90 results on '"Armeanu-Ebinger S"'
Search Results
2. Number of predicted tumour-neoantigens as biomarker for cancer immunotherapies
- Author
-
Armeanu-Ebinger, S., primary, Hadaschik, D., additional, Kyzirakos, C., additional, Mohr, C., additional, Battke, F., additional, Kohlbacher, O., additional, Nahnsen, S., additional, and Biskup, S., additional
- Published
- 2017
- Full Text
- View/download PDF
3. Wirkung von Sorafenib auf kindliche Lebertumore
- Author
-
Nagel, C, Warmann, SW, Armeanu-Ebinger, S, and Fuchs, J
- Subjects
ddc: 610 ,610 Medical sciences ,Medicine - Abstract
Einleitung: Hepatoblastome (HB) und pädiatrische hepatozelluläre Karzinome (HCC) sind die häufigsten malignen Lebertumore im Kindesalter. Die Prognose fortgeschrittener oder rezidivierender Tumoren wird im Wesentlichen von der Chemoresistenz bestimmt. Ziel war daher die Evaluation einer[for full text, please go to the a.m. URL], 131. Kongress der Deutschen Gesellschaft für Chirurgie
- Published
- 2014
- Full Text
- View/download PDF
4. Optimized neoantigen selection based on tumor exome data
- Author
-
Kyzirakos, C., primary, Mohr, C., additional, Armeanu-Ebinger, S., additional, Feldhahn, M., additional, Hadaschik, D., additional, Walzer, M., additional, Döcker, D., additional, Menzel, M., additional, Nahnsen, S., additional, Kohlbacher, O., additional, and Biskup, S., additional
- Published
- 2016
- Full Text
- View/download PDF
5. Implementation of an interdisciplinary molecular tumor board in managing of advanced stage breast cancer
- Author
-
Armeanu-Ebinger, S., primary, Döcker, D., additional, Kopic, A., additional, Brucker, S., additional, Wallwiener, D., additional, Martens, U., additional, Kyzirakos, C., additional, Menzel, M., additional, Rinkleb, A., additional, and Biskup, S., additional
- Published
- 2016
- Full Text
- View/download PDF
6. Das BH3-Mimetikum Obatoclax verhindert das Tumorwachstum im orthotopen Mausmodell für Hepatoblastome
- Author
-
Lieber, J, Ellerkamp, V, Vogt, F, Wenz, J, Warmann, S, Fuchs, J, and Armeanu-Ebinger, S
- Subjects
ddc: 610 ,610 Medical sciences ,Medicine - Abstract
Einleitung: Sowohl Resistenzen auf Chemotherapeutika als auch die Metastasierung repräsentieren die größten Herausforderungen bei der Behandlung von hoch-Risiko Hepatoblastomen (HB) und erfordern neue Behandlungsalternativen. Apoptose-Modulatoren wie die BH3-Mimetika Obatoclax und ABT-737[for full text, please go to the a.m. URL], 130. Kongress der Deutschen Gesellschaft für Chirurgie
- Published
- 2013
- Full Text
- View/download PDF
7. P063 Ultra deep panel-based NGS of cancer-relevant genes as treatment decision support
- Author
-
Döcker, D., primary, Menzel, M., additional, Armeanu-Ebinger, S., additional, Rinckleb, A., additional, and Biskup, S., additional
- Published
- 2015
- Full Text
- View/download PDF
8. Optimierte in vivo-Detektion von kindlichen Rhabdomyosarkomen mittels PET-MRT im Tiermodell
- Author
-
Seitz, G, Griesinger, C, Hermann, D, Gneiling, M, Warmann, S, Fuchs, J, Pichler, B, Armeanu-Ebinger, S, Seitz, G, Griesinger, C, Hermann, D, Gneiling, M, Warmann, S, Fuchs, J, Pichler, B, and Armeanu-Ebinger, S
- Published
- 2014
9. 29 - Number of predicted tumour-neoantigens as biomarker for cancer immunotherapies
- Author
-
Armeanu-Ebinger, S., Hadaschik, D., Kyzirakos, C., Mohr, C., Battke, F., Kohlbacher, O., Nahnsen, S., and Biskup, S.
- Published
- 2017
- Full Text
- View/download PDF
10. Panel-Based Next-Generation Sequencing of Cancer-Relevant Genes As Treatment Decision Support
- Author
-
Menzel, M., primary, Armeanu-Ebinger, S., additional, Feldhahn, M., additional, and Biskup, S., additional
- Published
- 2014
- Full Text
- View/download PDF
11. Glutamine depletion by crisantaspase hinders the growth of human hepatocellular carcinoma xenografts
- Author
-
Chiu, M, primary, Tardito, S, additional, Pillozzi, S, additional, Arcangeli, A, additional, Armento, A, additional, Uggeri, J, additional, Missale, G, additional, Bianchi, M G, additional, Barilli, A, additional, Dall'Asta, V, additional, Campanini, N, additional, Silini, E M, additional, Fuchs, J, additional, Armeanu-Ebinger, S, additional, and Bussolati, O, additional
- Published
- 2014
- Full Text
- View/download PDF
12. Synergistische Effekte von Beta-Catenin und YAP führen zur Entstehung von Hepatoblastomen in Mäusen und beim Menschen
- Author
-
Calvisi, DF, primary, Tao, J, additional, Ranganathan, S, additional, Cigliano, A, additional, Jiang, L, additional, Fan, B, additional, Armeanu-Ebinger, S, additional, Delogu, S, additional, Ribback, S, additional, Dombrowski, F, additional, Monga, SPS, additional, Chen, X, additional, and Evert, M, additional
- Published
- 2014
- Full Text
- View/download PDF
13. 1540P - Implementation of an interdisciplinary molecular tumor board in managing of advanced stage breast cancer
- Author
-
Armeanu-Ebinger, S., Döcker, D., Kopic, A., Brucker, S., Wallwiener, D., Martens, U., Kyzirakos, C., Menzel, M., Rinkleb, A., and Biskup, S.
- Published
- 2016
- Full Text
- View/download PDF
14. 1097P - Optimized neoantigen selection based on tumor exome data
- Author
-
Kyzirakos, C., Mohr, C., Armeanu-Ebinger, S., Feldhahn, M., Hadaschik, D., Walzer, M., Döcker, D., Menzel, M., Nahnsen, S., Kohlbacher, O., and Biskup, S.
- Published
- 2016
- Full Text
- View/download PDF
15. Pharmacological inhibition of beta-catenin in hepatoblastoma cells
- Author
-
Ellerkamp, V., primary, Lieber, J., additional, Nagel, C., additional, Wenz, J., additional, Warmann, S. W., additional, Fuchs, J., additional, and Armeanu-Ebinger, S., additional
- Published
- 2012
- Full Text
- View/download PDF
16. Effect of sorafenib combined with cytostatic agents on hepatoblastoma cell lines and xenografts
- Author
-
Eicher, C, primary, Dewerth, A, additional, Thomale, J, additional, Ellerkamp, V, additional, Hildenbrand, S, additional, Warmann, S W, additional, Fuchs, J, additional, and Armeanu-Ebinger, S, additional
- Published
- 2012
- Full Text
- View/download PDF
17. Onkolytische Sendai-Viren als neuartiger Ansatz zur virotherapeutischen Behandlung des HCC
- Author
-
Zimmermann, M, primary, Armeanu-Ebinger, S, additional, Bossow, S, additional, Lampe, J, additional, Smirnow, I, additional, Schenk, A, additional, Lange, S, additional, Weiss, TS, additional, Neubert, W, additional, Malek, NP, additional, Lauer, UM, additional, and Bitzer, M, additional
- Published
- 2012
- Full Text
- View/download PDF
18. Novel oncolytic Sendai viruses (SeV) for the virotherapeutic treatment of HCC
- Author
-
Zimmermann, M, primary, Armeanu-Ebinger, S, additional, Bossow, S, additional, Lampe, J, additional, Smirnow, I, additional, Lange, S, additional, Weiss, TS, additional, Neubert, W, additional, Malek, NP, additional, Lauer, UM, additional, and Bitzer, M, additional
- Published
- 2012
- Full Text
- View/download PDF
19. Sorafenib as a new therapeutic option for Hepatoblastoma
- Author
-
Dewerth, A, primary, Eicher, C, additional, Fuchs, J, additional, and Armeanu-Ebinger, S, additional
- Published
- 2011
- Full Text
- View/download PDF
20. Macrophage-mediated cytotoxicity against rhabdomyosarcoma cells
- Author
-
Herrmann, D, primary, Seitz, G, additional, Warmann, SW, additional, Fuchs, J, additional, and Armeanu-Ebinger, S, additional
- Published
- 2011
- Full Text
- View/download PDF
21. Development of a drug resistance model in hepatoblastoma
- Author
-
Dewerth, A, primary, Eicher, C, additional, Kirchner, B, additional, Warmann, SW, additional, Fuchs, J, additional, and Armeanu-Ebinger, S, additional
- Published
- 2010
- Full Text
- View/download PDF
22. Expression and functional role of miR-200c in rhabdomyosarcoma
- Author
-
Herrmann, D, primary, Seitz, G, additional, Warmann, S, additional, Bonin, M, additional, Leuschner, I, additional, Fuchs, J, additional, and Armeanu-Ebinger, S, additional
- Published
- 2010
- Full Text
- View/download PDF
23. 1169P - Panel-Based Next-Generation Sequencing of Cancer-Relevant Genes As Treatment Decision Support
- Author
-
Menzel, M., Armeanu-Ebinger, S., Feldhahn, M., and Biskup, S.
