1
|
Zobeck M, Khan J, Venkatramani R, Okcu MF, Scheurer ME, Lupo PJ. Improving Individualized Rhabdomyosarcoma Prognosis Predictions Using Somatic Molecular Biomarkers. JCO Precis Oncol 2025; 9:e2400556. [PMID: 39913888 PMCID: PMC11801453 DOI: 10.1200/po-24-00556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 11/19/2024] [Accepted: 01/06/2025] [Indexed: 02/09/2025] Open
Abstract
PURPOSE Molecular markers increasingly influence risk-stratified treatment selection for pediatric rhabdomyosarcoma (RMS). This study aims to integrate molecular and clinical data to produce individualized prognosis predictions that can further improve treatment selection. METHODS Clinical variables and somatic mutation data for 20 genes from 641 patients with RMS in the United Kingdom and the United States were used to develop three Cox proportional hazard models for predicting event-free survival (EFS). The Baseline Clinical (BC) model included treatment location, age, fusion status, and risk group. The Gene Enhanced 2 (GE2) model added TP53 and MYOD1 mutations to the BC predictors. The Gene Enhanced 6 (GE6) model further included NF1, MET, CDKN2A, and MYCN mutations, selected through least absolute shrinkage and selection operator regression. Model performance was assessed using likelihood ratio tests and optimism-adjusted, bootstrapped validation and calibration metrics. RESULTS The GE6 model demonstrated superior predictive performance compared with the BC model (P < .001) and GE2 model (P < .001). The GE6 model achieved the highest discrimination with a time-dependent area under the receiver operating characteristic curve of 0.766. Mutations in TP53, MYOD1, CDKN2A, MET, and MYCN were associated with higher hazards, while NF1 mutation correlated with lower hazard. Individual prognosis predictions varied between models in ways that may suggest different treatments for the same patient. For example, the 5-year EFS for a 10-year-old patient with high-risk, fusion-negative, NF1-positive disease was 50.0% (95% CI, 39 to 64) from BC but 76% (64 to 90) from GE6. CONCLUSION Incorporating molecular markers into RMS prognosis models improves prognosis predictions. Individualized prognosis predictions may suggest alternative treatment regimens compared with traditional risk-classification schemas. Improved clinical variables and external validation are required before implementing these models into clinical practice.
Collapse
Affiliation(s)
- Mark Zobeck
- Department of Pediatrics, Division of Hematology/Oncology, Baylor College of Medicine, Houston, TX
- Texas Children's Cancer and Hematology Centers, Texas Children's Hospital, Houston, TX
| | - Javed Khan
- Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Rajkumar Venkatramani
- Department of Pediatrics, Division of Hematology/Oncology, Baylor College of Medicine, Houston, TX
- Texas Children's Cancer and Hematology Centers, Texas Children's Hospital, Houston, TX
| | - M. Fatih Okcu
- Department of Pediatrics, Division of Hematology/Oncology, Baylor College of Medicine, Houston, TX
- Texas Children's Cancer and Hematology Centers, Texas Children's Hospital, Houston, TX
| | - Michael E. Scheurer
- Department of Pediatrics, Division of Hematology/Oncology, Baylor College of Medicine, Houston, TX
- Texas Children's Cancer and Hematology Centers, Texas Children's Hospital, Houston, TX
| | - Philip J. Lupo
- Department of Pediatrics, Division of Hematology/Oncology, Baylor College of Medicine, Houston, TX
- Texas Children's Cancer and Hematology Centers, Texas Children's Hospital, Houston, TX
| |
Collapse
|
2
|
Zobeck M, Khan J, Venkatramani R, Okcu MF, Scheurer ME, Lupo PJ. Improving Individualized Rhabdomyosarcoma Prognosis Predictions Using Somatic Molecular Biomarkers. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.04.24313032. [PMID: 39281734 PMCID: PMC11398450 DOI: 10.1101/2024.09.04.24313032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Abstract
Purpose Molecular markers, such as FOXO1 fusion genes and TP53 and MYOD1 mutations, increasingly influence risk-stratified treatment selection for pediatric rhabdomyosarcoma (RMS). This study aims to integrate molecular and clinical data to produce individualized prognosis predictions that can further improve treatment selection. Patients and Methods Clinical variables and somatic mutation data for 20 genes from 641 RMS patients in the United Kingdom and the United States were used to develop three Cox proportional hazard models for predicting event-free survival (EFS). The 'Baseline Clinical' (BC) model included treatment location, age, fusion status, and risk group. The 'Gene Enhanced 2' (GE2) model added TP53 and MYOD1 mutations to the BC predictors. The 'Gene Enhanced 6' (GE6) model further included NF1, MET, CDKN2A, and MYCN mutations, selected through LASSO regression. Model performance was assessed using likelihood ratio (LR) tests and optimism-adjusted, bootstrapped validation and calibration metrics. Results The GE6 model demonstrated superior predictive performance, offering 39% more predictive information than the BC model (LR p<0.001) and 15% more than the GE2 model (LR p<0.001). The GE6 model achieved the highest discrimination with a C-index of 0.7087, a Nagalkerke R2 of 0.205, and appropriate calibration. Mutations in TP53, MYOD1, CDKN2A, MET, and MYCN were associated with higher hazards, while NF1 mutation correlated with lower hazard. Individual prognosis predictions varied between models in ways that may suggest different treatments for the same patient. For example, the 5-year EFS for a 10-year-old patient with high-risk, fusion-negative, NF1-positive disease was 50.0% (95% confidence interval: 39-64%) from BC but 76% (64-90%) from GE6. Conclusion Incorporating molecular markers into RMS prognosis models improves prognosis predictions. Individualized prognosis predictions may suggest alternative treatment regimens compared to traditional risk-classification schemas. Improved clinical variables and external validation are required prior to implementing these models into clinical practice.
Collapse
Affiliation(s)
- Mark Zobeck
- Baylor College of Medicine, Department of Pediatrics, Division of Hematology/Oncology, Houston, Texas
- Texas Children’s Hospital, Texas Children’s Cancer and Hematology Centers, Houston, Texas
| | - Javed Khan
- Center for Cancer Research, National Cancer Institute, Bethesda, 004Daryland
| | - Rajkumar Venkatramani
- Baylor College of Medicine, Department of Pediatrics, Division of Hematology/Oncology, Houston, Texas
- Texas Children’s Hospital, Texas Children’s Cancer and Hematology Centers, Houston, Texas
| | - M. Fatih Okcu
- Baylor College of Medicine, Department of Pediatrics, Division of Hematology/Oncology, Houston, Texas
- Texas Children’s Hospital, Texas Children’s Cancer and Hematology Centers, Houston, Texas
| | - Michael E. Scheurer
- Baylor College of Medicine, Department of Pediatrics, Division of Hematology/Oncology, Houston, Texas
- Texas Children’s Hospital, Texas Children’s Cancer and Hematology Centers, Houston, Texas
| | - Philip J. Lupo
- Baylor College of Medicine, Department of Pediatrics, Division of Hematology/Oncology, Houston, Texas
- Texas Children’s Hospital, Texas Children’s Cancer and Hematology Centers, Houston, Texas
| |
Collapse
|
3
|
Talapatra J, Reddy MM. Lipid Metabolic Reprogramming in Embryonal Neoplasms with MYCN Amplification. Cancers (Basel) 2023; 15:cancers15072144. [PMID: 37046804 PMCID: PMC10093342 DOI: 10.3390/cancers15072144] [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: 02/27/2023] [Revised: 03/18/2023] [Accepted: 03/21/2023] [Indexed: 04/14/2023] Open
Abstract
Tumor cells reprogram their metabolism, including glucose, glutamine, nucleotide, lipid, and amino acids to meet their enhanced energy demands, redox balance, and requirement of biosynthetic substrates for uncontrolled cell proliferation. Altered lipid metabolism in cancer provides lipids for rapid membrane biogenesis, generates the energy required for unrestricted cell proliferation, and some of the lipids act as signaling pathway mediators. In this review, we focus on the role of lipid metabolism in embryonal neoplasms with MYCN dysregulation. We specifically review lipid metabolic reactions in neuroblastoma, retinoblastoma, medulloblastoma, Wilms tumor, and rhabdomyosarcoma and the possibility of targeting lipid metabolism. Additionally, the regulation of lipid metabolism by the MYCN oncogene is discussed.
Collapse
Affiliation(s)
- Jyotirmayee Talapatra
- The Operation Eyesight Universal Institute for Eye Cancer, L V Prasad Eye Institute, Bhubaneswar 751024, India
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India
| | - Mamatha M Reddy
- The Operation Eyesight Universal Institute for Eye Cancer, L V Prasad Eye Institute, Bhubaneswar 751024, India
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India
| |
Collapse
|
4
|
Ramadan F, Saab R, Hussein N, Clézardin P, Cohen PA, Ghayad SE. Non-coding RNA in rhabdomyosarcoma progression and metastasis. Front Oncol 2022; 12:971174. [PMID: 36033507 PMCID: PMC9403786 DOI: 10.3389/fonc.2022.971174] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 07/25/2022] [Indexed: 12/12/2022] Open
Abstract
Rhabdomyosarcoma (RMS) is a soft tissue sarcoma of skeletal muscle differentiation, with a predominant occurrence in children and adolescents. One of the major challenges facing treatment success is the presence of metastatic disease at the time of diagnosis, commonly associated with the more aggressive fusion-positive subtype. Non-coding RNA (ncRNA) can regulate gene transcription and translation, and their dysregulation has been associated with cancer development and progression. MicroRNA (miRNA) are short non-coding nucleic acid sequences involved in the regulation of gene expression that act by targeting messenger RNA (mRNA), and their aberrant expression has been associated with both RMS initiation and progression. Other ncRNA including long non-coding RNA (lncRNA), circular RNA (circRNA) and ribosomal RNA (rRNA) have also been associated with RMS revealing important mechanistic roles in RMS biology, but these studies are still limited and require further investigation. In this review, we discuss the established roles of ncRNA in RMS differentiation, growth and progression, highlighting their potential use in RMS prognosis, as therapeutic agents or as targets of treatment.
Collapse
Affiliation(s)
- Farah Ramadan
- Department of Biology, Faculty of Science II, Lebanese University, Beirut, Lebanon
- Université Claude Bernard Lyon 1, Lyon, France
- INSERM, Unit 1033, LYOS, Lyon, France
- Department of Chemistry and Biochemistry, Laboratory of Cancer Biology and Molecular Immunology, Faculty of Science I, Lebanese University, Hadat, Lebanon
| | - Raya Saab
- Department of Anatomy, Cell Biology and Physiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Department of Pediatric and Adolescent Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Nader Hussein
- Department of Chemistry and Biochemistry, Laboratory of Cancer Biology and Molecular Immunology, Faculty of Science I, Lebanese University, Hadat, Lebanon
| | - Philippe Clézardin
- Université Claude Bernard Lyon 1, Lyon, France
- INSERM, Unit 1033, LYOS, Lyon, France
| | - Pascale A. Cohen
- Université Claude Bernard Lyon 1, Lyon, France
- INSERM, Unit 1033, LYOS, Lyon, France
| | - Sandra E. Ghayad
- Department of Biology, Faculty of Science II, Lebanese University, Beirut, Lebanon
- Aix-Marseille University, INSERM 1263, INRAE 1260, C2VN, Marseille, France
| |
Collapse
|
5
|
Therapeutic targeting of ATR in alveolar rhabdomyosarcoma. Nat Commun 2022; 13:4297. [PMID: 35879366 PMCID: PMC9314382 DOI: 10.1038/s41467-022-32023-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 07/11/2022] [Indexed: 11/08/2022] Open
Abstract
Despite advances in multi-modal treatment approaches, clinical outcomes of patients suffering from PAX3-FOXO1 fusion oncogene-expressing alveolar rhabdomyosarcoma (ARMS) remain dismal. Here we show that PAX3-FOXO1-expressing ARMS cells are sensitive to pharmacological ataxia telangiectasia and Rad3 related protein (ATR) inhibition. Expression of PAX3-FOXO1 in muscle progenitor cells is not only sufficient to increase sensitivity to ATR inhibition, but PAX3-FOXO1-expressing rhabdomyosarcoma cells also exhibit increased sensitivity to structurally diverse inhibitors of ATR. Mechanistically, ATR inhibition leads to replication stress exacerbation, decreased BRCA1 phosphorylation and reduced homologous recombination-mediated DNA repair pathway activity. Consequently, ATR inhibitor treatment increases sensitivity of ARMS cells to PARP1 inhibition in vitro, and combined treatment with ATR and PARP1 inhibitors induces complete regression of primary patient-derived ARMS xenografts in vivo. Lastly, a genome-wide CRISPR activation screen (CRISPRa) in combination with transcriptional analyses of ATR inhibitor resistant ARMS cells identifies the RAS-MAPK pathway and its targets, the FOS gene family, as inducers of resistance to ATR inhibition. Our findings provide a rationale for upcoming biomarker-driven clinical trials of ATR inhibitors in patients suffering from ARMS.
