1
|
Li Y, Wu Z, Ding T, Zhang W, Guo H, Huang F. Comprehensive bioinformatics analysis and cell line experiments revealed the important role of CDCA3 in sarcoma. Heliyon 2024; 10:e32785. [PMID: 39035484 PMCID: PMC11259814 DOI: 10.1016/j.heliyon.2024.e32785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 06/03/2024] [Accepted: 06/10/2024] [Indexed: 07/23/2024] Open
Abstract
Background Sarcoma mainly originate from bone and soft tissue and are highly aggressive malignant tumors. Cell division cycle-related protein 3 (CDCA3) is a protein involved in the regulation of the cell cycle, which is highly expressed in a variety of malignant tumors. However, its role in sarcoma remains unclear. This study aims to investigate the function and potential mechanism of CDCA3 in sarcoma and to elucidate its importance in sarcoma. Methods We first studied the expression and prognosis of CDCA family members in sarcoma by Oncomine and the Gene Expression Profiling Interactive Analysis (GEPIA). The role of CDCA3 protein in sarcoma was further analyzed by the Cancer Genome Atlas Program (TCGA), the Cancer Cell Lineage Encyclopedia (CCLE), and Linke-dOmics. In addition, immunohistochemistry and Western blot were used to verify the expression of CDCA3 protein in clinical samples as well as sarcoma cell lines (U2OS, SAOS2, MG63, and HOS). Subsequently, in vitro experiments (cloning and scratching experiments) were performed using sh-NC as well as sh-CDCA3 group cells to reveal the biological functions of CDCA3. Results We found that the CDCA family (CDCA3, CDCA4, and CDCA8) is highly expressed in sarcoma, and the expression level of CDCA3, CDCA4, and CDCA8 negatively correlates with the prognosis of sarcoma patients. CDCA3 mRNA was highly expressed in pan-cancer by CCLE and TCGA database analysis. KEGG analysis showed that CDCA3 was mainly enriched in the cell cycle signaling pathway (It promoted the transition of the cell cycle from the G0/G1 phase to the S phase). In the level of immune infiltration, CDCA3 was negatively correlated with pDC cells, CD8+T cells, and cytotoxic cells. Finally, patients with high CDCA3 expression in sarcoma were analyzed for resistance to NU7441 and others, while sensitive to Fulvestrant and Dihydrorotenone. Furthermore, we demonstrated high expression of CDCA3 protein in sarcoma tissues and cell lines by immunohistochemistry and Western blot experiments. Cloning, EDU, scratching, and migration experiments showed that the knockdown of CDCA3 inhibited the Proliferation and progression of sarcoma cells. Conclusion These results suggest for the first time that knockdown of CDCA3 may inhibit sarcoma progression. CDCA3 may be an effective target for the treatment of sarcoma.