- Published
- 2014
- Full Text
- View/download PDF
24. Effect of sorafenib combined with cytostatic agents on hepatoblastoma cell lines and xenografts.
- Author
-
Eicher, C, Dewerth, A, Thomale, J, Ellerkamp, V, Hildenbrand, S, Warmann, S W, Fuchs, J, and Armeanu-Ebinger, S
- Subjects
LIVER tumors ,TUMOR treatment ,ANTINEOPLASTIC agents ,COMBINATION drug therapy ,CELL lines ,XENOGRAFTS ,IMMUNOLOGY - Abstract
Background:Sorafenib has recently been shown to reduce tumour growth in hepatoblastoma (HB) xenografts. The effect of a combined administration with cytostatic agents was now investigated.Methods:Cell viability after treatment with sorafenib and different cytostatic agents was evaluated in two HB cell lines (HUH6 and HepT1) using MTT assay. ERK signalling was investigated by western blot, NOXA expression by rt-PCR, and formation of DNA adducts using immunocytology. NMRI mice bearing subcutaneous HUH6-derived tumours were treated with sorafenib alone or in combination with cisplatin. Tumour progression, viability, apoptosis, and vascularisation were monitored by tumour volume, AFP levels, TUNEL assay, and CD31 immunostaining, respectively.Results:The combination of sorafenib and cisplatin led to a remarkable decrease in cell viability. The cisplatin-induced enhanced ERK1/2 activation, but not NOXA expression and the formation of DNA adducts was partly abrogated by sorafenib. In HB xenografts, both, sorafenib and alternated application of sorafenib and cisplatin significantly reduced tumour growth (P<0.05). Levels of AFP were lower in both treated groups (P=0.08). Relative apoptotic areas were increased (P=0.003). Mean vascular density was the lowest in the sorafenib/CDDP group (P=0.02).Conclusion:The combination of sorafenib with cisplatin might be a promising treatment option for high risk or recurrent HB. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
25. Effect of duplex drugs linking 2'-deoxy-5-fluorouridine (5-FdU) with 3'-C-ethynylcytidine (ECyd) on hepatoblastoma cell lines.
- Author
-
Eicher C, Dewerth A, Ellerkamp V, Fuchs J, Schott S, Armeanu-Ebinger S, Eicher, Carmen, Dewerth, Alexander, Ellerkamp, Verena, Fuchs, Joerg, Schott, Sarah, and Armeanu-Ebinger, Sorin
- Abstract
Purpose: Duplex drugs are promising anticancer agents. After in vivo cleavage into active nucleoside analogues, they exert their anti-tumour activity with reduced toxicity and side effects. Here we evaluated the impact of two duplex drugs on the viability of hepatoblastoma (HB) cells lines and their toxicity against human fibroblasts.Methods: The duplex drugs 2'-deoxy-5-fluorouridylyl-(3'-5')- 3'-C-ethynylcytidine (5-FdU(3'-5')ECyd) and 3'-C-ethynylcytidinylyl-(5'→1-O)-2-O-octadecyl-sn-glycerylyl-(3'-Ο→5')-2'-deoxy-5-fluorouridine (ECyd-lipid-5-FdU) were analysed in two HB cell lines (HUH6, HepT1) and fibroblasts by MTT assay. The treatment potential was compared to the single substances 2'-deoxy-5-fluorourindine (5-FdU), 3'-C-ethynylycytidine (ECyd) and an equimolar mixture of both. Cell cycle analyses were performed using flow cytometry after 7-AAD staining.Results: Both duplex drugs achieve a potent cytotoxic effect at low μM concentrations, which was more pronounced than the mixture of ECyd + 5-FdU. Further, both substances exert toxicity on fibroblasts of tumour samples, with less toxicity in foreskin fibroblasts cultures. Cell cycle analyses revealed a shift towards apoptotic cells for both drugs in HB cells.Conclusion: 5-FdU(3'-5')ECyd and ECyd-lipid-5-FdU exert a highly potent anti-tumoural effect on HB cells and might therefore be a treatment option in HB. Pharmacological formulations of both duplex drugs have to be evaluated in vivo to reduce possible side effects. [ABSTRACT FROM AUTHOR]- Published
- 2013
- Full Text
- View/download PDF
26. The BH3 mimetic ABT-737 increases treatment efficiency of paclitaxel against hepatoblastoma
- Author
-
Fuchs Jörg, Warmann Steven W, Kirchner Bettina, Wenz Julia, Eicher Carmen, Lieber Justus, and Armeanu-Ebinger Sorin
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background The primary goal of current chemotherapy in hepatoblastoma (HB) is reduction of tumour volume and vitality to enable complete surgical resection and reduce risk of recurrence or metastatic disease. Drug resistance remains a major challenge for HB treatment. In some malignancies inhibition of anti-apoptotic pathways using small BH3 mimetic molecules like ABT-737 shows synergistic effects in combination with cystotoxic agents in vitro. Now we analysed toxicology and synergistic effects of this approach in HB cells and HB xenografts. Methods Viability was monitored in HB cells (HUH6 and HepT1) and fibroblasts treated with paclitaxel, ABT-737 and a combination of both in a MTT assay. HUH6 xenotransplants in NOD/LtSz-scid IL2Rγnull mice (NSG) were treated accordingly. Tumour volume and body weight were monitored. Xenografted tumours were analysed by histology and immunohistochemistry (Ki-67 and TUNEL assay). Results ABT-737 reduced viability in HUH6 and HepT1 cells cultures at concentrations above 1 μM and also enhanced the cytotoxic effect of paclitaxel when used in combination. Thereby paclitaxel could be reduced tenfold to achieve similar reduction of viability of tumour cells. In contrast no toxicity in fibroblasts was observed at the same regiments. Subcutaneous HB (HUH6) treated with paclitaxel (12 mg/kg body weight, n = 7) led to delayed tumour growth in the beginning of the experiment. However, tumour volume was similar to controls (n = 5) at day 25. Combination treatment with paclitaxel and ABT-737 (100 mg/kg, n = 8) revealed significantly 10 fold lower relative tumour volumes compared to control and paclitaxel groups. Paclitaxel dependent toxicity was observed in this mice strain. Conclusions Our results demonstrate enhancement of chemotherapy by using modulators of apoptosis. Further analyses should include improved pharmacological formulations of paclitaxel and BH3 mimetics in order to reduce toxicological effects. Sensitising HB to apoptosis may also render resistant HB susceptible to established chemotherapy regimens.
- Published
- 2011
- Full Text
- View/download PDF
27. Tumour stromal cells derived from paediatric malignancies display MSC-like properties and impair NK cell cytotoxicity
- Author
-
Kluba Torsten, Armeanu-Ebinger Sorin, Mang Philippa, Gieseke Friederike, Vaegler Martin, Johann Pascal-David, Handgretinger Rupert, and Müller Ingo
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Tumour growth and metastatic infiltration are favoured by several components of the tumour microenvironment. Bone marrow-derived multipotent mesenchymal stromal cells (MSC) are known to contribute to the tumour stroma. When isolated from healthy bone marrow, MSC exert potent antiproliferative effects on immune effector cells. Due to phenotypic and morphological similarities of MSC and tumour stromal cells (TStrC), we speculated that immunotherapeutic approaches may be hampered if TStrC may still exhibit immunomodulatory properties of MSC. Methods In order to compare immunomodulatory properties of MSC and tumour stromal cells (TStrC), we established and analyzed TStrC cultures from eleven paediatric tumours and MSC preparations from bone marrow aspirates. Immunophenotyping, proliferation assays and NK cell cytotoxicity assays were employed to address the issue. Results While TStrC differed from MSC in terms of plasticity, they shared surface expression of CD105, CD73 and other markers used for MSC characterization. Furthermore, TStrC displayed a strong antiproliferative effect on peripheral blood mononuclear cells (PBMC) in coculture experiments similar to MSC. NK cell cytotoxicity was significantly impaired after co-culture with TStrC and expression of the activating NK cell receptors NKp44 and NKp46 was reduced. Conclusions Our data show that TStrC and MSC share important phenotypic and functional characteristics. The inhibitory effect of TStrC on PBMC and especially on NK cells may facilitate the immune evasion of paediatric tumours.
- Published
- 2010
- Full Text
- View/download PDF
28. Tumour stromal cells derived from paediatric malignancies display MSC-like properties and impair NK cell cytotoxicity.
- Author
-
Johann PD, Vaegler M, Gieseke F, Mang P, Armeanu-Ebinger S, Kluba T, Handgretinger R, Müller I, Johann, Pascal-David, Vaegler, Martin, Gieseke, Friederike, Mang, Philippa, Armeanu-Ebinger, Sorin, Kluba, Torsten, Handgretinger, Rupert, and Müller, Ingo
- Abstract
Background: Tumour growth and metastatic infiltration are favoured by several components of the tumour microenvironment. Bone marrow-derived multipotent mesenchymal stromal cells (MSC) are known to contribute to the tumour stroma. When isolated from healthy bone marrow, MSC exert potent antiproliferative effects on immune effector cells. Due to phenotypic and morphological similarities of MSC and tumour stromal cells (TStrC), we speculated that immunotherapeutic approaches may be hampered if TStrC may still exhibit immunomodulatory properties of MSC.Methods: In order to compare immunomodulatory properties of MSC and tumour stromal cells (TStrC), we established and analyzed TStrC cultures from eleven paediatric tumours and MSC preparations from bone marrow aspirates. Immunophenotyping, proliferation assays and NK cell cytotoxicity assays were employed to address the issue.Results: While TStrC differed from MSC in terms of plasticity, they shared surface expression of CD105, CD73 and other markers used for MSC characterization. Furthermore, TStrC displayed a strong antiproliferative effect on peripheral blood mononuclear cells (PBMC) in coculture experiments similar to MSC. NK cell cytotoxicity was significantly impaired after co-culture with TStrC and expression of the activating NK cell receptors NKp44 and NKp46 was reduced.Conclusions: Our data show that TStrC and MSC share important phenotypic and functional characteristics. The inhibitory effect of TStrC on PBMC and especially on NK cells may facilitate the immune evasion of paediatric tumours. [ABSTRACT FROM AUTHOR]- Published
