1
|
Heim C, Hartig L, Weinelt N, Moser LM, Salzmann-Manrique E, Merker M, Wels WS, Tonn T, Bader P, Klusmann JH, van Wijk SJ, Rettinger E. Bortezomib promotes the TRAIL-mediated killing of resistant rhabdomyosarcoma by ErbB2/Her2-targeted CAR-NK-92 cells via DR5 upregulation. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200802. [PMID: 38706988 PMCID: PMC11067460 DOI: 10.1016/j.omton.2024.200802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 04/08/2024] [Indexed: 05/07/2024]
Abstract
Treatment resistance and immune escape are hallmarks of metastatic rhabdomyosarcoma (RMS), underscoring the urgent medical need for therapeutic agents against this disease entity as a key challenge in pediatric oncology. Chimeric antigen receptor (CAR)-based immunotherapies, such as the ErbB2 (Her2)-CAR-engineered natural killer (NK) cell line NK-92/5.28.z, provide antitumor cytotoxicity primarily through CAR-mediated cytotoxic granule release and thereafter-even in cases with low surface antigen expression or tumor escape-by triggering intrinsic NK cell-mediated apoptosis induction via additional ligand/receptors. In this study, we showed that bortezomib increased susceptibility toward apoptosis in clinically relevant RMS cell lines RH30 and RH41, and patient-derived RMS tumor organoid RMS335, by upregulation of the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) receptor DR5 in these metastatic, relapsed/refractory (r/r) RMS tumors. Subsequent administration of NK-92/5.28.z cells significantly enhanced antitumor activity in vitro. Applying recombinant TRAIL instead of NK-92/5.28.z cells confirmed that the synergistic antitumor effects of the combination treatment were mediated via TRAIL. Western blot analyses indicated that the combination treatment with bortezomib and NK-92/5.28.z cells increased apoptosis by interacting with the nuclear factor κB, JNK, and caspase pathways. Overall, bortezomib pretreatment can sensitize r/r RMS tumors to CAR- and, by upregulating DR5, TRAIL-mediated cytotoxicity of NK-92/5.28.z cells.
Collapse
Affiliation(s)
- Catrin Heim
- Goethe University Frankfurt, Department of Pediatrics, Division of Stem Cell Transplantation and Immunology, 60590 Frankfurt am Main, Germany
- Goethe University Frankfurt, Department of Pediatrics, 60590 Frankfurt am Main, Germany
| | - Leonie Hartig
- Goethe University Frankfurt, Department of Pediatrics, Division of Stem Cell Transplantation and Immunology, 60590 Frankfurt am Main, Germany
- Goethe University Frankfurt, Department of Pediatrics, 60590 Frankfurt am Main, Germany
| | - Nadine Weinelt
- Institute for Experimental Paediatric Haematology and Oncology (EPOH), 60528 Frankfurt am Main, Germany
| | - Laura M. Moser
- Goethe University Frankfurt, Department of Pediatrics, Division of Stem Cell Transplantation and Immunology, 60590 Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), partner site Frankfurt am Main, a partnership between DKFZ and University Hospital and Georg-Speyer-Haus, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), 60596 Frankfurt am Main, Germany
- Universitäres Centrum für Tumorerkrankungen (UCT) Frankfurt Marburg, 60590 Frankfurt am Main, Germany
- Goethe University Frankfurt, Department of Pediatrics, 60590 Frankfurt am Main, Germany
| | - Emilia Salzmann-Manrique
- Goethe University Frankfurt, Department of Pediatrics, Division of Stem Cell Transplantation and Immunology, 60590 Frankfurt am Main, Germany
- Goethe University Frankfurt, Department of Pediatrics, 60590 Frankfurt am Main, Germany
| | - Michael Merker
- Goethe University Frankfurt, Department of Pediatrics, Division of Stem Cell Transplantation and Immunology, 60590 Frankfurt am Main, Germany
- Universitäres Centrum für Tumorerkrankungen (UCT) Frankfurt Marburg, 60590 Frankfurt am Main, Germany
- Goethe University Frankfurt, Department of Pediatrics, 60590 Frankfurt am Main, Germany
| | - Winfried S. Wels
- German Cancer Consortium (DKTK), partner site Frankfurt am Main, a partnership between DKFZ and University Hospital and Georg-Speyer-Haus, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), 60596 Frankfurt am Main, Germany
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, 60596 Frankfurt am Main, Germany
| | - Torsten Tonn
- DRK-Blutspendedienst Baden-Württemberg/Hessen gemeinnützige GmbH, 60505 Frankfurt am Main, Germany
| | - Peter Bader
- Goethe University Frankfurt, Department of Pediatrics, Division of Stem Cell Transplantation and Immunology, 60590 Frankfurt am Main, Germany
- Universitäres Centrum für Tumorerkrankungen (UCT) Frankfurt Marburg, 60590 Frankfurt am Main, Germany
- Goethe University Frankfurt, Department of Pediatrics, 60590 Frankfurt am Main, Germany
| | - Jan-Henning Klusmann
- German Cancer Consortium (DKTK), partner site Frankfurt am Main, a partnership between DKFZ and University Hospital and Georg-Speyer-Haus, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), 60596 Frankfurt am Main, Germany
- Universitäres Centrum für Tumorerkrankungen (UCT) Frankfurt Marburg, 60590 Frankfurt am Main, Germany
- Goethe University Frankfurt, Department of Pediatrics, 60590 Frankfurt am Main, Germany
| | - Sjoerd J.L. van Wijk
- Institute for Experimental Paediatric Haematology and Oncology (EPOH), 60528 Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), partner site Frankfurt am Main, a partnership between DKFZ and University Hospital and Georg-Speyer-Haus, Frankfurt am Main, Germany
- Universitäres Centrum für Tumorerkrankungen (UCT) Frankfurt Marburg, 60590 Frankfurt am Main, Germany
| | - Eva Rettinger
- Goethe University Frankfurt, Department of Pediatrics, Division of Stem Cell Transplantation and Immunology, 60590 Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), partner site Frankfurt am Main, a partnership between DKFZ and University Hospital and Georg-Speyer-Haus, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), 60596 Frankfurt am Main, Germany
- Universitäres Centrum für Tumorerkrankungen (UCT) Frankfurt Marburg, 60590 Frankfurt am Main, Germany
| |
Collapse
|
2
|
Milosevic E, Novkovic M, Cenni V, Bavelloni A, Kojic S, Jasnic J. Molecular characterization of ANKRD1 in rhabdomyosarcoma cell lines: expression, localization, and proteasomal degradation. Histochem Cell Biol 2024; 161:435-444. [PMID: 38396247 DOI: 10.1007/s00418-024-02272-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/11/2024] [Indexed: 02/25/2024]
Abstract
Rhabdomyosarcoma (RMS) is the most common soft tissue malignancy in children and adolescents. Respecting the age of the patients and the tumor aggressiveness, investigation of the molecular mechanisms of RMS tumorigenesis is directed toward the identification of novel therapeutic targets. To contribute to a better understanding of the molecular pathology of RMS, we investigated ankyrin repeat domain 1 (ANKRD1), designated as a potential marker for differential diagnostics. In this study, we used three RMS cell lines (SJRH30, RD, and HS-729) to assess its expression profile, intracellular localization, and turnover. They express wild-type ANKRD1, as judged by the sequencing of the open reading frame. Each cell line expressed a different amount of ANKRD1 protein, although the transcript level was similar. According to western blot analysis, ANKRD1 protein was expressed at detectable levels in the SJRH30 and RD cells (SJRH30 > RD), but not in the HS-729, even after immunoprecipitation. Immunocytochemistry revealed nuclear and cytoplasmic localization of ANKRD1 in all examined cell lines. Moreover, the punctate pattern of ANKRD1 staining in the nuclei of RD and HS-729 cells overlapped with coilin, indicating its association with Cajal bodies. We have shown that RMS cells are not able to overexpress ANKRD1 protein, which can be attributed to its proteasomal degradation. The unsuccessful attempt to overexpress ANKRD1 in RMS cells indicates the possibility that its overexpression may have detrimental effects for RMS cells and opens a window for further research into its role in RMS pathogenesis and for potential therapeutic targeting.
