1
|
Kumar H, Adegunlehin A, Chen Z, Kim P. FusionPub, a therapeutic landscape of human fusion genes. Brief Bioinform 2025; 26:bbaf145. [PMID: 40188496 PMCID: PMC11972630 DOI: 10.1093/bib/bbaf145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 02/22/2025] [Accepted: 03/14/2025] [Indexed: 04/08/2025] Open
Abstract
To advance the development of fusion protein-targeting therapeutics, it is crucial to understand how patients with fusion oncoproteins have been treated and what small molecules have been studied. To fill this gap, we developed FusionPub, a knowledgebase of the therapeutic landscape of human fusion genes. We searched PubMed abstracts for 107K human fusion genes with 14K drugs. We also searched PubMed abstracts for 18K human fusion proteins, considering all gene synonyms of individual partner genes with 14K drugs. After manual curation, we found 17 342 records from 9623 PubMed abstracts. For these fusion genes and drugs, we provide a summary sentence of their relationship using the generative AI, details of the drugs, clinical trial records, common drug records from patient treatment history, etc. FusionPub will be a valuable and unique resource providing knowledge of the development of fusion gene targeting therapeutics.
Collapse
Affiliation(s)
- Himansu Kumar
- Department of Bioinformatics and Systems Medicine, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, 7000 Fannin St., Houston, TX 77030, United States
| | - Abayomi Adegunlehin
- Department of Health Data Science and Artificial Intelligence, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, 7000 Fannin St., Houston, TX 77030, United States
| | - Zikang Chen
- Department of Computer Science, Rice University, 6100 Main St, Houston, TX 77005, United States
| | - Pora Kim
- Department of Bioinformatics and Systems Medicine, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, 7000 Fannin St., Houston, TX 77030, United States
| |
Collapse
|
2
|
Codenotti S, Asperti M, Poli M, Lorenzi L, Pietrantoni A, Cassandri M, Marampon F, Fanzani A. Synthetic inhibition of SREBP2 and the mevalonate pathway blocks rhabdomyosarcoma tumor growth in vitro and in vivo and promotes chemosensitization. Mol Metab 2025; 92:102085. [PMID: 39706565 PMCID: PMC11750561 DOI: 10.1016/j.molmet.2024.102085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/03/2024] [Accepted: 12/12/2024] [Indexed: 12/23/2024] Open
Abstract
OBJECTIVE The aim of the present study was to investigate the effects of targeting the mevalonate pathway (MVP) in rhabdomyosarcoma (RMS), a soft tissue tumor with a prevalence in young people. METHODS In silico analyses of RNA datasets were performed to correlate MVP with RMS patient survival. The sensitivity of RMS cell lines to MVP inhibitors was assessed in vitro by analysis of cell growth (crystal violet and clonogenic assays), cell migration (wound healing assay), cell survival (neutral red assay), and oxidative stress (ROS assay). The effects of MVP inhibitors were tested in vivo by analyzing RMS xenografts grown in NOD/SCID mice. Quantification of protein targets was performed using immunoblotting or immunohistochemistry analyses. RESULTS In silico analysis showed upregulation of sterol regulatory element-binding protein 2 (SREBP2) and MVP genes, including 3-Hydroxy-3-Methylglutaryl-CoA Reductase (HMGCR), farnesyl-diphosphate synthase (FDPS), squalene epoxidase (SQLE), which correlated with worse overall patient survival. Targeting of MVP in human RD and RH30 lines by inhibitors of SREBP2 (fatostatin), HMGCR (lovastatin and simvastatin), and FDPS (zoledronic acid) resulted in impaired cell growth, migration, and viability, and increased oxidative cell death in combination with actinomycin D. Conversely, cholesterol (CHO) supplementation enhanced cell growth and migration. Fatostatin and lovastatin produced rapid attenuation of Erk1/2 and Akt1 signaling in RMS lines, and oral administration of lovastatin reduced tumor mass growth of xenografted RD cells in NOD/SCID mice. Finally, we found that forced Akt1 activation in RD cells was sufficient to drive SREBP2, HMGCR and SQLE protein expression, promoting increased susceptibility to MVP inhibitors. CONCLUSIONS These data suggest that the Akt1, SREBP2 and MVP axis is critical for RMS tumor growth, migration, and oxidative stress protection primarily through maintaining adequate CHO levels that enable proper intracellular signaling. Therefore, stimulating CHO depletion via SREBP2 and MVP inhibition may represent a viable option to improve the combination therapy protocol, especially in pAkt1-positive RMS.
Collapse
Affiliation(s)
- Silvia Codenotti
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.
| | - Michela Asperti
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Maura Poli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Luisa Lorenzi
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy; ASST Spedali Civili di Brescia, 25123, Brescia, Italy
| | - Alberto Pietrantoni
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy; ASST Spedali Civili di Brescia, 25123, Brescia, Italy
| | - Matteo Cassandri
- Department of Radiological Sciences, Oncology and Anatomic Pathology, "Sapienza" University of Rome, 00161, Rome, Italy
| | | | - Alessandro Fanzani
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.
| |
Collapse
|
3
|
Song Z, Gong B, Qu T, Chen Y, Zhao G, Jin Y, Zhao Q. Anlotinib destabilizes PAX3-FOXO1 to induce rhabdomyosarcoma cell death via upregulating NEK2. Biomed Pharmacother 2024; 177:117126. [PMID: 38996706 DOI: 10.1016/j.biopha.2024.117126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/06/2024] [Accepted: 07/08/2024] [Indexed: 07/14/2024] Open
Abstract
BACKGROUND Rhabdomyosarcoma (RMS) is one of the most common soft tissue sarcomas in children and adolescents, in which PAX3-FOXO1 fusion gene positive patients have very poor prognosis. PAX3-FOXO1 has been identified as an independent prognostic predictor in RMS, with no currently available targeted therapeutic intervention. The novel tyrosine kinase inhibitor anlotinib exhibits a wide range of anticancer effects in multiple types of cancers; however, there have been no relevant studies regarding its application in RMS. MATERIALS AND METHODS We investigated the effects of PAX3-FOXO1 on the therapeutic efficacy of anlotinib using the CCK-8 assay, flow cytometry, invasion assay, wound healing assay, western blotting, quantitative polymerase chain reaction(qPCR), and xenograft experiments. RNA-seq and co-immunoprecipitation assays were conducted to determine the specific mechanism by which anlotinib regulates PAX3-FOXO1 expression. RESULTS Anlotinib effectively inhibited RMS cell proliferation and promoted apoptosis and G2/M phase arrest while impeding tumor growth in vivo. Downregulation of PAX3-FOXO1 enhances the antitumor effects of anlotinib. Anlotinib upregulates protein kinase NEK2 and increases the degradation of PAX3-FOXO1 via the ubiquitin-proteasome pathway, leading to a reduction in PAX3-FOXO1 protein levels. CONCLUSION Anlotinib effectively inhibited the malignant progression of RMS and promoted degradation of the fusion protein PAX3-FOXO1. Anlotinib could be a targeted therapeutic approach to treat PAX3-FOXO1 fusion-positive RMS.
Collapse
Affiliation(s)
- Zian Song
- Department of Pediatric Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Centre for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Baocheng Gong
- Department of Pediatric Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Centre for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Tongyuan Qu
- Department of Pediatric Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Centre for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Yankun Chen
- Department of Pediatric Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Centre for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Guangzong Zhao
- Department of Pediatric Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Centre for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Yan Jin
- Department of Pediatric Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Centre for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China.
| | - Qiang Zhao
- Department of Pediatric Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Centre for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China.
| |
Collapse
|
4
|
Rich J, Bennaroch M, Notel L, Patalakh P, Alberola J, Issa F, Opolon P, Bawa O, Rondof W, Marchais A, Dessen P, Meurice G, Le-Gall M, Polrot M, Ser-Le Roux K, Mamchaoui K, Droin N, Raslova H, Maire P, Geoerger B, Pirozhkova I. DiPRO1 distinctly reprograms muscle and mesenchymal cancer cells. EMBO Mol Med 2024; 16:1840-1885. [PMID: 39009887 PMCID: PMC11319797 DOI: 10.1038/s44321-024-00097-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 06/13/2024] [Accepted: 06/18/2024] [Indexed: 07/17/2024] Open
Abstract
We have recently identified the uncharacterized ZNF555 protein as a component of a productive complex involved in the morbid function of the 4qA locus in facioscapulohumeral dystrophy. Subsequently named DiPRO1 (Death, Differentiation, and PROliferation related PROtein 1), our study provides substantial evidence of its role in the differentiation and proliferation of human myoblasts. DiPRO1 operates through the regulatory binding regions of SIX1, a master regulator of myogenesis. Its relevance extends to mesenchymal tumors, such as rhabdomyosarcoma (RMS) and Ewing sarcoma, where DiPRO1 acts as a repressor via the epigenetic regulators TIF1B and UHRF1, maintaining methylation of cis-regulatory elements and gene promoters. Loss of DiPRO1 mimics the host defense response to virus, awakening retrotransposable repeats and the ZNF/KZFP gene family. This enables the eradication of cancer cells, reprogramming the cellular decision balance towards inflammation and/or apoptosis by controlling TNF-α via NF-kappaB signaling. Finally, our results highlight the vulnerability of mesenchymal cancer tumors to si/shDiPRO1-based nanomedicines, positioning DiPRO1 as a potential therapeutic target.
Collapse
Affiliation(s)
- Jeremy Rich
- UMR8126 CNRS, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France
| | - Melanie Bennaroch
- UMR8126 CNRS, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France
| | - Laura Notel
- UMR8126 CNRS, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France
| | - Polina Patalakh
- UMR8126 CNRS, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France
| | - Julien Alberola
- UMR8126 CNRS, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France
| | - Fayez Issa
- INSERM U1016, CNRS UMR 8104, Institut Cochin, Université Paris-Cité, Paris, France
| | - Paule Opolon
- Pathology and Cytology Section, UMS AMMICA, CNRS, INSERM, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France
| | - Olivia Bawa
- Pathology and Cytology Section, UMS AMMICA, CNRS, INSERM, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France
| | - Windy Rondof
- Bioinformatics Platform, UMS AMMICA, CNRS, INSERM, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer campus, INSERM U1015, Université Paris-Saclay, Villejuif, France
| | - Antonin Marchais
- Bioinformatics Platform, UMS AMMICA, CNRS, INSERM, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer campus, INSERM U1015, Université Paris-Saclay, Villejuif, France
| | - Philippe Dessen
- Bioinformatics Platform, UMS AMMICA, CNRS, INSERM, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France
| | - Guillaume Meurice
- Bioinformatics Platform, UMS AMMICA, CNRS, INSERM, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France
| | - Morgane Le-Gall
- Proteom'IC facility, Université Paris Cité, CNRS, INSERM, Institut Cochin, F-75014, Paris, France
| | - Melanie Polrot
- Pre-clinical Evaluation Unit (PFEP), INSERM, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France
| | - Karine Ser-Le Roux
- Pre-clinical Evaluation Unit (PFEP), INSERM, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France
| | - Kamel Mamchaoui
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, F-75013, Paris, France
| | - Nathalie Droin
- Genomic Platform, UMS AMMICA US 23 INSERM UAR 3655 CNRS, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France
- UMR1287 INSERM, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France
| | - Hana Raslova
- UMR1287 INSERM, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France
| | - Pascal Maire
- INSERM U1016, CNRS UMR 8104, Institut Cochin, Université Paris-Cité, Paris, France
| | - Birgit Geoerger
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer campus, INSERM U1015, Université Paris-Saclay, Villejuif, France
| | - Iryna Pirozhkova
- UMR8126 CNRS, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France.
- INSERM U1016, CNRS UMR 8104, Institut Cochin, Université Paris-Cité, Paris, France.
| |
Collapse
|
5
|
Ahmad Fahmi AK, Kamel S, Arasu K, Mianxin C, Mohan Singh AS. Silent Killer in the Nose: Two Cases of Nasal Alveolar Rhabdomyosarcoma in Adults. Cureus 2023; 15:e50430. [PMID: 38222161 PMCID: PMC10784867 DOI: 10.7759/cureus.50430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/13/2023] [Indexed: 01/16/2024] Open
Abstract
We report two cases of nasal alveolar rhabdomyosarcoma (ARMS) in adult patients from our center who presented with local mass effect and systemic involvement. Our first patient had spontaneous unilateral epistaxis. Her blood investigation showed severe thrombocytopenia, and the bone marrow biopsy result showed bone marrow infiltration by non-hematopoietic malignant cells. Nasal endoscopy showed a mass arising medial to the left middle turbinate. Our second patient presented with right eye proptosis, associated with blurring of vision. Nasal endoscopy showed a right whitish nasal mass arising lateral to the middle turbinate. Both patients were diagnosed by immunohistochemical analysis showing ARMS, a soft tissue malignancy uncommon in adults. RMS in adults has a worse prognosis. Hence, the management is challenging. Early diagnostic workup is essential for the commencement of early treatment for better oncological outcomes.
