1
|
Lacinski RA, Dziadowicz SA, Roth CA, Ma L, Melemai VK, Fitzpatrick B, Chaharbakhshi E, Heim T, Lohse I, Schoedel KE, Hu G, Llosa NJ, Weiss KR, Lindsey BA. Proteomic and transcriptomic analyses identify apo-transcobalamin-II as a biomarker of overall survival in osteosarcoma. Front Oncol 2024; 14:1417459. [PMID: 39493449 PMCID: PMC11527601 DOI: 10.3389/fonc.2024.1417459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 09/17/2024] [Indexed: 11/05/2024] Open
Abstract
Background The large-scale proteomic platform known as the SomaScan® assay is capable of simultaneously measuring thousands of proteins in patient specimens through next-generation aptamer-based multiplexed technology. While previous studies have utilized patient peripheral blood to suggest serum biomarkers of prognostic or diagnostic value in osteosarcoma (OSA), the most common primary pediatric bone cancer, they have ultimately been limited in the robustness of their analyses. We propose utilizing this aptamer-based technology to describe the systemic proteomic milieu in patients diagnosed with this disease. Methods To determine novel biomarkers associated with overall survival in OSA, we deployed the SomaLogic SomaScan® 7k assay to investigate the plasma proteomic profile of naive primary, recurrent, and metastatic OSA patients. Following identification of differentially expressed proteins (DEPs) between 2-year deceased and survivor cohorts, publicly available databases including Survival Genie, TIGER, and KM Plotter Immunotherapy, among others, were utilized to investigate the significance of our proteomic findings. Results Apo-transcobalamin-II (APO-TCN2) was identified as the most DEP between 2-year deceased and survivor cohorts (Log2 fold change = 6.8, P-value = 0.0017). Survival analysis using the Survival Genie web-based platform indicated that increased intratumoral TCN2 expression was associated with better overall survival in both OSA (TARGET-OS) and sarcoma (TCGA-SARC) datasets. Cell-cell communication analysis using the TIGER database suggested that TCN2+ Myeloid cells likely interact with marginal zone and immunoglobin-producing B lymphocytes expressing the TCN2 receptor (CD320) to promote their proliferation and survival in both non-small cell lung cancer and melanoma tumors. Analysis of publicly available OSA scRNA-sequencing datasets identified similar populations in naive primary tumors. Furthermore, circulating APO-TCN2 levels in OSA were then associated with a plasma proteomic profile likely necessary for robust B lymphocyte proliferation, infiltration, and formation of intratumoral tertiary lymphoid structures for improved anti-tumor immunity. Conclusions Overall, APO-TCN2, a circulatory protein previously described in various lymphoproliferative disorders, was associated with 2-year survival status in patients diagnosed with OSA. The relevance of this protein and apparent immunological function (anti-tumor B lymphocyte responses) was suggested using publicly available solid tumor RNA-sequencing datasets. Further studies characterizing the biological function of APO-TCN2 and its relevance in these diseases is warranted.
Collapse
Affiliation(s)
- Ryan A. Lacinski
- Department of Orthopaedics, West Virginia University School of Medicine, Morgantown, WV, United States
- West Virginia University Cancer Institute, West Virginia University School of Medicine, Morgantown, WV, United States
| | - Sebastian A. Dziadowicz
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States
- Bioinformatics Core, West Virginia University School of Medicine, Morgantown, WV, United States
| | - Clark A. Roth
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Li Ma
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States
- Bioinformatics Core, West Virginia University School of Medicine, Morgantown, WV, United States
| | - Vincent K. Melemai
- Department of Orthopaedics, West Virginia University School of Medicine, Morgantown, WV, United States
| | - Brody Fitzpatrick
- Department of Orthopaedics, West Virginia University School of Medicine, Morgantown, WV, United States
| | - Edwin Chaharbakhshi
- Department of Orthopaedics, West Virginia University School of Medicine, Morgantown, WV, United States
| | - Tanya Heim
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Ines Lohse
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Karen E. Schoedel
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Gangqing Hu
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States
- Bioinformatics Core, West Virginia University School of Medicine, Morgantown, WV, United States
| | - Nicolas J. Llosa
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Kurt R. Weiss
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Brock A. Lindsey
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
2
|
Zhou H, Dong Y, Alhaskawi A, Lai J, Wang Z, Ezzi SHA, Kota VG, Abdulla MHAH, Sun Z, Lu H. The Roles of TNF Signaling Pathways in Metabolism of Bone Tumors. Front Pharmacol 2022; 13:907629. [PMID: 35847045 PMCID: PMC9277014 DOI: 10.3389/fphar.2022.907629] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/30/2022] [Indexed: 12/15/2022] Open
Abstract
The metabolism of bone tumors is extraordinarily complex and involves many signaling pathways and processes, including the tumor necrosis factor (TNF) signaling pathway, which consists of TNF factors and the TNF receptors that belong to the TNF receptor superfamily (TNFRSF). It is appreciated that signaling events and pathways involving TNFRSF components are essential in coordinating the functions of multiple cell types that act as a host defense network against pathogens and malignant cells, the implications of TNFRSF-related signaling pathways on bone tumor metabolism remain to be summarized, which is one of the significant obstacles to the application of TNF-related treatment modalities in the domain of bone oncology. This review will discuss and summarize the anti-tumor properties of important TNFRSF components concerning osteosarcoma, chondrosarcoma, and Ewing sarcoma.
