1
|
Qin H, Qi T, Xu J, Wang T, Zeng H, Yang J, Yu F. Integration of ubiquitination-related genes in predictive signatures for prognosis and immunotherapy response in sarcoma. Front Oncol 2024; 14:1446522. [PMID: 39469643 PMCID: PMC11513255 DOI: 10.3389/fonc.2024.1446522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 09/19/2024] [Indexed: 10/30/2024] Open
Abstract
Background Ubiquitination is one of the most prevalent and complex post-translational modifications of proteins in eukaryotes, playing a critical role in regulating various physiological and pathological processes. Targeting ubiquitination pathways, either through inhibition or activation, holds promise as a novel therapeutic approach for cancer treatment. However, the expression patterns, prognostic significance, and underlying mechanisms of ubiquitination-related genes (URGs) in sarcoma (SARC) remain unclear. Methods We analyzed URG expression patterns and prognostic implications in TCGA-SARC using public databases, identifying DEGs related to ubiquitination among SARC molecular subtypes. Functional enrichment analysis elucidated their biological significance. Prognostic signatures were developed using LASSO-Cox regression, and a predictive nomogram was constructed. External validation was performed using GEO datasets and clinical tissue samples. The association between URG risk scores and various clinical parameters, immune response, drug sensitivity, and RNA modification regulators was investigated. Integration of data from multiple sources and RT-qPCR confirmed upregulated expression of prognostic URGs in SARC. Single-cell RNA sequencing data analyzed URG distribution across immune cell types. Prediction analysis identified potential target genes of microRNAs and long non-coding RNAs. Results We identified five valuable genes (CALR, CASP3, BCL10, PSMD7, PSMD10) and constructed a prognostic model, simultaneously identifying two URG-related subtypes in SARC. The UEGs between subtypes in SARC are mainly enriched in pathways such as Cell cycle, focal adhesion, and ECM-receptor interaction. Analysis of URG risk scores reveals that patients with a low-risk score have better prognoses compared to those with high-risk scores. There is a significant correlation between DRG riskscore and clinical features, immune therapy response, drug sensitivity, and genes related to pan-RNA epigenetic modifications. High-risk SARC patients were identified as potential beneficiaries of immune checkpoint inhibitor therapy. We established regulatory axes in SARC, including CALR/hsa-miR-29c-3p/LINC00943, CASP3/hsa-miR-143-3p/LINC00944, and MIR503HG. RT-qPCR data further confirmed the upregulation of prognostic URGs in SARC. Finally, we validated the prognostic model's excellent predictive performance in predicting outcomes for SARC patients. Conclusion We discovered a significant correlation between aberrant expression of URGs and prognosis in SARC patients, identifying a prognostic model related to ubiquitination. This model provides a basis for individualized treatment and immunotherapy decisions for SARC patients.
Collapse
Affiliation(s)
- Haotian Qin
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Tiantian Qi
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Juan Xu
- Department of Oncology, Chaohu Hospital of Anhui Medical University, Hefei, China
| | - Tianbing Wang
- Department of Orthopedics and Trauma, Peking University People’s Hospital, Beijing, China
| | - Hui Zeng
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Jun Yang
- Department of Radiology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Fei Yu
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
2
|
Tlemsani C, Heske CM, Elloumi F, Pongor L, Khandagale P, Varma S, Luna A, Meltzer PS, Khan J, Reinhold WC, Pommier Y. Sarcoma_CellminerCDB: A tool to interrogate the genomic and functional characteristics of a comprehensive collection of sarcoma cell lines. iScience 2024; 27:109781. [PMID: 38868205 PMCID: PMC11167437 DOI: 10.1016/j.isci.2024.109781] [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: 10/26/2023] [Revised: 12/28/2023] [Accepted: 04/15/2024] [Indexed: 06/14/2024] Open
Abstract
Sarcomas are a diverse group of rare malignancies composed of multiple different clinical and molecular subtypes. Due to their rarity and heterogeneity, basic, translational, and clinical research in sarcoma has trailed behind that of other cancers. Outcomes for patients remain generally poor due to an incomplete understanding of disease biology and a lack of novel therapies. To address some of the limitations impeding preclinical sarcoma research, we have developed Sarcoma_CellMinerCDB, a publicly available interactive tool that merges publicly available sarcoma cell line data and newly generated omics data to create a comprehensive database of genomic, transcriptomic, methylomic, proteomic, metabolic, and pharmacologic data on 133 annotated sarcoma cell lines. The reproducibility, functionality, biological relevance, and therapeutic applications of Sarcoma_CellMinerCDB described herein are powerful tools to address and generate biological questions and test hypotheses for translational research. Sarcoma_CellMinerCDB (https://discover.nci.nih.gov/SarcomaCellMinerCDB) aims to contribute to advancing the preclinical study of sarcoma.
Collapse
Affiliation(s)
- Camille Tlemsani
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
- Department of Medical Oncology, Cochin Hospital, Paris Cancer Institute CARPEM, Université Paris Cité, APHP. Centre, Paris, France
- Institut Cochin, INSERM U1016, CNRS UMR8104, Paris Cancer Institute CARPEM, Université Paris Cité, Paris, France
| | - Christine M. Heske
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Fathi Elloumi
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Lorinc Pongor
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
- Hungarian Centre of Excellence for Molecular Medicine, Cancer Genomics and Epigenetics Core Group, Szeged, Hungary
| | - Prashant Khandagale
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Sudhir Varma
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Augustin Luna
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
- Computational Biology Branch, National Library of Medicine, NIH, Bethesda, Maryland 20892, USA
| | - Paul S. Meltzer
- Genetics Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Javed Khan
- Genetics Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - William C. Reinhold
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Yves Pommier
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| |
Collapse
|
3
|
Xu J, Guo K, Sheng X, Huang Y, Wang X, Dong J, Qin H, Wang C. Correlation analysis of disulfidptosis-related gene signatures with clinical prognosis and immunotherapy response in sarcoma. Sci Rep 2024; 14:7158. [PMID: 38531930 DOI: 10.1038/s41598-024-57594-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 03/20/2024] [Indexed: 03/28/2024] Open
Abstract
Disulfidptosis, a newly discovered type of programmed cell death, could be a mechanism of cell death controlled by SLC7A11. This could be closely associated with tumor development and advancement. Nevertheless, the biological mechanism behind disulfidptosis-related genes (DRGs) in sarcoma (SARC) is uncertain. This study identified three valuable genes (SLC7A11, RPN1, GYS1) associated with disulfidptosis in sarcoma (SARC) and developed a prognostic model. The multiple databases and RT-qPCR data confirmed the upregulated expression of prognostic DRGs in SARC. The TCGA internal and ICGC external validation cohorts were utilized to validate the predictive model capacity. Our analysis of DRG riskscores revealed that the low-risk group exhibited a more favorable prognosis than the high-risk group. Furthermore, we observed a significant association between DRG riskscores and different clinical features, immune cell infiltration, immune therapeutic sensitivity, drug sensitivity, and RNA modification regulators. In addition, two external independent immunetherapy datasets and clinical tissue samples were collected, validating the value of the DRGs risk model in predicting immunotherapy response. Finally, the SLC7A11/hsa-miR-29c-3p/LINC00511, and RPN1/hsa-miR-143-3p/LINC00511 regulatory axes were constructed. This study provided DRG riskscore signatures to predict prognosis and response to immunotherapy in SARC, guiding personalized treatment decisions.
Collapse
Affiliation(s)
- Juan Xu
- Department of Oncology, Chaohu Hospital of Anhui Medical University, Hefei, China
| | - Kangwen Guo
- Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Xiaoan Sheng
- Department of Oncology, Chaohu Hospital of Anhui Medical University, Hefei, China
| | - Yuting Huang
- Department of Oncology, Chaohu Hospital of Anhui Medical University, Hefei, China
| | - Xuewei Wang
- Department of Oncology, Chaohu Hospital of Anhui Medical University, Hefei, China
| | - Juanjuan Dong
- Department of Oncology, Chaohu Hospital of Anhui Medical University, Hefei, China.
| | - Haotian Qin
- National and Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, China.
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, China.
| | - Chao Wang
- Department of Oncology, Chaohu Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
4
|
Murphy J, Resch EE, Leland C, Meyer CF, Llosa NJ, Gross JM, Pratilas CA. Clinical outcomes of patients with CIC-rearranged sarcoma: a single institution retrospective analysis. J Cancer Res Clin Oncol 2024; 150:112. [PMID: 38436779 PMCID: PMC10912249 DOI: 10.1007/s00432-024-05631-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 01/24/2024] [Indexed: 03/05/2024]
Abstract
PURPOSE CIC-rearranged sarcomas represent a type of undifferentiated small round cell sarcoma (USRCS) characterized by poor survival, rapid development of chemotherapy resistance, and high rates of metastasis. We aim to contribute to the growing body of knowledge regarding diagnosis, treatment, clinical course, and outcomes for these patients. METHODS This case series investigates the clinical courses of ten patients with CIC-rearranged sarcoma treated at the Johns Hopkins Hospital from July 2014 through January 2024. Clinical data were retrospectively extracted from electronic medical records. RESULTS Patients ranged from 10 to 67 years of age at diagnosis, with seven patients presenting with localized disease and three with metastatic disease. Tumors originated from soft tissues of various anatomic locations. Mean overall survival (OS) was 22.1 months (10.6-52.2), and mean progression-free survival (PFS) was 16.7 months (5.3-52.2). Seven patients received intensive systemic therapy with an Ewing sarcoma-directed regimen or a soft tissue sarcoma-directed regimen. Three patients experienced prolonged disease-free survival without systemic treatment. CONCLUSION Most patients in this case series demonstrated aggressive clinical courses consistent with those previously described in the literature, although we note a spectrum of clinical outcomes not previously reported. The diversity of clinical courses underscores the need for an improved understanding of individual tumor biology to enhance clinical decision-making and patient prognosis. Despite its limitations, this article broadens the spectrum of reported clinical outcomes, providing a valuable addition to the published literature on this rare cancer.
Collapse
Affiliation(s)
- Jacob Murphy
- Johns Hopkins University School of Medicine, 733 N Broadway, Baltimore, MD, 21205, USA
| | - Erin E Resch
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA
| | - Christopher Leland
- Johns Hopkins University School of Medicine, 733 N Broadway, Baltimore, MD, 21205, USA
- Department of Orthopedic Surgery, Massachusetts General Hospital, Boston, USA
| | - Christian F Meyer
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA
| | - Nicolas J Llosa
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA
| | - John M Gross
- Department of Pathology, Johns Hopkins University School of Medicine, 401 N Broadway, Baltimore, MD, 21231, USA
| | - Christine A Pratilas
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA.
| |
Collapse
|
5
|
Ciurej A, Lewis E, Gupte A, Al-Antary E. Checkpoint Immunotherapy in Pediatric Oncology: Will We Say Checkmate Soon? Vaccines (Basel) 2023; 11:1843. [PMID: 38140246 PMCID: PMC10748105 DOI: 10.3390/vaccines11121843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/30/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) are a relatively new class of immunotherapy which bolsters the host immune system by "turning off the brakes" of effector cells (e.g., CTLA-4, PD-1, PD-L1). Although their success in treating adult malignancy is well documented, their utility in pediatric cancer has not yet been shown to be as fruitful. We review ICIs, their use in pediatric malignancies, and active pediatric clinical trials, exemplifying some of adult efforts that could be related to pediatric future trials and complications of ICI therapy. Through our review, we propose the consideration of ICI as standard therapy in lymphoma and various solid tumor types, especially in relapsed or refractory (R/R) disease. However, further studies are needed to demonstrate ICI effectiveness in pediatric leukemia.
Collapse
Affiliation(s)
- Alexander Ciurej
- Pediatric Department, Children’s Hospital of Michigan, Detroit, MI 48201, USA; (A.C.)
| | - Elizabeth Lewis
- School of Medicine, Wayne State University, Detroit, MI 48201, USA
| | - Avanti Gupte
- Pediatric Department, Children’s Hospital of Michigan, Detroit, MI 48201, USA; (A.C.)
- Pediatric Blood and Marrow Transplantation Program, Division of Hematology/Oncology, Barbara Ann Karmanos Cancer Center, Children’s Hospital of Michigan, Detroit, MI 48201, USA
- Department of Pediatrics, Central Michigan University College of Medicine, Mt Clemons, MI 48859, USA
| | - Eman Al-Antary
- Pediatric Department, Children’s Hospital of Michigan, Detroit, MI 48201, USA; (A.C.)
- Pediatric Blood and Marrow Transplantation Program, Division of Hematology/Oncology, Barbara Ann Karmanos Cancer Center, Children’s Hospital of Michigan, Detroit, MI 48201, USA
- Department of Pediatrics, Central Michigan University College of Medicine, Mt Clemons, MI 48859, USA
| |
Collapse
|
6
|
Heesen P, Ranft A, Bhadri V, Brichard B, Collaud S, Cyprova S, Eich H, Ek T, Gelderblom H, Hardes J, Haveman L, Jabar S, Hartmann W, Andreou D, Hauser P, Kersting J, Juergens H, Kanerva J, Kühne T, Raciborska A, Rascon J, Streitbürger A, Timmermann B, Uhlenbruch Y, Dirksen U. Association between local treatment modalities and event-free survival, overall survival, and local recurrence in patients with localised Ewing Sarcoma. Report from the Ewing 2008 trial. Eur J Cancer 2023; 192:113260. [PMID: 37595489 DOI: 10.1016/j.ejca.2023.113260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/21/2023] [Accepted: 07/23/2023] [Indexed: 08/20/2023]
Abstract
BACKGROUND Local treatment is a crucial element in the standard of care for Ewing sarcoma (EWS). While systemic treatment is improved in randomised clinical trials, local treatment modalities are discussed controversially. We analysed the association between local therapy and event-free survival (EFS), overall survival (OS), and local recurrence (LR) in prospectively collected data of patients with localised EWS. PATIENTS AND METHODS We analysed data from the international Ewing 2008 study registered between 2009 and 2019 in 117 centres. After induction chemotherapy, patients received surgery, radiotherapy, or a combination thereof. We performed Cox regression, conducted propensity score-weighted sensitivity analysis, and performed subgroup analyses. Hazard ratios (HRs) and 95% confidence intervals are reported. RESULTS We included 863 patients with localised EWS (surgery alone: 331, combination therapy: 358, definitive radiotherapy: 174). In patients treated with combination therapy compared to surgery alone, EFS HR was 0.84 (0.57-1.24; p = 0.38), OS HR was 0.84 (0.57-1.23; p = 0.41), and LR HR was 0.58 (0.26-1.31; p = 0.19). Hazards of any event were increased in patients treated with definitive radiotherapy compared to surgery only, HR 1.53 (1.02-2.31; p = 0.04). Patients with poor responses to chemotherapy benefitted from combination therapy over definitive surgery with an EFS HR 0.49 (0.27-0.89; p = 0.02). Patients with pelvic tumours benefitted from combination therapy over surgery only regarding LR, HR 0.12 (0.02-0.72; p = 0.02). CONCLUSION Patients with poor responses to chemotherapy benefitted from radiotherapy added to surgery. In the whole group, radiotherapy alone as opposed to surgery alone increased the hazards of any event.