Collapse
Affiliation(s)
| | - Syafiqah Kamel
- Department of Otorhinolaryngology-Head & Neck Surgery, Taiping Hospital, Taiping, MYS
| | - Kanivannen Arasu
- Department of Otorhinolaryngology-Head & Neck Surgery, Taiping Hospital, Taiping, MYS
| | - Chew Mianxin
- Department of Pathology, Taiping Hospital, Taiping, MYS
| | | |
Collapse
|
6
|
Cornelison R, Marrah L, Fierti A, Piczak C, Glowczyk M, Tajammal A, Lynch S, Li H. The Potential for Targeting AVIL and Other Actin-Binding Proteins in Rhabdomyosarcoma. Int J Mol Sci 2023; 24:14196. [PMID: 37762498 PMCID: PMC10531751 DOI: 10.3390/ijms241814196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/12/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
Rhabdomyosarcoma (RMS) is the most common pediatric soft-tissue cancer with a survival rate below 27% for high-risk children despite aggressive multi-modal therapeutic interventions. After decades of research, no targeted therapies are currently available. Therapeutically targeting actin-binding proteins, although promising, has historically been challenging. Recent advances have made this possibility more salient, including our lab's identification of advillin (AVIL), a novel oncogenic actin-binding protein that plays a role in many cytoskeletal functions. AVIL is overexpressed in many RMS cell lines, patient-derived xenograft models, and a cohort of 30 clinical samples of both the alveolar (ARMS) and embryonal (ERMS) subtypes. Overexpression of AVIL in mesenchymal stem cells induces neoplastic transformation both in vitro and in vivo, and reversing overexpression through genetic modulation reverses the transformation. This suggests a critical role of AVIL in RMS tumorigenesis and maintenance. As an actin-binding protein, AVIL would not traditionally be considered a druggable target. This perspective will address the feasibility of targeting differentially expressed actin-binding proteins such as AVIL therapeutically, and how critical cell infrastructure can be damaged in a cancer-specific manner.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Hui Li
- Department of Pathology, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
7
|
Soloperto D, Gazzini S, Cerullo R. Molecular Mechanisms of Carcinogenesis in Pediatric Airways Tumors. Int J Mol Sci 2023; 24:ijms24032195. [PMID: 36768522 PMCID: PMC9916405 DOI: 10.3390/ijms24032195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 01/15/2023] [Accepted: 01/18/2023] [Indexed: 01/24/2023] Open
Abstract
Primary tumors of the airways in the pediatric population are very rare entities. For this reason, little is known about the pathogenesis of these neoplasms. Understanding the biology has different practical implications: for example, it could help in the differential diagnosis, have a prognostic significance, or may lead to the development of a targeted therapy. The aim of this article is to present the current knowledge about pediatric airways tumors, focusing on the molecular mechanisms that cause the onset and progression of these neoplasms. After a brief introduction of epidemiology and clinical presentation, the tumorigenesis of the most frequent pediatric airways tumors will be described: Juvenile-onset recurrent respiratory papillomatosis (JORRP), Subglottic Hemangiona (SH), Rhabdomyosarcoma (RMS), and Mucoepidermoid carcinoma (MEC).
Collapse
|
8
|
Zhang Y, Huang W, Li L, Qiu Y, Jiao H, Chen Z, Yang Q, Song L, Kang L. Retroperitoneal alveolar rhabdomyosarcoma intruding into spinal canal: A case report and literature review. Front Med (Lausanne) 2022; 9:1019964. [DOI: 10.3389/fmed.2022.1019964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 09/07/2022] [Indexed: 11/06/2022] Open
Abstract
BackgroundRhabdomyosarcoma (RMS) is the most frequent soft sarcoma in children and adolescents. Alveolar rhabdomyosarcoma (ARMS) is a relatively rare subtype that is characterized by aggressive behavior and an unsatisfactory prognosis. An ARMS can arise anywhere but most commonly occurs at extremity sites with a very small fraction in the retroperitoneum. The utility of 2-Deoxy-2-[fluorine-18]-fluoro-D-glucose (18F-FDG) positron emission tomography combined with computed tomography (PET/CT) remains to be established in ARMS.Case ReportA 3-year-old female child was accidentally found with a large left upper abdominal mass for a day. CT examination indicated a huge soft tissue mass in the left retroperitoneum extending superiorly to the level of the left hilus renalis and inferiorly to the left acetabulum in the pelvic cavity, with intrusion into the lumbar foramens. 18F-FDG PET/CT found a mass in the left retroperitoneum from the level of T12 to the left acetabulum, with the maximum standardized uptake value (SUVmax) of about 7.0, and a CT value of about 39 HU, invading the left L3-5 intervertebral foramina and protruding into the spinal canal, with unclear boundary with the spinal cord. Retroperitoneal tumor resection and the repair operation of vascular exploration were performed. An ARMS was confirmed by postoperative biopsy, immunohistochemical staining, and genetic detection with the rupture of the fork head in rhabdomyosarcoma (FKHR). The patient received chemotherapy and was in a good condition with no recurrence and obvious complications.ConclusionRetroperitoneal ARMS is rare and indicates a poor outcome with the potential to involve vital organs and intrude into the spinal canal. Accurate diagnosis and staging using PET/CT would contribute to better risk stratifications and appropriate treatment individually.
Collapse
|
9
|
Pomella S, Porrazzo A, Cassandri M, Camero S, Codenotti S, Milazzo L, Vulcano F, Barillari G, Cenci G, Marchese C, Fanzani A, Megiorni F, Rota R, Marampon F. Translational Implications for Radiosensitizing Strategies in Rhabdomyosarcoma. Int J Mol Sci 2022; 23:13281. [PMID: 36362070 PMCID: PMC9656983 DOI: 10.3390/ijms232113281] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 10/28/2022] [Accepted: 10/28/2022] [Indexed: 08/13/2024] Open
Abstract
Rhabdomyosarcoma (RMS) is the most common soft tissue sarcoma of childhood and adolescence that includes FP-RMS, harboring the fusion oncoprotein PAX3/7-FOXO1 and FN-RMS, often mutant in the RAS pathway. Risk stratifications of RMS patients determine different prognostic groups and related therapeutic treatment. Current multimodal therapeutic strategies involve surgery, chemotherapy (CHT) and radiotherapy (RT), but despite the deeper knowledge of response mechanisms underpinning CHT treatment and the technological improvements that characterize RT, local failures and recurrence frequently occur. This review sums up the RMS classification and the management of RMS patients, with special attention to RT treatment and possible radiosensitizing strategies for RMS tumors. Indeed, RMS radioresistance is a clinical problem and further studies aimed at dissecting radioresistant molecular mechanisms are needed to identify specific targets to hit, thus improving RT-induced cytotoxicity.
Collapse
Affiliation(s)
- Silvia Pomella
- Department of Oncohematology, Bambino Gesù Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, 00146 Rome, Italy
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Antonella Porrazzo
- Units of Molecular Genetics of Complex Phenotypes, Bambino Gesù Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, 00146 Rome, Italy
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza University of Rome, 00161 Rome, Italy
| | - Matteo Cassandri
- Department of Oncohematology, Bambino Gesù Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, 00146 Rome, Italy
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza University of Rome, 00161 Rome, Italy
| | - Simona Camero
- Department of Maternal, Infantile and Urological Sciences, Sapienza University of Rome, 00161 Rome, Italy
| | - Silvia Codenotti
- Department of Molecular and Translational Medicine, Division of Biotechnology, University of Brescia, 25123 Brescia, Italy
| | - Luisa Milazzo
- Department of Oncology and Molecular Medicine, Italian National Institute of Health, 00161 Rome, Italy
| | - Francesca Vulcano
- Department of Oncology and Molecular Medicine, Italian National Institute of Health, 00161 Rome, Italy
| | - Giovanni Barillari
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Giovanni Cenci
- Department of Biology and Biotechnology “C. Darwin”, Sapienza University of Rome, 00185 Rome, Italy
| | - Cinzia Marchese
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Alessandro Fanzani
- Department of Molecular and Translational Medicine, Division of Biotechnology, University of Brescia, 25123 Brescia, Italy
| | - Francesca Megiorni
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Rossella Rota
- Department of Oncohematology, Bambino Gesù Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, 00146 Rome, Italy
| | - Francesco Marampon
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza University of Rome, 00161 Rome, Italy
| |
Collapse
|
10
|
Machado ER, van de Vlekkert D, Sheppard HS, Perry S, Downing SM, Laxton J, Ashmun R, Finkelstein DB, Neale GA, Hu H, Harwood FC, Koo SC, Grosveld GC, d'Azzo A. Haploinsufficiency of the lysosomal sialidase NEU1 results in a model of pleomorphic rhabdomyosarcoma in mice. Commun Biol 2022; 5:992. [PMID: 36127469 PMCID: PMC9489700 DOI: 10.1038/s42003-022-03968-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 09/08/2022] [Indexed: 11/08/2022] Open
Abstract
Rhabdomyosarcoma, the most common pediatric sarcoma, has no effective treatment for the pleomorphic subtype. Still, what triggers transformation into this aggressive phenotype remains poorly understood. Here we used Ptch1+/-/ETV7TG/+/- mice with enhanced incidence of rhabdomyosarcoma to generate a model of pleomorphic rhabdomyosarcoma driven by haploinsufficiency of the lysosomal sialidase neuraminidase 1. These tumors share mostly features of embryonal and some of alveolar rhabdomyosarcoma. Mechanistically, we show that the transforming pathway is increased lysosomal exocytosis downstream of reduced neuraminidase 1, exemplified by the redistribution of the lysosomal associated membrane protein 1 at the plasma membrane of tumor and stromal cells. Here we exploit this unique feature for single cell analysis and define heterogeneous populations of exocytic, only partially differentiated cells that force tumors to pleomorphism and promote a fibrotic microenvironment. These data together with the identification of an adipogenic signature shared by human rhabdomyosarcoma, and likely fueling the tumor's metabolism, make this model of pleomorphic rhabdomyosarcoma ideal for diagnostic and therapeutic studies.
Collapse
Affiliation(s)
- Eda R Machado
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | | | - Heather S Sheppard
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Scott Perry
- Flow Cytometry Core Facility, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Susanna M Downing
- Department of Cell & Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Jonathan Laxton
- Flow Cytometry Core Facility, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Richard Ashmun
- Flow Cytometry Core Facility, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - David B Finkelstein
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Geoffrey A Neale
- Hartwell Center for Bioinformatics and Biotechnology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Huimin Hu
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Frank C Harwood
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Selene C Koo
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Gerard C Grosveld
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
| | - Alessandra d'Azzo
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
| |
Collapse
|
11
|
Duan C, Townley H. Exploitation of High Tumour GSH Levels for Targeted siRNA Delivery in Rhabdomyosarcoma Cells. Biomolecules 2022; 12:biom12081129. [PMID: 36009022 PMCID: PMC9405954 DOI: 10.3390/biom12081129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/09/2022] [Accepted: 08/15/2022] [Indexed: 11/16/2022] Open
Abstract
Metastatic alveolar rhabdomyosarcoma (aRMS) is an aggressive paediatric cancer with a poor prognosis. Downregulation of critical tumour genes using targeted siRNA remains an obstacle, but association with nanoparticles could help to deliver, protect, target, and enhance penetration. siRNA towards two genes was investigated: (i) Human αB-crystallin (CRYAB) and Heat Shock Protein Family B (Small) Member 2 (HSPB2), and (ii) Keratin 17 (KRT17). A mesoporous silica based nanosystem was linked to siRNA via disulfide bonds and loaded with IR820 dye. Transfection efficiency and signalling was evaluated, and the metabolic effects and cell proliferation were monitored in 2D culture and 3D spheroid models. The bound siRNA was protected from degradation with RNase I for at least 24 h. The delivered siRNA showed significant suppression of viability; 53.21 ± 23.40% for CRYAB and HSPB2 siRNA, and 88.06 ± 17.28% for KRT17 siRNA. After 72 h this increased to >50% cell apoptosis and necrosis. Intracellular total glutathione (GSH) levels were also compared with fibroblasts, and the RMS cell lines showed a several-fold increase. IR820 cellular uptake rate and penetration depth was significantly improved by nanoparticle delivery. Targetted siRNA delivery may pave the way for less invasive and more effective treatments of aRMS.
Collapse
Affiliation(s)
- Chengchen Duan
- Nuffield Department of Women’s and Reproductive Health, Oxford University John Radcliffe Hospital, Headington, Oxford OX3 9DU, UK
| | - Helen Townley
- Nuffield Department of Women’s and Reproductive Health, Oxford University John Radcliffe Hospital, Headington, Oxford OX3 9DU, UK
- Department of Engineering Science, Oxford University, Parks Road, Oxford OX1 3PJ, UK
- Correspondence: ; Tel.: +44-1865-283792
| |
Collapse
|
12
|
Urla C, Stagno MJ, Fuchs J, Warmann SW, Schmid E. Anticancer bioactivity of zerumbone on pediatric rhabdomyosarcoma cells. J Cancer Res Clin Oncol 2022:10.1007/s00432-022-04237-1. [PMID: 35931788 DOI: 10.1007/s00432-022-04237-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 07/25/2022] [Indexed: 10/16/2022]
Abstract
PURPOSE Natural products are generally regarded as safe and have been shown to mediate anticancer activities against a variety of cell types. Zerumbone is a natural cyclic sesquiterpene derived from the rhizome of Zingiber zerumbet, which has attracted extensive attention in the recent decade for anticancer activities. The present study investigates the in vitro effect of zerumbone on rhabdomyosarcoma cells. METHODS Two rhabdomyosarcoma cell lines (RD and RH30) were used as the model system. The growth inhibition of zerumbone was measured by MTT-assay, apoptosis via flow cytometry, gene expression by real-time PCR, the migration by transwell assay, and intracellular signaling by Western blotting. RESULTS Zerumbone shows anticancer effects on RD and RH30 cells in a dose-dependent manner via cell growth inhibition and induction of apoptosis. Exposure of RD and RH30 cells on zerumbone also resulted in a decrease of migration and downregulation of the hedgehog pathway. CONCLUSIONS Taken together, our study provided the first evidence that zerumbone imparted strong inhibitory and apoptotic effects on pediatric rhabdomyosarcoma cell lines and merit further investigation as a promising candidate for the anticancer therapy.