Collapse
Affiliation(s)
- Yang Li
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Zhiwei Wu
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Tao Ding
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Wenbiao Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Hongjuan Guo
- School of Life Sciences, Anhui Medical University, Hefei 230032, China
| | - Fei Huang
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| |
Collapse
|
2
|
Nyström H, Jönsson M, Nilbert M, Carneiro A. Immune-cell infiltration in high-grade soft tissue sarcomas; prognostic implications of tumor-associated macrophages and B-cells. Acta Oncol 2023; 62:33-39. [PMID: 36786033 DOI: 10.1080/0284186x.2023.2172688] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Abstract
BACKGROUND Soft tissue sarcomas are rare, morphologically, and genetically heterogenous. Though the tumors display abundant tumor stroma with infiltrating immune cells, the prognostic impact of various immunologic markers in sarcoma remains poorly defined. We aimed to characterize the immune landscape of a treatment-naïve cohort of soft tissue sarcoma of the extremities and the trunk wall with correlations to metastasis-free survival. MATERIALS AND METHODS We surveyed immunohistochemical expression patterns for CD163, CD20, CD3, CD8, and FOXP3 in 134 adult high-grade leiomyosarcomas, liposarcomas, and synovial sarcomas. RESULTS Macrophages outnumbered tumor-infiltrating lymphocytes. High CD163 infiltration was identified in 49% of the tumors and was overrepresented (66%) in leiomyosarcoma compared to liposarcoma (46%) and synovial sarcoma (9%). Tumor-grade also correlated with CD163 positivity with high expression in 53% of the high-grade lesions and 28% in low-grade tumors. Infiltrating CD3, CD8 and FOXP3-positive T-cells were significantly more prevalent in leiomyosarcomas than in liposarcomas/synovial sarcomas. CD20+ B-cells were identified only in 14% of the STS. Correlation to established prognostic factors revealed a correlation between CD163+ macrophages and necrosis and predicted an increased risk of metastases. No correlation between CD20+ B-cells and known prognostic factors could be established, though CD20+ B-cells infiltration predicted improved overall survival. CONCLUSION We confirm that tumor-infiltrating macrophages outnumber tumor-infiltrating lymphocytes in soft tissue sarcoma and signify an increased risk of metastasis. CD20+ B-cells are scarce in STS and correlate to improved survival. To date, immunotherapeutic strategies directed against T-cells have shown limited effect in soft tissue sarcoma. Our observations suggest that immunomodulatory agents focusing on macrophages may be worthwhile for further investigations in this tumor type. Further studies exploring the prognostic and predictive significance of CD20+ B cells are warranted.
Collapse
Affiliation(s)
- Helena Nyström
- Institute of Clinical Sciences, Division of Oncology and Pathology, Lund University, Lund, Sweden.,Department of Hematology, Oncology and Radiation Physics, Skåne University Hospital, Lund, Sweden
| | - Mats Jönsson
- Institute of Clinical Sciences, Division of Oncology and Pathology, Lund University, Lund, Sweden
| | - Mef Nilbert
- Institute of Clinical Sciences, Division of Oncology and Pathology, Lund University, Lund, Sweden.,Department of Hematology, Oncology and Radiation Physics, Skåne University Hospital, Lund, Sweden
| | - Ana Carneiro
- Institute of Clinical Sciences, Division of Oncology and Pathology, Lund University, Lund, Sweden.,Department of Hematology, Oncology and Radiation Physics, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
3
|
Li X, Hu Y, Xie Y, Lu R, Li Q, Tao H, Chen S. Whole-tumor histogram analysis of diffusion-weighted imaging and dynamic contrast-enhanced MRI for soft tissue sarcoma: correlation with HIF-1alpha expression. Eur Radiol 2022; 33:3961-3973. [PMID: 36462043 DOI: 10.1007/s00330-022-09296-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 09/02/2022] [Accepted: 11/10/2022] [Indexed: 12/04/2022]
Abstract
OBJECTIVE To investigate the correlation of histogram metrics from diffusion-weighted imaging (DWI) and dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) parameters with HIF-1alpha expression in soft tissue sarcoma (STS). METHODS We enrolled 71 patients with STS who underwent 3.0-T MRI, including conventional MRI, DWI, and DCE-MRI sequences. Location, maximum tumor diameter, envelope, T2-weighted tumor heterogeneity, peritumoral edema, peritumoral enhancement, necrosis, tail-like pattern, bone invasion, and vessel/nerve invasion and/or encasement were determined using conventional MRI images. The whole-tumor histogram metrics were calculated on the apparent diffusion coefficient (ADC), Ktrans, Kep, and Ve maps. Independent-samples t test and one-way ANOVA were used for testing the differences between normally distributed categorical data with HIF-1alpha expression. Pearson and Spearman correlations and multiple linear regression analyses were performed to determine the correlations between histogram metrics and HIF-1alpha expression. Survival curves were plotted using the Kaplan-Meier method. RESULTS Regarding conventional MRI features, only highly heterogeneous on T2-weighted images (55.6 ± 19.9% vs. 45.4 ± 20.5%, p = 0.041) and more than 50% necrotic area (57.3 ± 20.4% vs. 43.9 ± 19.7%, p = 0.002) were prone to indicate STS with higher HIF-1alpha expression. Histogram metrics obtained from ADC (mean, median, 10th, and 25th percentile values), Ktrans (mean, median, 75th, and 90th percentile values), and Kep (90th percentile values) were significantly correlated with HIF-1alpha expression. Multiple linear regression analysis demonstrated that more than 50% necrosis, ADCskewness, Ktrans90th, and grade III were independently associated with HIF-1alpha expression. CONCLUSION DWI and DCE-MRI histogram parameters were significantly correlated with HIF-1alpha expression in STS. KEY POINTS • DWI and DCE-MRI histogram parameters are correlated with HIF-1alpha expression in STS. • More than 50% necrosis, ADCskewness, Ktrans90th, and grade III were independently associated with HIF-1alpha expression in STS.
