masaya Kanahori
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka City, 812-8582, Japan
| | - Ryunosuke Oyama
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka City, 812-8582, Japan
| | - Suguru Fukushima
- Department of Musculoskeletal Oncology and Rehabilitation, National Cancer Center, Tokyo, Japan
| | - Makoto Nakagawa
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka City, 812-8582, Japan
| | - Nokitaka Setsu
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka City, 812-8582, Japan
| | - Makoto Endo
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka City, 812-8582, Japan
| | - Toshifumi Fujiwara
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka City, 812-8582, Japan
| | - Tomoya Matsunobu
- Department of Orthopaedic Surgery, Kyushu Rosai Hospital, Fukuoka, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Pathological Sciences, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yasuharu Nakashima
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka City, 812-8582, Japan
| |
Collapse
|
4
|
He Y, Cheng D, Lian C, Liu Y, Luo W, Wang Y, Ma C, Wu Q, Tian P, He D, Jia Z, Lv X, Zhang X, Pan Z, Lu J, Xiao Y, Zhang P, Liang Y, Yang Q, Hu G. Serglycin induces osteoclastogenesis and promotes tumor growth in giant cell tumor of bone. Cell Death Dis 2021; 12:868. [PMID: 34556636 PMCID: PMC8460728 DOI: 10.1038/s41419-021-04161-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 08/24/2021] [Accepted: 09/08/2021] [Indexed: 11/09/2022]
Abstract
Giant cell tumor of bone (GCTB) is an aggressive osteolytic bone tumor characterized by the within-tumor presence of osteoclast-like multinucleated giant cells (MGCs), which are induced by the neoplastic stromal cells and lead to extensive bone destruction. However, the underlying mechanism of the pathological process of osteoclastogenesis in GCTB is poorly understood. Here we show that the proteoglycan Serglycin (SRGN) secreted by neoplastic stromal cells plays a crucial role in the formation of MGCs and tumorigenesis in GCTB. Upregulated SRGN expression and secretion are observed in GCTB tumor cells and patients. Stromal-derived SRGN promotes osteoclast differentiation from monocytes. SRGN knockdown in stromal cells inhibits tumor growth and bone destruction in a patient-derived orthotopic xenograft model of mice. Mechanistically SRGN interacts with CD44 on the cell surface of monocytes and thus activates focal adhesion kinase (FAK), leading to osteoclast differentiation. Importantly, blocking CD44 with a neutralizing antibody reduces the number of MGCs and suppresses tumorigenesis in vivo. Overall, our data reveal a mechanism of MGC induction in GCTB and support CD44-targeting approaches for GCTB treatment.
Collapse
Affiliation(s)
- Yunfei He
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Dongdong Cheng
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Cheng Lian
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yingjie Liu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Wenqian Luo
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yuan Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Chengxin Ma
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qiuyao Wu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Pu Tian
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Dasa He
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zhenchang Jia
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xianzhe Lv
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xue Zhang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zhen Pan
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Jinxi Lu
- Department of General Surgery, Xinzhou District People's Hospital, Wuhan, China
| | - Yansen Xiao
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Peiyuan Zhang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yajun Liang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qingcheng Yang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.
| | - Guohong Hu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China. .,Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.
