Skip to main content

Main menu

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Editorial Policies
    • Subscribers
    • Advertisers
    • Editorial Board
    • Special Issues 2025
  • Journal Metrics
  • Other Publications
    • In Vivo
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
    • 2008 Nobel Laureates
  • About Us
    • General Policy
    • Contact
  • Other Publications
    • Anticancer Research
    • In Vivo
    • Cancer Genomics & Proteomics

User menu

  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart

Search

  • Advanced search
Anticancer Research
  • Other Publications
    • Anticancer Research
    • In Vivo
    • Cancer Genomics & Proteomics
  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart
Anticancer Research

Advanced Search

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Editorial Policies
    • Subscribers
    • Advertisers
    • Editorial Board
    • Special Issues 2025
  • Journal Metrics
  • Other Publications
    • In Vivo
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
    • 2008 Nobel Laureates
  • About Us
    • General Policy
    • Contact
  • Visit us on Facebook
  • Follow us on Linkedin
Research ArticleExperimental Studies

Preclinical Evaluation of Combined Targeted Approaches in Malignant Rhabdoid Tumors

NATALIA MORENO and KORNELIUS KERL
Anticancer Research August 2016, 36 (8) 3883-3887;
NATALIA MORENO
University Children's Hospital, Department of Hematology and Oncology, Münster, Germany
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
KORNELIUS KERL
University Children's Hospital, Department of Hematology and Oncology, Münster, Germany
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: kornelius.kerl@ukmuenster.de
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Background/Aim: Rhabdoid tumors (RT) are aggressive pediatric tumors, which show poor prognosis despite use of multimodal intensive therapy. In these tumors, several different oncogenic pathways and epigenetic regulators (like CDK4/6-cyclinD-Rb-signaling, EZH2, histone deacetylases) are contemporaneously deregulated as a consequence of biallelic SMARCB1/SNF5/INI1 alterations. Since these tumors are highly resistant to current therapies, alternative treatment strategies are urgently required. Materials and Methods: In this study, we evaluated cytotoxic effects (by MTT tests) of small molecular compounds, which specifically target these deregulated pathways, using either single-drug or combined approaches. Half-maximal inhibitory concentration (IC50) and combined index (CI) were calculated. Results: All target-directed inhibitors blocked cell growth of three different rhabdoid tumor cell lines in vitro. Several combinations of those target-specific drugs synergistically inhibited cell proliferation of rhabdoid tumors. Conclusion: Supporting earlier reports, combined target-directed approaches are a promising tool for the therapy of malignant rhabdoid tumors.

  • Rhabdoid tumors
  • cyclin-D
  • HDAC
  • EZH2
  • aurora kinase
  • bortezomib

Rhabdoid tumors (RT) are aggressive cancers affecting predominantly infants and young children. These neoplasms occur in different anatomical localizations, mainly in the kidneys (RTK, rhabdoid tumors of the kidney), in the brain (AT/RT, atypical teratoid, rhabdoid tumors) and in the soft tissues (MRT, malignant rhabdoid tumors) (1, 2).

The prognosis of patients has improved significantly in the last years as a result of implementing treatment protocols specifically designed for these tumor entities (3). Nevertheless, overall survival (OS) of patients, especially in relapses of rhabdoid tumors, is still poor, despite using incisive multimodal chemotherapeutic, radiotherapeutic and surgical interventions. Further intensification of therapy does not seem to improve prognosis but increases the rate of toxic side-effects (4) making the development of alternative therapeutic approaches necessary.

The majority of RTs exhibit homozygous deletions or mutations of the SMARCB1 (also known as hSNF, INI1, BAF47) tumor suppressor gene, localized on chromosome 22q11.2 (5). This gene encodes one of the core subunits of the human ATP-dependent chromatin remodeling complex SWI/SNF, which is involved in modulation of accessibility of chromatin to transcription factors and, thus, in regulation of gene transcription, as well as in a wide variety of other cellular processes (6).

Biallelic loss of SMARCB1 leads to deregulation of cell signaling pathways implicated in oncogenesis like CDK4/CDK6/cyclinD1 (7), aurora kinase A (8) or the Sonic hedgehog pathway (SHH) (9). Deregulated epigenetic mechanisms (e.g. EZH2 (10) and HDACs (11)) have also been described in these tumor entities. In addition, loss of SMARCB1 in RT led to increased phosphorylation of eIF2α, a central cytoplasmic unfolded protein response (UPR) component, suggesting a role for the UPR in these tumors (12).