Collapse
Affiliation(s)
- Xiangwen Li
- Department of Radiology and Institute of Medical Functional and Molecular Imaging, Huashan Hospital, Fudan University, 12 Wulumuqizhong Road, Shanghai, China
| | - Yiwen Hu
- Department of Radiology and Institute of Medical Functional and Molecular Imaging, Huashan Hospital, Fudan University, 12 Wulumuqizhong Road, Shanghai, China
| | - Yuxue Xie
- Department of Radiology and Institute of Medical Functional and Molecular Imaging, Huashan Hospital, Fudan University, 12 Wulumuqizhong Road, Shanghai, China
| | - Rong Lu
- Department of Radiology and Institute of Medical Functional and Molecular Imaging, Huashan Hospital, Fudan University, 12 Wulumuqizhong Road, Shanghai, China
| | - Qing Li
- MR Collaborations, Siemens Healthineers Ltd., Shanghai, China
| | - Hongyue Tao
- Department of Radiology and Institute of Medical Functional and Molecular Imaging, Huashan Hospital, Fudan University, 12 Wulumuqizhong Road, Shanghai, China.
| | - Shuang Chen
- Department of Radiology and Institute of Medical Functional and Molecular Imaging, Huashan Hospital, Fudan University, 12 Wulumuqizhong Road, Shanghai, China.
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, 2 middle Wulumuqizhong Road, Shanghai, China.
| |
Collapse
|
4
|
Arthur A, Johnston EW, Winfield JM, Blackledge MD, Jones RL, Huang PH, Messiou C. Virtual Biopsy in Soft Tissue Sarcoma. How Close Are We? Front Oncol 2022; 12:892620. [PMID: 35847882 PMCID: PMC9286756 DOI: 10.3389/fonc.2022.892620] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 05/31/2022] [Indexed: 12/13/2022] Open
Abstract
A shift in radiology to a data-driven specialty has been unlocked by synergistic developments in imaging biomarkers (IB) and computational science. This is advancing the capability to deliver "virtual biopsies" within oncology. The ability to non-invasively probe tumour biology both spatially and temporally would fulfil the potential of imaging to inform management of complex tumours; improving diagnostic accuracy, providing new insights into inter- and intra-tumoral heterogeneity and individualised treatment planning and monitoring. Soft tissue sarcomas (STS) are rare tumours of mesenchymal origin with over 150 histological subtypes and notorious heterogeneity. The combination of inter- and intra-tumoural heterogeneity and the rarity of the disease remain major barriers to effective treatments. We provide an overview of the process of successful IB development, the key imaging and computational advancements in STS including quantitative magnetic resonance imaging, radiomics and artificial intelligence, and the studies to date that have explored the potential biological surrogates to imaging metrics. We discuss the promising future directions of IBs in STS and illustrate how the routine clinical implementation of a virtual biopsy has the potential to revolutionise the management of this group of complex cancers and improve clinical outcomes.