Collapse
Affiliation(s)
- Haiying Zhou
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Yanzhao Dong
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Ahmad Alhaskawi
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Jingtian Lai
- Zhejiang University School of Medicine, Hangzhou, China
| | - Zewei Wang
- Zhejiang University School of Medicine, Hangzhou, China
| | | | | | | | - Zhenyu Sun
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Hui Lu
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
- Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Zhejiang University, Hangzhou, China
| |
Collapse
|
3
|
Li Z, Zhou C, Peng Q, Wang S, Qian G, Tang L, Zhou X, Yang Q, Shen Z, Huang G, Wang Y, Li H. Fibrinogen–Albumin Ratio Index Exhibits Predictive Value of Neoadjuvant Chemotherapy in Osteosarcoma. Cancer Manag Res 2022; 14:1671-1682. [PMID: 35547600 PMCID: PMC9084387 DOI: 10.2147/cmar.s358310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 04/15/2022] [Indexed: 11/23/2022] Open
Affiliation(s)
- Zhendong Li
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, People’s Republic of China
| | - Chenliang Zhou
- Department of Oncology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, People’s Republic of China
| | - Qing Peng
- Department of VIP Clinic, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, People’s Republic of China
| | - Suguo Wang
- Department of VIP Clinic, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, People’s Republic of China
| | - Guowei Qian
- Department of Oncology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, People’s Republic of China
| | - Lina Tang
- Department of Oncology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, People’s Republic of China
| | - Xin Zhou
- Cancer Research Institute of Jilin University, the First Hospital of Jilin University, Changchun, Jilin, 130021, People’s Republic of China
| | - Qingcheng Yang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, People’s Republic of China
| | - Zan Shen
- Department of Oncology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, People’s Republic of China
| | - GaoZhong Huang
- Department of VIP Clinic, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, People’s Republic of China
| | - Yonggang Wang
- Department of Oncology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, People’s Republic of China
- Correspondence: Yonggang Wang; Hongtao Li, Department of Oncology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, No. 600, Yishan Road, Xu Hui District, Shanghai, 200233, People’s Republic of China, Tel +086-021-24058408, Email ;
| | - Hongtao Li
- Department of Oncology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, People’s Republic of China
| |
Collapse
|
4
|
The smac mimetic LCL161 targets established pulmonary osteosarcoma metastases in mice. Clin Exp Metastasis 2021; 38:441-449. [PMID: 34398333 DOI: 10.1007/s10585-021-10116-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 08/05/2021] [Indexed: 10/20/2022]
Abstract
Osteosarcoma is the most common form of primary bone cancer and frequently metastasizes to the lungs. Current therapies fail to successfully treat over two thirds of patients with metastatic osteosarcoma, so there is an urgent imperative to develop therapies that effectively target established metastases. Smac mimetics are drugs that work by inhibiting the pro-survival activity of IAP proteins such as cIAP1 and cIAP2, which can be overexpressed in osteosarcomas. In vitro, osteosarcoma cells are sensitive to a range of Smac mimetics in combination with TNFα. This sensitivity has also been demonstrated in vivo using the Smac mimetic LCL161, which inhibited the growth of subcutaneous and intramuscular osteosarcomas. Here, we evaluated the efficacy of LCL161 using mice bearing osteosarcoma metastases without the presence of a primary tumor, modeling the scenario in which a patient's primary tumor had been surgically removed. We demonstrated the ability of LCL161 as a single agent and in combination with doxorubicin to inhibit the growth of, and in some cases eliminate, established pulmonary osteosarcoma metastases in vivo. Resected lung metastases from treated and untreated mice remained sensitive to LCL161 in combination with TNFα ex vivo. This suggested that there was little to no acquired resistance to LCL161 treatment in surviving osteosarcoma cells and implied that tumor microenvironmental factors underlie the observed variation in responses to LCL161.
Collapse
|
5
|
Yapar A, Tokgöz MA, Yapar D, Atalay İB, Ulucaköy C, Güngör BŞ. Diagnostic and prognostic role of neutrophil/lymphocyte ratio, platelet/lymphocyte ratio, and lymphocyte/monocyte ratio in patients with osteosarcoma. Jt Dis Relat Surg 2021; 32:489-496. [PMID: 34145828 PMCID: PMC8343865 DOI: 10.52312/jdrs.2021.79775] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 12/11/2020] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVES This study aims to evaluate the diagnostic and prognostic significance of neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), and lymphocyte-to-monocyte ratio (LMR) values in patients with osteosarcoma. PATIENTS AND METHODS A total of 172 patients (111 males, 61 females; mean age: 24.3±15.3 years; range, 7 to 82 years) diagnosed with osteosarcoma in our institution between January 2002 and December 2018 were retrospectively analyzed. A total of 165 healthy individuals (115 males, 50 females; mean age: 20.2±9.2 years; range, 10 to 65 years) who did not have infectious, rheumatological or hematological diseases or any pathological finding were assigned as the control group. The clinical, laboratory, and demographic findings of the patients were obtained from hospital records. Pre-treatment NLR, PLR, and LMR values were calculated in all patients. Diagnostic and prognostic values of pre-treatment NLR, PLR and LMR were assessed using receiver operating curve (ROC) analysis. The Kaplan-Meier method was used for survival analysis. RESULTS For diagnostic approach, the highest significance in area under the curve (AUC) values was obtained for NLR (AUC=0.763). The AUC for PLR and LMR was statistically significant, while the statistical power was weak compared to NLR (AUC=0.681 and 0.603). The NLR, PLR, and LMR were found to be predictors of mortality. The cut-off value was found to be 3.28 for NLR, 128 for PLR, and 4.22 for LMR. The prognostic value of NLR for mortality was higher than (AUC=0.749) PLR (AUC=0.688) and LMR (AUC=0.609). The NLR, PLR, and LMR were associated with overall survival (OS). There was a significant difference in the median OS time among the NLR, PLR, and LMR values (log-rank test order p<0.001, p=0.001, and p=0.004, respectively). CONCLUSION Based on our study results, pre-treatment NLR, PLR and MLR have diagnostic and prognostic values in osteosarcoma.
Collapse
Affiliation(s)
| | | | | | | | - Coşkun Ulucaköy
- SBÜ Dr. Abdurrahman Yurtaslan Onkoloji Eğitim ve Araştırma Hastanesi Ortopedi ve Travmatoloji Kliniği, 06105 Yenimahalle, Ankara, Türkiye.
| | | |
Collapse
|
6
|
Development of Secondary Osteosarcoma After TBI and Allogeneic Bone Marrow Transplant: A Case Series of 3 Patients. J Pediatr Hematol Oncol 2020; 42:e100-e103. [PMID: 30807391 DOI: 10.1097/mph.0000000000001442] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Osteosarcoma can rarely occur as a subsequent malignant neoplasm after cancer therapy. Children who underwent treatment for cancer and received an allogeneic hematopoietic cell transplant are at a higher risk to develop secondary malignancies. Radiation is also a known risk factor, but estimating the quantitative risk is difficult due to the rarity of the condition and long latency period between primary and secondary cancer. In this report, we present 3 patients diagnosed with leukemia as young children who received hematopoietic cell transplants with total body irradiation as part of the conditioning regimen, and later went on to develop secondary osteosarcoma.
Collapse
|
7
|
Cappariello A, Rucci N. Tumour-Derived Extracellular Vesicles (EVs): A Dangerous "Message in A Bottle" for Bone. Int J Mol Sci 2019; 20:E4805. [PMID: 31569680 PMCID: PMC6802008 DOI: 10.3390/ijms20194805] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 09/19/2019] [Accepted: 09/20/2019] [Indexed: 12/14/2022] Open
Abstract
Several studies have shown the importance of Extracellular Vesicles (EVs) in the intercellular communication between tumour and resident cells. Through EVs, tumour cells can trigger cell-signalling molecules and shuttle exogenous information to target cells, thus promoting spread of the disease. In fact, many processes are fuelled by EVs, such as tumour invasion and dormancy, drug-resistance, immune-surveillance escape, extravasation, extracellular matrix remodelling and metastasis. A key element is certainly the molecular profile of the shed cargo. Understanding the biochemical basis of EVs would help to predict the ability and propensity of cancer cells to metastasize a specific tissue, with the aim to target the release of EVs and to manipulate their content as a possible therapeutic approach. Moreover, EV profiling could help monitor the progression of cancer, providing a useful tool for more effective therapy. This review will focus on all the EV-mediated mentioned mechanisms in the context of both primary bone cancers and bone metastases.
