1
|
Perotti D, O'Sullivan MJ, Walz AL, Davick J, Al-Saadi R, Benedetti DJ, Brzezinski J, Ciceri S, Cost NG, Dome JS, Drost J, Evageliou N, Furtwängler R, Graf N, Maschietto M, Mullen EA, Murphy AJ, Ortiz MV, van der Beek JN, Verschuur A, Wegert J, Williams R, Spreafico F, Geller JI, van den Heuvel-Eibrink MM, Hong AL. Hallmark discoveries in the biology of non-Wilms tumour childhood kidney cancers. Nat Rev Urol 2025:10.1038/s41585-024-00993-6. [PMID: 39881003 DOI: 10.1038/s41585-024-00993-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/17/2024] [Indexed: 01/31/2025]
Abstract
Approximately 20% of paediatric and adolescent/young adult patients with renal tumours are diagnosed with non-Wilms tumour, a broad heterogeneous group of tumours that includes clear-cell sarcoma of the kidney, congenital mesoblastic nephroma, malignant rhabdoid tumour of the kidney, renal-cell carcinoma, renal medullary carcinoma and other rare histologies. The differential diagnosis of these tumours dates back many decades, when these pathologies were identified initially through clinicopathological observation of entities with outcomes that diverged from Wilms tumour, corroborated with immunohistochemistry and molecular cytogenetics and, subsequently, through next-generation sequencing. These advances enabled near-definitive recognition of different tumours and risk stratification of patients. In parallel, the generation of new renal-tumour models of some of these pathologies including cell lines, organoids, xenografts and genetically engineered mouse models improved our understanding of the development of these tumours and have facilitated the identification of new therapeutic targets. Despite these many achievements, paediatric and adolescent/young adult patients continue to die from such rare cancers at higher rates than patients with Wilms tumour. Thus, international coordinated efforts are needed to answer unresolved questions and improve outcomes.
Collapse
Affiliation(s)
- Daniela Perotti
- Predictive Medicine: Molecular Bases of Genetic Risk, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Maureen J O'Sullivan
- Histology Laboratory, Children's Health Ireland at Crumlin, Dublin, Ireland
- Histopathology, School of Medicine, Trinity College, Dublin, Ireland
- Departments of Histopathology and Paediatrics, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Amy L Walz
- Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jonathan Davick
- University of Iowa Hospitals and Clinics Stead Family Children's Hospital, Carver College of Medicine, Iowa City, IA, USA
| | - Reem Al-Saadi
- UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Daniel J Benedetti
- Division of Pediatric Hematology/Oncology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jack Brzezinski
- Division of Haematology/Oncology, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Paediatrics, University of Toronto, Toronto, Ontario, Canada
| | - Sara Ciceri
- Predictive Medicine: Molecular Bases of Genetic Risk, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Nicholas G Cost
- Department of Surgery, Division of Urology, University of Colorado School of Medicine and the Surgical Oncology Program at Children's Hospital Colorado, Denver, CO, USA
| | - Jeffrey S Dome
- Division of Oncology, Center for Cancer and Blood Disorders, Children's National Hospital, Washington, DC, USA
- Department of Pediatrics, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Jarno Drost
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
| | | | - Rhoikos Furtwängler
- Pediatric Hematology and Oncology, Children's Hospital, Inselspital Bern University, Bern, Switzerland
- Childhood Renal Tumour Center Saarland University, Homburg, Germany
| | - Norbert Graf
- Department Paediatric Oncology & Hematology, Saarland University, Homburg, Germany
| | | | - Elizabeth A Mullen
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
| | - Andrew J Murphy
- St. Jude Children's Research Hospital Memphis, Memphis, TN, USA
| | | | - Justine N van der Beek
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- University Medical Center Utrecht, Utrecht, the Netherlands
| | - Arnauld Verschuur
- Department of Pediatric Hematology and Oncology, Hôpital d'Enfants de la Timone, APHM, Marseille, France
| | - Jenny Wegert
- Theodor-Boveri-Institute/Biocenter, Developmental Biochemistry, Wuerzburg University, Wuerzburg, Germany
| | - Richard Williams
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
- Section of Genetics and Genomics, Faculty of Medicine, Imperial College London, London, UK
| | - Filippo Spreafico
- Paediatric Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - James I Geller
- Division of Oncology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| | | | - Andrew L Hong
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA.
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA.
| |
Collapse
|
2
|
Verrelle P, Gestraud P, Poyer F, Soria A, Tessier S, Lescure A, Anthony E, Corbé M, Heinrich S, Beauvineau C, Chaput L, Granzhan A, Piguel S, Perez F, Teulade-Fichou MP, Megnin-Chanet F, Del Nery E. Integrated High-Throughput Screening and Large-Scale Isobolographic Analysis to Accelerate the Discovery of Radiosensitizers With Greater Selectivity for Cancer Cells. Int J Radiat Oncol Biol Phys 2024; 118:1294-1307. [PMID: 37778425 DOI: 10.1016/j.ijrobp.2023.09.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 09/15/2023] [Accepted: 09/19/2023] [Indexed: 10/03/2023]
Abstract
PURPOSE High-throughput screening (HTS) platforms have been widely used to identify candidate anticancer drugs and drug-drug combinations; however, HTS-based identification of new drug-ionizing radiation (IR) combinations has rarely been reported. Herein, we developed an integrated approach including cell-based HTS and computational large-scale isobolographic analysis to accelerate the identification of radiosensitizing compounds acting strongly and more specifically on cancer cells. METHODS AND MATERIALS In a 384-well plate format, 160 compounds likely to interfere with the cell response to radiation were screened on human glioblastoma (U251-MG) and cervix carcinoma (ME-180) cell lines, as well as on normal fibroblasts (CCD-19Lu). After drug exposure, cells were irradiated or not and short-term cell survival was assessed by high-throughput cell microscopy. Computational large-scale dose-response and isobolographic approach were used to identify promising synergistic drugs radiosensitizing cancer cells rather than normal cells. Synergy of a promising compound was confirmed on ME-180 cells by an independent 96-well assay protocol, and finally, by the gold-standard colony forming assay. RESULTS We retained 4 compounds synergistic at 2 isoeffects in U251-MG and ME-180 cell lines and 11 compounds synergistically effective in only one cancer cell line. Among these 15 promising radiosensitizers, 5 compounds showed limited toxicity combined or not with IR on normal fibroblasts. CONCLUSIONS Overall, this study demonstrated that HTS chemoradiation screening together with large-scale computational analysis is an efficient tool to identify synergistic drug-IR combinations, with concomitant assessment of unwanted toxicity on normal fibroblasts. It sparks expectations to accelerate the discovery of highly desired agents improving the therapeutic index of radiation therapy.
Collapse
Affiliation(s)
- Pierre Verrelle
- Radiation Oncology Department, Institut Curie Hospital, Paris, France; Chemistry and Modelisation for the Biology of Cancer, CNRS UMR9187, INSERM U1196, Institut Curie, Université Paris Saclay, 91405 Orsay, France.
| | - Pierre Gestraud
- Chemistry and Modelisation for the Biology of Cancer, CNRS UMR9187, INSERM U1196, Institut Curie, Université Paris Saclay, 91405 Orsay, France
| | - Florent Poyer
- Chemistry and Modelisation for the Biology of Cancer, CNRS UMR9187, INSERM U1196, Institut Curie, Université Paris Saclay, 91405 Orsay, France
| | - Adèle Soria
- Biophenics High-Content Screening Laboratory, Department of Translational Research, PSL Research University, PICT-IBiSa, Institut Curie Research Center, Paris, France
| | - Sarah Tessier
- Biophenics High-Content Screening Laboratory, Department of Translational Research, PSL Research University, PICT-IBiSa, Institut Curie Research Center, Paris, France
| | - Aurianne Lescure
- Biophenics High-Content Screening Laboratory, Department of Translational Research, PSL Research University, PICT-IBiSa, Institut Curie Research Center, Paris, France
| | - Elodie Anthony
- Biophenics High-Content Screening Laboratory, Department of Translational Research, PSL Research University, PICT-IBiSa, Institut Curie Research Center, Paris, France
| | - Maxime Corbé
- Biophenics High-Content Screening Laboratory, Department of Translational Research, PSL Research University, PICT-IBiSa, Institut Curie Research Center, Paris, France
| | - Sophie Heinrich
- Experimental Radiotherapy Platform (RadeXp), Translational Research Department, Institut Curie, Orsay, France; Inserm U1021-CNRS UMR 3347, Institut Curie, Paris Saclay University
| | - Claire Beauvineau
- Bioinformatics and Computational Systems Biology of Cancer, PSL Research University, Mines Paris Tech, INSERM U900, Paris, France
| | - Ludovic Chaput
- Bioinformatics and Computational Systems Biology of Cancer, PSL Research University, Mines Paris Tech, INSERM U900, Paris, France
| | - Anton Granzhan
- Bioinformatics and Computational Systems Biology of Cancer, PSL Research University, Mines Paris Tech, INSERM U900, Paris, France
| | - Sandrine Piguel
- Bioinformatics and Computational Systems Biology of Cancer, PSL Research University, Mines Paris Tech, INSERM U900, Paris, France; BioCIS UMR8076, Université Paris-Saclay, Faculté de Pharmacie, Orsay, France
| | - Franck Perez
- Biophenics High-Content Screening Laboratory, Department of Translational Research, PSL Research University, PICT-IBiSa, Institut Curie Research Center, Paris, France; Cell Biology and Cancer UMR144, Institut Curie, PSL Research University, Paris, France
| | - Marie-Paule Teulade-Fichou
- Chemistry and Modelisation for the Biology of Cancer, CNRS UMR9187, INSERM U1196, Institut Curie, Université Paris Saclay, 91405 Orsay, France
| | - Frédérique Megnin-Chanet
- Bioinformatics and Computational Systems Biology of Cancer, PSL Research University, Mines Paris Tech, INSERM U900, Paris, France
| | - Elaine Del Nery
- Biophenics High-Content Screening Laboratory, Department of Translational Research, PSL Research University, PICT-IBiSa, Institut Curie Research Center, Paris, France.
| |
Collapse
|
3
|
Hua T, Xue Y, Sarker DB, Kiran S, Li Y, Sang QXA. Modeling human brain rhabdoid tumor by inactivating tumor suppressor genes in induced pluripotent stem cells. Bioact Mater 2024; 31:136-150. [PMID: 37637078 PMCID: PMC10448240 DOI: 10.1016/j.bioactmat.2023.08.009] [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: 04/24/2023] [Revised: 08/02/2023] [Accepted: 08/08/2023] [Indexed: 08/29/2023] Open
Abstract
Atypical teratoid/rhabdoid tumor (ATRT) is a rare childhood malignancy that originates in the central nervous system. Over ninety-five percent of ATRT patients have biallelic inactivation of the tumor suppressor gene SMARCB1. ATRT has no standard treatment, and a major limiting factor in therapeutic development is the lack of reliable ATRT models. We employed CRISPR/Cas9 gene-editing technology to knock out SMARCB1 and TP53 genes in human episomal induced pluripotent stem cells (Epi-iPSCs), followed by brief neural induction, to generate an ATRT-like model. The dual knockout Epi-iPSCs retained their stemness with the capacity to differentiate into three germ layers. High expression of OCT4 and NANOG in neurally induced knockout spheroids was comparable to that in two ATRT cell lines. Beta-catenin protein expression was higher in SMARCB1-deficient cells and spheroids than in normal Epi-iPSC-derived spheroids. Nucleophosmin, Osteopontin, and Ki-67 proteins were also expressed by the SMARCB1-deficient spheroids. In summary, the tumor model resembled embryonal features of ATRT and expressed ATRT biomarkers at mRNA and protein levels. Ribociclib, PTC-209, and the combination of clofilium tosylate and pazopanib decreased the viability of the ATRT-like cells. This disease modeling scheme may enable the establishment of individualized tumor models with patient-specific mutations and facilitate high-throughput drug testing.