Collapse
Affiliation(s)
- Amani Arthur
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, Sutton, United Kingdom
| | - Edward W. Johnston
- Sarcoma Unit, The Royal Marsden National Health Service (NHS) Foundation Trust, London, United Kingdom
| | - Jessica M. Winfield
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, Sutton, United Kingdom
- Sarcoma Unit, The Royal Marsden National Health Service (NHS) Foundation Trust, London, United Kingdom
| | - Matthew D. Blackledge
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, Sutton, United Kingdom
| | - Robin L. Jones
- Sarcoma Unit, The Royal Marsden National Health Service (NHS) Foundation Trust, London, United Kingdom
- Division of Clinical Studies, The Institute of Cancer Research, London, United Kingdom
| | - Paul H. Huang
- Division of Molecular Pathology, The Institute of Cancer Research, Sutton, United Kingdom
| | - Christina Messiou
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, Sutton, United Kingdom
- Sarcoma Unit, The Royal Marsden National Health Service (NHS) Foundation Trust, London, United Kingdom
| |
Collapse
|
5
|
Angeles CV, Velez A, Rios J, Laxa B, Shum D, Ruiz PD, Shen Y, Ostrovnaya I, Gularte-Mérida R, Nacev BA, Dickson MA, Djaballah H, Okada T, Singer S. A High-Content Screen for C/EBPα Expression Identifies Novel Therapeutic Agents in Dedifferentiated Liposarcoma. Clin Cancer Res 2021; 28:175-186. [PMID: 34667024 DOI: 10.1158/1078-0432.ccr-19-2486] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 06/21/2021] [Accepted: 10/15/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Dedifferentiated liposarcoma (DDLS), one of the most common and aggressive sarcomas, infrequently responds to chemotherapy. DDLS survival and growth depend on underexpression of C/EBPα, a tumor suppressor and transcriptional regulator controlling adipogenesis. We sought to screen and prioritize candidate drugs that increase C/EBPα expression and may therefore serve as differentiation-based therapies for DDLS. EXPERIMENTAL DESIGN We screened known bioactive compounds for the ability to restore C/EBPα expression and inhibit proliferation selectively in two DDLS cell lines but not in normal adipose-derived stem cells (ASC). Selected hits' activity was validated, and the mechanism of the most potent, SN-38, was investigated. The in vivo efficacy of irinotecan, the prodrug of SN-38, was evaluated in DDLS xenograft models. RESULTS Of 3,119 compounds, screen criteria were met by 19. Validation experiments confirmed the DDLS selectivity of deguelin, emetine, and SN-38 and showed that they induce apoptosis in DDLS cells. SN-38 had the lowest IC50 (approximately 10 nmol/L), and its pro-apoptotic effects were countered by knockdown of CEBPA but not of TP53. Irinotecan significantly inhibited tumor growth at well-tolerated doses, induced nuclear expression of C/EBPα, and inhibited HIF1α expression in DDLS patient-derived and cancer cell line xenograft models. In contrast, doxorubicin, the most common treatment for nonresectable DDLS, reduced tumor growth by 30% to 50% at a dose that caused weight loss. CONCLUSIONS This high-content screen revealed potential treatments for DDLS. These include irinotecan, which induces apoptosis of DDLS cells in a C/EBPα-dependent, p53-independent manner, and should be clinically evaluated in patients with advanced DDLS.
Collapse
Affiliation(s)
- Christina V Angeles
- Sarcoma Biology Laboratory, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ana Velez
- Sarcoma Biology Laboratory, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jordan Rios
- Sarcoma Biology Laboratory, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Bernadette Laxa
- Sarcoma Biology Laboratory, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - David Shum
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Penelope D Ruiz
- Sarcoma Biology Laboratory, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Yawei Shen
- Sarcoma Biology Laboratory, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Irina Ostrovnaya
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Rodrigo Gularte-Mérida
- Sarcoma Biology Laboratory, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Benjamin A Nacev
- Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, New York
| | - Mark A Dickson
- Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, New York
| | - Hakim Djaballah
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Tomoyo Okada
- Sarcoma Biology Laboratory, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York.