Collapse
Affiliation(s)
- Emilija Milosevic
- Laboratory for Molecular Biology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, 11042, Belgrade, Serbia
| | - Mirjana Novkovic
- Laboratory for Molecular Biology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, 11042, Belgrade, Serbia
| | - Vittoria Cenni
- CNR Institute of Molecular Genetics "Luigi-Luca Cavalli-Sforza" Unit of Bologna, Via di Barbiano 1/10, 40136, Bologna, Italy
- IRCCS, Istituto Ortopedico Rizzoli, 40136, Bologna, Italy
| | - Alberto Bavelloni
- Laboratory of Experimental Oncology, IRCCS, Istituto Ortopedico Rizzoli, 40136, Bologna, Italy
| | - Snezana Kojic
- Laboratory for Molecular Biology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, 11042, Belgrade, Serbia.
| | - Jovana Jasnic
- Laboratory for Molecular Biology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, 11042, Belgrade, Serbia.
| |
Collapse
|
3
|
Meister MT, Groot Koerkamp MJA, de Souza T, Breunis WB, Frazer‐Mendelewska E, Brok M, DeMartino J, Manders F, Calandrini C, Kerstens HHD, Janse A, Dolman MEM, Eising S, Langenberg KPS, van Tuil M, Knops RRG, van Scheltinga ST, Hiemcke‐Jiwa LS, Flucke U, Merks JHM, van Noesel MM, Tops BBJ, Hehir‐Kwa JY, Kemmeren P, Molenaar JJ, van de Wetering M, van Boxtel R, Drost J, Holstege FCP. Mesenchymal tumor organoid models recapitulate rhabdomyosarcoma subtypes. EMBO Mol Med 2022; 14:e16001. [PMID: 35916583 PMCID: PMC9549731 DOI: 10.15252/emmm.202216001] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 07/11/2022] [Accepted: 07/12/2022] [Indexed: 02/05/2023] Open
Abstract
Rhabdomyosarcomas (RMS) are mesenchyme-derived tumors and the most common childhood soft tissue sarcomas. Treatment is intense, with a nevertheless poor prognosis for high-risk patients. Discovery of new therapies would benefit from additional preclinical models. Here, we describe the generation of a collection of 19 pediatric RMS tumor organoid (tumoroid) models (success rate of 41%) comprising all major subtypes. For aggressive tumors, tumoroid models can often be established within 4-8 weeks, indicating the feasibility of personalized drug screening. Molecular, genetic, and histological characterization show that the models closely resemble the original tumors, with genetic stability over extended culture periods of up to 6 months. Importantly, drug screening reflects established sensitivities and the models can be modified by CRISPR/Cas9 with TP53 knockout in an embryonal RMS model resulting in replicative stress drug sensitivity. Tumors of mesenchymal origin can therefore be used to generate organoid models, relevant for a variety of preclinical and clinical research questions.
Collapse
Affiliation(s)
- Michael T Meister
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands,Oncode InstituteUtrechtThe Netherlands
| | - Marian J A Groot Koerkamp
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands,Oncode InstituteUtrechtThe Netherlands
| | - Terezinha de Souza
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands,Oncode InstituteUtrechtThe Netherlands
| | - Willemijn B Breunis
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands,Department of Oncology and Children's Research CenterUniversity Children's Hospital ZürichZürichSwitzerland
| | - Ewa Frazer‐Mendelewska
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands,Oncode InstituteUtrechtThe Netherlands
| | - Mariël Brok
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands,Oncode InstituteUtrechtThe Netherlands
| | - Jeff DeMartino
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands,Oncode InstituteUtrechtThe Netherlands
| | - Freek Manders
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands,Oncode InstituteUtrechtThe Netherlands
| | - Camilla Calandrini
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands,Oncode InstituteUtrechtThe Netherlands
| | | | - Alex Janse
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
| | - M Emmy M Dolman
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands,Children's Cancer Institute, Lowy Cancer CentreUNSW SydneyKensingtonNSWAustralia,School of Women's and Children's Health, Faculty of MedicineUNSW SydneyKensingtonNSWAustralia
| | - Selma Eising
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
| | | | - Marc van Tuil
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
| | - Rutger R G Knops
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
| | | | | | - Uta Flucke
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
| | | | - Max M van Noesel
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
| | | | | | - Patrick Kemmeren
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands,Center for Molecular MedicineUMC Utrecht and Utrecht UniversityUtrechtThe Netherlands
| | - Jan J Molenaar
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
| | - Marc van de Wetering
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands,Oncode InstituteUtrechtThe Netherlands
| | - Ruben van Boxtel
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands,Oncode InstituteUtrechtThe Netherlands
| | - Jarno Drost
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands,Oncode InstituteUtrechtThe Netherlands
| | - Frank C P Holstege
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands,Center for Molecular MedicineUMC Utrecht and Utrecht UniversityUtrechtThe Netherlands
| |
Collapse
|
4
|
Qi L, Xu R, Ren X, Zhang W, Yang Z, Tu C, Li Z. Comprehensive Profiling Reveals Prognostic and Immunogenic Characteristics of Necroptosis in Soft Tissue Sarcomas. Front Immunol 2022; 13:877815. [PMID: 35663937 PMCID: PMC9159500 DOI: 10.3389/fimmu.2022.877815] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 04/14/2022] [Indexed: 12/31/2022] Open
Abstract
Soft tissue sarcomas (STSs) are heterogeneous malignancies derived from mesenchymal cells. Due to its rarity, heterogeneity, and limited overall response to chemotherapy, STSs represent a therapeutic challenge. Necroptosis is a novel therapeutic strategy for enhancing immunotherapy of cancer. Nevertheless, no research has explored the relationship between necroptosis-related genes (NRGs) and STSs. In this study, differentially expressed NRGs were identified using The Cancer Genome Atlas (TCGA) and The Cancer Genotype-Tissue Expression (GTEx) project. The expression levels of 34 NRGs were significantly different. Several key NRGs were validated using RT-qPCR and our own sequencing data. Patients with STSs were divided into two clusters using consensus cluster analysis, and significant differences were observed in their survival (p=0.002). We found the differentially expressed genes (DEGs) between the two clusters and carried out subsequent analysis. The necroptosis-related gene signatures with 10 key DEGs were identified with a risk score constructed. The prognosis of TCGA-SARC cohort with low necroptosis-related risk score was better (p<0.001). Meanwhile, the low-risk group had a significantly increased immune infiltration. Using the data of GSE17118 and another immunotherapy cohort as external validations, we observed significant survival differences between the two risk groups (p=0.019). The necroptosis-related risk score proved to be an independent prognostic factor, and a nomogram was further established and integrated with other clinical features. Notably, the necroptosis-related gene signature could also act as the prognostic indicator in other malignancies based on pan-cancer analysis. In summary, the study outlines NRGs in STSs and their potential role in prognosis and will be one of the important directions for future research.