Collapse
|
6
|
Singh S, Abu-Zaid A, Jin H, Fang J, Wu Q, Wang T, Feng H, Quarni W, Shao Y, Maxham L, Abdolvahabi A, Yun MK, Vaithiyalingam S, Tan H, Bowling J, Honnell V, Young B, Guo Y, Bajpai R, Pruett-Miller SM, Grosveld GC, Hatley M, Xu B, Fan Y, Wu G, Chen EY, Chen T, Lewis PW, Rankovic Z, Li Y, Murphy AJ, Easton J, Peng J, Chen X, Wang R, White SW, Davidoff AM, Yang J. Targeting KDM4 for treating PAX3-FOXO1-driven alveolar rhabdomyosarcoma. Sci Transl Med 2022; 14:eabq2096. [PMID: 35857643 PMCID: PMC9548378 DOI: 10.1126/scitranslmed.abq2096] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Chimeric transcription factors drive lineage-specific oncogenesis but are notoriously difficult to target. Alveolar rhabdomyosarcoma (RMS) is an aggressive childhood soft tissue sarcoma transformed by the pathognomonic Paired Box 3-Forkhead Box O1 (PAX3-FOXO1) fusion protein, which governs a core regulatory circuitry transcription factor network. Here, we show that the histone lysine demethylase 4B (KDM4B) is a therapeutic vulnerability for PAX3-FOXO1+ RMS. Genetic and pharmacologic inhibition of KDM4B substantially delayed tumor growth. Suppression of KDM4 proteins inhibited the expression of core oncogenic transcription factors and caused epigenetic alterations of PAX3-FOXO1-governed superenhancers. Combining KDM4 inhibition with cytotoxic chemotherapy led to tumor regression in preclinical PAX3-FOXO1+ RMS subcutaneous xenograft models. In summary, we identified a targetable mechanism required for maintenance of the PAX3-FOXO1-related transcription factor network, which may translate to a therapeutic approach for fusion-positive RMS.
Collapse
Affiliation(s)
- Shivendra Singh
- Department of Surgery, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Ahmed Abu-Zaid
- Department of Surgery, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Hongjian Jin
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Jie Fang
- Department of Surgery, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Qiong Wu
- Department of Surgery, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Tingting Wang
- Center for Childhood Cancer and Blood Disease, Abigail Wexner Research Institute, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH 43205, USA
| | - Helin Feng
- Department of Orthopedics, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China
| | - Waise Quarni
- Department of Surgery, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Ying Shao
- Department of Computational Biology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Lily Maxham
- Department of Computational Biology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Alireza Abdolvahabi
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Mi-Kyung Yun
- Department of Structural Biology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Sivaraja Vaithiyalingam
- Department of Structural Biology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
- Protein Technologies Center, Molecular Interaction Analysis, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Haiyan Tan
- Department of Structural Biology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - John Bowling
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Victoria Honnell
- Graduate School of Biomedical Sciences, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Brandon Young
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Yian Guo
- Department of Biostatistics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Richa Bajpai
- Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Shondra M Pruett-Miller
- Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Gerard C Grosveld
- Department of Genetics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Mark Hatley
- Department of Oncology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Beisi Xu
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Yiping Fan
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Gang Wu
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Eleanor Y Chen
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| | - Taosheng Chen
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Peter W Lewis
- Department of Biomolecular Chemistry, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53706, USA
| | - Zoran Rankovic
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Yimei Li
- Department of Biostatistics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Andrew J Murphy
- Department of Surgery, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - John Easton
- Department of Computational Biology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Junmin Peng
- Department of Structural Biology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Xiang Chen
- Department of Computational Biology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Ruoning Wang
- Center for Childhood Cancer and Blood Disease, Abigail Wexner Research Institute, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH 43205, USA
| | - Stephen W White
- Department of Structural Biology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
- Graduate School of Biomedical Sciences, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Andrew M Davidoff
- Department of Surgery, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Jun Yang
- Department of Surgery, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
- Graduate School of Biomedical Sciences, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
- Department of Pathology, College of Medicine, The University of Tennessee Health Science Center, 930 Madison Ave., Suite 500, Memphis, TN 38163, USA
| |
Collapse
|
7
|
FOXF1 is required for the oncogenic properties of PAX3-FOXO1 in rhabdomyosarcoma. Oncogene 2021; 40:2182-2199. [PMID: 33627785 PMCID: PMC8005492 DOI: 10.1038/s41388-021-01694-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 01/13/2021] [Accepted: 02/01/2021] [Indexed: 01/31/2023]
Abstract
The PAX3-FOXO1 fusion protein is the key oncogenic driver in fusion positive rhabdomyosarcoma (FP-RMS), an aggressive soft tissue malignancy with a particularly poor prognosis. Identifying key downstream targets of PAX3-FOXO1 will provide new therapeutic opportunities for treatment of FP-RMS. Herein, we demonstrate that Forkhead Box F1 (FOXF1) transcription factor is uniquely expressed in FP-RMS and is required for FP-RMS tumorigenesis. The PAX3-FOXO1 directly binds to FOXF1 enhancers and induces FOXF1 gene expression. CRISPR/Cas9 mediated inactivation of either FOXF1 coding sequence or FOXF1 enhancers suppresses FP-RMS tumorigenesis even in the presence of PAX3-FOXO1 oncogene. Knockdown or genetic knockout of FOXF1 induces myogenic differentiation in PAX3-FOXO1-positive FP-RMS. Over-expression of FOXF1 decreases myogenic differentiation in primary human myoblasts. In FP-RMS tumor cells, FOXF1 protein binds chromatin near enhancers associated with FP-RMS gene signature. FOXF1 cooperates with PAX3-FOXO1 and E-box transcription factors MYOD1 and MYOG to regulate FP-RMS-specific gene expression. Altogether, FOXF1 functions downstream of PAX3-FOXO1 to promote FP-RMS tumorigenesis.
Collapse
|
8
|
Petragnano F, Pietrantoni I, Camero S, Codenotti S, Milazzo L, Vulcano F, Macioce G, Giordani I, Tini P, Cheleschi S, Gravina GL, Festuccia C, Rossetti A, Delle Monache S, Ordinelli A, Ciccarelli C, Mauro A, Barbara B, Antinozzi C, Schiavetti A, Maggio R, Di Luigi L, Polimeni A, Marchese C, Tombolini V, Fanzani A, Bernabò N, Megiorni F, Marampon F. Clinically relevant radioresistant rhabdomyosarcoma cell lines: functional, molecular and immune-related characterization. J Biomed Sci 2020; 27:90. [PMID: 32854690 PMCID: PMC7453562 DOI: 10.1186/s12929-020-00683-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 08/24/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The probability of local tumor control after radiotherapy (RT) remains still miserably poor in pediatric rhabdomyosarcoma (RMS). Thus, understanding the molecular mechanisms responsible of tumor relapse is essential to identify personalized RT-based strategies. Contrary to what has been done so far, a correct characterization of cellular radioresistance should be performed comparing radioresistant and radiosensitive cells with the same isogenic background. METHODS Clinically relevant radioresistant (RR) embryonal (RD) and alveolar (RH30) RMS cell lines have been developed by irradiating them with clinical-like hypo-fractionated schedule. RMS-RR cells were compared to parental isogenic counterpart (RMS-PR) and studied following the radiobiological concept of the "6Rs", which stand for repair, redistribution, repopulation, reoxygenation, intrinsic radioresistance and radio-immuno-biology. RESULTS RMS-RR cell lines, characterized by a more aggressive and in vitro pro-metastatic phenotype, showed a higher ability to i) detoxify from reactive oxygen species; ii) repair DNA damage by differently activating non-homologous end joining and homologous recombination pathways; iii) counteract RT-induced G2/M cell cycle arrest by re-starting growth and repopulating after irradiation; iv) express cancer stem-like profile. Bioinformatic analyses, performed to assess the role of 41 cytokines after RT exposure and their network interactions, suggested TGF-β, MIF, CCL2, CXCL5, CXCL8 and CXCL12 as master regulators of cancer immune escape in RMS tumors. CONCLUSIONS These results suggest that RMS could sustain intrinsic and acquire radioresistance by different mechanisms and indicate potential targets for future combined radiosensitizing strategies.
Collapse
Affiliation(s)
- Francesco Petragnano
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Ilaria Pietrantoni
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Simona Camero
- Department of Maternal, Infantile, and Urological Sciences, "Sapienza" University of Rome, Rome, Italy
| | - Silvia Codenotti
- Department of Molecular and Translational Medicine, Division of Biotechnology, University of Brescia, Brescia, Italy
| | - Luisa Milazzo
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena, Rome, Italy
| | - Francesca Vulcano
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena, Rome, Italy
| | - Giampiero Macioce
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena, Rome, Italy
| | - Ilenia Giordani
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Section of Histology and Medical Embryology, "Sapienza" University, Rome, Italy
| | - Paolo Tini
- Sbarro Health Research Organization, Temple University, Philadelphia, PA, USA
- Unit of Radiation Oncology, University Hospital of Siena, Siena, Italy
| | - Sara Cheleschi
- Department of Medicine, Surgery and Neuroscience, Rheumatology Unit, University of Siena, Policlinico Le Scotte, Siena, Italy
| | - Giovanni Luca Gravina
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Claudio Festuccia
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Alessandra Rossetti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Simona Delle Monache
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Alessandra Ordinelli
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Carmela Ciccarelli
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Annunziata Mauro
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Barboni Barbara
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Cristina Antinozzi
- Unit of Endocrinology, Department of Movement, Human and Health Sciences, University of Rome "Foro Italico", Rome, Italy
| | - Amalia Schiavetti
- Department of Maternal, Infantile, and Urological Sciences, "Sapienza" University of Rome, Rome, Italy
| | - Roberto Maggio
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Luigi Di Luigi
- Unit of Endocrinology, Department of Movement, Human and Health Sciences, University of Rome "Foro Italico", Rome, Italy
| | - Antonella Polimeni
- Department of Oral and Maxillo-Facial Sciences, Sapienza University of Rome, Rome, Italy
| | - Cinzia Marchese
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Vincenzo Tombolini
- Department of Radiotherapy, Policlinico Umberto I, "Sapienza" University of Rome, Rome, Italy
| | - Alessandro Fanzani
- Department of Molecular and Translational Medicine, Division of Biotechnology, University of Brescia, Brescia, Italy
| | - Nicola Bernabò
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Francesca Megiorni
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Francesco Marampon
- Department of Radiotherapy, Policlinico Umberto I, "Sapienza" University of Rome, Rome, Italy.
| |
Collapse
|
9
|
Andersson N, Bakker B, Karlsson J, Valind A, Holmquist Mengelbier L, Spierings DCJ, Foijer F, Gisselsson D. Extensive Clonal Branching Shapes the Evolutionary History of High-Risk Pediatric Cancers. Cancer Res 2020; 80:1512-1523. [PMID: 32041836 DOI: 10.1158/0008-5472.can-19-3468] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 12/18/2019] [Accepted: 02/04/2020] [Indexed: 11/16/2022]
Abstract
Darwinian evolution of tumor cells remains underexplored in childhood cancer. We here reconstruct the evolutionary histories of 56 pediatric primary tumors, including 24 neuroblastomas, 24 Wilms tumors, and 8 rhabdomyosarcomas. Whole-genome copy-number and whole-exome mutational profiling of multiple regions per tumor were performed, followed by clonal deconvolution to reconstruct a phylogenetic tree for each tumor. Overall, 88% of the tumors exhibited genetic variation among primary tumor regions. This variability typically emerged through collateral phylogenetic branching, leading to spatial variability in the distribution of more than 50% (96/173) of detected diagnostically informative genetic aberrations. Single-cell sequencing of 547 individual cancer cells from eight solid pediatric tumors confirmed branching evolution to be a fundamental underlying principle of genetic variation in all cases. Strikingly, cell-to-cell genetic diversity was almost twice as high in aggressive compared with clinically favorable tumors (median Simpson index of diversity 0.45 vs. 0.88; P = 0.029). Similarly, a comparison of multiregional sampling data from a total of 274 tumor regions showed that new phylogenetic branches emerge at a higher frequency per sample and carry a higher mutational load in high-risk than in low-risk tumors. Timelines based on spatial genetic variation showed that the mutations most influencing relapse risk occur at initiation of clonal expansion in neuroblastoma and rhabdomyosarcoma, whereas in Wilms tumor, they are late events. Thus, from an evolutionary standpoint, some high-risk childhood cancers are born bad, whereas others grow worse over time. SIGNIFICANCE: Different pediatric cancers with a high risk of relapse share a common generic pattern of extensively branching evolution of somatic mutations. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/7/1512/F1.large.jpg.