Collapse
Affiliation(s)
- Alfredo Cappariello
- Department of Onco-haematology IRCCS Bambino Gesù Children's Hospital, 00152 Rome, Italy.
| | - Nadia Rucci
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy.
| |
Collapse
|
8
|
Shekhar TM, Burvenich IJG, Harris MA, Rigopoulos A, Zanker D, Spurling A, Parker BS, Walkley CR, Scott AM, Hawkins CJ. Smac mimetics LCL161 and GDC-0152 inhibit osteosarcoma growth and metastasis in mice. BMC Cancer 2019; 19:924. [PMID: 31521127 PMCID: PMC6744692 DOI: 10.1186/s12885-019-6103-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 08/28/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Current therapies fail to cure over a third of osteosarcoma patients and around three quarters of those with metastatic disease. "Smac mimetics" (also known as "IAP antagonists") are a new class of anti-cancer agents. Previous work revealed that cells from murine osteosarcomas were efficiently sensitized by physiologically achievable concentrations of some Smac mimetics (including GDC-0152 and LCL161) to killing by the inflammatory cytokine TNFα in vitro, but survived exposure to Smac mimetics as sole agents. METHODS Nude mice were subcutaneously or intramuscularly implanted with luciferase-expressing murine 1029H or human KRIB osteosarcoma cells. The impacts of treatment with GDC-0152, LCL161 and/or doxorubicin were assessed by caliper measurements, bioluminescence, 18FDG-PET and MRI imaging, and by weighing resected tumors at the experimental endpoint. Metastatic burden was examined by quantitative PCR, through amplification of a region of the luciferase gene from lung DNA. ATP levels in treated and untreated osteosarcoma cells were compared to assess in vitro sensitivity. Immunophenotyping of cells within treated and untreated tumors was performed by flow cytometry, and TNFα levels in blood and tumors were measured using cytokine bead arrays. RESULTS Treatment with GDC-0152 or LCL161 suppressed the growth of subcutaneously or intramuscularly implanted osteosarcomas. In both models, co-treatment with doxorubicin and Smac mimetics impeded average osteosarcoma growth to a greater extent than either drug alone, although these differences were not statistically significant. Co-treatments were also more toxic. Co-treatment with LCL161 and doxorubicin was particularly effective in the KRIB intramuscular model, impeding primary tumor growth and delaying or preventing metastasis. Although the Smac mimetics were effective in vivo, in vitro they only efficiently killed osteosarcoma cells when TNFα was supplied. Implanted tumors contained high levels of TNFα, produced by infiltrating immune cells. Spontaneous osteosarcomas that arose in genetically-engineered immunocompetent mice also contained abundant TNFα. CONCLUSIONS These data imply that Smac mimetics can cooperate with TNFα secreted by tumor-associated immune cells to kill osteosarcoma cells in vivo. Smac mimetics may therefore benefit osteosarcoma patients whose tumors contain Smac mimetic-responsive cancer cells and TNFα-producing infiltrating cells.
Collapse
Affiliation(s)
- Tanmay M. Shekhar
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria 3086 Australia
| | - Ingrid J. G. Burvenich
- Tumour Targeting Laboratory, Ludwig Institute for Cancer Research and Olivia Newton-John Cancer Research Institute, Melbourne, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, Australia
| | - Michael A. Harris
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria 3086 Australia
| | - Angela Rigopoulos
- Tumour Targeting Laboratory, Ludwig Institute for Cancer Research and Olivia Newton-John Cancer Research Institute, Melbourne, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, Australia
| | - Damien Zanker
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria 3086 Australia
| | - Alex Spurling
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria 3086 Australia
| | - Belinda S. Parker
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria 3086 Australia
| | - Carl R. Walkley
- St. Vincent’s Institute, Fitzroy, Victoria 3065 Australia
- Department of Medicine, St. Vincent’s Hospital, University of Melbourne, Fitzroy, Victoria 3065 Australia
- Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, Victoria 3000 Australia
| | - Andrew M. Scott
- Tumour Targeting Laboratory, Ludwig Institute for Cancer Research and Olivia Newton-John Cancer Research Institute, Melbourne, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, Australia
- Departments of Medical Oncology and Molecular Imaging & Therapy, Austin Health, Heidelberg, Melbourne, Australia
- Department of Medicine, University of Melbourne, Melbourne, Australia
| | - Christine J. Hawkins
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria 3086 Australia
| |
Collapse
|
9
|
Tan GJS, Gerrand CH, Rankin KS. Blood-borne biomarkers of osteosarcoma: A systematic review. Pediatr Blood Cancer 2019; 66:e27462. [PMID: 30251311 DOI: 10.1002/pbc.27462] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 08/29/2018] [Accepted: 08/30/2018] [Indexed: 12/27/2022]
Abstract
Osteosarcoma is the most common type of primary malignant bone tumor in children and young adults. Development of clinically useful biomarkers has the potential to improve treatments. The aim of this review was to investigate the recent literature assessing the utility of biomarkers for osteosarcoma. A detailed literature search was performed, with hand searches for related research publications. The search was limited to publications in English between January 2007 and February 2017. Of 286 studies identified, 24 met the inclusion criteria. There is a wide range of osteosarcoma biomarkers identified which act as clinical prognostic factors in patient outcome.
Collapse
Affiliation(s)
- Gerald J S Tan
- The Ipswich Hospital, East Suffolk and North Essex NHS Foundation Trust, Ipswich, United Kingdom
| | - C H Gerrand
- The London Sarcoma Service, Royal National Orthopaedic Hospital, Stanmore, United Kingdom
| | - K S Rankin
- North of England Bone and Soft Tissue Tumour Service, Freeman Hospital, Newcastle Upon Tyne, United Kingdom
| |
Collapse
|
10
|
Brady JV, Troyer RM, Ramsey SA, Leeper H, Yang L, Maier CS, Goodall CP, Ruby CE, Albarqi HAM, Taratula O, Bracha S. A Preliminary Proteomic Investigation of Circulating Exosomes and Discovery of Biomarkers Associated with the Progression of Osteosarcoma in a Clinical Model of Spontaneous Disease. Transl Oncol 2018; 11:1137-1146. [PMID: 30053712 PMCID: PMC6077151 DOI: 10.1016/j.tranon.2018.07.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 07/02/2018] [Accepted: 07/06/2018] [Indexed: 12/21/2022] Open
Abstract
Circulating cancer exosomes are microvesicles which originate from malignant cells and other organs influenced by the disease and can be found in blood. The exosomal proteomic cargo can often be traced to the cells from which they originated, reflecting the physiological status of these cells. The similarities between cancer exosomes and the tumor cells they originate from exhibit the potential of these vesicles as an invaluable target for liquid biopsies. Exosomes were isolated from the serum of eight osteosarcoma-bearing dogs, five healthy dogs, and five dogs with traumatic fractures. We also characterized exosomes which were collected longitudinally from patients with osteosarcoma prior and 2 weeks after amputation, and eventually upon detection of lung metastasis. Exosomal proteins fraction were analyzed by label-free mass spectrometry proteomics and were validated with immunoblots of selected proteins. Ten exosomal proteins were found that collectively discriminate serum of osteosarcoma patients from serum healthy or fractured dogs with an accuracy of 85%. Additionally, serum from different disease stages could be distinguished with an accuracy of 77% based on exosomal proteomic composition. The most discriminating protein changes for both sample group comparisons were related to complement regulation, suggesting an immune evasion mechanism in early stages of osteosarcoma as well as in advanced disease.