Collapse
Affiliation(s)
- Lin Qi
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Changsha, China
| | - Ruiling Xu
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Changsha, China
| | - Xiaolei Ren
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Changsha, China
| | - Wenchao Zhang
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Changsha, China
| | - Zhimin Yang
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Changsha, China.,Department of Microbiology, Immunology & Molecular Genetics, UT Health Science Center, University of Texas Long School of Medicine, San Antonio, TX, United States
| | - Chao Tu
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Changsha, China
| | - Zhihong Li
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Changsha, China
| |
Collapse
|
5
|
Nefedova E, Koptev V, Bobikova AS, Cherepushkina V, Mironova T, Afonyushkin V, Shkil N, Donchenko N, Kozlova Y, Sigareva N, Davidova N, Bogdanchikova N, Pestryakov A, Toledano-Magaña Y. The Infectious Bronchitis Coronavirus Pneumonia Model Presenting a Novel Insight for the SARS-CoV-2 Dissemination Route. Vet Sci 2021; 8:239. [PMID: 34679068 PMCID: PMC8540477 DOI: 10.3390/vetsci8100239] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/10/2021] [Accepted: 10/12/2021] [Indexed: 12/13/2022] Open
Abstract
Infectious bronchitis (IB) of chickens is a highly contagious disease characterized by damage of the respiratory system and reproductive organs in young animals caused by a virus of the genus Gamma coronavirus. The condition of the respiratory system caused by the IB virus in chickens has many similarities with the pathology of the respiratory system caused by SARS-CoV-2 in humans. The effectiveness of virucidal drugs (Argovit, Triviron, Ecocid, and lauric acid monoglyceride) was tested on chickens inoculated with a tenfold dose of a vaccine strain based on the attenuated virus H120 against IB of chickens. On the 6th day after inoculation, inflammatory changes in the intestines, lungs, and thymus were observed in the control group. The experimental groups were characterized by less pronounced inflammatory reactions and a lower proportion of thymus and lung probes containing genomic IB virus RNA. Since the virucidal activity of four orally administrated formulations was possible only in the intestine, the experimental data indirectly confirmed the hypothesis of the possibility of the predominant accumulation of coronaviruses in the intestine and subsequent lung damage due to the hematogenous redistribution of viral particles and IBV antigens. It was suggested that other coronaviruses including SARS-CoV-2 can implement a similar mechanism.
Collapse
Affiliation(s)
- Ekaterina Nefedova
- Siberian Federal Scientific Center of Agro-BioTechnologies of Russian Academy of Sciences, Novosibirsk Region, Novosibirsk District, 630501 Krasnoobsk, Russia; (E.N.); (V.K.); (A.S.B.); (V.C.); (T.M.); (V.A.); (N.S.); (N.D.); (N.D.)
| | - Vyacheslav Koptev
- Siberian Federal Scientific Center of Agro-BioTechnologies of Russian Academy of Sciences, Novosibirsk Region, Novosibirsk District, 630501 Krasnoobsk, Russia; (E.N.); (V.K.); (A.S.B.); (V.C.); (T.M.); (V.A.); (N.S.); (N.D.); (N.D.)
| | - Anna S. Bobikova
- Siberian Federal Scientific Center of Agro-BioTechnologies of Russian Academy of Sciences, Novosibirsk Region, Novosibirsk District, 630501 Krasnoobsk, Russia; (E.N.); (V.K.); (A.S.B.); (V.C.); (T.M.); (V.A.); (N.S.); (N.D.); (N.D.)
- Molecular Biology Department, Federal State Budgetary Educational Institution Higher Education Novosibirsk State Agrarian University, 630090 Novosibirsk, Russia;
| | - Viktoria Cherepushkina
- Siberian Federal Scientific Center of Agro-BioTechnologies of Russian Academy of Sciences, Novosibirsk Region, Novosibirsk District, 630501 Krasnoobsk, Russia; (E.N.); (V.K.); (A.S.B.); (V.C.); (T.M.); (V.A.); (N.S.); (N.D.); (N.D.)
| | - Tatyana Mironova
- Siberian Federal Scientific Center of Agro-BioTechnologies of Russian Academy of Sciences, Novosibirsk Region, Novosibirsk District, 630501 Krasnoobsk, Russia; (E.N.); (V.K.); (A.S.B.); (V.C.); (T.M.); (V.A.); (N.S.); (N.D.); (N.D.)
- Molecular Biology Department, Federal State Budgetary Educational Institution Higher Education Novosibirsk State Agrarian University, 630090 Novosibirsk, Russia;
| | - Vasily Afonyushkin
- Siberian Federal Scientific Center of Agro-BioTechnologies of Russian Academy of Sciences, Novosibirsk Region, Novosibirsk District, 630501 Krasnoobsk, Russia; (E.N.); (V.K.); (A.S.B.); (V.C.); (T.M.); (V.A.); (N.S.); (N.D.); (N.D.)
- Institute of Chemical Biology and Fundamental Medicine SB RAS, 630090 Novosibirsk, Russia;
| | - Nikolai Shkil
- Siberian Federal Scientific Center of Agro-BioTechnologies of Russian Academy of Sciences, Novosibirsk Region, Novosibirsk District, 630501 Krasnoobsk, Russia; (E.N.); (V.K.); (A.S.B.); (V.C.); (T.M.); (V.A.); (N.S.); (N.D.); (N.D.)
| | - Nikolai Donchenko
- Siberian Federal Scientific Center of Agro-BioTechnologies of Russian Academy of Sciences, Novosibirsk Region, Novosibirsk District, 630501 Krasnoobsk, Russia; (E.N.); (V.K.); (A.S.B.); (V.C.); (T.M.); (V.A.); (N.S.); (N.D.); (N.D.)
| | - Yulia Kozlova
- Institute of Chemical Biology and Fundamental Medicine SB RAS, 630090 Novosibirsk, Russia;
| | - Natalia Sigareva
- Molecular Biology Department, Federal State Budgetary Educational Institution Higher Education Novosibirsk State Agrarian University, 630090 Novosibirsk, Russia;
| | - Natalia Davidova
- Siberian Federal Scientific Center of Agro-BioTechnologies of Russian Academy of Sciences, Novosibirsk Region, Novosibirsk District, 630501 Krasnoobsk, Russia; (E.N.); (V.K.); (A.S.B.); (V.C.); (T.M.); (V.A.); (N.S.); (N.D.); (N.D.)