- 2010
- Full Text
- View/download PDF
29. Benchmarking whole exome sequencing in the German network for personalized medicine.
- Author
-
Menzel M, Martis-Thiele M, Goldschmid H, Ott A, Romanovsky E, Siemanowski-Hrach J, Seillier L, Brüchle NO, Maurer A, Lehmann KV, Begemann M, Elbracht M, Meyer R, Dintner S, Claus R, Meier-Kolthoff JP, Blanc E, Möbs M, Joosten M, Benary M, Basitta P, Hölscher F, Tischler V, Groß T, Kutz O, Prause R, William D, Horny K, Goering W, Sivalingam S, Borkhardt A, Blank C, Junk SV, Yasin L, Moskalev EA, Carta MG, Ferrazzi F, Tögel L, Wolter S, Adam E, Matysiak U, Rosenthal T, Dönitz J, Lehmann U, Schmidt G, Bartels S, Hofmann W, Hirsch S, Dikow N, Göbel K, Banan R, Hamelmann S, Fink A, Ball M, Neumann O, Rehker J, Kloth M, Murtagh J, Hartmann N, Jurmeister P, Mock A, Kumbrink J, Jung A, Mayr EM, Jacob A, Trautmann M, Kirmse S, Falkenberg K, Ruckert C, Hirsch D, Immel A, Dietmaier W, Haack T, Marienfeld R, Fürstberger A, Niewöhner J, Gerstenmaier U, Eberhardt T, Greif PA, Appenzeller S, Maurus K, Doll J, Jelting Y, Jonigk D, Märkl B, Beule D, Horst D, Wulf AL, Aust D, Werner M, Reuter-Jessen K, Ströbel P, Auber B, Sahm F, Merkelbach-Bruse S, Siebolts U, Roth W, Lassmann S, Klauschen F, Gaisa NT, Weichert W, Evert M, Armeanu-Ebinger S, Ossowski S, Schroeder C, Schaaf CP, Malek N, Schirmacher P, Kazdal D, Pfarr N, Budczies J, and Stenzinger A
- Subjects
- Humans, Germany, Biomarkers, Tumor genetics, Computational Biology methods, Exome Sequencing methods, Precision Medicine methods, Precision Medicine standards, Benchmarking, Neoplasms genetics, DNA Copy Number Variations
- Abstract
Introduction: Whole Exome Sequencing (WES) has emerged as an efficient tool in clinical cancer diagnostics to broaden the scope from panel-based diagnostics to screening of all genes and enabling robust determination of complex biomarkers in a single analysis., Methods: To assess concordance, six formalin-fixed paraffin-embedded (FFPE) tissue specimens and four commercial reference standards were analyzed by WES as matched tumor-normal DNA at 21 NGS centers in Germany, each employing local wet-lab and bioinformatics. Somatic and germline variants, copy-number alterations (CNAs), and complex biomarkers were investigated. Somatic variant calling was performed in 494 diagnostically relevant cancer genes. The raw data were collected and re-analyzed with a central bioinformatic pipeline to separate wet- and dry-lab variability., Results: The mean positive percentage agreement (PPA) of somatic variant calling was 76 % while the positive predictive value (PPV) was 89 % in relation to a consensus list of variants found by at least five centers. Variant filtering was identified as the main cause for divergent variant calls. Adjusting filter criteria and re-analysis increased the PPA to 88 % for all and 97 % for the clinically relevant variants. CNA calls were concordant for 82 % of genomic regions. Homologous recombination deficiency (HRD), tumor mutational burden (TMB), and microsatellite instability (MSI) status were concordant for 94 %, 93 %, and 93 % of calls, respectively. Variability of CNAs and complex biomarkers did not decrease considerably after harmonization of the bioinformatic processing and was hence attributed mainly to wet-lab differences., Conclusion: Continuous optimization of bioinformatic workflows and participating in round robin tests are recommended., Competing Interests: Declaration of Competing Interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: MMT reports speaker and travel Expenses from Twist. JS reports speaker honoraria from DLS, Molecular Health, AstraZeneca and Biocartis, outside the submitted work. UL reports speaker fees from AstraZeneca, GSK, Novartis, Menarini, advisory board from AstraZeneca and Novartis. DH reports speaker honorary AstraZeneca, adboard BMS, WD speaker honoraries BMS & Novartis. SMB reports speaker honoraria, advisory board fees and research grants from AstraZeneca, Daiichi, Menarini, Novartis, Roche, BMS, Pfizer, Bayer, MSD, Merck, Amgen, Molecular Health, Targos, DLS, Janssen, GSK, QuIP, outside the submitted work. SL reports research grant from BMS, advisory board/speaker invitation from AstraZeneca, Eli Lilly, Roche and Takeda outside of this work. NTG reports research support from Janssen-Cilag and Advisory Boards from Janssen-Cilag, AstraZeneca, Daiichi-Sankyo and BMS outside the submitted work. WW reports research grants from Roche, MSD, BMS and AstraZeneca. Advisory board, lectures and speaker bureau fees from Roche, MSD, BMS, AstraZeneca, Pfizer, Merck, Lilly, Boehringer, Novartis, Takeda, Bayer, Janssen, Amgen, Astellas, Illumina, Eisai, Siemens, Agilent, ADC, GSK und Molecular Health. SO received reimbursement for travel expenses and payment for conference presentations from Illumina Inc. and Oxford Nanopore Technologies. CS reports research funding from BMS Stiftung Immunonkologie and institutional grants from Illumina outside the submitted work. CPS reports an investigator-initiated grant from Illumnia outside of the submitted work. PS reports grants from Inctye, BMS, Gilead, Falk, speakers bureau/advisory board from MSD, BMS, AstraZeneca, Incyte, Astellas, Janssen, Eisai, Amgen, Boehringer Ingelheim. DK reports personal fees for speaker honoraria from AstraZeneca, and Pfizer, personal fees for Advisory Board from Bristol-Myers Squibb, outside the submitted work. NP reports speaker fees from Novartis, Bayer, Roche, AstraZeneca, Illumina, BMS, MSD, PGDX/Labcorp, advisory board from Novartis, Lilly, Roche, Janssen, travel expenses from Novartis, AstraZeneca, Illumina, BMS, MSD, PGDX/Labcorp, Research grants from Illumina. JB reports grants from German Cancer Aid and consulting from MSD, outside the submitted work. AS reports participation in Advisory Board/Speaker’s Bureau for Astra Zeneca, AGCT, Bayer, Bristol-Myers Squibb, Eli Lilly, Illumina, Janssen, MSD, Novartis, Pfizer, Roche, Seattle Genetics, Takeda, and Thermo Fisher, grants from Bayer, Bristol-Myers Squibb, and Chugai, outside the submitted work. All other authors report no conflicts of interest., (Copyright © 2024 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
30. Tumour-informed liquid biopsies to monitor advanced melanoma patients under immune checkpoint inhibition.
- Author
-
Schroeder C, Gatidis S, Kelemen O, Schütz L, Bonzheim I, Muyas F, Martus P, Admard J, Armeanu-Ebinger S, Gückel B, Küstner T, Garbe C, Flatz L, Pfannenberg C, Ossowski S, and Forschner A
- Subjects
- Humans, Liquid Biopsy methods, Male, Female, Middle Aged, Aged, Prospective Studies, Adult, Biomarkers, Tumor genetics, Positron Emission Tomography Computed Tomography methods, Aged, 80 and over, Melanoma drug therapy, Melanoma genetics, Melanoma pathology, Immune Checkpoint Inhibitors therapeutic use, Circulating Tumor DNA genetics, Circulating Tumor DNA blood
- Abstract
Immune checkpoint inhibitors (ICI) have significantly improved overall survival in melanoma patients. However, 60% experience severe adverse events and early response markers are lacking. Circulating tumour DNA (ctDNA) is a promising biomarker for treatment-response and recurrence detection. The prospective PET/LIT study included 104 patients with palliative combined or adjuvant ICI. Tumour-informed sequencing panels to monitor 30 patient-specific variants were designed and 321 liquid biopsies of 87 patients sequenced. Mean sequencing depth after deduplication using UMIs was 6000x and the error rate of UMI-corrected reads was 2.47×10
-4 . Variant allele fractions correlated with PET/CT MTV (rho=0.69), S100 (rho=0.72), and LDH (rho=0.54). A decrease of allele fractions between T1 and T2 was associated with improved PFS and OS in the palliative cohort (p = 0.008 and p < 0.001). ctDNA was detected in 76.9% of adjuvant patients with relapse (n = 10/13), while all patients without progression (n = 9) remained ctDNA negative. Tumour-informed liquid biopsies are a reliable tool for monitoring treatment response and early relapse in melanoma patients with ICI., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
31. Case Report: Long-Term Survival of a Patient with Cerebral Metastasized Ovarian Carcinoma Treated with a Personalized Peptide Vaccine and Anti-PD-1 Therapy.
- Author
-
Zelba H, Kyzirakos C, Kayser S, Shao B, Reinhardt A, Pieper N, Rabsteyn A, Döcker D, Armeanu-Ebinger S, Kloor M, Hadaschik D, Schulze M, Battke F, Golf A, and Biskup S
- Abstract
Ovarian cancer is one of the most common cancers among women and the most lethal malignancy of all gynecological cancers. Surgery is promising in the early stages; however, most patients are first diagnosed in the advanced stages, where treatment options are limited. Here, we present a 49-year-old patient who was first diagnosed with stage III ovarian cancer. After the tumor progressed several times under guideline therapies with no more treatment options available at that time, the patient received a fully individualized neoantigen-derived peptide vaccine in the setting of an individual healing attempt. The tumor was analyzed for somatic mutations via whole exome sequencing and potential neoepitopes were vaccinated over a period of 50 months. During vaccination, the patient additionally received anti-PD-1 therapy to prevent further disease progression. Vaccine-induced T-cell responses were detected using intracellular cytokine staining. After eleven days of in vitro expansion, four T-cell activation markers (namely IFN-ɣ, TNF-α, IL-2, and CD154) were measured. The proliferation capacity of neoantigen-specific T-cells was determined using a CFSE proliferation assay. Immune monitoring revealed a very strong CD4+ T-cell response against one of the vaccinated peptides. The vaccine-induced T-cells simultaneously expressed CD154, TNF, IL-2, and IFN-ɣ and showed a strong proliferation capacity upon neoantigen stimulation. Next-generation sequencing, as well as immunohistochemical analysis, revealed a loss of Beta-2 microglobulin (B2M), which is essential for MHC class I presentation. The results presented here implicate that the application of neoantigen-derived peptide vaccines might be considered for those cancer stages, where promising therapeutic options are lacking. Furthermore, we provide more data that endorse the intensive investigation of B2M loss as a tumor escape mechanism in clinical trials using anti-cancer vaccines together with immune-checkpoint inhibitors.