Collapse
Affiliation(s)
- Natalie Andersson
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Bjorn Bakker
- European Research Institute for the Biology of Ageing (ERIBA), University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Jenny Karlsson
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Anders Valind
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden.,Department of Pediatrics, Skåne University Hospital, Lund, Sweden
| | | | - Diana C J Spierings
- European Research Institute for the Biology of Ageing (ERIBA), University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Floris Foijer
- European Research Institute for the Biology of Ageing (ERIBA), University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - David Gisselsson
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden. .,Division of Oncology-Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden.,Clinical Genetics and Pathology, Laboratory Medicine, Lund University Hospital, Skåne Healthcare Region, Lund, Sweden
| |
Collapse
|
10
|
Ommer J, Selfe JL, Wachtel M, O'Brien EM, Laubscher D, Roemmele M, Kasper S, Delattre O, Surdez D, Petts G, Kelsey A, Shipley J, Schäfer BW. Aurora A Kinase Inhibition Destabilizes PAX3-FOXO1 and MYCN and Synergizes with Navitoclax to Induce Rhabdomyosarcoma Cell Death. Cancer Res 2019; 80:832-842. [PMID: 31888889 DOI: 10.1158/0008-5472.can-19-1479] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 09/12/2019] [Accepted: 12/18/2019] [Indexed: 11/16/2022]
Abstract
The clinically aggressive alveolar rhabdomyosarcoma (RMS) subtype is characterized by expression of the oncogenic fusion protein PAX3-FOXO1, which is critical for tumorigenesis and cell survival. Here, we studied the mechanism of cell death induced by loss of PAX3-FOXO1 expression and identified a novel pharmacologic combination therapy that interferes with PAX3-FOXO1 biology at different levels. Depletion of PAX3-FOXO1 in fusion-positive (FP)-RMS cells induced intrinsic apoptosis in a NOXA-dependent manner. This was pharmacologically mimicked by the BH3 mimetic navitoclax, identified as top compound in a screen from 208 targeted compounds. In a parallel approach, and to identify drugs that alter the stability of PAX3-FOXO1 protein, the same drug library was screened and fusion protein levels were directly measured as a read-out. This revealed that inhibition of Aurora kinase A most efficiently negatively affected PAX3-FOXO1 protein levels. Interestingly, this occurred through a novel specific phosphorylation event in and binding to the fusion protein. Aurora kinase A inhibition also destabilized MYCN, which is both a functionally important oncogene and transcriptional target of PAX3-FOXO1. Combined treatment with an Aurora kinase A inhibitor and navitoclax in FP-RMS cell lines and patient-derived xenografts synergistically induced cell death and significantly slowed tumor growth. These studies identify a novel functional interaction of Aurora kinase A with both PAX3-FOXO1 and its effector MYCN, and reveal new opportunities for targeted combination treatment of FP-RMS. SIGNIFICANCE: These findings show that Aurora kinase A and Bcl-2 family proteins are potential targets for FP-RMS.
Collapse
Affiliation(s)
- Johannes Ommer
- Department of Oncology and Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Joanna L Selfe
- Sarcoma Molecular Pathology Laboratory, The Institute of Cancer Research, London, United Kingdom
| | - Marco Wachtel
- Department of Oncology and Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Eleanor M O'Brien
- Sarcoma Molecular Pathology Laboratory, The Institute of Cancer Research, London, United Kingdom
| | - Dominik Laubscher
- Department of Oncology and Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Michaela Roemmele
- Department of Oncology and Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Stephanie Kasper
- Department of Oncology and Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Olivier Delattre
- France INSERM U830, Équipe Labellisé LNCC, PSL Université, SIREDO Oncology Centre, Institut Curie, Paris, France
| | - Didier Surdez
- France INSERM U830, Équipe Labellisé LNCC, PSL Université, SIREDO Oncology Centre, Institut Curie, Paris, France
| | - Gemma Petts
- Department of Diagnostic Paediatric Histopathology, Royal Manchester Children's Hospital, Manchester, United Kingdom
| | - Anna Kelsey
- Department of Diagnostic Paediatric Histopathology, Royal Manchester Children's Hospital, Manchester, United Kingdom
| | - Janet Shipley
- Sarcoma Molecular Pathology Laboratory, The Institute of Cancer Research, London, United Kingdom
| | - Beat W Schäfer
- Department of Oncology and Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland.
| |
Collapse
|
11
|
Gatz SA, Aladowicz E, Casanova M, Chisholm JC, Kearns PR, Fulda S, Geoerger B, Schäfer BW, Shipley JM. A Perspective on Polo-Like Kinase-1 Inhibition for the Treatment of Rhabdomyosarcomas. Front Oncol 2019; 9:1271. [PMID: 31824851 PMCID: PMC6882953 DOI: 10.3389/fonc.2019.01271] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 11/04/2019] [Indexed: 12/13/2022] Open
Abstract
Rhabdomyosarcomas are the most common pediatric soft tissue sarcoma and are a major cause of death from cancer in young patients requiring new treatment options to improve outcomes. High-risk patients include those with metastatic or relapsed disease and tumors with PAX3-FOXO1 fusion genes that encode a potent transcription factor that drives tumourigenesis through transcriptional reprogramming. Polo-Like Kinase-1 (PLK1) is a serine/threonine kinase that phosphorylates a wide range of target substrates and alters their activity. PLK1 functions as a pleiotropic master regulator of mitosis and regulates DNA replication after stress. Taken together with high levels of expression that correlate with poor outcomes in many cancers, including rhabdomyosarcomas, it is an attractive therapeutic target. This is supported in rhabdomyosarcoma models by characterization of molecular and phenotypic effects of reducing and inhibiting PLK1, including changes to the PAX3-FOXO1 fusion protein. However, as tumor re-growth has been observed, combination strategies are required. Here we review preclinical evidence and consider biological rationale for PLK1 inhibition in combination with drugs that promote apoptosis, interfere with activity of PAX3-FOXO1 and are synergistic with microtubule-destabilizing drugs such as vincristine. The preclinical effects of low doses of the PLK1 inhibitor volasertib in combination with vincristine, which is widely used in rhabdomyosarcoma treatment, show particular promise in light of recent clinical data in the pediatric setting that support achievable volasertib doses predicted to be effective. Further development of novel therapeutic strategies including PLK1 inhibition may ultimately benefit young patients with rhabdomyosarcoma and other cancers.
Collapse
Affiliation(s)
- Susanne A. Gatz
- Cancer Research UK Clinical Trials Unit (CRCTU), Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
- Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Ewa Aladowicz
- Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | | | - Julia C. Chisholm
- Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
- Children and Young People's Unit, The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Pamela R. Kearns
- Cancer Research UK Clinical Trials Unit (CRCTU), Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Simone Fulda
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University Frankfurt, Frankfurt, Germany
| | - Birgit Geoerger
- Gustave Roussy Cancer Campus, Department of Paediatric and Adolescent Oncology, Université Paris-Saclay, Villejuif, France
| | - Beat W. Schäfer
- Department of Oncology and Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Janet M. Shipley
- Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| |
Collapse
|
12
|
The PAX3-FOXO1 oncogene alters exosome miRNA content and leads to paracrine effects mediated by exosomal miR-486. Sci Rep 2019; 9:14242. [PMID: 31578374 PMCID: PMC6775163 DOI: 10.1038/s41598-019-50592-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 09/16/2019] [Indexed: 12/11/2022] Open
Abstract
Rhabdomyosarcoma (RMS) is the most common soft tissue sarcoma in children. The alveolar subtype (ARMS) is clinically more aggressive, and characterized by an oncogenic fusion protein PAX3-FOXO1 that drives oncogenic cellular properties. Exosomes are small, secreted vesicles that affect paracrine signaling. We show that PAX3-FOXO1 transcript alters exosome content of C2C12 myoblasts, leading to pro-tumorigenic paracrine effects in recipient cells. Microarray analysis revealed alteration in miRNA content of exosomes, affecting cellular networks involved in cell metabolism, growth signaling, and cellular invasion. Overexpression and knockdown studies showed that miR-486-5p is an effector of PAX3-FOXO1, and mediates its paracrine effects in exosomes, including promoting recipient cell migration, invasion, and colony formation. Analysis of human RMS cells showed miR-486-5p is enriched in both cells and exosomes, and to a higher extent in ARMS subtypes. Analysis of human serum samples showed that miR-486-5p is enriched in exosomes of patients with RMS, and follow-up after chemotherapy showed decrease to control values. Our findings identify a novel role of both PAX3-FOXO1 and its downstream effector miR-486-5p in exosome-mediated oncogenic paracrine effects of RMS, and suggest its possible use as a biomarker.
Collapse
|
13
|
Bhat AV, Palanichamy Kala M, Rao VK, Pignata L, Lim HJ, Suriyamurthy S, Chang KT, Lee VK, Guccione E, Taneja R. Epigenetic Regulation of the PTEN-AKT-RAC1 Axis by G9a Is Critical for Tumor Growth in Alveolar Rhabdomyosarcoma. Cancer Res 2019; 79:2232-2243. [PMID: 30833420 DOI: 10.1158/0008-5472.can-18-2676] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 12/17/2018] [Accepted: 02/26/2019] [Indexed: 11/16/2022]
Abstract
Alveolar rhabdomyosarcoma (ARMS) is an aggressive pediatric cancer with poor prognosis. As transient and stable modifications to chromatin have emerged as critical mechanisms in oncogenic signaling, efforts to target epigenetic modifiers as a therapeutic strategy have accelerated in recent years. To identify chromatin modifiers that sustain tumor growth, we performed an epigenetic screen and found that inhibition of lysine methyltransferase G9a significantly affected the viability of ARMS cell lines. Targeting expression or activity of G9a reduced cellular proliferation and motility in vitro and tumor growth in vivo. Transcriptome and chromatin immunoprecipitation-sequencing analysis provided mechanistic evidence that the tumor-suppressor PTEN was a direct target gene of G9a. G9a repressed PTEN expression in a methyltransferase activity-dependent manner, resulting in increased AKT and RAC1 activity. Re-expression of constitutively active RAC1 in G9a-deficient tumor cells restored oncogenic phenotypes, demonstrating its critical functions downstream of G9a. Collectively, our study provides evidence for a G9a-dependent epigenetic program that regulates tumor growth and suggests targeting G9a as a therapeutic strategy in ARMS. SIGNIFICANCE: These findings demonstrate that RAC1 is an effector of G9a oncogenic functions and highlight the potential of G9a inhibitors in the treatment of ARMS.