Collapse
Affiliation(s)
- Timothy Hua
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, FL, 32306-4390, USA
| | - Yu Xue
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, FL, 32306-4390, USA
| | - Drishty B. Sarker
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, FL, 32306-4390, USA
| | - Sonia Kiran
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, FL, 32306-4390, USA
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, 32310-6046, USA
- Institute of Molecular Biophysics, Florida State University, Tallahassee, FL, 32306-4380, USA
| | - Qing-Xiang Amy Sang
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, FL, 32306-4390, USA
- Institute of Molecular Biophysics, Florida State University, Tallahassee, FL, 32306-4380, USA
| |
Collapse
|
4
|
Vasev YA, Nasibullina ER, Makarov AS, Uchuskin MG. Interrupted Furan-Yne Cyclization: Access to Unsaturated Dicarbonyl Compounds and Their Subsequent Transformation into Functionalized Pyridazines. Org Lett 2023; 25:7780-7785. [PMID: 37862046 DOI: 10.1021/acs.orglett.3c02794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2023]
Abstract
The key carbenoid intermediate of transition-metal-catalyzed furan-yne cyclization in Hashmi phenol synthesis could be efficiently intercepted with water under the developed reaction conditions in order to provide access to functionalized unsaturated dicarbonyl compounds that might serve as convenient precursors for the straightforward synthesis of annulated pyridazines.
Collapse
Affiliation(s)
- Yury A Vasev
- Department of Chemistry, Perm State University, Bukireva 15, 614990 Perm, Russia
| | | | - Anton S Makarov
- Department of Chemistry, Perm State University, Bukireva 15, 614990 Perm, Russia
| | - Maxim G Uchuskin
- Department of Chemistry, Perm State University, Bukireva 15, 614990 Perm, Russia
| |
Collapse
|
5
|
Fevre R, Mary G, Vertti-Quintero N, Durand A, Tomasi RFX, Del Nery E, Baroud CN. Combinatorial drug screening on 3D Ewing sarcoma spheroids using droplet-based microfluidics. iScience 2023; 26:106651. [PMID: 37168549 PMCID: PMC10165258 DOI: 10.1016/j.isci.2023.106651] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 02/02/2023] [Accepted: 04/05/2023] [Indexed: 05/13/2023] Open
Abstract
Culturing and screening cells in microfluidics, particularly in three-dimensional formats, has the potential to impact diverse areas from fundamental biology to cancer precision medicine. Here, we use a platform based on anchored droplets for drug screening. The response of spheroids of Ewing sarcoma (EwS) A673 cells to simultaneous or sequential combinations of etoposide and cisplatin was evaluated. This was done by culturing spheroids of EwS cells inside 500 nL droplets then merging them with secondary droplets containing fluorescent-barcoded drugs at different concentrations. Differences in EwS spheroid growth and viability were measured by microscopy. After drug exposure such measurements enabled estimation of their IC50 values, which were in agreement with values obtained in standard multiwell plates. Then, synergistic drug combination was evaluated. Sequential combination treatment of EwS with etoposide applied 24 h before cisplatin resulted in amplified synergistic effect. As such, droplet-based microfluidics offers the modularity required for evaluation of drug combinations.
Collapse
Affiliation(s)
- Romain Fevre
- Laboratoire d’ Hydrodynamique (LadHyX), CNRS, EcolePolytechnique, InstitutPolytechnique de Paris, 91128 Palaiseau, France
- Institut Pasteur, Université Paris Cité, Physical microfluidics and Bioengineering, 25-28 Rue du Dr. Roux, 75015 Paris, France
| | - Gaëtan Mary
- Okomera, iPEPS, the HealthTech Hub, Paris Brain Institute, HôpitalPitiéSalpêtrière, 75013 Paris, France
| | - Nadia Vertti-Quintero
- Institut Pasteur, Université Paris Cité, Physical microfluidics and Bioengineering, 25-28 Rue du Dr. Roux, 75015 Paris, France
| | - Aude Durand
- Institut Pasteur, Université Paris Cité, Physical microfluidics and Bioengineering, 25-28 Rue du Dr. Roux, 75015 Paris, France
| | - Raphaël F.-X. Tomasi
- Laboratoire d’ Hydrodynamique (LadHyX), CNRS, EcolePolytechnique, InstitutPolytechnique de Paris, 91128 Palaiseau, France
- Institut Pasteur, Université Paris Cité, Physical microfluidics and Bioengineering, 25-28 Rue du Dr. Roux, 75015 Paris, France
- Okomera, iPEPS, the HealthTech Hub, Paris Brain Institute, HôpitalPitiéSalpêtrière, 75013 Paris, France
| | - Elaine Del Nery
- Biophenics High-Content Screening Laboratory, Translational Research Department, PICT-IBiSA, Institut Curie, PSL Research University, 75005 Paris, France
- Corresponding author
| | - Charles N. Baroud
- Laboratoire d’ Hydrodynamique (LadHyX), CNRS, EcolePolytechnique, InstitutPolytechnique de Paris, 91128 Palaiseau, France
- Institut Pasteur, Université Paris Cité, Physical microfluidics and Bioengineering, 25-28 Rue du Dr. Roux, 75015 Paris, France
- Corresponding author
| |
Collapse
|
6
|
Zhang C, Li H. Molecular targeted therapies for pediatric atypical teratoid/rhabdoid tumors. Pediatr Investig 2022; 6:111-122. [PMID: 35774526 PMCID: PMC9218972 DOI: 10.1002/ped4.12325] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 04/26/2022] [Indexed: 01/01/2023] Open
Abstract
Atypical teratoid/rhabdoid tumors (AT/RTs) are lethal central nervous system tumors, which are primarily diagnosed in infants. Current treatments for AT/RTs include surgery, radiotherapy, and chemotherapy; these treatments have poor prognoses and challenging side effects. The pivotal genetic event in AT/RT pathogenesis comprises the inactivation of SMARCB1 or SMARCA4. Recent epigenetic studies have demonstrated mutual and subtype-specific epigenetic derangements that drive tumorigenesis; the exploitation of these potential targets might improve the dismal treatment outcomes of AT/RTs. This review aims to summarize the literature concerning targeted molecular therapies for pediatric AT/RTs.
Collapse
Affiliation(s)
- Chang Zhang
- Department of NeurosurgeryChildren's Hospital of Fudan UniversityShanghaiChina
| | - Hao Li
- Department of NeurosurgeryChildren's Hospital of Fudan UniversityShanghaiChina
| |
Collapse
|
7
|
Buchou C, Laud-Duval K, van der Ent W, Grossetête S, Zaidi S, Gentric G, Corbé M, Müller K, Del Nery E, Surdez D, Delattre O. Upregulation of the Mevalonate Pathway through EWSR1-FLI1/EGR2 Regulatory Axis Confers Ewing Cells Exquisite Sensitivity to Statins. Cancers (Basel) 2022; 14:2327. [PMID: 35565457 PMCID: PMC9100622 DOI: 10.3390/cancers14092327] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/02/2022] [Accepted: 05/05/2022] [Indexed: 02/04/2023] Open
Abstract
Ewing sarcoma (EwS) is an aggressive primary bone cancer in children and young adults characterized by oncogenic fusions between genes encoding FET-RNA-binding proteins and ETS transcription factors, the most frequent fusion being EWSR1-FLI1. We show that EGR2, an Ewing-susceptibility gene and an essential direct target of EWSR1-FLI1, directly regulates the transcription of genes encoding key enzymes of the mevalonate (MVA) pathway. Consequently, Ewing sarcoma is one of the tumors that expresses the highest levels of mevalonate pathway genes. Moreover, genome-wide screens indicate that MVA pathway genes constitute major dependencies of Ewing cells. Accordingly, the statin inhibitors of HMG-CoA-reductase, a rate-limiting enzyme of the MVA pathway, demonstrate cytotoxicity in EwS. Statins induce increased ROS and lipid peroxidation levels, as well as decreased membrane localization of prenylated proteins, such as small GTP proteins. These metabolic effects lead to an alteration in the dynamics of S-phase progression and to apoptosis. Statin-induced effects can be rescued by downstream products of the MVA pathway. Finally, we further show that statins impair tumor growth in different Ewing PDX models. Altogether, the data show that statins, which are off-patent, well-tolerated, and inexpensive compounds, should be strongly considered in the therapeutic arsenal against this deadly childhood disease.