Collapse
Affiliation(s)
- Cristian Urla
- Department of Pediatric Surgery and Pediatric Urology, University Children's Hospital of Tuebingen, Hoppe-Seyler-Str. 3, 72076, Tuebingen, Germany
| | - Matias Julian Stagno
- Department of Pediatric Surgery and Pediatric Urology, University Children's Hospital of Tuebingen, Hoppe-Seyler-Str. 3, 72076, Tuebingen, Germany
| | - Jörg Fuchs
- Department of Pediatric Surgery and Pediatric Urology, University Children's Hospital of Tuebingen, Hoppe-Seyler-Str. 3, 72076, Tuebingen, Germany
| | - Steven W Warmann
- Department of Pediatric Surgery and Pediatric Urology, University Children's Hospital of Tuebingen, Hoppe-Seyler-Str. 3, 72076, Tuebingen, Germany
| | - Evi Schmid
- Department of Pediatric Surgery and Pediatric Urology, University Children's Hospital of Tuebingen, Hoppe-Seyler-Str. 3, 72076, Tuebingen, Germany.
| |
Collapse
|
13
|
Lanzi C, Cassinelli G. Combinatorial strategies to potentiate the efficacy of HDAC inhibitors in fusion-positive sarcomas. Biochem Pharmacol 2022; 198:114944. [DOI: 10.1016/j.bcp.2022.114944] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/02/2022] [Accepted: 02/02/2022] [Indexed: 12/12/2022]
|
14
|
Shrestha R, Mohankumar K, Martin G, Hailemariam A, Lee SO, Jin UH, Burghardt R, Safe S. Flavonoids kaempferol and quercetin are nuclear receptor 4A1 (NR4A1, Nur77) ligands and inhibit rhabdomyosarcoma cell and tumor growth. J Exp Clin Cancer Res 2021; 40:392. [PMID: 34906197 PMCID: PMC8670039 DOI: 10.1186/s13046-021-02199-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 11/26/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Flavonoids exhibit both chemopreventive and chemotherapeutic activity for multiple tumor types, however, their mechanisms of action are not well defined. Based on some of their functional and gene modifying activities as anticancer agents, we hypothesized that kaempferol and quercetin were nuclear receptor 4A1 (NR4A1, Nur77) ligands and confirmed that both compounds directly bound NR4A1 with KD values of 3.1 and 0.93 μM, respectively. METHODS The activities of kaempferol and quercetin were determined in direct binding to NR4A1 protein and in NR4A1-dependent transactivation assays in Rh30 and Rh41 rhabdomyosarcoma (RMS) cells. Flavonoid-dependent effects as inhibitors of cell growth, survival and invasion were determined in XTT and Boyden chamber assays respectively and changes in protein levels were determined by western blots. Tumor growth inhibition studies were carried out in athymic nude mice bearing Rh30 cells as xenografts. RESULTS Kaempferol and quercetin bind NR4A1 protein and inhibit NR4A1-dependent transactivation in RMS cells. NR4A1 also regulates RMS cell growth, survival, mTOR signaling and invasion. The pro-oncogenic PAX3-FOXO1 and G9a genes are also regulated by NR4A1 and, these pathways and genes are all inhibited by kaempferol and quercetin. Moreover, at a dose of 50 mg/kg/d kaempferol and quercetin inhibited tumor growth in an athymic nude mouse xenograft model bearing Rh30 cells. CONCLUSION These results demonstrate the clinical potential for repurposing kaempferol and quercetin for clinical applications as precision medicine for treating RMS patients that express NR4A1 in order to increase the efficacy and decrease dosages of currently used cytotoxic drugs.
Collapse
Affiliation(s)
- Rupesh Shrestha
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX, 77843, USA
| | - Kumaravel Mohankumar
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, 4466 TAMU, College Station, TX, 77843-4466, USA
| | - Greg Martin
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, 4466 TAMU, College Station, TX, 77843-4466, USA
| | - Amanuel Hailemariam
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, 4466 TAMU, College Station, TX, 77843-4466, USA
| | - Syng-Ook Lee
- Department of Food Science and Technology, Keimyung University, Daegu, 42601, Republic of Korea
| | - Un-Ho Jin
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, 4466 TAMU, College Station, TX, 77843-4466, USA
| | - Robert Burghardt
- Department of Veterinary Integrated Biosciences, Texas A&M University, College Station, TX, 77843, USA
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, 4466 TAMU, College Station, TX, 77843-4466, USA.
| |
Collapse
|
15
|
Laubscher D, Gryder BE, Sunkel BD, Andresson T, Wachtel M, Das S, Roschitzki B, Wolski W, Wu XS, Chou HC, Song YK, Wang C, Wei JS, Wang M, Wen X, Ngo QA, Marques JG, Vakoc CR, Schäfer BW, Stanton BZ, Khan J. BAF complexes drive proliferation and block myogenic differentiation in fusion-positive rhabdomyosarcoma. Nat Commun 2021; 12:6924. [PMID: 34836971 PMCID: PMC8626462 DOI: 10.1038/s41467-021-27176-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 10/25/2021] [Indexed: 12/12/2022] Open
Abstract
Rhabdomyosarcoma (RMS) is a pediatric malignancy of skeletal muscle lineage. The aggressive alveolar subtype is characterized by t(2;13) or t(1;13) translocations encoding for PAX3- or PAX7-FOXO1 chimeric transcription factors, respectively, and are referred to as fusion positive RMS (FP-RMS). The fusion gene alters the myogenic program and maintains the proliferative state while blocking terminal differentiation. Here, we investigated the contributions of chromatin regulatory complexes to FP-RMS tumor maintenance. We define the mSWI/SNF functional repertoire in FP-RMS. We find that SMARCA4 (encoding BRG1) is overexpressed in this malignancy compared to skeletal muscle and is essential for cell proliferation. Proteomic studies suggest proximity between PAX3-FOXO1 and BAF complexes, which is further supported by genome-wide binding profiles revealing enhancer colocalization of BAF with core regulatory transcription factors. Further, mSWI/SNF complexes localize to sites of de novo histone acetylation. Phenotypically, interference with mSWI/SNF complex function induces transcriptional activation of the skeletal muscle differentiation program associated with MYCN enhancer invasion at myogenic target genes, which is recapitulated by BRG1 targeting compounds. We conclude that inhibition of BRG1 overcomes the differentiation blockade of FP-RMS cells and may provide a therapeutic strategy for this lethal childhood tumor.
Collapse
Affiliation(s)
- Dominik Laubscher
- grid.412341.10000 0001 0726 4330Department of Oncology and Children’s Research Center, University Children’s Hospital, Zurich, Switzerland
| | - Berkley E. Gryder
- grid.48336.3a0000 0004 1936 8075Genetics Branch, NCI, NIH, Bethesda, MD USA ,grid.67105.350000 0001 2164 3847Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH USA
| | - Benjamin D. Sunkel
- grid.240344.50000 0004 0392 3476Nationwide Children’s Hospital, Center for Childhood Cancer and Blood Diseases, Columbus, OH USA
| | - Thorkell Andresson
- grid.418021.e0000 0004 0535 8394Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD USA
| | - Marco Wachtel
- grid.412341.10000 0001 0726 4330Department of Oncology and Children’s Research Center, University Children’s Hospital, Zurich, Switzerland
| | - Sudipto Das
- grid.418021.e0000 0004 0535 8394Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD USA
| | - Bernd Roschitzki
- grid.7400.30000 0004 1937 0650Functional Genomics Center, University of Zurich/ETH Zurich, Zurich, Switzerland
| | - Witold Wolski
- grid.7400.30000 0004 1937 0650Functional Genomics Center, University of Zurich/ETH Zurich, Zurich, Switzerland
| | - Xiaoli S. Wu
- grid.225279.90000 0004 0387 3667Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724 USA
| | - Hsien-Chao Chou
- grid.48336.3a0000 0004 1936 8075Genetics Branch, NCI, NIH, Bethesda, MD USA
| | - Young K. Song
- grid.48336.3a0000 0004 1936 8075Genetics Branch, NCI, NIH, Bethesda, MD USA
| | - Chaoyu Wang
- grid.48336.3a0000 0004 1936 8075Genetics Branch, NCI, NIH, Bethesda, MD USA
| | - Jun S. Wei
- grid.48336.3a0000 0004 1936 8075Genetics Branch, NCI, NIH, Bethesda, MD USA
| | - Meng Wang
- grid.240344.50000 0004 0392 3476Nationwide Children’s Hospital, Center for Childhood Cancer and Blood Diseases, Columbus, OH USA
| | - Xinyu Wen
- grid.48336.3a0000 0004 1936 8075Genetics Branch, NCI, NIH, Bethesda, MD USA
| | - Quy Ai Ngo
- grid.412341.10000 0001 0726 4330Department of Oncology and Children’s Research Center, University Children’s Hospital, Zurich, Switzerland
| | - Joana G. Marques
- grid.412341.10000 0001 0726 4330Department of Oncology and Children’s Research Center, University Children’s Hospital, Zurich, Switzerland
| | - Christopher R. Vakoc
- grid.225279.90000 0004 0387 3667Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724 USA
| | - Beat W. Schäfer
- grid.412341.10000 0001 0726 4330Department of Oncology and Children’s Research Center, University Children’s Hospital, Zurich, Switzerland
| | - Benjamin Z. Stanton
- grid.240344.50000 0004 0392 3476Nationwide Children’s Hospital, Center for Childhood Cancer and Blood Diseases, Columbus, OH USA ,grid.261331.40000 0001 2285 7943Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH USA ,grid.261331.40000 0001 2285 7943Department of Biological Chemistry & Pharmacology, The Ohio State University College of Medicine, Columbus, OH USA
| | - Javed Khan
- Genetics Branch, NCI, NIH, Bethesda, MD, USA.
| |
Collapse
|
16
|
Panicker S, Venkatabalasubramanian S, Pathak S, Ramalingam S. The impact of fusion genes on cancer stem cells and drug resistance. Mol Cell Biochem 2021; 476:3771-3783. [PMID: 34095988 DOI: 10.1007/s11010-021-04203-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 05/29/2021] [Indexed: 12/12/2022]
Abstract
With ever increasing evidences on the role of fusion genes as the oncogenic protagonists in myriad cancers, it's time to explore if fusion genes can be the next generational drug targets in meeting the current demands of higher drug efficacy. Eliminating cancer stem cells (CSC) has become the current focus; however, we have reached a standstill in drug development owing to the lack of effective strategies to eradicate CSC. We believe that fusion genes could be the novel targets to overcome this limitation. The intriguing feature of fusion genes is that it dominantly impacts every aspect of CSC including self-renewal, differentiation, lineage commitment, tumorigenicity and stemness. Given the clinical success of fusion gene-based drugs in hematological cancers, our attempt to target fusion genes in eradicating CSC can be rewarding. As fusion genes are expressed explicitly in cancer cells, eradicating CSC by targeting fusion genes provides yet an another advantage of negligible patient side effects since normal cells remain unaffected by the drug. We hereby delineate the latest evidences on how fusion genes regulate CSC and drug resistance.
Collapse
Affiliation(s)
- Saurav Panicker
- Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Kanchipuram, 603203, Tamil Nadu, India
| | | | - Surajit Pathak
- Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Kelambakkam, Chennai, 603103, Tamil Nadu, India
| | - Satish Ramalingam
- Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Kanchipuram, 603203, Tamil Nadu, India.
| |
Collapse
|
17
|
Adamus A, Ali I, Vasileiadis V, Al-Hileh L, Lisec J, Frank M, Seitz G, Engel N. Vincetoxicum arnottianum modulates motility features and metastatic marker expression in pediatric rhabdomyosarcoma by stabilizing the actin cytoskeleton. BMC Complement Med Ther 2021; 21:136. [PMID: 33947373 PMCID: PMC8097906 DOI: 10.1186/s12906-021-03299-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 04/07/2021] [Indexed: 12/28/2022] Open
Abstract
Background Prevention of metastatic invasion is one of the main challenges in the treatment of alveolar rhabdomyosarcoma. Still the therapeutic options are limited. Therefore, an anti-tumor screening was initiated focusing on the anti-metastatic and anti-invasion properties of selected medicinal plant extracts and phytoestrogens, already known to be effective in the prevention and treatment of different cancer entities. Methods Treatment effects were first evaluated by cell viability, migration, invasion, and colony forming assays on the alveolar rhabdomyosarcoma cell line RH-30 in comparison with healthy primary cells. Results Initial anti-tumor screenings of all substances analyzed in this study, identified the plant extract of Vincetoxicum arnottianum (VSM) as the most promising candidate, harboring the highest anti-metastatic potential. Those significant anti-motility properties were proven by a reduced ability for migration (60%), invasion (99%) and colony formation (61%) under 48 h exposure to 25 μg/ml VSM. The restricted motility features were due to an induction of the stabilization of the cytoskeleton – actin fibers were 2.5-fold longer and were spanning the entire cell. Decreased proliferation (PCNA, AMT, GCSH) and altered metastasis (e. g. SGPL1, CXCR4, stathmin) marker expression on transcript and protein level confirmed the significant lowered tumorigenicity under VSM treatment. Finally, significant alterations in the cell metabolism were detected for 25 metabolites, with levels of uracil, N-acetyl serine and propanoyl phosphate harboring the greatest alterations. Compared to the conventional therapy with cisplatin, VSM treated cells demonstrated a similar metabolic shutdown of the primary cell metabolism. Primary control cells were not affected by the VSM treatment. Conclusions This study revealed the VSM root extract as a potential, new migrastatic drug candidate for the putative treatment of pediatric alveolar rhabdomyosarcoma with actin filament stabilizing properties and accompanied by a marginal effect on the vitality of primary cells. Graphical abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12906-021-03299-x.