| | - Nina Bogdanchikova
- Centro de Nanociencias y Nanotecnología, Universidad Nacional Autónoma de México, Ensenada 22860, BC, Mexico
| | - Alexey Pestryakov
- Research School of Chemistry & Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634050 Tomsk, Russia;
| | - Yanis Toledano-Magaña
- Escuela de Ciencias de la Salud Unidad Valle Dorado, Universidad Autónoma de Baja California, Ensenada 22890, BC, Mexico
| |
Collapse
|
6
|
Tsuchiya R, Yoshimatsu Y, Noguchi R, Sin Y, Ono T, Sei A, Takeshita F, Sugaya J, Nakatani F, Yoshida A, Ohtori S, Kawai A, Kondo T. Establishment and characterization of NCC-ssRMS2-C1: a novel patient-derived cell line of spindle cell/sclerosing rhabdomyosarcoma. Hum Cell 2021; 34:1569-1578. [PMID: 34164773 DOI: 10.1007/s13577-021-00569-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Accepted: 06/15/2021] [Indexed: 11/24/2022]
Abstract
Spindle cell/sclerosing rhabdomyosarcoma (ssRMS) is a rare subtype of rhabdomyosarcoma (RMS) that has fascicular spindle cell and/or sclerosing morphology. SsRMS has a diverse molecular background and is categorized into three groups: congenital/infantile ssRMS with a gene fusion involving the NCOA2 and VGLL2, ssRMS with the MYOD1 mutation, and ssRMS with no recurrent identifiable genetic alterations. Because ssRMS is a newly defined disease concept of RMS, the optimal treatment methods have not been determined. This results in unfavorable prognosis and consequently signals the urgent need for continuous research. Patient-derived cell lines are essential tools in basic and translational research. However, only two ssRMS cell lines with the MYOD1 mutation have been reported to date. Thus, we established a novel ssRMS cell line named NCC-ssRMS2-C1 using a surgically resected tumor tissue from an adult ssRMS patient. NCC-ssRMS2-C1 cells retained the copy number alterations corresponding to the original tumor and are categorized into the group with no recurrent identifiable genetic alterations. NCC-ssRMS2-C1 cells demonstrated constant proliferation, spheroid formation, and capability for invasion in vitro, reflecting the malignant features of the original tumor tissue. In a drug screening test, ssRMS demonstrated remarkable sensitivity to romidepsin, trabectedin, actinomycin D, and bortezomib. Hence, we conclude that the NCC-ssRMS2-C1 cell line is the first ssRMS cell line which belongs to the group with no recurrent identifiable genetic alterations, and it will be a useful resource in both basic and translational studies for ssRMS.
Collapse
Affiliation(s)
- Ryuto Tsuchiya
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan.,Department of Orthopaedic Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan
| | - Yuki Yoshimatsu
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Rei Noguchi
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Yooksil Sin
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Takuya Ono
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Akane Sei
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Fumitaka Takeshita
- Department of Translational Oncology, Fundamental Innovative Oncology Core Center, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Jun Sugaya
- Department of Musculoskeletal Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Fumihiko Nakatani
- Department of Musculoskeletal Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Akihiko Yoshida
- Department of Diagnostic Pathology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Seiji Ohtori
- Department of Orthopaedic Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan
| | - Akira Kawai
- Department of Musculoskeletal Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Tadashi Kondo
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan.
| |
Collapse
|
7
|
Harris MA, Miles MA, Shekhar TM, Cerra C, Georgy SR, Ryan SD, Cannon CM, Hawkins CJ. The Proteasome Inhibitor Ixazomib Inhibits the Formation and Growth of Pulmonary and Abdominal Osteosarcoma Metastases in Mice. Cancers (Basel) 2020; 12:cancers12051207. [PMID: 32403415 PMCID: PMC7281181 DOI: 10.3390/cancers12051207] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/07/2020] [Accepted: 05/08/2020] [Indexed: 12/17/2022] Open
Abstract
Osteosarcoma is the most common form of primary bone cancer. Over 20% of osteosarcoma patients present with pulmonary metastases at diagnosis, and nearly 70% of these patients fail to respond to treatment. Previous work revealed that human and canine osteosarcoma cell lines are extremely sensitive to the therapeutic proteasome inhibitor bortezomib in vitro. However, bortezomib has proven disappointingly ineffective against solid tumors including sarcomas in animal experiments and clinical trials. Poor tumor penetration has been speculated to account for the inconsistency between in vitro and in vivo responses of solid tumors to bortezomib. Here we show that the second-generation proteasome inhibitor ixazomib, which reportedly has enhanced solid tumor penetration compared to bortezomib, is toxic to human and canine osteosarcoma cells in vitro. We used experimental osteosarcoma metastasis models to compare the efficacies of ixazomib and bortezomib against primary tumors and metastases derived from luciferase-expressing KRIB or 143B human osteosarcoma cell lines in athymic mice. Neither proteasome inhibitor reduced the growth of primary intramuscular KRIB tumors, however both drugs inhibited the growth of established pulmonary metastases created via intravenous inoculation with KRIB cells, which were significantly better vascularized than the primary tumors. Only ixazomib slowed metastases from KRIB primary tumors and inhibited the growth of 143B pulmonary and abdominal metastases, significantly enhancing the survival of mice intravenously injected with 143B cells. Taken together, these results suggest ixazomib exerts better single agent activity against osteosarcoma metastases than bortezomib. These data provide hope that incorporation of ixazomib, or other proteasome inhibitors that penetrate efficiently into solid tumors, into current regimens may improve outcomes for patients diagnosed with metastatic osteosarcoma.
Collapse
Affiliation(s)
- Michael A. Harris
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, 3086 Victoria, Australia; (M.A.H.); (M.A.M.); (T.M.S.); (C.C.)
| | - Mark A. Miles
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, 3086 Victoria, Australia; (M.A.H.); (M.A.M.); (T.M.S.); (C.C.)
| | - Tanmay M. Shekhar
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, 3086 Victoria, Australia; (M.A.H.); (M.A.M.); (T.M.S.); (C.C.)
| | - Carmelo Cerra
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, 3086 Victoria, Australia; (M.A.H.); (M.A.M.); (T.M.S.); (C.C.)
| | - Smitha R. Georgy
- Department of Anatomic Pathology, Faculty of Veterinary and Agricultural Sciences, University of Melbourne, 3010 Victoria, Australia;
| | - Stewart D. Ryan
- Translational Research and Animal Clinical Trial Study Group (TRACTS), Faculty of Veterinary and Agricultural Sciences, University of Melbourne, 3010 Melbourne, Australia; (S.D.R.); (C.M.C.)
| | - Claire M. Cannon
- Translational Research and Animal Clinical Trial Study Group (TRACTS), Faculty of Veterinary and Agricultural Sciences, University of Melbourne, 3010 Melbourne, Australia; (S.D.R.); (C.M.C.)
| | - Christine J. Hawkins
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, 3086 Victoria, Australia; (M.A.H.); (M.A.M.); (T.M.S.); (C.C.)