Collapse
Affiliation(s)
- Akshay V Bhat
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Monica Palanichamy Kala
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Vinay Kumar Rao
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Luca Pignata
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Huey Jin Lim
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Sudha Suriyamurthy
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Kenneth T Chang
- Department of Pathology, KK Women and Children's Hospital, Singapore, Singapore
| | - Victor K Lee
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Ernesto Guccione
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Reshma Taneja
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
14
|
Kasiappan R, Jutooru I, Mohankumar K, Karki K, Lacey A, Safe S. Reactive Oxygen Species (ROS)-Inducing Triterpenoid Inhibits Rhabdomyosarcoma Cell and Tumor Growth through Targeting Sp Transcription Factors. Mol Cancer Res 2019; 17:794-805. [PMID: 30610105 PMCID: PMC6397684 DOI: 10.1158/1541-7786.mcr-18-1071] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 11/13/2018] [Accepted: 12/17/2018] [Indexed: 12/13/2022]
Abstract
Methyl 2-trifluoromethyl-3,11-dioxo-18β-olean-1,12-dien-3-oate (CF3DODA-Me) is derived synthetically from glycyrrhetinic acid, a major component of licorice, and this compound induced reactive oxygen species (ROS) in RD and Rh30 rhabdomyosarcoma (RMS) cells. CF3DODA-Me also inhibited growth and invasion and induced apoptosis in RMS cells, and these responses were attenuated after cotreatment with the antioxidant glutathione, demonstrating the effective anticancer activity of ROS in RMS. CF3DODA-Me also downregulated expression of specificity protein (Sp) transcription factors Sp1, Sp3, and Sp4 and prooncogenic Sp-regulated genes including PAX3-FOXO1 (in Rh30 cells). The mechanism of CF3DODA-Me-induced Sp-downregulation involved ROS-dependent repression of c-Myc and cMyc-regulated miR-27a and miR-17/20a, and this resulted in induction of the miRNA-regulated Sp repressors ZBTB4, ZBTB10, and ZBTB34. The cell and tumor growth effects of CF3DODA-Me further emphasize the sensitivity of RMS cells to ROS inducers and their potential clinical applications for treating this deadly disease. IMPLICATIONS: CF3DODA-Me and HDAC inhibitors that induce ROS-dependent Sp downregulation could be developed for clinical applications in treating rhabdomyosarcoma.
Collapse
Affiliation(s)
- Ravi Kasiappan
- Department of Biochemistry, CSIR-Central Food Technological Research Institute, Mysore, Karnataka, India
| | - Indira Jutooru
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas
| | - Kumaravel Mohankumar
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas
| | - Keshav Karki
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas
| | - Alexandra Lacey
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas.
| |
Collapse
|
15
|
Nguyen TH, Barr FG. Therapeutic Approaches Targeting PAX3-FOXO1 and Its Regulatory and Transcriptional Pathways in Rhabdomyosarcoma. Molecules 2018; 23:E2798. [PMID: 30373318 PMCID: PMC6278278 DOI: 10.3390/molecules23112798] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 10/24/2018] [Accepted: 10/26/2018] [Indexed: 02/06/2023] Open
Abstract
Rhabdomyosarcoma (RMS) is a family of soft tissue cancers that are related to the skeletal muscle lineage and predominantly occur in children and young adults. A specific chromosomal translocation t(2;13)(q35;q14) that gives rise to the chimeric oncogenic transcription factor PAX3-FOXO1 has been identified as a hallmark of the aggressive alveolar subtype of RMS. PAX3-FOXO1 cooperates with additional molecular changes to promote oncogenic transformation and tumorigenesis in various human and murine models. Its expression is generally restricted to RMS tumor cells, thus providing a very specific target for therapeutic approaches for these RMS tumors. In this article, we review the recent understanding of PAX3-FOXO1 as a transcription factor in the pathogenesis of this cancer and discuss recent developments to target this oncoprotein for treatment of RMS.
Collapse
Affiliation(s)
| | - Frederic G. Barr
- Laboratory of Pathology, National Cancer Institute, 10 Center Drive, Bethesda, MD 20892, USA;
| |
Collapse
|
16
|
Fanzani A, Poli M. Iron, Oxidative Damage and Ferroptosis in Rhabdomyosarcoma. Int J Mol Sci 2017; 18:ijms18081718. [PMID: 28783123 PMCID: PMC5578108 DOI: 10.3390/ijms18081718] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 08/02/2017] [Accepted: 08/03/2017] [Indexed: 12/14/2022] Open
Abstract
Recent data have indicated a fundamental role of iron in mediating a non-apoptotic and non-necrotic oxidative form of programmed cell death termed ferroptosis that requires abundant cytosolic free labile iron to promote membrane lipid peroxidation. Different scavenger molecules and detoxifying enzymes, such as glutathione (GSH) and glutathione peroxidase 4 (GPX4), have been shown to overwhelm or exacerbate ferroptosis depending on their expression magnitude. Ferroptosis is emerging as a potential weapon against tumor growth since it has been shown to potentiate cell death in some malignancies. However, this mechanism has been poorly studied in Rhabdomyosarcoma (RMS), a myogenic tumor affecting childhood and adolescence. One of the main drivers of RMS genesis is the Retrovirus Associated DNA Sequences/Extracellular signal Regulated Kinases (RAS/ERK)signaling pathway, the deliberate activation of which correlates with tumor aggressiveness and oxidative stress levels. Since recent studies have indicated that treatment with oxidative inducers can significantly halt RMS tumor progression, in this review we covered different aspects, ranging from iron metabolism in carcinogenesis and tumor growth, to mechanisms of iron-mediated cell death, to highlight the potential role of ferroptosis in counteracting RMS growth.
Collapse
Affiliation(s)
- Alessandro Fanzani
- Department of Molecular and Translational Medicine (DMMT), University of Brescia, Viale Europa 11, 25123 Brescia, Italy.
| | - Maura Poli
- Department of Molecular and Translational Medicine (DMMT), University of Brescia, Viale Europa 11, 25123 Brescia, Italy.
| |
Collapse
|
17
|
Zhang J, Song N, Zang D, Yu J, Li J, Di W, Guo R, Zhao W, Wang H. c-Myc promotes tumor proliferation and anti‑apoptosis by repressing p21 in rhabdomyosarcomas. Mol Med Rep 2017; 16:4089-4094. [PMID: 28765944 DOI: 10.3892/mmr.2017.7101] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Accepted: 01/05/2017] [Indexed: 11/05/2022] Open
Abstract
v-myc avian myelocytomatosis viral oncogene homolog (c-Myc) is an important member protein of the Myc family that is important in cell cycle progression, apoptosis and tumorigenesis. In the present study, the role of c‑Myc in rhabdomyosarcoma (RMS) was assessed. Firstly, expression of endogenous c‑Myc and cyclin dependent kinase inhibitor 1A (p21) was examined in normal skeletal muscle, RMS specimens and TE671 RMS cells by immunohistochemistry, reverse transcription‑quantitative polymerase chain reaction and western blotting. Furthermore, cell cycle progression and apoptosis were assessed in TE671 RMS cells following treatment with a c‑Myc inhibitor, 10058‑F4. The results demonstrated that c‑Myc was overexpressed in clinical RMS tissues and TE671 cells, with the highest expression observed in the most RMS samples. Expression of p21 protein and apoptosis function were increased following treatment with 10058‑F4, but no difference was observed in cell cycle progression. In conclusion, the present study indicated that c‑Myc promotes RMS development by inhibiting apoptosis through repression of p21 transcription. Further studies will be required to evaluate c‑Myc as a target for RMS clinical treatment.
Collapse
Affiliation(s)
- Jinghang Zhang
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, P.R. China
| | - Na Song
- Department of Molecular Biology and Biochemistry, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, P.R. China
| | - Dan Zang
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, P.R. China
| | - Jian Yu
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, P.R. China
| | - Jinsong Li
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, P.R. China
| | - Wenyu Di
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, P.R. China
| | - Ruina Guo
- Department of Pathology, Puyang Oilfield General Hospital, Puyang 457000, P.R. China
| | - Weixing Zhao
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, P.R. China
| | - Haijun Wang
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, P.R. China
| |
Collapse
|
18
|
Farhan M, Wang H, Gaur U, Little PJ, Xu J, Zheng W. FOXO Signaling Pathways as Therapeutic Targets in Cancer. Int J Biol Sci 2017; 13:815-827. [PMID: 28808415 PMCID: PMC5555100 DOI: 10.7150/ijbs.20052] [Citation(s) in RCA: 364] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 04/11/2017] [Indexed: 12/11/2022] Open
Abstract
Many transcription factors play a key role in cellular differentiation and the delineation of cell phenotype. Transcription factors are regulated by phosphorylation, ubiquitination, acetylation/deacetylation and interactions between two or more proteins controlling multiple signaling pathways. These pathways regulate different physiological processes and pathological events, such as cancer and other diseases. The Forkhead box O (FOXO) is one subfamily of the fork head transcription factor family with important roles in cell fate decisions and this subfamily is also suggested to play a pivotal functional role as a tumor suppressor in a wide range of cancers. During apoptosis, FOXOs are involved in mitochondria-dependent and -independent processes triggering the expression of death receptor ligands like Fas ligand, TNF apoptosis ligand and Bcl‑XL, bNIP3, Bim from Bcl-2 family members. Different types of growth factors like insulin play a vital role in the regulation of FOXOs. The most important pathway interacting with FOXO in different types of cancers is the PI3K/AKT pathway. Some other important pathways such as the Ras-MEK-ERK, IKK and AMPK pathways are also associated with FOXOs in tumorigenesis. Therapeutically targeting the FOXO signaling pathway(s) could lead to the discovery and development of efficacious agents against some cancers, but this requires an enhanced understanding and knowledge of FOXO transcription factors and their regulation and functioning. This review focused on the current understanding of cell biology of FOXO transcription factors which relates to their potential role as targets for the treatment and prevention of human cancers. We also discuss drugs which are currently being used for cancer treatment along with their target pathways and also point out some potential drawbacks of those drugs, which further signifies the need for development of new drug strategies in the field of cancer treatment.
Collapse
Affiliation(s)
- Mohd Farhan
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Haitao Wang
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Uma Gaur
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Peter J Little
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Queensland, 4102 Australia and Xin Hua College, Sun Yat- Sen University, China
| | - Jiangping Xu
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Wenhua Zheng
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| |
Collapse
|
19
|
El Demellawy D, McGowan-Jordan J, de Nanassy J, Chernetsova E, Nasr A. Update on molecular findings in rhabdomyosarcoma. Pathology 2017; 49:238-246. [PMID: 28256213 DOI: 10.1016/j.pathol.2016.12.345] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 12/12/2016] [Accepted: 12/20/2016] [Indexed: 12/13/2022]
Abstract
Rhabdomyosarcoma (RMS) is the most common malignant soft tissue tumour in children and adolescents. Histologically RMS resembles developing fetal striated skeletal muscle. RMS is stratified into different histological subtypes which appear to influence management plans and patient outcome. Importantly, molecular classification of RMS seems to more accurately capture the true biology and clinical course and prognosis of RMS to guide therapeutic decisions. The identification of PAX-FOXO1 fusion status in RMS is one of the most important updates in the risk stratification of RMS. There are several genes close to PAX that are frequently altered including the RAS family, FGFR4, PIK3CA, CTNNB1, FBXW7, and BCOR. As with most paediatric blue round cell tumours and sarcomas, chemotherapy is the key regimen for RMS therapy. Currently there are no direct inhibitors against PAX-FOXO1 fusion oncoproteins and targeting epigenetic cofactors is limited to clinical trials. Failure of therapy in RMS is usually related to drug resistance and metastatic disease. Through this review we have highlighted most of the molecular aspects in RMS and have attempted to correlate with RMS classification, treatment and prognosis.
Collapse
Affiliation(s)
- Dina El Demellawy
- Faculty of Medicine, University of Ottawa, Ontario, Canada; Pediatric Pathology, Children's Hospital of Eastern Ontario, Ontario, Canada.
| | - Jean McGowan-Jordan
- Faculty of Medicine, University of Ottawa, Ontario, Canada; Genetics, Children's Hospital of Eastern Ontario, Ontario, Canada
| | - Joseph de Nanassy
- Faculty of Medicine, University of Ottawa, Ontario, Canada; Pediatric Pathology, Children's Hospital of Eastern Ontario, Ontario, Canada
| | | | - Ahmed Nasr
- Faculty of Medicine, University of Ottawa, Ontario, Canada; Pediatric Surgery, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| |
Collapse
|
20
|
Lacey A, Rodrigues-Hoffman A, Safe S. PAX3-FOXO1A Expression in Rhabdomyosarcoma Is Driven by the Targetable Nuclear Receptor NR4A1. Cancer Res 2017; 77:732-741. [PMID: 27864345 PMCID: PMC5290192 DOI: 10.1158/0008-5472.can-16-1546] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 10/14/2016] [Accepted: 10/21/2016] [Indexed: 12/24/2022]
Abstract
Alveolar rhabdomyosarcoma (ARMS) is a devastating pediatric disease driven by expression of the oncogenic fusion gene PAX3-FOXO1A. In this study, we report overexpression of the nuclear receptor NR4A1 in rhabdomyosarcomas that is sufficient to drive high expression of PAX3-FOXO1A there. RNAi-mediated silencing of NR4A1 decreased expression of PAX3-FOXO1A and its downstream effector genes. Similarly, cell treatment with the NR4A1 small-molecule antagonists 1,1-bis(3-indolyl)-1-(p-hydroxy or p-carbomethoxyphenyl)methane (C-DIM) decreased PAX3-FOXO1A. Mechanistic investigations revealed a requirement for the NR4A1/Sp4 complex to bind GC-rich promoter regions to elevate transcription of the PAX3-FOXO1A gene. In parallel, NR4A1 also regulated expression of β1-integrin, which with PAX3-FOXO1A, contributed to tumor cell migration that was blocked by C-DIM/NR4A1 antagonists. Taken together, our results provide a preclinical rationale for the use of NR4A1 small-molecule antagonists to treat ARMS and other rhabdomyosarcomas driven by PAX3-FOXO1A. Cancer Res; 77(3); 732-41. ©2016 AACR.
