1
|
Hayes-Jordan AA, Ma X, Menegaz BA, Lamhamedi-Cherradi SE, Kingsley CV, Benson JA, Camacho PE, Ludwig JA, Lockworth CR, Garcia GE, Craig SL. Efficacy of ONC201 in Desmoplastic Small Round Cell Tumor. Neoplasia 2018; 20:524-532. [PMID: 29626752 PMCID: PMC5915995 DOI: 10.1016/j.neo.2018.02.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 02/13/2018] [Accepted: 02/19/2018] [Indexed: 12/30/2022] Open
Abstract
Desmoplastic Small Round Cell Tumor (DSRCT) is a rare sarcoma tumor of adolescence and young adulthood, which harbors a recurrent chromosomal translocation between the Ewing’s sarcoma gene (EWSR1) and the Wilms’ tumor suppressor gene (WT1). Patients usually develop multiple abdominal tumors with liver and lymph node metastasis developing later. Survival is poor using a multimodal therapy that includes chemotherapy, radiation and surgical resection, new therapies are needed for better management of DSRCT. Triggering cell apoptosis is the scientific rationale of many cancer therapies. Here, we characterized for the first time the expression of pro-apoptotic receptors, tumor necrosis-related apoptosis-inducing ligand receptors (TRAILR1-4) within an established human DSRCT cell line and clinical samples. The molecular induction of TRAIL-mediated apoptosis using agonistic small molecule, ONC201 in vitro cell-based proliferation assay and in vivo novel orthotopic xenograft animal models of DSRCT, was able to inhibit cell proliferation that was associated with caspase activation, and tumor growth, indicating that a cell-based delivery of an apoptosis-inducing factor could be relevant therapeutic agent to control DSRCT.
Collapse
Affiliation(s)
- Andrea A Hayes-Jordan
- Division of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit, 1484, Houston, TX, USA.
| | - Xiao Ma
- Division of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit, 1484, Houston, TX, USA
| | - Brian A Menegaz
- Division of Sarcoma Medical Oncology-Research, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit, 1952, Houston, TX, USA
| | - Salah-Eddine Lamhamedi-Cherradi
- Division of Sarcoma Medical Oncology-Research, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit, 1952, Houston, TX, USA
| | - Charles V Kingsley
- Department of Imaging Physics-Research, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit, 1902, Houston, TX, USA
| | - Jalen A Benson
- Division of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit, 1484, Houston, TX, USA
| | - Pamela E Camacho
- Department of Pediatrics-Patient Care, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit, 0087, Houston, TX, USA
| | - Joseph A Ludwig
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit, 0450, Houston, TX, USA
| | - Cynthia R Lockworth
- Department of Veterinary Medicine & Surgery, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit, 0063, Houston, TX, USA
| | - Gloria E Garcia
- Department of Veterinary Medicine & Surgery, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit, 0063, Houston, TX, USA
| | - Suzanne L Craig
- Department of Veterinary Medicine & Surgery, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit, 0063, Houston, TX, USA
| |
Collapse
|
2
|
Anderson PM, Bielack SS, Gorlick RG, Skubitz K, Daw NC, Herzog CE, Monge OR, Lassaletta A, Boldrini E, Pápai Z, Rubino J, Pathiraja K, Hille DA, Ayers M, Yao S, Nebozhyn M, Lu B, Mauro D. A phase II study of clinical activity of SCH 717454 (robatumumab) in patients with relapsed osteosarcoma and Ewing sarcoma. Pediatr Blood Cancer 2016; 63:1761-70. [PMID: 27362300 PMCID: PMC5129487 DOI: 10.1002/pbc.26087] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 04/19/2016] [Accepted: 04/28/2016] [Indexed: 01/18/2023]
Abstract