Collapse
Affiliation(s)
- Philip Heesen
- Pediatrics III, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Hufelandstrasse 55, 45147 Essen, Germany; University of Zurich, Raemistrasse 71, 8006 Zurich, Switzerland
| | - Andreas Ranft
- Pediatrics III, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Hufelandstrasse 55, 45147 Essen, Germany; German Cancer Consortium (DKTK), and National Center for Tumor diseases (NCT), Partnersite Essen, Essen, Germany
| | - Vivek Bhadri
- Chris O´ Brien Lifehouse, Camperdown, Australia; Health, University of Sydney, Camperdown, New South Wales 2006, Australia
| | - Benedicte Brichard
- Department of Pediatric Haematology and Oncology, Cliniques Universitaires Saint Luc, Université Catholique de Louvain, Av. Hippocrate 10, 1200 Brussels, Belgium
| | - Stephane Collaud
- German Cancer Consortium (DKTK), and National Center for Tumor diseases (NCT), Partnersite Essen, Essen, Germany; Department of Thoracic Surgery, Ruhrlandklinik, University Hospital Essen, West German Cancer Center, Tüschener Weg 40, 45239 Essen, Germany
| | - Sona Cyprova
- Charles University, Motol Children's Hospital, V Uvalu 84, 150 06 Prague 5, Czech Republic
| | - Hans Eich
- Radiotherapy and Radiooncology, University Hospital Muenster, West German Cancer Center Network, Albert-Schweitzer-Campus 1, 48149 Muenster, Germany
| | - Torben Ek
- Childhood Cancer Center Queen Silvia Children's Hospital, Behandlingsvägen 7, 416 50 Gothenburg, Sweden
| | - Hans Gelderblom
- Department of Medical Oncology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden Netherlands
| | - Jendrik Hardes
- German Cancer Consortium (DKTK), and National Center for Tumor diseases (NCT), Partnersite Essen, Essen, Germany; Clinic of Orthopedics, University Hospital Essen, West German Cancer Centre, Hufelandstrasse 55, 45147 Essen, Germany
| | - Lianne Haveman
- Department of Solid Tumors, Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, the Netherlands
| | - Susanne Jabar
- Pediatrics III, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Hufelandstrasse 55, 45147 Essen, Germany; German Cancer Consortium (DKTK), and National Center for Tumor diseases (NCT), Partnersite Essen, Essen, Germany
| | - Wolfgang Hartmann
- Gerhard Domagk Institute for Pathology, University Hospital Muenster, West German Cancer Center Network, Albert-Schweitzer-Campus 1, 48149 Muenster, Germany
| | - Dimosthenis Andreou
- Department of Orthopedics and Trauma, Medical University of Graz, Auenbruggerpl. 2, 8036 Graz, Austria
| | - Peter Hauser
- Velkey László Child's, Health Center, Borsod-Abaúj-Zemplén County University Teaching Hospital, Miskolc, Hungary
| | - Josephine Kersting
- Pediatrics III, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Hufelandstrasse 55, 45147 Essen, Germany
| | - Heribert Juergens
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, West German Cancer Center Network, Albert-Schweitzer-Campus 1, 48149 Muenster, Germany
| | - Jukka Kanerva
- HUS Helsinki University Hospital, New Children's Hospital, Div. Hematology and Stem Cell Transplantation, Stenbäckinkatu 9 Hallintokeskus, rakennus 5, 00290 Helsinki, Finnland
| | - Thomas Kühne
- Department of Oncology/ Haematology, University Children's Hospital Basel, Spitalstrasse 33, 4056 Basel, Switzerland
| | - Anna Raciborska
- Mother and Child Institute, Department of Oncology and Surgical Oncology for Children and Youth, Marcina Kasprzaka 17a, 01-211 Warszawa, Poland
| | - Jelena Rascon
- Center for Pediatric Oncology and Hematology, Vilnius University Hospital Santaros Klinikos, Vilnius University, Santariškių g. 2, 08661 Vilnius, Lithuania
| | - Arne Streitbürger
- German Cancer Consortium (DKTK), and National Center for Tumor diseases (NCT), Partnersite Essen, Essen, Germany; Department of Solid Tumors, Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, the Netherlands
| | - Beate Timmermann
- German Cancer Consortium (DKTK), and National Center for Tumor diseases (NCT), Partnersite Essen, Essen, Germany; Clinic for Particle Therapy, West German Proton Beam Centre, University Hospital Essen, West German Cancer Centre, German Cancer Research Centre (DKTK), Hufelandstrasse 55, 45147 Essen, Germany
| | - Yasmin Uhlenbruch
- Patient Representative, St. Josefs Hospital Bochum, University Hospital, Gudrunstraße 56, 44791 Bochum, Germany
| | - Uta Dirksen
- Pediatrics III, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Hufelandstrasse 55, 45147 Essen, Germany; German Cancer Consortium (DKTK), and National Center for Tumor diseases (NCT), Partnersite Essen, Essen, Germany.
| |
Collapse
|
7
|
Baracani R, Bhaskaran M, Davis SM, Morford L, Luffer-Atlas D. PDGFRα monoclonal antibody: Assessment of toxicity in juvenile mice administered a murine surrogate antibody of olaratumab. Birth Defects Res 2023; 115:782-796. [PMID: 36916488 DOI: 10.1002/bdr2.2169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/22/2023] [Accepted: 02/24/2023] [Indexed: 03/16/2023]
Abstract
BACKGROUND Olaratumab (Lartruvo™) is a recombinant human IgG1 monoclonal antibody that specifically binds PDGFRα. In order to support use of Lartruvo in pediatric patients, a definitive juvenile animal study in neonatal mice was conducted with a human anti-mouse PDGFRα antibody analog of olaratumab (LSN3338786). METHODS A pilot study was used to set doses for the definitive juvenile mouse study. In the definitive study, juvenile mice were administered vehicle, 50, 100, or 150 mg/kg LSN3338786 by subcutaneous (SC) injection every 3 days between postnatal days (PND) 1 and 49, for a total of 17 doses. Blood samples were collected on PND 49 for antibody analysis and toxicokinetic evaluation. Tissues were collected on PND 52 for histopathologic examination. RESULTS Results of the pilot study indicated that dosing neonatal mice starting on PND 1 via SC administration every 3 days was logistically feasible, produced exposures consistent with prior animal studies, and the selected dose levels were well tolerated by juvenile mice. In the definitive juvenile study, there were no LSN3338786-related deaths, clinical findings, and no effects on mean body weights, body weight gains, or food consumption. Additionally, there were no adverse LSN3338786-related hematology findings, and no macroscopic, organ weight, or microscopic findings of note. The highest dose evaluated, 150 mg/kg, was considered the NOAEL for juvenile toxicity. CONCLUSIONS In conclusion, the juvenile animal studies did not identify any new toxicities or increased sensitivities for the intended pediatric patient population. The use of the surrogate antibody approach in a standard rodent model enabled the de-risking of theoretical concerns for toxicity in pediatric patients due to disruption of the PDGFRα pathway during early human development, such as pulmonary development.
Collapse
Affiliation(s)
| | - Manoj Bhaskaran
- Lilly Research Laboratories, Corporate Center, Indianapolis, Indiana, USA
| | | | - LaRonda Morford
- Lilly Research Laboratories, Corporate Center, Indianapolis, Indiana, USA
| | - Debra Luffer-Atlas
- Lilly Research Laboratories, Corporate Center, Indianapolis, Indiana, USA
| |
Collapse
|
8
|
Trends in Overall Survival among Patients Treated for Sarcoma at a Large Tertiary Cancer Center between 1986 and 2014. Cancers (Basel) 2023; 15:cancers15020514. [PMID: 36672463 PMCID: PMC9856368 DOI: 10.3390/cancers15020514] [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: 11/11/2022] [Revised: 01/10/2023] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
Sarcomas are relatively rare malignancies accounting for about 1% of all cancer diagnoses. Studies on sarcomas comprising large cohorts covering extended time periods are lacking. Therefore, this study aimed to evaluate the impact of demographic, behavioral, and clinical characteristics on overall survival (OS) among individuals diagnosed with soft tissue sarcoma (STS) or bone sarcoma at the Moffitt Cancer Center between 1986 and 2014. Unadjusted and multivariable Cox proportional hazard regression (CPHR) models were constructed to generate hazard ratios (HRs) and 95% confidence intervals (CIs) to evaluate associations between a range of demographic, behavioral, and clinical characteristics, and OS. Additionally, Kaplan-Meier survival curves, associated log-rank statistics, and adjusted CPHR models were generated by time periods based on the year of first contact (1986-1994, 1995-1999, 2000-2005, 2006-2010, 2011-2014) to evaluate for temporal differences in OS. Of the 2570 patients, 2037 were diagnosed with STS, whereas 533 were diagnosed with bone sarcoma. At the time of analysis, 50% of the population were alive. In multivariable analyses, we observed poorer survival for patients ≥ 40 years of age (HR = 1.54, 95% CI = 1.34-1.78), current smokers (HR = 1.18, 95% CI = 1.01-1.37), patients with metastasis (HR = 2.19, 95% CI = 1.95-2.47), and patients not receiving first-line surgery treatment (HR = 2.11, 95% CI = 1.82-2.45). We discovered limited improvements in OS over time among individuals diagnosed with STS or bone sarcomas with the exception of gastrointestinal stromal tumors (GIST), which showed a significant improvement in OS across time periods (p = 0.0034). Overall, we identified well-established characteristics associated with OS (e.g., metastasis) in addition to factors (e.g., smoking status) not previously reported to impact OS. Improvements in survival over time have been relatively modest, suggesting the need for improved therapeutic options, especially for those diagnosed with less frequent sarcomas.
Collapse
|
9
|
Affiliation(s)
- Anuradha Rajagopalan
- Department of Child Health, School of Medicine, University of Missouri, Columbia, MO
| | - Sarah C Christenberry
- Department of Child Health, School of Medicine, University of Missouri, Columbia, MO
| | | |
Collapse
|
10
|
Zhou C, Chen Z, Xiao B, Xiang C, Li A, Zhao Z, Li H. Comprehensive analysis of GINS subunits prognostic value and ceRNA network in sarcoma. Front Cell Dev Biol 2022; 10:951363. [PMID: 36092720 PMCID: PMC9462653 DOI: 10.3389/fcell.2022.951363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 07/29/2022] [Indexed: 11/13/2022] Open
Abstract
Background: The GINS complex, composed of GINS1/2/3/4 subunits, is an essential structure of Cdc45-MCM-GINS (CMG) helicase and plays a vital role in establishing the DNA replication fork and chromosome replication. Meanwhile, GINS genes have been associated with the poor prognosis of various malignancies. However, the abnormal expression of GINS genes and their diagnostic and prognostic value in sarcomas (SARC) remain unclear. Methods: Oncomine, Gene Expression Profiling Interactive Analysis (GEPIA), Kaplan-Meier Plotter, Cancer cell line encyclopedia (CCLE), The University of Alabama at Birmingham Cancer Data Analysis Portal (UALCAN), R studio, and Tumor Immune Estimation Resource (TIMER) were used to analyze the expression profiles, prognostic value, biological function, ceRNA, and immune infiltration associated with GINS genes in sarcomas. Results: We found that GINS1/2/3/4 genes exhibited significantly upregulated transcription levels in SARC samples compared to non-tumor tissues and exhibited high expression levels in sarcoma cell lines. In addition, SARC patients with increased expression levels of GINS1/2/3/4 showed poorer survival rates. Immune infiltration analysis showed that GINS subunits were closely associated with the infiltration of immune cells in sarcomas. Conclusion: Our research identified GINS subunits as potential diagnostic and prognostic biological targets in SARC and elucidated their underlying effects in the genesis and progression of SARC. These results may provide new opportunities and research directions for targeted sarcoma therapy.
Collapse
Affiliation(s)
- Chuqiao Zhou
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Orthopedic Biomedical Materials Engineering Laboratory of Hunan Province, Changsha, China
| | - Zhuoyuan Chen
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Orthopedic Biomedical Materials Engineering Laboratory of Hunan Province, Changsha, China
| | - Bo Xiao
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Orthopedic Biomedical Materials Engineering Laboratory of Hunan Province, Changsha, China
| | - Cheng Xiang
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Orthopedic Biomedical Materials Engineering Laboratory of Hunan Province, Changsha, China
| | - Aoyu Li
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Orthopedic Biomedical Materials Engineering Laboratory of Hunan Province, Changsha, China
| | - Ziyue Zhao
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Orthopedic Biomedical Materials Engineering Laboratory of Hunan Province, Changsha, China
| | - Hui Li
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Orthopedic Biomedical Materials Engineering Laboratory of Hunan Province, Changsha, China
- *Correspondence: Hui Li,
| |
Collapse
|
11
|
Munjewar PK, Sharma DR, Wanjari MB, Kantode VV. A Rare Occurrence of Relapse Malignant Vascular Mesenchymal Tumors (Rhabdomyosarcoma). Cureus 2022; 14:e27525. [PMID: 36060346 PMCID: PMC9427127 DOI: 10.7759/cureus.27525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 07/31/2022] [Indexed: 11/05/2022] Open
Abstract
Relapsed rhabdomyosarcoma (RMS) has several therapeutic challenges. The novel treatment for relapsed RMS was surgical management, chemotherapy, and radiotherapy. Reoccurrence significantly occurs in children and adolescents. RMS occurs anywhere in the body but mostly occurs in the legs, head, neck, urinary, and reproductive systems. Here, we present the case of a 19-year-old female who came to the emergency department with complaints of swelling in the left side of the neck that extended toward the face and left eye, breathlessness, and vomiting for one month. She has a history of peripheral nerve sheath tumor and type 1 diabetes mellitus. Surgical management was done through excision of the mesenchymal tumor surgery, and the patient's prognosis was good.
Collapse
|
12
|
Yao W, Du X, Wang J, Wang X, Zhang P, Niu X. Long-Term Efficacy and Safety of Anlotinib as a Monotherapy and Combined Therapy for Advanced Sarcoma. Onco Targets Ther 2022; 15:669-679. [PMID: 35726279 PMCID: PMC9206457 DOI: 10.2147/ott.s365506] [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: 03/15/2022] [Accepted: 06/09/2022] [Indexed: 11/23/2022] Open
Abstract
Objective To analyze the effectiveness of the long-term (> 12 months) administration of anlotinib as a monotherapy or combined therapy in patients with advanced sarcomas. Methods A retrospective analysis was conducted of patients with advanced sarcomas with measurable target lesions since 2018. Twenty-two of the patients had taken anlotinib regularly for > 12 months. The patients’ general information and the drug’s clinical efficacy and toxicity data were collected and statistically analyzed using RECIST 1.1 to measure the target lesions and tumor PFS time as the main endpoints. We used a swimmer plot to observe the drug’s efficacy and duration, and employed a waterfall plot to express the best treatment effect. Results The study included 14 male and 8 female patients, ranging in age from 14 to 75 (mean: 44.82) years. The primary diseases included alveolar soft part sarcoma, synovial sarcoma, leiomyosarcoma, and others. The metastasis sites were the lungs in fifteen cases, lymph nodes in four cases, and multiple sites in three cases. Fourteen patients had previously undergone chemotherapy. The current therapy protocol was oral anlotinib alone for nine cases, combination chemotherapy for nine cases, and combination immunotherapy (anti-PD-1) for four cases. The highest clinical efficacy was complete remission (CR) in four (18.18%) cases, partial response (PR) in five (22.73%) cases, and stable disease in 13 (59.09%) cases, with an odds ratio of response of 40.91%. The mean PFS for the CR, PR, and stable disease groups was 16.50, 14.50, and 29.31 months, respectively (p < 0.05). The main adverse effects included hand-foot syndrome, hypertension, and leukopenia. Conclusion Anlotinib monotherapy or combination therapy can be more effective and safer for certain advanced sarcomas, with more extended maintenance and acceptable side effects. Clinical efficacy at the CR and PR levels might predict the long-term PFS in certain advanced sarcomas.