- Published
- 2024
- Full Text
- View/download PDF
32. EGFR expression is associated with relapse in a melanoma cohort receiving adjuvant PD-1-based immunotherapy.
- Author
-
Amaral T, Pop OT, Chatziioannou E, Sinnberg T, Niessner H, Zhao J, Ring SS, Joerger M, Schroeder C, Armeanu-Ebinger S, Cozzio A, Leiter U, Thomas I, Jochum W, Garbe C, Forchhammer S, Levesque M, Mangana J, Hölzel M, Dummer R, Schürch CM, Forschner A, and Flatz L
- Abstract
Competing Interests: Conflicts of interest Dr Amaral reports personal honoraria from BMS, CeCaVa, Novartis, and Pierre Fabre, institutional financial support from iFIT, NeraCare, Novartis, Sanofi, and SkylineDx, and institutional research grants from Novartis, outside the submitted work. Drs Sinnberg and Niessner report institutional funding from Novartis and Pierre Fabre outside the submitted work. Dr Ring listed as inventors of the patent entitled “Arenavirus particles to treat solid tumors” (patent number WO2018/185307 A1) describing the application of artLCMV vectors in the treatment of tumors, outside the submitted work. Dr Joerger reports advisory role (institutional): Novartis, AstraZeneca, Basilea Pharmaceutica, Bayer, BMS, Debiopharm, MSD, Roche, and Sanofi, research funding: Swiss Cancer Research, and travel grants: Roche, Sanofi, and Takeda. Dr Schroeder reports institutional grants from Illumina and research grants from BMS Stiftung Immunonkologie outside the submitted work. Dr Cozzio has served as adviser for and/or received speaking fees from AbbVie, Almirall, Amgen, BMS, Eli Lilly, Galderma, LEO Pharma, Janssen-Cilag, Novartis, Pfizer, Pierre Fabre Pharma, Sanofi, and UCB, outside the submitted work. Dr Leiter reports research support from MSD, consulting fees and honoraria from Sun Pharma, Sanofi (personal and institutional), MSD (personal and institutional), Novartis, Roche, and Almirall Hermal, support for attending meeting from Sun Pharma and participation on a Data Safety Monitoring Board or Advisory Board from Sun Pharma, Sanofi, MSD, Novartis, Roche, and Almirall Hermal, outside the submitted work. Dr Thomas reports honoraria for talks or advisory boards from BMS and Pierre Fabre and also reports research funding (institutional) from BMS, Pfizer, MSD, Amgen, argenx, LEO, Novartis, UCB, 4SC, AstraZeneca, BioNtech, Genentech, Roche, Biotech, CureVac, HUYA, Incyte, Idera, Iovance, InflaRx, CerpassRx, Kartos, Nektar, Philogen, Pierre Fabre, Regeneron, Replimune, and Sanofi. Dr Garbe reports personal fees from Amgen, MSD, and Philogen and grants and personal fees from Novartis, NeraCare, BMS, Roche, and Sanofi, outside the submitted work. Dr Forchhammer received personal fees from Recordati, Kyowa Kirin, and Takeda Pharmaceuticals (speakers’ honoraria) as well as institutional grants from NeraCare, SkylineDx, and BioNTech outside the submitted work. Dr Levesque has received project-specific research funding outside the scope of this work from Novartis, Roche, Molecular Partners, and Oncobit. Dr Mangana has intermittent projects focused consultancy or advisory relationships with Merck/Pfizer, MSD, Amgen, Novartis, Roche, BMS, and Pierre Fabre and has received travel support from Ultrasun, L’Oreal, MSD, BMS, and Pierre Fabre outside of the submitted work. Dr Dummer reports research funding to their institution for clinical studies from MSD and consulting or advisory roles with MSD, Novartis, Roche, BMS, Amgen, Takeda, Pierre Fabre, Sun Pharma, Sanofi, Catalym, Second Genome, Regeneron, Alligator, T3 Pharma, MaxiVAX, Pfizer, and touchIME outside the submitted work. Dr Schürch is on the Scientific Advisory Board of and has received research funding from Enable Medicine, Inc, both outside the current work. Dr Forschner served as consultant to Roche, Novartis, MSD, BMS, and Pierre Fabre, received travel support from Roche, Novartis, BMS, and Pierre Fabre, received speaker fees from Roche, Novartis, BMS, MSD, and CeGaT and also reports institutional research grants from BMS Stiftung Immunonkologie. Dr Flatz reports grants or contracts from Hookipa Pharma, SAKK/Immunophotonics, DFG grant (Deutsche Forschungsgemeinschaft), Philogen, and Mundipharma, consulting fees from Philogen, Sanofi, Novartis, and BMS, participation on Data Safety Board University of Basel, outside the submitted work. The other authors have no conflicts of interest to declare.
- Published
- 2023
- Full Text
- View/download PDF
33. Multicentric pilot study to standardize clinical whole exome sequencing (WES) for cancer patients.
- Author
-
Menzel M, Ossowski S, Kral S, Metzger P, Horak P, Marienfeld R, Boerries M, Wolter S, Ball M, Neumann O, Armeanu-Ebinger S, Schroeder C, Matysiak U, Goldschmid H, Schipperges V, Fürstberger A, Allgäuer M, Eberhardt T, Niewöhner J, Blaumeiser A, Ploeger C, Haack TB, Tay TKY, Kelemen O, Pauli T, Kirchner M, Kluck K, Ott A, Renner M, Admard J, Gschwind A, Lassmann S, Kestler H, Fend F, Illert AL, Werner M, Möller P, Seufferlein TTW, Malek N, Schirmacher P, Fröhling S, Kazdal D, Budczies J, and Stenzinger A
- Abstract
A growing number of druggable targets and national initiatives for precision oncology necessitate broad genomic profiling for many cancer patients. Whole exome sequencing (WES) offers unbiased analysis of the entire coding sequence, segmentation-based detection of copy number alterations (CNAs), and accurate determination of complex biomarkers including tumor mutational burden (TMB), homologous recombination repair deficiency (HRD), and microsatellite instability (MSI). To assess the inter-institution variability of clinical WES, we performed a comparative pilot study between German Centers of Personalized Medicine (ZPMs) from five participating institutions. Tumor and matched normal DNA from 30 patients were analyzed using custom sequencing protocols and bioinformatic pipelines. Calling of somatic variants was highly concordant with a positive percentage agreement (PPA) between 91 and 95% and a positive predictive value (PPV) between 82 and 95% compared with a three-institution consensus and full agreement for 16 of 17 druggable targets. Explanations for deviations included low VAF or coverage, differing annotations, and different filter protocols. CNAs showed overall agreement in 76% for the genomic sequence with high wet-lab variability. Complex biomarkers correlated strongly between institutions (HRD: 0.79-1, TMB: 0.97-0.99) and all institutions agreed on microsatellite instability. This study will contribute to the development of quality control frameworks for comprehensive genomic profiling and sheds light onto parameters that require stringent standardization., (© 2023. Nature Publishing Group UK.)
- Published
- 2023
- Full Text
- View/download PDF
34. UV-radiation and MC1R germline mutations are risk factors for the development of conventional and spitzoid melanomas in children and adolescents.
- Author
-
Liebmann A, Admard J, Armeanu-Ebinger S, Wild H, Abele M, Gschwind A, Seibel-Kelemen O, Seitz C, Bonzheim I, Riess O, Demidov G, Sturm M, Schadeck M, Pogoda M, Bien E, Krawczyk M, Jüttner E, Mentzel T, Cesen M, Pfaff E, Kunc M, Forchhammer S, Forschner A, Leiter-Stöppke U, Eigentler TK, Schneider DT, Schroeder C, Ossowski S, and Brecht IB
- Abstract
Background: Genomic characterisation has led to an improved understanding of adult melanoma. However, the aetiology of melanoma in children is still unclear and identifying the correct diagnosis and therapeutic strategies remains challenging., Methods: Exome sequencing of matched tumour-normal pairs from 26 paediatric patients was performed to study the mutational spectrum of melanomas. The cohort was grouped into different categories: spitzoid melanoma (SM), conventional melanoma (CM), and other melanomas (OT)., Findings: In all patients with CM (n = 10) germline variants associated with melanoma were found in low to moderate melanoma risk genes: in 8 patients MC1R variants, in 2 patients variants in MITF, PTEN and BRCA2. Somatic BRAF mutations were detected in 60% of CMs, homozygous deletions of CDKN2A in 20%, TERTp mutations in 30%. In the SM group (n = 12), 5 patients carried at least one MC1R variant; somatic BRAF mutations were detected in 8.3%, fusions in 25% of the cases. No SM showed a homozygous CDKN2A deletion nor a TERTp mutation. In 81.8% of the CM/SM cases the UV damage signatures SBS7 and/or DBS1 were detected. The patient with melanoma arising in giant congenital nevus (CNM) demonstrated the characteristic NRAS Q61K mutation., Interpretation: UV-radiation and MC1R germline variants are risk factors in the development of conventional and spitzoid paediatric melanomas. Paediatric CMs share genomic similarities with adult CMs while the SMs differ genetically from the CM group. Consistent genetic characterization of all paediatric melanomas will potentially lead to better subtype differentiation, treatment, and prevention in the future., Funding: Found in Acknowledgement., Competing Interests: Declaration of interests Outside of submitted work: IBB: Institutional grant from Biontech. O.R. and C.Sc.: grants: Deutsche Forschungsgemeinschaft (DFG), Deutsche Krebshilfe (DKH), European Union (EU), grants to the institution from Illumina and BMS Stiftung Immunonkologie. U.L.S: grants: MSD; support for attending meeting and/or travel: Sun Pharma, Pierre Fabre; participation on a Data Safety Monitoring Board or Advisory Board: MSD, Novartis, Sun Pharma, Almirall, Roche, Sanofi; ADO board member. T.E.: Consulting fees: Bristol-Myers Squibb, MSD, Novartis, Almirall, Hermal, CureVac, Immunocore, Sanofi. S.O.: grants: DFG, Deutsche Luft und Raumfahrt Gesellschaft (DLR), European Union (EU), Bundesministerium für Bildung und Forschung (BMBF), Deutsche Krebshilfe, Bundesministerium für Arbeit und Soziales (BMSA); presentation: Illumina; support for attending meeting and/or travel: Oxford Nanopore Technologies. S.F.: grants: NeraCare, Skyline Dx, Biontech,; speakers honoraria: Kyowa Kirin, Recordati, Takeda Pharmaceuticals., (Copyright © 2023 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