BACKGROUND Robatumumab (19D12; MK-7454 otherwise known as SCH717454) is a fully human antibody that binds to and inhibits insulin-like growth factor receptor-1 (IGF-1R). This multiinstitutional study (P04720) determined the safety and clinical efficacy of robatumumab in three separate patient groups with resectable osteosarcoma metastases (Group 1), unresectable osteosarcoma metastases (Group 2), and Ewing sarcoma metastases (Group 3). PROCEDURE Robatumumab infusions were administered every 2 weeks and were well tolerated with minimal toxicity. Centrally reviewed response data were available for 144 patients. RESULTS Low disease burden was important for osteosarcoma response: three of 31 patients had complete response or partial response (PR) by Response Evaluation Criteria in Solid Tumors (RECIST) in resectable patients (Group 1) versus zero of 29 in unresectable patients (Group 2); median overall survival was 20 months in Group 1 versus 8.2 months in Group 2. In centrally reviewed patients with Ewing sarcoma with PET-CT data (N = 84/115), there were six PR, 23 stable disease, and 55 progression of disease by RECIST at 2 months. Patients with Ewing sarcoma had a median overall survival of 6.9 months. However, responding patients with Ewing sarcoma were allowed to continue on treatment after study closure. A minority of patients with metastatic Ewing sarcoma showed clinical responses and have remained healthy after receiving 25-115 doses of robatumumab with remissions of >4 years duration (N = 6). CONCLUSIONS These findings show that although the IGF-1R remains an attractive treatment target, additional research is needed to identify responders and/or means to achieve durable remissions in order to successfully exploit IGF-1R signal blockade in Ewing sarcoma (clinicaltrials.gov: NCT00617890).
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Brian Lu
- Merck & Co., IncKenilworthNew Jersey
| | | |
Collapse
|
3
|
Peterse EFP, Cleven AHG, De Jong Y, Briaire-de Bruijn I, Fletcher JA, Danen EHJ, Cleton-Jansen AM, Bovée JVMG. No preclinical rationale for IGF1R directed therapy in chondrosarcoma of bone. BMC Cancer 2016; 16:475. [PMID: 27418340 PMCID: PMC4946092 DOI: 10.1186/s12885-016-2522-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 07/05/2016] [Indexed: 12/23/2022] Open
Abstract
Background Chondrosarcoma is a malignant cartilage forming bone tumour for which no effective systemic treatment is available. Previous studies illustrate the need for a better understanding of the role of the IGF pathway in chondrosarcoma to determine if it can be a target for therapy, which was therefore explored in this study. Methods Expression of mediators of IGF1R signalling and phosphorylation status of IRS1 was determined in chondrosarcoma cell lines by qRT-PCR and western blot. The effect of activation and inhibition of IGF1R signalling on downstream targets was assessed by western blot. Ten chondrosarcoma cell lines were treated with OSI-906 (IGF1R and IR dual inhibitor) after which cell proliferation and migration were determined by a viability assay and the xCELLigence system, respectively. In addition, four chondrosarcoma cell lines were treated with a combination of doxorubicin and OSI-906. By immunohistochemistry, IGF1R expression levels were determined in tissue microarrays of 187 cartilage tumours and ten paraffin embedded cell lines. Results Mediators of IGF1R signalling are heterogeneously expressed and phosphorylated IRS1 was detected in 67 % of the tested chondrosarcoma cell lines, suggesting that IGF1R signalling is active in a subset of chondrosarcoma cell lines. In the cell lines with phosphorylated IRS1, inhibition of IGF1R signalling decreased phosphorylated Akt levels and increased IGF1R expression, but it did not influence MAPK or S6 activity. In line with these findings, treatment with IGF1R/IR inhibitors did not impact proliferation or migration in any of the chondrosarcoma cell lines, even upon stimulation with IGF1. Although synergistic effects of IGF1R/IR inhibition with doxorubicin are described for other cancers, our results demonstrate that this was not the case for chondrosarcoma. In addition, we found minimal IGF1R expression in primary tumours in contrast to the high expression detected in chondrosarcoma cell lines, even if both were derived from the same tumour, suggesting that in vitro culturing upregulates IGF1R expression. Conclusions The results from this study indicate that the IGF pathway is not essential for chondrosarcoma growth, migration or chemoresistance. Furthermore, IGF1R is only minimally expressed in chondrosarcoma primary tumours. Therefore, the IGF pathway is not expected to be an effective therapeutic target for chondrosarcoma of bone. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2522-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Arjen H G Cleven