Collapse
Affiliation(s)
- Weitao Yao
- Bone and Soft Department, The Affiliated Cancer Hospital of Zheng Zhou University, He Nan Cancer Hospital, Zheng Zhou City, 450000, People's Republic of China
| | - Xinhui Du
- Bone and Soft Department, The Affiliated Cancer Hospital of Zheng Zhou University, He Nan Cancer Hospital, Zheng Zhou City, 450000, People's Republic of China
| | - Jiaqiang Wang
- Bone and Soft Department, The Affiliated Cancer Hospital of Zheng Zhou University, He Nan Cancer Hospital, Zheng Zhou City, 450000, People's Republic of China
| | - Xin Wang
- Bone and Soft Department, The Affiliated Cancer Hospital of Zheng Zhou University, He Nan Cancer Hospital, Zheng Zhou City, 450000, People's Republic of China
| | - Peng Zhang
- Bone and Soft Department, The Affiliated Cancer Hospital of Zheng Zhou University, He Nan Cancer Hospital, Zheng Zhou City, 450000, People's Republic of China
| | - Xiaohui Niu
- Department of Orthopedic Oncology Surgery, Beijing Ji Shui Tan Hospital, Peking University, Beijing, 100035, People's Republic of China
| |
Collapse
|
13
|
Torres-Zárate C, Vences-Mejía A, Espinosa-Aguirre JJ, Díaz-Díaz E, Palacios-Acosta JM, Cárdenas-Cardós R, Hernández-Arrazola D, Shalkow-Klincovstein J, Jurado RR, Santes-Palacios R, Molina-Ortiz D. Expression of Cytochrome P450 Enzymes in Pediatric Non-Rhabdomyosarcoma Soft Tissue Sarcomas: Possible Role in Carcinogenesis and Treatment Response. Int J Toxicol 2022; 41:234-242. [PMID: 35437033 DOI: 10.1177/10915818221085909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The 5-year relative survival rate estimate of treated patients with non-rhabdomyosarcoma soft tissue sarcomas (NRSTS) is ∼50% since they generally present with tumor progression, relapse, metastasis, and/or chemoresistance. The expression of cytochrome P450 (CYP) enzymes in malignancies can affect the pharmacology of drugs commonly used in chemotherapy or confer susceptibility to development of chemical carcinogenesis; in addition, their specific tumor expression can be used as a therapeutic target. Using qPCR and Western blot assays, the expression of CYP1B1, CYP2E1, CYP3A4, and CYP3A5 were analyzed in a cohort of tumor tissue paired with non-malignant adjacent tissue of patients with NRSTS. The mRNA and protein expression of CYP1B1, CYP2E1, and CYP3A4 were significantly increased in tumor tissue. We propose that the expression of these isoforms is related to carcinogenesis and chemoresistance frequently observed in these neoplasms.
Collapse
Affiliation(s)
- Carmen Torres-Zárate
- Laboratorio de Toxicología Genética, Instituto Nacional de Pediatría, Mexico City, Mexico
| | - Araceli Vences-Mejía
- Laboratorio de Toxicología Genética, Instituto Nacional de Pediatría, Mexico City, Mexico
| | | | - Eduardo Díaz-Díaz
- Laboratorio de Toxicología Genética, Instituto Nacional de Pediatría, Mexico City, Mexico
| | | | | | | | | | - Rodolfo R Jurado
- Departamento de Anatomía Patológica, Instituto Nacional de Pediatría, Mexico City, Mexico
| | - Rebeca Santes-Palacios
- Laboratorio de Toxicología Genética, Instituto Nacional de Pediatría, Mexico City, Mexico
| | - Dora Molina-Ortiz
- Laboratorio de Toxicología Genética, Instituto Nacional de Pediatría, Mexico City, Mexico
| |
Collapse
|
14
|
Wu G, Tian Q, Liu J, Zhou Q, Zou D, Chen Z, Wu T, Wang W, Xia H, Zhou J. Comprehensive analysis of expression and prognosis for LMNB family genes in human sarcoma. Medicine (Baltimore) 2022; 101:e28933. [PMID: 35356902 PMCID: PMC10513290 DOI: 10.1097/md.0000000000028933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 02/08/2022] [Indexed: 11/26/2022] Open
Abstract
ABSTRACT Previous studies indicated that lamin proteins were thought to be related to gene expression, chromatin structure, and unclear stability. There are 2 types of vertebrate lamins, including A and B. The 2 B type proteins are encoded by lamin B1 (LMNB1) and lamin B2 (LMNB2). The LMNBs factor has been found to be associated with the development of multiple tumors, but its association with sarcoma has been barely mentioned.The transcription levels of LMNBs were analyzed via Oncomine database. Gene Expression Profiling Interactive Analysis (GEPIA) dataset was adopted to analyze the differential expression of LMNBs in sarcoma. Cancer Cell Line Encyclopedia dataset was used to explore the expression of LMNBs in sarcoma cell line. We analyzed the prognostic value of LMNBs in GEPIA and Kaplan-Meier Plotter. Oncomine and GEPIA datasets were also used to detect the relationship between LMNBs and their co-expressed genes. We used the Database for Annotation, Visualization and Integrated Discovery to conduct the Gene Ontology analysis of LMNBs and their co-expressed genes. Kyoto Encyclopedia of Genes and Genomes was also used to analyze the pathway of LMNBs.LMNB1 and LMNB2 were reported to be hyperexpressed in sarcoma. The expression of LMNBs was elevated in various sarcoma cell lines. According to the results, we observed that LMNBs were connected to the poor overall survival, recurrence-free survival, and disease-free survival of sarcoma patients.This study indicated that hyperexpression of LMNBs was significantly related to worse outcome of sarcoma, LMNB1 and LMNB2 were expected to become potential biomarkers for human.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Hong Xia
- * Correspondence: Hong Xia, Department of Orthopedics, Xiangtan Central Hospital, Xiangtan, Hunan 411100, China (e-mail: ); Jian Zhou,Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China (e-mail: ).
| | - Jian Zhou
- * Correspondence: Hong Xia, Department of Orthopedics, Xiangtan Central Hospital, Xiangtan, Hunan 411100, China (e-mail: ); Jian Zhou,Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China (e-mail: ).
| |
Collapse
|
15
|
Huot M, Caron M, Richer C, Djibo R, Najmanovich R, St-Onge P, Sinnett D, Raynal NJM. Repurposing proscillaridin A in combination with decitabine against embryonal rhabdomyosarcoma RD cells. Cancer Chemother Pharmacol 2021; 88:845-856. [PMID: 34331108 DOI: 10.1007/s00280-021-04339-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 07/15/2021] [Indexed: 12/13/2022]
Abstract
PURPOSE Embryonal rhabdomyosarcoma (eRMS) is the most common type of rhabdomyosarcoma in children. eRMS is characterized by malignant skeletal muscle cells driven by hyperactivation of several oncogenic pathways including the MYC pathway. Targeting MYC in cancer has been extremely challenging. Recently, we have demonstrated that the heart failure drug, proscillaridin A, produced anticancer effects with specificity toward MYC expressing leukemia cells. We also reported that decitabine, a hypomethylating drug, synergizes with proscillaridin A in colon cancer cells. Here, we investigated whether proscillaridin A exhibits epigenetic and anticancer activity against eRMS RD cells, overexpressing MYC oncogene, and its combination with decitabine. METHODS We investigated the anticancer effects of proscillaridin A in eRMS RD cells in vitro. In response to drug treatment, we measured growth inhibition, cell cycle arrest, loss of clonogenicity and self-renewal capacity. We further evaluated the impact of proscillaridin A on MYC expression and its downstream transcriptomic effects by RNA sequencing. Then, we measured protein expression of epigenetic regulators and their associated chromatin post-translational modifications in response to drug treatment. Chromatin immunoprecipitation sequencing data sets were coupled with transcriptomic results to pinpoint the impact of proscillaridin A on gene pathways associated with specific chromatin modifications. Lastly, we evaluated the effect of the combination of proscillaridin A and the DNA demethylating drug decitabine on eRMS RD cell growth and clonogenic potential. RESULTS Clinically relevant concentration of proscillaridin A (5 nM) produced growth inhibition, cell cycle arrest and loss of clonogenicity in eRMS RD cells. Proscillaridin A produced a significant downregulation of MYC protein expression and inhibition of oncogenic transcriptional programs controlled by MYC, involved in cell replication. Interestingly, significant reduction in total histone 3 acetylation and on specific lysine residues (lysine 9, 14, 18, and 27 on histone 3) was associated with significant protein downregulation of a series of lysine acetyltransferases (KAT3A, KAT3B, KAT2A, KAT2B, and KAT5). In addition, proscillaridin A produced synergistic growth inhibition and loss of clonogenicity when combined with the approved DNA demethylating drug decitabine. CONCLUSION Proscillaridin A produces anticancer and epigenetic effects in the low nanomolar range and its combination with decitabine warrants further investigation for the treatment of eRMS.
Collapse
Affiliation(s)
- Marielle Huot
- Département de Pharmacologie et Physiologie, Université de Montréal, Montréal, QC, Canada.,Sainte-Justine University Hospital Research Center, 3175, Chemin de la Côte-Sainte-Catherine, Montréal, QC, H3T 1C5, Canada
| | - Maxime Caron
- Sainte-Justine University Hospital Research Center, 3175, Chemin de la Côte-Sainte-Catherine, Montréal, QC, H3T 1C5, Canada
| | - Chantal Richer
- Sainte-Justine University Hospital Research Center, 3175, Chemin de la Côte-Sainte-Catherine, Montréal, QC, H3T 1C5, Canada
| | - Rahinatou Djibo
- Département de Pharmacologie et Physiologie, Université de Montréal, Montréal, QC, Canada.,Sainte-Justine University Hospital Research Center, 3175, Chemin de la Côte-Sainte-Catherine, Montréal, QC, H3T 1C5, Canada
| | - Rafael Najmanovich
- Département de Pharmacologie et Physiologie, Université de Montréal, Montréal, QC, Canada
| | - Pascal St-Onge
- Sainte-Justine University Hospital Research Center, 3175, Chemin de la Côte-Sainte-Catherine, Montréal, QC, H3T 1C5, Canada
| | - Daniel Sinnett
- Sainte-Justine University Hospital Research Center, 3175, Chemin de la Côte-Sainte-Catherine, Montréal, QC, H3T 1C5, Canada.,Département de Pédiatrie, Université de Montréal, Montréal, QC, Canada
| | - Noël J M Raynal
- Département de Pharmacologie et Physiologie, Université de Montréal, Montréal, QC, Canada. .,Sainte-Justine University Hospital Research Center, 3175, Chemin de la Côte-Sainte-Catherine, Montréal, QC, H3T 1C5, Canada.
| |
Collapse
|
16
|
Heske CM, Mascarenhas L. Relapsed Rhabdomyosarcoma. J Clin Med 2021; 10:804. [PMID: 33671214 PMCID: PMC7922213 DOI: 10.3390/jcm10040804] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/02/2021] [Accepted: 02/13/2021] [Indexed: 12/14/2022] Open
Abstract
Relapsed rhabdomyosarcoma (RMS) represents a significant therapeutic challenge. Nearly one-third of patients diagnosed with localized RMS and over two-thirds of patients with metastatic RMS will experience disease recurrence following primary treatment, generally within three years. Clinical features at diagnosis, including primary site, tumor invasiveness, size, stage, and histology impact likelihood of relapse and prognosis post-relapse. Aspects of initial treatment, including extent of surgical resection, use of radiotherapy, and chemotherapy regimen, are also associated with post-relapse outcomes, as are features of the relapse itself, including time to relapse and extent of disease involvement. Although there is no standard treatment for patients with relapsed RMS, several general principles, including tissue biopsy confirmation of diagnosis, assessment of post-relapse prognosis, determination of the feasibility of additional local control measures, and discussion of patient goals, should all be part of the approach to care. Patients with features suggestive of a favorable prognosis, which include those with botryoid RMS or stage 1 or group I embryonal RMS (ERMS) who have had no prior treatment with cyclophosphamide, have the highest chance of achieving long-term cure when treated with a multiagent chemotherapy regimen at relapse. Unfortunately, patients who do not meet these criteria represent the majority and have poor outcomes when treated with such regimens. For this group, strong consideration should be given for enrollment on a clinical trial.
Collapse
Affiliation(s)
- Christine M. Heske
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Leo Mascarenhas
- Cancer and Blood Disease Institute, Children’s Hospital Los Angeles, Division of Hematology/Oncology, Department of Pediatrics and Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90027, USA;
| |
Collapse
|
17
|
Westermann C, Weller J, Pedroso F, Canner J, Pratilas CA, Rhee DS. Socioeconomic and health care coverage disparities in children, adolescents, and young adults with sarcoma. Pediatr Blood Cancer 2020; 67:e28708. [PMID: 32939963 DOI: 10.1002/pbc.28708] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 08/31/2020] [Accepted: 09/01/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Socioeconomic and health care coverage disparities are established as poor prognostic markers in adults with sarcoma, but few studies examine these differences among pediatric, adolescents and young adults (AYA). This study examines the association between socioeconomic status (SES), insurance status, and disease presentation among children and AYA patients with sarcoma. METHODS This is a retrospective cohort study of patients aged 0-25 years with bone or soft tissue sarcoma from the National Cancer Database. SES assignments were based on estimated median income and education level. Patient demographics and clinical factors were compared by SES and insurance status. Multivariate logistic regression models were fitted to determine adjusted odds ratios of SES and insurance status on metastatic disease or tumor size ≥5 cm at time of presentation. RESULTS In a cohort of 9112 patients, 2932 (32.1%) had low, 2084 (22.8%) middle, and 4096 (44.9%) high SES. For insurance status, 5864 (64.3%) had private, 2737 (30.0%) public, and 511 (5.6%) were uninsured. Compared to high SES, patients with low SES were more likely to have metastatic disease (OR = 1.16, P = .03) and tumors ≥5 cm (OR = 1.29, P < .01). Compared to private insurance, public and no insurance were associated with metastatic disease (OR = 1.35, P < .01 and OR = 1.32, P = .02) and increased tumors ≥5 cm (OR = 1.28, P < .01 and OR = 1.67, P < .01). CONCLUSIONS SES disparities exist among children and AYA patients with sarcoma. Low SES and public or no insurance are associated with advanced disease at presentation. Further studies are needed to identify interventions to improve earlier detection of sarcomas in at-risk children and young adults.
Collapse
Affiliation(s)
- Carly Westermann
- Virginia Polytechnic Institute and State University, Blacksburg, Virginia
| | - Jennine Weller
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Felipe Pedroso
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Joe Canner
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Christine A Pratilas
- Division of Pediatric Oncology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
| | - Daniel S Rhee
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
18
|
Li YL, Gao YL, Niu XL, Wu YT, Du YM, Tang MS, Li JY, Guan XH, Song B. Identification of Subtype-Specific Metastasis-Related Genetic Signatures in Sarcoma. Front Oncol 2020; 10:544956. [PMID: 33123466 PMCID: PMC7573283 DOI: 10.3389/fonc.2020.544956] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 08/28/2020] [Indexed: 12/18/2022] Open
Abstract
Background: Sarcomas are heterogeneous rare malignancies constituting approximately 1% of all solid cancers in adults and including more than 70 histological and molecular subtypes with different pathological and clinical development characteristics. Method: We identified prognostic biomarkers of sarcomas by integrating clinical information and RNA-seq data from TCGA and GEO databases. In addition, results obtained from cell cycle, cell migration, and invasion assays were used to assess the capacity for Tanespimycin to inhibit the proliferation and metastasis of sarcoma. Results: Sarcoma samples (N = 536) were divided into four pathological subtypes including DL (dedifferentiated liposarcoma), LMS (leiomyosarcoma), UPS (undifferentiated pleomorphic sarcomas), and MFS (myxofibrosarcoma). RNA-seq expression profile data from the TCGA dataset were used to analyze differentially expressed genes (DEGs) within metastatic and non-metastatic samples of these four sarcoma pathological subtypes with DEGs defined as metastatic-related signatures (MRS). Prognostic analysis of MRS identified a group of genes significantly associated with prognosis in three pathological subtypes: DL, LMS, and UPS. ISG15, NUP50, PTTG1, SERPINE1, and TSR1 were found to be more likely associated with adverse prognosis. We also identified Tanespimycin as a drug exerting inhibitory effects on metastatic LMS subtype and therefore can serve a potential treatment for this type of sarcoma. Conclusions: These results provide new insights into the pathogenesis, diagnosis, treatment, and prognosis of sarcomas and provide new directions for further study of sarcoma.