Collapse
Affiliation(s)
- Anna Adamus
- Department of Pediatric Surgery, University Hospital, Marburg, Germany
| | - Iftikhar Ali
- Department of Chemistry, Karakoram International University, Gilgit, Pakistan.,Shandong Key Laboratory of TCM Quality Control Technology, Shandong Analysis and Test Center, Jinan, Shandong Province, P.R. China
| | | | - Luai Al-Hileh
- Department of Pediatric Surgery, University Hospital, Marburg, Germany
| | - Jan Lisec
- Division 1.7 Analytical Chemistry, Federal Institute for Materials Research and Testing (BAM), Berlin, Germany
| | - Marcus Frank
- Medical Biology and Electron Microscopy Center, Rostock University Medical Center, Rostock, Germany.,Department of Life, Light & Matter, University of Rostock, Rostock, Germany
| | - Guido Seitz
- Department of Pediatric Surgery, University Hospital, Marburg, Germany
| | - Nadja Engel
- Department of Oral and Maxillofacial Surgery, Facial Plastic Surgery, Rostock University Medical Center, Rostock, Germany.
| |
Collapse
|
18
|
Almasi S, Crawford Parks TE, Ravel-Chapuis A, MacKenzie A, Côté J, Cowan KN, Jasmin BJ. Differential regulation of autophagy by STAU1 in alveolar rhabdomyosarcoma and non-transformed skeletal muscle cells. Cell Oncol (Dordr) 2021; 44:851-870. [PMID: 33899158 DOI: 10.1007/s13402-021-00607-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2021] [Indexed: 11/30/2022] Open
Abstract
PURPOSE Recent work has highlighted the therapeutic potential of targeting autophagy to modulate cell survival in a variety of diseases including cancer. Recently, we found that the RNA-binding protein Staufen1 (STAU1) is highly expressed in alveolar rhabdomyosarcoma (ARMS) and that this abnormal expression promotes tumorigenesis. Here, we asked whether STAU1 is involved in the regulation of autophagy in ARMS cells. METHODS We assessed the impact of STAU1 expression modulation in ARMS cell lines (RH30 and RH41), non-transformed skeletal muscle cells (C2C12) and STAU1-transgenic mice using complementary techniques. RESULTS We found that STAU1 silencing reduces autophagy in the ARMS cell lines RH30 and RH41, while increasing their apoptosis. Mechanistically, this inhibitory effect was found to be caused by a direct negative impact of STAU1 depletion on the stability of Beclin-1 (BECN1) and ATG16L1 mRNAs, as well as by an indirect inhibition of JNK signaling via increased expression of Dual specificity phosphatase 8 (DUSP8). Pharmacological activation of JNK or expression silencing of DUSP8 was sufficient to restore autophagy in STAU1-depleted cells. By contrast, we found that STAU1 downregulation in non-transformed skeletal muscle cells activates autophagy in a mTOR-dependent manner, without promoting apoptosis. A similar effect was observed in skeletal muscles obtained from STAU1-overexpressing transgenic mice. CONCLUSIONS Together, our data indicate an effect of STAU1 on autophagy regulation in ARMS cells and its differential role in non-transformed skeletal muscle cells. Our findings suggest a cancer-specific potential of targeting STAU1 for the treatment of ARMS.
Collapse
Affiliation(s)
- Shekoufeh Almasi
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, K1H 8M5, Canada.,The Eric J. Poulin Centre for Neuromuscular Diseases, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Tara E Crawford Parks
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, K1H 8M5, Canada.,The Eric J. Poulin Centre for Neuromuscular Diseases, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Aymeric Ravel-Chapuis
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, K1H 8M5, Canada.,The Eric J. Poulin Centre for Neuromuscular Diseases, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Alex MacKenzie
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, K1H 8M5, Canada.,The Eric J. Poulin Centre for Neuromuscular Diseases, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.,Division of Endocrinology, Department of Paediatric, CHEO, University of Ottawa, Ottawa, ON, Canada
| | - Jocelyn Côté
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, K1H 8M5, Canada.,The Eric J. Poulin Centre for Neuromuscular Diseases, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Kyle N Cowan
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, K1H 8M5, Canada.,Division of Paediatric Surgery, Department of Surgery, CHEO, University of Ottawa, Ottawa, ON, Canada.,Molecular Biomedicine Program, CHEO, Ottawa, ON, Canada
| | - Bernard J Jasmin
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, K1H 8M5, Canada. .,The Eric J. Poulin Centre for Neuromuscular Diseases, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
19
|
Berlow NE, Crawford KA, Bult CJ, Noakes C, Sloma I, Rudzinski ER, Keller C. Functional impact of a germline RET mutation in alveolar rhabdomyosarcoma. Cold Spring Harb Mol Case Stud 2021; 7:mcs.a006049. [PMID: 33722797 PMCID: PMC8208040 DOI: 10.1101/mcs.a006049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 03/01/2021] [Indexed: 11/24/2022] Open
Abstract
Specific mutations in the RET proto-oncogene are associated with multiple endocrine neoplasia type 2A, a hereditary syndrome characterized by tumorigenesis in multiple glandular elements. In rare instances, MEN2A-associated germline RET mutations have also occurred with non-MEN2A associated cancers. One such germline mutant RET mutation occurred concomitantly in a young adult diagnosed with alveolar rhabdomyosarcoma, a pediatric and young adult soft-tissue cancer with a generally poor prognosis. Although tumor tissue samples were initially unable to provide a viable cell culture for study, tumor tissues were sequenced for molecular characteristics. Through a hierarchical clustering approach, the index case sample was matched to several genetically similar cell models, which were transformed to express the same mutant RET as the index case and used to explore potential therapeutic options for mutant RET-bearing alveolar rhabdomyosarcoma. We also determined whether the RET mutation associated with the index case caused synthetic lethality to select clinical agents. From our investigation, we did not identify synthetic lethality associated with the expression of that patient's RET variant, and overall we did not find experimental evidence for the role of RET in rhabdomyosarcoma progression.
Collapse
Affiliation(s)
- Noah E Berlow
- Children's Cancer Therapy Development Institute, Beaverton, Oregon 97005, USA
| | - Kenneth A Crawford
- Children's Cancer Therapy Development Institute, Beaverton, Oregon 97005, USA
| | - Carol J Bult
- The Jackson Laboratory, Bar Harbor, Maine 04609, USA
| | | | - Ido Sloma
- Champions Oncology, Hackensack, New Jersey 07601, USA
| | | | - Charles Keller
- Children's Cancer Therapy Development Institute, Beaverton, Oregon 97005, USA
| |
Collapse
|
20
|
Straccia P, Martini M, Pierconti F. Pericardial metastasis from alveolar rhabdomyosarcoma: A case report. Diagn Cytopathol 2021; 49:E297-E300. [PMID: 33560576 DOI: 10.1002/dc.24717] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 01/05/2021] [Accepted: 01/26/2021] [Indexed: 11/10/2022]
Abstract
Alveolar rhabdomyosarcoma (ARMS) commonly occurring in children and adolescents, is a rare and aggressive soft tissue malignancy demonstrating rapid growth and dissemination. Although their histopathologic and immunohistochemical findings are well known, cytomorphological features on serous effusions have not been well documented. A case of metastatic rhabdomyosarcoma diagnosed on cytological examination of the pericardial fluid cytology and discuss the diagnostic difficulties that were encountered in this case has been presented.
Collapse
Affiliation(s)
- Patrizia Straccia
- Division of Anatomic Pathology and Histology, Università Cattolica del Sacro Cuore, Foundation "A. Gemelli" University Hospital, Rome, Italy
| | - Maurizio Martini
- Division of Anatomic Pathology and Histology, Università Cattolica del Sacro Cuore, Foundation "A. Gemelli" University Hospital, Rome, Italy
| | - Francesco Pierconti
- Division of Anatomic Pathology and Histology, Università Cattolica del Sacro Cuore, Foundation "A. Gemelli" University Hospital, Rome, Italy
| |
Collapse
|
21
|
Williams J, Xu B, Putnam D, Thrasher A, Li C, Yang J, Chen X. MethylationToActivity: a deep-learning framework that reveals promoter activity landscapes from DNA methylomes in individual tumors. Genome Biol 2021; 22:24. [PMID: 33461601 PMCID: PMC7814737 DOI: 10.1186/s13059-020-02220-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 12/06/2020] [Indexed: 12/12/2022] Open
Abstract
Although genome-wide DNA methylomes have demonstrated their clinical value as reliable biomarkers for tumor detection, subtyping, and classification, their direct biological impacts at the individual gene level remain elusive. Here we present MethylationToActivity (M2A), a machine learning framework that uses convolutional neural networks to infer promoter activities based on H3K4me3 and H3K27ac enrichment, from DNA methylation patterns for individual genes. Using publicly available datasets in real-world test scenarios, we demonstrate that M2A is highly accurate and robust in revealing promoter activity landscapes in various pediatric and adult cancers, including both solid and hematologic malignant neoplasms.
Collapse
Affiliation(s)
- Justin Williams
- Department of Computational Biology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Mail Stop 1135, Memphis, TN, 38105, USA
| | - Beisi Xu
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Daniel Putnam
- Department of Computational Biology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Mail Stop 1135, Memphis, TN, 38105, USA
| | - Andrew Thrasher
- Department of Computational Biology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Mail Stop 1135, Memphis, TN, 38105, USA
| | - Chunliang Li
- Department of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jun Yang
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Xiang Chen
- Department of Computational Biology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Mail Stop 1135, Memphis, TN, 38105, USA.
| |
Collapse
|
22
|
Usefulness of 18f-FDG PET-CT in Staging, Restaging, and Response Assessment in Pediatric Rhabdomyosarcoma. Diagnostics (Basel) 2020; 10:diagnostics10121112. [PMID: 33371506 PMCID: PMC7767528 DOI: 10.3390/diagnostics10121112] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 11/23/2020] [Accepted: 12/11/2020] [Indexed: 12/18/2022] Open
Abstract
Rhabdomyosarcoma is the most common soft-tissue sarcoma of childhood. Despite clinical advances, subsets of these patients continue to suffer high morbidity and mortality rates associated with their disease. Following the European guidelines for 18F-FDG PET and PET-CT imaging in pediatric oncology, the routine use of 18F-FDG PET-CT may be useful for patients affected by rhabdomyosarcoma, in staging, in the evaluation of response to therapy, and for restaging/detection of relapse. The European Pediatric Protocols are very old, and for staging and restaging, they recommend only radionuclide bone scan. The 18F-FDG PET-CT exam is listed as an optional investigation prescribed according to local availability and local protocols in the investigations panel required at the end of the treatment. We present two cases highlighting the usefulness of 18F-FDG PET-CT in managing pediatric patients affected by rhabdomyosarcoma, providing some bibliographic references.
Collapse
|
23
|
Sugito N, Heishima K, Ito Y, Akao Y. Synthetic MIR143-3p Suppresses Cell Growth in Rhabdomyosarcoma Cells by Interrupting RAS Pathways Including PAX3-FOXO1. Cancers (Basel) 2020; 12:cancers12113312. [PMID: 33182548 PMCID: PMC7696565 DOI: 10.3390/cancers12113312] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Accepted: 11/06/2020] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Rhabdomyosarcoma (RMS) is a soft tissue sarcoma with embryonal (ERMS) and alveoral (ARMS) features, most frequently found in children. ARMS has the worse prognosis due to the formation of the chimeric PAX3–FOXO1 gene. New therapies are needed for the treatment of ARMS. The aim of this study is to evaluate the anticancer effect of chemically-modified MIR143-3p#12 (CM-MIR143#12) on RMS. The ectopic expression of CM-MIR143#12 induced a cell growth suppression by silencing not only KRAS, AKT, and ERK but also the PAX3–FOXO1 chimeric gene, and KRAS networks could control the expression of chimeric PAX3–FOXO1 in ARMS cells. Moreover, CM-MIR143#12 also silenced NRAS mutant in ERMS RD cells. CM-MIR143#12 can be a new nucleic acid medicine for the treatment of RMS by impairing the RAS networks including PAX3–FOXO1. Abstract Rhabdomyosarcoma (RMS) is a soft tissue sarcoma most frequently found in children. In RMS, there are two major subtypes, embryonal RMS (ERMS) and alveolar RMS (ARMS). ARMS has the worse prognosis of the two owing to the formation of the chimeric PAX3–FOXO1 gene. A novel therapeutic method is required for treating ARMS. In our previous study, we found that the ectopic expression of chemically modified MIR143-3p#12 (CM-MIR143#12), which is RNase-resistant and shows the highest anti-proliferation activity among the synthesized MIR143 derivatives that were tested, induces significant cell growth suppression by targeting KRAS, AKT, and ERK in colorectal cancer cells. The expression of MIR143-3p in RMS was dramatically downregulated compared with that of normal tissue. Ectopic expression of CM-MIR143#12 in RMS cells resulted in a significant growth inhibitory effect through the induction of apoptosis and autophagy. Interestingly, we found that CM-MIR143#12 also silenced the expression of chimeric PAX3–FOXO1 directly and, using siR-KRAS or siR-AKT, that KRAS networks regulated the expression of PAX3–FOXO1 in ARMS cells. In ERMS harboring NRAS mutation, CM-MIR143#12 silenced mutated NRAS. These findings indicate that CM-MIR143#12 efficiently perturbed the RAS signaling pathway, including the ARMS-specific KRAS/PAX3–FOXO1 networks.
Collapse
Affiliation(s)
- Nobuhiko Sugito
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1194, Japan; (N.S.); (K.H.)
| | - Kazuki Heishima
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1194, Japan; (N.S.); (K.H.)
| | - Yuko Ito
- Department of Anatomy and Cell Biology, Division of Life Sciences, Osaka Medical College, 2-7 Daigaku-machi, Takatsuki, Osaka 569-8686, Japan;
| | - Yukihiro Akao
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1194, Japan; (N.S.); (K.H.)