Collapse
Affiliation(s)
- Charlie Buchou
- INSERM U830, Équipe Labellisée LNCC, Diversity & Plasticity of Childhood Tumors Laboratory, PSL Research University, SIREDO Oncology Center, Institut Curie Research Center, 26 rue d’ULM, 75005 Paris, France; (C.B.); (K.L.-D.); (W.v.d.E.); (S.G.); (S.Z.); (D.S.)
| | - Karine Laud-Duval
- INSERM U830, Équipe Labellisée LNCC, Diversity & Plasticity of Childhood Tumors Laboratory, PSL Research University, SIREDO Oncology Center, Institut Curie Research Center, 26 rue d’ULM, 75005 Paris, France; (C.B.); (K.L.-D.); (W.v.d.E.); (S.G.); (S.Z.); (D.S.)
| | - Wietske van der Ent
- INSERM U830, Équipe Labellisée LNCC, Diversity & Plasticity of Childhood Tumors Laboratory, PSL Research University, SIREDO Oncology Center, Institut Curie Research Center, 26 rue d’ULM, 75005 Paris, France; (C.B.); (K.L.-D.); (W.v.d.E.); (S.G.); (S.Z.); (D.S.)
| | - Sandrine Grossetête
- INSERM U830, Équipe Labellisée LNCC, Diversity & Plasticity of Childhood Tumors Laboratory, PSL Research University, SIREDO Oncology Center, Institut Curie Research Center, 26 rue d’ULM, 75005 Paris, France; (C.B.); (K.L.-D.); (W.v.d.E.); (S.G.); (S.Z.); (D.S.)
| | - Sakina Zaidi
- INSERM U830, Équipe Labellisée LNCC, Diversity & Plasticity of Childhood Tumors Laboratory, PSL Research University, SIREDO Oncology Center, Institut Curie Research Center, 26 rue d’ULM, 75005 Paris, France; (C.B.); (K.L.-D.); (W.v.d.E.); (S.G.); (S.Z.); (D.S.)
| | - Géraldine Gentric
- INSERM U830, Équipe Labellisée LNCC, Stress and Cancer Laboratory, PSL Research University, Institut Curie Research Center, 26 rue d’ULM, 75005 Paris, France;
| | - Maxime Corbé
- Department of Translational Research, The Biophenics High-Content Screening Laboratory, PSL Research University, Institut Curie Research Center, 26 rue d’ULM, 75005 Paris, France; (M.C.); (K.M.); (E.D.N.)
| | - Kévin Müller
- Department of Translational Research, The Biophenics High-Content Screening Laboratory, PSL Research University, Institut Curie Research Center, 26 rue d’ULM, 75005 Paris, France; (M.C.); (K.M.); (E.D.N.)
| | - Elaine Del Nery
- Department of Translational Research, The Biophenics High-Content Screening Laboratory, PSL Research University, Institut Curie Research Center, 26 rue d’ULM, 75005 Paris, France; (M.C.); (K.M.); (E.D.N.)
| | - Didier Surdez
- INSERM U830, Équipe Labellisée LNCC, Diversity & Plasticity of Childhood Tumors Laboratory, PSL Research University, SIREDO Oncology Center, Institut Curie Research Center, 26 rue d’ULM, 75005 Paris, France; (C.B.); (K.L.-D.); (W.v.d.E.); (S.G.); (S.Z.); (D.S.)
- Balgrist University Hospital, University of Zurich, Zurich, Forchstrasse 340, 8008 Zürich, Switzerland
| | - Olivier Delattre
- INSERM U830, Équipe Labellisée LNCC, Diversity & Plasticity of Childhood Tumors Laboratory, PSL Research University, SIREDO Oncology Center, Institut Curie Research Center, 26 rue d’ULM, 75005 Paris, France; (C.B.); (K.L.-D.); (W.v.d.E.); (S.G.); (S.Z.); (D.S.)
| |
Collapse
|
8
|
Nemes K, Johann PD, Tüchert S, Melchior P, Vokuhl C, Siebert R, Furtwängler R, Frühwald MC. Current and Emerging Therapeutic Approaches for Extracranial Malignant Rhabdoid Tumors. Cancer Manag Res 2022; 14:479-498. [PMID: 35173482 PMCID: PMC8841298 DOI: 10.2147/cmar.s289544] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 01/11/2022] [Indexed: 12/12/2022] Open
Abstract
Extracranial malignant rhabdoid tumors (extracranial MRT) are rare, highly aggressive malignancies affecting mainly infants and children younger than 3 years. Common anatomic sites comprise the kidneys (RTK – rhabdoid tumor of kidney) and other soft tissues (eMRT – extracranial, extrarenal malignant rhabdoid tumor). The genetic origin of these diseases is linked to biallelic pathogenic variants in the genes SMARCB1, or rarely SMARCA4, encoding subunits of the SWI/SNF chromatin-remodeling complex. Even if extracranial MRT seem to be quite homogeneous, recent epigenome analyses reveal a certain degree of epigenetic heterogeneity. Use of intensified therapies has modestly improved survival for extracranial MRT. Patients at standard risk profit from conventional therapies; most high-risk patients still experience a dismal course and often therapy resistance. Discoveries of clinical and molecular hallmarks and the exploration of experimental therapeutic approaches open exciting perspectives for clinical and molecularly stratified experimental treatment approaches. To ultimately improve the outcome of patients with extracranial MRTs, they need to be characterized and stratified clinically and molecularly. High-risk patients need novel therapeutic approaches including selective experimental agents in phase I/II clinical trials.
Collapse
Affiliation(s)
- Karolina Nemes
- Paediatrics and Adolescent Medicine, Swabian Children's Cancer Center, University Medical Center Augsburg, Augsburg, Germany
| | - Pascal D Johann
- Paediatrics and Adolescent Medicine, Swabian Children's Cancer Center, University Medical Center Augsburg, Augsburg, Germany.,Division of Pediatric Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefanie Tüchert
- Department of Diagnostic and Interventional Radiology, University Hospital Augsburg, Augsburg, Germany
| | - Patrick Melchior
- Department of Radiation Oncology, University of Saarland, Homburg, Germany
| | - Christian Vokuhl
- Section of Pediatric Pathology, Department of Pathology, University Hospital Bonn, Bonn, Germany
| | - Reiner Siebert
- Institute of Human Genetics, Ulm University & Ulm University Medical Center, Ulm, Germany
| | - Rhoikos Furtwängler
- Department of Pediatric Hematology and Oncology, University of Saarland, Homburg, Germany
| | - Michael C Frühwald
- Paediatrics and Adolescent Medicine, Swabian Children's Cancer Center, University Medical Center Augsburg, Augsburg, Germany
| |
Collapse
|
9
|
Functional Therapeutic Target Validation Using Pediatric Zebrafish Xenograft Models. Cancers (Basel) 2022; 14:cancers14030849. [PMID: 35159116 PMCID: PMC8834194 DOI: 10.3390/cancers14030849] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/29/2022] [Accepted: 02/03/2022] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Despite the major progress of precision and personalized oncology, a significant therapeutic benefit is only achieved in cases with directly druggable genetic alterations. This highlights the need for additional methods that reliably predict each individual patient’s response in a clinically meaningful time, e.g., through ex vivo functional drug screen profiling. Moreover, patient-derived xenograft (PDX) models are more predictive than cell culture studies, as they reconstruct cell–cell and cell–extracellular matrix (ECM) interactions and consider the tumor microenvironment, drug metabolism and toxicities. Zebrafish PDXs (zPDX) are nowadays emerging as a fast model allowing for multiple drugs to be tested at the same time in a clinically relevant time window. Here, we show that functional drug response profiling of zPDX from primary material obtained through the INdividualized Therapy FOr Relapsed Malignancies in Childhood (INFORM) pediatric precision oncology pipeline provides additional key information for personalized precision oncology. Abstract The survival rate among children with relapsed tumors remains poor, due to tumor heterogeneity, lack of directly actionable tumor drivers and multidrug resistance. Novel personalized medicine approaches tailored to each tumor are urgently needed to improve cancer treatment. Current pediatric precision oncology platforms, such as the INFORM (INdividualized Therapy FOr Relapsed Malignancies in Childhood) study, reveal that molecular profiling of tumor tissue identifies targets associated with clinical benefit in a subgroup of patients only and should be complemented with functional drug testing. In such an approach, patient-derived tumor cells are exposed to a library of approved oncological drugs in a physiological setting, e.g., in the form of animal avatars injected with patient tumor cells. We used molecularly fully characterized tumor samples from the INFORM study to compare drug screen results of individual patient-derived cell models in functional assays: (i) patient-derived spheroid cultures within a few days after tumor dissociation; (ii) tumor cells reisolated from the corresponding mouse PDX; (iii) corresponding long-term organoid-like cultures and (iv) drug evaluation with the corresponding zebrafish PDX (zPDX) model. Each model had its advantage and complemented the others for drug hit and drug combination selection. Our results provide evidence that in vivo zPDX drug screening is a promising add-on to current functional drug screening in precision medicine platforms.
Collapse
|
10
|
Davidson K, Grevitt P, Contreras-Gerenas MF, Bridge KS, Hermida M, Shah KM, Mardakheh FK, Stubbs M, Burke R, Casado P, Cutillas PR, Martin SA, Sharp TV. Targeted therapy for LIMD1-deficient non-small cell lung cancer subtypes. Cell Death Dis 2021; 12:1075. [PMID: 34764236 PMCID: PMC8586256 DOI: 10.1038/s41419-021-04355-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 10/13/2021] [Accepted: 10/25/2021] [Indexed: 12/15/2022]
Abstract
An early event in lung oncogenesis is loss of the tumour suppressor gene LIMD1 (LIM domains containing 1); this encodes a scaffold protein, which suppresses tumorigenesis via a number of different mechanisms. Approximately 45% of non-small cell lung cancers (NSCLC) are deficient in LIMD1, yet this subtype of NSCLC has been overlooked in preclinical and clinical investigations. Defining therapeutic targets in these LIMD1 loss-of-function patients is difficult due to a lack of 'druggable' targets, thus alternative approaches are required. To this end, we performed the first drug repurposing screen to identify compounds that confer synthetic lethality with LIMD1 loss in NSCLC cells. PF-477736 was shown to selectively target LIMD1-deficient cells in vitro through inhibition of multiple kinases, inducing cell death via apoptosis. Furthermore, PF-477736 was effective in treating LIMD1-/- tumours in subcutaneous xenograft models, with no significant effect in LIMD1+/+ cells. We have identified a novel drug tool with significant preclinical characterisation that serves as an excellent candidate to explore and define LIMD1-deficient cancers as a new therapeutic subgroup of critical unmet need.