| | - Samuel Singer
- Sarcoma Biology Laboratory, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York. .,Department of Surgery, Weill Cornell Medical College, New York, New York
| |
Collapse
|
6
|
Kannan S, Lock I, Ozenberger BB, Jones KB. Genetic drivers and cells of origin in sarcomagenesis. J Pathol 2021; 254:474-493. [DOI: 10.1002/path.5617] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/01/2020] [Accepted: 01/06/2021] [Indexed: 02/06/2023]
Affiliation(s)
- Sarmishta Kannan
- Departments of Orthopaedics and Oncological Sciences Huntsman Cancer Institute, University of Utah School of Medicine Salt Lake City UT USA
| | - Ian Lock
- Departments of Orthopaedics and Oncological Sciences Huntsman Cancer Institute, University of Utah School of Medicine Salt Lake City UT USA
| | - Benjamin B Ozenberger
- Departments of Orthopaedics and Oncological Sciences Huntsman Cancer Institute, University of Utah School of Medicine Salt Lake City UT USA
| | - Kevin B Jones
- Departments of Orthopaedics and Oncological Sciences Huntsman Cancer Institute, University of Utah School of Medicine Salt Lake City UT USA
| |
Collapse
|
7
|
Schaner PE, Pettus JR, Flood AB, Williams BB, Jarvis LA, Chen EY, Pastel DA, Zuurbier RA, diFlorio-Alexander RM, Swartz HM, Kuppusamy P. OxyChip Implantation and Subsequent Electron Paramagnetic Resonance Oximetry in Human Tumors Is Safe and Feasible: First Experience in 24 Patients. Front Oncol 2020; 10:572060. [PMID: 33194670 PMCID: PMC7653093 DOI: 10.3389/fonc.2020.572060] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 09/21/2020] [Indexed: 12/18/2022] Open
Abstract
Introduction: Tumor hypoxia confers both a poor prognosis and increased resistance to oncologic therapies, and therefore, hypoxia modification with reliable oxygen profiling during anticancer treatment is desirable. The OxyChip is an implantable oxygen sensor that can detect tumor oxygen levels using electron paramagnetic resonance (EPR) oximetry. We report initial safety and feasibility outcomes after OxyChip implantation in a first-in-humans clinical trial (NCT02706197, www.clinicaltrials.gov). Materials and Methods: Twenty-four patients were enrolled. Eligible patients had a tumor ≤ 3 cm from the skin surface with planned surgical resection as part of standard-of-care therapy. Most patients had a squamous cell carcinoma of the skin (33%) or a breast malignancy (33%). After an initial cohort of six patients who received surgery alone, eligibility was expanded to patients receiving either chemotherapy or radiotherapy prior to surgical resection. The OxyChip was implanted into the tumor using an 18-G needle; a subset of patients had ultrasound-guided implantation. Electron paramagnetic resonance oximetry was carried out using a custom-built clinical EPR scanner. Patients were evaluated for associated toxicity using the Common Terminology Criteria for Adverse Events (CTCAE); evaluations started immediately after OxyChip placement, occurred during every EPR oximetry measurement, and continued periodically after removal. The OxyChip was removed during standard-of-care surgery, and pathologic analysis of the tissue surrounding the OxyChip was performed. Results: Eighteen patients received surgery alone, while five underwent chemotherapy and one underwent radiotherapy prior to surgery. No unanticipated serious adverse device events occurred. The maximum severity of any adverse event as graded by the CTCAE was 1 (least severe), and all were related to events typically associated with implantation. After surgical resection, 45% of the patients had no histopathologic findings specifically associated with the OxyChip. All tissue pathology was "anticipated" excepting a patient with greater than expected inflammatory findings, which was assessed to be related to the tumor as opposed to the OxyChip. Conclusion: This report of the first-in-humans trial of OxyChip implantation and EPR oximetry demonstrated no significant clinical pathology or unanticipated serious adverse device events. Use of the OxyChip in the clinic was thus safe and feasible.