Collapse
Affiliation(s)
- Jacqueline V Brady
- Carlson College of Veterinary Medicine, Department of Clinical Sciences, Oregon State University, Corvallis, OR, USA
| | - Ryan M Troyer
- Carlson College of Veterinary Medicine, Department of Clinical Sciences, Oregon State University, Corvallis, OR, USA
| | - Stephen A Ramsey
- Carlson College of Veterinary Medicine, Department of Biomedical Sciences, Oregon State University, Corvallis, OR, USA
| | - Haley Leeper
- Carlson College of Veterinary Medicine, Department of Clinical Sciences, Oregon State University, Corvallis, OR, USA
| | - Liping Yang
- College of Science, Department of Chemistry, Oregon State University, Corvallis, OR, USA
| | - Claudia S Maier
- College of Science, Department of Chemistry, Oregon State University, Corvallis, OR, USA
| | - Cheri P Goodall
- Carlson College of Veterinary Medicine, Department of Clinical Sciences, Oregon State University, Corvallis, OR, USA
| | - Carl E Ruby
- Carlson College of Veterinary Medicine, Department of Clinical Sciences, Oregon State University, Corvallis, OR, USA
| | | | - Oleh Taratula
- College of Pharmacy, Oregon State University, Corvallis, OR, USA
| | - Shay Bracha
- Carlson College of Veterinary Medicine, Department of Clinical Sciences, Oregon State University, Corvallis, OR, USA.
| |
Collapse
|
11
|
Hou CH, Yang RS, Tsao YT. Connective tissue growth factor stimulates osteosarcoma cell migration and induces osteosarcoma metastasis by upregulating VCAM-1 expression. Biochem Pharmacol 2018; 155:71-81. [PMID: 29909077 DOI: 10.1016/j.bcp.2018.06.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 06/13/2018] [Indexed: 12/22/2022]
Abstract
Osteosarcoma is the most common bone malignancy that occurs in the young population. After osteosarcoma cells metastasize to the lung, prognosis is very poor owing to difficulties in early diagnosis and effective treatment. Recently, connective tissue growth factor (CTGF) was reported to be a critical contributor to osteosarcoma metastasis. However, the detailed mechanism associated with CTGF-directed migration in bone neoplasms is still mostly unknown. Through the in vivo and in vitro examination of osteosarcoma cells, this study suggests that VCAM-1 up-regulation and increased osteosarcoma cell migration are involved in this process. Antagonizing αvβ3 integrin inhibited cell migration. Moreover, FAK, PI3K, Akt and NF-κB activation were also shown to be involved in CTGF-mediated osteosarcoma metastasis. Taken together, CTGF promotes VCAM-1 production and further induces osteosarcoma metastasis via the αvβ3 integrin/FAK/PI3K/Akt/NF-κB signaling pathway, which could represent a promising clinical target to improve patient outcome.
Collapse
Affiliation(s)
- Chun-Han Hou
- Department of Orthopedic Surgery, National Taiwan University Hospital, NO 1, Jen-Ai Road, Taipei 100, Taiwan.
| | - Rong-Sen Yang
- Department of Orthopedic Surgery, National Taiwan University Hospital, NO 1, Jen-Ai Road, Taipei 100, Taiwan
| | - Ya-Ting Tsao
- Department of Orthopedic Surgery, National Taiwan University Hospital, NO 1, Jen-Ai Road, Taipei 100, Taiwan
| |
Collapse
|
12
|
Shores DR, Everett AD. Children as Biomarker Orphans: Progress in the Field of Pediatric Biomarkers. J Pediatr 2018; 193:14-20.e31. [PMID: 29031860 PMCID: PMC5794519 DOI: 10.1016/j.jpeds.2017.08.077] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 08/04/2017] [Accepted: 08/30/2017] [Indexed: 12/20/2022]
Affiliation(s)
- Darla R Shores
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD.
| | - Allen D Everett
- Division of Cardiology, Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD
| |
Collapse
|
13
|
Evola FR, Costarella L, Pavone V, Caff G, Cannavò L, Sessa A, Avondo S, Sessa G. Biomarkers of Osteosarcoma, Chondrosarcoma, and Ewing Sarcoma. Front Pharmacol 2017; 8:150. [PMID: 28439237 PMCID: PMC5383728 DOI: 10.3389/fphar.2017.00150] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Accepted: 03/09/2017] [Indexed: 02/03/2023] Open
Abstract
Osteosarcoma is the most frequent malignant bone neoplasm, followed by chondrosarcoma and Ewing sarcoma. The diagnosis of bone neoplasms is generally made through histological evaluation of a biopsy. Clinical and radiological features are also important in aiding diagnosis and to complete the staging of bone cancer. In addition to these, there are several non-specific serological or specific molecular markers for bone neoplasms. In bone tumors, molecular markers increase the accuracy of the diagnosis and assist in subtyping bone tumors. Here, we review these markers and discuss their role in the diagnosis and prognosis of the three most frequent malignant bone neoplasms, namely osteosarcoma, chondrosarcoma, and Ewing sarcoma.
Collapse
Affiliation(s)
- Francesco R. Evola
- Clinica Ortopedica, Dipartimento di Chirurgia, Azienda Ospedaliera-Universitaria Policlinico Vittorio Emanuele di CataniaCatania, Italy
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Zhou Y, Zhang W, Tang F, Luo Y, Min L, Zhang W, Shi R, Duan H, Tu C. A case report of apatinib in treating osteosarcoma with pulmonary metastases. Medicine (Baltimore) 2017; 96:e6578. [PMID: 28403086 PMCID: PMC5403083 DOI: 10.1097/md.0000000000006578] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
RATIONALE Osteosarcoma is the most common malignant bone tumor in children and adolescents. Pulmonary metastases lead to a significantly increased risk of death. Apatinib, a new potent oral small-molecule tyrosine kinase inhibitor targeting the intracellular domain of vascular endothelial growth factor receptor 2 (VEGFR-2), shows survival benefits in treating advanced or metastatic gastric adenocarcinoma, non-squamous non-small cell lung cancer and metastatic breast cancer. However, its efficacy in metastatic osteosarcoma has not been reported yet. PATIENT CONCERNS Herein, we presented a 50-year-old man patient who visited hospital due to local bone pain in the left leg. DIAGNOSES He was initially diagnosed with osteoblastic osteosarcoma. INTERVENTIONS The patient suffered repeated resection surgeries but developed multiple lung metastases. Positive staining for CD31, CD34, and VEGFR-2 were detected in the tumor section. As he refused to receive chemotherapy due to concerns regarding the chemotherapy toxicities and sorafenib due to high cost, apatinib was given at a dose of 500 mg daily. OUTCOMES Eleven months following apatinib administration, the patient achieved a partial response according to the RECIST 1.1 standard. No severe toxicity or drug-related side effect was observed during the treatment. LESSONS Therefore, apatinib could be a new option for the treatment of metastatic osteosarcoma. Clinical trials are required to further confirm the efficacy and safety of apatinib in treating pulmonary metastases from osteosarcoma.