- Correspondence: ; Tel.: +61-3-9479-2339
| |
Collapse
|
8
|
Kahen E, Yu D, Harrison DJ, Clark J, Hingorani P, Cubitt CL, Reed DR. Identification of clinically achievable combination therapies in childhood rhabdomyosarcoma. Cancer Chemother Pharmacol 2016; 78:313-23. [PMID: 27324022 PMCID: PMC4965487 DOI: 10.1007/s00280-016-3077-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 06/03/2016] [Indexed: 11/29/2022]
Abstract
Purpose Systemic therapy has improved rhabdomyosarcoma event-free and overall survival; however, approximately 40 % of patients will have progressive or recurrent disease which is difficult to cure and remains a considerable challenge. Minimal progress has been made in improving outcomes for metastatic or relapsed RMS due to a lack of effective therapeutic agents. Targeted therapies are likely to be incorporated into regimens which rely on conventional cytotoxic chemotherapy. A system to evaluate novel combinations of interest is needed. Methods In this study, we explored 8 agents, 5 that are routinely used or similar to agents used in the clinical management of RMS and 3 biologically targeted agents with novel mechanisms of action, the Wee1 inhibitor AZD1775, the tyrosine kinase inhibitor cabozantinib, and the proteasome inhibitor bortezomib. All were tested individually at clinically achievable concentrations for activity in 4 RMS cell lines and then for potential synergy in two-drug combinations. Results We found single-agent activity in five of the agents (or their active metabolites) that constitute the standard of care in RMS and for AZD1775 with mean IC50 values of 207 ng/ml, well below clinically achievable levels. In addition, the combination of individual cytotoxic chemotherapeutics currently used for RMS demonstrated largely synergistic activity with higher, but clinically achievable concentrations of AZD1775 in our assays. Conclusions Prioritization of chemotherapeutics in RMS is possible using an in vitro system that can define novel drug combinations worthy of future investigation. AZD1775 exhibits single-agent activity, as well as synergy with conventional cytotoxic chemotherapy, and is a novel targeted agent that warrants further study in RMS. Electronic supplementary material The online version of this article (doi:10.1007/s00280-016-3077-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Elliot Kahen
- Sunshine Project Translational Research Lab, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Diana Yu
- Sunshine Project Translational Research Lab, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | | | - Justine Clark
- Sunshine Project Translational Research Lab, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Pooja Hingorani
- Center for Cancer and Blood Disorders, Phoenix Children's Hospital, Phoenix, AZ, USA
| | - Christopher L Cubitt
- Sunshine Project Translational Research Lab, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.,Chemical Biology and Molecular Medicine Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Damon R Reed
- Sunshine Project Translational Research Lab, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA. .,Chemical Biology and Molecular Medicine Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA. .,Sarcoma Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA. .,Adolescent and Young Adult Program, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, 33612, Tampa, FL, USA.
| |
Collapse
|
9
|
Rello-Varona S, Herrero-Martín D, Lagares-Tena L, López-Alemany R, Mulet-Margalef N, Huertas-Martínez J, Garcia-Monclús S, García Del Muro X, Muñoz-Pinedo C, Tirado OM. The importance of being dead: cell death mechanisms assessment in anti-sarcoma therapy. Front Oncol 2015; 5:82. [PMID: 25905041 PMCID: PMC4387920 DOI: 10.3389/fonc.2015.00082] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 03/21/2015] [Indexed: 12/23/2022] Open
Abstract
Cell death can occur through different mechanisms, defined by their nature and physiological implications. Correct assessment of cell death is crucial for cancer therapy success. Sarcomas are a large and diverse group of neoplasias from mesenchymal origin. Among cell death types, apoptosis is by far the most studied in sarcomas. Albeit very promising in other fields, regulated necrosis and other cell death circumstances (as so-called "autophagic cell death" or "mitotic catastrophe") have not been yet properly addressed in sarcomas. Cell death is usually quantified in sarcomas by unspecific assays and in most cases the precise sequence of events remains poorly characterized. In this review, our main objective is to put into context the most recent sarcoma cell death findings in the more general landscape of different cell death modalities.
Collapse
Affiliation(s)
- Santiago Rello-Varona
- Sarcoma Research Group, Molecular Oncology Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat , Barcelona , Spain
| | - David Herrero-Martín
- Sarcoma Research Group, Molecular Oncology Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat , Barcelona , Spain
| | - Laura Lagares-Tena
- Sarcoma Research Group, Molecular Oncology Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat , Barcelona , Spain
| | - Roser López-Alemany
- Sarcoma Research Group, Molecular Oncology Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat , Barcelona , Spain
| | - Núria Mulet-Margalef
- Sarcoma Research Group, Molecular Oncology Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat , Barcelona , Spain
| | - Juan Huertas-Martínez
- Sarcoma Research Group, Molecular Oncology Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat , Barcelona , Spain
| | - Silvia Garcia-Monclús
- Sarcoma Research Group, Molecular Oncology Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat , Barcelona , Spain
| | - Xavier García Del Muro
- Sarcoma Research Group, Molecular Oncology Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat , Barcelona , Spain
| | - Cristina Muñoz-Pinedo
- Cell Death Regulation Group, Molecular Oncology Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat , Barcelona , Spain
| | - Oscar Martínez Tirado
- Sarcoma Research Group, Molecular Oncology Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat , Barcelona , Spain
| |
Collapse
|
10
|
Mairinger FD, Walter RFH, Theegarten D, Hager T, Vollbrecht C, Christoph DC, Worm K, Ting S, Werner R, Stamatis G, Mairinger T, Baba H, Zarogoulidis K, Huang H, Li Q, Tsakiridis K, Zarogoulidis P, Schmid KW, Wohlschlaeger J. Gene Expression Analysis of the 26S Proteasome Subunit PSMB4 Reveals Significant Upregulation, Different Expression and Association with Proliferation in Human Pulmonary Neuroendocrine Tumours. J Cancer 2014; 5:646-54. [PMID: 25157275 PMCID: PMC4142326 DOI: 10.7150/jca.9955] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2014] [Accepted: 06/20/2014] [Indexed: 12/23/2022] Open
Abstract
Background: Proteasomal subunit PSMB4 was suggested to be a survival gene in an animal model of hepatocellular carcinoma and in glioblastoma cell lines. In pulmonary adenocarcinoma, a high expression of these genes was found to be associated with poor differentiation and survival. This study investigates the gene expression levels of 26S proteasome subunits in human pulmonary neuroendocrine tumours including typical (TC) and atypical (AC) carcinoid tumours as well as small cell (SCLC) and large cell (LCNEC) neuroendocrine carcinomas. Material and methods: Gene expression levels of proteasomal subunits (PSMA1, PSMA5, PSMB4, PSMB5 and PSMD1) were investigated in 80 neuroendocrine pulmonary tumours (each 20 TC, AC, LCNLC and SCLC) and compared to controls. mRNA levels were determined by using TaqMan assays. Immunohistochemistry on tissue microarrays (TMA) was performed to determine the expression of ki67, cleaved caspase 3 and PSMB4. Results: All proteasomal subunit gene expressions were significantly upregulated in TC, AC, SCLC and LCNEC compared to controls. PSMB4 mRNA is differently expressed between all neuroendocrine tumour subtypes demonstrating the highest expression and greatest range in LCNEC (p=0.043), and is significantly associated with proliferative activity (p=0.039). Conclusion: In line with other 26S proteasomal subunits PSMB4 is significantly increased, but differently expressed between pulmonary neuroendocrine tumours and is associated with the proliferative activity. Unlike in pulmonary adenocarcinomas, no association with biological behaviour was observed, suggesting that increased proteasomal subunit gene expression is a common and probably early event in the tumorigenesis of pulmonary neuroendocrine tumours regardless of their differentiation.