Collapse
Affiliation(s)
- Kathryn Davidson
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M6 BQ, UK
| | - Paul Grevitt
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M6 BQ, UK
| | - Maria F Contreras-Gerenas
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M6 BQ, UK
| | - Katherine S Bridge
- York Biomedical Research Institute, University of York, Wentworth Way, York, YO10 5DD, UK
| | - Miguel Hermida
- Department of Bioengineering, Imperial College, London, UK
| | - Kunal M Shah
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M6 BQ, UK
| | - Faraz K Mardakheh
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M6 BQ, UK
| | - Mark Stubbs
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, 15 Cotswold Road, Sutton, SM2 5NG, UK
| | - Rosemary Burke
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, 15 Cotswold Road, Sutton, SM2 5NG, UK
| | - Pedro Casado
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M6 BQ, UK
| | - Pedro R Cutillas
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M6 BQ, UK
| | - Sarah A Martin
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M6 BQ, UK.
| | - Tyson V Sharp
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M6 BQ, UK.
| |
Collapse
|
11
|
Epigenetic remodelling upon FGFR inhibition. Nat Cell Biol 2021; 23:1115-1116. [PMID: 34737444 DOI: 10.1038/s41556-021-00782-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
12
|
Harttrampf AC, da Costa MEM, Renoult A, Daudigeos-Dubus E, Geoerger B. Histone deacetylase inhibitor panobinostat induces antitumor activity in epithelioid sarcoma and rhabdoid tumor by growth factor receptor modulation. BMC Cancer 2021; 21:833. [PMID: 34281526 PMCID: PMC8290558 DOI: 10.1186/s12885-021-08579-w] [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: 12/18/2020] [Accepted: 07/10/2021] [Indexed: 01/06/2023] Open
Abstract
Background Epithelioid sarcomas and rhabdoid tumors are rare, aggressive malignancies with poor prognosis. Both are characterized by INI1 alterations and deregulation of growth factor receptors albeit their interaction has not been elucidated. Methods In this study, we investigated the activity of a panel of epigenetic modulators and receptor tyrosine kinase inhibitors in vitro on respective cell lines as well as on primary patient-derived epithelioid sarcoma cells, and in vivo on xenografted mice. Focusing on histone deacetylase (HDAC) inhibitors, we studied the mechanism of action of this class of agents, its effect on growth factor receptor regulation, and changes in epithelial-to-mesenchymal transition by using cell- and RT-qPCR-based assays. Results Pan-HDAC inhibitor panobinostat exhibited potent anti-proliferative activity at low nanomolar concentrations in A204 rhabdoid tumor, and VAESBJ/GRU1 epithelioid sarcoma cell lines, strongly induced apoptosis, and resulted in significant tumor growth inhibition in VAESBJ xenografts. It differentially regulated EGFR, FGFR1 and FGFR2, leading to downregulation of EGFR in epithelioid sarcoma and to mesenchymal-to-epithelial transition whereas in rhabdoid tumor cells, EGFR was strongly upregulated and reinforced the mesenchymal phenotype. All three cell lines were rendered more susceptible towards combination with EGFF inhibitor erlotinib, further enhancing apoptosis. Conclusions HDAC inhibitors exhibit significant anticancer activity due to their multifaceted actions on cytotoxicity, differentiation and drug sensitization. Our data suggest that the tailored, tissue-specific combination of HDAC inhibitors with therapeutics which target cellular salvage mechanisms might increase their therapeutic relevance. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-08579-w.
Collapse
Affiliation(s)
- Anne Catherine Harttrampf
- Gustave Roussy Cancer Center, INSERM U1015, Université Paris-Saclay, Villejuif, France.,Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Center, 114 Rue Edouard Vaillant, 94805, Villejuif, France
| | | | - Aline Renoult
- Gustave Roussy Cancer Center, INSERM U1015, Université Paris-Saclay, Villejuif, France.,Present address: Institute of Research in Immunology and Cancer, Dr Trang Hoang Laboratory, Université de Montréal, Montreal, Québec, Canada
| | - Estelle Daudigeos-Dubus
- Gustave Roussy Cancer Center, INSERM U1015, Université Paris-Saclay, Villejuif, France.,Present address: AP-HP Nord, DMU Neurosciences, Service de Neurologie, FHU NeuroVasc, Université de Paris, Paris, France
| | - Birgit Geoerger
- Gustave Roussy Cancer Center, INSERM U1015, Université Paris-Saclay, Villejuif, France. .,Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Center, 114 Rue Edouard Vaillant, 94805, Villejuif, France.
| |
Collapse
|
13
|
Napolitano A, Ostler AE, Jones RL, Huang PH. Fibroblast Growth Factor Receptor (FGFR) Signaling in GIST and Soft Tissue Sarcomas. Cells 2021; 10:cells10061533. [PMID: 34204560 PMCID: PMC8235236 DOI: 10.3390/cells10061533] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 06/11/2021] [Accepted: 06/15/2021] [Indexed: 12/20/2022] Open
Abstract
Sarcomas are a heterogeneous group of rare malignancies originating from mesenchymal tissues with limited therapeutic options. Recently, alterations in components of the fibroblast growth factor receptor (FGFR) signaling pathway have been identified in a range of different sarcoma subtypes, most notably gastrointestinal stromal tumors, rhabdomyosarcomas, and liposarcomas. These alterations include genetic events such as translocations, mutations, and amplifications as well as transcriptional overexpression. Targeting FGFR has therefore been proposed as a novel potential therapeutic approach, also in light of the clinical activity shown by multi-target tyrosine kinase inhibitors in specific subtypes of sarcomas. Despite promising preclinical evidence, thus far, clinical trials have enrolled very few sarcoma patients and the efficacy of selective FGFR inhibitors appears relatively low. Here, we review the known alterations of the FGFR pathway in sarcoma patients as well as the preclinical and clinical evidence for the use of FGFR inhibitors in these diseases. Finally, we discuss the possible reasons behind the current clinical data and highlight the need for biomarker stratification to select patients more likely to benefit from FGFR targeted therapies.
Collapse
Affiliation(s)
- Andrea Napolitano
- Sarcoma Unit, The Royal Marsden NHS Foundation Trust, 203 Fulham Road, London SW3 6JJ, UK; (A.N.); (A.E.O.); (R.L.J.)
- Department of Medical Oncology, University Campus Bio-Medico, 00128 Rome, Italy
| | - Alexandra E. Ostler
- Sarcoma Unit, The Royal Marsden NHS Foundation Trust, 203 Fulham Road, London SW3 6JJ, UK; (A.N.); (A.E.O.); (R.L.J.)
| | - Robin L. Jones
- Sarcoma Unit, The Royal Marsden NHS Foundation Trust, 203 Fulham Road, London SW3 6JJ, UK; (A.N.); (A.E.O.); (R.L.J.)
- The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | - Paul H. Huang
- The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
- Correspondence: ; Tel.: +44-207-153-5554
| |
Collapse
|
14
|
Ho B, Johann PD, Grabovska Y, De Dieu Andrianteranagna MJ, Yao F, Frühwald M, Hasselblatt M, Bourdeaut F, Williamson D, Huang A, Kool M. Molecular subgrouping of atypical teratoid/rhabdoid tumors-a reinvestigation and current consensus. Neuro Oncol 2021; 22:613-624. [PMID: 31889194 PMCID: PMC7229260 DOI: 10.1093/neuonc/noz235] [Citation(s) in RCA: 126] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background Atypical teratoid/rhabdoid tumors (ATRTs) are known to exhibit molecular and clinical heterogeneity even though SMARCB1 inactivation is the sole recurrent genetic event present in nearly all cases. Indeed, recent studies demonstrated 3 molecular subgroups of ATRTs that are genetically, epigenetically, and clinically distinct. As these studies included different numbers of tumors, various subgrouping techniques, and naming, an international working group sought to align previous findings and to reach a consensus on nomenclature and clinicopathological significance of ATRT subgroups. Methods We integrated various methods to perform a meta-analysis on published and unpublished DNA methylation and gene expression datasets of ATRTs and associated clinicopathological data. Results In concordance with previous studies, the analyses identified 3 main molecular subgroups of ATRTs, for which a consensus was reached to name them ATRT-TYR, ATRT-SHH, and ATRT-MYC. The ATRT-SHH subgroup exhibited further heterogeneity, segregating further into 2 subtypes associated with a predominant supratentorial (ATRT-SHH-1) or infratentorial (ATRT-SHH-2) location. For each ATRT subgroup we provide an overview of its main molecular and clinical characteristics, including SMARCB1 alterations and pathway activation. Conclusions The introduction of a common classification, characterization, and nomenclature of ATRT subgroups will facilitate future research and serve as a common ground for subgrouping patient samples and ATRT models, which will aid in refining subgroup-based therapies for ATRT patients.
Collapse
Affiliation(s)
- Ben Ho
- Division of Hematology and Oncology, Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Pascal D Johann
- Hopp Children's Cancer Center, Heidelberg, Germany.,Division of Pediatric Neuro-oncology, German Cancer Research Center and German Cancer Research Consortium, Heidelberg, Germany.,Department of Pediatric Hematology and Oncology, University Hospital Heidelberg, Heidelberg, Germany
| | - Yura Grabovska
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, UK
| | - Mamy Jean De Dieu Andrianteranagna
- Departments of Genetics and of Oncopediatry and Young Adults, Curie Institute, Paris, France.,INSERM U830, Laboratory of Translational Research in Pediatric Oncology, SIREDO Pediatric Oncology Center, Curie Institute, Paris, France
| | - Fupan Yao
- Department of Medical Biophysics, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Michael Frühwald
- University Children's Hospital Augsburg, Swabian Children's Cancer Center, Augsburg, Germany
| | - Martin Hasselblatt
- Institute of Neuropathology, University Hospital Münster, Münster, Germany
| | - Franck Bourdeaut
- Departments of Genetics and of Oncopediatry and Young Adults, Curie Institute, Paris, France.,INSERM U830, Laboratory of Translational Research in Pediatric Oncology, SIREDO Pediatric Oncology Center, Curie Institute, Paris, France
| | - Daniel Williamson
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, UK
| | - Annie Huang
- Division of Hematology and Oncology, Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Marcel Kool
- Hopp Children's Cancer Center, Heidelberg, Germany.,Division of Pediatric Neuro-oncology, German Cancer Research Center and German Cancer Research Consortium, Heidelberg, Germany
| |
Collapse
|
15
|
Hoffman LM, Richardson EA, Ho B, Margol A, Reddy A, Lafay-Cousin L, Chi S, Slavc I, Judkins A, Hasselblatt M, Bourdeaut F, Frühwald MC, Vibhakar R, Bouffet E, Huang A. Advancing biology-based therapeutic approaches for atypical teratoid rhabdoid tumors. Neuro Oncol 2021; 22:944-954. [PMID: 32129445 DOI: 10.1093/neuonc/noaa046] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Atypical teratoid rhabdoid tumor (ATRT) is a rare, highly malignant central nervous system cancer arising in infants and younger children, historically considered to be homogeneous, monogenic, and incurable. Recent use of intensified therapies has modestly improved survival for ATRT; however, a majority of patients will still succumb to their disease. While ATRTs almost universally exhibit loss of SMARCB1 (BAF47/INI1/SNF5), recent whole genome, transcriptome, and epigenomic analyses of large cohorts reveal previously underappreciated molecular heterogeneity. These discoveries provide novel insights into how SMARCB1 loss drives oncogenesis and confer specific therapeutic vulnerabilities, raising exciting prospects for molecularly stratified treatment for patients with ATRT.