Collapse
Affiliation(s)
| | - Wengeng Zhang
- Precision Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | | | - Yi Luo
- Department of Orthopedics
| | - Li Min
- Department of Orthopedics
| | | | | | | | | |
Collapse
|
15
|
Chaiyawat P, Settakorn J, Sangsin A, Teeyakasem P, Klangjorhor J, Soongkhaw A, Pruksakorn D. Exploring targeted therapy of osteosarcoma using proteomics data. Onco Targets Ther 2017; 10:565-577. [PMID: 28203090 PMCID: PMC5295800 DOI: 10.2147/ott.s119993] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Despite multimodal therapeutic treatments of osteosarcoma (OS), some patients develop resistance to currently available regimens and eventually end up with recurrent or metastatic outcomes. Many attempts have been made to discover effective drugs for improving outcome; however, due to the heterogeneity of the disease, new therapeutic options have not yet been identified. This study aims to explore potential targeted therapy related to protein profiles of OS. In this review of proteomics studies, we extracted data on differentially expressed proteins (DEPs) from archived literature in PubMed and our in-house repository. The data were divided into three experimental groups, DEPs in 1) OS/OB: OS vs osteoblastic (OB) cells, 2) metastasis: metastatic vs non-metastatic sublines plus fresh tissues from primary OS with and without pulmonary metastasis, and 3) chemoresistance: spheroid (higher chemoresistance) vs monolayer cells plus fresh tissues from biopsies from good and poor responders. All up-regulated protein entities in the list of DEPs were sorted and cross-referenced with identifiers of targets of US Food and Drug Administration (FDA)-approved agents and chemical inhibitors. We found that many targets of FDA-approved antineoplastic agents, mainly a group of epigenetic regulators, kinases, and proteasomes, were highly expressed in OS cells. Additionally, some overexpressed proteins were targets of FDA-approved non-cancer drugs, including immunosuppressive and antiarrhythmic drugs. The resulting list of chemical agents showed that some transferase enzyme inhibitors might have anticancer activity. We also explored common targets of OS/OB and metastasis groups, including amidophosphoribosyltransferase (PPAT), l-lactate dehydrogenase B chain (LDHB), and pyruvate kinase M2 (PKM2) as well as the common target of all categories, cathepsin D (CTSD). This study demonstrates the benefits of a text mining approach to exploring therapeutic targets related to protein expression patterns. These results suggest possible repurposing of some FDA-approved medicines for the treatment of OS and using chemical inhibitors in drug screening tests.
Collapse
Affiliation(s)
- Parunya Chaiyawat
- Orthopedic Laboratory and Research Netting Center, Department of Orthopedics
| | | | - Apiruk Sangsin
- Orthopedic Laboratory and Research Netting Center, Department of Orthopedics
| | - Pimpisa Teeyakasem
- Orthopedic Laboratory and Research Netting Center, Department of Orthopedics
| | | | | | - Dumnoensun Pruksakorn
- Orthopedic Laboratory and Research Netting Center, Department of Orthopedics; Excellence Center in Osteology Research and Training Center, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
16
|
Simard FA, Richert I, Vandermoeten A, Decouvelaere AV, Michot JP, Caux C, Blay JY, Dutour A. Description of the immune microenvironment of chondrosarcoma and contribution to progression. Oncoimmunology 2016; 6:e1265716. [PMID: 28344871 DOI: 10.1080/2162402x.2016.1265716] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 11/18/2016] [Accepted: 11/21/2016] [Indexed: 01/12/2023] Open
Abstract
Chondrosarcoma (CHS) is a rare bone malignancy characterized by its resistance to conventional systemic and radiation therapies. Whether immunotherapy targeting immune checkpoints may be active in these tumors remains unknown. To explore the role of the immune system in this tumor, we analyzed the immune environment of chondrosarcomas both in human sample, and in a syngeneic rat model, and tested the contribution of T lymphocytes and macrophages in chondrosarcoma progression. Immunohistochemical stainings were performed on human chondrosarcoma samples and on Swarm rat chondrosarcoma (SRC) model. Selective immunodepletion assays were performed in SRC to evaluate immune population's involvement in tumor progression. In human and rat chondrosarcoma, immune infiltrates composed of lymphocytes and macrophages were identified in the peritumoral area. Immune infiltrates composition was found correlated with tumors characteristics and evolution (grade, invasiveness and size). In SRC, selective depletion of T lymphocytes resulted in an accelerated growth rates, whereas depletion of CD163+ macrophages slowed down tumor progression. Splenocytes isolated from CHS-bearing SRC showed a specific cytotoxicity directed against chondrosarcoma cells (27%), which significantly decreased in CD3-depleted SRC (11%). The immune environment contributes to CHS progression in both human and animal models, suggesting that immunomodulatory approaches could be tested in bone chondrosarcoma.