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Yvonne De Jong
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Jonathan A Fletcher
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Erik H J Danen
- Division of Toxicology, Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands
| | | | - Judith V M G Bovée
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
4
|
Brownhill S, Cohen D, Burchill S. Proliferation index: a continuous model to predict prognosis in patients with tumours of the Ewing's sarcoma family. PLoS One 2014; 9:e104106. [PMID: 25157404 PMCID: PMC4144797 DOI: 10.1371/journal.pone.0104106] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 07/09/2014] [Indexed: 01/03/2023] Open
Abstract
The prognostic value of proliferation index (PI) and apoptotic index (AI), caspase-8, -9 and -10 expression have been investigated in primary Ewing's sarcoma family of tumours (ESFT). Proliferating cells, detected by immunohistochemistry for Ki-67, were identified in 91% (91/100) of tumours with a median PI of 14 (range 0-87). Apoptotic cells, identified using the TUNEL assay, were detected in 96% (76/79) of ESFT; the median AI was 3 (range 0-33). Caspase-8 protein expression was negative (0) in 14% (11/79), low (1) in 33% (26/79), medium (2) in 38% (30/79) and high (3) in 15% (12/79) of tumours, caspase-9 expression was low (1) in 66% (39/59) and high (3) in 34% (20/59), and caspase-10 protein was low (1) in 37% (23/62) and negative (0) in 63% (39/62) of primary ESFT. There was no apparent relationship between caspase-8, -9 and -10 expression, PI and AI. PI was predictive of relapse-free survival (RFS; p = 0.011) and overall survival (OS; p = <0.001) in a continuous model, whereas AI did not predict outcome. Patients with tumours expressing low levels of caspase-9 protein had a trend towards a worse RFS than patients with tumours expressing higher levels of caspase-9 protein (p = 0.054, log rank test), although expression of caspases-8, -9 and/or -10 did not significantly predict RFS or OS. In a multivariate analysis model that included tumour site, tumour volume, the presence of metastatic disease at diagnosis, PI and AI, PI independently predicts OS (p = 0.003). Consistent with previous publications, patients with pelvic tumours had a significantly worse OS than patients with tumours at other sites (p = 0.028); patients with a pelvic tumour and a PI≥20 had a 6 fold-increased risk of death. These studies advocate the evaluation of PI in a risk model of outcome for patients with ESFT.
Collapse
Affiliation(s)
- Samantha Brownhill
- Children's Cancer Research Group, Leeds Institute of Cancer and Pathology, St James's University Hospital, Leeds, United Kingdom
- * E-mail:
| | - Dena Cohen
- Clinical Trials Research Unit, Leeds Institute of Cancer and Pathology, St James's University Hospital, Leeds, United Kingdom
| | - Sue Burchill
- Children's Cancer Research Group, Leeds Institute of Cancer and Pathology, St James's University Hospital, Leeds, United Kingdom
| |
Collapse
|
5
|
Targeted therapies for bone sarcomas. BONEKEY REPORTS 2013; 2:378. [PMID: 24422100 DOI: 10.1038/bonekey.2013.112] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Revised: 06/07/2013] [Accepted: 06/11/2013] [Indexed: 02/08/2023]
Abstract
Bone sarcomas include a very large number of tumour subtypes, which originate form bone and more particularly from mesenchymal stem cell lineage. Osteosarcoma, Ewing's sarcoma and chondrosarcoma, the three main bone sarcoma entities develop in a favourable microenvironment composed by bone cells, blood vessels, immune cells, based on the 'seed and soil theory'. Current therapy associates surgery and chemotherapy, however, bone sarcomas remain diseases with high morbidity and mortality especially in children and adolescents. In the past decade, various new therapeutic approaches emerged and target the tumour niche or/and directly the tumour cells by acting on signalling/metabolic pathways involved in cell proliferation, apoptosis or drug resistance. The present review gives a brief overview from basic to clinical assessment of the main targeted therapies of bone sarcoma cells.
Collapse
|