In a previous study, we could demonstrate that targeting one deregulated pathway in RT might lead to further pronounced up-regulation of a second mechanism of tumorigenesis (13). This observation makes combined therapeutical targeted approaches reasonable. The purpose of our study was to investigate the cytotoxic activity of specific molecular inhibitors affecting known deregulated pathways in RT. Therefore, cytotoxicity on different tumor cell lines derived from primary RT was explored by using single and combined approaches of multiple target-specific small molecular inhibitors. Since RT presents contemporaneously a deregulation of not only one, but multiple molecular pathways, we hypothesized that synergistic effects of these substances may be a valuable tool for their treatment.

Materials and Methods

Cell lines. RT cell lines LM (RT of liver), G401 (RT of the kidney (RTK) and A204 (RT of the liver) were cultured in DMEM high-glucose formulation (Invitrogen, Karlsruhe, Germany), supplemented with 10% fetal bovine serum (South American, Invitrogen, Carlsbad, CA, USA), 2% glutamine (Invitrogen) and 1% of penicillin/streptomycin (Invitrogen). The cells were cultured at 37°C in a humidified atmosphere with 5% CO2. A204 and G401 were obtained from ATCC (Wesel, Germany). LM cells were a gift from Prof. Handgretinger (Tübingen, Germany). The identity of all cell lines was verified using single-tube polymerase chain reaction (ST-PCR). All experiments in this publication were performed using at least three independent replicates.

Inhibitors. These inhibitors were used in this study: suberoylanilindehydroxamic acid (SAHA), histondeacetylase (HDAC) inhibitor (SML-0061; Sigma, Taufkirchen Germany,); DZNep, EZH2 inhibitor (#252790; Merck Millipore, Darmstadt, Germany); bortezomib (BZ), proteasome inhibitor (#5.04314.0001; Merck Millipore), CDK4 inhibitor (#219476; Merck Millipore); MLN 8054, aurora kinase inhibitor (Seleckchem, Huston, TX, USA).

All inhibitors were reconstituted according to manufacturers' recommendations either in ethanol or dimethyl sulfoxide (DMSO). All compounds were stored until further usage as a 10 mM solution.

Cytotoxicity assay. Cell suspensions (5,000 cells/100 μl) were seeded into four 96-well-plates. Cells were allowed to reach exponential growth before 100 μl of cell culture medium containing the drugs at different concentrations were added. Each drug concentration (0, 0.01, 0.1, 1, 10 and 100 μM) was tested in 3 biological replicates. For experiments with combined treatment (Table II), we used compound 1 in increasing concentrations as in single compound experiments (0, 0.01, 0.1, 1, 10 and 100 μM). DZNep was assayed in a range from 0.001 to 10 μM. Compound 2 was used at 1/10 of the concentration of compound 1. After 72 h, cells were incubated for 4 h with 10 μl MTT reagent (5 mg/ml MTT dissolved in PBS). Metabolically active cells cleaved the yellow tetrazolium salt to a purple formazan dye. The resulting crystals were dissolved in 100 μl isopropanol - 0.04 N HCl. The specimen was evaluated spectrophotometrically at 570 nm and a reference of 650 nm using a Multiskan Ascent multiplate reader (Labsystems, Helsinki, Finland).

Analysis of combined drug effects on cytotoxicity. To evaluate drug combination effects we analyzed cytotoxicity assay data using the median effect method by Chou and Talalay (14). The fraction of unaffected cells was defined as the proportion of living cells compared to the control. The combination index (CI) indicates synergism if CI <1, antagonism for CI >1 and an additive effect for CI=1. Values of the CI were determined at the half-maximal inhibitory concentration (IC50) concentration (fraction affected=0.5). The method was implemented in the statistical software R (Version 2.15.1; https://cran.r-project.org/bin/windows/base/old/2.15.1/).

Results

Small molecular compounds, which target deregulated signaling, inhibit proliferation of rhabdoid tumor cells. In RT multiple signaling, pathways, as well as epigenetic modulating mechanisms, have been found to be deregulated (7, 10, 11, 15). The aim of this study was to evaluate the cytotoxic effects of different small molecular compounds that specifically inhibit these altered pathways. The CDK4/6-cyclinD-Rb pathway, targeted by a specific CDK4 inhibitor (CDK4i) and aurora kinase A inhibited by MLN 8054 are both implicated in cell cycle progression and, thus, in cellular proliferation. Epigenetic regulators were targeted by SAHA inhibiting HDACs and by DZNep inhibiting the histone methyltransferase EZH2, which silences genes by trimethylating histone H3K27. BZ was included in this study as a proteasome inhibitor because loss of SMARCB1 led to increased phosphorylation of eIF2α in rhabdoid tumors (12).