Collapse
Affiliation(s)
| | - Robert Fred Henry Walter
- 1. Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Germany. ; 2. Ruhrlandklinik, West German Lung Centre, University Hospital Essen, University of Duisburg-Essen, Germany
| | - Dirk Theegarten
- 1. Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Germany
| | - Thomas Hager
- 1. Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Germany
| | - Claudia Vollbrecht
- 3. Institute of Pathology, University Hospital Cologne, University of Cologne, Germany
| | | | - Karl Worm
- 1. Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Germany
| | - Saskia Ting
- 1. Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Germany
| | - Robert Werner
- 1. Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Germany
| | - Georgios Stamatis
- 5. Department of Thoracic Surgery, Ruhrlandklinik, West German Lung Centre, University Hospital Essen, University of Duisburg-Essen, Germany
| | - Thomas Mairinger
- 6. Department of Pathology, Helios Klinikum Emil von Behring, Berlin, Germany
| | - Hideo Baba
- 1. Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Germany
| | - Konstantinos Zarogoulidis
- 7. Pulmonary-Oncology, ``G. Papanikolaou`` General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Haidong Huang
- 8. Department of Respiratory Diseases, Changhai Hospital/First Affiliated Hospital of the Second Military Medical University, Shanghai, People's Republic of China, China
| | - Qiang Li
- 8. Department of Respiratory Diseases, Changhai Hospital/First Affiliated Hospital of the Second Military Medical University, Shanghai, People's Republic of China, China
| | - Kosmas Tsakiridis
- 9. Thoracic Surgery Department, ``Saint Luke`` Private Clinic, Thessaloniki, Panorama, Greece
| | - Paul Zarogoulidis
- 7. Pulmonary-Oncology, ``G. Papanikolaou`` General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Kurt Werner Schmid
- 1. Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Germany
| | - Jeremias Wohlschlaeger
- 1. Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Germany
| |
Collapse
|
11
|
Anti-cancer drug KP1019 induces Hog1 phosphorylation and protein ubiquitylation in Saccharomyces cerevisiae. Eur J Pharmacol 2014; 736:77-85. [DOI: 10.1016/j.ejphar.2014.04.032] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 04/22/2014] [Accepted: 04/23/2014] [Indexed: 01/29/2023]
|
12
|
Cao MN, Zhou YB, Gao AH, Cao JY, Gao LX, Sheng L, Xu L, Su MB, Cao XC, Han MM, Wang MK, Li J. Curcusone D, a novel ubiquitin–proteasome pathway inhibitor via ROS-induced DUB inhibition, is synergistic with bortezomib against multiple myeloma cell growth. Biochim Biophys Acta Gen Subj 2014; 1840:2004-13. [DOI: 10.1016/j.bbagen.2014.02.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2013] [Revised: 01/24/2014] [Accepted: 02/04/2014] [Indexed: 12/13/2022]
|
13
|
Škrott Z, Cvek B. Linking the activity of bortezomib in multiple myeloma and autoimmune diseases. Crit Rev Oncol Hematol 2014; 92:61-70. [PMID: 24890785 DOI: 10.1016/j.critrevonc.2014.05.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 04/25/2014] [Accepted: 05/02/2014] [Indexed: 01/12/2023] Open
Abstract
Since their introduction to the clinic 10 years ago, proteasome inhibitors have become the cornerstone of anti-multiple myeloma therapy. Despite significant progress in understanding the consequences of proteasome inhibition, the unique activity of bortezomib is still unclear. Disappointing results from clinical trials with bortezomib in other malignancies raise the question of what makes multiple myeloma so sensitive to proteasome inhibition. Successful administration of bortezomib in various immunological disorders that exhibit high antibody production suggests that the balance between protein synthesis and degradation is a key determinant of sensitivity to proteasome inhibition because a high rate of protein production is a shared characteristic in plasma and myeloma cells. Initial or acquired resistance to bortezomib remains a major obstacle in the clinic as in vitro data from cell lines suggest a key role for the β5 subunit mutation in resistance; however the mutation was not found in patient samples. Recent studies indicate the importance of selecting for a subpopulation of cells that produce lower amounts of paraprotein during bortezomib therapy.
Collapse
Affiliation(s)
- Zdeněk Škrott
- Department of Cell Biology and Genetics, Faculty of Science, Palacky University, Slechtitelu 11, 78371 Olomouc, Czech Republic
| | - Boris Cvek
- Department of Cell Biology and Genetics, Faculty of Science, Palacky University, Slechtitelu 11, 78371 Olomouc, Czech Republic.
| |
Collapse
|
14
|
PAX3-FOXO1 induces up-regulation of Noxa sensitizing alveolar rhabdomyosarcoma cells to apoptosis. Neoplasia 2014; 15:738-48. [PMID: 23814486 DOI: 10.1593/neo.121888] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Revised: 04/05/2013] [Accepted: 04/11/2013] [Indexed: 02/08/2023] Open
Abstract
Alveolar rhabdomyosarcoma (ARMS) has a much poorer prognosis than the more common embryonal subtype. Most ARMS tumors characteristically possess a specific genomic translocation between the genes of PAX3/7 and FOXO1 (FKHR), which forms fusion proteins possessing the DNA binding domains of PAX3/7 and the more transcriptionally potent transactivation domain of FOXO1. We have shown that the proapoptotic BH3-only family member Noxa is upregulated by the PAX3-FOXO1 fusion transcription factor in a p53-independent manner. The increased expression of Noxa renders PAX3-FOXO1-expressing cells more susceptible to apoptosis induced by a γ-secretase inhibitor (GSI1, Z-LLNle-CHO), the proteasome inhibitor bortezomib, and BH3 mimetic ABT-737. Apoptosis in response to bortezomib can be overcome by shRNA knockdown of Noxa. In vivo treatment with bortezomib reduced the growth of tumors derived from a PAX3-FOXO1-expressing primary myoblast tumor model and RH41 xenografts. We therefore demonstrate that PAX3-FOXO1 up-regulation of Noxa represents an unanticipated aspect of ARMS tumor biology that creates a therapeutic window to allow induction of apoptosis in ARMS cells.
Collapse
|
15
|
Constantinidou A, Pollack S, Loggers E, Rodler E, Jones RL. The evolution of systemic therapy in sarcoma. Expert Rev Anticancer Ther 2013; 13:211-23. [PMID: 23406562 DOI: 10.1586/era.12.161] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Approximately 50% of patients with localized soft tissue sarcomas will develop recurrent disease after complete surgical resection, requiring alternative means of treatment. Conventional chemotherapy comprising of doxorubicin and ifosfamide has shown benefit in advanced disease, however, there remains a clear need for more effective, less toxic, therapies for the treatment of this heterogeneous group of mesenchymal malignancies. Recently, greater emphasis has been placed on the underlying biology of individual sarcoma subtypes, with the development and evaluation of novel therapies both in common and in rare subtypes. In addition, there is a greater specificity in the selection of chemotherapy agents based on activity in individual histological subtypes. Despite these advances the management of sarcoma, and in particular of rare subtypes, remains a major challenge. Some histological subtypes are resistant to conventional chemotherapy and patients with these diseases should be offered participation in early phase clinical trials of novel drugs.
Collapse
|
16
|
BAG3 induction is required to mitigate proteotoxicity via selective autophagy following inhibition of constitutive protein degradation pathways. Oncogene 2013; 33:1713-24. [PMID: 23644654 DOI: 10.1038/onc.2013.110] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Revised: 01/21/2013] [Accepted: 02/07/2013] [Indexed: 01/02/2023]
Abstract
Simultaneous inhibition of the two major constitutive protein quality control (PQC) pathways, that is, the ubiquitin-proteasome system (UPS) and the aggresome-autophagy system, has been suggested as a promising strategy to trigger cell death in cancer cells. However, we observed that one third of rhabdomyosarcoma (RMS) cells survives parallel inhibition of the UPS by Bortezomib and the aggresome-autophagy pathway by the cytoplasmic histone deacetylase 6 inhibitor ST80, and is able to regrow upon drug removal, thus pointing to the induction of compensatory pathways. Here, we identify Bcl-2-associated athanogene 3 (BAG3) as a critical mediator of inducible resistance in surviving cells after concomitant blockage of constitutive PQC pathways by mitigating ST80/Bortezomib-triggered proteotoxicity via selective autophagy. ST80/Bortezomib cotreatment upregulates BAG3 mRNA and protein levels in surviving cells in addition to triggering the accumulation of insoluble protein aggregates. Intriguingly, knockdown of BAG3 by RNA interference severely impairs clearance of protein aggregates, significantly increases cell death and reduces long-term survival and clonogenic growth during recovery after ST80/Bortezomib cotreatment. Similarly, inhibition of autophagy by inducible autophagy-related protein 7 knockdown prevents removal of protein aggregates and cell regrowth during recovery after ST80/Bortezomib cotreatment. Also, the inhibition of lysosomal degradation using the V-ATPase pump inhibitor Bafilomycin A1 enhances accumulation of protein aggregates, and completely abolishes regrowth after Bortezomib/ST80-induced proteotoxic stress. By identifying BAG3 as a key mediator of inducible resistance by mitigating proteotoxicity via selective autophagy after inhibition of constitutive PQC systems, our study provides new insights into the regulation of PQC pathways in cancer cells and identifies new targets for therapeutic intervention.