Collapse
Affiliation(s)
- Lindsey M Hoffman
- Center for Cancer and Blood Disorders, Phoenix Children's Hospital, Phoenix, Arizona, USA
| | - Elizabeth Anne Richardson
- Arthur and Sonia Labatt Brain Tumour Research Centre, Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Ben Ho
- Arthur and Sonia Labatt Brain Tumour Research Centre, Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Ashley Margol
- Cancer and Blood Disease Institute, Children's Hospital Los Angeles, Los Angeles, California, USA.,Department of Pediatrics, Keck School of Medicine of University of Southern California, Los Angeles, California, USA
| | - Alyssa Reddy
- Departments of Neurology and Pediatrics, University of California San Francisco, San Francisco, California, USA
| | - Lucie Lafay-Cousin
- Department of Pediatric Hematology Oncology and Blood and Marrow Transplantation, Alberta Children's Hospital, Calgary, Alberta, Canada.,Department of Paediatrics and Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Susan Chi
- Pediatric Medical Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts, USA.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Irene Slavc
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria.,Division of Neonatology, Pediatric Intensive Care and Neuropediatrics, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Alexander Judkins
- Center for Personalized Medicine, Children's Hospital of Los Angeles.,Pathology and Laboratory Medicine, Children's Hospital of Los Angeles.,Department of Pathology, Keck School of Medicine University of Southern California, Los Angeles, California, USA
| | - Martin Hasselblatt
- Institute of Neuropathology, University Hospital Münster, Münster, Germany
| | - Franck Bourdeaut
- Curie Institute, Integrated Cancer Research Site, Paris, France.,Departments of Genetics and of Oncopediatry and Young Adults, Curie Institute, Paris, France.,INSERM U830, Laboratory of Translational Research in Pediatric Oncology, SIREDO Pediatric Oncology Center, Curie Institute, Paris, France
| | - Michael C Frühwald
- Swabian Children's Cancer Center, University Children's Hospital, University Hospital Augsburg, Augsburg, Germany.,Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, University of Münster, Münster, Germany.,EU-RHAB Registry Working Group, Augsburg, Germany
| | - Rajeev Vibhakar
- Department of Pediatrics, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado, USA.,Morgan Adams Foundation Pediatric Brain Tumor Research Program, Children's Hospital Colorado, Aurora, Colorado, USA.,Department of Neurosurgery, University of Colorado Denver, Aurora, Colorado, USA
| | - Eric Bouffet
- Child Health Evaluative Sciences, SickKids Research Institute, Toronto, Ontario, Canada.,Department of Paediatrics, University of Toronto, Toronto, Ontario, Canada.,Division of Hematology/Oncology, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Annie Huang
- Department of Paediatrics, University of Toronto, Toronto, Ontario, Canada.,Division of Hematology/Oncology, Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Medical Biophysics, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
16
|
Oberlick EM, Rees MG, Seashore-Ludlow B, Vazquez F, Nelson GM, Dharia NV, Weir BA, Tsherniak A, Ghandi M, Krill-Burger JM, Meyers RM, Wang X, Montgomery P, Root DE, Bieber JM, Radko S, Cheah JH, Hon CSY, Shamji AF, Clemons PA, Park PJ, Dyer MA, Golub TR, Stegmaier K, Hahn WC, Stewart EA, Schreiber SL, Roberts CWM. Small-Molecule and CRISPR Screening Converge to Reveal Receptor Tyrosine Kinase Dependencies in Pediatric Rhabdoid Tumors. Cell Rep 2020; 28:2331-2344.e8. [PMID: 31461650 DOI: 10.1016/j.celrep.2019.07.021] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 04/19/2019] [Accepted: 07/08/2019] [Indexed: 02/09/2023] Open
Abstract
Cancer is often seen as a disease of mutations and chromosomal abnormalities. However, some cancers, including pediatric rhabdoid tumors (RTs), lack recurrent alterations targetable by current drugs and need alternative, informed therapeutic options. To nominate potential targets, we performed a high-throughput small-molecule screen complemented by a genome-scale CRISPR-Cas9 gene-knockout screen in a large number of RT and control cell lines. These approaches converged to reveal several receptor tyrosine kinases (RTKs) as therapeutic targets, with RTK inhibition effective in suppressing RT cell growth in vitro and against a xenograft model in vivo. RT cell lines highly express and activate (phosphorylate) different RTKs, creating dependency without mutation or amplification. Downstream of RTK signaling, we identified PTPN11, encoding the pro-growth signaling protein SHP2, as a shared dependency across all RT cell lines. This study demonstrates that large-scale perturbational screening can uncover vulnerabilities in cancers with "quiet" genomes.
Collapse
Affiliation(s)
- Elaine M Oberlick
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Biological and Biomedical Sciences Program, Harvard Medical School, Boston, MA 02115, USA; Broad Institute, Cambridge, MA 02142, USA
| | | | - Brinton Seashore-Ludlow
- Broad Institute, Cambridge, MA 02142, USA; Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institute, 171 77 Stockholm, Sweden
| | | | - Geoffrey M Nelson
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| | - Neekesh V Dharia
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Broad Institute, Cambridge, MA 02142, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02215, USA; Boston Children's Hospital, Boston, MA 02115, USA
| | | | | | | | | | | | - Xiaofeng Wang
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | | | | | | | - Sandi Radko
- Comprehensive Cancer Center and Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | | | | | | | | | - Peter J Park
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA; Harvard Ludwig Center, Harvard Medical School, Boston, MA 02115, USA
| | - Michael A Dyer
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Todd R Golub
- Broad Institute, Cambridge, MA 02142, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02215, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Kimberly Stegmaier
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Broad Institute, Cambridge, MA 02142, USA; Boston Children's Hospital, Boston, MA 02115, USA
| | - William C Hahn
- Broad Institute, Cambridge, MA 02142, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Elizabeth A Stewart
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Stuart L Schreiber
- Broad Institute, Cambridge, MA 02142, USA; Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Charles W M Roberts
- Comprehensive Cancer Center and Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
17
|
Georgi F, Kuttler F, Murer L, Andriasyan V, Witte R, Yakimovich A, Turcatti G, Greber UF. A high-content image-based drug screen of clinical compounds against cell transmission of adenovirus. Sci Data 2020; 7:265. [PMID: 32788590 PMCID: PMC7423605 DOI: 10.1038/s41597-020-00604-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 07/16/2020] [Indexed: 12/21/2022] Open
Abstract
Human adenoviruses (HAdVs) are fatal to immuno-suppressed individuals, but no effective anti-HAdV therapy is available. Here, we present a novel image-based high-throughput screening (HTS) platform, which scores the full viral replication cycle from virus entry to dissemination of progeny and second-round infections. We analysed 1,280 small molecular weight compounds of the Prestwick Chemical Library (PCL) for interference with HAdV-C2 infection in a quadruplicate, blinded format, and performed robust image analyses and hit filtering. We present the entire set of the screening data including all images, image analyses and data processing pipelines. The data are made available at the Image Data Resource (IDR, idr0081). Our screen identified Nelfinavir mesylate as an inhibitor of HAdV-C2 multi-round plaque formation, but not single round infection. Nelfinavir has been FDA-approved for anti-retroviral therapy in humans. Our results underscore the power of image-based full cycle infection assays in identifying viral inhibitors with clinical potential.
Collapse
Affiliation(s)
- Fanny Georgi
- Department of Molecular Life Sciences, University of Zurich (UZH), Winterthurerstrasse, 190, 8057, Zurich, Switzerland
| | - Fabien Kuttler
- Biomolecular Screening Facility, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Station 15, Lausanne, 1015, Switzerland
| | - Luca Murer
- Department of Molecular Life Sciences, University of Zurich (UZH), Winterthurerstrasse, 190, 8057, Zurich, Switzerland
| | - Vardan Andriasyan
- Department of Molecular Life Sciences, University of Zurich (UZH), Winterthurerstrasse, 190, 8057, Zurich, Switzerland
| | - Robert Witte
- Department of Molecular Life Sciences, University of Zurich (UZH), Winterthurerstrasse, 190, 8057, Zurich, Switzerland
| | - Artur Yakimovich
- MRC Laboratory for Molecular Cell Biology, University College London, Gower St, London, WC1E 6BT, United Kingdom
- Artificial Intelligence for Life Sciences CIC, 40 Gowers walk, London, E1 8BH, United Kingdom
| | - Gerardo Turcatti
- Biomolecular Screening Facility, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Station 15, Lausanne, 1015, Switzerland
| | - Urs F Greber
- Department of Molecular Life Sciences, University of Zurich (UZH), Winterthurerstrasse, 190, 8057, Zurich, Switzerland.
| |
Collapse
|
18
|
Howard TP, Oberlick EM, Rees MG, Arnoff TE, Pham MT, Brenan L, DoCarmo M, Hong AL, Kugener G, Chou HC, Drosos Y, Mathias KM, Ramos P, Seashore-Ludlow B, Giacomelli AO, Wang X, Freeman BB, Blankenship K, Hoffmann L, Tiv HL, Gokhale PC, Johannessen CM, Stewart EA, Schreiber SL, Hahn WC, Roberts CWM. Rhabdoid Tumors Are Sensitive to the Protein-Translation Inhibitor Homoharringtonine. Clin Cancer Res 2020; 26:4995-5006. [PMID: 32631955 DOI: 10.1158/1078-0432.ccr-19-2717] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Revised: 05/30/2020] [Accepted: 06/29/2020] [Indexed: 12/31/2022]
Abstract
PURPOSE Rhabdoid tumors are devastating pediatric cancers in need of improved therapies. We sought to identify small molecules that exhibit in vitro and in vivo efficacy against preclinical models of rhabdoid tumor. EXPERIMENTAL DESIGN We screened eight rhabdoid tumor cell lines with 481 small molecules and compared their sensitivity with that of 879 other cancer cell lines. Genome-scale CRISPR-Cas9 inactivation screens in rhabdoid tumors were analyzed to confirm target vulnerabilities. Gene expression and CRISPR-Cas9 data were queried across cell lines and primary rhabdoid tumors to discover biomarkers of small-molecule sensitivity. Molecular correlates were validated by manipulating gene expression. Subcutaneous rhabdoid tumor xenografts were treated with the most effective drug to confirm in vitro results. RESULTS Small-molecule screening identified the protein-translation inhibitor homoharringtonine (HHT), an FDA-approved treatment for chronic myelogenous leukemia (CML), as the sole drug to which all rhabdoid tumor cell lines were selectively sensitive. Validation studies confirmed the sensitivity of rhabdoid tumor to HHT was comparable with that of CML cell lines. Low expression of the antiapoptotic gene BCL2L1, which encodes Bcl-XL, was the strongest predictor of HHT sensitivity, and HHT treatment consistently depleted Mcl-1, the synthetic-lethal antiapoptotic partner of Bcl-XL. Rhabdoid tumor cell lines and primary-tumor samples expressed low BCL2L1, and overexpression of BCL2L1 induced resistance to HHT in rhabdoid tumor cells. Furthermore, HHT treatment inhibited rhabdoid tumor cell line and patient-derived xenograft growth in vivo. CONCLUSIONS Rhabdoid tumor cell lines and xenografts are highly sensitive to HHT, at least partially due to their low expression of BCL2L1. HHT may have therapeutic potential against rhabdoid tumors.