View this table:
  • View inline
  • View popup
  • Download powerpoint
Table I.

IC50 values of single drug treatments. Summarizes results of proliferation assays (after 72 h of treatment) of five different small molecular inhibitors (bortezomib, CDK4i, MLN8054, SAHA, DZNep) in three different rhabdoid tumor cells lines (A204, G401, LM).

In proliferation assays, all used small molecular compounds were able to inhibit proliferation of all three different RT cell lines (A204, G401 and LM) in a nanomolar to micromolar range (Figure 1 and Table I). All cell lines tested exhibited a significant dose-dependent reduction of cell numbers after the treatment. G401 and LM were more sensitive to all the inhibitors evaluated, in comparison to the RT cell line A204.

Administration of BZ very strongly decreased cell proliferation of all three cell lines being the most cytotoxic compound of all tested substances with an IC50 ranging from 0.01 to 0.07 μM, depending on the studied cell line.

Treatment with DZNep was able to inhibit very efficiently cell proliferation, especially of LM cells, which presented an IC50=0.26 μM.

Combinations of small molecular inhibitors, which target deregulated signaling pathways in rhabdoid tumor, act synergistically on blocking tumor cell proliferation. In this study, we aimed to evaluate in vitro combined approaches of compounds targeting different deregulated signaling pathways in RTs. We included five different compounds (BZ, CDK4i, DZNep, MLN8054 and SAHA) in ten different combinations and tested them on three different RT cell lines (G401, A204, LM). Five of those drug combination inhibited synergistically tumor growth in all cell lines used in these experiments (BZ+CDK4i; BZ+DZNep; BZ+MLN; CDK4i+MLN; SAHA+BZ) (Figure 2 and Table II). Interestingly, in the other five drug combinations, we observed differences between the three treated cell lines. A204 and LM cells (both liver-derived) showed comparable synergistic/antagonistic drug profiles of the different compound combinations (e.g. antagonistic: DZNep+MLN; SAHA+DZNep). On G401, all tested combinations showed synergistic effects in tumor cell growth inhibition except of SAHA+DZNep and SAHA+MLN, which acted antagonistically.

Figure 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1.

Effect of single-compound treatment on proliferation assays of RT cell lines. Concentration-dependent response to treatment with the indicated inhibitors performed on rhabdoid tumor cell lines A204 (A), G401 (B) and LM (C) after 72 h. Dotted line indicates 50% of living population. Every experiment was performed in triplicate.

Figure 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 2.

Simultaneous treatment with different target molecule inhibitors act synergistically on inhibiting cell growth of RT. Combined index (CI) of three rhabdoid tumor cell lines A204 (A), G401 (B) and LM (C). Proliferation assay (after 72 h of treatment) of stated combination were used. CI below 1.0 indicated synergistic effects; CI equal or above 1.0 indicated non-synergistic (additive or antagonistic) effects.

Discussion

RT are aggressive pediatric malignancies characterized by the biallelic inactivation of the tumor suppressor SMARCB1 (16), one of the core subunits of the SWI/SNF complex. Alterations in the SWI/SNF complex have been found to be implicated in cell differentiation, developmental diseases and cancer (17).

Different mechanisms driving oncogenesis, including cell signaling transduction and cell cycle regulating pathways, like CyclinD1-Rb (7), aurora kinase A (12) and Sonic hedgehog signaling (9), as well as enzymes controlling other cellular events like proteosomal pathways, are known to be deregulated in RT. Different epigenetic mechanisms, including the trimethylation of histone H3K27 and histone deacetylation, are altered in RTs. Consequently, substances targeting molecules implicated on these pathways have to be systematically evaluated as potential therapeutic approaches for these tumors. In this study, we performed a preclinical in vitro screen using multiple small molecular inhibitors for combined approaches to inhibit RT cell proliferation. In this screen, all RT cell lines showed tumor cell growth inhibition to these target directed inhibitors.

View this table:
  • View inline
  • View popup
  • Download powerpoint
Table II.

IC50 and CI values of combinatorial drug treatments. Results summary of proliferation assays (after 72 h of treatment) of ten combinations of five different small molecular inhibitors (bortezomib, CDK4i, MLN8054, SAHA, DZNep) in three different rhabdoid tumor cells lines (A204, G401, LM). The CI values have been determined at the perspective at the respective IC50 value. CI<1 indicates synergism; CI>1 indicates additive or antagonistic effects. R2 denotes the coefficient of determination of the linear regression in the median effect plot.