35. Clinical outcome of biomarker-guided therapies in adult patients with tumors of the nervous system.
- Author
-
Renovanz M, Kurz SC, Rieger J, Walter B, Becker H, Hille H, Bombach P, Rieger D, Grosse L, Häusser L, Skardelly M, Merk DJ, Paulsen F, Hoffmann E, Gani C, Neumann M, Beschorner R, Rieß O, Roggia C, Schroeder C, Ossowski S, Armeanu-Ebinger S, Gschwind A, Biskup S, Schulze M, Fend F, Singer S, Zender L, Lengerke C, Brucker SY, Engler T, Forschner A, Stenzl A, Kohlbacher O, Nahnsen S, Gabernet G, Fillinger S, Bender B, Ernemann U, Öner Ö, Beha J, Malek HS, Möller Y, Ruhm K, Tatagiba M, Schittenhelm J, Bitzer M, Malek N, Zips D, and Tabatabai G
- Abstract
Background: The clinical utility of molecular profiling and targeted therapies for neuro-oncology patients outside of clinical trials is not established. We aimed at investigating feasibility and clinical utility of molecular profiling and targeted therapy in adult patients with advanced tumors in the nervous system within a prospective observational study., Methods: molecular tumor board (MTB)@ZPM (NCT03503149) is a prospective observational precision medicine study for patients with advanced tumors. After inclusion of patients, we performed comprehensive molecular profiling, formulated ranked biomarker-guided therapy recommendations based on consensus by the MTB, and collected prospective clinical outcome data., Results: Here, we present initial data of 661 adult patients with tumors of the nervous system enrolled by December 31, 2021. Of these, 408 patients were presented at the MTB. Molecular-instructed therapy recommendations could be made in 380/408 (93.1%) cases and were prioritized by evidence levels. Therapies were initiated in 86/380 (22.6%) cases until data cutoff. We observed a progression-free survival ratio >1.3 in 31.3% of patients., Conclusions: Our study supports the clinical utility of biomarker-guided therapies for neuro-oncology patients and indicates clinical benefit in a subset of patients. Our data might inform future clinical trials, translational studies, and even clinical care., (© The Author(s) 2023. Published by Oxford University Press, the Society for Neuro-Oncology and the European Association of Neuro-Oncology.)
- Published
- 2023
- Full Text
- View/download PDF
36. TMB and BRAF mutation status are independent predictive factors in high-risk melanoma patients with adjuvant anti-PD-1 therapy.
- Author
-
Eckardt J, Schroeder C, Martus P, Armeanu-Ebinger S, Kelemen O, Gschwind A, Bonzheim I, Eigentler T, Amaral T, Ossowski S, Rieß O, Flatz L, Garbe C, and Forschner A
- Subjects
- Humans, Proto-Oncogene Proteins B-raf genetics, Neoplasm Recurrence, Local drug therapy, Neoplasm Recurrence, Local genetics, Adjuvants, Immunologic, Mutation, Skin Neoplasms pathology, Melanoma drug therapy, Melanoma genetics, Melanoma pathology
- Abstract
Background: High tumor mutational burden (TMB) is associated with a favorable outcome in metastatic melanoma patients treated with immune checkpoint inhibitors. However, data are limited in the adjuvant setting. As BRAF mutated patients have an alternative with targeted adjuvant therapy, it is important to identify predictive factors for relapse and recurrence-free survival (RFS) in patients receiving adjuvant anti-PD-1 antibodies., Methods: We evaluated 165 melanoma patients who started adjuvant anti-PD-1 antibody therapy at our center between March 2018 and September 2019. The initial tumor stage was assessed at the beginning of therapy according to the 8th edition of the AJCC Cancer Staging Manual. Tumor and normal tissue of the high-risk stages IIIC/D/IV were sequenced using a 700 gene NGS panel., Results: The tumor stages at the beginning of adjuvant anti-PD-1 therapy were as follows: N = 80 stage IIIA/B (48%), N = 85 stage IIIC/D/IV (52%). 72/165 patients (44%) suffered a relapse, 44/72 (61%) with only loco regional and 28/72 (39%) with distant metastases. Sequencing results were available from 83 to 85 patients with stage IIIC/D/IV. BRAF mutation status (HR 2.12, 95% CI 1.12-4.08; p = 0.022) and TMB (HR 7.11, 95% CI 2.19-23.11; p = 0.001) were significant and independent predictive factors for relapse-free survival (RFS)., Conclusion: BRAF mutation status and TMB were independent predictive factors for RFS. Patients with BRAF V600E/K mutation and TMB high had the best outcome. A classification based on BRAF mutation status and TMB is proposed to predict RFS in melanoma patients with adjuvant anti-PD-1 therapy., (© 2022. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
37. Germline findings in patients with advanced malignancies screened with paired blood-tumour testing for personalised treatment approaches.
- Author
-
Roggia C, Armeanu-Ebinger S, Gschwind A, Seibel-Kelemen O, Hertler S, Faust U, Liebmann A, Haack TB, Neumann M, Bonzheim I, Forschner A, Kopp HG, Herster F, Hartkopf A, Bitzer M, Malek NP, Brecht IB, Ruhm K, Möller Y, Löwenheim H, Ossowski S, Rieß OH, and Schroeder C
- Subjects
- Humans, Precision Medicine, Germ-Line Mutation, Mutation, Genes, BRCA2, Genetic Predisposition to Disease, Genetic Testing methods, Neoplasms diagnosis, Neoplasms genetics, Neoplasms therapy, Hematologic Neoplasms
- Abstract
Background: Sequencing of tumour tissue with comprehensive gene panels is increasingly used to guide treatment in precision oncology. Analysis of tumour-normal pairs allows in contrast to tumour-only assessment direct discrimination between somatic and germline alterations, which might have important implications not only for the patients but also their families., Methods: We performed tumour normal sequencing with a large gene panel in 1048 patients with advanced cancer to support treatment decision. Sequencing results were correlated with clinical and family data., Results: We identified 156 likely pathogenic or pathogenic (LP/P) germline variants in cancer predisposition genes (CPGs) in 144 cases (13.7%). Of all patients, 8.8% had a LP/P variant in autosomal-dominant cancer predisposition genes (AD-CPGs), most of them being genes with high or moderate penetrance (ATM, BRCA2, CHEK2 and BRCA1). In 48 cases, the P/LP variant matched the expected tumour spectrum. A second variant in tumour tissue was found in 31 patients with AD-CPG variants. Low frequency mutations in either TP53, ATM or DNMT3A in the normal sample indicated clonal haematopoiesis in five cases., Conclusions: Tumour-normal testing for personalised treatment identifies germline LP/P variants in a relevant proportion of patients with cancer. The majority of them would not have been referred to genetic counselling based on family history. Indirect functional readouts of tumour-normal sequencing can provide novel links between CPGs and unexpected cancers. The interpretation of increasingly complex datasets in precision oncology is challenging and concepts of interdisciplinary personalised cancer prevention are needed to support patients and their families., (Copyright © 2022 Elsevier Ltd. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
38. Detection of mobile elements insertions for routine clinical diagnostics in targeted sequencing data.
- Author
-
Demidov G, Park J, Armeanu-Ebinger S, Roggia C, Faust U, Cordts I, Blandfort M, Haack TB, Schroeder C, and Ossowski S
- Subjects
- Computational Biology methods, Genetic Predisposition to Disease, Genetics, Medical methods, Germ-Line Mutation, High-Throughput Nucleotide Sequencing, Humans, Molecular Sequence Annotation, Oncogenes, Rare Diseases diagnosis, Rare Diseases genetics, Sequence Analysis, DNA, Exome Sequencing, DNA Transposable Elements, Diagnostic Tests, Routine, Genetic Testing, Mutagenesis, Insertional
- Abstract
Background: Targeted sequencing approaches such as gene panel or exome sequencing have become standard of care for the diagnosis of rare and common genetic disease. The detection and interpretation of point mutations, small insertions and deletions, and even exon-level copy number variants are well established in clinical genetic testing. Other types of genetic variation such as mobile elements insertions (MEIs) are technically difficult to detect. In addition, their downstream clinical interpretation is more complex compared to point mutations due to a larger genomic footprint that can not only predict a clear loss of protein function but might disturb gene regulation and splicing even when located within the non-coding regions. As a consequence, the contribution of MEIs to disease and tumor development remains largely unexplored in routine diagnostics., Methods: In this study, we investigated the occurrence of MEIs in 7,693 exome datasets from individuals with rare diseases and healthy relatives as well as 788 cancer patients analyzed by panel sequencing., Results: We present several exemplary cases highlighting the diagnostic value of MEIs and propose a strategy for the detection, prioritization, and clinical interpretation of MEIs in routine clinical diagnostics., Conclusion: In this paper, we state that detection and interpretation of MEIs in clinical practice in targeted NGS data can be performed relatively easy despite the fact that MEIs very rarely occur in coding parts of the human genome. Large scale reanalysis of MEIs in existing cohorts may solve otherwise unsolvable cases., (© 2021 The Authors. Molecular Genetics & Genomic Medicine published by Wiley Periodicals LLC.)