- Correspondence:
| |
Collapse
|
24
|
Gonzalez Curto G, Der Vartanian A, Frarma YEM, Manceau L, Baldi L, Prisco S, Elarouci N, Causeret F, Korenkov D, Rigolet M, Aurade F, De Reynies A, Contremoulins V, Relaix F, Faklaris O, Briscoe J, Gilardi-Hebenstreit P, Ribes V. The PAX-FOXO1s trigger fast trans-differentiation of chick embryonic neural cells into alveolar rhabdomyosarcoma with tissue invasive properties limited by S phase entry inhibition. PLoS Genet 2020; 16:e1009164. [PMID: 33175861 PMCID: PMC7682867 DOI: 10.1371/journal.pgen.1009164] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 11/23/2020] [Accepted: 10/02/2020] [Indexed: 12/18/2022] Open
Abstract
The chromosome translocations generating PAX3-FOXO1 and PAX7-FOXO1 chimeric proteins are the primary hallmarks of the paediatric fusion-positive alveolar subtype of Rhabdomyosarcoma (FP-RMS). Despite the ability of these transcription factors to remodel chromatin landscapes and promote the expression of tumour driver genes, they only inefficiently promote malignant transformation in vivo. The reason for this is unclear. To address this, we developed an in ovo model to follow the response of spinal cord progenitors to PAX-FOXO1s. Our data demonstrate that PAX-FOXO1s, but not wild-type PAX3 or PAX7, trigger the trans-differentiation of neural cells into FP-RMS-like cells with myogenic characteristics. In parallel, PAX-FOXO1s remodel the neural pseudo-stratified epithelium into a cohesive mesenchyme capable of tissue invasion. Surprisingly, expression of PAX-FOXO1s, similar to wild-type PAX3/7, reduce the levels of CDK-CYCLIN activity and increase the fraction of cells in G1. Introduction of CYCLIN D1 or MYCN overcomes this PAX-FOXO1-mediated cell cycle inhibition and promotes tumour growth. Together, our findings reveal a mechanism that can explain the apparent limited oncogenicity of PAX-FOXO1 fusion transcription factors. They are also consistent with certain clinical reports indicative of a neural origin of FP-RMS.
Collapse
Affiliation(s)
| | | | | | - Line Manceau
- Université de Paris, CNRS, Institut Jacques Monod, Paris, France
| | - Lorenzo Baldi
- Université de Paris, CNRS, Institut Jacques Monod, Paris, France
| | - Selene Prisco
- Université de Paris, CNRS, Institut Jacques Monod, Paris, France
| | - Nabila Elarouci
- Programme Cartes d'Identité des Tumeurs, Ligue Nationale Contre le Cancer, Paris, France
| | - Frédéric Causeret
- Université de Paris, Imagine Institute, Team Genetics and Development of the Cerebral Cortex, Paris, France
- Université de Paris, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, Paris, France
| | - Daniil Korenkov
- Université de Paris, CNRS, Institut Jacques Monod, Paris, France
| | - Muriel Rigolet
- Univ Paris Est Créteil, INSERM, EnVA, EFS, IMRB, Créteil, France
| | - Frédéric Aurade
- Univ Paris Est Créteil, INSERM, EnVA, EFS, IMRB, Créteil, France
- Sorbonne Université, INSERM, UMRS974, Center for Research in Myology, Paris, France
| | - Aurélien De Reynies
- Programme Cartes d'Identité des Tumeurs, Ligue Nationale Contre le Cancer, Paris, France
| | - Vincent Contremoulins
- ImagoSeine core facility of Institut Jacques Monod and member of France-BioImaging, France
| | - Frédéric Relaix
- Univ Paris Est Créteil, INSERM, EnVA, EFS, IMRB, Créteil, France
| | - Orestis Faklaris
- ImagoSeine core facility of Institut Jacques Monod and member of France-BioImaging, France
| | - James Briscoe
- The Francis Crick Institute, 1 Midland Road, London, United Kingdom
| | | | - Vanessa Ribes
- Université de Paris, CNRS, Institut Jacques Monod, Paris, France
| |
Collapse
|
25
|
McCarthy N, Dolgikh N, Logue S, Patterson JB, Zeng Q, Gorman AM, Samali A, Fulda S. The IRE1 and PERK arms of the unfolded protein response promote survival of rhabdomyosarcoma cells. Cancer Lett 2020; 490:76-88. [PMID: 32679165 DOI: 10.1016/j.canlet.2020.07.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/25/2020] [Accepted: 07/08/2020] [Indexed: 02/07/2023]
Abstract
Rhabdomyosarcoma (RMS), the most common soft-tissue sarcoma, is associated with a low 5-year survival and harsh treatment side effects, underscoring an urgent need for therapy. The unfolded protein response (UPR) is activated in response to endoplasmic reticulum (ER) stress, where three ER stress receptors, IRE1, PERK and ATF6, aim to restore cellular homeostasis. The UPR is pro-tumourigenic in many cancers. In this study, we investigate basal UPR activity in RMS. Basal activation of IRE1 and PERK was observed in RMS cell lines, which was diminished upon addition of the IRE1 RNase inhibitor, MKC8866, or PERK inhibitor, AMGEN44. UPR inhibition caused a reduction in cell viability, cell proliferation and inhibition of long-term colony formation in both subtypes of RMS. Alveolar RMS (ARMS) subtype was highly sensitive to IRE1 inhibition, whereas embryonal RMS (ERMS) subtypes responded more markedly to PERK inhibition. Further investigation revealed a robust activation of senescence upon UPR inhibition. For the first time, the UPR is implicated in RMS biology and phenotype, and inhibition of UPR signalling reduces cell growth, suggesting that the UPR may be a promising target in RMS.
Collapse
Affiliation(s)
- Nicole McCarthy
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University Frankfurt, Komturstr. 3a, 60528, Frankfurt, Germany
| | - Nadezda Dolgikh
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University Frankfurt, Komturstr. 3a, 60528, Frankfurt, Germany
| | - Susan Logue
- Rady Faculty of Health Sciences, University of Manitoba, Canada
| | | | - Qinping Zeng
- Fosun Orinove PharmaTech Inc., Suzhou, Jiangsu, China
| | - Adrienne M Gorman
- Apoptosis Research Centre, National University of Ireland Galway, Galway, Ireland; School of Natural Sciences, National University of Ireland Galway, Galway, Ireland
| | - Afshin Samali
- Apoptosis Research Centre, National University of Ireland Galway, Galway, Ireland; School of Natural Sciences, National University of Ireland Galway, Galway, Ireland
| | - Simone Fulda
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University Frankfurt, Komturstr. 3a, 60528, Frankfurt, Germany; German Cancer Consortium (DKTK), Partner Site Frankfurt, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
26
|
Partner independent fusion gene detection by multiplexed CRISPR-Cas9 enrichment and long read nanopore sequencing. Nat Commun 2020; 11:2861. [PMID: 32504042 PMCID: PMC7275081 DOI: 10.1038/s41467-020-16641-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 05/12/2020] [Indexed: 02/06/2023] Open
Abstract
Fusion genes are hallmarks of various cancer types and important determinants for diagnosis, prognosis and treatment. Fusion gene partner choice and breakpoint-position promiscuity restricts diagnostic detection, even for known and recurrent configurations. Here, we develop FUDGE (FUsion Detection from Gene Enrichment) to accurately and impartially identify fusions. FUDGE couples target-selected and strand-specific CRISPR-Cas9 activity for fusion gene driver enrichment — without prior knowledge of fusion partner or breakpoint-location — to long read nanopore sequencing with the bioinformatics pipeline NanoFG. FUDGE has flexible target-loci choices and enables multiplexed enrichment for simultaneous analysis of several genes in multiple samples in one sequencing run. We observe on-average 665 fold breakpoint-site enrichment and identify nucleotide resolution fusion breakpoints within 2 days. The assay identifies cancer cell line and tumor sample fusions irrespective of partner gene or breakpoint-position. FUDGE is a rapid and versatile fusion detection assay for diagnostic pan-cancer fusion detection. Fusion genes are a hallmarks of cancer, though breakpoint-position promiscuity restricts diagnostic detection. Here, the authors present FUDGE, a CRISPR-Cas9-based enrichment strategy for nanopore sequencing to identify target fusions irrespective of genomic breakpoint or fusion partner.
Collapse
|
27
|
Camero S, Camicia L, Marampon F, Ceccarelli S, Shukla R, Mannarino O, Pizer B, Schiavetti A, Pizzuti A, Tombolini V, Marchese C, Dominici C, Megiorni F. BET inhibition therapy counteracts cancer cell survival, clonogenic potential and radioresistance mechanisms in rhabdomyosarcoma cells. Cancer Lett 2020; 479:71-88. [PMID: 32200036 DOI: 10.1016/j.canlet.2020.03.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/04/2020] [Accepted: 03/10/2020] [Indexed: 12/15/2022]
Abstract
The antitumour effects of OTX015, a first-in-class BET inhibitor (BETi), were investigated as a single agent or in combination with ionizing radiation (IR) in preclinical in vitro models of rhabdomyosarcoma (RMS), the most common childhood soft tissue sarcoma. Herein, we demonstrated the upregulation of BET Bromodomain gene expression in RMS tumour biopsies and cell lines compared to normal skeletal muscle. In vitro experiments showed that OTX015 significantly reduced RMS cell proliferation by altering cell cycle modulators and apoptotic related proteins due to the accumulation of DNA breaks that cells are unable to repair. Interestingly, OTX015 also impaired migration capacity and tumour-sphere architecture by downregulating pro-stemness genes and was able to potentiate ionizing radiation effects by reducing the expression of different drivers of tumour dissemination and resistance mechanisms, including the GNL3 gene, that we correlated for the first time with the RMS phenotype. In conclusion, our research sheds further light on the molecular events of OTX015 action against RMS cells and indicates this novel BETi as an effective option to improve therapeutic strategies and overcome the development of resistant cancer cells in patients with RMS.
Collapse
Affiliation(s)
- Simona Camero
- Department of Maternal, Infantile, and Urological Sciences, "Sapienza" University of Rome, Rome, Italy.
| | - Lucrezia Camicia
- Department of Maternal, Infantile, and Urological Sciences, "Sapienza" University of Rome, Rome, Italy.
| | - Francesco Marampon
- Department of Radiological, Oncological and Pathological Sciences, "Sapienza" University of Rome, Rome, Italy.
| | - Simona Ceccarelli
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy.
| | - Rajeev Shukla
- Department of Perinatal and Paediatric Pathology, Alder Hey Children's NHS Foundation Trust, Liverpool, UK.
| | - Olga Mannarino
- Department of Maternal, Infantile, and Urological Sciences, "Sapienza" University of Rome, Rome, Italy.
| | - Barry Pizer
- Department of Oncology, Alder Hey Children's NHS Foundation Trust, Eaton Road, Liverpool, L12 2AP, UK.
| | - Amalia Schiavetti
- Department of Maternal, Infantile, and Urological Sciences, "Sapienza" University of Rome, Rome, Italy.
| | - Antonio Pizzuti
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy.
| | - Vincenzo Tombolini
- Department of Radiological, Oncological and Pathological Sciences, "Sapienza" University of Rome, Rome, Italy.
| | - Cinzia Marchese
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy.
| | - Carlo Dominici
- Department of Maternal, Infantile, and Urological Sciences, "Sapienza" University of Rome, Rome, Italy.
| | - Francesca Megiorni
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy.
| |
Collapse
|
28
|
Hypericin and its radio iodinated derivatives – A novel combined approach for the treatment of pediatric alveolar rhabdomyosarcoma cells in vitro. Photodiagnosis Photodyn Ther 2020; 29:101588. [DOI: 10.1016/j.pdpdt.2019.101588] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 10/17/2019] [Accepted: 10/24/2019] [Indexed: 02/06/2023]
|
29
|
Calvão J, Cardoso JC, Ramos L, Figueiredo A. Primary cutaneous alveolar rhabdomyosarcoma in an adolescent - A challenging diagnosis. Pediatr Dermatol 2020; 37:184-186. [PMID: 31797424 DOI: 10.1111/pde.14055] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Primary cutaneous alveolar rhabdomyosarcoma is an extremely rare and highly aggressive soft tissue sarcoma that predominantly arises on the extremities and perineum of adolescents and young adults. Dermatologists should be aware of these tumors in order to promptly make the diagnosis and initiate treatment.
Collapse
Affiliation(s)
- Joana Calvão
- Dermatovenereology Department, Coimbra University Hospital, Coimbra, Portugal
| | - José Carlos Cardoso
- Dermatovenereology Department, Coimbra University Hospital, Coimbra, Portugal
| | - Leonor Ramos
- Dermatovenereology Department, Coimbra University Hospital, Coimbra, Portugal
| | - Américo Figueiredo
- Dermatovenereology Department, Coimbra University Hospital, Coimbra, Portugal
| |
Collapse
|
30
|
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
|
31
|
Chen C, Dorado Garcia H, Scheer M, Henssen AG. Current and Future Treatment Strategies for Rhabdomyosarcoma. Front Oncol 2019; 9:1458. [PMID: 31921698 PMCID: PMC6933601 DOI: 10.3389/fonc.2019.01458] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 12/05/2019] [Indexed: 12/31/2022] Open
Abstract
Rhabdomyosarcoma (RMS) is the most common soft tissue sarcoma in children, and can be subcategorized histologically and/or based on PAX-FOXO1 fusion gene status. Over the last four decades, there have been no significant improvements in clinical outcomes for advanced and metastatic RMS patients, underscoring a need for new treatment options for these groups. Despite significant advancements in our understanding of the genomic landscape and underlying biological mechanisms governing RMS that have informed the identification of novel therapeutic targets, development of these therapies in clinical trials has lagged far behind. In this review, we summarize the current frontline multi-modality therapy for RMS according to pediatric protocols, highlight emerging targeted therapies and immunotherapies identified by preclinical studies, and discuss early clinical trial data and the implications they hold for future clinical development.