Collapse
Affiliation(s)
- Alexandra Lacey
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas
| | | | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas.
| |
Collapse
|
21
|
Missiaglia E, Shepherd CJ, Aladowicz E, Olmos D, Selfe J, Pierron G, Delattre O, Walters Z, Shipley J. MicroRNA and gene co-expression networks characterize biological and clinical behavior of rhabdomyosarcomas. Cancer Lett 2016; 385:251-260. [PMID: 27984116 PMCID: PMC5157784 DOI: 10.1016/j.canlet.2016.10.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 10/02/2016] [Accepted: 10/03/2016] [Indexed: 11/29/2022]
Abstract
Rhabdomyosarcomas (RMS) in children and adolescents are heterogeneous sarcomas broadly defined by skeletal muscle features and the presence/absence of PAX3/7-FOXO1 fusion genes. MicroRNAs are small non-coding RNAs that regulate gene expression in a cell context specific manner. Sequencing analyses of microRNAs in 64 RMS revealed expression patterns separating skeletal muscle, fusion gene positive and negative RMS. Integration with parallel gene expression data assigned biological functions to 12 co-expression networks/modules that reassuringly included myogenic roles strongly correlated with microRNAs known in myogenesis and RMS development. Modules also correlated with clinical outcome and fusion status. Regulation of microRNAs by the fusion protein was demonstrated after PAX3-FOXO1 reduction, exemplified by miR-9-5p. MiR-9-5p levels correlated with poor outcome, even within fusion gene positive RMS, and were higher in metastatic versus non-metastatic disease. MiR-9-5p reduction inhibited RMS cell migration. Our findings reveal microRNAs in a regulatory framework of biological and clinical significance in RMS. RNAseq profiled miRNA expression in 64 rhabdomyosarcomas (RMS). MiRNA expression distinguished muscle and RMS on the basis of fusion gene status. Co-expression networks linked to function, clinical data and fusion gene status. Identified miRNAs, including miR-9-5p, altered by the PAX3-FOXO1 fusion protein. Demonstrated clinical and functional role for miR-9-5p in RMS.
Collapse
Affiliation(s)
- Edoardo Missiaglia
- Sarcoma Molecular Pathology Team, Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, Surrey, SM2 5NG, UK
| | - Chris J Shepherd
- Sarcoma Molecular Pathology Team, Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, Surrey, SM2 5NG, UK
| | - Ewa Aladowicz
- Sarcoma Molecular Pathology Team, Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, Surrey, SM2 5NG, UK
| | - David Olmos
- Sarcoma Molecular Pathology Team, Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, Surrey, SM2 5NG, UK
| | - Joanna Selfe
- Sarcoma Molecular Pathology Team, Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, Surrey, SM2 5NG, UK
| | - Gaëlle Pierron
- Unité de Génétique Somatique, Institut Curie, 26 Rue d'Ulm, 75248, Paris Cedex 05, France
| | - Olivier Delattre
- Unité de Génétique Somatique, Institut Curie, 26 Rue d'Ulm, 75248, Paris Cedex 05, France
| | - Zoe Walters
- Sarcoma Molecular Pathology Team, Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, Surrey, SM2 5NG, UK
| | - Janet Shipley
- Sarcoma Molecular Pathology Team, Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, Surrey, SM2 5NG, UK.
| |
Collapse
|
22
|
Abstract
Rhabdomyosarcoma (RMS) is a myogenic tumor classified as the most frequent soft tissue sarcoma affecting children and adolescents. The histopathological classification includes 5 different histotypes, with 2 most predominant referred as to embryonal and alveolar, the latter being characterized by adverse outcome. The current molecular classification identifies 2 major subsets, those harboring the fused Pax3-Foxo1 transcription factor generating from a recurrent specific translocation (fusion-positive RMS), and those lacking this signature but harboring mutations in the RAS/PI3K/AKT signaling axis (fusion-negative RMS). Since little attention has been devoted to RMS metabolism until now, in this review we summarize the "state of art" of metabolism and discuss how some of the molecular signatures found in this cancer, as observed in other more common tumors, can predict important metabolic challenges underlying continuous cell growth, oxidative stress resistance and metastasis, which could be the subject of future targeted therapies.
Collapse
Affiliation(s)
- Eugenio Monti
- a Department of Molecular and Translational Medicine , University of Brescia , Brescia , Italy
| | - Alessandro Fanzani
- a Department of Molecular and Translational Medicine , University of Brescia , Brescia , Italy.,b Interuniversity Institute of Myology , Rome , Italy
| |
Collapse
|
23
|
Böhm M, Wachtel M, Marques JG, Streiff N, Laubscher D, Nanni P, Mamchaoui K, Santoro R, Schäfer BW. Helicase CHD4 is an epigenetic coregulator of PAX3-FOXO1 in alveolar rhabdomyosarcoma. J Clin Invest 2016; 126:4237-4249. [PMID: 27760049 DOI: 10.1172/jci85057] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 09/08/2016] [Indexed: 12/19/2022] Open
Abstract
A vast number of cancer genes are transcription factors that drive tumorigenesis as oncogenic fusion proteins. Although the direct targeting of transcription factors remains challenging, therapies aimed at oncogenic fusion proteins are attractive as potential treatments for cancer. There is particular interest in targeting the oncogenic PAX3-FOXO1 fusion transcription factor, which induces alveolar rhabdomyosarcoma (aRMS), an aggressive cancer of skeletal muscle cells for which patient outcomes remain dismal. In this work, we have defined the interactome of PAX3-FOXO1 and screened 60 candidate interactors using siRNA-mediated depletion to identify candidates that affect fusion protein activity in aRMS cells. We report that chromodomain helicase DNA binding protein 4 (CHD4), an ATP-dependent chromatin remodeler, acts as crucial coregulator of PAX3-FOXO1 activity. CHD4 interacts with PAX3-FOXO1 via short DNA fragments. Together, they bind to regulatory regions of PAX3-FOXO1 target genes. Gene expression analysis suggested that CHD4 coregulatory activity is essential for a subset of PAX3-FOXO1 target genes. Depletion of CHD4 reduced cell viability of fusion-positive but not of fusion-negative RMS in vitro, which resembled loss of PAX3-FOXO1. It also caused specific regression of fusion-positive xenograft tumors in vivo. Therefore, this work identifies CHD4 as an epigenetic coregulator of PAX3-FOXO1 activity, providing rational evidence for CHD4 as a potential therapeutic target in aRMS.
Collapse
MESH Headings
- Animals
- Autoantigens/genetics
- Autoantigens/metabolism
- Cell Line, Tumor
- Epigenesis, Genetic
- Female
- Gene Expression Regulation, Neoplastic
- Heterografts
- Humans
- Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics
- Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism
- Mice, Inbred NOD
- Mice, SCID
- Neoplasm Transplantation
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/metabolism
- Paired Box Transcription Factors/genetics
- Paired Box Transcription Factors/metabolism
- Rhabdomyosarcoma, Alveolar/genetics
- Rhabdomyosarcoma, Alveolar/metabolism
- Rhabdomyosarcoma, Alveolar/pathology
Collapse
|
24
|
Bharathy N, Suriyamurthy S, Rao VK, Ow JR, Lim HJ, Chakraborty P, Vasudevan M, Dhamne CA, Chang KTE, Min VLK, Kundu TK, Taneja R. P/CAF mediates PAX3-FOXO1-dependent oncogenesis in alveolar rhabdomyosarcoma. J Pathol 2016; 240:269-281. [PMID: 27453350 DOI: 10.1002/path.4773] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 07/19/2016] [Accepted: 07/21/2016] [Indexed: 12/29/2022]
Abstract
Alveolar rhabdomyosarcoma (ARMS) is an aggressive paediatric cancer of skeletal muscle with poor prognosis. A PAX3-FOXO1 fusion protein acts as a driver of malignancy in ARMS by disrupting tightly coupled but mutually exclusive pathways of proliferation and differentiation. While PAX3-FOXO1 is an attractive therapeutic target, no current treatments are designed to block its oncogenic activity. The present work shows that the histone acetyltransferase P/CAF (KAT2B) is overexpressed in primary tumours from ARMS patients. Interestingly, in fusion-positive ARMS cell lines, P/CAF acetylates and stabilizes PAX3-FOXO1 rather than MyoD, a master regulator of muscle differentiation. Silencing P/CAF, or pharmacological inhibition of its acetyltransferase activity, down-regulates PAX3-FOXO1 levels concomitant with reduced proliferation and tumour burden in xenograft mouse models. Our studies identify a P/CAF-PAX3-FOXO1 signalling node that promotes oncogenesis and may contribute to MyoD dysfunction in ARMS. This work exemplifies the therapeutic potential of targeting chromatin-modifying enzymes to inhibit fusion oncoproteins that are a frequent event in sarcomas. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Narendra Bharathy
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Sudha Suriyamurthy
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Vinay Kumar Rao
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Jin Rong Ow
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Huey Jin Lim
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Payal Chakraborty
- Bionivid Technology Pvt Ltd, 401-4 AB Cross, 1st Main, Kasturi Nagar, Bangalore, India
| | - Madavan Vasudevan
- Bionivid Technology Pvt Ltd, 401-4 AB Cross, 1st Main, Kasturi Nagar, Bangalore, India
| | | | | | - Victor Lee Kwan Min
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Tapas K Kundu
- Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India
| | - Reshma Taneja
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
25
|
Coomans de Brachène A, Demoulin JB. FOXO transcription factors in cancer development and therapy. Cell Mol Life Sci 2016; 73:1159-72. [PMID: 26686861 PMCID: PMC11108379 DOI: 10.1007/s00018-015-2112-y] [Citation(s) in RCA: 220] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 11/30/2015] [Accepted: 12/07/2015] [Indexed: 12/19/2022]
Abstract
The forkhead box O (FOXO) transcription factors are considered as tumor suppressors that limit cell proliferation and induce apoptosis. FOXO gene alterations have been described in a limited number of human cancers, such as rhabdomyosarcoma, leukemia and lymphoma. In addition, FOXO proteins are inactivated by major oncogenic signals such as the phosphatidylinositol-3 kinase pathway and MAP kinases. Their expression is also repressed by micro-RNAs in multiple cancer types. FOXOs are mediators of the tumor response to various therapies. However, paradoxical roles of FOXOs in cancer progression were recently described. FOXOs contribute to the maintenance of leukemia-initiating cells in acute and chronic myeloid leukemia. These factors may also promote invasion and metastasis of subsets of colon and breast cancers. Resistance to treatment was also ascribed to FOXO activation in multiple cases, including targeted therapies. In this review, we discuss the complex role of FOXOs in cancer development and response to therapy.
Collapse
Affiliation(s)
- Alexandra Coomans de Brachène
- de Duve Institute, MEXP-UCL 74.30, Université catholique de Louvain, Avenue Hippocrate 75, B1.74.05, 1200, Brussels, Belgium
| | - Jean-Baptiste Demoulin
- de Duve Institute, MEXP-UCL 74.30, Université catholique de Louvain, Avenue Hippocrate 75, B1.74.05, 1200, Brussels, Belgium.
| |
Collapse
|
26
|
Harel M, Ferrer FA, Shapiro LH, Makari JH. Future directions in risk stratification and therapy for advanced pediatric genitourinary rhabdomyosarcoma. Urol Oncol 2016; 34:103-15. [DOI: 10.1016/j.urolonc.2015.09.013] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 09/19/2015] [Accepted: 09/22/2015] [Indexed: 11/17/2022]
|
27
|
Unpeaceful roles of mutant PAX proteins in cancer. Semin Cell Dev Biol 2015; 44:126-34. [DOI: 10.1016/j.semcdb.2015.09.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 09/10/2015] [Accepted: 09/16/2015] [Indexed: 01/07/2023]
|
28
|
Recurrent PAX3-MAML3 fusion in biphenotypic sinonasal sarcoma. Nat Genet 2014; 46:666-8. [PMID: 24859338 DOI: 10.1038/ng.2989] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Accepted: 04/30/2014] [Indexed: 12/27/2022]
Abstract
Biphenotypic sinonasal sarcoma (SNS) is a newly described tumor of the nasal and paranasal areas. Here we report a recurrent chromosomal translocation in SNS, t(2;4)(q35;q31.1), resulting in a PAX3-MAML3 fusion protein that is a potent transcriptional activator of PAX3 response elements. The SNS phenotype is characterized by aberrant expression of genes involved in neuroectodermal and myogenic differentiation, closely simulating the developmental roles of PAX3.