- Published
- 2021
- Full Text
- View/download PDF
39. Targeting extracellular and juxtamembrane FGFR2 mutations in chemotherapy-refractory cholangiocarcinoma.
- Author
-
Bitzer M, Spahn S, Babaei S, Horger M, Singer S, Schulze-Osthoff K, Missios P, Gatidis S, Nann D, Mattern S, Scheble V, Nikolaou K, Armeanu-Ebinger S, Schulze M, Schroeder C, Biskup S, Beha J, Claassen M, Ruhm K, Poso A, and Malek NP
- Abstract
Intrahepatic cholangiocarcinoma (iCCA) has emerged as a promising candidate for precision medicine, especially in the case of activating FGFR2 gene fusions. In addition to fusions, a considerable fraction of iCCA patients reveals FGFR2 mutations, which might lead to uncontrolled activation of the FGFR2 pathway but are mostly of unknown functional significance. A current challenge for molecular tumor boards (MTB) is to predict the functional consequences of such FGFR2 alterations to guide potential treatment decisions. We report two iCCA patients with extracellular and juxtamembrane FGFR2 mutations. After in silico investigation of the alterations and identification of activated FGFR2 downstream targets in tumor specimens by immunohistochemistry and transcriptome analysis, the MTB recommended treatment with an FGFR-inhibiting tyrosine kinase inhibitor. Both patients developed a rapidly detectable and prolonged partial response to treatment. These two cases suggest an approach to characterize further detected FGFR2 mutations in iCCA to enable patients´ selection for a successful application of the FGFR -inhibiting drugs., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
40. Case Report: Combined CDK4/6 and MEK Inhibition in Refractory CDKN2A and NRAS Mutant Melanoma.
- Author
-
Forschner A, Sinnberg T, Mroz G, Schroeder C, Reinert CP, Gatidis S, Bitzer M, Eigentler T, Garbe C, Niessner H, Röcken M, Roggia C, Armeanu-Ebinger S, Riess O, Mattern S, Nann D, and Bonzheim I
- Abstract
There are only limited treatment options for metastatic NRAS mutant melanoma patients with resistance to immune checkpoint inhibitors. Besides activation of the mitogen-activated protein (MAP) kinase pathway, they often have additional disturbances in cell cycle regulation. However, unlike BRAF mutant melanoma, no targeted therapy has yet been approved for NRAS mutant melanoma so far. Here we present a NRAS mutant melanoma patient with response to combined binimetinib and ribociclib therapy following characterization of the molecular defects of the tumor by panel sequencing. Next generation sequencing (708 cancer genes) of a soft tissue metastasis revealed a homozygous deletion of CDKN2A in addition to the previously known NRAS mutation, as well as amplification of CCNE1 and CDK6. Immunohistochemical staining of the altered cell cycle genes confirmed loss of p16, reduced expression of p21 and high expression of CDK6 and cyclin D1. As the patient had been progressive on combined immunotherapy, targeted therapy with combined MEK and CDK4/6 inhibition was initiated as recommended by the molecular tumor board. Response to treatment was monitored with PET/CT and liquid biopsy, serum LDH, and S100. In addition, a patient-derived xenograft (PDX) was used to prove the efficacy of the two drugs in combination. Furthermore, senescence-associated beta-galactosidase staining showed that more cells were senescent under the combination treatment of binimetinib and ribociclib. Our case demonstrates how an individualized, molecular-based therapeutic approach could be found based on next-generation sequencing results. Furthermore our report highlights the fruitful and efficient collaboration of dermatooncologists, human geneticists, molecular pathologists, biochemists, radiologists, and nuclear physicians. Further studies are urgently needed to expand the very limited therapeutic landscape of NRAS mutated melanoma., Competing Interests: AF served as consultant to Roche, Novartis, MSD, BMS, Pierre-Fabre; received travel support from Roche, Novartis, BMS, Pierre-Fabre, received speaker fees from Roche, Novartis, BMS, MSD, and CeGaT outside the submitted work. She reports institutional research grants from BMS Stiftung Immunonkologie outside the submitted work. CS reports institutional grants from Novartis and grants from BMS Stiftung Immunonkologie outside the submitted work. MB served in advisory committees for Bayer, BMS, EISAI, IPSEN, and MSD outside the submitted work. TE reports personal fees from Amgen, grants and personal fees from Novartis, personal fees from Philogen, grants and personal fees from Roche, grants and personal fees from Sanofi, personal fees from BMS, personal fees from MSD, outside the submitted work. CG reports personal fees from Amgen, grants and personal fees from NeraCare, grants and personal fees from Novartis, personal fees from Philogen, grants and personal fees from Roche, grants and personal fees from Sanofi, personal fees from BMS, personal fees from MSD, outside the submitted work. IB received speaker fees from Novartis, Bayer and AstraZeneca and honoraria for advisory board participation from BMS and Novartis. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Forschner, Sinnberg, Mroz, Schroeder, Reinert, Gatidis, Bitzer, Eigentler, Garbe, Niessner, Röcken, Roggia, Armeanu-Ebinger, Riess, Mattern, Nann and Bonzheim.)
- Published
- 2021
- Full Text
- View/download PDF
41. Macrophage migration inhibitory factor (MIF) is induced by cytotoxic drugs and is involved in immune escape and migration in childhood rhabdomyosarcoma.
- Author
-
Johler SM, Fuchs J, Seitz G, and Armeanu-Ebinger S
- Subjects
- Antineoplastic Agents administration & dosage, Antineoplastic Agents pharmacology, Humans, Macrophage Migration-Inhibitory Factors administration & dosage, Macrophage Migration-Inhibitory Factors pharmacology, Antineoplastic Agents therapeutic use, Immunotherapy methods, Macrophage Migration-Inhibitory Factors therapeutic use, Rhabdomyosarcoma therapy
- Abstract
Macrophage migration inhibitory factor (MIF) is known to be involved in oncogenic transformation, tumour progression, and immunosuppression and is overexpressed in many solid tumours, including paediatric rhabdomyosarcoma (RMS). We investigated the function of MIF in RMS during treatment with cytotoxic drugs. RMS cell lines were analysed by flow cytometry, immunofluorescence staining, and ELISA. We demonstrated the overexpression of MIF in RMS cells and the enhanced expression and secretion after treatment with cytotoxic agents. Migration assays of RMS cells revealed that inhibitors of MIF (ISO-1, Ant.III 4-IPP, Ant.V, sulforaphane (SF)) and blocking antibodies caused reduced migration, indicating a role for MIF in metastatic invasion. Additionally, we investigated the function of MIF in immune escape. The development of a population containing immunosuppressive myeloid-derived suppressor cells was promoted by incubation in conditioned medium of RMS cells comprising MIF and was reversed by MIF inhibitors but not by antibodies. Although most inhibitors may restore immune activity, Ant.III and 10 µM SF disturbed T cell proliferation in a CFSE assay, whereas T cell proliferation was not reduced by 3 µM SF, ISO-1 or antibodies. However, the inhibition of MIF by blocking antibodies did not increase the killing activity of allogenic PBMCs co-cultured with RMS cells. Our results reveal that MIF may be involved in an immune escape mechanism and demonstrate the involvement of MIF in immunogenic cell death during treatment with cytotoxic drugs. Targeting MIF may contribute to the restoration of immune sensitivity and the control of migration and metastatic invasion.
- Published
- 2016
- Full Text
- View/download PDF
42. Photodynamic Therapy Potentiates the Effects of Curcumin on Pediatric Epithelial Liver Tumor Cells.
- Author
-
Ellerkamp V, Bortel N, Schmid E, Kirchner B, Armeanu-Ebinger S, and Fuchs J
- Subjects
- Antineoplastic Agents pharmacology, Carcinoma, Hepatocellular pathology, Cell Line, Tumor, Cell Survival drug effects, Child, Drug Screening Assays, Antitumor, Drug Synergism, Hep G2 Cells, Humans, Liver Neoplasms pathology, Carcinoma, Hepatocellular drug therapy, Curcumin pharmacology, Liver Neoplasms drug therapy, Photochemotherapy methods
- Abstract
Background/aim: Curcumin (CUM) is a promising agent in complementary oncology. The present study analyzed the photoactive properties of curcumin on pediatric epithelial liver tumor cell lines., Materials and Methods: Hepatoblastoma cell lines (HuH6, HepT1) and hepatocellular carcinoma cell lines (HepG2, HC-AFW1) were treated with curcumin and exposed to blue light (phototherapy, 480 nm, 300 W). Cell viability (MTT tests), cellular oxidative stress (production of reactive oxygen species (ROS)) and cellular uptake/degradation of curcumin were analyzed., Results: Significant loss of viability resulted from 24-48 h incubation with curcumin. With photodynamic therapy (PDT), even short time incubation (1 h) with curcumin resulted in significantly lower half maximal inhibitory concentration (IC50) (p<0.001, two-way ANOVA). Significant ROS production was observed with PDT and curcumin., Conclusion: Phototherapy strongly enhances the anticancer properties of curcumin in pediatric solid liver tumors in vitro., (Copyright© 2016 International Institute of Anticancer Research (Dr. John G. Delinassios), All rights reserved.)
- Published
- 2016
43. Effects of curcumin in pediatric epithelial liver tumors: inhibition of tumor growth and alpha-fetoprotein in vitro and in vivo involving the NFkappaB- and the beta-catenin pathways.
- Author
-
Bortel N, Armeanu-Ebinger S, Schmid E, Kirchner B, Frank J, Kocher A, Schiborr C, Warmann S, Fuchs J, and Ellerkamp V
- Subjects
- Animals, Antineoplastic Agents pharmacology, Apoptosis drug effects, Blotting, Western, Carcinoma, Hepatocellular metabolism, Carcinoma, Hepatocellular pathology, Child, Female, Humans, Immunoenzyme Techniques, In Vitro Techniques, Liver Neoplasms metabolism, Liver Neoplasms pathology, Male, Mice, Mice, Inbred NOD, Mice, SCID, NF-kappa B genetics, Neovascularization, Pathologic prevention & control, RNA, Messenger genetics, Real-Time Polymerase Chain Reaction, Reverse Transcriptase Polymerase Chain Reaction, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, alpha-Fetoproteins genetics, beta Catenin genetics, Carcinoma, Hepatocellular prevention & control, Cell Proliferation drug effects, Curcumin pharmacology, Liver Neoplasms prevention & control, NF-kappa B metabolism, alpha-Fetoproteins metabolism, beta Catenin metabolism
- Abstract
In children with hepatocellular carcinoma (pHCC) the 5-year overall survival rate is poor. Effects of cytostatic therapies such as cisplatin and doxorubicin are limited due to chemoresistance and tumor relapse. In adult HCC, several antitumor properties are described for the use of curcumin. Curcumin is one of the best-investigated phytochemicals in complementary oncology without relevant side effects. Its use is limited by low bioavailability. Little is known about the influence of curcumin on pediatric epithelial hepatic malignancies. We investigated the effects of curcumin in combination with cisplatin on two pediatric epithelial liver tumor cell lines. As mechanisms of action inhibition of NFkappaB, beta-catenin, and decrease of cyclin D were identified. Using a mouse xenograft model we could show a significant decrease of alpha-fetoprotein after combination therapy of oral micellar curcumin and cisplatin. Significant concentrations of curcuminoids were found in blood samples, organ lysates, and tumor tissue after oral micellar curcumin administration. Micellar curcumin in combination with cisplatin can be a promising strategy for treatment of pediatric HCC.