Nowadays, performing whole-genome sequencing, genome-wide gene expression and methylome analyses allow the examination of deregulated pathways for individual patients and, thus, the application of a personalized molecular targeted therapy, which may be a promising tool in the treatment of rhabdoid tumors.

In other tumor entities, including a subset of medulloblastomas (MB), individual pathways, such as the Sonic hedgehog pathway, which drive tumorigenesis (18), are detected in clinical trials for personalized treatments. On the one hand, SHH-activated MB with PTCH mutations has been shown to be highly responsive to SMOOTHENED (SMO) receptor antagonists (19). On the other hand, mutations in the SMO receptor during treatment, making these tumors resistant to this kind of therapy, have been reported (20-22). Due to these known mechanisms of developing resistances to target directed drugs, a combined approach makes sense in the therapy of RT. The diversity of deregulated signal pathways presented by this tumor entity, including different subgroups (23), is another argument to use combinations of target-specific inhibitors.

In summary, in this study we showed that diverse combinations of target-specific drugs exert strong synergistic effects on tumor cell proliferation inhibition of RT.

Compounds of all tested classes of inhibitors are used in clinical trials (24-27) making a rapid transfer after further preclinical evaluation into clinical trials for the treatment of RT patients feasible.

Acknowledgements

KK is supported by the “Gesellschaft für KinderKrebsForschung” and the “Sonja-Wasowicz” foundation.

  • Received May 23, 2016.
  • Revision received June 15, 2016.
  • Accepted June 17, 2016.
  • Copyright© 2016 International Institute of Anticancer Research (Dr. John G. Delinassios), All rights reserved