Collapse
|
17
|
Xu S, De Becker A, De Raeve H, Van Camp B, Vanderkerken K, Van Riet I. In vitro expanded bone marrow-derived murine (C57Bl/KaLwRij) mesenchymal stem cells can acquire CD34 expression and induce sarcoma formation in vivo. Biochem Biophys Res Commun 2012; 424:391-7. [PMID: 22771324 DOI: 10.1016/j.bbrc.2012.06.118] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Accepted: 06/25/2012] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cells (MSCs) have currently generated numerous interests in pre-clinical and clinical applications due to their multiple lineages differentiation potential and immunomodulary effects. However, accumulating evidence indicates that MSCs, especially murine MSCs (mMSCs), can undergo spontaneous transformation after long-term in vitro culturing, which might reduce the therapeutic application possibilities of these stem cells. In the present study, we observed that in vitro expanded bone marrow (BM) derived mMSCs from the C57Bl/KaLwRij mouse strain can lose their specific stem cells markers (CD90 and CD105) and acquire CD34 expression, accompanied with an altered morphology and an impaired tri-lineages differentiation capacity. Compared to normal mMSCs, these transformed mMSCs exhibited an increased proliferation rate, an enhanced colony formation and migration ability as well as a higher sensitivity to anti-tumor drugs. Transformed mMSCs were highly tumorigenic in vivo, resulting in aggressive sarcoma formation when transplanted in non-immunocompromised mice. Furthermore, we found that Notch signaling downstream genes (hey1, hey2 and heyL) were significantly upregulated in transformed mMSCs, while Hedgehog signaling downstream genes Gli1 and Ptch1 and the Wnt signaling downstream gene beta-catenin were all decreased. Taken together, we observed that murine in vitro expanded BM-MSCs can transform into CD34 expressing cells that induce sarcoma formation in vivo. We assume that dysregulation of the Notch(+)/Hh(-)/Wnt(-) signaling pathway is associated with the malignant phenotype of the transformed mMSCs.
Collapse
Affiliation(s)
- Song Xu
- Department of Lung Cancer Surgery, Lung Cancer Institute, Tianjin Medical University General Hospital, 300052 Tianjin, China
| | | | | | | | | | | |
Collapse
|
18
|
Peron M, Bonvini P, Rosolen A. Effect of inhibition of the ubiquitin-proteasome system and Hsp90 on growth and survival of rhabdomyosarcoma cells in vitro. BMC Cancer 2012; 12:233. [PMID: 22691173 PMCID: PMC3480867 DOI: 10.1186/1471-2407-12-233] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Accepted: 05/21/2012] [Indexed: 01/04/2023] Open
Abstract
Background The ubiquitin-proteasome system (UPS) and the heat shock response (HSR) are two critical regulators of cell homeostasis, as their inhibition affects growth and survival of normal cells, as well as stress response and invasiveness of cancer cells. We evaluated the effects of the proteasome inhibitor Bortezomib and of 17-DMAG, a competitive inhibitor of Hsp90, in rhabdomyosarcoma (RMS) cells, and analyzed the efficacy of single-agent exposures with combination treatments. Methods To assess cytotoxicity induced by Bortezomib and 17-DMAG in RMS cells, viability was measured by MTT assay after 24, 48 and 72 hours. Western blotting and immunofluorescence analyses were carried out to elucidate the mechanisms of action. Apoptosis was measured by FACS with Annexin-V-FITC and Propidium Iodide. Results Bortezomib and 17-DMAG, when combined at single low-toxic concentrations, enhanced growth inhibition of RMS cells, with signs of autophagy that included intensive cytoplasmic vacuolization and conversion of cytosolic LC3-I protein to its autophagosome-associated form. Treatment with lysosomal inhibitor chloroquine facilitates apoptosis, whereas stimulation of autophagy by rapamycin prevents LC3-I conversion and cell death, suggesting that autophagy is a resistance mechanism in RMS cells exposed to proteotoxic drugs. However, combination treatment also causes caspase-dependent apoptosis, PARP cleavage and Annexin V staining, as simultaneous inhibition of both UPS and HSR systems limits cytoprotective autophagy, exacerbating stress resulting from accumulation of misfolded proteins. Conclusion The combination of proteasome inhibitor Bortezomib with Hsp90 inhibitor 17-DMAG, appears to have important therapeutic advantages in the treatment of RMS cells compared with single-agent exposure, because compensatory survival mechanisms that occur as side effects of treatment may be prevented.
Collapse
Affiliation(s)
- Marica Peron
- Clinica di Oncoematologia Pediatrica, Azienda Ospedaliera-Università di Padova, Via Giustiniani 3, Padova 35128, Italy
| | | | | |
Collapse
|
19
|
Mandili G, Khadjavi A, Gallo V, Minero VG, Bessone L, Carta F, Giribaldi G, Turrini F. Characterization of the protein ubiquitination response induced by Doxorubicin. FEBS J 2012; 279:2182-91. [PMID: 22536838 DOI: 10.1111/j.1742-4658.2012.08602.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Doxorubicin is commonly considered to exert its anti-tumor activity by triggering apoptosis of cancer cells through DNA damage. Recent reports have shown that Doxorubicin elicits a marked heat shock response, and that either inhibition or silencing of heat shock proteins enhance the Doxorubicin apoptotic effect in neuroblastoma cells. In order to investigate whether Doxorubicin may also act through protein modification, we performed a proteomic analysis of ubiquitinated proteins. Here we show that nanomolar Doxorubicin treatment of neuroblastoma cells caused: (a) dose-dependent over-ubiquitination of a specific set of proteins in the absence of measurable inhibition of proteasome, (b) protein ubiquination patterns similar to those with Bortezomib, a proteasome inhibitor, (c) depletion and loss of activity of ubiquitinated enzymes such as lactate dehydrogenase and α-enolase, and (d) a decrease in HSP27 solubility, probably a consequence of its binding to denatured proteins. These data strongly reinforce the hypothesis that Doxorubicin may also exert its effect by damaging proteins.
Collapse
Affiliation(s)
- Giorgia Mandili
- Department of Genetics, Biology and Biochemistry, University of Turin, Turin, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Abstract
PURPOSE OF REVIEW To review the 2008-2009 literature on pediatric genitourinary tumors and highlight the most significant publications. RECENT FINDINGS New techniques such as gene expression profiling, PET, nephron-sparing surgery, and stem cell transplantation are being incorporated into contemporary treatments for pediatric patients with genitourinary tumors. The WTX gene is the most commonly mutated gene in Wilms tumor, and its product enhances Wilms tumor gene 1-mediated transcription. Germline WTX mutations cause an X-linked sclerosing bone dysplasia but do not appear to predispose to Wilms tumor formation. Protocadherin gene clusters on chromosome 5q31 may act as tumor suppressors. In rhabdomyosarcoma, ILK and platelet-derived growth factor receptor-A join the paired box gene 7 and 3-forkhead box O1 fusions as potential therapeutic targets, and muscle-specific microRNAs offer promise as adjuvant therapy. Despite the high cure rate of Wilms tumor, long-term survivors remain at risk of death from various causes. SUMMARY In general, the prognosis for patients with pediatric genitourinary tumors is favorable. The elucidation of the molecular abnormalities in these tumors is determining risk stratification, treatment strategies, and candidates for new drug development.