| |
Collapse
|
5
|
Shibuya I, Takami M, Kawamoto M, Karakawa A, Nakamura S, Kamijo R. Immunohistochemical Analysis of the Distribution of RANKL-Expressing Cells and the Expression of Osteoclast-Related Markers in Giant Cell Tumor of Bone. J HARD TISSUE BIOL 2020. [DOI: 10.2485/jhtb.29.137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- Isao Shibuya
- Department of Biochemistry, Showa University School of Dentistry
- Department of Orthopaedic Surgery, Teikyo University Mizonokuchi Hospital
| | - Masamichi Takami
- Department of Pharmacology, Showa University School of Dentistry
| | - Masashi Kawamoto
- Department of Diagnostic Pathology, Teikyo University Mizonokuchi Hospital
| | - Akiko Karakawa
- Department of Pharmacology, Showa University School of Dentistry
| | - Shigeru Nakamura
- Department of Orthopaedic Surgery, Teikyo University Mizonokuchi Hospital
| | - Ryutaro Kamijo
- Department of Biochemistry, Showa University School of Dentistry
| |
Collapse
|
6
|
Omlor GW, Lange J, Streit M, Gantz S, Merle C, Germann T, Mechtersheimer G, Fellenberg J, Lehner B. Retrospective analysis of 51 intralesionally treated cases with progressed giant cell tumor of the bone: local adjuvant use of hydrogen peroxide reduces the risk for tumor recurrence. World J Surg Oncol 2019; 17:73. [PMID: 31014317 PMCID: PMC6480805 DOI: 10.1186/s12957-019-1613-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Accepted: 04/08/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Giant cell tumor of the bone (GCT) has high local recurrence rates and the prognosis is hard to predict. We therefore retrospectively analyzed clinical outcome and recurrences of 51 GCT cases focusing on the effects of adjuvant local use of hydrogen peroxide. METHODS The series enclosed 51 advanced GCT cases of the upper and lower extremities (n = 27 Campanacci grade III; n = 24 grade II; n = 39 surgery at our institution, n = 12 elsewhere). Mean follow-up was 88.3 (± 62.0) months. Surgical details, histology, metastases, recurrences, and interview-based data on satisfaction and function including the Musculoskeletal Tumor Society (MSTS) score were evaluated. It was investigated whether hydrogen peroxide was additionally used or not to clean the tumor cavity after curettage as we hypothesized influence on recurrences. To analyze the underlying mechanisms, GCT-derived stromal cell lines were cultured in vitro and tested for cell viability and apoptosis after treatment with hydrogen peroxide. Statistical analysis was performed with Student's t tests, analysis of variance (ANOVA) with post hoc testing, Mann-Whitney U tests, chi-square tests, Kaplan-Meier analysis, and multivariate Cox regression analysis. RESULTS The whole series had 21 recurrences (41%). Eleven recurrences were found (28%) after surgery at our institution. Kaplan-Meier analysis of cumulative recurrence-free survival revealed at 2 years follow-up 69% (72%, only our institution) and at 10 years follow-up 54% (68%, only our institution). Intralesional resection was performed by vigorous curettage, burring, and defect filling with either polymethylmethacrylate bone cement (n = 45) or cancellous bone from the iliac crest (n = 6). Univariate chi-square analysis showed significantly lower recurrence rate after bone cement filling (2.3-fold, p = 0.024). Cleaning of the lesion cavity with hydrogen peroxide significantly reduced recurrence rate (whole collective 2.9-fold, p = 0.004; our institution 2.8-fold, p = 0.04) and significantly increased cumulative recurrence-free survival rate (whole collective at 10 years follow-up 74% versus 31%, p = 0.002; our institution 79% versus 48%, p = 0.02) compared to cases without hydrogen peroxide treatment. In multivariate analysis, significant risk factors for recurrence were pathological fracture (hazard ratio 3.7; p = 0.04), high mitosis rate (hazard ratio 15.6; p = 0.01), and lack of hydrogen peroxide use (hazard ratio 6.0; p = 0.02). In vitro cell culture analyses found apoptotic nature of hydrogen peroxide induced GCT cell death. CONCLUSIONS The present series proved for the first time that additional cleaning of the tumor cavity with hydrogen peroxide before defect filling significantly reduced recurrence rate and significantly increased recurrence-free survival in advanced but intralesionally treated GCT cases.
Collapse
Affiliation(s)
- Georg W Omlor
- Center of Orthopaedics, Trauma Surgery and Paraplegiology, Heidelberg University Hospital, Schlierbacher Landstrasse 200a, 69118, Heidelberg, Germany.