Collapse
Affiliation(s)
- Philip E Schaner
- Department of Medicine, Dartmouth Hitchcock Medical Center, Lebanon, NH, United States.,Geisel School of Medicine, Dartmouth College, Hanover, NH, United States.,Norris Cotton Cancer Center, Lebanon, NH, United States
| | - Jason R Pettus
- Department of Medicine, Dartmouth Hitchcock Medical Center, Lebanon, NH, United States.,Geisel School of Medicine, Dartmouth College, Hanover, NH, United States.,Norris Cotton Cancer Center, Lebanon, NH, United States.,Department of Pathology, Dartmouth Hitchcock Medical Center, Lebanon, NH, United States
| | - Ann Barry Flood
- Geisel School of Medicine, Dartmouth College, Hanover, NH, United States.,Norris Cotton Cancer Center, Lebanon, NH, United States.,Department of Radiology, Dartmouth Hitchcock Medical Center, Lebanon, NH, United States
| | - Benjamin B Williams
- Department of Medicine, Dartmouth Hitchcock Medical Center, Lebanon, NH, United States.,Geisel School of Medicine, Dartmouth College, Hanover, NH, United States.,Norris Cotton Cancer Center, Lebanon, NH, United States.,Department of Radiology, Dartmouth Hitchcock Medical Center, Lebanon, NH, United States
| | - Lesley A Jarvis
- Department of Medicine, Dartmouth Hitchcock Medical Center, Lebanon, NH, United States.,Geisel School of Medicine, Dartmouth College, Hanover, NH, United States.,Norris Cotton Cancer Center, Lebanon, NH, United States
| | - Eunice Y Chen
- Geisel School of Medicine, Dartmouth College, Hanover, NH, United States.,Norris Cotton Cancer Center, Lebanon, NH, United States.,Department of Surgery, Dartmouth Hitchcock Medical Center, Lebanon, NH, United States
| | - David A Pastel
- Geisel School of Medicine, Dartmouth College, Hanover, NH, United States.,Norris Cotton Cancer Center, Lebanon, NH, United States.,Department of Radiology, Dartmouth Hitchcock Medical Center, Lebanon, NH, United States
| | - Rebecca A Zuurbier
- Geisel School of Medicine, Dartmouth College, Hanover, NH, United States.,Norris Cotton Cancer Center, Lebanon, NH, United States.,Department of Radiology, Dartmouth Hitchcock Medical Center, Lebanon, NH, United States
| | - Roberta M diFlorio-Alexander
- Geisel School of Medicine, Dartmouth College, Hanover, NH, United States.,Norris Cotton Cancer Center, Lebanon, NH, United States.,Department of Radiology, Dartmouth Hitchcock Medical Center, Lebanon, NH, United States
| | - Harold M Swartz
- Department of Medicine, Dartmouth Hitchcock Medical Center, Lebanon, NH, United States.,Geisel School of Medicine, Dartmouth College, Hanover, NH, United States.,Norris Cotton Cancer Center, Lebanon, NH, United States.,Department of Radiology, Dartmouth Hitchcock Medical Center, Lebanon, NH, United States
| | - Periannan Kuppusamy
- Department of Medicine, Dartmouth Hitchcock Medical Center, Lebanon, NH, United States.,Geisel School of Medicine, Dartmouth College, Hanover, NH, United States.,Norris Cotton Cancer Center, Lebanon, NH, United States.,Department of Radiology, Dartmouth Hitchcock Medical Center, Lebanon, NH, United States
| |
Collapse
|
8
|
Han S, Huang T, Hou F, Yao L, Wang X, Wu X. The prognostic value of hypoxia-inducible factor-1α in advanced cancer survivors: a meta-analysis with trial sequential analysis. Ther Adv Med Oncol 2019; 11:1758835919875851. [PMID: 31579115 PMCID: PMC6759726 DOI: 10.1177/1758835919875851] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 08/19/2019] [Indexed: 12/26/2022] Open
Abstract