- Published
- 2015
- Full Text
- View/download PDF
44. Successful in vivo tumor visualization using fluorescence laparoscopy in a mouse model of disseminated alveolar rhabdomyosarcoma.
- Author
-
Urla C, Armeanu-Ebinger S, Fuchs J, and Seitz G
- Subjects
- Animals, Anthracenes, Apoptosis, Fluorescence, In Situ Nick-End Labeling, Mice, Mice, Inbred NOD, Perylene analogs & derivatives, Perylene therapeutic use, Protein Kinase C antagonists & inhibitors, Radiation-Sensitizing Agents therapeutic use, Rhabdomyosarcoma, Alveolar drug therapy, Rhabdomyosarcoma, Alveolar surgery, Soft Tissue Neoplasms drug therapy, Soft Tissue Neoplasms surgery, Tumor Cells, Cultured, Laparoscopy methods, Neoplasms, Experimental, Photochemotherapy methods, Rhabdomyosarcoma, Alveolar pathology, Soft Tissue Neoplasms pathology
- Abstract
Background: Surgery for rhabdomyosarcoma is challenging due to a lack of clear delineation between tumor and surrounding tissue. Mutilating surgery can be necessary in difficult tumor localizations. Therefore, novel diagnostic and therapeutic modalities are required. The aim of this study was to evaluate the in vivo tumor detection of RMS using fluorescence laparoscopy and to analyze the efficacy of hypericin-induced photodynamic therapy in a mouse model., Methods: Seventeen NOD/LtSz-scid IL2Rγnull-mice were divided into four groups. In group 1, mCherry-expressing tumor cells and in group 2-4 non-transfected tumor cells were xenotransplanted. Three weeks later, one fluorochrome per group (ICG, ICG-cetuximab, hypericin) was injected. Fluorescence laparoscopy was carried out and tumors were resected using fluorescence guidance. In the hypericin group, photodynamic therapy was performed using blue light and apoptosis was evaluated by TUNEL test., Results: A clear discrimination between healthy and tumor tissue was feasible by fluorescending properties with mCherry expressing tumor cells and after injection of hypericin. No fluorescence was detected in mice injected with ICG and ICG-labeled cetuximab. Hypericin photodynamic therapy induced apoptosis of tumor cells after exposure to blue light., Conclusions: Intraoperative photodynamic diagnosis was feasible using mCherry-transfected tumor cells or hypericin. Additionally, intraoperative photodynamic therapy was possible and effective.
- Published
- 2015
- Full Text
- View/download PDF
45. The role of BH3-mimetic drugs in the treatment of pediatric hepatoblastoma.
- Author
-
Lieber J, Armeanu-Ebinger S, and Fuchs J
- Subjects
- Antibodies, Monoclonal therapeutic use, Apoptosis drug effects, Biphenyl Compounds pharmacology, Child, Drug Resistance, Neoplasm, Hepatoblastoma pathology, Humans, Indoles, Liver Neoplasms pathology, Nitrophenols pharmacology, Piperazines pharmacology, Piperazines therapeutic use, Proto-Oncogene Proteins c-bcl-2 antagonists & inhibitors, Proto-Oncogene Proteins c-bcl-2 metabolism, Pyrroles pharmacology, Sulfonamides pharmacology, TNF-Related Apoptosis-Inducing Ligand immunology, TNF-Related Apoptosis-Inducing Ligand metabolism, Biphenyl Compounds therapeutic use, Hepatoblastoma drug therapy, Liver Neoplasms drug therapy, Nitrophenols therapeutic use, Pyrroles therapeutic use, Sulfonamides therapeutic use
- Abstract
Pediatric hepatoblastoma (HB) is commonly treated by neoadjuvant chemotherapy and surgical tumor resection according to international multicenter trial protocols. Complete tumor resection is essential and survival rates up to 95% have now been achieved in those tumors classified as standard-risk HB. Drug resistance and occurrence of metastases remain the major challenges in the treatment of HB, especially in high-risk tumors. These conditions urgently require the development of alternative therapeutic strategies. One of those alternatives is the modulation of apoptosis in HB cells. HBs regularly overexpress anti-apoptotic proteins of the Bcl-family in comparison to healthy liver tissue. This fact may contribute to the development of chemoresistance of HB cells. Synthetic small inhibitory molecules with BH3-mimetic effects, such as ABT-737 and obatoclax, enhance the susceptibility of tumor cells to different cytotoxic drugs and thereby affect initiator proteins of the apoptosis cascade via the intrinsic pathway. Besides additive effects on HB cell viability when used in combination with cytotoxic drugs, BH3-mimetics also play a role in preventing metastasation by reducing adhesion and inhibiting cell migration abilities. Presumably, including additive BH3-mimetic drugs into existing therapeutic regimens in HB patients might allow dose reduction of established cytotoxic drugs and thereby associated immanent side effects, while maintaining the antitumor activity. Furthermore, reduction of tumor growth and inhibition of tumor cell dissemination may facilitate complete surgical tumor resection, which is mandatory in this tumor type resulting in improved survival rates in high-risk HB. Currently, there are phase I and phase II clinical trials in several cancer entities using this potential target. This paper reviews the available literature regarding the use of BH3-mimetic drugs as single agents or in combination with chemotherapy in various malignancies and focuses on results in HB cells.
- Published
- 2015
- Full Text
- View/download PDF
46. Anti-tumor activity of sorafenib in a model of a pediatric hepatocellular carcinoma.
- Author
-
Nagel C, Armeanu-Ebinger S, Dewerth A, Warmann SW, and Fuchs J
- Subjects
- Adult, Animals, Blotting, Western, Carcinoma, Hepatocellular metabolism, Carcinoma, Hepatocellular pathology, Child, Epithelial-Mesenchymal Transition, Female, Humans, Liver Neoplasms metabolism, Liver Neoplasms pathology, Male, Mice, Mice, Inbred NOD, Mice, SCID, Niacinamide therapeutic use, Sorafenib, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, beta Catenin metabolism, Carcinoma, Hepatocellular drug therapy, Cell Proliferation drug effects, Liver Neoplasms drug therapy, Neovascularization, Pathologic drug therapy, Niacinamide analogs & derivatives, Phenylurea Compounds therapeutic use, Protein Kinase Inhibitors therapeutic use
- Abstract
Background: Treatment outcome of children with pediatric hepatocellular carcinoma (pHCC) is poor. Therefore, we evaluated the tyrosine kinase inhibitor sorafenib in a model of pHCC., Methods: Cell viability after treatment with sorafenib was evaluated in HC-AFW1 cells (pHCC) using MTT assay and compared to an adult HCC (aHCC) and two hepatoblastoma (HB) cell lines. ERK, pERK, E-cadherin, and vimentin expression were investigated using Western Blot. Sorafenib (60 mg/kg) was administered orally to NOD.Cg-Prkdcscid-IL2rgtmWjl/Sz mice bearing subcutaneous HC-AFW1-derived tumors. Tumor progression, viability, and vascularization were monitored by tumor volume, AFP levels, and CD31 immunostaining, respectively. Sensitization to sorafenib was evaluated using the β-catenin inhibitor ICG001., Results: Sorafenib reduced cell viability in HC-AFW1 (IC50: 8 µM), comparable to HB cells, however less pronounced in aHCC cells (IC50: 23 µM). Sorafenib inhibited ERK signaling in both, HC-AFW1 cells and -xenografts. In vivo, sorafenib treatment only led to a moderate tumor growth inhibition, although significant reduction of vascularization and tumor growth kinetics was observed. Long-term treatment with sorafenib decreased E-cadherin, but showed no induction of vimentin expression. Combining sorafenib with a β-catenin inhibitor led to an additional reduction of cell viability., Conclusion: Sorafenib together with inhibitors of the β-catenin pathway might be an effective tool in the treatment of pediatric HCC., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