Collapse
Affiliation(s)
- Ya-Ling Li
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, China.,National Health Commission Key Laboratory of Immunodermatology (China Medical University), Shenyang, China.,Key Laboratory of Immunodermatology, Ministry of Education, Shenyang, China
| | - Ya-Li Gao
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, China.,National Health Commission Key Laboratory of Immunodermatology (China Medical University), Shenyang, China.,Key Laboratory of Immunodermatology, Ministry of Education, Shenyang, China
| | - Xue-Li Niu
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, China.,National Health Commission Key Laboratory of Immunodermatology (China Medical University), Shenyang, China.,Key Laboratory of Immunodermatology, Ministry of Education, Shenyang, China
| | - Yu-Tong Wu
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, China.,National Health Commission Key Laboratory of Immunodermatology (China Medical University), Shenyang, China.,Key Laboratory of Immunodermatology, Ministry of Education, Shenyang, China
| | - Yi-Mei Du
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, China.,National Health Commission Key Laboratory of Immunodermatology (China Medical University), Shenyang, China.,Key Laboratory of Immunodermatology, Ministry of Education, Shenyang, China
| | - Ming-Sui Tang
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, China.,National Health Commission Key Laboratory of Immunodermatology (China Medical University), Shenyang, China.,Key Laboratory of Immunodermatology, Ministry of Education, Shenyang, China
| | - Jing-Yi Li
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, China.,National Health Commission Key Laboratory of Immunodermatology (China Medical University), Shenyang, China.,Key Laboratory of Immunodermatology, Ministry of Education, Shenyang, China
| | - Xiu-Hao Guan
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, China.,National Health Commission Key Laboratory of Immunodermatology (China Medical University), Shenyang, China.,Key Laboratory of Immunodermatology, Ministry of Education, Shenyang, China
| | - Bing Song
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, China.,School of Dentistry, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
19
|
Cabrera-Andrade A, López-Cortés A, Munteanu CR, Pazos A, Pérez-Castillo Y, Tejera E, Arrasate S, González-Díaz H. Perturbation-Theory Machine Learning (PTML) Multilabel Model of the ChEMBL Dataset of Preclinical Assays for Antisarcoma Compounds. ACS OMEGA 2020; 5:27211-27220. [PMID: 33134682 PMCID: PMC7594149 DOI: 10.1021/acsomega.0c03356] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 10/06/2020] [Indexed: 06/11/2023]
Abstract
Sarcomas are a group of malignant neoplasms of connective tissue with a different etiology than carcinomas. The efforts to discover new drugs with antisarcoma activity have generated large datasets of multiple preclinical assays with different experimental conditions. For instance, the ChEMBL database contains outcomes of 37,919 different antisarcoma assays with 34,955 different chemical compounds. Furthermore, the experimental conditions reported in this dataset include 157 types of biological activity parameters, 36 drug targets, 43 cell lines, and 17 assay organisms. Considering this information, we propose combining perturbation theory (PT) principles with machine learning (ML) to develop a PTML model to predict antisarcoma compounds. PTML models use one function of reference that measures the probability of a drug being active under certain conditions (protein, cell line, organism, etc.). In this paper, we used a linear discriminant analysis and neural network to train and compare PT and non-PT models. All the explored models have an accuracy of 89.19-95.25% for training and 89.22-95.46% in validation sets. PTML-based strategies have similar accuracy but generate simplest models. Therefore, they may become a versatile tool for predicting antisarcoma compounds.
Collapse
Affiliation(s)
- Alejandro Cabrera-Andrade
- Grupo
de Bio-Quimioinformática, Universidad
de Las Américas, de los Granados Avenue, Quito 170125, Ecuador
- Carrera
de Enfermería, Facultad de Ciencias de la Salud, Universidad de Las Américas, de los Granados Avenue, Quito 170125, Ecuador
- RNASA-IMEDIR,
Computer Sciences Faculty, University of
A Coruña, A Coruña 15071, Spain
| | - Andrés López-Cortés
- RNASA-IMEDIR,
Computer Sciences Faculty, University of
A Coruña, A Coruña 15071, Spain
- Centro
de Investigación Genética y Genómica, Facultad
de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Mariscal Sucre Avenue, Quito 170129, Ecuador
| | - Cristian R. Munteanu
- RNASA-IMEDIR,
Computer Sciences Faculty, University of
A Coruña, A Coruña 15071, Spain
- Biomedical
Research Institute of A Coruña (INIBIC), University Hospital Complex of A Coruña (CHUAC), A Coruña 15006, Spain
- Centro de
Investigación en Tecnologías de la Información
y las Comunicaciones (CITIC), Campus de
Elviña s/n, A Coruña 15071, Spain
| | - Alejandro Pazos
- RNASA-IMEDIR,
Computer Sciences Faculty, University of
A Coruña, A Coruña 15071, Spain
- Biomedical
Research Institute of A Coruña (INIBIC), University Hospital Complex of A Coruña (CHUAC), A Coruña 15006, Spain
| | - Yunierkis Pérez-Castillo
- Grupo
de Bio-Quimioinformática, Universidad
de Las Américas, de los Granados Avenue, Quito 170125, Ecuador
- Escuela
de Ciencias Físicas y Matemáticas, Universidad de Las Américas, de los Granados Avenue, Quito 170125, Ecuador
| | - Eduardo Tejera
- Grupo
de Bio-Quimioinformática, Universidad
de Las Américas, de los Granados Avenue, Quito 170125, Ecuador
- Facultad
de Ingeniería y Ciencias Aplicadas, Universidad de Las Américas, de los Granados Avenue, Quito 170125, Ecuador
| | - Sonia Arrasate
- Department
of Organic Chemistry II and Basque Center for Biophysics, University of Basque Country UPV/EHU, Leioa 48940, Biscay, Spain
| | - Humbert González-Díaz
- Department
of Organic Chemistry II and Basque Center for Biophysics, University of Basque Country UPV/EHU, Leioa 48940, Biscay, Spain
- Ikerbasque,
Basque Foundation for Science, Bilbao 48011, Biscay, Spain
| |
Collapse
|
20
|
Dionísio FCF, Oliveira LS, Hernandes MA, Engel EE, Rangayyan RM, Azevedo-Marques PM, Nogueira-Barbosa MH. Manual and semiautomatic segmentation of bone sarcomas on MRI have high similarity. ACTA ACUST UNITED AC 2020; 53:e8962. [PMID: 32022102 PMCID: PMC6993358 DOI: 10.1590/1414-431x20198962] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 10/14/2019] [Indexed: 02/07/2023]
Abstract
The aims of this study were to evaluate the intra- and interobserver reproducibility of manual segmentation of bone sarcomas in magnetic resonance imaging (MRI) studies and to compare manual and semiautomatic segmentation methods. This retrospective study included twelve osteosarcoma and eight Ewing sarcoma MRI studies performed prior to any therapeutic intervention. All cases were histopathologically confirmed. Three radiologists used 3D-Slicer software to perform manual segmentation of bone sarcomas in a blinded and independent manner. One radiologist segmented manually and also performed semiautomatic segmentation with the GrowCut tool. Segmentation exercises were timed for comparison. The dice similarity coefficient (DSC) and Hausdorff distance (HD) were used to evaluate similarity between the segmentation results and further statistical analyses were performed to compare DSC, HD, and volumetric results. Manual segmentation was reproducible with intraobserver DSC varying from 0.83 to 0.97 and HD from 3.37 to 28.73 mm. Interobserver DSC of manual segmentation showed variation from 0.73 to 0.97 and HD from 3.93 to 33.40 mm. Semiautomatic segmentation compared to manual segmentation resulted in DSCs of 0.71−0.96 and HDs of 5.38−31.54 mm. Semiautomatic segmentation required significantly less time compared to manual segmentation (P value ≤0.05). Among all situations compared, tumor volumetry did not show significant statistical differences (P value >0.05). We found excellent intra- and interobserver agreement for manual segmentation of osteosarcoma and Ewing sarcoma. There was high similarity between manual and semiautomatic segmentation, with a significant reduction of segmentation time using the semiautomatic method.
Collapse
Affiliation(s)
- F C F Dionísio
- Departamento de Imagens Médicas, Hematologia e Oncologia Clínica, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil.,Laboratório de Pesquisa em Imagens Musculoesqueléticas, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| | - L S Oliveira
- Departamento de Imagens Médicas, Hematologia e Oncologia Clínica, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil.,Laboratório de Pesquisa em Imagens Musculoesqueléticas, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| | - M A Hernandes
- Departamento de Imagens Médicas, Hematologia e Oncologia Clínica, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| | - E E Engel
- Departamento de Imagens Médicas, Hematologia e Oncologia Clínica, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| | - R M Rangayyan
- Department of Electrical and Computer Engineering, University of Calgary, Calgary, Alberta, Canada
| | - P M Azevedo-Marques
- Departamento de Imagens Médicas, Hematologia e Oncologia Clínica, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| | - M H Nogueira-Barbosa
- Departamento de Imagens Médicas, Hematologia e Oncologia Clínica, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil.,Laboratório de Pesquisa em Imagens Musculoesqueléticas, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| |
Collapse
|
21
|
Penumarthy NL, Goldsby RE, Shiboski SC, Wustrack R, Murphy P, Winestone LE. Insurance impacts survival for children, adolescents, and young adults with bone and soft tissue sarcomas. Cancer Med 2019; 9:951-958. [PMID: 31838786 PMCID: PMC6997066 DOI: 10.1002/cam4.2739] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 11/15/2019] [Indexed: 12/18/2022] Open
Abstract
Background While racial/ethnic survival disparities have been described in pediatric oncology, the impact of income has not been extensively explored. We analyzed how public insurance influences 5‐year overall survival (OS) in young patients with sarcomas. Methods The University of California San Francisco Cancer Registry was used to identify patients aged 0‐39 diagnosed with bone or soft tissue sarcomas between 2000 and 2015. Low‐income patients were defined as those with no insurance or Medicaid, a means‐tested form of public insurance. Survival curves were computed using the Kaplan‐Meier method and compared using log‐rank tests and Cox models. Causal mediation was used to assess whether the association between public insurance and mortality is mediated by metastatic disease. Results Of 1106 patients, 39% patients were classified as low‐income. Low‐income patients were more likely to be racial/ethnic minorities and to present with metastatic disease (OR 1.96, 95% CI 1.35‐2.86). Low‐income patients had significantly worse OS (61% vs 71%). Age at diagnosis and extent of disease at diagnosis were also independent predictors of OS. When stratified by extent of disease, low‐income patients consistently had significantly worse OS (localized: 78% vs 84%, regional: 64% vs 73%, metastatic: 23% vs 30%, respectively). Mediation analysis indicated that metastatic disease at diagnosis mediated 15% of the effect of public insurance on OS. Conclusions Low‐income patients with bone and soft tissue sarcomas had decreased OS regardless of disease stage at presentation. The mechanism by which insurance status impacts survival requires additional investigation, but may be through reduced access to care.
Collapse
Affiliation(s)
- Neela L Penumarthy
- Division of Bioethics and Palliative Care, Department of Pediatrics, University of Washington School of Medicine, Seattle Children's Hospital, Seattle, WA, USA
| | - Robert E Goldsby
- Division of Oncology, Department of Pediatrics, UCSF Benioff Children's Hospital, San Francisco, CA, USA.,Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, CA, USA
| | - Stephen C Shiboski
- Department of Epidemiology and Biostatistics, UCSF, San Francisco, CA, USA
| | | | - Patricia Murphy
- Division of Oncology, Department of Pediatrics, UCSF Benioff Children's Hospital, San Francisco, CA, USA
| | - Lena E Winestone
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, CA, USA.,Division of Allergy, Immunology, and BMT, Department of Pediatrics, UCSF Benioff Children's Hospital, San Francisco, CA, USA
| |
Collapse
|
22
|
Panagopoulos D, Themistocleous M, Apostolopoulou K, Sfakianos G. Primary, Dural-Based, Ewing Sarcoma Manifesting with Seizure Activity: Presentation of a Rare Tumor Entity with Literature Review. World Neurosurg 2019; 129:216-220. [DOI: 10.1016/j.wneu.2019.06.036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 06/01/2019] [Accepted: 06/03/2019] [Indexed: 12/29/2022]
|
23
|
Xu Q, Gao T, Zhang B, Zeng J, Dai M. Primary osteosarcoma in elderly patients: A report of three cases. Oncol Lett 2019; 18:990-996. [PMID: 31423158 PMCID: PMC6607338 DOI: 10.3892/ol.2019.10446] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Accepted: 03/07/2019] [Indexed: 02/04/2023] Open
Abstract
Osteosarcoma is the most common type of primary malignant bone tumor in children and young adults. However, primary osteosarcoma in elderly patients is rare. The present study reports 3 cases of advanced osteosarcoma in elderly patients. The pathological findings in all 3 cases confirmed the diagnosis of primary osteosarcoma. Notably, each patient received different treatment options. Chemoradiotherapy was recommended in case 1 due to the age of the patient. However, the patient requested to be discharged and was lost to follow-up. Conversely, in case 2, the 62-year-old female patient underwent systemic chemotherapy, but no surgical treatment, and in case 3, the 51-year-old male patient underwent complete tumor resection and received systemic chemotherapy for late tumor recurrence. Early diagnosis of osteosarcoma in elderly patients is difficult, and misdiagnosis or a missed diagnosis is common. In clinical practice, bone tumors in elderly patients should be investigated carefully. Imaging examinations are essential for diagnosis, and biopsy is required for confirmation. However, the efficacy of chemotherapy for elderly patients with primary osteosarcoma remains uncertain. Collectively, due to the small number of reports of osteosarcoma in the elderly population, the 3 cases in the present study raise awareness of this rare condition.
Collapse
Affiliation(s)
- Qiang Xu
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, Jiangxi 330006, P.R. China
| | - Tian Gao
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, Jiangxi 330006, P.R. China
| | - Bin Zhang
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, Jiangxi 330006, P.R. China
| | - Jin Zeng
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, Jiangxi 330006, P.R. China
| | - Min Dai
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
24
|
Rainusso N, Cleveland H, Hernandez JA, Quintanilla NM, Hicks J, Vasudevan S, Marco RAW, Allen-Rhoades W, Wang LL, Yustein JT. Generation of patient-derived tumor xenografts from percutaneous tumor biopsies in children with bone sarcomas. Pediatr Blood Cancer 2019; 66:e27579. [PMID: 30548185 DOI: 10.1002/pbc.27579] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 10/26/2018] [Accepted: 11/21/2018] [Indexed: 11/07/2022]
Abstract
One of the limitations of performing percutaneous biopsies in patients with bone sarcomas is the small amount of tumor that can be obtained for research purposes. Here, we describe our experience developing patient-derived tumor xenografts (PDXs) using percutaneous tumor biopsies in children with bone sarcomas. We generated 14 bone sarcoma PDXs from percutaneous tumor biopsies. We also developed eight bone sarcoma PDXs from surgical resection of primary bone tumors and pulmonary metastases. A multidisciplinary team approach was critical to establish an accurate diagnosis and to provide adequate tumor samples for PDX generation.