Collapse
Affiliation(s)
- Celine Chen
- Department of Pediatric Oncology/Hematology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Heathcliff Dorado Garcia
- Department of Pediatric Oncology/Hematology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Monika Scheer
- Pediatrics 5, Klinikum Stuttgart, Olgahospital, Stuttgart, Germany
| | - Anton G. Henssen
- Department of Pediatric Oncology/Hematology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Experimental and Clinical Research Center (ECRC) of the MDC and Charité Berlin, Berlin, Germany
| |
Collapse
|
32
|
Pasvolsky O, Heiman L, Popovtzer A, Zimra Y, Rabizadeh E, Barshack I, Mardoukh C, Raanani P, Rozovski U. Genomic analysis of metastatic rhabdomyosarcoma masquerading as acute leukemia. Pathol Res Pract 2019; 216:152779. [PMID: 31813602 DOI: 10.1016/j.prp.2019.152779] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/01/2019] [Accepted: 12/01/2019] [Indexed: 10/25/2022]
Abstract
Blast appearing cells in the peripheral blood and bone marrow may occasionally arise from non-hematopoietic tissues. We present a 58 year old female who presented at our emergency room with symptomatic pancytopenia. Several months earlier she was diagnosed and treated for rhabdomyosracoma of the nasopharynx and entered remission. When we examined the bone-marrow aspirate we estimated the number of blasts at 25 %. Based on this evaluation, a provisional diagnosis of acute leukemia was made. However, immunohistochemistry and flow cytometry analysis revealed that the cells presumed to be blasts were in fact rhabdomyosarcoma cells masquerading as leukemia. The mutational landscapes of the primary tumor and the bone marrow metastasis had similar yet distinct profiles. Annotation analysis suggested that the primary and metastatic tumors use alternate mutations to activate the RAS/AKT signaling pathways. In this case, looking beyond the mutational profiling revealed an additional layer of similarity between both the original and metastatic samples, exposing a common and possibly targetable pathway. Application of annotation tools in clinical practice could enable extraction of valuable information from somatic mutational gene panels.
Collapse
Affiliation(s)
- Oren Pasvolsky
- Institute of Hematology, Davidoff Center, Beilinson Hospital, Rabin Medical Center, Petah Tikva, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Lucille Heiman
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; Department of Pathology, Rabin Medical Center, Petah Tikva, Israel
| | - Aron Popovtzer
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; Head and Neck Tumor Unit, Davidoff Cancer Center, Beilinson Hospital, Rabin Medical Center, Petah Tikva, Israel
| | - Yael Zimra
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; Hemato-Oncology Laboratory, Felsenstein Medical Research Center, Rabin Medical Center, Petah Tikva, Israel
| | - Esther Rabizadeh
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; Hemato-Oncology Laboratory, Felsenstein Medical Research Center, Rabin Medical Center, Petah Tikva, Israel
| | - Iris Barshack
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; Department of Pathology, Sheba Medical Center, Israel
| | - Corine Mardoukh
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; Department of Pathology, Sheba Medical Center, Israel
| | - Pia Raanani
- Institute of Hematology, Davidoff Center, Beilinson Hospital, Rabin Medical Center, Petah Tikva, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Uri Rozovski
- Institute of Hematology, Davidoff Center, Beilinson Hospital, Rabin Medical Center, Petah Tikva, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
33
|
Der Vartanian A, Chabanais J, Carrion C, Maftah A, Germot A. Downregulation of POFUT1 Impairs Secondary Myogenic Fusion Through a Reduced NFATc2/IL-4 Signaling Pathway. Int J Mol Sci 2019; 20:ijms20184396. [PMID: 31500188 PMCID: PMC6770550 DOI: 10.3390/ijms20184396] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Revised: 08/26/2019] [Accepted: 09/05/2019] [Indexed: 12/25/2022] Open
Abstract
Past work has shown that the protein O-fucosyltransferase 1 (POFUT1) is involved in mammal myogenic differentiation program. Pofut1 knockdown (Po –) in murine C2C12 cells leads to numerous elongated and thin myotubes, suggesting significant defects in secondary fusion. Among the few pathways involved in this process, NFATc2/IL-4 is described as the major one. To unravel the impact of POFUT1 on secondary fusion, we used wild-type (WT) C2C12 and Po – cell lines to follow Myf6, Nfatc2, Il-4 and Il-4rα expressions during a 120 h myogenic differentiation time course. Secreted IL-4 was quantified by ELISA. IL-4Rα expression and its labeling on myogenic cell types were investigated by Western blot and immunofluorescence, respectively. Phenotypic observations of cells treated with IL-4Rα blocking antibody were performed. In Po –, we found a decrease in nuclei number per myotube and a downexpression of Myf6. The observed downregulation of Nfatc2 is correlated to a diminution of secreted IL-4 and to the low level of IL-4Rα for reserve cells. Neutralization of IL-4Rα on WT C2C12 promotes myonuclear accretion defects, similarly to those identified in Po –. Thus, POFUT1 could be a new controller of myotube growth during myogenesis, especially through NFATc2/IL-4 signaling pathway.
Collapse
Affiliation(s)
- Audrey Der Vartanian
- PEIRENE, EA 7500, Glycosylation et différenciation cellulaire, Université de Limoges, F-87000 Limoges, France
- present address: INSERM, IMRB U955-E10, Faculté de Médecine, Université Paris Est Créteil, F-94000 Créteil, France
| | - Julien Chabanais
- PEIRENE, EA 7500, Glycosylation et différenciation cellulaire, Université de Limoges, F-87000 Limoges, France
| | - Claire Carrion
- UMR CNRS 7276, Contrôle de la Réponse Immune et des Lymphoproliférations, Université de Limoges, F-87000 Limoges, France
| | - Abderrahman Maftah
- PEIRENE, EA 7500, Glycosylation et différenciation cellulaire, Université de Limoges, F-87000 Limoges, France
| | - Agnès Germot
- PEIRENE, EA 7500, Glycosylation et différenciation cellulaire, Université de Limoges, F-87000 Limoges, France
- Correspondence: ; Tel.: +33-(0)5-55-45-76-57
| |
Collapse
|
34
|
Hutzen B, Ghonime M, Lee J, Mardis ER, Wang R, Lee DA, Cairo MS, Roberts RD, Cripe TP, Cassady KA. Immunotherapeutic Challenges for Pediatric Cancers. MOLECULAR THERAPY-ONCOLYTICS 2019; 15:38-48. [PMID: 31650024 PMCID: PMC6804520 DOI: 10.1016/j.omto.2019.08.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Solid tumors contain a mixture of malignant cells and non-malignant infiltrating cells that often create a chronic inflammatory and immunosuppressive microenvironment that restricts immunotherapeutic approaches. Although childhood and adult cancers share some similarities related to microenvironmental changes, pediatric cancers are unique, and adult cancer practices may not be wholly applicable to our pediatric patients. This review highlights the differences in tumorigenesis, viral infection, and immunologic response between children and adults that need to be considered when trying to apply experiences from experimental therapies in adult cancer patients to pediatric cancers.
Collapse
Affiliation(s)
- Brian Hutzen
- The Research Institute at Nationwide Children's Hospital, Center for Childhood Cancer and Blood Diseases, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Mohammed Ghonime
- The Research Institute at Nationwide Children's Hospital, Center for Childhood Cancer and Blood Diseases, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Joel Lee
- The Ohio State University, Columbus, OH, USA
| | - Elaine R Mardis
- The Research Institute at Nationwide Children's Hospital, Center for Childhood Cancer and Blood Diseases, The Ohio State University College of Medicine, Columbus, OH, USA.,The Ohio State University, Columbus, OH, USA.,Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA.,Institute for Genomic Medicine, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Ruoning Wang
- The Research Institute at Nationwide Children's Hospital, Center for Childhood Cancer and Blood Diseases, The Ohio State University College of Medicine, Columbus, OH, USA.,The Ohio State University, Columbus, OH, USA.,Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Dean A Lee
- The Research Institute at Nationwide Children's Hospital, Center for Childhood Cancer and Blood Diseases, The Ohio State University College of Medicine, Columbus, OH, USA.,The Ohio State University, Columbus, OH, USA.,Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Mitchell S Cairo
- Department of Pediatrics, Cancer and Blood Diseases Center, New York Medical College, Valhalla, NY, USA
| | - Ryan D Roberts
- The Research Institute at Nationwide Children's Hospital, Center for Childhood Cancer and Blood Diseases, The Ohio State University College of Medicine, Columbus, OH, USA.,The Ohio State University, Columbus, OH, USA.,Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Timothy P Cripe
- The Research Institute at Nationwide Children's Hospital, Center for Childhood Cancer and Blood Diseases, The Ohio State University College of Medicine, Columbus, OH, USA.,The Ohio State University, Columbus, OH, USA.,Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Kevin A Cassady
- The Research Institute at Nationwide Children's Hospital, Center for Childhood Cancer and Blood Diseases, The Ohio State University College of Medicine, Columbus, OH, USA.,The Ohio State University, Columbus, OH, USA.,Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA.,Division of Pediatric Infection Diseases, New York Medical College, Valhalla, NY, USA
| |
Collapse
|
35
|
c-Myb regulates tumorigenic potential of embryonal rhabdomyosarcoma cells. Sci Rep 2019; 9:6342. [PMID: 31004084 PMCID: PMC6474878 DOI: 10.1038/s41598-019-42684-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 04/04/2019] [Indexed: 02/08/2023] Open
Abstract
Rhabdomyosarcomas (RMS) are a heterogeneous group of mesodermal tumors, the most common sub-types are embryonal (eRMS) and alveolar (aRMS) rhabdomyosarcoma. Immunohistochemical analysis revealed c-Myb expression in both eRMS and aRMS. c-Myb has been reported to be often associated with malignant human cancers. We therefore investigated the c-Myb role in RMS using cellular models of RMS. Specific suppression of c-Myb by a lentiviral vector expressing doxycycline (Dox)-inducible c-Myb shRNA inhibited proliferation, colony formation, and migration of the eRMS cell line (RD), but not of the aRMS cell line (RH30). Upon c-Myb knockdown in eRMS cells, cells accumulated in G0/G1 phase, the invasive behaviour of cells was repressed, and elevated levels of myosin heavy chain, marker of muscle differentiation, was detected. Next, we used an RD-based xenograft model to investigate the role of c-Myb in eRMS tumorigenesis in vivo. We found that Dox administration did not result in efficient suppression of c-Myb in growing tumors. However, when c-Myb-deficient RD cells were implanted into SCID mice, we observed inefficient tumor grafting and attenuation of tumor growth during the initial stages of tumor expansion. The presented study suggests that c-Myb could be a therapeutic target in embryonal rhabdomyosarcoma assuming that its expression is ablated.
Collapse
|
36
|
Disialoganglioside GD2 Expression in Pediatric Rhabdomyosarcoma: A Case Series and Review of the Literature. J Pediatr Hematol Oncol 2019; 41:118-120. [PMID: 30198960 DOI: 10.1097/mph.0000000000001311] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Rhabdomyosarcoma (RMS) is the most common pediatric soft tissue sarcoma. Despite aggressive therapy, patients with metastatic or relapsed disease experience dismal outcomes and novel therapies are urgently needed. In this study, we evaluated expression of disialoganglioside (GD2), a cell surface antigen with therapeutic implication, in 16 RMS patient samples. Scoring revealed GD2 positivity in 25% of the samples. These data suggest that a small subset of RMS tumors express GD2, which may be a therapeutic target in these patients.
Collapse
|
37
|
Adamus A, Peer K, Ali I, Lisec J, Falodun A, Frank M, Seitz G, Engel N. Berberis orthobotrys - A promising herbal anti-tumorigenic candidate for the treatment of pediatric alveolar rhabdomyosarcoma. JOURNAL OF ETHNOPHARMACOLOGY 2019; 229:262-271. [PMID: 30315865 DOI: 10.1016/j.jep.2018.10.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 10/04/2018] [Accepted: 10/07/2018] [Indexed: 06/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Berberis orthobotrys (BORM) is a medical plant with a long history in traditional usage for the treatment of wounds, cancer, gastrointestinal malady and several other diseases. Our previous studies identified the endemic Pakistani plant Berberis orthobotrys Bien. ex Aitch. as promising source for the treatment of breast cancer and osteosarcoma. AIM OF THE STUDY The present study was aimed to evaluate the anti-cancer properties of 26 plant derived extracts and compounds including the methanolic root extract of Berberis orthobotrys (BORM) on pediatric alveolar rhabdomyosarcoma (RMA), which is known to develop drug resistance, metastatic invasion and potential tumor progression. MATERIALS AND METHODS The main anti-tumor activity of BORM was verified by focusing on morphological, cell structural and metabolic alterations via metabolic profiling, cell viability measurements, flow cytometry, western blotting and diverse microscopy-based methods using the human RMA cell line Rh30. RESULTS Exposure of 25 µg/ml BORM exerts an influence on the cell stability, the degradation of oncosomes as well as the shutdown of the metabolic activity of RMA cells, primarily by downregulation of the energy metabolism. Therefore glycyl-aspartic acid and N-acetyl serine decreased moderately, and uracil increased intracellularly. On healthy, non-transformed muscle cells BORM revealed very low metabolic alterations and nearly no cytotoxic impact. Furthermore, BORM is also capable to reduce Rh30 cell migration (~50%) and proliferation (induced G2/M cycle arrest) as well as to initiate apoptosis confirmed by reduced Bcl-2, Bax and PCNA expression and induced PARP-1 cleavage. CONCLUSIONS The study provides the first evidence, that BORM treatment is effective against RMA cells with low side effects on healthy cells.