References

  1. ↵
    1. Biegel JA
    : Molecular genetics of atypical teratoid/rhabdoid tumor. Neurosurg Focus 20(1): E11, 2006.
    OpenUrlCrossRefPubMed
  2. ↵
    1. Biegel JA,
    2. Kalpana G,
    3. Knudsen ES,
    4. Packer RJ,
    5. Roberts CW,
    6. Thiele CJ,
    7. Weissman B,
    8. Smith M
    : The role of INI1 and the SWI/SNF complex in the development of rhabdoid tumors: meeting summary from the workshop on childhood atypical teratoid/rhabdoid tumors. Cancer Res 62(1): 323-328, 2002.
    OpenUrlAbstract
  3. ↵
    1. Chi SN,
    2. Zimmerman MA,
    3. Yao X,
    4. Cohen KJ,
    5. Burger P,
    6. Biegel JA,
    7. Rorke-Adams LB,
    8. Fisher MJ,
    9. Janss A,
    10. Mazewski C,
    11. Goldman S,
    12. Manley PE,
    13. Bowers DC,
    14. Bendel A,
    15. Rubin J,
    16. Turner CD,
    17. Marcus KJ,
    18. Goumnerova L,
    19. Ullrich NJ,
    20. Kieran MW
    : Intensive multimodality treatment for children with newly diagnosed CNS atypical teratoid rhabdoid tumor. J Clin Oncol 27(3): 385-389, 2009.
    OpenUrlAbstract/FREE Full Text
  4. ↵
    1. Zaky W,
    2. Dhall G,
    3. Ji L,
    4. Haley K,
    5. Allen J,
    6. Atlas M,
    7. Bertolone S,
    8. Cornelius A,
    9. Gardner S,
    10. Patel R,
    11. Pradhan K,
    12. Shen V,
    13. Thompson S,
    14. Torkildson J,
    15. Sposto R,
    16. Finlay JL
    : Intensive induction chemotherapy followed by myeloablative chemotherapy with autologous hematopoietic progenitor cell rescue for young children newly-diagnosed with central nervous system atypical teratoid/rhabdoid tumors: The head start III experience. Pediatr Blood Cancer 61(1): 95-101, 2013.
    OpenUrlPubMed
  5. ↵
    1. Biegel JA,
    2. Tan L,
    3. Zhang F,
    4. Wainwright L,
    5. Russo P,
    6. Rorke LB
    : Alterations of the hSNF5/INI1 gene in central nervous system atypical teratoid/rhabdoid tumors and renal and extrarenal rhabdoid tumors. Clin Cancer Res 8(11): 3461-3467, 2000.
    OpenUrl
  6. ↵
    1. Euskirchen GM,
    2. Auerbach RK,
    3. Davidov E,
    4. Gianoulis TA,
    5. Zhong G,
    6. Rozowsky J,
    7. Bhardwaj N,
    8. Gerstein MB,
    9. Snyder M
    : Diverse roles and interactions of the SWI/SNF chromatin remodeling complex revealed using global approaches. PLoS Genet 7(3): e1002008, 2011.
    OpenUrlCrossRefPubMed
  7. ↵
    1. Alarcon-Vargas D,
    2. Zhang Z,
    3. Agarwal B,
    4. Challagulla K,
    5. Mani S,
    6. Kalpana GV
    : Targeting cyclin D1, a downstream effector of INI1/hSNF5, in rhabdoid tumors. Oncogene 25(5): 722-734, 2006.
    OpenUrlCrossRefPubMed
  8. ↵
    1. Lee S,
    2. Cimica V,
    3. Ramachandra N,
    4. Zagzag D,
    5. Kalpana GV
    : Aurora A is a repressed effector target of the chromatin remodeling protein INI1/hSNF5 required for rhabdoid tumor cell survival. Cancer Res 71(9): 3225-3235, 2011.
    OpenUrlAbstract/FREE Full Text
  9. ↵
    1. Jagani Z,
    2. Mora-Blanco EL,
    3. Sansam CG,
    4. McKenna ES,
    5. Wilson B,
    6. Chen D,
    7. Klekota J,
    8. Tamayo P,
    9. Nguyen PT,
    10. Tolstorukov M,
    11. Park PJ,
    12. Cho YJ,
    13. Hsiao K,
    14. Buonamici S,
    15. Pomeroy SL,
    16. Mesirov JP,
    17. Ruffner H,
    18. Bouwmeester T,
    19. Luchansky SJ,
    20. Murtie J,
    21. Kelleher JF,
    22. Warmuth M,
    23. Sellers WR,
    24. Roberts CW,
    25. Dorsch M
    : Loss of the tumor suppressor Snf5 leads to aberrant activation of the Hedgehog-Gli pathway. Nat Med 16(12): 1429-1433, 2010.
    OpenUrlCrossRefPubMed
  10. ↵
    1. Wilson BG,
    2. Wang X,
    3. Shen X,
    4. McKenna ES,
    5. Lemieux ME,
    6. Cho YJ,
    7. Koellhoffer EC,
    8. Pomeroy SL,
    9. Orkin SH,
    10. Roberts CW
    : Epigenetic antagonism between polycomb and SWI/SNF complexes during oncogenic transformation. Cancer Cell 18(4): 316-328, 2010.
    OpenUrlCrossRefPubMed
  11. ↵
    1. Furchert SE,
    2. Lanvers-Kaminsky C,
    3. Juurgens H,
    4. Jung M,
    5. Loidl A,
    6. Fruhwald MC
    : Inhibitors of histone deacetylases as potential therapeutic tools for high-risk embryonal tumors of the nervous system of childhood. Int J Cancer 120(8): 1787-1794, 2007.
    OpenUrlCrossRefPubMed
  12. ↵
    1. Hertwig F,
    2. Meyer K,
    3. Braun S,