Collapse
Affiliation(s)
- Nino Rainusso
- Texas Children's Cancer & Hematology Centers, Houston, Texas.,Department of Pediatrics, Division of Hematology-Oncology, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Heather Cleveland
- Department of Interventional Radiology, Texas Children's Hospital, Houston, Texas
| | - J Alberto Hernandez
- Department of Radiology, Interventional Radiology Section, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas
| | - Norma M Quintanilla
- Department of Pathology and Immunology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas
| | - John Hicks
- Department of Pathology and Immunology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas
| | - Sanjeev Vasudevan
- Pediatric Surgical Oncology Laboratory, Texas Children's Surgical Oncology Program, Houston, Texas.,M.E. DeBakey Department of Surgery, Division of Pediatric Surgery, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Rex A W Marco
- Department of Orthopedics, Houston Methodist Hospital, Houston, Texas
| | - Wendy Allen-Rhoades
- Texas Children's Cancer & Hematology Centers, Houston, Texas.,Department of Pediatrics, Division of Hematology-Oncology, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Lisa L Wang
- Texas Children's Cancer & Hematology Centers, Houston, Texas.,Department of Pediatrics, Division of Hematology-Oncology, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Jason T Yustein
- Texas Children's Cancer & Hematology Centers, Houston, Texas.,Department of Pediatrics, Division of Hematology-Oncology, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
25
|
Cheng L, Pandya PH, Liu E, Chandra P, Wang L, Murray ME, Carter J, Ferguson M, Saadatzadeh MR, Bijangi-Visheshsaraei K, Marshall M, Li L, Pollok KE, Renbarger JL. Integration of genomic copy number variations and chemotherapy-response biomarkers in pediatric sarcoma. BMC Med Genomics 2019; 12:23. [PMID: 30704460 PMCID: PMC6357363 DOI: 10.1186/s12920-018-0456-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background While most pediatric sarcomas respond to front-line therapy, some bone sarcomas do not show radiographic response like soft-tissue sarcomas (rhabdomyosarccomas) but do show 90% necrosis. Though, new therapies are urgently needed to improve survival and quality of life in pediatric patients with sarcomas. Complex chromosomal aberrations such as amplifications and deletions of DNA sequences are frequently observed in pediatric sarcomas. Evaluation of copy number variations (CNVs) associated with pediatric sarcoma patients at the time of diagnosis or following therapy offers an opportunity to assess dysregulated molecular targets and signaling pathways that may drive sarcoma development, progression, or relapse. The objective of this study was to utilize publicly available data sets to identify potential predictive biomarkers of chemotherapeutic response in pediatric Osteosarcoma (OS), Rhabdomyosarcoma (RMS) and Ewing’s Sarcoma Family of Tumors (ESFTs) based on CNVs following chemotherapy (OS n = 117, RMS n = 64, ESFTs n = 25 tumor biopsies). Methods There were 206 CNV profiles derived from pediatric sarcoma biopsies collected from the public databases TARGET and NCBI-Gene Expression Omnibus (GEO). Through our comparative genomic analyses of OS, RMS, and ESFTs and 22,255 healthy individuals called from the Database of Genomic Variants (DGV), we identified CNVs (amplifications and deletions) pattern of genomic instability in these pediatric sarcomas. By integrating CNVs of Cancer Cell Line Encyclopedia (CCLE) identified in the pool of genes with drug-response data from sarcoma cell lines (n = 27) from Cancer Therapeutics Response Portal (CTRP) Version 2, potential predictive biomarkers of therapeutic response were identified. Results Genes associated with survival and/recurrence of these sarcomas with statistical significance were found on long arm of chromosome 8 and smaller aberrations were also identified at chromosomes 1q, 12q and x in OS, RMS, and ESFTs. A pool of 63 genes that harbored amplifications and/or deletions were frequently associated with recurrence across OS, RMS, and ESFTs. Correlation analysis of CNVs from CCLE with drug-response data of CTRP in 27 sarcoma cell lines, 33 CNVs out of 63 genes correlated with either sensitivity or resistance to 17 chemotherapies from which actionable CNV signatures such as IGF1R, MYC, MAPK1, ATF1, and MDM2 were identified. These CNV signatures could potentially be used to delineate patient populations that will respond versus those that will not respond to a particular chemotherapy. Conclusions The large-scale analyses of CNV-drug screening provides a platform to evaluate genetic alterations across aggressive pediatric sarcomas. Additionally, this study provides novel insights into the potential utilization of CNVs as not only prognostic but also as predictive biomarkers of therapeutic response. Information obtained in this study may help guide and prioritize patient-specific therapeutic options in pediatric bone and soft-tissue sarcomas. Electronic supplementary material The online version of this article (10.1186/s12920-018-0456-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lijun Cheng
- Department of Biomedical Informatics, College of Medicine, Ohio State University, Columbus, OH, 43210, USA
| | - Pankita H Pandya
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA.,Division of Hematology/Oncology, Department of Pediatrics, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA
| | - Enze Liu
- Department of Biomedical Informatics, College of Medicine, Ohio State University, Columbus, OH, 43210, USA.,Center for Computational Biology and Bioinformatics, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA
| | - Pooja Chandra
- Center for Computational Biology and Bioinformatics, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA
| | - Limei Wang
- Department of Biomedical Informatics, College of Medicine, Ohio State University, Columbus, OH, 43210, USA
| | - Mary E Murray
- Division of Hematology/Oncology, Department of Pediatrics, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA
| | - Jacquelyn Carter
- Division of Hematology/Oncology, Department of Pediatrics, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA
| | - Michael Ferguson
- Division of Hematology/Oncology, Department of Pediatrics, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA
| | - Mohammad Reza Saadatzadeh
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA.,Division of Hematology/Oncology, Department of Pediatrics, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA
| | - Khadijeh Bijangi-Visheshsaraei
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA.,Division of Hematology/Oncology, Department of Pediatrics, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA
| | - Mark Marshall
- Division of Hematology/Oncology, Department of Pediatrics, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA
| | - Lang Li
- Department of Biomedical Informatics, College of Medicine, Ohio State University, Columbus, OH, 43210, USA. .,Herman B Wells Center for Pediatric Research, Department of Pediatrics, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA.
| | - Karen E Pollok
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA. .,Division of Hematology/Oncology, Department of Pediatrics, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA. .,Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN, 46202, USA.
| | - Jamie L Renbarger
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA. .,Division of Hematology/Oncology, Department of Pediatrics, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA. .,Center for Computational Biology and Bioinformatics, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA. .,Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN, 46202, USA. .,Indiana Institute of Personalized Medicine, Indiana University, Indianapolis, IN, 46202, USA.
| |
Collapse
|
26
|
Aydin S, Yuksel O, Tanritanir R, Aydin AE, Celik SE. Primary Ewing Sarcoma of Frontotemporal Bone in Geriatric Patient. World Neurosurg 2018; 115:278-281. [PMID: 29715573 DOI: 10.1016/j.wneu.2018.04.158] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 04/20/2018] [Accepted: 04/21/2018] [Indexed: 11/28/2022]
Abstract
BACKGROUND Ewing sarcoma is a rare primary malignant bone tumor, which mainly affects children and adolescents. Calvarial bone involvement and its appearance in elderly patients are extremely rare. CASE DESCRIPTION We presented a 68-year-old female patient with headache and right frontotemporal swelling. Imaging studies showed a right frontotemporal mass expanding to the Sylvian fissure. The patient underwent total resection of the mass, and pathologic evaluation ensured the diagnosis of primary Ewing sarcoma. The patient had adjuvant radiotherapy and chemotherapy after surgery. CONCLUSIONS Prognosis and efficiency of treatment for primary Ewing sarcoma of skull are unclear in elderly patients because of the sarcoma's rare appearance. Therefore more clinical evaluation is necessary. This case is the oldest patient presented in the literature.
Collapse
Affiliation(s)
- Seckin Aydin
- Department of Neurosurgery, T. C. Ministry of Health Okmeydani Education and Research Hospital, Istanbul, Turkey
| | - Odhan Yuksel
- Department of Neurosurgery, Aspendos Anadolu Hospital, Antalya, Turkey.
| | - Rahime Tanritanir
- Department of Pathology, T. C. Ministry of Health Okmeydani Education and Research Hospital, Istanbul, Turkey
| | - Aysegul Esen Aydin
- T. C. Ministry of Health Bakirkoy Research and Training Hospital for Neurology Neurosurgery and Psychiatry, Istanbul, Turkey
| | - Suat Erol Celik
- Department of Neurosurgery, T. C. Ministry of Health Okmeydani Education and Research Hospital, Istanbul, Turkey
| |
Collapse
|
27
|
Kim C, Choi YB, Lee JW, Yoo KH, Sung KW, Koo HH. Excellent treatment outcomes in children younger than 18 months with stage 4 MYCN nonamplified neuroblastoma. KOREAN JOURNAL OF PEDIATRICS 2018; 61:53-58. [PMID: 29563945 PMCID: PMC5854843 DOI: 10.3345/kjp.2018.61.2.53] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 09/11/2017] [Accepted: 09/26/2017] [Indexed: 11/27/2022]
Abstract
Purpose Although the prognosis is generally good in patients with intermediate-risk neuroblastoma, no consensus has been reached on the ideal treatment regimen. This study analyzed treatment outcomes and toxicities in patients younger than 18 months with stage 4 MYCN nonamplified neuroblastoma. Methods We retrospectively analyzed 20 patients younger than 18 months newly diagnosed with stage 4 MYCN nonamplified neuroblastoma between January 2009 and December 2015. Patients received 9 cycles of chemotherapy and surgery, with or without local radiotherapy, followed by 12 cycles of differentiation therapy with 13-cis-retinoic acid. Chemotherapy consisted of alternating cycles of cisplatin, etoposide, doxorubicin, and cyclophosphamide (CEDC) and ifosfamide, carboplatin, and etoposide (ICE) regimens. Results The most common primary tumor site was the abdomen (85%), and the most common metastatic sites were the lymph nodes (65%), followed by the bones (60%), liver (55%), skin (45%), and bone marrow (25%). At the end of induction therapy, 14 patients (70%) achieved complete response, with 1 achieving very good partial response, 4 achieving partial response, and 1 showing mixed response. Nine patients (45%) received local radiotherapy. At a median follow-up of 47 months (range, 17–91 months), none of these patients experienced relapse, progression, or secondary malignancy, or died. Three years after chemotherapy completion, none of the patients had experienced grade ≥3 late adverse effects. Conclusion Patients younger than 18 months with stage 4 MYCN nonamplified neuroblastoma showed excellent outcomes, without significant late adverse effects, when treated with alternating cycles of CEDC and ICE, followed by surgery and differentiation therapy.
Collapse
Affiliation(s)
- Chiwoo Kim
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Young Bae Choi
- Department of Pediatrics, Chung-Ang University Hospital, Seoul, Korea
| | - Ji Won Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Keon Hee Yoo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ki Woong Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hong Hoe Koo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
28
|
Cassinelli G, Favini E, Dal Bo L, Tortoreto M, De Maglie M, Dagrada G, Pilotti S, Zunino F, Zaffaroni N, Lanzi C. Antitumor efficacy of the heparan sulfate mimic roneparstat (SST0001) against sarcoma models involves multi-target inhibition of receptor tyrosine kinases. Oncotarget 2018; 7:47848-47863. [PMID: 27374103 PMCID: PMC5216983 DOI: 10.18632/oncotarget.10292] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 05/08/2016] [Indexed: 12/20/2022] Open
Abstract
The heparan sulfate (HS) mimic/heparanase inhibitor roneparstat (SST0001) shows antitumor activity in preclinical sarcoma models. We hypothesized that this 100% N-acetylated and glycol-split heparin could interfere with the functions of several receptor tyrosine kinases (RTK) coexpressed in sarcomas and activated by heparin-binding growth factors. Using a phospho-proteomic approach, we investigated the drug effects on RTK activation in human cell lines representative of different sarcoma subtypes. Inhibition of FGF, IGF, ERBB and PDGF receptors by the drug was biochemically and functionally validated. Roneparstat counteracted the autocrine loop induced by the COL1A1/PDGFB fusion oncogene, expressed in a human dermatofibrosarcoma protuberans primary culture and in NIH3T3COL1A1/PDGFB transfectants, inhibiting cell anchorage-independent growth and invasion. In addition, roneparstat inhibited the activation of cell surface PDGFR and PDGFR-associated FAK, likely contributing to the reversion of NIH3T3COL1A1/PDGFB cell transformed and pro-invasive phenotype. Biochemical and histological/immunohistochemical ex vivo analyses confirmed a reduced activation of ERBB4, EGFR, INSR, IGF1R, associated with apoptosis induction and angiogenesis inhibition in a drug-treated Ewing's sarcoma family tumor xenograft. The combination of roneparstat with irinotecan significantly improved the antitumor effect against A204 rhabdoid xenografts resulting in a high rate of complete responses and cures. These findings reveal that roneparstat exerts a multi-target inhibition of RTKs relevant in the pathobiology of different sarcoma subtypes. These effects, likely cooperating with heparanase inhibition, contribute to the antitumor efficacy of the drug. The study supports heparanase/HS axis targeting as a valuable approach in combination therapies of different sarcoma subtypes providing a preclinical rationale for clinical investigation.
Collapse
Affiliation(s)
- Giuliana Cassinelli
- Molecular Pharmacology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Enrica Favini
- Molecular Pharmacology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Laura Dal Bo
- Molecular Pharmacology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Monica Tortoreto
- Molecular Pharmacology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Marcella De Maglie
- Department of Veterinary Sciences and Public Health, Università Degli Studi di Milano, Milan, Italy.,Mouse and Animal Pathology Laboratory, Fondazione Filarete, Milan, Italy
| | - Gianpaolo Dagrada
- Laboratory of Experimental Molecular Pathology, Department of Diagnostic Pathology and Laboratory, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Silvana Pilotti
- Laboratory of Experimental Molecular Pathology, Department of Diagnostic Pathology and Laboratory, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Franco Zunino
- Molecular Pharmacology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Nadia Zaffaroni
- Molecular Pharmacology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Cinzia Lanzi
- Molecular Pharmacology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
29
|
Xin M, Qiao Z, Li J, Liu J, Song S, Zhao X, Miao P, Tang T, Wang L, Liu W, Yang X, Dai K, Huang G. miR-22 inhibits tumor growth and metastasis by targeting ATP citrate lyase: evidence in osteosarcoma, prostate cancer, cervical cancer and lung cancer. Oncotarget 2018; 7:44252-44265. [PMID: 27317765 PMCID: PMC5190093 DOI: 10.18632/oncotarget.10020] [Citation(s) in RCA: 131] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 05/11/2016] [Indexed: 12/15/2022] Open
Abstract
MicroRNAs (miRNAs) are non-coding small RNAs that function as negative regulators of gene expression involving in the tumor biology. ATP citrate lyase (ACLY), a key enzyme initiating de novo lipid synthesis, has been found to be upregulated in cancer cells, and its inhibition causes suppressive effects in a variety of tumors. At present, although several ACLY inhibitors have been reported, the potential role of miRNAs in interfering ACLY still needs further clarification. Herein, four different types of tumor cells including osteosarcoma, prostate, cervical and lung cancers were adopted in our study, and we have demonstrated that miR-22 directly downregulated ACLY. Moreover, miR-22 was proved to attenuate cancer cell proliferation and invasion, as well as promote cell apoptosis via inhibiting ACLY. Additionally, we confirmed the higher ACLY protein levels and the lower miR-22 expressions in hundreds of clinical samples of the four primary tumors, and a negative correlation relationship between ACLY and miR-22 was clarified. Finally, in the four animal models, we found that along with the loss of the ACLY expression, the miR-22-treated mice developed rather smaller tumors, less probabilities of distant metastasis, and fairly longer survivals. De novo lipogenesis suppression triggered by miR-22-ACLY axis may contribute to the inhibition of tumor growth and metastasis. These findings provide unequivocal proofs that miR-22 is responsible for the posttranscriptional regulation of ACLY, which yields promising therapeutic effects in osteosarcoma, prostate, cervical and lung cancers.