Collapse
Affiliation(s)
- Anna Adamus
- Department of Pediatric Surgery, University Hospital Marburg, Baldingerstraße, 35033 Marburg, Germany
| | - Katharina Peer
- Department of Pediatric Surgery, University Hospital Marburg, Baldingerstraße, 35033 Marburg, Germany
| | - Iftikhar Ali
- Department of Chemistry, Karakoram International University, 15100 Gilgit, Pakistan
| | - Jan Lisec
- Division 1.7 Analytical Chemistry, Federal Institute for Materials Research and Testing (BAM), Richard-Willstätter-Straße 11, Berlin 12489 Germany
| | - Abiodun Falodun
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Benin, Benin City 300001, Nigeria
| | - Marcus Frank
- Medical Biology and Electron Microscopy Centre, Rostock University Medical Center, Strempelstraße 14, Rostock 18057, Germany
| | - Guido Seitz
- Department of Pediatric Surgery, University Hospital Marburg, Baldingerstraße, 35033 Marburg, Germany
| | - Nadja Engel
- Department of Pediatric Surgery, University Hospital Marburg, Baldingerstraße, 35033 Marburg, Germany; Department of Oral and Maxillofacial Surgery, Facial Plastic Surgery, Rostock University Medical Center, Schillingallee 35, 18057 Rostock, Germany.
| |
Collapse
|
38
|
Abstract
Rhabdomyosarcoma (RMS) is the most common soft tissue sarcoma in children and represents a high-grade neoplasm of skeletal myoblast-like cells. Decades of clinical and basic research have gradually improved our understanding of the pathophysiology of RMS and helped to optimize clinical care. The two major subtypes of RMS, originally characterized on the basis of light microscopic features, are driven by fundamentally different molecular mechanisms and pose distinct clinical challenges. Curative therapy depends on control of the primary tumour, which can arise at many distinct anatomical sites, as well as controlling disseminated disease that is known or assumed to be present in every case. Sophisticated risk stratification for children with RMS incorporates various clinical, pathological and molecular features, and that information is used to guide the application of multifaceted therapy. Such therapy has historically included cytotoxic chemotherapy as well as surgery, ionizing radiation or both. This Primer describes our current understanding of RMS epidemiology, disease susceptibility factors, disease mechanisms and elements of clinical care, including diagnostics, risk-based care of newly diagnosed and relapsed disease and the prevention and management of late effects in survivors. We also outline potential opportunities to further translate new biological insights into improved clinical outcomes.
Collapse
Affiliation(s)
- Stephen X Skapek
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Andrea Ferrari
- Pediatric Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Abha A Gupta
- Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Philip J Lupo
- Department of Pediatrics, Section of Hematology-Oncology, Baylor College of Medicine, Houston, TX, USA
| | - Erin Butler
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Janet Shipley
- Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, Belmont, UK
| | - Frederic G Barr
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Douglas S Hawkins
- Seattle Children's Hospital, University of Washington, and Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| |
Collapse
|
39
|
Camero S, Ceccarelli S, De Felice F, Marampon F, Mannarino O, Camicia L, Vescarelli E, Pontecorvi P, Pizer B, Shukla R, Schiavetti A, Mollace MG, Pizzuti A, Tombolini V, Marchese C, Megiorni F, Dominici C. PARP inhibitors affect growth, survival and radiation susceptibility of human alveolar and embryonal rhabdomyosarcoma cell lines. J Cancer Res Clin Oncol 2019; 145:137-152. [PMID: 30357520 PMCID: PMC6326011 DOI: 10.1007/s00432-018-2774-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 10/16/2018] [Indexed: 12/26/2022]
Abstract
PURPOSE PARP inhibitors (PARPi) are used in a wide range of human solid tumours but a limited evidence is reported in rhabdomyosarcoma (RMS), the most frequent childhood soft-tissue sarcoma. The cellular and molecular effects of Olaparib, a specific PARP1/2 inhibitor, and AZD2461, a newly synthesized PARP1/2/3 inhibitor, were assessed in alveolar and embryonal RMS cells both as single-agent and in combination with ionizing radiation (IR). METHODS Cell viability was monitored by trypan blue exclusion dye assays. Cell cycle progression and apoptosis were measured by flow cytometry, and alterations of specific molecular markers were investigated by, Real Time PCR, Western blotting and immunofluorescence experiments. Irradiations were carried out at a dose rate of 2 Gy (190 UM/min) or 4 Gy (380 UM/min). Radiosensitivity was assessed by using clonogenic assays. RESULTS Olaparib and AZD2461 dose-dependently reduced growth of both RH30 and RD cells by arresting growth at G2/M phase and by modulating the expression, activation and subcellular localization of specific cell cycle regulators. Downregulation of phospho-AKT levels and accumulation of γH2AX, a specific marker of DNA damage, were significantly and persistently induced by Olaparib and AZD2461 exposure, this leading to apoptosis-related cell death. Both PARPi significantly enhanced the effects of IR by accumulating DNA damage, increasing G2 arrest and drastically reducing the clonogenic capacity of RMS-cotreated cells. CONCLUSIONS This study suggests that the combined exposure to PARPi and IR might display a role in the treatment of RMS tumours compared with single-agent exposure, since stronger cytotoxic effects are induced, and compensatory survival mechanisms are prevented.
Collapse
Affiliation(s)
- Simona Camero
- Department of Paediatrics, “Sapienza” University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Simona Ceccarelli
- Department of Experimental Medicine, “Sapienza” University of Rome, Rome, Italy
| | - Francesca De Felice
- Department of Radiological, Oncological and Pathological Sciences, “Sapienza” University of Rome, Rome, Italy
| | - Francesco Marampon
- Department of Radiological, Oncological and Pathological Sciences, “Sapienza” University of Rome, Rome, Italy
| | - Olga Mannarino
- Department of Paediatrics, “Sapienza” University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Lucrezia Camicia
- Department of Paediatrics, “Sapienza” University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Enrica Vescarelli
- Department of Experimental Medicine, “Sapienza” University of Rome, Rome, Italy
| | - Paola Pontecorvi
- Department of Experimental Medicine, “Sapienza” University of Rome, Rome, Italy
| | - Barry Pizer
- Department of Oncology, Alder Hey Children’s NHS Foundation Trust, Eaton Road, Liverpool, L12 2AP UK
| | - Rajeev Shukla
- Department of Perinatal and Paediatric Pathology, Alder Hey Children’s NHS Foundation Trust, Liverpool, UK
| | - Amalia Schiavetti
- Department of Paediatrics, “Sapienza” University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Maria Giovanna Mollace
- Department of Paediatrics, “Sapienza” University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Antonio Pizzuti
- Department of Experimental Medicine, “Sapienza” University of Rome, Rome, Italy
| | - Vincenzo Tombolini
- Department of Radiological, Oncological and Pathological Sciences, “Sapienza” University of Rome, Rome, Italy
| | - Cinzia Marchese
- Department of Experimental Medicine, “Sapienza” University of Rome, Rome, Italy
| | - Francesca Megiorni
- Department of Paediatrics, “Sapienza” University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Carlo Dominici
- Department of Paediatrics, “Sapienza” University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| |
Collapse
|
40
|
Dupain C, Harttrampf AC, Boursin Y, Lebeurrier M, Rondof W, Robert-Siegwald G, Khoueiry P, Geoerger B, Massaad-Massade L. Discovery of New Fusion Transcripts in a Cohort of Pediatric Solid Cancers at Relapse and Relevance for Personalized Medicine. Mol Ther 2018; 27:200-218. [PMID: 30509566 DOI: 10.1016/j.ymthe.2018.10.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 10/18/2018] [Accepted: 10/26/2018] [Indexed: 12/16/2022] Open
Abstract
We hypothetized that pediatric cancers would more likely harbor fusion transcripts. To dissect the complexity of the fusions landscape in recurrent solid pediatric cancers, we conducted a study on 48 patients with different relapsing or resistant malignancies. By analyzing RNA sequencing data with a new in-house pipeline for fusions detection named ChimComp, followed by verification by real-time PCR, we identified and classified the most confident fusion transcripts (FTs) according to their potential biological function and druggability. The majority of FTs were predicted to affect key cancer pathways and described to be involved in oncogenesis. Contrary to previous descriptions, we found no significant correlation between the number of fusions and mutations, emphasizing the particularity to study pre-treated pediatric patients. A considerable proportion of FTs containing tumor suppressor genes was detected, reflecting their importance in pediatric cancers. FTs containing non-receptor tyrosine kinases occurred at low incidence and predominantly in brain tumors. Remarkably, more than 30% of patients presented a potentially druggable high-confidence fusion. In conclusion, we detected new oncogenic FTs in relapsing pediatric cancer patients by establishing a robust pipeline that can be applied to other malignancies, to detect and prioritize experimental validation studies leading to the development of new therapeutic options.
Collapse
Affiliation(s)
- Célia Dupain
- Université Paris-Sud 11, Laboratoire de Vectorologie et Thérapeutiques Anticancéreuses, UMR 8203, Villejuif 94805, France; CNRS, Villejuif, Laboratoire de Vectorologie et Thérapeutiques Anticancéreuses, UMR 8203, Villejuif 94805, France; Gustave Roussy, Laboratoire de Vectorologie et Thérapeutiques Anticancéreuses, UMR 8203, Villejuif 94805, France
| | - Anne C Harttrampf
- Université Paris-Sud 11, Laboratoire de Vectorologie et Thérapeutiques Anticancéreuses, UMR 8203, Villejuif 94805, France; CNRS, Villejuif, Laboratoire de Vectorologie et Thérapeutiques Anticancéreuses, UMR 8203, Villejuif 94805, France; Gustave Roussy, Laboratoire de Vectorologie et Thérapeutiques Anticancéreuses, UMR 8203, Villejuif 94805, France; Gustave Roussy, Department of Pediatric and Adolescent Oncology, Villejuif 94805, France
| | - Yannick Boursin
- Gustave Roussy, Bioinformatics Platform, AMMICA, INSERM US23/CNRS UMS3655, Villejuif 94805, France
| | - Manuel Lebeurrier
- Gustave Roussy, Bioinformatics Platform, AMMICA, INSERM US23/CNRS UMS3655, Villejuif 94805, France
| | - Windy Rondof
- Université Paris-Sud 11, Laboratoire de Vectorologie et Thérapeutiques Anticancéreuses, UMR 8203, Villejuif 94805, France; CNRS, Villejuif, Laboratoire de Vectorologie et Thérapeutiques Anticancéreuses, UMR 8203, Villejuif 94805, France; Gustave Roussy, Laboratoire de Vectorologie et Thérapeutiques Anticancéreuses, UMR 8203, Villejuif 94805, France; Gustave Roussy, Bioinformatics Platform, AMMICA, INSERM US23/CNRS UMS3655, Villejuif 94805, France
| | | | - Pierre Khoueiry
- American University of Beirut, Faculty of Medicine, Department of Biochemistry and Molecular Genetics, P.O. Box 11-0236 DTS 419-B, Bliss Street, Beirut, Lebanon
| | - Birgit Geoerger
- Université Paris-Sud 11, Laboratoire de Vectorologie et Thérapeutiques Anticancéreuses, UMR 8203, Villejuif 94805, France; CNRS, Villejuif, Laboratoire de Vectorologie et Thérapeutiques Anticancéreuses, UMR 8203, Villejuif 94805, France; Gustave Roussy, Laboratoire de Vectorologie et Thérapeutiques Anticancéreuses, UMR 8203, Villejuif 94805, France; Gustave Roussy, Department of Pediatric and Adolescent Oncology, Villejuif 94805, France
| | - Liliane Massaad-Massade
- Université Paris-Sud 11, Laboratoire de Vectorologie et Thérapeutiques Anticancéreuses, UMR 8203, Villejuif 94805, France; CNRS, Villejuif, Laboratoire de Vectorologie et Thérapeutiques Anticancéreuses, UMR 8203, Villejuif 94805, France; Gustave Roussy, Laboratoire de Vectorologie et Thérapeutiques Anticancéreuses, UMR 8203, Villejuif 94805, France.
| |
Collapse
|
41
|
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
|
42
|
Lacey A, Hedrick E, Cheng Y, Mohankumar K, Warren M, Safe S. Interleukin-24 (IL24) Is Suppressed by PAX3-FOXO1 and Is a Novel Therapy for Rhabdomyosarcoma. Mol Cancer Ther 2018; 17:2756-2766. [PMID: 30190424 DOI: 10.1158/1535-7163.mct-18-0118] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 05/08/2018] [Accepted: 08/29/2018] [Indexed: 12/17/2022]
Abstract
Alveolar rhabdomyosarcoma (ARMS) patients have a poor prognosis, and this is primarily due to overexpression of the oncogenic fusion protein PAX3-FOXO1. Results of RNA-sequencing studies show that PAX3-FOXO1 represses expression of interleukin-24 (IL24), and these two genes are inversely expressed in patient tumors. PAX3-FOXO1 also regulates histone deacetylase 5 (HDAC5) in ARMS cells, and results of RNA interference studies confirmed that PAX3-FOXO1-mediated repression of IL24 is HDAC5-dependent. Knockdown of PAX3-FOXO1 decreases ARMS cell proliferation, survival, and migration, and we also observed similar responses in cells after overexpression of IL24, consistent with results reported for this tumor suppressor-like cytokine in other solid tumors. We also observed in double knockdown studies that the inhibition of ARMS cell proliferation, survival, and migration after knockdown of PAX3-FOXO1 was significantly (>75%) reversed by knockdown of IL24. Adenoviral-expressed IL24 was directly injected into ARMS tumors in athymic nude mice, and this resulted in decreased tumor growth and weight. Because adenoviral IL24 has already successfully undergone phase I in clinical trials, this represents an alternative approach (alone and/or combination) for treating ARMS patients who currently undergo cytotoxic drug therapies.