| | - Jessica Lange
- Center of Orthopaedics, Trauma Surgery and Paraplegiology, Heidelberg University Hospital, Schlierbacher Landstrasse 200a, 69118, Heidelberg, Germany
| | - Marcus Streit
- Center of Orthopaedics, Trauma Surgery and Paraplegiology, Heidelberg University Hospital, Schlierbacher Landstrasse 200a, 69118, Heidelberg, Germany
| | - Simone Gantz
- Center of Orthopaedics, Trauma Surgery and Paraplegiology, Heidelberg University Hospital, Schlierbacher Landstrasse 200a, 69118, Heidelberg, Germany
| | - Christian Merle
- Center of Orthopaedics, Trauma Surgery and Paraplegiology, Heidelberg University Hospital, Schlierbacher Landstrasse 200a, 69118, Heidelberg, Germany
| | - Thomas Germann
- Institute of Pathology Heidelberg, University of Heidelberg, 69120, Heidelberg, Germany
| | - Gunhild Mechtersheimer
- Department of Diagnostic and Interventional Radiology, University of Heidelberg, 69120, Heidelberg, Germany
| | - Jörg Fellenberg
- Center of Orthopaedics, Trauma Surgery and Paraplegiology, Heidelberg University Hospital, Schlierbacher Landstrasse 200a, 69118, Heidelberg, Germany
| | - Burkhard Lehner
- Center of Orthopaedics, Trauma Surgery and Paraplegiology, Heidelberg University Hospital, Schlierbacher Landstrasse 200a, 69118, Heidelberg, Germany
| |
Collapse
|
7
|
Zhang L, Lv Z, Xu J, Chen C, Ge Q, Li P, Wei D, Wu Z, Sun X. Micro
RNA
‐134 inhibits osteosarcoma angiogenesis and proliferation by targeting the
VEGFA
/
VEGFR
1 pathway. FEBS J 2018; 285:1359-1371. [PMID: 29474747 DOI: 10.1111/febs.14416] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Revised: 01/31/2018] [Accepted: 02/19/2018] [Indexed: 12/16/2022]
Affiliation(s)
| | - Zhi Lv
- Department of Orthopaedics The Second Hospital of Shanxi Medical University Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair Taiyuan China
| | - Jing Xu
- Shanxi Medical University Taiyuan China
| | | | | | - Pengcui Li
- Department of Orthopaedics The Second Hospital of Shanxi Medical University Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair Taiyuan China
| | | | - Zhuangzhuang Wu
- Department of Orthopaedics The Second Hospital of Shanxi Medical University Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair Taiyuan China
| | - Xiaojuan Sun
- Department of Orthopaedics The Second Hospital of Shanxi Medical University Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair Taiyuan China
| |
Collapse
|
8
|
Colia V, Provenzano S, Hindi N, Casali PG, Stacchiotti S. Systemic therapy for selected skull base sarcomas: Chondrosarcoma, chordoma, giant cell tumour and solitary fibrous tumour/hemangiopericytoma. Rep Pract Oncol Radiother 2016; 21:361-9. [PMID: 27330421 DOI: 10.1016/j.rpor.2015.12.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 07/10/2015] [Accepted: 12/18/2015] [Indexed: 12/14/2022] Open
Abstract
This review highlights the data currently available on the activity of systemic therapy in chondrosarcoma, chordoma, giant cell tumour of the bone (GCTB) and solitary fibrous tumour, i.e., four rare sarcomas amongst mesenchymal malignancy arising from the skull base.
Collapse
Affiliation(s)
- Vittoria Colia
- Adult Mesenchymal Tumour & Rare Cancer Medical Oncology Unit, Cancer Medicine Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Salvatore Provenzano
- Adult Mesenchymal Tumour & Rare Cancer Medical Oncology Unit, Cancer Medicine Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Nadia Hindi
- Adult Mesenchymal Tumour & Rare Cancer Medical Oncology Unit, Cancer Medicine Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Paolo G Casali
- Adult Mesenchymal Tumour & Rare Cancer Medical Oncology Unit, Cancer Medicine Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Silvia Stacchiotti
- Adult Mesenchymal Tumour & Rare Cancer Medical Oncology Unit, Cancer Medicine Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
9
|
Conceição ALG, Babeto E, Candido NM, Franco FC, de Campos Zuccari DAP, Bonilha JL, Cordeiro JA, Calmon MF, Rahal P. Differential Expression of ADAM23, CDKN2A (P16), MMP14 and VIM Associated with Giant Cell Tumor of Bone. J Cancer 2015; 6:593-603. [PMID: 26078788 PMCID: PMC4466407 DOI: 10.7150/jca.11238] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 02/10/2015] [Indexed: 12/17/2022] Open
Abstract
Though benign, giant cell tumor of bone (GCTB) can become aggressive and can exhibit a high mitotic rate, necrosis and rarely vascular invasion and metastasis. GCTB has unique histologic characteristics, a high rate of multinucleated cells, a variable and unpredictable growth potential and uncertain biological behavior. In this study, we sought to identify genes differentially expressed in GCTB, thus building a molecular profile of this tumor. We performed quantitative real-time polymerase chain reaction (qPCR), immunohistochemistry and analyses of methylation to identify genes that are putatively associated with GCTB. The expression of the ADAM23 and CDKN2A genes was decreased in GCTB samples compared to normal bone tissue, measured by qPCR. Additionally, a high hypermethylation frequency of the promoter regions of ADAM23 and CDKN2A in GCTB was observed. The expression of the MAP2K3, MMP14, TIMP2 and VIM genes was significantly higher in GCTB than in normal bone tissue, a fact that was confirmed by qPCR and immunohistochemistry. The set of genes identified here furthers our understanding of the molecular basis of GCTB.