Collapse
|
29
|
Sarcomas. Mol Oncol 2013. [DOI: 10.1017/cbo9781139046947.068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
30
|
Hipp NI, Christner L, Wirth T, Mueller-Klieser W, Walenta S, Schröck E, Debatin KM, Beltinger C. MYCN and survivin cooperatively contribute to malignant transformation of fibroblasts. Carcinogenesis 2013; 35:479-88. [PMID: 24130166 DOI: 10.1093/carcin/bgt341] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The oncogenes MYCN and survivin (BIRC5) maintain aggressiveness of diverse cancers including sarcomas. To investigate whether these oncogenes cooperate in initial malignant transformation, we transduced them into Rat-1 fibroblasts. Indeed, survivin enhanced MYCN-driven contact-uninhibited and anchorage-independent growth in vitro. Importantly, upon subcutaneous transplantation into mice, cells overexpressing both instead of either one of the oncogenes generated tumors with shortened latency, marked anaplasia and an increased proliferation-to-apoptosis ratio resulting in accelerated growth. Mechanistically, the increased tumorigenicity was associated with an enhanced Warburg effect and a hypoxia inducible factor 1α linked vascular remodeling. This cooperation between MYCN and survivin may be important in the genesis of several cancers.
Collapse
Affiliation(s)
- Nora I Hipp
- Department of Paediatrics and Adolescent Medicine, University Medical Centre Ulm, Ulm89075, Germany
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Keller C, Guttridge DC. Mechanisms of impaired differentiation in rhabdomyosarcoma. FEBS J 2013; 280:4323-34. [PMID: 23822136 DOI: 10.1111/febs.12421] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Revised: 06/17/2013] [Accepted: 07/01/2013] [Indexed: 12/22/2022]
Abstract
Rhabdomyosarcoma (RMS) is the most common soft tissue sarcoma of childhood, with presumed skeletal muscle origins, because of its myogenic phenotype. RMS is composed of two main subtypes, embryonal RMS (eRMS) and alveolar RMS (aRMS). Whereas eRMS histologically resembles embryonic skeletal muscle, the aRMS subtype is more aggressive and has a poorer prognosis. In addition, whereas the genetic profile of eRMS is not well established, aRMS is commonly associated with distinct chromosome translocations that fuse domains of the transcription factors Pax3 and Pax7 to the forkhead family member FOXO1A. Both eRMS and aRMS tumor cells express myogenic markers such as MyoD, but their ability to complete differentiation is impaired. How this impairment occurs is the subject of this review, which will focus on several themes, including signaling pathways that converge on Pax-forkhead gene targets, alterations in MyoD function, epigenetic modifications of myogenic promoters, and microRNAs whose expression patterns in RMS alter key regulatory circuits to help maintain tumor cells in an opportunistically less differentiated state.
Collapse
Affiliation(s)
- Charles Keller
- Pediatric Cancer Biology Program, Papé Family Pediatric Research Institute, Department of Pediatrics, Oregon Health & Science University, Portland, OR, USA
| | | |
Collapse
|
32
|
Yamamoto Y, Fukuda K, Fuchimoto Y, Matsuzaki Y, Saikawa Y, Kitagawa Y, Morikawa Y, Kuroda T. Cetuximab promotes anticancer drug toxicity in rhabdomyosarcomas with EGFR amplification in vitro. Oncol Rep 2013; 30:1081-6. [PMID: 23828214 DOI: 10.3892/or.2013.2588] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 04/02/2013] [Indexed: 11/06/2022] Open
Abstract
Overexpression of human epidermal growth factor receptor (EGFR) has been detected in various tumors and is associated with poor outcomes. Combination treatment regimens with EGFR-targeting and cytotoxic agents are a potential therapeutic option for rhabdomyosarcoma (RMS) with EGFR amplification. We investigated the effects of combination treatment with actinomycin D and the EGFR-targeting agent cetuximab in 4 RMS cell lines. All 4 RMS cell lines expressed wild-type K-ras, and 2 of the 4 overexpressed EGFR, as determined by flow cytometry, real-time PCR and direct sequencing. Combination of cetuximab and actinomycin D was highly effective, synergistically inhibiting cell growth with a combination index of less than 1. Moreover, combination treatment with these two reagents increased the rate of apoptosis in EGFR-positive cells. Cetuximab has antitumor activity in EGFR-amplified RMS cells when combined with antitumor reagents, indicating that cetuximab is a potential therapeutic reagent for RMS with EGFR amplification.
Collapse
Affiliation(s)
- Yuki Yamamoto
- Department of Pediatric Surgery, Keio University School of Medicine, Tokyo 160-858, Japan
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Ahn EH, Mercado GE, Laé M, Ladanyi M. Identification of target genes of PAX3-FOXO1 in alveolar rhabdomyosarcoma. Oncol Rep 2013; 30:968-78. [PMID: 23733015 PMCID: PMC3776721 DOI: 10.3892/or.2013.2513] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Accepted: 03/28/2013] [Indexed: 01/07/2023] Open
Abstract
Rhabdomyosarcoma (RMS) is a soft tissue sarcoma categorized into two major subtypes: alveolar RMS (ARMS) and embryonal RMS (ERMS). Most ARMS express the PAX3-FOXO1 (P3F) fusion oncoprotein generated by the 2;13 chromosomal translocation. In the present study, the downstream target genes of P3F were identified by analyzing two independent sets of gene expression profiles: primary RMS tumors and RD ERMS cells transduced with inducible P3F constructs. We found 34 potential target genes (27 upregulated and 7 downregulated) that were significantly and differentially expressed between P3F-positive and P3F-negative categories, both in primary RMS tumors and in the inducible P3F cell culture system. Gene ontology analysis of microarray data of the inducible P3F cell culture system employed indicated apoptosis, cell death, development, and signal transduction as overrepresented significant functional categories found in both upregulated and downregulated genes. Therefore, among the 34 potential target genes, the expression of cell death-related [Gremlin1, cysteine knot superfamily 1, BMP antagonist 1 (GREM1) and death-associated protein kinase 1 (DAPK1)] and development-related [myogenic differentiation 1 (MYOD1) and hairy/enhancer-of-split related with YRPW motif 1 (HEY1)] genes were further investigated. The differential expression of GREM1, DAPK1, MYOD1 and HEY1 was confirmed in independent tumors and inducible cell culture systems. The expression of GREM1, DAPK1 and MYOD1 were significantly upregulated; HEY1 was significantly downregulated in independent P3F-positive ARMS tumors and transcriptionally active P3F cells, compared to those in ERMS tumors and transcriptionally inactive P3F cells. This study identified target genes of P3F and suggested that four downstream targets (GREM1, DAPK1, MYOD1 and HEY1) can contribute to the biological activities of P3F involved in growth suppression or cell death and myogenic differentiation.
Collapse
Affiliation(s)
- Eun Hyun Ahn
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA.
| | | | | | | |
Collapse
|
34
|
Olanich ME, Barr FG. A call to ARMS: targeting the PAX3-FOXO1 gene in alveolar rhabdomyosarcoma. Expert Opin Ther Targets 2013; 17:607-23. [PMID: 23432728 DOI: 10.1517/14728222.2013.772136] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Expression of fusion oncoproteins generated by recurrent chromosomal translocations represents a major tumorigenic mechanism characteristic of multiple cancers, including one-third of all sarcomas. Oncogenic fusion genes provide novel targets for therapeutic intervention. The PAX3-FOXO1 oncoprotein in alveolar rhabdomyosarcoma (ARMS) is presented as a paradigm to examine therapeutic strategies for targeting sarcoma-associated fusion genes. AREAS COVERED This review discusses the role of PAX3-FOXO1 in ARMS tumors. Besides evaluating various approaches to molecularly target PAX3-FOXO1 itself, this review highlights therapeutically attractive downstream genes activated by PAX3-FOXO1. EXPERT OPINION Oncogenic fusion proteins represent desirable therapeutic targets because their expression is specific to tumor cells, but these fusions generally characterize rare malignancies. Full development and testing of potential drugs targeted to these fusions are complicated by the small numbers of patients in these disease categories. Although efforts to develop targeted therapies against fusion proteins should continue, molecular targets that are applicable to a broader tumor landscape should be pursued. A shift of the traditional paradigm to view therapeutic intervention as target-specific rather than tumor-specific will help to circumvent the challenges posed by rare tumors and maximize the possibility of developing successful new treatments for patients with these rare translocation-associated sarcomas.
Collapse
Affiliation(s)
- Mary E Olanich
- National Institutes of Health, National Cancer Institute, Center for Cancer Research, Laboratory of Pathology , Bethesda, MD 20892, USA
| | | |
Collapse
|
35
|
Marshall AD, Grosveld GC. Alveolar rhabdomyosarcoma - The molecular drivers of PAX3/7-FOXO1-induced tumorigenesis. Skelet Muscle 2012. [PMID: 23206814 PMCID: PMC3564712 DOI: 10.1186/2044-5040-2-25] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Rhabdomyosarcoma is a soft tissue sarcoma arising from cells of a mesenchymal or skeletal muscle lineage. Alveolar rhabdomyosarcoma (ARMS) is more aggressive than the more common embryonal (ERMS) subtype. ARMS is more prone to metastasis and carries a poorer prognosis. In contrast to ERMS, the majority of ARMS tumors carry one of several characteristic chromosomal translocations, such as t(2;13)(q35;q14), which results in the expression of a PAX3-FOXO1 fusion transcription factor. In this review we discuss the genes that cooperate with PAX3-FOXO1, as well as the target genes of the fusion transcription factor that contribute to various aspects of ARMS tumorigenesis. The characterization of these pathways will lead to a better understanding of ARMS tumorigenesis and will allow the design of novel targeted therapies that will lead to better treatment for this aggressive pediatric tumor.
Collapse
Affiliation(s)
- Amy D Marshall
- Department of Genetics, St Jude Children's Research Hospital, Memphis, TN, 38105, USA.
| | | |
Collapse
|
36
|
Davicioni E, Wai DH, Anderson MJ. Diagnostic and Prognostic Sarcoma Signatures. Mol Diagn Ther 2012; 12:359-74. [DOI: 10.1007/bf03256302] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
37
|
Abstract
Soft tissue sarcomas are an uncommon and diverse group of more than 50 mesenchymal malignancies. The pathogenesis of many of these is poorly understood, but others have begun to reveal the secrets of their underlying mechanisms. With considerable effort over recent years, soft tissue sarcomas have increasingly been classified on the basis of underlying molecular alterations. In turn, this has allowed the development and application of targeted agents in several specific, molecularly defined, sarcoma subtypes. This review will focus on the rationale for targeted therapy in sarcoma, with emphasis on the relevance of specific molecular factors and pathways in both translocation-associated sarcomas and in genetically complex tumors. In addition, we will address some of the early successes in sarcoma-targeted therapy as well as a few challenges and disappointments in this field. Finally, we will discuss several possible opportunities represented by poorly understood, but potentially promising new therapeutic targets, as well as several novel biological agents currently in preclinical and early phase I/II trials. This will provide the reader with the context for understanding the current state of this field and a sense of where it may be headed in the coming years.
Collapse
Affiliation(s)
- Elizabeth G Demicco
- Department of Pathology, The University of Texas M.D. Anderson Cancer Center, Houston, 77030-4009, USA
| | | | | | | |
Collapse
|
38
|
Ray A, Huh WW. Current State-of-the-Art Systemic Therapy for Pediatric Soft Tissue Sarcomas. Curr Oncol Rep 2012; 14:311-9. [DOI: 10.1007/s11912-012-0243-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
39
|
Abstract
To address the poor outcomes in rhabdomyosarcoma, particularly the alveolar subtype, new therapies are needed. Potential cancer-specific alterations that may be molecular targets include gene fusions or copy number changes. Following the latter strategy, an attractive antigene approach was developed to inhibit MYCN oncogene expression in rhabdomyosarcoma.