Background: Expression of hypoxia-inducible factors (HIFs) has been observed, but their prognostic role in advanced cancers remains uncertain. We conducted a meta-analysis to establish the prognostic effect of HIFs and to better guide treatment planning for advanced cancers. Methods: Pooled hazard ratios (HRs) and 95% confidence intervals (CIs) were calculated. Trial sequential analysis (TSA) was also performed. The clinical outcomes included overall survival (OS), disease-free survival (DFS), progression-free survival (PFS), cancer-specific survival (CSS), relapse/recurrence-free survival (RFS), and metastasis-free survival (MFS) in patients with advanced tumors according to multivariate analysis. Results: A total of 31 studies including 3453 cases who received chemotherapy, radiotherapy, or chemoradiotherapy were identified. Pooled analyses revealed that HIF-1α expression was correlated with worse OS (HR = 1.61, p < 0.001), DFS (HR = 1.61, p < 0.001), PFS (HR = 1.49, p = 0.01), CSS (HR = 1.65, p = 0.056), RFS (HR = 2.10, p = 0.015), or MFS (HR = 2.36, p = 0.002) in advanced cancers. HIF-1α expression was linked to shorter OS in the digestive tract, epithelial ovarian, breast, non-small cell lung, and clear cell renal cell carcinomas. Subgroup analysis by study region showed that HIF-1α expression was correlated with poor OS in Europeans and Asians, while an analysis by histologic subtypes found that HIF-1α expression was not associated with OS in squamous cell carcinoma. No relationship was found between HIF-2α expression and OS, DFS, PFS, or CSS. Conclusions: Targeting HIF-1α may be a useful therapeutic approach to improve survival for advanced cancer patients. Based on TSA, more randomized controlled trials are strongly suggested.
Collapse
Affiliation(s)
- Susu Han
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Zhijiang Road, 200071, People's Republic of China
| | - Tao Huang
- The Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, People's Republic of China
| | - Fenggang Hou
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, People's Republic of China
| | - Liting Yao
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, People's Republic of China
| | - Xiyu Wang
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, People's Republic of China
| | - Xing Wu
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, People's Republic of China
| |
Collapse
|
9
|
Morrell MB, Alvarez Florez C, Zhang A, Kleinerman ES, Savage H, Marmonti E, Park M, Shaw A, Schadler KL. Vascular modulation through exercise improves chemotherapy efficacy in Ewing sarcoma. Pediatr Blood Cancer 2019; 66:e27835. [PMID: 31136074 PMCID: PMC6646082 DOI: 10.1002/pbc.27835] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 05/06/2019] [Accepted: 05/10/2019] [Indexed: 12/15/2022]
Abstract
Recent studies in mouse models of cancer have shown that exercise improves tumor vascular function, thereby improving chemotherapy delivery and efficacy. However, the mechanisms underlying this improvement remain unclear and the effect of exercise on Ewing sarcoma (ES), a pediatric bone and soft tissue cancer, is unknown. The effect of exercise on tumor vascular hyperpermeability, which inversely correlates with drug delivery to the tumor, has also not been evaluated. We hypothesized that exercise improves chemotherapy efficacy by enhancing its delivery through improving tumor vascular permeability. We treated ES-bearing mice with doxorubicin with or without moderate treadmill exercise. Exercise did not significantly alter ES tumor vessel morphology. However, compared to control mice, tumors of exercised mice had significantly reduced hyperpermeability, significantly decreased hypoxia, and higher doxorubicin penetration. Compared to doxorubicin alone, doxorubicin plus exercise inhibited tumor growth more efficiently. We evaluated endothelial cell sphingosine-1-phosphate receptors 1 and 2 (S1PR1 and S1PR2) as potential mediators of the improved vascular permeability and increased function afforded by exercise. Relative to tumors from control mice, vessels in tumors from exercised mice had increased S1PR1 and decreased S1PR2 expression. Our results support a model in which exercise remodels ES vasculature to reduce vessel hyperpermeability, potentially via modulation of S1PR1 and S1PR2, thereby improving doxorubicin delivery and inhibiting tumor growth more than doxorubicin alone does. Our data suggest moderate aerobic exercise should be tested in clinical trials as a potentially useful adjuvant to standard chemotherapy for patients with ES.