Collapse
Affiliation(s)
- Thomas P Howard
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Division of Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Broad Institute of Harvard and MIT, Cambridge, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Elaine M Oberlick
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Division of Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts.,Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Matthew G Rees
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Taylor E Arnoff
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Division of Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Minh-Tam Pham
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Division of Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Lisa Brenan
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Mariana DoCarmo
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Andrew L Hong
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Division of Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Broad Institute of Harvard and MIT, Cambridge, Massachusetts.,Department of Pediatrics, Emory University, Atlanta, Georgia
| | | | - Hsien-Chao Chou
- Comprehensive Cancer Center, St. Jude Children's Research Hospital, Memphis, Tennessee.,Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Yiannis Drosos
- Comprehensive Cancer Center, St. Jude Children's Research Hospital, Memphis, Tennessee.,Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Kaeli M Mathias
- Comprehensive Cancer Center, St. Jude Children's Research Hospital, Memphis, Tennessee.,Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Pilar Ramos
- Comprehensive Cancer Center, St. Jude Children's Research Hospital, Memphis, Tennessee.,Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | | | - Andrew O Giacomelli
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Broad Institute of Harvard and MIT, Cambridge, Massachusetts.,Tumor Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Xiaofeng Wang
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Division of Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts.,Department of Molecular and Systems Biology, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire
| | - Burgess B Freeman
- Preclinical Pharmacokinetics Shared Resource, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Kaley Blankenship
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Lauren Hoffmann
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Hong L Tiv
- Experimental Therapeutics Core and Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Prafulla C Gokhale
- Experimental Therapeutics Core and Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Elizabeth A Stewart
- Comprehensive Cancer Center, St. Jude Children's Research Hospital, Memphis, Tennessee. .,Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee.,Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Stuart L Schreiber
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts.,Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts
| | - William C Hahn
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. .,Broad Institute of Harvard and MIT, Cambridge, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Charles W M Roberts
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Division of Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts. .,Broad Institute of Harvard and MIT, Cambridge, Massachusetts.,Harvard Medical School, Boston, Massachusetts.,Comprehensive Cancer Center, St. Jude Children's Research Hospital, Memphis, Tennessee.,Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee
| |
Collapse
|
19
|
Zhang H, Qi L, Du Y, Huang LF, Braun FK, Kogiso M, Zhao Y, Li C, Lindsay H, Zhao S, Injac SG, Baxter PA, Su JM, Stephan C, Keller C, Heck KA, Harmanci A, Harmanci AO, Yang J, Klisch TJ, Li XN, Patel AJ. Patient-Derived Orthotopic Xenograft (PDOX) Mouse Models of Primary and Recurrent Meningioma. Cancers (Basel) 2020; 12:cancers12061478. [PMID: 32517016 PMCID: PMC7352400 DOI: 10.3390/cancers12061478] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 05/26/2020] [Accepted: 06/01/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Meningiomas constitute one-third of all primary brain tumors. Although typically benign, about 20% of these tumors recur despite surgery and radiation, and may ultimately prove fatal. There are currently no effective chemotherapies for meningioma. We, therefore, set out to develop patient-derived orthotopic xenograft (PDOX) mouse models of human meningioma using tumor. METHOD Of nine patients, four had World Health Organization (WHO) grade I tumors, five had WHO grade II tumors, and in this second group two patients also had recurrent (WHO grade III) meningioma. We also classified the tumors according to our recently developed molecular classification system (Types A, B, and C, with C being the most aggressive). We transplanted all 11 surgical samples into the skull base of immunodeficient (SCID) mice. Only the primary and recurrent tumor cells from one patient-both molecular Type C, despite being WHO grades II and III, respectively-led to the formation of meningioma in the resulting mouse models. We characterized the xenografts by histopathology and RNA-seq and compared them with the original tumors. We performed an in vitro drug screen using 60 anti-cancer drugs followed by in vivo validation. RESULTS The PDOX models established from the primary and recurrent tumors from patient K29 (K29P-PDOX and K29R-PDOX, respectively) replicated the histopathology and key gene expression profiles of the original samples. Although these xenografts could not be subtransplanted, the cryopreserved primary tumor cells were able to reliably generate PDOX tumors. Drug screening in K29P and K29R tumor cell lines revealed eight compounds that were active on both tumors, including three histone deacetylase (HDAC) inhibitors. We tested the HDAC inhibitor Panobinostat in K29R-PDOX mice, and it significantly prolonged mouse survival (p < 0.05) by inducing histone H3 acetylation and apoptosis. CONCLUSION Meningiomas are not very amenable to PDOX modeling, for reasons that remain unclear. Yet at least some of the most malignant tumors can be modeled, and cryopreserved primary tumor cells can create large panels of tumors that can be used for preclinical drug testing.
Collapse
Affiliation(s)
- Huiyuan Zhang
- Laboratory of Molecular Neuro-Oncology, Department of Pediatrics, Preclinical Neuro-Oncology Research Program, Baylor College of Medicine, Houston, TX 77030, USA; (H.Z.); (L.Q.); (Y.D.); (F.K.B.); (M.K.); (H.L.); (S.Z.); (S.G.I.); (P.A.B.)
- Department of Pediatrics, Texas Children’s Cancer Center, Texas Children’s Hospital, Houston, TX 77030, USA; (Y.Z.); (J.M.S.); (J.Y.)
| | - Lin Qi
- Laboratory of Molecular Neuro-Oncology, Department of Pediatrics, Preclinical Neuro-Oncology Research Program, Baylor College of Medicine, Houston, TX 77030, USA; (H.Z.); (L.Q.); (Y.D.); (F.K.B.); (M.K.); (H.L.); (S.Z.); (S.G.I.); (P.A.B.)
- Department of Pediatrics, Texas Children’s Cancer Center, Texas Children’s Hospital, Houston, TX 77030, USA; (Y.Z.); (J.M.S.); (J.Y.)
- Program of Precision Medicine PDOX Modeling of Pediatric Tumors, Ann and Robert H. Lurie Children’s Hospital of Chicago and Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Yuchen Du
- Laboratory of Molecular Neuro-Oncology, Department of Pediatrics, Preclinical Neuro-Oncology Research Program, Baylor College of Medicine, Houston, TX 77030, USA; (H.Z.); (L.Q.); (Y.D.); (F.K.B.); (M.K.); (H.L.); (S.Z.); (S.G.I.); (P.A.B.)
- Department of Pediatrics, Texas Children’s Cancer Center, Texas Children’s Hospital, Houston, TX 77030, USA; (Y.Z.); (J.M.S.); (J.Y.)
- Program of Precision Medicine PDOX Modeling of Pediatric Tumors, Ann and Robert H. Lurie Children’s Hospital of Chicago and Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - L. Frank Huang
- Division of Experimental Hematology and Cancer Biology, Brain Tumor Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA;
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Frank K. Braun
- Laboratory of Molecular Neuro-Oncology, Department of Pediatrics, Preclinical Neuro-Oncology Research Program, Baylor College of Medicine, Houston, TX 77030, USA; (H.Z.); (L.Q.); (Y.D.); (F.K.B.); (M.K.); (H.L.); (S.Z.); (S.G.I.); (P.A.B.)
- Department of Pediatrics, Texas Children’s Cancer Center, Texas Children’s Hospital, Houston, TX 77030, USA; (Y.Z.); (J.M.S.); (J.Y.)
| | - Mari Kogiso
- Laboratory of Molecular Neuro-Oncology, Department of Pediatrics, Preclinical Neuro-Oncology Research Program, Baylor College of Medicine, Houston, TX 77030, USA; (H.Z.); (L.Q.); (Y.D.); (F.K.B.); (M.K.); (H.L.); (S.Z.); (S.G.I.); (P.A.B.)
- Department of Pediatrics, Texas Children’s Cancer Center, Texas Children’s Hospital, Houston, TX 77030, USA; (Y.Z.); (J.M.S.); (J.Y.)
| | - Yanling Zhao
- Department of Pediatrics, Texas Children’s Cancer Center, Texas Children’s Hospital, Houston, TX 77030, USA; (Y.Z.); (J.M.S.); (J.Y.)
| | - Can Li
- Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX 77030, USA; (C.L.); (C.S.)
| | - Holly Lindsay
- Laboratory of Molecular Neuro-Oncology, Department of Pediatrics, Preclinical Neuro-Oncology Research Program, Baylor College of Medicine, Houston, TX 77030, USA; (H.Z.); (L.Q.); (Y.D.); (F.K.B.); (M.K.); (H.L.); (S.Z.); (S.G.I.); (P.A.B.)
- Department of Pediatrics, Texas Children’s Cancer Center, Texas Children’s Hospital, Houston, TX 77030, USA; (Y.Z.); (J.M.S.); (J.Y.)
| | - Sibo Zhao
- Laboratory of Molecular Neuro-Oncology, Department of Pediatrics, Preclinical Neuro-Oncology Research Program, Baylor College of Medicine, Houston, TX 77030, USA; (H.Z.); (L.Q.); (Y.D.); (F.K.B.); (M.K.); (H.L.); (S.Z.); (S.G.I.); (P.A.B.)
- Department of Pediatrics, Texas Children’s Cancer Center, Texas Children’s Hospital, Houston, TX 77030, USA; (Y.Z.); (J.M.S.); (J.Y.)
| | - Sarah G. Injac
- Laboratory of Molecular Neuro-Oncology, Department of Pediatrics, Preclinical Neuro-Oncology Research Program, Baylor College of Medicine, Houston, TX 77030, USA; (H.Z.); (L.Q.); (Y.D.); (F.K.B.); (M.K.); (H.L.); (S.Z.); (S.G.I.); (P.A.B.)
- Department of Pediatrics, Texas Children’s Cancer Center, Texas Children’s Hospital, Houston, TX 77030, USA; (Y.Z.); (J.M.S.); (J.Y.)
| | - Patricia A. Baxter
- Laboratory of Molecular Neuro-Oncology, Department of Pediatrics, Preclinical Neuro-Oncology Research Program, Baylor College of Medicine, Houston, TX 77030, USA; (H.Z.); (L.Q.); (Y.D.); (F.K.B.); (M.K.); (H.L.); (S.Z.); (S.G.I.); (P.A.B.)