Collapse
Affiliation(s)
| | - Erica Babeto
- 1. Laboratory of Genomics Studies, UNESP, São José do Rio Preto, Brazil
| | | | | | | | | | - José Antônio Cordeiro
- 4. Department of Epidemiology and Collective Health, FAMERP, São José do Rio Preto, Brazil
| | | | - Paula Rahal
- 1. Laboratory of Genomics Studies, UNESP, São José do Rio Preto, Brazil
| |
Collapse
|
10
|
Zhang J, Dong J, Yang Z, Ma X, Zhang J, Li M, Chen Y, Ding Y, Li K, Zhang Z. Expression of ezrin, CD44, and VEGF in giant cell tumor of bone and its significance. World J Surg Oncol 2015; 13:168. [PMID: 25929323 PMCID: PMC4434870 DOI: 10.1186/s12957-015-0579-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 04/08/2015] [Indexed: 11/23/2022] Open
Abstract
Background This research aimed to study the role of ezrin, CD44, and VEGF in invasion, metastasis, recurrence, and prognosis of giant cell tumor of bone (GCTB) and its association with the clinical and pathological features of GCTB. Methods Expression status of ezrin, CD44, and VEGF in 80 GCTB tissues and its adjacent noncancerous tissue samples were measured with immunohistochemical and Elivison staining. Their correlation with the clinical and pathologic factors was statistically analyzed by chi-square test. Results The expression status of ezrin, CD44, and VEGF were significantly higher in GCTB tissue samples than in its adjacent noncancerous tissue samples and in GCTB at Campanacci stage III than in Campanacci stages I and II (P < 0.05). No significant difference was found in age and sex of the patients and locations of the tumor (P > 0.05). Survival analysis showed that the expression status of ezrin, CD44, VEGF, and Campanacci clinical stages of GCTB were positively associated with the survival rate of GCTB patients and negatively associated with ezrin and Campanacci stages of GCTB, indicating that ezrin, CD44, VEGF, and Campanacci clinical stages of GCTB are the independent factors for GCTB. Conclusions Ezrin, CD44, and VEGF are over-expressed in GCTB tissue and its adjacent noncancerous tissue samples and may play an important role in the occurrence, invasion, metastasis, and recurrence of GCTB. Measurement of ezrin, CD44, and VEGF expression status may contribute to the judgment of prognosis of GCTB patients.
Collapse
Affiliation(s)
- Jing Zhang
- Bone and Soft Tissue Tumors Research Center of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming, Yunnan, 650118, People's Republic of China.
| | - Jian Dong
- Bone and Soft Tissue Tumors Research Center of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming, Yunnan, 650118, People's Republic of China.
| | - Zuozhang Yang
- Bone and Soft Tissue Tumors Research Center of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming, Yunnan, 650118, People's Republic of China.
| | - Xiang Ma
- Bone and Soft Tissue Tumors Research Center of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming, Yunnan, 650118, People's Republic of China.
| | - Jinlei Zhang
- Bone and Soft Tissue Tumors Research Center of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming, Yunnan, 650118, People's Republic of China.
| | - Mei Li
- Department of Pathology, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming, Yunnan, 650118, People's Republic of China.
| | - Yun Chen
- Department of Pathology, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming, Yunnan, 650118, People's Republic of China.
| | - Yingying Ding
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming, 650118, People's Republic China.
| | - Kun Li
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming, 650118, People's Republic China.
| | - Zhiping Zhang
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming, 650118, People's Republic China.