Collapse
Affiliation(s)
- Frederic G Barr
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD 20892
| |
Collapse
|
40
|
van Gaal JC, Flucke UE, Roeffen MH, de Bont ES, Sleijfer S, Mavinkurve-Groothuis AM, Suurmeijer AJ, van der Graaf WT, Versleijen-Jonkers YM. Anaplastic Lymphoma Kinase Aberrations in Rhabdomyosarcoma: Clinical and Prognostic Implications. J Clin Oncol 2012; 30:308-15. [DOI: 10.1200/jco.2011.37.8588] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Purpose The aim of this study is to investigate anaplastic lymphoma kinase (ALK) protein expression and underlying genetic aberrations in rhabdomyosarcoma (RMS), with special attention to clinical and prognostic implications. Patients and Methods A total of 189 paraffin-embedded RMS tumor specimens from 145 patients were collected on tissue microarray. ALK protein expression was evaluated by immunohistochemistry. ALK gene (2p23) copy number and translocations were determined by in situ hybridization. cDNA sequencing of the receptor tyrosine kinase domain of the ALK gene was assessed in 43 samples. Results Strong cytoplasmic ALK protein expression was more frequently observed in alveolar RMS (ARMS) than in embryonal RMS (ERMS) (81% v 32%, respectively; P < .001). ALK gene copy number gain was detected in the vast majority of ARMS (88%), compared with 52% of ERMS (P < .001). ALK copy number correlated with protein expression in primary tumors (n = 107). We identified one point mutation (2%) and seven tumors harboring whole exon deletions (16%). In ERMS, specific ALK gain in the primary tumor correlated with metastatic disease (100% in metastatic disease v 29% in nonmetastatic disease; P = .004) and poor disease-specific survival (5-year disease-specific survival: 62% v 82% for nonspecific or no gain; P = .046). Conclusion Because ALK aberrations on genomic and protein levels are frequently found in RMSs, in particular ARMS, and are associated with disease progression and outcome in ERMS, ALK may play a role in tumor biology and may provide a potential therapeutic target for these tumors. Future research should aim at the oncogenic role of ALK and the potential effect of ALK inhibitors in RMS.
Collapse
Affiliation(s)
- J. Carlijn van Gaal
- J. Carlijn van Gaal, Uta E. Flucke, Melissa H.S. Roeffen, Annelies M.C. Mavinkurve-Groothuis, Winette T.A. van der Graaf, and Yvonne M.H. Versleijen-Jonkers, Radboud University Nijmegen Medical Centre, Nijmegen; Eveline S.J.M. de Bont, Beatrix Children's Hospital, University Medical Center Groningen; Albert J.H. Suurmeijer, University Medical Center Groningen, Groningen; and Stefan Sleijfer, Daniel den Hoed Cancer Center, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Uta E. Flucke
- J. Carlijn van Gaal, Uta E. Flucke, Melissa H.S. Roeffen, Annelies M.C. Mavinkurve-Groothuis, Winette T.A. van der Graaf, and Yvonne M.H. Versleijen-Jonkers, Radboud University Nijmegen Medical Centre, Nijmegen; Eveline S.J.M. de Bont, Beatrix Children's Hospital, University Medical Center Groningen; Albert J.H. Suurmeijer, University Medical Center Groningen, Groningen; and Stefan Sleijfer, Daniel den Hoed Cancer Center, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Melissa H.S. Roeffen
- J. Carlijn van Gaal, Uta E. Flucke, Melissa H.S. Roeffen, Annelies M.C. Mavinkurve-Groothuis, Winette T.A. van der Graaf, and Yvonne M.H. Versleijen-Jonkers, Radboud University Nijmegen Medical Centre, Nijmegen; Eveline S.J.M. de Bont, Beatrix Children's Hospital, University Medical Center Groningen; Albert J.H. Suurmeijer, University Medical Center Groningen, Groningen; and Stefan Sleijfer, Daniel den Hoed Cancer Center, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Eveline S.J.M. de Bont
- J. Carlijn van Gaal, Uta E. Flucke, Melissa H.S. Roeffen, Annelies M.C. Mavinkurve-Groothuis, Winette T.A. van der Graaf, and Yvonne M.H. Versleijen-Jonkers, Radboud University Nijmegen Medical Centre, Nijmegen; Eveline S.J.M. de Bont, Beatrix Children's Hospital, University Medical Center Groningen; Albert J.H. Suurmeijer, University Medical Center Groningen, Groningen; and Stefan Sleijfer, Daniel den Hoed Cancer Center, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Stefan Sleijfer
- J. Carlijn van Gaal, Uta E. Flucke, Melissa H.S. Roeffen, Annelies M.C. Mavinkurve-Groothuis, Winette T.A. van der Graaf, and Yvonne M.H. Versleijen-Jonkers, Radboud University Nijmegen Medical Centre, Nijmegen; Eveline S.J.M. de Bont, Beatrix Children's Hospital, University Medical Center Groningen; Albert J.H. Suurmeijer, University Medical Center Groningen, Groningen; and Stefan Sleijfer, Daniel den Hoed Cancer Center, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Annelies M.C. Mavinkurve-Groothuis
- J. Carlijn van Gaal, Uta E. Flucke, Melissa H.S. Roeffen, Annelies M.C. Mavinkurve-Groothuis, Winette T.A. van der Graaf, and Yvonne M.H. Versleijen-Jonkers, Radboud University Nijmegen Medical Centre, Nijmegen; Eveline S.J.M. de Bont, Beatrix Children's Hospital, University Medical Center Groningen; Albert J.H. Suurmeijer, University Medical Center Groningen, Groningen; and Stefan Sleijfer, Daniel den Hoed Cancer Center, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Albert J.H. Suurmeijer
- J. Carlijn van Gaal, Uta E. Flucke, Melissa H.S. Roeffen, Annelies M.C. Mavinkurve-Groothuis, Winette T.A. van der Graaf, and Yvonne M.H. Versleijen-Jonkers, Radboud University Nijmegen Medical Centre, Nijmegen; Eveline S.J.M. de Bont, Beatrix Children's Hospital, University Medical Center Groningen; Albert J.H. Suurmeijer, University Medical Center Groningen, Groningen; and Stefan Sleijfer, Daniel den Hoed Cancer Center, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Winette T.A. van der Graaf
- J. Carlijn van Gaal, Uta E. Flucke, Melissa H.S. Roeffen, Annelies M.C. Mavinkurve-Groothuis, Winette T.A. van der Graaf, and Yvonne M.H. Versleijen-Jonkers, Radboud University Nijmegen Medical Centre, Nijmegen; Eveline S.J.M. de Bont, Beatrix Children's Hospital, University Medical Center Groningen; Albert J.H. Suurmeijer, University Medical Center Groningen, Groningen; and Stefan Sleijfer, Daniel den Hoed Cancer Center, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Yvonne M.H. Versleijen-Jonkers
- J. Carlijn van Gaal, Uta E. Flucke, Melissa H.S. Roeffen, Annelies M.C. Mavinkurve-Groothuis, Winette T.A. van der Graaf, and Yvonne M.H. Versleijen-Jonkers, Radboud University Nijmegen Medical Centre, Nijmegen; Eveline S.J.M. de Bont, Beatrix Children's Hospital, University Medical Center Groningen; Albert J.H. Suurmeijer, University Medical Center Groningen, Groningen; and Stefan Sleijfer, Daniel den Hoed Cancer Center, Erasmus Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
41
|
Tonelli R, McIntyre A, Camerin C, Walters ZS, Di Leo K, Selfe J, Purgato S, Missiaglia E, Tortori A, Renshaw J, Astolfi A, Taylor KR, Serravalle S, Bishop R, Nanni C, Valentijn LJ, Faccini A, Leuschner I, Formica S, Reis-Filho JS, Ambrosini V, Thway K, Franzoni M, Summersgill B, Marchelli R, Hrelia P, Cantelli-Forti G, Fanti S, Corradini R, Pession A, Shipley J. Antitumor activity of sustained N-myc reduction in rhabdomyosarcomas and transcriptional block by antigene therapy. Clin Cancer Res 2011; 18:796-807. [PMID: 22065083 DOI: 10.1158/1078-0432.ccr-11-1981] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Rhabdomyosarcomas are a major cause of cancer death in children, described with MYCN amplification and, in the alveolar subtype, transcription driven by the PAX3-FOXO1 fusion protein. Our aim was to determine the prevalence of N-Myc protein expression and the potential therapeutic effects of reducing expression in rhabdomyosarcomas, including use of an antigene strategy that inhibits transcription. EXPERIMENTAL DESIGN Protein expression was assessed by immunohistochemistry. MYCN expression was reduced in representative cell lines by RNA interference and an antigene peptide nucleic acid (PNA) oligonucleotide conjugated to a nuclear localization signal peptide. Associated gene expression changes, cell viability, and apoptosis were analyzed in vitro. As a paradigm for antigene therapy, the effects of systemic treatment of mice with rhabdomyosarcoma cell line xenografts were determined. RESULTS High N-Myc levels were significantly associated with genomic amplification, presence of the PAX3/7-FOXO1 fusion genes, and proliferative capacity. Sustained reduction of N-Myc levels in all rhabdomyosarcoma cell lines that express the protein decreased cell proliferation and increased apoptosis. Positive feedback was shown to regulate PAX3-FOXO1 and N-Myc levels in the alveolar subtype that critically decrease PAX3-FOXO1 levels on reducing N-Myc. Pharmacologic systemic administration of the antigene PNA can eliminate alveolar rhabdomyosarcoma xenografts in mice, without relapse or toxicity. CONCLUSION N-Myc, with its restricted expression in non-fetal tissues, is a therapeutic target to treat rhabdomyosarcomas, and blocking gene transcription using antigene oligonucleotide strategies has therapeutic potential in the treatment of cancer and other diseases that has not been previously realized in vivo.
Collapse
Affiliation(s)
- Roberto Tonelli
- Department of Pediatric Hematology, University of Bologna, Bologna, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
Rhabdomyosarcoma (RMS) is a morphologically and clinically heterogeneous group of malignant tumors that resemble developing skeletal muscle and is the most common soft-tissue sarcoma in children and adolescents. The most prominent sites involve head and neck structures (~40%), genito-urinary track (~25%), and extremities (~20%). Embryonal (ERMS) and alveolar (ARMS) are the two major RMS subtypes that are distinct in their morphology and genetic make-up. The prognosis for this cancer depends strongly on tumor size, location, staging, and child's age. In general, ERMS has a more favorable outcome, whereas the mortality rate remains high in patients with ARMS, because of its aggressive and metastatic nature. Over the past two decades, researchers have made concerted efforts to delineate genetic and epigenetic changes associated with RMS pathogenesis. These molecular signatures have presented golden opportunities to design targeted therapies for treating this aggressive cancer. This article highlights recent advances in understanding the molecular pathogenesis of RMS, and addresses promising research areas for further exploration.
Collapse
Affiliation(s)
- C Wang
- Department of Oral Biology and Center for Molecular Biology of Oral Diseases, University of Illinois at Chicago, 801 South Paulina Street, RM530CB, m/c 860, Chicago, IL 60612, USA.
| |
Collapse
|
43
|
Rengaswamy V, Kontny U, Rössler J. New approaches for pediatric rhabdomyosarcoma drug discovery: targeting combinatorial signaling. Expert Opin Drug Discov 2011; 6:1103-25. [PMID: 22646865 DOI: 10.1517/17460441.2011.611498] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Rhabdomyosarcomas (RMS) are rare heterogeneous pediatric tumors that are treated by surgery, chemotherapy and irradiation. New therapeutic approaches are needed, especially in the advanced stages to target the pro-oncogenic signals. Exploring the molecular interactions of the regulatory signals and their roles in the developmental aspects of different subtypes of RMS is essential to identify potential targets and develop new therapeutic drugs. AREAS COVERED Insights into different drug discovery approaches are discussed with specific emphasis on gene expression profiling, fusion protein, role of small interfering RNA (siRNA)- and microRNA (miRNA)-based discovery approaches, targeting cancer stem cells, and in vitro and in vivo model systems. Targeting some overexpressed signals along with the possibilities of combination therapy of validated drug targets is discussed. Additionally, methods to overcome the limitations of discovery-based research are briefly discussed. EXPERT OPINION Due to drug resistance, ineffective therapy in advanced stages and relapse, there is a demand to explore new drug targets and discovery approaches. Implementing miRNA-based profiling would reveal the extent of miR-based regulation, various biomarkers and potential targets in RMS. A suitable combination of innovative techniques and the use of model systems might assist the identification and validation of novel targets and drug discovery methods. Combining specific drugs along with type-specific target inhibition of overexpressed mRNAs through siRNA approaches would enable the development of personalized therapy.