- Department of Pediatrics, Texas Children’s Cancer Center, Texas Children’s Hospital, Houston, TX 77030, USA; (Y.Z.); (J.M.S.); (J.Y.)
| | - Jack M. Su
- Department of Pediatrics, Texas Children’s Cancer Center, Texas Children’s Hospital, Houston, TX 77030, USA; (Y.Z.); (J.M.S.); (J.Y.)
| | - Clifford Stephan
- Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX 77030, USA; (C.L.); (C.S.)
| | - Charles Keller
- Children’s Cancer Therapy Development Institute, Beaverton, OR 97005, USA;
| | - Kent A. Heck
- Department of Pathology, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Akdes Harmanci
- Center for Computational Systems Medicine, School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX 77030, USA;
| | - Arif O. Harmanci
- Center for Precision Health, School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX 77030, USA;
| | - Jianhua Yang
- Department of Pediatrics, Texas Children’s Cancer Center, Texas Children’s Hospital, Houston, TX 77030, USA; (Y.Z.); (J.M.S.); (J.Y.)
| | - Tiemo J. Klisch
- Jan and Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX 77030, USA;
| | - Xiao-Nan Li
- Laboratory of Molecular Neuro-Oncology, Department of Pediatrics, Preclinical Neuro-Oncology Research Program, Baylor College of Medicine, Houston, TX 77030, USA; (H.Z.); (L.Q.); (Y.D.); (F.K.B.); (M.K.); (H.L.); (S.Z.); (S.G.I.); (P.A.B.)
- Department of Pediatrics, Texas Children’s Cancer Center, Texas Children’s Hospital, Houston, TX 77030, USA; (Y.Z.); (J.M.S.); (J.Y.)
- Program of Precision Medicine PDOX Modeling of Pediatric Tumors, Ann and Robert H. Lurie Children’s Hospital of Chicago and Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Correspondence: (X.-N.L.); (A.J.P.)
| | - Akash J. Patel
- Jan and Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX 77030, USA;
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX 77030, USA
- Correspondence: (X.-N.L.); (A.J.P.)
| |
Collapse
|
20
|
Theruvath J, Sotillo E, Mount CW, Graef CM, Delaidelli A, Heitzeneder S, Labanieh L, Dhingra S, Leruste A, Majzner RG, Xu P, Mueller S, Yecies DW, Finetti MA, Williamson D, Johann PD, Kool M, Pfister S, Hasselblatt M, Frühwald MC, Delattre O, Surdez D, Bourdeaut F, Puget S, Zaidi S, Mitra SS, Cheshier S, Sorensen PH, Monje M, Mackall CL. Locoregionally administered B7-H3-targeted CAR T cells for treatment of atypical teratoid/rhabdoid tumors. Nat Med 2020; 26:712-719. [PMID: 32341579 DOI: 10.1038/s41591-020-0821-8] [Citation(s) in RCA: 206] [Impact Index Per Article: 41.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 03/06/2020] [Indexed: 01/01/2023]
Abstract
Atypical teratoid/rhabdoid tumors (ATRTs) typically arise in the central nervous system (CNS) of children under 3 years of age. Despite intensive multimodal therapy (surgery, chemotherapy and, if age permits, radiotherapy), median survival is 17 months1,2. We show that ATRTs robustly express B7-H3/CD276 that does not result from the inactivating mutations in SMARCB1 (refs. 3,4), which drive oncogenesis in ATRT, but requires residual SWItch/Sucrose Non-Fermentable (SWI/SNF) activity mediated by BRG1/SMARCA4. Consistent with the embryonic origin of ATRT5,6, B7-H3 is highly expressed on the prenatal, but not postnatal, brain. B7-H3.BB.z-chimeric antigen receptor (CAR) T cells administered intracerebroventricularly or intratumorally mediate potent antitumor effects against cerebral ATRT xenografts in mice, with faster kinetics, greater potency and reduced systemic levels of inflammatory cytokines compared to CAR T cells administered intravenously. CAR T cells administered ICV also traffic from the CNS into the periphery; following clearance of ATRT xenografts, B7-H3.BB.z-CAR T cells administered intracerebroventricularly or intravenously mediate antigen-specific protection from tumor rechallenge, both in the brain and periphery. These results identify B7-H3 as a compelling therapeutic target for this largely incurable pediatric tumor and demonstrate important advantages of locoregional compared to systemic delivery of CAR T cells for the treatment of CNS malignancies.
Collapse
Affiliation(s)
- Johanna Theruvath
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Elena Sotillo
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Christopher W Mount
- Department of Neurology, Stanford University School of Medicine, Stanford, CA, USA
| | - Claus Moritz Graef
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Alberto Delaidelli
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Sabine Heitzeneder
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Louai Labanieh
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA.,Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Shaurya Dhingra
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Amaury Leruste
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Robbie G Majzner
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA.,Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Peng Xu
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Sabine Mueller
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA.,Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA.,Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | - Derek W Yecies
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Martina A Finetti
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, UK
| | - Daniel Williamson
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, UK
| | - Pascal D Johann
- Hopp Children's Cancer Center Heidelberg, Heidelberg, Germany.,Division of Pediatric Neurooncology, German Cancer Research Center and German Cancer Consortium, Heidelberg, Germany.,Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Marcel Kool
- Hopp Children's Cancer Center Heidelberg, Heidelberg, Germany.,Division of Pediatric Neurooncology, German Cancer Research Center and German Cancer Consortium, Heidelberg, Germany
| | - Stefan Pfister
- Hopp Children's Cancer Center Heidelberg, Heidelberg, Germany.,Division of Pediatric Neurooncology, German Cancer Research Center and German Cancer Consortium, Heidelberg, Germany.,Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Martin Hasselblatt
- Institute of Neuropathology, Münster University Hospital, Münster, Germany
| | - Michael C Frühwald
- University Children's Hospital Augsburg, Swabian Children's Cancer Center, Augsburg, Germany.,EU-RHAB Registry Center, Augsburg, Germany
| | - Olivier Delattre
- Paris Sciences Lettres Research University, INSERM U830, Paris, France.,Paris Sciences Lettres Research University, SIREDO: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, Paris, France
| | - Didier Surdez
- Paris Sciences Lettres Research University, INSERM U830, Paris, France.,Paris Sciences Lettres Research University, SIREDO: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, Paris, France
| | - Franck Bourdeaut
- Paris Sciences Lettres Research University, INSERM U830, Paris, France.,Paris Sciences Lettres Research University, SIREDO: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, Paris, France
| | - Stephanie Puget
- Paris University, Necker-Enfants Malades Hospital, Department of Neurosurgery, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Sakina Zaidi
- Paris Sciences Lettres Research University, INSERM U830, Paris, France.,Paris Sciences Lettres Research University, SIREDO: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, Paris, France
| | - Siddhartha S Mitra
- Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Samuel Cheshier
- Division of Pediatric Neurosurgery, Department of Neurosurgery, Primary Children's Hospital and Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Poul H Sorensen
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Michelle Monje
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA.,Department of Neurology, Stanford University School of Medicine, Stanford, CA, USA.,Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA.,Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA.,Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.,Stanford Institute for Stem Cell and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Crystal L Mackall
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA. .,Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA. .,Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
21
|
Federico SM, Caldwell KJ, McCarville MB, Daryani VM, Stewart CF, Mao S, Wu J, Davidoff AM, Santana VM, Furman WL, Pappo AS, Navid F. Phase I expansion cohort to evaluate the combination of bevacizumab, sorafenib and low-dose cyclophosphamide in children and young adults with refractory or recurrent solid tumours. Eur J Cancer 2020; 132:35-42. [PMID: 32325418 DOI: 10.1016/j.ejca.2020.03.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 02/15/2020] [Accepted: 03/06/2020] [Indexed: 10/24/2022]
Abstract
BACKGROUND Angiogenesis is critical for tumour growth and metastasis. Dual inhibition of vascular endothelial growth factors and platelet-derived growth factor receptors suppresses angiogenesis. This expansion cohort of a phase I study targeted angiogenesis with sorafenib, bevacizumab and low-dose cyclophosphamide in children and young adults with recurrent solid tumours. METHODS An expansion cohort including patients with refractory or recurrent solid tumours was enrolled and received bevacizumab (15 mg/kg IV, day 1), sorafenib (90 mg/m2 po twice daily, days 1-21) and low-dose cyclophosphamide (50 mg/m2 po daily, days 1-21). Each course was 21 days. Toxicities were assessed using Common Terminology Criteria for Adverse Events, v3.0, and responses were evaluated by Response Evaluation Criteria in Solid Tumors criteria. Serial bevacizumab pharmacokinetic (PK) studies were performed during course 1. RESULTS Twenty-four patients (15 males; median age 14.5 yrs; range 1-22 yr) received a median of 6 courses (range 1-18). Twelve patients had a bone or soft tissue sarcoma. The most common grade III/IV non-haematologic toxicities were hypertension (N = 4), hand/foot rash (N = 3) and elevated lipase (N = 3). The most common grade III/IV haematologic toxicities were neutropenia (N = 7) and lymphopenia (N = 17). Three patients (2 synovial sarcoma, 1 rhabdoid tumour) achieved a partial response and 18 had stable disease. The progression-free survival at 3 and 6 months were 78.1% (95% confidence interval [CI] 60.6-95.6%) and 54% (95% CI 30.2-78.2%), respectively. Bevacizumab PKs in 15 patients was similar to published adult PK results. CONCLUSIONS Intravenous bevacizumab combined with oral sorafenib and low-dose cyclophosphamide was tolerated and demonstrated promising activity in a subset of childhood solid tumours.
Collapse
Affiliation(s)
- Sara M Federico
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Kenneth J Caldwell
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Mary B McCarville
- Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Vinay M Daryani
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Clinton F Stewart
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Shenghua Mao
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jianrong Wu
- Department of Biostatistics and Bioinformatics Shared Resource Facility, Markey Cancer Center, University of Kentucky, Lexington, KY 40504, USA
| | - Andrew M Davidoff
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Victor M Santana
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Wayne L Furman
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Alberto S Pappo
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Fariba Navid
- Department of Pediatrics, Children's Hospital Los Angeles, University of Southern California, Keck School of Medicine, Los Angeles, CA 90027, USA
| |
Collapse
|
22
|
The SWI/SNF complex in cancer - biology, biomarkers and therapy. Nat Rev Clin Oncol 2020; 17:435-448. [PMID: 32303701 DOI: 10.1038/s41571-020-0357-3] [Citation(s) in RCA: 399] [Impact Index Per Article: 79.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2020] [Indexed: 12/11/2022]
Abstract
Cancer genome-sequencing studies have revealed a remarkably high prevalence of mutations in genes encoding subunits of the SWI/SNF chromatin-remodelling complexes, with nearly 25% of all cancers harbouring aberrations in one or more of these genes. A role for such aberrations in tumorigenesis is evidenced by cancer predisposition in both carriers of germline loss-of-function mutations and genetically engineered mouse models with inactivation of any of several SWI/SNF subunits. Whereas many of the most frequently mutated oncogenes and tumour-suppressor genes have been studied for several decades, the cancer-promoting role of mutations in SWI/SNF genes has been recognized only more recently, and thus comparatively less is known about these alterations. Consequently, increasing research interest is being focused on understanding the prognostic and, in particular, the potential therapeutic implications of mutations in genes encoding SWI/SNF subunits. Herein, we review the burgeoning data on the mechanisms by which mutations affecting SWI/SNF complexes promote cancer and describe promising emerging opportunities for targeted therapy, including immunotherapy with immune-checkpoint inhibitors, presented by these mutations. We also highlight ongoing clinical trials open specifically to patients with cancers harbouring mutations in certain SWI/SNF genes.