Collapse
Affiliation(s)
- Mei Xin
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Zhiguang Qiao
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, China
| | - Jing Li
- Bone and Joint Research Center, The First Affiliated Hospital, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an 710061, China.,Department of Orthopaedics, Alpert Medical School/Rhode Island Hospital, Brown University, Providence, RI 02903, USA
| | - Jianjun Liu
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Shaoli Song
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Xiaoping Zhao
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Ping Miao
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Tingting Tang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, China
| | - Lei Wang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, China
| | - Weichun Liu
- Department of Gynecology and Obstetrics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Xiaodi Yang
- Department of Anesthesiology, Zhongshan Hospital, School of Medicine, Fudan University, Shanghai 200032, China
| | - Kerong Dai
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, China.,The Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine (SJTUSM) & Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai 200031, China
| | - Gang Huang
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.,Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine (SJTUSM) & Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai, 200031, China
| |
Collapse
|
30
|
Heske CM, Mendoza A, Edessa LD, Baumgart JT, Lee S, Trepel J, Proia DA, Neckers L, Helman LJ. STA-8666, a novel HSP90 inhibitor/SN-38 drug conjugate, causes complete tumor regression in preclinical mouse models of pediatric sarcoma. Oncotarget 2018; 7:65540-65552. [PMID: 27608846 PMCID: PMC5323173 DOI: 10.18632/oncotarget.11869] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 08/30/2016] [Indexed: 11/25/2022] Open
Abstract
Long-term survival in patients with metastatic, relapsed, or recurrent Ewing sarcoma and rhabdomyosarcoma is dismal. Irinotecan, a topoisomerase 1 inhibitor, has activity in these sarcomas, but due to poor bioavailability of its active metabolite (SN-38) has had limited clinical efficacy. In this study we have evaluated the efficacy and toxicity of STA-8666, a novel drug conjugate which uses an HSP90 inhibitor to facilitate intracellular, tumor-targeted delivery of the topoisomerase 1 inhibitor SN-38, thus preferentially delivering and concentrating SN-38 within tumor tissue. We present in vivo evidence from mouse xenograft models that STA-8666 results in more persistent inhibition of topoisomerase 1 and prolonged DNA damage compared to irinotecan. This translates into superior antitumor efficacy and survival in multiple aggressive models of both diseases in mouse xenografts, as well as in an irinotecan-resistant model of pediatric osteosarcoma, demonstrated by dramatic tumor shrinkage, durable remission and prolonged complete regressions following short-term treatment, compared to conventional irinotecan. Gene expression analysis performed on xenograft tumors treated with either irinotecan or STA-8666 showed that STA-8666 affected expression of DNA damage and repair genes more robustly than irinotecan. These results suggest that STA-8666 may be a promising new agent for patients with pediatric-type sarcoma.
Collapse
Affiliation(s)
- Christine M Heske
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Arnulfo Mendoza
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Leah D Edessa
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Joshua T Baumgart
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sunmin Lee
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jane Trepel
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | - Len Neckers
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Lee J Helman
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
31
|
Histone deacetylase inhibitor ITF2357 leads to apoptosis and enhances doxorubicin cytotoxicity in preclinical models of human sarcoma. Oncogenesis 2018; 7:20. [PMID: 29472530 PMCID: PMC5833676 DOI: 10.1038/s41389-018-0026-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 09/26/2017] [Accepted: 11/09/2017] [Indexed: 12/14/2022] Open
Abstract
Sarcomas are rare tumors with generally poor prognosis, for which current therapies have shown limited efficacy. Histone deacetylase inhibitors (HDACi) are emerging anti-tumor agents; however, little is known about their effect in sarcomas. By using established and patient-derived sarcoma cells with different subtypes, we showed that the pan-HDACi, ITF2357, potently inhibited in vitro survival in a p53-independent manner. ITF2357-mediated cell death implied the activation of mitochondrial apoptosis, as attested by induction of pro-apoptotic BH3-only proteins and a caspases-dependent mechanism. ITF2357 also induced autophagy, which protected sarcoma cells from apoptotic cell death. ITF2357 activated forkhead box (FOXO) 1 and 3a transcription factors and their downstream target genes, however, silencing of both FOXO1 and 3a did not protect sarcoma cells against ITF2357-induced apoptosis and upregulated FOXO4 and 6. Notably, ITF2357 synergized with Doxorubicin to induce cell death of established and patient-derived sarcoma cells. Furthermore, combination treatment strongly impaired xenograft tumor growth in vivo, when compared to single treatments, suggesting that combination of ITF2357 with Doxorubicin has the potential to enhance sensitization in different preclinical models of sarcoma. Overall, our study highlights the therapeutic potential of ITF2357, alone or in rational combination therapies, for bone and soft tissue sarcomas management.
Collapse
|
32
|
Bailly C, Leforestier R, Campion L, Thebaud E, Moreau A, Kraeber-Bodere F, Carlier T, Bodet-Milin C. Prognostic value of FDG-PET indices for the assessment of histological response to neoadjuvant chemotherapy and outcome in pediatric patients with Ewing sarcoma and osteosarcoma. PLoS One 2017; 12:e0183841. [PMID: 28841702 PMCID: PMC5571925 DOI: 10.1371/journal.pone.0183841] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 08/11/2017] [Indexed: 12/11/2022] Open
Abstract
Purpose The objective of this retrospective work was to evaluate the prognostic value on histological response and survival of quantitative indices derived from FDG-PET performed before and after chemotherapy (CHT), in a homogeneous pediatric Ewing sarcoma (EWS) and Osteosarcoma (OST) population. Methods Thirty-one patients with EWS and 31 with OST were included. All patients were treated with neoadjuvant CHT, and underwent surgery for local control. All patients had FDG-PET at diagnosis and after CHT, prior to surgery. Several parameters were evaluated: SUVmax, SUVpeak, SUVmean, metabolic tumor volume, total lesion glycolysis, 7 textural features and 3 shape features (SF). The segmentation was performed using an adaptive approach. Results were compared to histopathological regression of the resected tumor and to clinical follow-up for survival evaluation. Results For EWS, univariate analysis did not highlight any prognostic value on histological response, or survival regardless of all the considered metrics. For OST, only one of the SF, namely elongation, was significantly associated with PFS and OS on both univariate and multivariate analysis (PFS: p = 0.019, HR = 5.583; OS: p = 0.0062, HR = 7.113). Conclusion Only elongation determined on initial FDG-PET has a potential interest as a prognostic factor of PFS and OS in pediatric OST patients. Unlike recent studies of the literature realized in adult population, all the metrics reveal limited additional prognostic value in pediatric EWS patients. This seems to reinforce the question of whether children experience different subtypes of the same pathologies than older patients, with different outcomes.
Collapse
Affiliation(s)
- Clement Bailly
- Nuclear Medicine Department, University Hospital, Nantes, France
- Nantes-Angers Cancer Research Center CRCNA, University of Nantes, INSERM UMR892, CNRS-UMR6299, Nantes, France
| | | | - Loic Campion
- Biometrics Department, ICO René Gauducheau Cancer Center, Saint Herblain, France
| | - Estelle Thebaud
- Pediatric Oncology Department, University Hospital, Nantes, France
| | - Anne Moreau
- Pathology Department, University Hospital, Nantes, France
| | - Francoise Kraeber-Bodere
- Nuclear Medicine Department, University Hospital, Nantes, France
- Nantes-Angers Cancer Research Center CRCNA, University of Nantes, INSERM UMR892, CNRS-UMR6299, Nantes, France
| | - Thomas Carlier
- Nuclear Medicine Department, University Hospital, Nantes, France
- Nantes-Angers Cancer Research Center CRCNA, University of Nantes, INSERM UMR892, CNRS-UMR6299, Nantes, France
| | - Caroline Bodet-Milin
- Nuclear Medicine Department, University Hospital, Nantes, France
- Nantes-Angers Cancer Research Center CRCNA, University of Nantes, INSERM UMR892, CNRS-UMR6299, Nantes, France
- * E-mail:
| |
Collapse
|
33
|
Inuwa MM, Zakariyau LY, Ismail DI, Friday ES, Ibrahim AA, Mohammed AA. Overview of extremity musculoskeletal neoplasms at the Ahmadu Bello University Teaching Hospital Zaria, Nigeria. Ann Afr Med 2017; 16:141-144. [PMID: 28671156 PMCID: PMC5579899 DOI: 10.4103/aam.aam_5_17] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Background: The burden of managing patients with musculoskeletal neoplasms in the West African sub-region is quite significant. This is largely due to late presentation, delay in making diagnosis, and failure of obtaining consent to certain surgical procedures. Improvements in diagnosis and treatment over the years have however increased life and limb survival for many patients. This study was aimed at evaluating the clinicopathologic pattern of neoplasm as it affects the upper and lower limbs with a view to determining the most common types, the most involved sites of the body and the age at presentation of these tumors. Materials and Methods: A total of 65 patients with histologically confirmed neoplasm of the upper and lower limbs arising from bone, cartilage, skeletal muscle, synovium, and tendon sheath were retrospectively selected 7 years from January 2008 to 2015. Results: Forty-one (63.1%) patients were males, whereas 24 (36.9%) were females. Age range is between 5 and 75 years, average of 30.8 years. Lower limb involvement was recorded in 40 (61.5%) patients, with remaining 25 (38.5%) patients in upper limbs. 28 (43.1%) patients had benign lesion, whereas 37 (56.9%) were malignant. Giant cell tumor (GCT) was the most frequent benign tumor (50%) while osteogenic sarcomas top the list on the malignant variety (32.4%). The most commonly affected bones were tibia, carpal-metacarpal, and femur in that order. Conclusion: In our center, GCT and osteogenic sarcoma are the most frequently encountered benign and malignant musculoskeletal extremity neoplasms, respectively, with the tibial bone being most commonly affected.
Collapse
Affiliation(s)
| | - Lawal Yau Zakariyau
- Department of Traumatic and Orthopedic Surgery, Ahmadu Bello University, Zaria, Nigeria
| | - Dahiru I Ismail
- Department of Traumatic and Orthopedic Surgery, Ahmadu Bello University, Zaria, Nigeria
| | - Ejagwulu S Friday
- Department of Traumatic and Orthopedic Surgery, Ahmadu Bello University, Zaria, Nigeria
| | - Aniko A Ibrahim
- Department of Traumatic and Orthopedic Surgery, Ahmadu Bello University, Zaria, Nigeria
| | - Abdulmalik A Mohammed
- Department of Traumatic and Orthopedic Surgery, Ahmadu Bello University, Zaria, Nigeria
| |
Collapse
|
34
|
Abstract
Nuclear medicine has an important role in the management of many cancers in pediatric age group with multiple imaging modalities and radiopharmaceuticals targeting various biological uptake mechanisms. 18-Flourodeoxyglucose is the radiotracer of choice especially in patients with sarcoma and lymphoma. (18)FDG-PET, for sarcoma and lymphomas, is proved to be superior to conventional imaging in staging and therapy response. Although studies are limited in pediatric population, (18)FDG-PET/CT has found its way through international guidelines. Limitations and strengths of PET imaging must be noticed before adapting PET imaging in clinical protocols. Established new response criteria using multiple parameters derived from (18)FDG-PET would increase the accuracy and repeatability of response evaluation. Current data suggest that I-123 metaiodobenzylguanidine (MIBG) remains the tracer of choice in the evaluation of neuroblastoma (NB) because of its high sensitivity, specificity, diagnostic accuracy, and prognostic value. It is valuable in determining the response to therapy, surveillance for disease recurrence, and in selecting patients for I-131 therapy. SPECT/CT improves the diagnostic accuracy and the interpretation confidence of MIBG scans. (18)FDG-PET/CT is an important complementary to MIBG imaging despite its lack of specificity to NB. It is valuable in cases of negative or inconclusive MIBG scans and when MIBG findings underestimate the disease status as determined from clinical and radiological findings. F-18 DOPA is promising tracer that reflects catecholamine metabolism and is both sensitive and specific. F-18 DOPA scintigraphy provides the advantages of PET/CT imaging with early and short imaging times, high spatial resolution, inherent morphologic correlation with CT, and quantitation. Regulatory and production issues currently limit the tracer's availability. PET/CT with Ga-68 DOTA appears to be useful in NB imaging and may have a unique role in selecting patients for peptide receptor radionuclide therapy with somatostatin analogues. C-11 hydroxyephedrine PET/CT is a specific PET tracer for NB, but the C-11 label that requires an on-site cyclotron production and the high physiologic uptake in the liver and kidneys limit its use. I-124 MIBG is useful for I-131 MIBG pretherapeutic dosimetry planning. Its use for diagnostic imaging as well as the use of F-18 labeled MIBG analogues is currently experimental. PET/MR imaging is emerging and is likely to become an important tool in the evaluation. It provides metabolic and superior morphological data in one imaging session, expediting the diagnosis and lowering the radiation exposure. Radioactive iodines not only detect residual tissue and metastatic disease but also are used in the treatment of differentiated thyroid cancer. However, these are not well documented in pediatric age group like adult patients. Use of radioactivity in pediatric population is very important and strictly controlled because of the possibility of secondary malignities; therefore, management of oncological cases requires detailed literature knowledge. This article aims to review the literature on the use of radionuclide imaging and therapy in pediatric population with thyroid cancer, sarcomas, lymphoma, and NB.
Collapse
Affiliation(s)
- Pınar Özgen Kiratli
- Department of Nuclear Medicine, Hacettepe University Medical Center, Ankara, Turkey.
| | - Murat Tuncel
- Department of Nuclear Medicine, Hacettepe University Medical Center, Ankara, Turkey
| | - Zvi Bar-Sever
- Department of Nuclear Medicine, Schneider Children's Medical Center, Petah Tikva, Israel
| |
Collapse
|
35
|
Li F, Liao Z, Zhao J, Zhao G, Li X, Du X, Yang Y, Yang J. Efficacy and safety of Apatinib in stage IV sarcomas: experience of a major sarcoma center in China. Oncotarget 2017; 8:64471-64480. [PMID: 28969086 PMCID: PMC5610018 DOI: 10.18632/oncotarget.16293] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 03/01/2017] [Indexed: 12/31/2022] Open
Abstract
Purpose This study was conducted to review the efficacy and safety of Apatinib in stage IV sarcoma patients who failed previous chemotherapy. Materials and Methods The clinical information on 16 patients with stage IV sarcomas who failed in prior chemotherapy and subsequently received Apatinib treatment was collected. Apatinib was given 500mg/daily and 4 weeks as a cycle. All patients had at least one measurable extracranial tumor according to Response Evaluation Criteria In Solid Tumors 1.0 criteria. Progression free survival (PFS), overall survival (OS), objective response rate (ORR), disease control rate (DCR) and treatment-related adverse effects (AEs) were reviewed and evaluated. Results Patients was administered Apatinib for 0 to 9 cycles with the median of 3.2 cycles. Median follow-up time was 8.4 months (1 to 12 months). Ten of 16 patients received at least 1 complete cycle of Apatinib treatment were eligible for the efficacy analysis. The median PFS was 8.84 months. Two patients achieved partial response (PR) and 6 patients achieved stable disease (SD). Two patients were evaluated as progression disease (PD) and one patient died of disease progression. The ORR was 20.0% (2/10) and the DCR was 80.0% (8/10). The most common grade 3/4 treatment-related AEs were hypertension (18.7%), hand-foot syndrome (12.5%) and proteinuria (6.3%). No drug-related severe AEs occurred. Conclusion CApatinib treatment in this exploratory study exhibited objective efficacy and manageable toxicity in stage IV sarcoma patients who failed in chemotherapy. This result supports future random controlled trial to further define Apatinib activity in stage IV sarcomas.
Collapse
Affiliation(s)
- Feng Li
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute & Hospital, Tianjin, People's Republic of China.,National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute & Hospital, Tianjin, People's Republic of China
| | - Zhichao Liao
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute & Hospital, Tianjin, People's Republic of China.,National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute & Hospital, Tianjin, People's Republic of China
| | - Jun Zhao
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute & Hospital, Tianjin, People's Republic of China.,National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute & Hospital, Tianjin, People's Republic of China
| | - Gang Zhao
- National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute & Hospital, Tianjin, People's Republic of China.,Department of Pathology, Tianjin Medical University Cancer Institute & Hospital, Tianjin, People's Republic of China
| | - Xubin Li
- National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute & Hospital, Tianjin, People's Republic of China.,Department of Radiology, Tianjin Medical University Cancer Institute & Hospital, Tianjin, People's Republic of China
| | - Xiaoling Du
- Department of Diagnostics, Tianjin Medical University, Tianjin, People's Republic of China
| | - Yun Yang
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute & Hospital, Tianjin, People's Republic of China.,National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute & Hospital, Tianjin, People's Republic of China
| | - Jilong Yang
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute & Hospital, Tianjin, People's Republic of China.,National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute & Hospital, Tianjin, People's Republic of China
| |
Collapse
|
36
|
Cell-to-cell heterogeneity of EWSR1-FLI1 activity determines proliferation/migration choices in Ewing sarcoma cells. Oncogene 2017; 36:3505-3514. [PMID: 28135250 PMCID: PMC5541267 DOI: 10.1038/onc.2016.498] [Citation(s) in RCA: 146] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 11/30/2016] [Accepted: 12/01/2016] [Indexed: 12/22/2022]
Abstract
Ewing sarcoma is characterized by the expression of the chimeric EWSR1-FLI1 transcription factor. Proteomic analyses indicate that the decrease of EWSR1-FLI1 expression leads to major changes in effectors of the dynamics of the actin cytoskeleton and the adhesion processes with a shift from cell-to-cell to cell-matrix adhesion. These changes are associated with a dramatic increase of in vivo cell migration and invasion potential. Importantly, EWSR1-FLI1 expression, evaluated by single-cell RT-ddPCR/immunofluorescence analyses, and activity, assessed by expression of EWSR1-FLI1 downstream targets, are heterogeneous in cell lines and in tumours and can fluctuate along time in a fully reversible process between EWSR1-FLI1high states, characterized by highly active cell proliferation, and EWSR1-FLI1low states where cells have a strong propensity to migrate, invade and metastasize. This new model of phenotypic plasticity proposes that the dynamic fluctuation of the expression level of a dominant oncogene is an intrinsic characteristic of its oncogenic potential.