Collapse
Affiliation(s)
- François A Simard
- Cancer Research Center of Lyon, INSERM UMR 1052, CNRS UMR 5286, Centre Léon Berard , Lyon, France
| | - Iseulys Richert
- Cancer Research Center of Lyon, INSERM UMR 1052, CNRS UMR 5286, Centre Léon Berard , Lyon, France
| | - Alexandra Vandermoeten
- SCAR, Université Claude Bernard Lyon1, Faculté de médecine et de pharmacie de Rockefeller , Lyon, France
| | | | | | - Christophe Caux
- Cancer Research Center of Lyon, INSERM UMR 1052, CNRS UMR 5286, Centre Léon Berard , Lyon, France
| | - Jean-Yves Blay
- Medical Oncology Department, Centre Leon Berard , Lyon, France
| | - Aurélie Dutour
- Cancer Research Center of Lyon, INSERM UMR 1052, CNRS UMR 5286, Centre Léon Berard , Lyon, France
| |
Collapse
|
17
|
Conry RM, Rodriguez MG, Pressey JG. Zoledronic acid in metastatic osteosarcoma: encouraging progression free survival in four consecutive patients. Clin Sarcoma Res 2016; 6:6. [PMID: 27127605 PMCID: PMC4848872 DOI: 10.1186/s13569-016-0046-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Accepted: 03/29/2016] [Indexed: 12/29/2022] Open
Abstract
Background Zoledronic acid (ZA) is a third-generation bisphosphonate in widespread clinical use to reduce pain and skeletal events in patients from a variety of malignancies with bone metastases. Pre-clinical studies indicate that ZA inhibits osteosarcoma through direct anti-proliferative effects, immune activation and anti-angiogenic activity. Methods The purpose of this study was to evaluate the antitumor efficacy of ZA at standard dose until progression in patients with stage IV osteosarcoma lacking a standard of care treatment option proven to influence survival. Researchers retrospectively reviewed medical records of all patients at our institution with high-grade osteosarcoma presumed to be incurable due to metastases progressive after primary combination chemotherapy who received single agent ZA in an effort to delay progression. Results In our four-patient cohort following initiation of ZA, the median progression-free survival was 19 months, and median overall survival was 56+ months. Two of four patients have remained progression-free since starting ZA. The other two initially progressed after 18–20 months on ZA followed by metastasectomy of lung or dural metastases and further stability for over a year following resumption of ZA. After a 20-month progression-free interval on ZA alone, one patient had partial response following addition of pazopanib to ZA that likely contributed to long term disease control. The four patients experienced no significant toxicities despite protracted dosing of ZA for up to 5 years, and none have required chemotherapy since beginning ZA. Conclusions Single agent ZA was associated with encouraging progression-free survival in four consecutive patients with metastatic osteosarcoma. Prospective trials of single agent ZA are warranted as protracted maintenance therapy in surgically incurable osteosarcoma relapsed or refractory to first line combination chemotherapy with radiographically measurable metastases.
Collapse
Affiliation(s)
- Robert M Conry
- Division of Hematology Oncology, University of Alabama at Birmingham, 2145 Bonner Way, Birmingham, AL 35243 USA
| | - Michael G Rodriguez
- Department of Radiology, University of Alabama at Birmingham, 619 19th St South, Birmingham, AL 35249 USA
| | - Joseph G Pressey
- Department of Pediatrics, University of Alabama at Birmingham, 1600 7th Avenue South, Birmingham, AL 35233 USA ; Cancer & Blood Disorders Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH USA
| |
Collapse
|
18
|
Hu ZB, Liao XH, Xu ZY, Yang X, Dong C, Jin AM, Lu H. PLK2 phosphorylates and inhibits enriched TAp73 in human osteosarcoma cells. Cancer Med 2015; 5:74-87. [PMID: 26625870 PMCID: PMC4708894 DOI: 10.1002/cam4.558] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 09/12/2015] [Accepted: 09/14/2015] [Indexed: 01/01/2023] Open
Abstract
TAp73, a member of the p53 tumor suppressor family, can substitute for p53 function, especially in p53‐null and p53‐mutant cells. However, TAp73 enrichment and phosphorylation change its transcriptional activity. Previously, we found that the antitumor function of TAp73 was reactivated by dephosphorylation. Polo‐like kinase 2 (PLK2) plays an important role in bone development. Using a biological information database and phosphorylation prediction software, we hypothesized that PLK2 phosphorylates TAp73 and inhibits TAp73 function in osteosarcomas. Actually,we determined that PLK2 physically binds to and phosphorylates TAp73 when TAp73 protein abundance is up‐regulated by cisplatin. PLK2‐phosphorylated TAp73 at residue Ser48 within the TA domain; phosphorylation of TAp73 was abolished by mutating this residue. Moreover, PLK2 inhibition combined with cisplatin treatment in osteosarcoma Saos2 cells up‐regulated p21 and puma mRNA expression to a greater extent than cisplatin treatment alone. Inhibiting PLK2 in TAp73‐enriched Saos2 cells resulted in inhibited cell proliferation, increased apoptosis, G1 phase arrest, and decreased cell invasion. However, these changes did not occur in TAp73 knockdown Saos2 cells. In conclusion, these findings reveal a novel PLK2 function in the phosphorylation of TAp73, which prevents TAp73 activity in osteosarcoma cells. Thereby, this research provides an insight into the clinical treatment of malignant tumors overexpressing TAp73.
Collapse
Affiliation(s)
- Zheng Bo Hu
- Department of Orthopedics, Zhu Jiang Hospital of Southern Medical University, Guangzhou, Guangdong, 510280, China
| | - Xiao Hong Liao
- The State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510280, China
| | - Zun Ying Xu
- Department of Orthopedics, Zhu Jiang Hospital of Southern Medical University, Guangzhou, Guangdong, 510280, China
| | - Xiao Yang
- Department of Orthopedics, Zhu Jiang Hospital of Southern Medical University, Guangzhou, Guangdong, 510280, China
| | - Chao Dong
- Department of Orthopedics, Zhu Jiang Hospital of Southern Medical University, Guangzhou, Guangdong, 510280, China
| | - An Min Jin
- Department of Orthopedics, Zhu Jiang Hospital of Southern Medical University, Guangzhou, Guangdong, 510280, China
| | - Hai Lu
- Department of Orthopaedics, The Third Affiliated Hospital of Southern Medical University, Academy of Orthopedics, Guangzhou, Guangdong, 510665, China
| |
Collapse
|
19
|
Hu T, Yang Q, Xu J, Zhang Z, He N, Du Y. Role of β-isomerized C-terminal telopeptides (β-CTx) and total procollagen type 1 amino-terminal propeptide (tP1NP) as osteosarcoma biomarkers. Int J Clin Exp Med 2015; 8:890-896. [PMID: 25785071 PMCID: PMC4358526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Accepted: 01/08/2015] [Indexed: 06/04/2023]
Abstract
INTRODUCTION Few serum markers are valid and useful for the diagnosis or therapeutic effect monitoring of osteosarcoma. This study aimed to investigate the role of β-isomerized C-terminal telopeptides (β-CTx) and total procollagen type 1 amino-terminal propeptide (tP1NP) as serological biomarkers for osteosarcoma patients. MATERIALS AND METHODS A total of 48 patients with osteosarcoma and 55 healthy volunteers were investigated. Serum β-CTx and tP1NP levels were measured by electrochemiluminescence immunoassay. Data were analyzed by t test with Walth's correction and receiver operating characteristic (ROC) curve analysis. RESULTS The baseline levels of β-CTx and tP1NP were found to be significantly higher in patients with osteosarcoma than the healthy volunteers. The mean areas under the ROC curves were 0.919 (range, 0.864-0.973) for β-CTx and 0.866 (range, 0.792-0.939) for tP1NP. The levels of β-CTx and tP1NP were lower in patients with stable disease after operation than those before operation. CONCLUSION These findings support our hypothesis that β-CTx and tP1NP are promising serum biomarkers for diagnosing or monitoring osteosarcoma.