| |
Collapse
|
11
|
Krishnan B, Smith TL, Dubey P, Zapadka ME, Torti FM, Willingham MC, Tallant EA, Gallagher PE. Angiotensin-(1-7) attenuates metastatic prostate cancer and reduces osteoclastogenesis. Prostate 2013; 73:71-82. [PMID: 22644942 PMCID: PMC3842188 DOI: 10.1002/pros.22542] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Accepted: 05/03/2012] [Indexed: 12/19/2022]
Abstract
BACKGROUND Angiotensin-(1-7) [Ang-(1-7)] is an endogenous, heptapeptide hormone with anti-proliferative and anti-angiogenic properties. The primary objective of this study was to determine whether Ang-(1-7) effectively reduces prostate cancer metastasis in mice. METHODS Human PC3 prostate cancer cells were injected into the aortic arch via the carotid artery of SCID mice pre-treated with Ang-(1-7) or injected into the tibia of athymic mice, administered Ang-(1-7) for 5 weeks beginning 2 weeks post-injection. Tumor growth and volume were determined by bioluminescent and magnetic resonance imaging. The presence of tumors was confirmed by hematoxylin and eosin staining; TRAP histochemistry was used to identify osteolytic lesions. The effect of Ang-(1-7) on osteoclastogenesis was assessed in differentiated bone marrow cells. RESULTS Pre-treatment with Ang-(1-7) prevented metastatic tumor formation following intra-aortic injection of PC3 cells, while 83% of untreated mice developed tumors in metastatic sites. Circulating VEGF was significantly higher in control mice compared to mice administered Ang-(1-7). A 5-week regimen of the heptapeptide hormone attenuated intra-tibial tumor growth; Ang-(1-7) was significantly higher in the tibia of treated mice than in control animals. Osteoclastogenesis was reduced by 50% in bone marrow cells differentiated in the presence of Ang-(1-7), suggesting that the heptapeptide hormone prevents the formation of osteolytic lesions to reduce tumor survival in the bone microenvironment. CONCLUSIONS These findings suggest that Ang-(1-7) may serve as an anti-angiogenic and anti-metastatic agent for advanced prostate cancer. By extension, the heptapeptide hormone may provide effective therapy for bone metastasis produced from primary tumors of the lung and breast.
Collapse
Affiliation(s)
- Bhavani Krishnan
- Hypertension and Vascular Research Center, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157
- Molecular Genetics & Genomics Program, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157
| | - Thomas L. Smith
- Department of Orthopedic Surgery, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157
| | - Purnima Dubey
- Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157
| | - Michael. E. Zapadka
- Department of Radiology, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157
| | - Frank M. Torti
- Department of Cancer Biology Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157
| | - Mark C. Willingham
- Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157
| | - E. Ann Tallant
- Hypertension and Vascular Research Center, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157
- Molecular Genetics & Genomics Program, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157
| | - Patricia E. Gallagher
- Hypertension and Vascular Research Center, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157
| |
Collapse
|
12
|
Cowan RW, Singh G. Giant cell tumor of bone: a basic science perspective. Bone 2013; 52:238-46. [PMID: 23063845 DOI: 10.1016/j.bone.2012.10.002] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Revised: 09/27/2012] [Accepted: 10/01/2012] [Indexed: 12/26/2022]
Abstract
Comprehending the pathogenesis of giant cell tumor of bone (GCT) is of critical importance for developing novel targeted treatments for this locally-aggressive primary bone tumor. GCT is characterized by the presence of large multinucleated osteoclast-like giant cells distributed amongst mononuclear spindle-like stromal cells and other monocytes. The giant cells are principally responsible for the extensive bone resorption by the tumor. However, the spindle-like stromal cells chiefly direct the pathology of the tumor by recruiting monocytes and promoting their fusion into giant cells. The stromal cells also enhance the resorptive ability of the giant cells. This review encompasses many of the attributes of GCT, including the process of giant cell formation and the mechanisms of bone resorption. The significance of the receptor activator of nuclear factor-κB ligand (RANKL) in the development of GCT and the importance of proteases, including numerous matrix metalloproteinases, are highlighted. The mesenchymal lineage of the stromal cells and the origin of the hematopoietic monocytes are also discussed. Several aspects of GCT that require further understanding, including the etiology of the tumor, the mechanisms of metastases, and the development of an appropriate animal model, are also considered. By exploring the current status of GCT research, this review accentuates the significant progress made in understanding the biology of the tumor, and discusses important areas for future investigation.