Collapse
Affiliation(s)
- Alexandra Lacey
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas
| | - Erik Hedrick
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas
| | - Yating Cheng
- 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
| | - Melanie Warren
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas.
| |
Collapse
|
43
|
Wierdl M, Tsurkan L, Chi L, Hatfield MJ, Tollemar V, Bradley C, Chen X, Qu C, Potter PM. Targeting ALK in pediatric RMS does not induce antitumor activity in vivo. Cancer Chemother Pharmacol 2018; 82:251-263. [PMID: 29855693 PMCID: PMC6054567 DOI: 10.1007/s00280-018-3615-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 05/29/2018] [Indexed: 12/22/2022]
Abstract
PURPOSE The anaplastic lymphoma kinase (ALK) has been demonstrated to be a valid clinical target in diseases such as anaplastic large cell lymphoma and non-small cell lung cancer. Recent studies have indicated that ALK is overexpressed in pediatric rhabdomyosarcoma (RMS) and hence we hypothesized that this kinase may be a suitable candidate for therapeutic intervention in this tumor. METHODS We evaluated the expression of ALK in a panel of pediatric RMS cell lines and patient-derived xenografts (PDX), and sensitivity to ALK inhibitors was assessed both in vitro and in vivo. RESULTS Essentially, all RMS lines were sensitive to crizotinib, NVP-TAE684 or LDK-378 in vitro, and molecular analyses demonstrated inhibition of RMS cell proliferation following siRNA-mediated reduction of ALK expression. However, in vivo PDX studies using ALK kinase inhibitors demonstrated no antitumor activity when used as single agents or when combined with standard of care therapy (vincristine, actinomycin D and cyclophosphamide). More alarmingly, however, crizotinib actually accelerated the growth of these tumors in vivo. CONCLUSIONS While ALK appears to be a relevant target in RMS in vitro, targeting this kinase in vivo yields no therapeutic efficacy, warranting extreme caution when considering the use of these agents in pediatric RMS patients.
Collapse
Affiliation(s)
- Monika Wierdl
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105-2794, USA
| | - Lyudmila Tsurkan
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105-2794, USA
| | - Liying Chi
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105-2794, USA
| | - M Jason Hatfield
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105-2794, USA
| | - Viktor Tollemar
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105-2794, USA
| | - Cori Bradley
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105-2794, USA
| | - Xiang Chen
- Department of Computational Biology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105-2794, USA
| | - Chunxu Qu
- Department of Computational Biology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105-2794, USA
| | - Philip M Potter
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105-2794, USA.
| |
Collapse
|
44
|
Du J, Wang Y, Meng L, Liu Y, Pang Y, Cui W, Zhang L, Li Z, Liu Q, Shang H, Liu C, Li F. c-MET expression potentially contributes to the poor prognosis of rhabdomyosarcoma. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2018; 11:4083-4092. [PMID: 31949799 PMCID: PMC6962817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 06/29/2018] [Indexed: 06/10/2023]
Abstract
Rhabdomyosarcoma (RMS) is one of the most common soft-tissue sarcomas with a poor prognosis. c-MET is a prognostic biomarker associated with growth, proliferation, invasion, and metastasis in various carcinomas. In this study, we aim to investigate the expression of c-MET in RMS and its effect on the prognosis of patients with rhabdomyosarcoma. We performed immunohistochemistry and a quantitative real-time polymerase chain reaction (qRT-PCR) to determine the expression levels of c-MET proteins and mRNAs. Results indicated that the c-MET protein and mRNA expression levels in the RMS samples were significantly higher than those in normal controls (P<0.01 and P=0.0492). However, the correlation between c-MET expression and any other clinicopathological parameter and survival was not significant (P=0.837). Nevertheless, c-MET expression had a significant influence on the overall survival rates of patients with ERMS (χ2=9.673, P=0.002) and fusion gene-negative patients (χ2=5.400, P=0.020). These findings suggest that c-MET may serve as a promising biomarker capable of predicting poor prognosis in patients with RMS.
Collapse
Affiliation(s)
- Juan Du
- Department of Pathology, School of Medicine, Shihezi UniversityShihezi, Xinjiang, China
| | - Yuanyuan Wang
- Department of Pathology, School of Medicine, Shihezi UniversityShihezi, Xinjiang, China
| | - Lian Meng
- Department of Pathology, School of Medicine, Shihezi UniversityShihezi, Xinjiang, China
| | - Yang Liu
- Department of Pathology, School of Medicine, Shihezi UniversityShihezi, Xinjiang, China
| | - Yuwen Pang
- Department of Pathology, School of Medicine, Shihezi UniversityShihezi, Xinjiang, China
| | - Wenwen Cui
- Department of Pathology, School of Medicine, Shihezi UniversityShihezi, Xinjiang, China
| | - Liang Zhang
- Department of Pathology, School of Medicine, Shihezi UniversityShihezi, Xinjiang, China
| | - Zhenzhen Li
- Department of Pathology, School of Medicine, Shihezi UniversityShihezi, Xinjiang, China
| | - Qianqian Liu
- Department of Pathology, School of Medicine, Shihezi UniversityShihezi, Xinjiang, China
| | - Hao Shang
- Department of Pathology, School of Medicine, Shihezi UniversityShihezi, Xinjiang, China
| | - Chunxia Liu
- Department of Pathology, School of Medicine, Shihezi UniversityShihezi, Xinjiang, China
| | - Feng Li
- Department of Pathology, School of Medicine, Shihezi UniversityShihezi, Xinjiang, China
- Department of Pathology, Beijing Chaoyang Hospital, Capital Medical UniversityBeijing, China
| |
Collapse
|
45
|
Wachtel M, Schäfer BW. PAX3-FOXO1: Zooming in on an “undruggable” target. Semin Cancer Biol 2018; 50:115-123. [DOI: 10.1016/j.semcancer.2017.11.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 10/31/2017] [Accepted: 11/13/2017] [Indexed: 12/17/2022]
|
46
|
PAX3-FOXO1 drives miR-486-5p and represses miR-221 contributing to pathogenesis of alveolar rhabdomyosarcoma. Oncogene 2018; 37:1991-2007. [PMID: 29367756 PMCID: PMC5895609 DOI: 10.1038/s41388-017-0081-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 10/26/2017] [Accepted: 12/01/2017] [Indexed: 01/02/2023]
Abstract
Rhabdomyosarcoma is the most common soft-tissue sarcoma in childhood and histologically resembles developing skeletal muscle. Alveolar rhabdomyosarcoma (ARMS) is an aggressive subtype with a higher rate of metastasis and poorer prognosis. The majority of ARMS tumors (80%) harbor a PAX3-FOXO1 or less commonly a PAX7-FOXO1 fusion gene. The presence of either the PAX3-FOXO1 or PAX7-FOXO1 fusion gene foretells a poorer prognosis resulting in clinical re-classification as either fusion-positive (FP-RMS) or fusion-negative RMS (FN-RMS). The PAX3/7-FOXO1 fusion genes result in the production of a rogue transcription factors that drive FP-RMS pathogenesis and block myogenic differentiation. Despite knowing the molecular driver of FP-RMS, targeted therapies have yet to make an impact for patients, highlighting the need for a greater understanding of the molecular consequences of PAX3-FOXO1 and its target genes including microRNAs. Here we show FP-RMS patient-derived xenografts and cell lines display a distinct microRNA expression pattern. We utilized both loss- and gain-of function approaches in human cell lines with knockdown of PAX3-FOXO1 in FP-RMS cell lines and expression of PAX3-FOXO1 in human myoblasts and identified microRNAs both positively and negatively regulated by the PAX3-FOXO1 fusion protein. We demonstrate PAX3-FOXO1 represses miR-221/222 that functions as a tumor suppressing microRNA through the negative regulation of CCND2, CDK6, and ERBB3. In contrast, miR-486-5p is transcriptionally activated by PAX3-FOXO1 and promotes FP-RMS proliferation, invasion, and clonogenic growth. Inhibition of miR-486-5p in FP-RMS xenografts decreased tumor growth, illustrating a proof of principle for future therapeutic intervention. Therefore, PAX3-FOXO1 regulates key microRNAs that may represent novel therapeutic vulnerabilities in FP-RMS.
Collapse
|
47
|
Sugito N, Taniguchi K, Kuranaga Y, Ohishi M, Soga T, Ito Y, Miyachi M, Kikuchi K, Hosoi H, Akao Y. Cancer-Specific Energy Metabolism in Rhabdomyosarcoma Cells Is Regulated by MicroRNA. Nucleic Acid Ther 2017; 27:365-377. [DOI: 10.1089/nat.2017.0673] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Nobuhiko Sugito
- United Graduate School of Drug Discovery and Medical Information Science, Gifu University, Gifu, Japan
| | - Kohei Taniguchi
- Department of General and Gastroenterological Surgery, Osaka Medical College, Takatsuki, Japan
| | - Yuki Kuranaga
- United Graduate School of Drug Discovery and Medical Information Science, Gifu University, Gifu, Japan
| | - Maki Ohishi
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan
| | - Yuko Ito
- Division of Life Sciences, Department of Anatomy and Cell Biology, Osaka Medical College, Takatsuki, Japan
| | - Mitsuru Miyachi
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Ken Kikuchi
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hajime Hosoi
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yukihiro Akao
- United Graduate School of Drug Discovery and Medical Information Science, Gifu University, Gifu, Japan
| |
Collapse
|
48
|
Abstract
Sarcomas arise from primitive mesenchymal cells, which are classified, into two main groups: Bone and soft tissue sarcomas. We have searched all-important electronic databases including Google scholar and PubMed for the collection of latest literature pertaining to pediatric sarcomas. Latest literature confirmed that these tumors are relatively rare and represent only 1% of all malignancies but they have higher incidence in children. Pediatric sarcomas comprise about 13% of all pediatric malignancies and are ranked third in childhood cancers. The highest incidence rates are reported among rhabdomyosarcoma, osteosarcoma and Ewing's sarcomas in children. All of these neoplasms often display highly aggressive behavior with tendency to form metastases. Important globally used management avenues include surgery with systemic chemotherapy and have success rate of 70% at 5-years. Furthermore, in the cases of advanced stages, the prognosis is poor, chances of treatment failure and recurrence are quite high. Utilization of cancer stem cells is the latest approach with great potential in management of above pathological state. The present review article discuss all-important aspects of commonly found pediatric sarcomas throughout the world.
Collapse
Affiliation(s)
- Junhua Cao
- Department of Pediatric Internal Medicine, Xuzhou Children's Hospital, Xuzhou, Jiangsu 221002, P.R. China
| | - Qi An
- Department of Pediatric Internal Medicine, Xuzhou Children's Hospital, Xuzhou, Jiangsu 221002, P.R. China
| | - Lei Wang
- Department of Pediatric Internal Medicine, Xuzhou Children's Hospital, Xuzhou, Jiangsu 221002, P.R. China
| |
Collapse
|
49
|
Yu Y, Fu L, Wang S, Jin Y, Han S, Chu P, Lu J, Guo Y, He L, Ni X. Investigation of IGF2, IGFBP2 and p63 proteins in rhabdomyosarcoma tumors. Growth Horm IGF Res 2017; 33:17-22. [PMID: 28129571 DOI: 10.1016/j.ghir.2017.01.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 01/12/2017] [Accepted: 01/12/2017] [Indexed: 12/21/2022]
Abstract
UNLABELLED Many efforts have been made to address involvement of the insulin-like growth-factor (IGF) pathway in rhabdomyosarcoma (RMS) pathogenesis, but the actual role of IGF in RMS is still controversial. OBJECTIVE To investigate the implications of IGF2, IGFBP2 and p63 in RMS, and further explored their potential interaction. DESIGN A total of 114 specimens of RMS along with clinic-pathologic characteristics were collected from the year of 2003 to 2013. Protein abundance was detected by immunohistochemical staining, potential relationships between protein levels and clinic-pathological parameters were applied using correlation analysis. RESULTS The results showed positive correlation between IGFBP2 and p63 (r=0.271, p=0.003), suggesting that the interaction of IGFBP2 and p63 might account for the pathogenesis of RMS. In the subtype analysis, positive correlation was still found in embryonal rhabdomyosarcoma (ERMS, r=0.214, p=0.034) and alveolar rhabdomyosarcoma (ARMS, r=0.498, p=0.048). By focusing on the interaction of IGF pathway and p63, our results reveal additional signs to elucidate difference of pathogenesis and severity between ERMS and ARMS. CONCLUSIONS The present study provides novel evidence to elucidate RMS pathogenesis and may be beneficial to clinical diagnosis and therapy for RMS.
Collapse
Affiliation(s)
- Yongbo Yu
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Libing Fu
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Shen Wang
- Clinical Diagnostic Center, 302nd Hospital of the People's Liberation Army, Beijing, China
| | - Yaqiong Jin
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Shujing Han
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Ping Chu
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Jie Lu
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Yongli Guo
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Lejian He
- Department of Pathology, Beijing Children's Hospital, Capital Medical University, Beijing, China.
| | - Xin Ni
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing, China; Department of Otolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
50
|
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
|