47. Activation of β-catenin and Yap1 in human hepatoblastoma and induction of hepatocarcinogenesis in mice.
- Author
-
Tao J, Calvisi DF, Ranganathan S, Cigliano A, Zhou L, Singh S, Jiang L, Fan B, Terracciano L, Armeanu-Ebinger S, Ribback S, Dombrowski F, Evert M, Chen X, and Monga SPS
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Animals, Bile Duct Neoplasms metabolism, Bile Duct Neoplasms pathology, Bile Ducts, Intrahepatic metabolism, Bile Ducts, Intrahepatic pathology, Carcinoma, Hepatocellular metabolism, Carcinoma, Hepatocellular pathology, Cell Death, Cell Proliferation, Cell Transformation, Neoplastic genetics, Cell Transformation, Neoplastic pathology, Cholangiocarcinoma metabolism, Cholangiocarcinoma pathology, Europe, Gene Expression Regulation, Neoplastic, Hep G2 Cells, Hepatoblastoma genetics, Hepatoblastoma pathology, Humans, Liver Neoplasms genetics, Liver Neoplasms pathology, Mice, Phosphoproteins genetics, Protein Binding, RNA Interference, Signal Transduction, Time Factors, Transcription Factors, Transcription, Genetic, Transfection, United States, YAP-Signaling Proteins, beta Catenin genetics, Adaptor Proteins, Signal Transducing metabolism, Cell Transformation, Neoplastic metabolism, Hepatoblastoma metabolism, Liver Neoplasms metabolism, Phosphoproteins metabolism, beta Catenin metabolism
- Abstract
Background & Aims: Aberrant activation of β-catenin and Yes-associated protein 1 (Yap1) signaling pathways have been associated with the development of multiple tumor types. Yap functions as a transcriptional coactivator by interacting with TEA domain DNA binding proteins. We investigated the interactions among these pathways during hepatic tumorigenesis., Methods: We used immunohistochemical analysis to determine expression of β-catenin and Yap1 in liver cancer specimens collected from patients in Europe and the United States, consisting of 104 hepatocellular carcinoma, 62 intrahepatic cholangiocarcinoma, and 94 hepatoblastoma samples. We assessed β-catenin and Yap1 signaling and interactions in hepatoblastoma cell lines ((HuH6, HepG2, HepT1, HC-AFW1, HepG2, and HC-AFW1); proteins were knocked down with small interfering RNAs, and effects on proliferation and cell death were measured. Sleeping beauty-mediated hydrodynamic transfection was used to overexpress constitutively active forms of β-catenin (ΔN90/β-catenin) and Yap1 (YapS127A) in livers of mice; tissues were collected, and histological and immunohistochemical analyses were performed., Results: We observed nuclear localization of β-catenin and Yap1 in 79% of hepatoblastoma samples but not in most hepatocellular carcinoma or intrahepatic cholangiocarcinoma samples. Yap1 and β-catenin coprecipitated in hepatoblastoma but not hepatocellular carcinoma cells. Small interfering RNA-mediated knockdown of Yap1 or β-catenin in hepatoblastoma cells reduced proliferation in an additive manner. Knockdown of Yap1 reduced its ability to coactivate transcription with β-catenin; β-catenin inhibitors inactivated Yap1. Overexpression of constitutively active forms of Yap1 and β-catenin in mouse liver led to rapid tumorigenesis, with 100% mortality by 11 weeks. Tumor cells expressed both proteins, and human hepatoblastoma cells expressed common targets of their 2 signaling pathways. Yap1 binding of TEA domain factors was required for tumorigenesis in mice., Conclusions: β-catenin and the transcriptional regulator Yap1 interact physically and are activated in most human hepatoblastoma tissues; overexpression of activated forms of these proteins in livers of mice leads to rapid tumor development. Further analysis of these mice will allow further studies of these pathways in hepatoblastoma pathogenesis and could lead to the identification of new therapeutic targets., (Copyright © 2014 AGA Institute. Published by Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
48. PET/MR imaging and optical imaging of metastatic rhabdomyosarcoma in mice.
- Author
-
Armeanu-Ebinger S, Griessinger CM, Herrmann D, Fuchs J, Kneilling M, Pichler BJ, and Seitz G
- Subjects
- Animals, Mice, Rhabdomyosarcoma pathology, Rhabdomyosarcoma secondary, Dideoxynucleosides, Fluorodeoxyglucose F18, Magnetic Resonance Imaging methods, Positron-Emission Tomography methods, Radiopharmaceuticals, Rhabdomyosarcoma diagnosis
- Abstract
Unlabelled: The combination of PET and MR imaging synergizes molecular and morphologic information, allowing better diagnosis in cancer patients. The diagnosis of tumor recurrence in rhabdomyosarcoma is extremely challenging and could be improved with PET/MR imaging. The aim of this study was to validate PET/MR imaging in a disseminated rhabdomyosarcoma mouse model., Methods: One million alveolar (Rh30) and embryonal (RD) rhabdomyosarcoma cells with stably transfected mCherry and Gaussia luciferase were injected intraperitoneally into NOD/LtSz-scid-IL2Rγnull mice. Nine animals were treated with vincristine (0.75 μg/g/d). Tumor growth was monitored on the basis of serum luciferase activity, optical imaging (OI) of the fluorescent protein mCherry, and sequential PET/MR imaging with 3'-deoxy-3'-(18)F-fluorothymidine ((18)F-FLT) and (18)F-FDG. Immunohistochemical Ki-67 and glucose transporter 1 analysis was used to evaluate tumor cell density and proliferative and metabolic activity., Results: The injection of rhabdomyosarcoma cells led to intraperitoneal tumor growth in 34 of 37 mice (Rh30) and 4 of 9 animals (RD). OI revealed inconsistent results for tumors located near the liver. The detection of tumors in the peritoneal cavity was exclusively possible with sequential PET/MR imaging. PET studies with (18)F-FLT MR imaging were more reliable than (18)F-FDG comparing the tracer uptake and correlation with tumor weight. Treatment with vincristine led to reduced tumor growth, which was efficiently detected with (18)F-FDG PET and MR imaging. Total tumor burden as estimated by PET/MR imaging correlated with the serum luciferase activity., Conclusion: We established a unique model of metastatic rhabdomyosarcoma with a high frequency of tumor occurrence and easy monitoring of the tumor growth based on reporter gene expression. The accurate detection of rhabdomyosarcoma requires high soft-tissue contrast provided by the MR imaging and high tracer uptake for PET, which was achieved with (18)F-FLT as the tracer before and (18)F-FDG after treatment with vincristine. PET/MR imaging allows improved diagnosis of experimental rhabdomyosarcoma and therefore might influence clinical therapeutic decisions in the future., (© 2014 by the Society of Nuclear Medicine and Molecular Imaging, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
49. BH3-mimetic drugs prevent tumour onset in an orthotopic mouse model of hepatoblastoma.
- Author
-
Lieber J, Ellerkamp V, Vogt F, Wenz J, Warmann SW, Fuchs J, and Armeanu-Ebinger S
- Subjects
- Animals, Biomimetic Materials chemistry, Biphenyl Compounds chemistry, Cell Transformation, Neoplastic pathology, Cells, Cultured, Disease Models, Animal, Drug Evaluation, Preclinical, Hepatoblastoma pathology, Humans, Indoles, Liver Neoplasms pathology, Mice, Mice, Inbred C57BL, Mice, Inbred NOD, Mice, Nude, Mice, Transgenic, Nitrophenols chemistry, Peptide Fragments chemistry, Piperazines chemistry, Piperazines pharmacology, Proto-Oncogene Proteins chemistry, Pyrroles chemistry, Sulfonamides chemistry, Biomimetic Materials pharmacology, Biphenyl Compounds pharmacology, Cell Transformation, Neoplastic drug effects, Hepatoblastoma prevention & control, Liver Neoplasms prevention & control, Nitrophenols pharmacology, Peptide Fragments pharmacology, Proto-Oncogene Proteins pharmacology, Pyrroles pharmacology, Sulfonamides pharmacology
- Abstract
Drug resistance and metastasis remain major challenges in the treatment of high-risk hepatoblastoma (HB) and require the development of alternative therapeutic strategies. Modulation of apoptosis in HB cells enhances the sensitivity of these cells towards various drugs and has been discussed to enforce treatment. We investigated the impact of apoptosis sensitisers, BH3-mimetics, on the interaction between the host and HB to reduce tumour growth and dissemination while enhancing immunity. BH3-mimetics, such as obatoclax and ABT-737, enhanced the apoptosis-inducing effect of TRAIL and TNF-α resistant HB cells (HepT1 and HUH6). Tumour cell migration was inhibited by ABT-737 and more markedly by obatoclax. In an orthotopic model of HB, tumour uptake was reduced when the cells were pretreated with low concentrations of obatoclax. Only 1 of 7 mice developed HB in the liver, compared with an incidence of 0.8 in the control group. In summary, our study showed that apoptosis sensitisers had broader effects on HB cells than expected including migration and susceptibility to cytokines in addition to the known effects on drug sensitization. Sensitising HB to apoptosis may also allow resistant HB to be targeted by immune cells and prevent tumour cell dissemination., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
50. Attenuated and protease-profile modified sendai virus vectors as a new tool for virotherapy of solid tumors.
- Author
-
Zimmermann M, Armeanu-Ebinger S, Bossow S, Lampe J, Smirnow I, Schenk A, Lange S, Weiss TS, Neubert W, Lauer UM, and Bitzer M
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, Cell Line, Tumor, Cell Survival, Chlorocebus aethiops, Genetic Vectors, Humans, Mice, Inbred BALB C, Mice, Nude, Neoplasm Proteins metabolism, Neoplasm Transplantation, Neoplasms enzymology, Oncolytic Viruses genetics, Oncolytic Viruses physiology, Peptide Hydrolases metabolism, Protein Engineering, Proteolysis, Sendai virus physiology, Vero Cells, Viral Load, Viral Proteins metabolism, Virus Replication, Neoplasms therapy, Oncolytic Virotherapy, Sendai virus genetics
- Abstract
Multiple types of oncolytic viruses are currently under investigation in clinical trials. To optimize therapeutic outcomes it is believed that the plethora of different tumor types will require a diversity of different virus types. Sendai virus (SeV), a murine parainfluenza virus, displays a broad host range, enters cells within minutes and already has been applied safely as a gene transfer vector in gene therapy patients. However, SeV spreading naturally is abrogated in human cells due to a lack of virus activating proteases. To enable oncolytic applications of SeV we here engineered a set of novel recombinant vectors by a two-step approach: (i) introduction of an ubiquitously recognized cleavage-motive into SeV fusion protein now enabling continuous spreading in human tissues, and (ii) profound attenuation of these rSeV by the knockout of viral immune modulating accessory proteins. When employing human hepatoma cell lines, newly generated SeV variants now reached high titers and induced a profound tumor cell lysis. In contrast, virus release from untransformed human fibroblasts or primary human hepatocytes was found to be reduced by about three log steps in a time course experiment which enables the cumulation of kinetic differences of the distinct phases of viral replication such as primary target cell infection, target cell replication, and progeny virus particle release. In a hepatoma xenograft animal model we found a tumor-specific spreading of our novel recombinant SeV vectors without evidence of biodistribution into non-malignant tissues. In conclusion, we successfully developed novel tumor-selective oncolytic rSeV vectors, constituting a new tool for virotherapy of solid tumors being ready for further preclinical and clinical development to address distinct tumor types.
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.