Collapse
Affiliation(s)
- Tu Hu
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People’s HospitalShanghai, China
| | - Qingcheng Yang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People’s HospitalShanghai, China
| | - Jing Xu
- Department of Laboratory Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s HospitalShanghai, China
| | - Zhichang Zhang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People’s HospitalShanghai, China
| | - Nengbin He
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People’s HospitalShanghai, China
| | - Yuzhen Du
- Department of Laboratory Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s HospitalShanghai, China
| |
Collapse
|
20
|
Hsieh IS, Yang RS, Fu WM. Osteopontin upregulates the expression of glucose transporters in osteosarcoma cells. PLoS One 2014; 9:e109550. [PMID: 25310823 PMCID: PMC4195676 DOI: 10.1371/journal.pone.0109550] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2014] [Accepted: 09/11/2014] [Indexed: 11/28/2022] Open
Abstract
Osteosarcoma is the most common primary malignancy of bone. Even after the traditional standard surgical therapy, metastasis still occurs in a high percentage of patients. Glucose is an important source of metabolic energy for tumor proliferation and survival. Tumors usually overexpress glucose transporters, especially hypoxia-responsive glucose transporter 1 and glucose transporter 3. Osteopontin, hypoxia-responsive glucose transporter 1, and glucose transporter 3 are overexpressed in many types of tumors and have been linked to tumorigenesis and metastasis. In this study, we investigated the regulation of glucose transporters by osteopontin in osteosarcoma. We observed that both glucose transporters and osteopontin were upregulated in hypoxic human osteosarcoma cells. Endogenously released osteopontin regulated the expression of glucose transporter 1 and glucose transporter 3 in osteosarcoma and enhanced glucose uptake into cells via the αvβ3 integrin. Knockdown of osteopontin induced cell death in 20% of osteosarcoma cells. Phloretin, a glucose transporter inhibitor, also caused cell death by treatment alone. The phloretin-induced cell death was significantly enhanced in osteopontin knockdown osteosarcoma cells. Combination of a low dose of phloretin and chemotherapeutic drugs, such as daunomycin, 5-Fu, etoposide, and methotrexate, exhibited synergistic cytotoxic effects in three osteosarcoma cell lines. Inhibition of glucose transporters markedly potentiated the apoptotic sensitivity of chemotherapeutic drugs in osteosarcoma. These results indicate that the combination of a low dose of a glucose transporter inhibitor with cytotoxic drugs may be beneficial for treating osteosarcoma patients.
Collapse
Affiliation(s)
- I-Shan Hsieh
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Rong-Sen Yang
- Department of Orthopedic Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Wen-Mei Fu
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
21
|
Ohba T, Cates JMM, Cole HA, Slosky DA, Haro H, Ichikawa J, Ando T, Schwartz HS, Schoenecker JG. Pleiotropic effects of bisphosphonates on osteosarcoma. Bone 2014; 63:110-20. [PMID: 24636958 DOI: 10.1016/j.bone.2014.03.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2014] [Revised: 03/06/2014] [Accepted: 03/08/2014] [Indexed: 12/12/2022]
Abstract
Osteosarcoma is the most common primary malignant tumor of bone and accounts for half of all primary skeletal malignancies in children and teenagers. The prognosis for patients who fail or progress on first-line chemotherapy protocols is poor, therefore, additional adjuvant therapeutic strategies are needed. A recent feasibility study has demonstrated that the nitrogen-containing bisphosphonate zoledronic acid (ZOL) can be combined safely with conventional chemotherapy. However, the pharmacodynamics of bisphosphonate therapy is not well characterized. Osteosarcoma is a highly angiogenic tumor. Recent reports of the anti-angiogenic effects of bisphosphonates prompted us to determine whether nitrogen-containing bisphosphonate (ZOL and alendronate) treatment attenuates osteosarcoma growth by inhibition of osteoclast activity, tumor-mediated angiogenesis, or direct inhibitory effects on osteosarcoma. Here, we demonstrate that bisphosphonates directly inhibit VEGFR2 expression in endothelial cells, as well as endothelial cell proliferation and migration. Additionally, bisphosphonates also decrease VEGF-A expression in osteosarcoma (K7M3) cells, resulting in reduced stimulation of endothelial cell migration in co-culture assays. ZOL also decreases VEGFR1 expression in aggressive osteosarcoma cell lines (K7M3, 143B) and induces apoptosis of these cells, but has negligible effects on less aggressive osteosarcoma cell lines (K12 and TE85). In vivo ZOL treatment results in significant reduction in osteosarcoma-initiated angiogenesis and tumor growth in a murine model of osteosarcoma. In conclusion, bisphosphonates have diverse growth inhibitory effects on osteosarcoma through: (1) activation of apoptosis and inhibition of cell proliferation, (2) inhibition of VEGF-A and VEGFR1 expression by tumor cells, (3) inhibition of tumor-induced angiogenesis, and (4) direct inhibitory actions on endothelial cells.
Collapse
Affiliation(s)
- Tetsuro Ohba
- Vanderbilt University Medical Center, Department of Orthopaedics, 2200 Children's Way, Nashville, TN 37232-9565, USA; Department of Orthopaedic Surgery, Faculty of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi 409-3898, Japan
| | - Justin M M Cates
- Vanderbilt University Medical Center, Department of Pathology, Microbiology and Immunology, 2200 Children's Way, Nashville, TN 37232-9565, USA
| | - Heather A Cole
- Vanderbilt University Medical Center, Department of Orthopaedics, 2200 Children's Way, Nashville, TN 37232-9565, USA
| | - David A Slosky
- Vanderbilt University Medical Center, Department of Cardio-Oncology, 2200 Children's Way, Nashville, TN 37232-9565, USA
| | - Hirotaka Haro
- Department of Orthopaedic Surgery, Faculty of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi 409-3898, Japan
| | - Jiro Ichikawa
- Department of Orthopaedic Surgery, Faculty of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi 409-3898, Japan
| | - Takashi Ando
- Department of Orthopaedic Surgery, Faculty of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi 409-3898, Japan
| | - Herbert S Schwartz
- Vanderbilt University Medical Center, Department of Orthopaedics, 2200 Children's Way, Nashville, TN 37232-9565, USA
| | - Jonathan G Schoenecker
- Vanderbilt University Medical Center, Department of Orthopaedics, 2200 Children's Way, Nashville, TN 37232-9565, USA; Vanderbilt University Medical Center, Department of Center for Bone Biology, 2200 Children's Way, Nashville, TN 37232-9565, USA; Vanderbilt University Medical Center, Department of Pathology, Microbiology and Immunology, 2200 Children's Way, Nashville, TN 37232-9565, USA; Vanderbilt University Medical Center, Department of Pharmacology, 2200 Children's Way, Nashville, TN 37232-9565, USA; Vanderbilt University Medical Center, Department of Pediatrics, 2200 Children's Way, Nashville, TN 37232-9565, USA.