Collapse
Affiliation(s)
- Venkatesh Rengaswamy
- University Hospital Freiburg, Center for Pediatrics and Adolescent Medicine, Clinic IV: Pediatric Hematology and Oncology, Mathildenstr. 1, 79106 Freiburg , Germany +49 761 270 43000 ; +49 761 270 45180 ;
| | | | | |
Collapse
|
44
|
Taylor BS, Barretina J, Maki RG, Antonescu CR, Singer S, Ladanyi M. Advances in sarcoma genomics and new therapeutic targets. Nat Rev Cancer 2011; 11:541-57. [PMID: 21753790 PMCID: PMC3361898 DOI: 10.1038/nrc3087] [Citation(s) in RCA: 320] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Increasingly, human mesenchymal malignancies are being classified by the abnormalities that drive their pathogenesis. Although many of these aberrations are highly prevalent within particular sarcoma subtypes, few are currently targeted therapeutically. Indeed, most subtypes of sarcoma are still treated with traditional therapeutic modalities, and in many cases sarcomas are resistant to adjuvant therapies. In this Review, we discuss the core molecular determinants of sarcomagenesis and emphasize the emerging genomic and functional genetic approaches that, coupled with novel therapeutic strategies, have the potential to transform the care of patients with sarcoma.
Collapse
Affiliation(s)
- Barry S Taylor
- Memorial Sloan-Kettering Cancer Center, New York, New York 10065, USA
| | | | | | | | | | | |
Collapse
|
45
|
Lee MH, Jothi M, Gudkov AV, Mal AK. Histone methyltransferase KMT1A restrains entry of alveolar rhabdomyosarcoma cells into a myogenic differentiated state. Cancer Res 2011; 71:3921-31. [PMID: 21493592 PMCID: PMC3107367 DOI: 10.1158/0008-5472.can-10-3358] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Alveolar rhabdomyosarcoma (ARMS) is an aggressive pediatric muscle cancer, which arrested during the process of skeletal muscle differentiation. In muscle myoblast cells, ectopic expression of the histone H3 lysine 9 (H3K9) methytransferase KMT1A blocks differentiation by repressing a myogenic gene expression program. In this study, we tested the hypothesis that activation of a KMT1A-mediated program of transcriptional repression prevents ARMS cells from differentiating. We investigated whether KMT1A represses the expression of differentiation-associated genes in ARMS cells, thereby blocking muscle differentiation. Our results show that expression of KMT1A is induced in human ARMS cancer cell lines when cultured under differentiation-permissible conditions. shRNA-mediated knockdown of KMT1A decreased anchorage dependent and independent cell proliferation and tumor xenograft growth, increased expression of differentiation-associated genes, and promoted the appearance of a terminally differentiated-like phenotype. Finally, shRNA-directed KMT1A knockdown restored the impaired transcriptional activity of the myogenic regulator MyoD. Together, our results suggested that high levels of KMT1A in ARMS cells under differentiation conditions impairs MyoD function, thereby arresting myogenic differentiation in these tumor cells. Thus, targeting KMT1A may be a novel strategy for the treatment of this disease.
Collapse
MESH Headings
- Animals
- Cell Differentiation/physiology
- Cell Growth Processes/physiology
- Cell Line, Tumor
- Child
- Gene Expression Regulation, Neoplastic
- Gene Knockdown Techniques
- Histone Methyltransferases
- Histone-Lysine N-Methyltransferase/biosynthesis
- Histone-Lysine N-Methyltransferase/deficiency
- Histone-Lysine N-Methyltransferase/genetics
- Humans
- Mice
- MyoD Protein/metabolism
- Myogenin/genetics
- Promoter Regions, Genetic
- RNA, Small Interfering/administration & dosage
- RNA, Small Interfering/genetics
- Rhabdomyosarcoma, Alveolar/enzymology
- Rhabdomyosarcoma, Alveolar/genetics
- Rhabdomyosarcoma, Alveolar/pathology
- Transduction, Genetic
- Transplantation, Heterologous
Collapse
Affiliation(s)
- Min-Hyung Lee
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, New York
| | - Mathivanan Jothi
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, New York
| | - Andrei V. Gudkov
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, New York
| | - Asoke K. Mal
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, New York
| |
Collapse
|
46
|
Zhang Y, Wang C. Nephroblastoma overexpressed (NOV/CCN3) gene: a paired-domain-specific PAX3-FKHR transcription target that promotes survival and motility in alveolar rhabdomyosarcoma cells. Oncogene 2011; 30:3549-62. [PMID: 21423212 DOI: 10.1038/onc.2011.69] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The CCN (Cy61, CTGF and NOV) family of proteins is a group of matricellular biomolecules involved in both physiological and pathological processes. Elevated expression of the CCN3 (also known as NOV, Nephroblastoma overexpressed) gene has been detected in clinical samples of the skeletal muscle cancer rhabdomyosarcoma, with the highest expression found in the alveolar subtype (aRMS). Over 80% of aRMSs are characterized by a chromosomal translocation-derived fusion transcription factor PAX3-FKHR. In this study, we linked elevated CCN3 levels in aRMS cells to PAX3-FKHR expression. We found reduced CCN3 levels in aRMS cells following small interfering RNA knockdown of PAX3-FKHR, and increased CCN3 levels in C2 myoblasts following ectopic expression of PAX3-FKHR. Promoter, electrophoretic mobility shift assay and chromatin immunoprecipitation analyses confirmed that the CCN3 gene was a direct target for PAX3-FKHR transcriptional activation through a paired-domain DNA sequence in the first intron of the CCN3 gene. To determine the function of CCN3, we showed that knockdown and ectopic expression of CCN3 decreased survival and increased differentiation in aRMS cells, respectively. In addition, we found that exogenously supplied CCN3 protein promoted aRMS cell adhesion, migration and Matrigel invasion. Taken together, data from this study have (1) provided a mechanistic basis for the CCN3 overexpression in aRMS cells, and (2) identified CCN3 as an autocrine/paracrine factor that contributes to the aggressive behavior of aRMS cells, perhaps through a positive feedback loop. Thus, CCN3 may be an attractive target for therapeutic intervention in aRMS.
Collapse
Affiliation(s)
- Y Zhang
- Center for Molecular Biology of Oral Diseases, University of Illinois at Chicago, Chicago, IL 60612, USA
| | | |
Collapse
|
47
|
Abstract
Small cell malignancies of children constitute a continuing diagnostic challenge for surgical pathologists, although modern methods of ancillary diagnosis provide powerful tools that resolve most difficult cases. Current techniques range from identification of DNA alternations, including gene fusions, chromosome translocations, and genetic deletions, to recognition of characteristic patterns of protein expression, usually visualized with immunohistochemistry. In spite of these advances, recognition of key cellular and histologic features remains the keystone of diagnosis but requires adequately fixed and carefully stained histologic sections. Cytologic features now suffice for diagnosis if confirmed by appropriate testing. This article outlines key histologic features of pediatric small cell neoplasms and the algorithms that allow diagnostic confirmation and the initiation of appropriate therapy.
Collapse
Affiliation(s)
- David M Parham
- Department of Pathology, College of Medicine, University of Oklahoma Health Sciences Center, 940 Stanton L. Young Boulevard, Room BMSB 451, Oklahoma City, OK 70104, USA.
| |
Collapse
|
48
|
Cao L, Yu Y, Bilke S, Walker RL, Mayeenuddin LH, Azorsa DO, Yang F, Pineda M, Helman LJ, Meltzer PS. Genome-wide identification of PAX3-FKHR binding sites in rhabdomyosarcoma reveals candidate target genes important for development and cancer. Cancer Res 2010; 70:6497-508. [PMID: 20663909 PMCID: PMC2922412 DOI: 10.1158/0008-5472.can-10-0582] [Citation(s) in RCA: 193] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The PAX3-FKHR fusion protein is present in a majority of alveolar rhabdomyosarcomas associated with increased aggressiveness and poor prognosis. To better understand the molecular pathogenesis of PAX3-FKHR, we carried out the first, unbiased genome-wide identification of PAX3-FKHR binding sites and associated target genes in alveolar rhabdomyosarcoma. The data shows that PAX3-FKHR binds to the same sites as PAX3 at both MYF5 and MYOD enhancers. The genome-wide analysis reveals that the PAX3-FKHR sites are (a) mostly distal to transcription start sites, (b) conserved, (c) enriched for PAX3 motifs, and (d) strongly associated with genes overexpressed in PAX3-FKHR-positive rhabdomyosarcoma cells and tumors. There is little evidence in our data set for PAX3-FKHR binding at the promoter sequences. The genome-wide analysis further illustrates a strong association between PAX3 and E-box motifs in these binding sites, suggestive of a common coregulation for many target genes. We also provide the first direct evidence that FGFR4 and IGF1R are the targets for PAX3-FKHR. The map of PAX3-FKHR binding sites provides a framework for understanding the pathogenic roles of PAX3-FKHR, as well as its molecular targets to allow a systematic evaluation of agents against this aggressive rhabdomyosarcoma.
Collapse
MESH Headings
- Anaplastic Lymphoma Kinase
- Binding Sites
- Cell Line, Tumor
- E-Box Elements
- Genome, Human
- Genome-Wide Association Study
- Humans
- MyoD Protein/genetics
- MyoD Protein/metabolism
- N-Myc Proto-Oncogene Protein
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Oncogene Proteins/genetics
- Oncogene Proteins/metabolism
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/metabolism
- Protein-Tyrosine Kinases/genetics
- Protein-Tyrosine Kinases/metabolism
- Receptor Protein-Tyrosine Kinases
- Receptor, Fibroblast Growth Factor, Type 4/genetics
- Receptor, IGF Type 1/antagonists & inhibitors
- Receptor, IGF Type 1/biosynthesis
- Receptor, IGF Type 1/genetics
- Regulatory Elements, Transcriptional
- Rhabdomyosarcoma, Alveolar/genetics
- Rhabdomyosarcoma, Alveolar/metabolism
- Sarcoma, Ewing/genetics
- Sarcoma, Ewing/metabolism
- Transcription Initiation Site
- Up-Regulation
Collapse
Affiliation(s)
- Liang Cao
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Human Genome Research Institute, Bethesda, Maryland 20892, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
Multidrug resistance is a common problem in the treatment of childhood rhabdomyosarcoma (RMS). Therefore, novel treatment regimes such as immunotherapy have to be evaluated. The aim of this study was to detect possible targets on RMS cells and to investigate whether corresponding humanized antibodies could be used to treat RMS. Screening for potential targets common for different subtypes of RMS was carried out with Affymetrix mRNA expression arrays on 12 primary RMS samples. Subsequent pathway analysis revealed the epidermal growth factor receptor (EGFR) as a potential target for antibody therapy. Expression of EGFR and binding of its specific antibody Cetuximab to embryonal RMS cell lines RD and A-204 and alveolar RMS Rh30 were monitored by flow cytometry. Cetuximab activity was quantified by proliferation assay on RMS cells, and by antibody dependent cellular cytotoxicity assay with peripheral blood mononuclear cells (PBMCs). Gene expression analysis revealed a high expression of EGFR in all embryonal RMS compared with alveolar RMS. The EGFR specific antibody, Cetuximab binds to Rh30 and to RD but not to A-204 cells. Proliferation of these cells was influenced neither by Cetuximab nor by the growth factor EGF. However, cell dependent cytotoxicity of PBMCs to RMS cells such as Rh30 and RD was enhanced specifically by Cetuximab. These promising Cetuximab effects justify analysis under in vivo conditions using suitable models.
Collapse
|
50
|
Schäfer BW, Niggli F. Multidisciplinary management of childhood sarcoma: time to expand. Expert Rev Anticancer Ther 2010; 10:1163-6. [DOI: 10.1586/era.10.95] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|