Collapse
Affiliation(s)
- Robert W Cowan
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | | |
Collapse
|
13
|
Lan J, Liu X, Rong W, Wei F, Jiang L, Yu H, Dang G, Liu Z. Stro-1(+) stromal cells have stem-like features in giant cell tumor of bone. J Surg Oncol 2012; 106:826-36. [PMID: 22605660 DOI: 10.1002/jso.23151] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2011] [Accepted: 04/17/2012] [Indexed: 11/07/2022]
Abstract
BACKGROUND Giant cell tumor of bone (GCTB) is an aggressive benign bone tumor with poor prognosis whose neoplastic component is stromal cells (SCs). Tumor stem-like cells (TSCs) have been demonstrated as precursors for tumor genesis and growth. The aim of this study is to identify TSCs in GCTB. METHODS Stro-1(+) and Stro-1(-) cells were isolated by fluorescence-activated cell sorting (FACS). Stem-like properties of both Stro-1(+) and Stro-1(-) subpopulations were assessed using MTT colorimetric assays, cell cycle analyses, sphere formation assays, and differentiation assays. Molecular profiles were analyzed by flow cytometry, immunofluorescence, and qRT-PCR. RESULTS The existence of rare Stro-1(+) cells was confirmed in vitro using FACS and in vivo by immunohistochemistry. These Stro-1(+) cells exhibited higher proliferative and cisplatin-resistant potentials than Stro-1(-) cells. In serum-free suspension cultures, Stro-1(+) SCs could form cell spheres and maintain self-renewal. Furthermore, Stro-1(+) SCs could differentiate into two mesenchymal lineage cells: osteoblasts and adipocytes. Cell surface markers CD44, CD117, and CD133 and stem cell-associated genes OCT3/4, NANOG, and ABCG2 were significantly higher in the Stro-1(+) subpopulation. CONCLUSIONS This study demonstrates that Stro-1(+) SCs in GCTB possess stem-like biological and molecular phenotypes, indicating that they are the TSCs of GCTB.
Collapse
Affiliation(s)
- Jie Lan
- Department of Orthopaedics, Peking University Third Hospital, Beijing, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Yang X, Li Y, Huang Q, Yang J, Shen B, Pei F. Evaluation of a biodegradable graft substitute in rabbit bone defect model. Indian J Orthop 2012; 46:266-73. [PMID: 22719111 PMCID: PMC3377135 DOI: 10.4103/0019-5413.96371] [Citation(s) in RCA: 4] [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] [Indexed: 02/05/2023]
Abstract
OBJECTIVE To evaluate a new biodegradable copolymer calcium sulfate/poly amino acid (CS/PAA) as a graft substitute for the repair of the surgically created cancellous bone defects in rabbits and its biological properties in vivo. MATERIALS AND METHODS Cancellous bone defects were created by drilling holes in the unilateral lateral aspect of the femoral condyle of New Zealand white rabbits. Three groups were assigned: Group A rabbits were grafted with 80% CS/PAA and group B rabbits were grafted with 95% CS/PAA as two treatment groups; group C was sham-operation control group. To study the osteogenic capability in vivo, specimens were harvested at 4, 8, 12, and 16 weeks after implantation and were evaluated by gross assessment, X-ray, histological examination, and histomorphometry. In order to identify the molecular mechanism of bone defect repair, the expression of bone morphogenetic protein-2 (BMP-2) and vascular endothelial growth factor (VEGF) was detected using Western blot at 4 weeks. RESULTS Group A and group B showed more vigorous and rapid repair leading to regeneration of cancellous bone than sham-operation control group on gross observation, radiology, and histomorphometry. There was no significant difference between groups A and B. Morphological observation and histological examination showed that the copolymers degraded in sync with the new bone formation process. The expression of BMP-2 and VEGF in implantation groups was higher than that in control group by western blot. CONCLUSION These findings demonstrated that the novel biodegradable copolymers can repair large areas of cancellous bone defects. With its controllable degradation rate, it suggests that CS/PAA may be a series of useful therapeutic substitute for bone defects.
Collapse
Affiliation(s)
- XiaoBo Yang
- Department of Orthopaedics, West China Hospital, Sichuan University, Chengdu, China
| | - Yong Li
- Department of Orthopaedics, West China Hospital, Sichuan University, Chengdu, China
| | - Qiang Huang
- Department of Orthopaedics, West China Hospital, Sichuan University, Chengdu, China
| | - Jing Yang
- Department of Orthopaedics, West China Hospital, Sichuan University, Chengdu, China
| | - Bing Shen
- Department of Orthopaedics, West China Hospital, Sichuan University, Chengdu, China
| | - FuXing Pei
- Department of Orthopaedics, West China Hospital, Sichuan University, Chengdu, China,Address for correspondence: Prof. FuXing Pei, Director of the Orthopaedics Department, West China Hospital, Sichuan University, No. 37, Guoxuexiang Street, Chengdu - 610 041, China. E-mail:
| |
Collapse
|