| |
Collapse
|
22
|
Ohba T, Cates JM, Cole HA, Slosky DA, Haro H, Ando T, Schwartz HS, Schoenecker JG. Autocrine VEGF/VEGFR1 Signaling in a Subpopulation of Cells Associates with Aggressive Osteosarcoma. Mol Cancer Res 2014; 12:1100-11. [DOI: 10.1158/1541-7786.mcr-14-0037] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
23
|
Xu M, Xu CX, Bi WZ, Song ZG, Jia JP, Chai W, Zhang LH, Wang Y. Effects of endostar combined multidrug chemotherapy in osteosarcoma. Bone 2013; 57:111-5. [PMID: 23912049 DOI: 10.1016/j.bone.2013.07.035] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Revised: 07/04/2013] [Accepted: 07/19/2013] [Indexed: 11/27/2022]
Abstract
Angiogenesis is closely related to tumor development and metastasis. Osteosarcoma is an angiogenesis-dependent tumor, and studies have shown that chemotherapy often induces angiogenesis. Endostatin is a broad spectrum angiogenesis inhibitor and, while pre-clinical trials have shown that the combination of endostatin with chemotherapy can enhance anti-tumor effects, this effect has not yet been shown in clinical trials. Here, we aimed to evaluate the clinical efficacy of endostar (ES, human recombinant endostatin) combined with chemotherapy in the treatment of osteosarcoma patients. A total of 116 newly diagnosed patients with osteosarcoma were enrolled in this study. All patients received 4cycles of chemotherapy with (54 cases) or without (62 cases) ES. ES was administered intravenously at a dose of 15mg/day for 2weeks during each cycle of chemotherapy. The tumors were removed by surgery after 2cycles of chemotherapy treatment, and their histologic response to chemotherapy was evaluated. Immunohistochemistry was used to measure VEGF and CD 31 expression. Chemotherapy increased VEGF expression and the presence of microvessels in osteosarcoma tissues compared with pre-chemotherapy. No significant difference was observed in the histologic response between the ES treatment and non-treatment groups. However, ES treatment significantly inhibited the chemotherapy-induced VEGF expression and presence of microvessels. The ES treatment did not affect the overall survival rate but did increase the event-free survival rate and decreased the occurrence of metastases. In conclusion, our results indicate that antiangiogenic therapy using ES has the potential to prevent the progression of metastases.
Collapse
Affiliation(s)
- Meng Xu
- Department of Orthopaedics, The General Hospital of Chinese People's Liberation Army, Beijing100853, China
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Song SH, Lee H, Song HR, Kim MJ, Park JH. Fibrocartilaginous intramedullary bone forming tumor of the distal femur mimicking osteosarcoma. J Korean Med Sci 2013; 28:631-5. [PMID: 23580060 PMCID: PMC3617321 DOI: 10.3346/jkms.2013.28.4.631] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Accepted: 01/11/2013] [Indexed: 12/19/2022] Open
Abstract
Fibrocartilaginous dysplasia (FCD) has occasionally led to a misdiagnosis and wrong decision which can significantly alter the outcome of the patients. A 9-yr-old boy presented with pain on his left distal thigh for 6 months without any trauma history. Initial radiographs showed moth eaten both osteolytic and osteosclerotic lesions and biopsy findings showed that the lesion revealed many irregular shaped and sclerotic mature and immature bony trabeculae. Initial diagnostic suggestions were varied from the conventional osteosarcoma to low grade central osteosarcoma or benign intramedullary bone forming lesion, but close observation was done. This study demonstrated a case of unusual fibrocartilaginous intramedullary bone forming tumor mimicking osteosarcoma, so that possible misdiagnosis might be made and unnecessary extensive surgical treatment could be performed. In conclusion, the role of orthopaedic oncologist as a decision maker is very important when the diagnosis is uncertain.
Collapse
Affiliation(s)
- Sang-Heon Song
- Department of Orthopaedic Surgery, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Hanna Lee
- Institute for rare diseases and Department of Orthopaedic Surgery, Korea University Medical Center, Guro Hospital, Seoul, Korea
| | - Hae-Ryong Song
- Institute for rare diseases and Department of Orthopaedic Surgery, Korea University Medical Center, Guro Hospital, Seoul, Korea
| | - Myo-Jong Kim
- Department of Orthopaedic Surgery, Korea University Medical Center, Anam Hospital, Seoul, Korea
| | - Jong-Hoon Park
- Department of Orthopaedic Surgery, Korea University Medical Center, Anam Hospital, Seoul, Korea
| |
Collapse
|
25
|
Chen CY, Tsai MM, Chi HC, Lin KH. Biological significance of a thyroid hormone-regulated secretome. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2013; 1834:2271-84. [PMID: 23429180 DOI: 10.1016/j.bbapap.2013.02.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2012] [Revised: 02/07/2013] [Accepted: 02/11/2013] [Indexed: 01/18/2023]
Abstract
The thyroid hormone, 3,3,5-triiodo-L-thyronine (T3), modulates several physiological processes, including cellular growth, differentiation, metabolism and proliferation, via interactions with thyroid hormone response elements (TREs) in the regulatory regions of target genes. Several intracellular and extracellular protein candidates are regulated by T3. Moreover, T3-regulated secreted proteins participate in physiological processes or cellular transformation. T3 has been employed as a marker in several disorders, such as cardiovascular disorder in chronic kidney disease, as well as diseases of the liver, immune system, endocrine hormone metabolism and coronary artery. Our group subsequently showed that T3 regulates several tumor-related secretory proteins, leading to cancer progression via alterations in extracellular matrix proteases and tumor-associated signaling pathways in hepatocellular carcinomas. Therefore, elucidation of T3/thyroid hormone receptor-regulated secretory proteins and their underlying mechanisms in cancers should facilitate the identification of novel therapeutic targets. This review provides a detailed summary on the known secretory proteins regulated by T3 and their physiological significance. This article is part of a Special Issue entitled: An Updated Secretome.
Collapse
Affiliation(s)
- Cheng-Yi Chen
- Department of Biochemistry, College of Medicine, Chang-Gung University, Taoyuan 333, Taiwan
| | | | | | | |
Collapse
|
26
|
Leckenby JI, Grobbelaar AO, Aston W. The use of a free vascularised fibula to reconstruct the radius following the resection of an osteosarcoma in a paediatric patient. J Plast Reconstr Aesthet Surg 2012; 66:427-9. [PMID: 22947773 DOI: 10.1016/j.bjps.2012.08.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2012] [Revised: 08/08/2012] [Accepted: 08/08/2012] [Indexed: 11/17/2022]
Abstract
Osteosarcoma is the most common high grade bone malignancy in children and the surgical treatment traditionally involves amputation. In our case, a 6-year-old girl was diagnosed with an osteosarcoma of the left distal radius after presenting with forearm pain. After initially being offered an amputation, a second opinion was sought and a limb salvage procedure was offered using a free vascularised fibula bone flap. This resulted in limb preserving surgery which allowed the potential for growth with the maximal preservation of function.
Collapse
Affiliation(s)
- J I Leckenby
- Department of Plastic and Reconstructive Surgery, The Royal Free London NHS Foundation Trust, Pond Street, Hampstead, London NW3 2QG, UK.
| | | | | |
Collapse
|