Collapse
Affiliation(s)
- Miriam B.G. Morrell
- Department of Pediatric Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Claudia Alvarez Florez
- Department of Pediatric Research, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Aiqian Zhang
- Department of Pediatric Research, The University of Texas MD Anderson Cancer Center, Houston, Texas,Department of Gynecology, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Eugenie S. Kleinerman
- Department of Pediatric Research, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hannah Savage
- Department of Pediatric Research, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Enrica Marmonti
- Department of Pediatric Research, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Minjeong Park
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Angela Shaw
- Department of Pediatric Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Keri L. Schadler
- Department of Pediatric Research, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
10
|
Yang L, Forker L, Irlam JJ, Pillay N, Choudhury A, West CML. Validation of a hypoxia related gene signature in multiple soft tissue sarcoma cohorts. Oncotarget 2018; 9:3946-3955. [PMID: 29423096 PMCID: PMC5790513 DOI: 10.18632/oncotarget.23280] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 11/13/2017] [Indexed: 12/12/2022] Open
Abstract
PURPOSE There is a need for adjuvant/neo-adjuvant treatment strategies to prevent metastatic relapse in soft tissue sarcoma (STS). Tumor hypoxia is associated with a high-risk of metastasis and is potentially targetable. This study aimed to derive and validate a hypoxia mRNA signature for STS for future biomarker-driven trials of hypoxia targeted therapy. MATERIALS AND METHODS RNA sequencing was used to identify seed genes induced by hypoxia in seven STS cell lines. Primary tumors in a training cohort (French training) were clustered into two phenotypes by seed gene expression and a de novo hypoxia signature derived. Prognostic significance of the de novo signature was evaluated in the training and two independent validation (French validation and The Cancer Genome Atlas) cohorts. RESULTS 37 genes were up-regulated by hypoxia in all seven cell lines, and a 24-gene signature was derived. The high-hypoxia phenotype defined by the signature was enriched for well-established hypoxia genes reported in the literature. The signature was prognostic in univariable analysis, and in multivariable analysis in the training (n = 183, HR 2.16, P = 0.0054) and two independent validation (n = 127, HR 3.06, P = 0.0019; n = 258, HR 2.05, P = 0.0098) cohorts. Combining information from the de novo hypoxia signature and a genome instability signature significantly improved prognostication. Transcriptomic analyses showed high-hypoxia tumors had more genome instability and lower immune scores. CONCLUSIONS A 24-gene STS-specific hypoxia signature may be useful for prognostication and identifying patients for hypoxia-targeted therapy in clinical trials.
Collapse
Affiliation(s)
- Lingjian Yang
- Translational Radiobiology Group, Division of Cancer Sciences, University of Manchester, Manchester Academic Health Science Centre, Christie Hospital, Manchester, UK
| | - Laura Forker
- Translational Radiobiology Group, Division of Cancer Sciences, University of Manchester, Manchester Academic Health Science Centre, Christie Hospital, Manchester, UK
| | - Joely J. Irlam
- Translational Radiobiology Group, Division of Cancer Sciences, University of Manchester, Manchester Academic Health Science Centre, Christie Hospital, Manchester, UK
| | - Nischalan Pillay
- Cancer Institute, University College London, London, UK
- Histopathology, Royal National Orthopaedic Hospital, Stanmore, UK
| | - Ananya Choudhury
- Translational Radiobiology Group, Division of Cancer Sciences, University of Manchester, Manchester Academic Health Science Centre, Christie Hospital, Manchester, UK
| | - Catharine M. L. West
- Translational Radiobiology Group, Division of Cancer Sciences, University of Manchester, Manchester Academic Health Science Centre, Christie Hospital, Manchester, UK
- NIHR Manchester Biomedical Research Centre, Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| |
Collapse
|