Collapse
|
23
|
Finetti MA, Grabovska Y, Bailey S, Williamson D. Translational genomics of malignant rhabdoid tumours: Current impact and future possibilities. Semin Cancer Biol 2020; 61:30-41. [PMID: 31923457 DOI: 10.1016/j.semcancer.2019.12.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 12/15/2019] [Accepted: 12/16/2019] [Indexed: 12/24/2022]
Abstract
Malignant Rhabdoid Tumours (MRT) are the quintessential example of an epigenetic cancer. Mutation of a single gene, SMARCB1 or more rarely SMARCA4, is capable of causing one of the most aggressive and lethal cancers of early childhood and infancy. SMARCB1 encodes a core subunit of the SWI/SNF complex and its mutation evokes genome-wide downstream effects which may be counteracted therapeutically. Here we review and discuss the use of translational genomics in the study of MRT biology and the ways in which this has impacted clinical practice or may do so in the future. First, the diagnosis and definition of MRT and the transition from a histopathological to a molecular definition. Second, epigenetic and transcriptomic subgroups within MRT, their defining features and potential prognostic or therapeutic significance. Third, functional genomic studies of MRT by mouse modelling and forced re-expression of SMARCB1 in MRT cells. Fourth, studies of underlying epigenetic mechanisms (e.g. EZH2, HDACs) or deregulated kinases (e.g. PDGFR, FGFR1) and the potential therapeutic opportunities these provide. Finally, we discuss likely future directions and proffer opinion on how future translational genomics should be integrated into future biological/clinical studies to select and evaluate the best anti-MRT therapeutic agents.
Collapse
Affiliation(s)
- Martina A Finetti
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle Upon Tyne, UK
| | - Yura Grabovska
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle Upon Tyne, UK
| | - Simon Bailey
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle Upon Tyne, UK
| | - Daniel Williamson
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle Upon Tyne, UK.
| |
Collapse
|
24
|
Roskoski R. The role of fibroblast growth factor receptor (FGFR) protein-tyrosine kinase inhibitors in the treatment of cancers including those of the urinary bladder. Pharmacol Res 2020; 151:104567. [DOI: 10.1016/j.phrs.2019.104567] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 11/20/2019] [Indexed: 12/31/2022]
|
25
|
Vinet M, Suresh S, Maire V, Monchecourt C, Némati F, Lesage L, Pierre F, Ye M, Lescure A, Brisson A, Meseure D, Nicolas A, Rigaill G, Marangoni E, Del Nery E, Roman-Roman S, Dubois T. Protein arginine methyltransferase 5: A novel therapeutic target for triple-negative breast cancers. Cancer Med 2019; 8:2414-2428. [PMID: 30957988 PMCID: PMC6537044 DOI: 10.1002/cam4.2114] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 03/07/2019] [Accepted: 03/07/2019] [Indexed: 01/15/2023] Open
Abstract
TNBC is a highly heterogeneous and aggressive breast cancer subtype associated with high relapse rates, and for which no targeted therapy yet exists. Protein arginine methyltransferase 5 (PRMT5), an enzyme which catalyzes the methylation of arginines on histone and non‐histone proteins, has recently emerged as a putative target for cancer therapy. Potent and specific PRMT5 inhibitors have been developed, but the therapeutic efficacy of PRMT5 targeting in TNBC has not yet been demonstrated. Here, we examine the expression of PRMT5 in a human breast cancer cohort obtained from the Institut Curie, and evaluate the therapeutic potential of pharmacological inhibition of PRMT5 in TNBC. We find that PRMT5 mRNA and protein are expressed at comparable levels in TNBC, luminal breast tumors, and healthy mammary tissues. However, immunohistochemistry analyses reveal that PRMT5 is differentially localized in TNBC compared to other breast cancer subtypes and to normal breast tissues. PRMT5 is heterogeneously expressed in TNBC and high PRMT5 expression correlates with poor prognosis within this breast cancer subtype. Using the small‐molecule inhibitor EPZ015666, we show that PRMT5 inhibition impairs cell proliferation in a subset of TNBC cell lines. PRMT5 inhibition triggers apoptosis, regulates cell cycle progression and decreases mammosphere formation. Furthermore, EPZ015666 administration to a patient‐derived xenograft model of TNBC significantly deters tumor progression. Finally, we reveal potentiation between EGFR and PRMT5 targeting, suggestive of a beneficial combination therapy. Our findings highlight a distinctive subcellular localization of PRMT5 in TNBC, and uphold PRMT5 targeting, alone or in combination, as a relevant treatment strategy for a subset of TNBC.
Collapse
Affiliation(s)
- Mathilde Vinet
- Translational Research Department, Institut Curie, PSL Research University, Paris, France.,Breast Cancer Biology Group, Institut Curie, Paris, France
| | - Samyuktha Suresh
- Translational Research Department, Institut Curie, PSL Research University, Paris, France.,Breast Cancer Biology Group, Institut Curie, Paris, France
| | - Virginie Maire
- Translational Research Department, Institut Curie, PSL Research University, Paris, France.,Breast Cancer Biology Group, Institut Curie, Paris, France
| | - Clarisse Monchecourt
- Translational Research Department, Institut Curie, PSL Research University, Paris, France.,Breast Cancer Biology Group, Institut Curie, Paris, France
| | - Fariba Némati
- Translational Research Department, Institut Curie, PSL Research University, Paris, France.,Preclinical Investigation Laboratory, Institut Curie, Paris, France
| | - Laetitia Lesage
- Platform of Investigative Pathology, Department of Pathology, Institut Curie, Paris, France
| | - Fabienne Pierre
- Translational Research Department, Institut Curie, PSL Research University, Paris, France.,Breast Cancer Biology Group, Institut Curie, Paris, France.,Biophenics High-Content Screening Laboratory, Cell and Tissue Imaging Facility (PICT-IBiSA), Institut Curie, Paris, France
| | - Mengliang Ye
- Translational Research Department, Institut Curie, PSL Research University, Paris, France.,Breast Cancer Biology Group, Institut Curie, Paris, France
| | - Auriane Lescure
- Translational Research Department, Institut Curie, PSL Research University, Paris, France.,Biophenics High-Content Screening Laboratory, Cell and Tissue Imaging Facility (PICT-IBiSA), Institut Curie, Paris, France
| | - Amélie Brisson
- Translational Research Department, Institut Curie, PSL Research University, Paris, France.,Breast Cancer Biology Group, Institut Curie, Paris, France
| | - Didier Meseure
- Platform of Investigative Pathology, Department of Pathology, Institut Curie, Paris, France
| | - André Nicolas
- Platform of Investigative Pathology, Department of Pathology, Institut Curie, Paris, France
| | - Guillem Rigaill
- Institute of Plant Sciences Paris-Saclay (IPS2), UMR 9213, UMR1403, CNRS, INRA, Université Paris-Sud, Université d'Evry, Université Paris-Diderot, Sorbonne, Paris-Cité, Orsay, France.,Laboratoire de Mathématiques et Modélisation d'Evry (LaMME), Université d'Evry Val d'Essonne, UMR CNRS 8071, ENSIIE, USC INRA, Evry, France
| | - Elisabetta Marangoni
- Translational Research Department, Institut Curie, PSL Research University, Paris, France.,Preclinical Investigation Laboratory, Institut Curie, Paris, France
| | - Elaine Del Nery
- Translational Research Department, Institut Curie, PSL Research University, Paris, France.,Biophenics High-Content Screening Laboratory, Cell and Tissue Imaging Facility (PICT-IBiSA), Institut Curie, Paris, France
| | - Sergio Roman-Roman
- Translational Research Department, Institut Curie, PSL Research University, Paris, France
| | - Thierry Dubois
- Translational Research Department, Institut Curie, PSL Research University, Paris, France.,Breast Cancer Biology Group, Institut Curie, Paris, France
| |
Collapse
|
26
|
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
|
27
|
Katoh M. Fibroblast growth factor receptors as treatment targets in clinical oncology. Nat Rev Clin Oncol 2018; 16:105-122. [DOI: 10.1038/s41571-018-0115-y] [Citation(s) in RCA: 250] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
28
|
Nemes K, Frühwald MC. Emerging therapeutic targets for the treatment of malignant rhabdoid tumors. Expert Opin Ther Targets 2018. [PMID: 29528755 DOI: 10.1080/14728222.2018.1451839] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Malignant Rhabdoid Tumor (MRT) is a rare and highly aggressive malignancy primarily affecting infants and young children. The most common anatomic locations are the central nervous system (AT/RT), the kidneys (RTK) and other soft tissues (eMRT). The genetic origin of this disease is linked to mutations in SMARCB1, a gene encoding a core subunit of the SWI/SNF chromatin-remodeling complex. Areas covered: Conventional multimodal treatment may offer a significant survival benefit to certain patients. It remains to be determined, however, which patients will prove resistant to chemotherapy and need novel therapeutic approaches. Herein we discuss key signal transduction pathways involved in the pathogenesis of rhabdoid tumors for potential targeted therapy (EZH2, DNMT, HDAC, CDK4/6/Cyclin D1/Rb, AURKA, SHH/GLI1, Wnt/ß-Catenin, immunotherapy). Additional agents currently evaluated in preclinical settings and experimental clinical trials are discussed. Expert opinion: MRTs are genetically homogeneous, but epigenetically distinct malignancies. While there is an abundance of experimental in vitro studies evaluating potential therapeutic avenues, a dearth of clinical trials specifically for this entity persists. In order to improve outcome patients need to be carefully stratified and treated by targeted therapies combined with conventional chemotherapy or with new, less selective experimental agents in phase I/II clinical trials.
Collapse
Affiliation(s)
- Karolina Nemes
- a Swabian Children's Cancer Center , Children's Hospital, Klinikum Augsburg , Augsburg , Germany
| | - Michael C Frühwald
- a Swabian Children's Cancer Center , Children's Hospital, Klinikum Augsburg , Augsburg , Germany
| |
Collapse
|