Collapse
|
37
|
Chakrabarty S, Foderingham N, O’Hara H. Selected Disorders of the Musculoskeletal System. Fam Med 2017. [DOI: 10.1007/978-3-319-04414-9_121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
38
|
Harris JC, Coburn JM, Kajdacsy-Balla A, Kaplan DL, Chiu B. Sustained delivery of vincristine inside an orthotopic mouse sarcoma model decreases tumor growth. J Pediatr Surg 2016; 51:2058-2062. [PMID: 27680598 PMCID: PMC5138133 DOI: 10.1016/j.jpedsurg.2016.09.040] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 09/12/2016] [Indexed: 12/26/2022]
Abstract
BACKGROUND Sarcoma accounts for 20% of solid tumors in children. Surgery has significant morbidity. We hypothesized that delivering chemotherapy directly into tumors through sustained release silk systems could slow tumor growth. METHODS Human Ewing sarcoma cells A673 were cultured with vincristine and doxorubicin to determine half maximal inhibitory concentration (IC50). Cells were injected into mouse hind leg to create orthotopic tumors. Tumor volumes were measured using ultrasound. When volume reached >250mm3, interventions included: implantation of drug-free silk foam (Control-F), doxorubicin 400μg foam (Dox400-F), vincristine 50μg foam (Vin50-F), drug-free silk gel (Control-G), vincristine 50μg gel (Vin50-G), or single dose intravenous vincristine 50μg (Vin50-IV). End-point was volume>1000mm3. Kaplan Meier and ANOVA were used. RESULTS IC50 for vincristine and doxorubicin was 0.5ng/mL and 200ng/mL, respectively. There was no difference between Dox400-F [6±1days to end point (DTEP)] and Control-F (5±1.3 DTEP). Vin50-F (12.4±3.5 DTEP) had slower growth compared to Control-F (p<0.001), and there was no difference between Vin50-F and Vin50-IV (14±0 DTEP). Growth was slowest with Vin50-G, 28±10.3 DTEP compared to all other treatment groups (p<0.05). CONCLUSION Sustained delivery of vincristine inside the sarcoma tumor with silk gel decreased tumor growth. Applying this intratumoral treatment strategy may potentially decrease the extent of surgical excision.
Collapse
Affiliation(s)
- Jamie C Harris
- Department of Surgery, Rush University Medical Center, 1750 W. Harrison St, Suite 785, Chicago, IL, 60612, USA
| | - Jeannine M Coburn
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA, 02155, USA
| | - Andre Kajdacsy-Balla
- Division of Pathology, University of Illinois Chicago, 840 S. Wood St. Suite 130 CSN, Chicago, IL, 60612
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA, 02155, USA
| | - Bill Chiu
- Department of Surgery, University of Illinois at Chicago, 840 S. Wood Street, Suite 416, Chicago, IL, 60612, USA.
| |
Collapse
|
39
|
Shim J, Staruch R, Koral K, Xie XJ, Chopra R, Laetsch TW. Pediatric Sarcomas Are Targetable by MR-Guided High Intensity Focused Ultrasound (MR-HIFU): Anatomical Distribution and Radiological Characteristics. Pediatr Blood Cancer 2016; 63:1753-60. [PMID: 27199087 PMCID: PMC6016837 DOI: 10.1002/pbc.26079] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Revised: 05/01/2016] [Accepted: 05/03/2016] [Indexed: 12/28/2022]
Abstract
BACKGROUND Despite intensive therapy, children with metastatic and recurrent sarcoma or neuroblastoma have a poor prognosis. Magnetic resonance guided high intensity focused ultrasound (MR-HIFU) is a noninvasive technique allowing the delivery of targeted ultrasound energy under MR imaging guidance. MR-HIFU may be used to ablate tumors without ionizing radiation or target chemotherapy using hyperthermia. Here, we evaluated the anatomic locations of tumors to assess the technical feasibility of MR-HIFU therapy for children with solid tumors. PROCEDURE Patients with sarcoma or neuroblastoma with available cross-sectional imaging were studied. Tumors were classified based on the location and surrounding structures within the ultrasound beam path as (i) not targetable, (ii) completely or partially targetable with the currently available MR-HIFU system, and (iii) potentially targetable if a respiratory motion compensation technique was used. RESULTS Of the 121 patients with sarcoma and 61 patients with neuroblastoma, 64% and 25% of primary tumors were targetable at diagnosis, respectively. Less than 20% of metastases at diagnosis or relapse were targetable for both sarcoma and neuroblastoma. Most targetable lesions were located in extremities or in the pelvis. Respiratory motion compensation may increase the percentage of targetable tumors by 4% for sarcomas and 10% for neuroblastoma. CONCLUSIONS Many pediatric sarcomas are localized at diagnosis and are targetable by current MR-HIFU technology. Some children with neuroblastoma have bony tumors targetable by MR-HIFU at relapse, but few newly diagnosed children with neuroblastoma have tumors amenable to MR-HIFU therapy. Clinical trials of MR-HIFU should focus on patients with anatomically targetable tumors.
Collapse
Affiliation(s)
- Jenny Shim
- Department of Pediatrics, Children’s Health, Dallas, Texas, USA
| | - Robert Staruch
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA,Philips Research North America, Cambridge, Massachusetts, USA
| | - Korgun Koral
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Xian-Jin Xie
- Department of Clinical Sciences, Simmons Comprehensive Cancer Center, Dallas, Texas, USA
| | - Rajiv Chopra
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA,Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Texas, USA
| | - Theodore W. Laetsch
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA,Pauline Allen Gill Center for Cancer and Blood Disorders, Children’s Health, Dallas, Texas, USA
| |
Collapse
|
40
|
Waters AM, Stafman LL, Garner EF, Mruthyunjayappa S, Stewart JE, Friedman GK, Coleman JM, Markert JM, Gillespie GY, Beierle EA. Effect of Repeat Dosing of Engineered Oncolytic Herpes Simplex Virus on Preclinical Models of Rhabdomyosarcoma. Transl Oncol 2016; 9:419-430. [PMID: 27751346 PMCID: PMC5067929 DOI: 10.1016/j.tranon.2016.07.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 07/14/2016] [Accepted: 07/18/2016] [Indexed: 12/16/2022] Open
Abstract
Rhabdomyosarcoma (RMS), a tumor of skeletal muscle origin, is the most common sarcoma of childhood. Despite multidrug chemotherapy regimens, surgical intervention, and radiation treatment, outcomes remain poor, especially in advanced disease, and novel therapies are needed for the treatment of these aggressive malignancies. Genetically engineered oncolytic viruses, such as herpes simplex virus-1 (HSV), are currently being explored as treatments for pediatric tumors. M002, an oncolytic HSV, has both copies of the γ134.5 gene deleted, enabling replication in tumor cells but thwarting infection of normal, postmitotic cells. We hypothesized that M002 would infect human RMS tumor cells and lead to decreased tumor cell survival in vitro and impede tumor growth in vivo. In the current study, we demonstrated that M002 could infect, replicate in, and decrease cell survival in both embryonal (ERMS) and alveolar rhabdomyosarcoma (ARMS) cells. Additionally, M002 reduced xenograft tumor growth and increased animal survival in both ARMS and ERMS. Most importantly, we showed for the first time that repeated dosing of oncolytic virus coupled with low-dose radiation provided improved tumor response in RMS. These findings provide support for the clinical investigation of oncolytic HSV in pediatric RMS.
Collapse
Affiliation(s)
- Alicia M Waters
- Department of Surgery, Division of Pediatric Surgery, University of Alabama, Birmingham, Birmingham, AL, USA 35233
| | - Laura L Stafman
- Department of Surgery, Division of Pediatric Surgery, University of Alabama, Birmingham, Birmingham, AL, USA 35233
| | - Evan F Garner
- Department of Surgery, Division of Pediatric Surgery, University of Alabama, Birmingham, Birmingham, AL, USA 35233
| | - Smitha Mruthyunjayappa
- Department of Surgery, Division of Pediatric Surgery, University of Alabama, Birmingham, Birmingham, AL, USA 35233
| | - Jerry E Stewart
- Department of Surgery, Division of Pediatric Surgery, University of Alabama, Birmingham, Birmingham, AL, USA 35233
| | - Gregory K Friedman
- Department of Pediatrics, Division of Hematology/Oncology, University of Alabama, Birmingham, Birmingham, AL, USA 35233
| | - Jennifer M Coleman
- Department of Surgery, Division of Neurosurgery, University of Alabama, Birmingham, Birmingham, AL, USA 35233
| | - James M Markert
- Department of Surgery, Division of Neurosurgery, University of Alabama, Birmingham, Birmingham, AL, USA 35233
| | - G Yancey Gillespie
- Department of Surgery, Division of Neurosurgery, University of Alabama, Birmingham, Birmingham, AL, USA 35233
| | - Elizabeth A Beierle
- Department of Surgery, Division of Pediatric Surgery, University of Alabama, Birmingham, Birmingham, AL, USA 35233.
| |
Collapse
|
41
|
The heparanase/heparan sulfate proteoglycan axis: A potential new therapeutic target in sarcomas. Cancer Lett 2016; 382:245-254. [PMID: 27666777 DOI: 10.1016/j.canlet.2016.09.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 09/08/2016] [Accepted: 09/08/2016] [Indexed: 12/29/2022]
Abstract
Heparanase, the only known mammalian endoglycosidase degrading heparan sulfate (HS) chains of HS proteoglycans (HSPG), is a highly versatile protein affecting multiple events in tumor cells and their microenvironment. In several malignancies, deregulation of the heparanase/HSPG system has been implicated in tumor progression, hence representing a valuable therapeutic target. Currently, multiple agents interfering with the heparanase/HSPG axis are under clinical investigation. Sarcomas are characterized by a high biomolecular complexity and multiple levels of interconnection with microenvironment sustaining their growth and progression. The clinical management of advanced diseases remains a challenge. In several sarcoma subtypes, high levels of heparanase expression have been correlated with poor prognosis associated factors. On the other hand, expression of cell surface-associated HSPGs (i.e. glypicans and syndecans) has been found altered in specific sarcoma subtypes. Recent studies provided the preclinical proof-of-principle of the role of the heparanase/HSPG axis as therapeutic target in various sarcoma subtypes. Although currently there are no clinical trials evaluating agents targeting heparanase and/or HSPGs in sarcomas, we here provide arguments for this strategy as potentially able to implement the therapeutic options for sarcoma patients.
Collapse
|
42
|
Choi YB, Yi ES, Lee JW, Yoo KH, Sung KW, Koo HH. High-Dose Chemotherapy and Autologous Stem Cell Transplantation in Children with High-Risk or Recurrent Bone and Soft Tissue Sarcomas. J Korean Med Sci 2016; 31:1055-62. [PMID: 27366002 PMCID: PMC4900996 DOI: 10.3346/jkms.2016.31.7.1055] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Accepted: 04/09/2016] [Indexed: 01/09/2023] Open
Abstract
Despite increasing evidence that high-dose chemotherapy and autologous stem cell transplantation (HDCT/auto-SCT) might improve the survival of patients with high-risk or recurrent solid tumors, therapy effectiveness for bone and soft tissue sarcoma treatment remains unclear. This study retrospectively investigated the feasibility and effectiveness of HDCT/auto-SCT for high-risk or recurrent bone and soft tissue sarcoma. A total of 28 patients (18 high-risk and 10 recurrent) underwent single or tandem HDCT/auto-SCT between October 2004 and September 2014. During follow-up of a median 15.3 months, 18 patients exhibited disease progression and 2 died of treatment-related toxicities (1 veno-occlusive disease and 1 sepsis). Overall, 8 patients remained alive and progression-free. The 3-year overall survival (OS) and event-free survival (EFS) rates for all 28 patients were 28.7% and 26.3%, respectively. In the subgroup analysis, OS and EFS rates were higher in patients with complete or partial remission prior to HDCT/auto-SCT than in those with worse responses (OS, 39.1% vs. 0.0%, P = 0.002; EFS, 36.8% vs. 0.0%, P < 0.001). Therefore, careful selection of patients who can benefit from HDCT/auto-SCT and maximal effort to reduce tumor burden prior to treatment will be important to achieve favorable outcomes in patients with high-risk or recurrent bone and soft tissue sarcomas.
Collapse
Affiliation(s)
- Young Bae Choi
- Department of Pediatrics, Chung-Ang University Hospital, Seoul, Korea
| | - Eun Sang Yi
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ji Won Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Keon Hee Yoo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ki Woong Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hong Hoe Koo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
43
|
Edelmann MN, Daryani VM, Bishop MW, Liu W, Brinkman TM, Stewart CF, Mulrooney DA, Kimberg C, Ness KK, Cheung YT, Srivastava DK, Robison LL, Hudson MM, Krull KR. Neurocognitive and Patient-Reported Outcomes in Adult Survivors of Childhood Osteosarcoma. JAMA Oncol 2016; 2:201-8. [PMID: 26583357 DOI: 10.1001/jamaoncol.2015.4398] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
IMPORTANCE This study provides the first objective data documenting neurocognitive impairment in long-term survivors of childhood osteosarcoma. OBJECTIVE To examine neurocognitive, neurobehavioral, emotional, and quality-of-life outcomes in long-term survivors of childhood osteosarcoma. DESIGN, SETTING, AND PARTICIPANTS Cross-sectional cohort study at an academic research hospital, with prospective treatment and chronic health predictors. Outcome data were collected from June 2008 to August 2014. Data analysis was completed in April 2015. Survivors of osteosarcoma recruited from the St Jude Lifetime Cohort Study were compared with community controls. MAIN OUTCOMES AND MEASURES Neurocognitive function, neurobehavioral symptoms, emotional distress, and quality of life. Outcomes were examined in relation to pharmacokinetic indices of methotrexate exposure and current chronic health conditions, which were assessed through medical examination and coded according to Common Terminology Criteria for Adverse Events, Version 4.03. RESULTS Eighty survivors of osteosarcoma (mean [SD] age, 38.9 [7.6] years; time since diagnosis, 24.7 [6.6] years; 42% female) were compared with 39 community controls (age, 39.0 [11.7] years; 56% female). Survivors demonstrated lower mean scores in reading skills (-0.21 [95% CI, -0.32 to -0.10] vs 0.05 [95% CI, -0.13 to 0.23]; P = .01), attention (-0.78 [95% CI, -1.32 to -0.24] vs 0.24 [95% CI, -0.07 to 0.55]; P = .002), memory (-0.24 [95% CI, -0.48 to 0] vs 0.27 [95% CI, -0.08 to 0.62]; P = .01), and processing speed (-0.15 [95% CI, -0.35 to 0.05] vs 0.74 [95% CI, 0.44 to 1.03]; P < .001). Results of pharmacokinetic analysis showed that high-dose methotrexate maximum plasma concentration (estimate = 0; P = .48), median clearance (estimate = -0.11; P = .76), and median/cumulative exposure (estimate = 0; P = .45) were not associated with neurocognitive outcomes. Any grade 3 or 4 Common Terminology Criteria for Adverse Events cardiac, pulmonary, or endocrine condition was associated with poorer memory (t = 2.93; P = .006) and slower processing speed (t = 3.03; P = .002). Survivor-reported poor general health was associated with decreased sustained attention (estimate = 0.24; P = .05) and processing speed (estimate = 0.34; P = .005). CONCLUSIONS AND RELEVANCE Long-term survivors of osteosarcoma are at risk for neurocognitive impairment, which is related to current chronic health conditions and not to original treatment with high-dose methotrexate. Prospective longitudinal studies are needed to identify onset and progression of impairment to inform optimal interventions.