Collapse
|
21
|
Abstract
Development of chemotherapeutic treatment modalities resulted in a dramatic increase in the survival of children with many types of cancer. Still, in case of some pediatric cancer entities including rhabdomyosarcoma, osteosarcoma and Ewing's sarcoma, survival of patients remains dismal and novel treatment approaches are urgently needed. Therefore, based on the concept of targeted therapy, numerous potential targets for the treatment of these cancers have been evaluated pre-clinically or in some cases even clinically during the last decade. This review gives an overview over many different potential therapeutic targets for treatment of these childhood sarcomas, including receptor tyrosine kinases, intracellular signaling molecules, cell cycle and apoptosis regulators, proteasome, hsp90, histone deacetylases, angiogenesis regulators and sarcoma specific fusion proteins. The large number of potential therapeutic targets suggests that improved comparability of pre-clinical models might be necessary to prioritize the most effective ones for future clinical trials.
Collapse
Affiliation(s)
- Marco Wachtel
- University Children's Hospital, Department of Oncology, Zürich, Switzerland
| | | |
Collapse
|
22
|
Wu WKK, Cho CH, Lee CW, Wu K, Fan D, Yu J, Sung JJY. Proteasome inhibition: a new therapeutic strategy to cancer treatment. Cancer Lett 2010; 293:15-22. [PMID: 20133049 DOI: 10.1016/j.canlet.2009.12.002] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2009] [Revised: 12/01/2009] [Accepted: 12/02/2009] [Indexed: 02/03/2023]
Abstract
The ubiquitin-proteasome system is a major pathway for protein degradation. Targeting this pathway using proteasome inhibitors represents a novel approach for the treatment of cancer. Proteasome inhibitors lower cell proliferation and induce apoptosis in solid and hematologic malignancies through multiple mechanisms, including stabilization of cell cycle regulators and pro-apoptotic factors, stimulation of bone morphogenetic protein signaling, inhibition of protein translation, and sensitization to ligand-induced apoptosis. In this connection, proteasome inhibition activates macroautophagy, a compensatory protein degradation system, as well as other pro-survival signaling pathways. Inhibition of these auto-protective responses sensitizes cancer cells to the anti-proliferative effects of proteasome inhibitors.
Collapse
Affiliation(s)
- William Ka Kei Wu
- Institute of Digestive Diseases, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | | | | | | | | | | | | |
Collapse
|
23
|
Park JA, Kim EK, Kang HJ, Shin HY, Kim IH, Ahn HS. Initial response to treatment was highly associated with the prognosis of childhood rhabdomyosarcoma: a retrospective analysis of a single center experience in Korea. Cancer Res Treat 2009; 40:111-5. [PMID: 19688116 DOI: 10.4143/crt.2008.40.3.111] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2008] [Accepted: 08/07/2008] [Indexed: 02/08/2023] Open
Abstract
PURPOSE Following the introduction of a multimodal approach to diagnosis and treatment, the prognosis of rhabdomyosarcoma (RMS) has markedly improved over the last three decades. However, there are few data on treatment outcomes in Korean patients. MATERIALS AND METHODS We performed a retrospective analysis of 77 patients with RMS diagnosed and treated at Seoul National University Children's Hospital between 1986 and 2005. RESULTS The overall 5-year survival and event-free survival rates for all patients were 77% and 59%, respectively. The Intergroup Rhabdomyosarcoma Study clinical grouping and initial response to treatment (20-week response) were important prognostic factors. CONCLUSIONS The outcome of childhood RMS was closely associated with the initial staging and the initial response to treatment. Modulating therapies according to initial responses and risk factors is critical, and new treatment strategies for high-risk patients are needed.
Collapse
Affiliation(s)
- Jeong A Park
- Department of Pediatrics and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | | | | | | | | | | |
Collapse
|
24
|
PAX3-FKHR sensitizes human alveolar rhabdomyosarcoma cells to camptothecin-mediated growth inhibition and apoptosis. Cancer Lett 2009; 284:157-64. [PMID: 19442434 DOI: 10.1016/j.canlet.2009.04.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2009] [Revised: 03/25/2009] [Accepted: 04/15/2009] [Indexed: 11/22/2022]
Abstract
Patients with alveolar rhabdomyosarcoma (ARMS) have poorer response to conventional chemotherapy and lower survival rates than those with embryonal RMS (ERMS). By high-throughput screening, we identified camptothecin as an ARMS-selective inhibitor. Camptothecin more efficiently inhibited proliferation and induced apoptosis in Rh30 (ARMS) than RD (ERMS) cells. Ectopic expression of the PAX3-FKHR (PF) fusion protein in RD cells significantly increased sensitivity, whereas siRNA knockdown of PF decreased sensitivity of Rh30 cells to camptothecin. The sensitization required a transcriptionally active PF, and camptothecin downregulated levels of PF protein. These findings suggest that it is feasible to develop agents that preferentially block the growth of ARMS.
Collapse
|
25
|
Maruwge W, D'Arcy P, Folin A, Brnjic S, Wejde J, Davis A, Erlandsson F, Bergh J, Brodin B. Sorafenib inhibits tumor growth and vascularization of rhabdomyosarcoma cells by blocking IGF-1R-mediated signaling. Onco Targets Ther 2008; 1:67-78. [PMID: 21127754 PMCID: PMC2994208 DOI: 10.2147/ott.s3833] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The growth of many soft tissue sarcomas is dependent on aberrant growth factor signaling, which promotes their proliferation and motility. With this in mind, we evaluated the effect of sorafenib, a receptor tyrosine kinase inhibitor, on cell growth and apoptosis in sarcoma cell lines of various histological subtypes. We found that sorafenib effectively inhibited cell proliferation in rhabdomyosarcoma, synovial sarcoma and Ewing’s sarcoma with IC50 values <5 μM. Sorafenib effectively induced growth arrest in rhabdomyosarcoma cells, which was concurrent with inhibition of Akt and Erk signaling. Studies of ligand-induced phosphorylation of Erk and Akt in rhabdomyosarcoma cells showed that insulin-like growth factor-1 is a potent activator, which can be blocked by treatment with sorafenib. In vivo sorafenib treatment of rhabdomyosarcoma xenografts had a significant inhibitory effect on tumor growth, which was associated with inhibited vascularization and enhanced necrosis in the adjacent tumor stroma. Our results demonstrate that in vitro and in vivo growth of rhabdomyosarcoma can be suppressed by treatment with sorafenib, and suggests the possibilities of using sorafenib as a potential adjuvant therapy for the treatment of rhabdomyosarcoma.
Collapse
Affiliation(s)
- Wessen Maruwge
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Ordóñez JL, Martins AS, Osuna D, Madoz–Gúrpide J, de Alava E. Targeting sarcomas: therapeutic targets and their rational. Semin Diagn Pathol 2008; 25:304-16. [DOI: 10.1053/j.semdp.2008.07.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|