Collapse
Affiliation(s)
- Michelle N Edelmann
- Department of Epidemiology and Cancer Control, St Jude Children's Research Hospital, Memphis, Tennessee
| | - Vinay M Daryani
- Department of Pharmaceutical Sciences, St Jude Children's Research Hospital, Memphis, Tennessee
| | - Michael W Bishop
- Department of Oncology, St Jude Children's Research Hospital, Memphis, Tennessee
| | - Wei Liu
- Department of Biostatistics, St Jude Children's Research Hospital, Memphis, Tennessee
| | - Tara M Brinkman
- Department of Epidemiology and Cancer Control, St Jude Children's Research Hospital, Memphis, Tennessee5Department of Psychology, St Jude Children's Research Hospital, Memphis, Tennessee
| | - Clinton F Stewart
- Department of Pharmaceutical Sciences, St Jude Children's Research Hospital, Memphis, Tennessee
| | - Daniel A Mulrooney
- Department of Epidemiology and Cancer Control, St Jude Children's Research Hospital, Memphis, Tennessee3Department of Oncology, St Jude Children's Research Hospital, Memphis, Tennessee
| | - Cara Kimberg
- Department of Epidemiology and Cancer Control, St Jude Children's Research Hospital, Memphis, Tennessee
| | - Kirsten K Ness
- Department of Epidemiology and Cancer Control, St Jude Children's Research Hospital, Memphis, Tennessee
| | - Yin Ting Cheung
- Department of Epidemiology and Cancer Control, St Jude Children's Research Hospital, Memphis, Tennessee
| | - Deo Kumar Srivastava
- Department of Biostatistics, St Jude Children's Research Hospital, Memphis, Tennessee
| | - Leslie L Robison
- Department of Epidemiology and Cancer Control, St Jude Children's Research Hospital, Memphis, Tennessee
| | - Melissa M Hudson
- Department of Epidemiology and Cancer Control, St Jude Children's Research Hospital, Memphis, Tennessee3Department of Oncology, St Jude Children's Research Hospital, Memphis, Tennessee
| | - Kevin R Krull
- Department of Epidemiology and Cancer Control, St Jude Children's Research Hospital, Memphis, Tennessee5Department of Psychology, St Jude Children's Research Hospital, Memphis, Tennessee
| |
Collapse
|
44
|
Schiffman JD, Breen M. Comparative oncology: what dogs and other species can teach us about humans with cancer. Philos Trans R Soc Lond B Biol Sci 2016; 370:rstb.2014.0231. [PMID: 26056372 DOI: 10.1098/rstb.2014.0231] [Citation(s) in RCA: 244] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Over 1.66 million humans (approx. 500/100,000 population rate) and over 4.2 million dogs (approx. 5300/100,000 population rate) are diagnosed with cancer annually in the USA. The interdisciplinary field of comparative oncology offers a unique and strong opportunity to learn more about universal cancer risk and development through epidemiology, genetic and genomic investigations. Working across species, researchers from human and veterinary medicine can combine scientific findings to understand more quickly the origins of cancer and translate these findings to novel therapies to benefit both human and animals. This review begins with the genetic origins of canines and their advantage in cancer research. We next focus on recent findings in comparative oncology related to inherited, or genetic, risk for tumour development. We then detail the somatic, or genomic, changes within tumours and the similarities between species. The shared cancers between humans and dogs that we discuss include sarcoma (osteosarcoma, soft tissue sarcoma, histiocytic sarcoma, hemangiosarcoma), haematological malignancies (lymphoma, leukaemia), bladder cancer, intracranial neoplasms (meningioma, glioma) and melanoma. Tumour risk in other animal species is also briefly discussed. As the field of genomics advances, we predict that comparative oncology will continue to benefit both humans and the animals that live among us.
Collapse
Affiliation(s)
- Joshua D Schiffman
- Department of Pediatrics and Oncological Sciences, Primary Children's Hospital, Intermountain Healthcare, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Matthew Breen
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, Center for Comparative Medicine and Translational Research, Center for Human Health and the Environment, Cancer Genetics, UNC Lineberger Comprehensive Cancer Center, North Carolina State University, Raleigh, NC, USA
| |
Collapse
|
45
|
Irreversible Electroporation as an Effective Technique for Ablating Human Metastatic Osteosarcoma. J Pediatr Hematol Oncol 2016; 38:182-6. [PMID: 26950088 DOI: 10.1097/mph.0000000000000516] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Irreversible electroporation (IRE) induces apoptosis in tumor cells with electric energy, allowing treatment of unresectable tumors. One potential application is metastatic osteosarcoma (OS) in the pediatric population. A 12-year-old underwent thoracotomy with resection of metastatic OS. IRE was applied to 1 resected tumor section. Using 2 probes, 100 pulses with width of 90 ms were delivered. Efficacy was measured by increase in current draw during treatment. The treated sample was analyzed with hematoxylin and eosin and transmission electron microscopy. Default voltage of 1800 kV was ineffective. Voltage of 2700 kV caused excessive current draw and was aborted to prevent thermal injury. At 2200 kV, current draw rise was 9 amps, signifying successful treatment. Untreated specimen showed viable OS, normal surrounding lung tissue. Treated tumor had edema within the tumor and in surrounding lung tissue, with intra-alveolar hemorrhage and cellular architecture destruction. There was also evidence for cellular destruction such as disruption of lipid bilayer and release of intracellular fluid. Optimal voltage for treatment was 2200 kV, likely higher due to electrical conduction variation in the aerated lung. IRE may be an option for pediatric patients with unresectable metastatic OS.
Collapse
|
46
|
Natural compounds for pediatric cancer treatment. Naunyn Schmiedebergs Arch Pharmacol 2015; 389:131-49. [DOI: 10.1007/s00210-015-1191-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 11/08/2015] [Indexed: 12/13/2022]
|
47
|
Deel MD, Li JJ, Crose LES, Linardic CM. A Review: Molecular Aberrations within Hippo Signaling in Bone and Soft-Tissue Sarcomas. Front Oncol 2015; 5:190. [PMID: 26389076 PMCID: PMC4557106 DOI: 10.3389/fonc.2015.00190] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 08/10/2015] [Indexed: 12/14/2022] Open
Abstract
The Hippo signaling pathway is an evolutionarily conserved developmental network vital for the regulation of organ size, tissue homeostasis, repair and regeneration, and cell fate. The Hippo pathway has also been shown to have tumor suppressor properties. Hippo transduction involves a series of kinases and scaffolding proteins that are intricately connected to proteins in developmental cascades and in the tissue microenvironment. This network governs the downstream Hippo transcriptional co-activators, YAP and TAZ, which bind to and activate the output of TEADs, as well as other transcription factors responsible for cellular proliferation, self-renewal, differentiation, and survival. Surprisingly, there are few oncogenic mutations within the core components of the Hippo pathway. Instead, dysregulated Hippo signaling is a versatile accomplice to commonly mutated cancer pathways. For example, YAP and TAZ can be activated by oncogenic signaling from other pathways, or serve as co-activators for classical oncogenes. Emerging evidence suggests that Hippo signaling couples cell density and cytoskeletal structural changes to morphogenic signals and conveys a mesenchymal phenotype. While much of Hippo biology has been described in epithelial cell systems, it is clear that dysregulated Hippo signaling also contributes to malignancies of mesenchymal origin. This review will summarize the known molecular alterations within the Hippo pathway in sarcomas and highlight how several pharmacologic compounds have shown activity in modulating Hippo components, providing proof-of-principle that Hippo signaling may be harnessed for therapeutic application in sarcomas.
Collapse
Affiliation(s)
- Michael D Deel
- Division of Hematology-Oncology, Department of Pediatrics, Duke University School of Medicine , Durham, NC , USA
| | - Jenny J Li
- Duke University School of Medicine , Durham, NC , USA
| | - Lisa E S Crose
- Division of Hematology-Oncology, Department of Pediatrics, Duke University School of Medicine , Durham, NC , USA
| | - Corinne M Linardic
- Division of Hematology-Oncology, Department of Pediatrics, Duke University School of Medicine , Durham, NC , USA ; Department of Pharmacology and Cancer Biology, Duke University School of Medicine , Durham, NC , USA
| |
Collapse
|
48
|
Abstract
Abnormal vaginal bleeding in a postmenarchal adolescent patient is most often related to dysfunctional uterine bleeding. However, there are other potential etiologies, including hematologic disorders, infections, and oncologic problems. We present a 12-year-old girl who presented with prolonged vaginal bleeding and was ultimately diagnosed with rhabdomyosarcoma. In this article, we discuss the approach to a patient with vaginal bleeding along with a more in-depth review of risk stratification in rhabdomyosarcoma, including treatment options such as chemotherapy, surgery, and radiation therapy.
Collapse
|
49
|
El-Naggar AM, Veinotte CJ, Cheng H, Grunewald TGP, Negri GL, Somasekharan SP, Corkery DP, Tirode F, Mathers J, Khan D, Kyle AH, Baker JH, LePard NE, McKinney S, Hajee S, Bosiljcic M, Leprivier G, Tognon CE, Minchinton AI, Bennewith KL, Delattre O, Wang Y, Dellaire G, Berman JN, Sorensen PH. Translational Activation of HIF1α by YB-1 Promotes Sarcoma Metastasis. Cancer Cell 2015; 27:682-97. [PMID: 25965573 DOI: 10.1016/j.ccell.2015.04.003] [Citation(s) in RCA: 207] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 02/28/2015] [Accepted: 04/10/2015] [Indexed: 12/27/2022]
Abstract
Metastatic dissemination is the leading cause of death in cancer patients, which is particularly evident for high-risk sarcomas such as Ewing sarcoma, osteosarcoma, and rhabdomyosarcoma. Previous research identified a crucial role for YB-1 in the epithelial-to-mesenchymal transition (EMT) and metastasis of epithelial malignancies. Based on clinical data and two distinct animal models, we now report that YB-1 is also a major metastatic driver in high-risk sarcomas. Our data establish YB-1 as a critical regulator of hypoxia-inducible factor 1α (HIF1α) expression in sarcoma cells. YB-1 enhances HIF1α protein expression by directly binding to and activating translation of HIF1A messages. This leads to HIF1α-mediated sarcoma cell invasion and enhanced metastatic capacity in vivo, highlighting a translationally regulated YB-1-HIF1α axis in sarcoma metastasis.
Collapse
Affiliation(s)
- Amal M El-Naggar
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 2B5, Canada; Department of Molecular Oncology, British Columbia Cancer Research Center, Vancouver, BC V5Z 1L3, Canada
| | - Chansey J Veinotte
- Department of Pediatrics, IWK Health Centre, Halifax, NS B3K 6R8, Canada
| | - Hongwei Cheng
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Thomas G P Grunewald
- Laboratory for Pediatric Sarcoma Biology, Institute of Pathology, LMU Munich, Thalkirchner Strasse 36, 80337 Munich, Germany
| | - Gian Luca Negri
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 2B5, Canada; Department of Molecular Oncology, British Columbia Cancer Research Center, Vancouver, BC V5Z 1L3, Canada
| | - Syam Prakash Somasekharan
- Department of Molecular Oncology, British Columbia Cancer Research Center, Vancouver, BC V5Z 1L3, Canada
| | - Dale P Corkery
- Department of Pediatrics, IWK Health Centre, Halifax, NS B3K 6R8, Canada
| | - Franck Tirode
- INSERM U830, Laboratoire de génétique et biologie des cancers, Institut Curie, 75248 Paris, France
| | - Joan Mathers
- Department of Molecular Oncology, British Columbia Cancer Research Center, Vancouver, BC V5Z 1L3, Canada
| | - Debjit Khan
- Department of Molecular Oncology, British Columbia Cancer Research Center, Vancouver, BC V5Z 1L3, Canada
| | - Alastair H Kyle
- Department of Integrative Oncology, Radiation Biology Unit, British Columbia Cancer Research Center, Vancouver, BC V5Z 1L3, Canada
| | - Jennifer H Baker
- Department of Integrative Oncology, Radiation Biology Unit, British Columbia Cancer Research Center, Vancouver, BC V5Z 1L3, Canada
| | - Nancy E LePard
- Department of Integrative Oncology, Radiation Biology Unit, British Columbia Cancer Research Center, Vancouver, BC V5Z 1L3, Canada
| | - Steven McKinney
- Department of Molecular Oncology, British Columbia Cancer Research Center, Vancouver, BC V5Z 1L3, Canada
| | - Shamil Hajee
- Department of Molecular Oncology, British Columbia Cancer Research Center, Vancouver, BC V5Z 1L3, Canada
| | - Momir Bosiljcic
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 2B5, Canada; Department of Integrative Oncology, Radiation Biology Unit, British Columbia Cancer Research Center, Vancouver, BC V5Z 1L3, Canada
| | - Gabriel Leprivier
- Department of Molecular Oncology, British Columbia Cancer Research Center, Vancouver, BC V5Z 1L3, Canada
| | - Cristina E Tognon
- Department of Molecular Oncology, British Columbia Cancer Research Center, Vancouver, BC V5Z 1L3, Canada
| | - Andrew I Minchinton
- Department of Integrative Oncology, Radiation Biology Unit, British Columbia Cancer Research Center, Vancouver, BC V5Z 1L3, Canada
| | - Kevin L Bennewith
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 2B5, Canada; Department of Integrative Oncology, Radiation Biology Unit, British Columbia Cancer Research Center, Vancouver, BC V5Z 1L3, Canada
| | - Olivier Delattre
- INSERM U830, Laboratoire de génétique et biologie des cancers, Institut Curie, 75248 Paris, France
| | - Yuzhuo Wang
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Graham Dellaire
- Department of Pathology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Jason N Berman
- Department of Pediatrics, IWK Health Centre, Halifax, NS B3K 6R8, Canada; Department of Pathology, Dalhousie University, Halifax, NS B3H 4R2, Canada; Department of Pediatrics, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Poul H Sorensen
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 2B5, Canada; Department of Molecular Oncology, British Columbia Cancer Research Center, Vancouver, BC V5Z 1L3, Canada.
| |
Collapse
|
50
|
Abstract
Cancer is a disease of the genome with diverse aetiologies including the accumulation of acquired mutations throughout the genome. There has been a flood of knowledge improving our understanding of the biology and molecular genetics of melanoma, lung and colorectal cancer since the genomics era started. Translation of this knowledge into a better understanding of cell proliferation, survival and apoptosis has produced a paradigm shift in medical oncology enabling gene-based cancer treatment (called personalised or precision medicine). Somatic mutation analysis is crucial for a genomics approach since it can identify driver mutations-the "Achilles' heel" of cancer, and support clinical decision-making through targeted therapy. Nevertheless, the applications of somatic DNA testing in cancer face many challenges such as obtaining comprehensive coverage of the cancer genome with limited DNA being available, and delivering an accurate report in a timely fashion without false-negative and false-positive results. Further advances in DNA technologies and bioinformatics will overcome these issues and maximise opportunities for targeted therapy. Somatic mutation analysis will then become an integral part of cancer management for all malignancies.
Collapse
Affiliation(s)
- Bing Yu
- 1 Department of Medical Genomics, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia ; 2 Sydney Medical School (Central), the University of Sydney, NSW 2006, Australia ; 3 Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia
| | - Sandra A O'Toole
- 1 Department of Medical Genomics, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia ; 2 Sydney Medical School (Central), the University of Sydney, NSW 2006, Australia ; 3 Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia
| | - Ronald J Trent
- 1 Department of Medical Genomics, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia ; 2 Sydney Medical School (Central), the University of Sydney, NSW 2006, Australia ; 3 Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia
| |
Collapse
|