    4. Ek S,
    5. Spang R,
    6. Pfenninger CV,
    7. Artner I,
    8. Prost G,
    9. Chen X,
    10. Biegel JA,
    11. Judkins AR,
    12. Englund E,
    13. Nuber UA
    : Definition of genetic events directing the development of distinct types of brain tumors from postnatal neural stem/progenitor cells. Cancer Res 72(13): 3381-3392, 2012.
    OpenUrlAbstract/FREE Full Text
  13. ↵
    1. Kerl K,
    2. Ries D,
    3. Unland R,
    4. Borchert C,
    5. Moreno N,
    6. Hasselblatt M,
    7. Jürgens H,
    8. Kool M,
    9. Görlich D,
    10. Eveslage M,
    11. Jung M,
    12. Meisterernst M,
    13. Frühwald M
    : The histone deacetylase inhibitor SAHA acts in synergism with fenretinide and doxorubicin to control growth of rhabdoid tumor cells. BMC Cancer 13: 286, 2013.
    OpenUrlCrossRefPubMed
  14. ↵
    1. Chou TC
    : Drug combination studies and their synergy quantification using the Chou-Talalay method. Cancer Res 70(2): 440-446, 2010.
    OpenUrlAbstract/FREE Full Text
  15. ↵
    1. Kerl K,
    2. Holsten T,
    3. Fruhwald MC
    : Rhabdoid Tumors: Clinical Approaches and Molecular Targets for Innovative Therapy. Pediatr Hematol Oncol 30(7): 587-604, 2013.
    OpenUrlCrossRefPubMed
  16. ↵
    1. Versteege I,
    2. Sévenet N,
    3. Lange J,
    4. Rousseau-Merck MF,
    5. Ambros P,
    6. Handgretinger R,
    7. Aurias A,
    8. Delattre O
    : Truncating mutations of hSNF5/INI1 in aggressive paediatric cancer. Nature 394(6689): 203-206, 1998.
    OpenUrlCrossRefPubMed
  17. ↵
    1. Romero OA,
    2. Sanchez-Cespedes M
    : The SWI/SNF genetic blockade: effects in cell differentiation, cancer and developmental diseases. Oncogene 33(21): 2681-2689, 2013.
    OpenUrlPubMed
  18. ↵
    1. Northcott PA,
    2. Korshunov A,
    3. Pfister SM,
    4. Taylor MD
    : The clinical implications of medulloblastoma subgroups. Nat Rev Neurol 8(6): 340-351, 2012.
    OpenUrlCrossRefPubMed
  19. ↵
    1. Rudin CM,
    2. Hann CL,
    3. Laterra J,
    4. Yauch RL,
    5. Callahan CA,
    6. Fu L,
    7. Holcomb T,
    8. Stinson J,
    9. Gould SE,
    10. Coleman B,
    11. LoRusso PM,
    12. Von Hoff DD,
    13. de Sauvage FJ,
    14. Low JA
    : Treatment of medulloblastoma with hedgehog pathway inhibitor GDC-0449. N Engl J Med 361(12): 1173-1178, 2009.
    OpenUrlCrossRefPubMed
  20. ↵
    1. Buonamici S,
    2. Williams J,
    3. Morrissey M,
    4. Wang A,
    5. Guo R,
    6. Vattay A,
    7. Hsiao K,
    8. Yuan J,
    9. Green J,
    10. Ospina B,
    11. Yu Q,
    12. Ostrom L,
    13. Fordjour P,
    14. Anderson DL,
    15. Monahan JE,
    16. Kelleher JF,
    17. Peukert S,
    18. Pan S,
    19. Wu X,
    20. Maira SM,
    21. García-Echeverría C,
    22. Briggs KJ,
    23. Watkins DN,
    24. Yao YM,
    25. Lengauer C,
    26. Warmuth M,
    27. Sellers WR,
    28. Dorsch M
    : Interfering with resistance to smoothened antagonists by inhibition of the PI3K pathway in medulloblastoma. Sci Transl Med 2(51): 51ra70, 2010.
    OpenUrlAbstract/FREE Full Text
    1. Kim J,
    2. Tang JY,
    3. Gong R,
    4. Kim J,
    5. Lee JJ,
    6. Clemons KV,
    7. Chong CR,
    8. Chang KS,
    9. Fereshteh M,
    10. Gardner D,
    11. Reya T,
    12. Liu JO,
    13. Epstein EH,
    14. Stevens DA,
    15. Beachy PA
    : Itraconazole, a commonly used antifungal that inhibits Hedgehog pathway activity and cancer growth. Cancer Cell 17(4): 388-399, 2010.
    OpenUrlCrossRefPubMed
  21. ↵
    1. Yauch RL,
    2. Dijkgraaf GJ,
    3. Alicke B,
    4. Januario T,
    5. Ahn CP,
    6. Holcomb T,
    7. Pujara K,
    8. Stinson J,
    9. Callahan CA,
    10. Tang T,
    11. Bazan JF,
    12. Kan Z,
    13. Seshagiri S,
    14. Hann CL,
    15. Gould SE,
    16. Low JA,
    17. Rudin CM,
    18. de Sauvage FJ
    : Smoothened mutation confers resistance to a Hedgehog pathway inhibitor in medulloblastoma. Science 326(5952): 572-574, 2009.
    OpenUrlAbstract/FREE Full Text
  22. ↵
    1. Johann PD,
    2. Erkek S,
    3. Zapatka M,
    4. Kerl K 4.,
    5. Buchhalter I 5.,
    6. Hovestadt V 3.,
    7. Jones DT 6.,
    8. Sturm D 1.,
    9. Hermann C 5.,
    10. Segura Wang M 7.,
    11. Korshunov A 8.,
    12. Rhyzova M 9.,
    13. Gröbner S 10.,
    14. Brabetz S 10.,
    15. Chavez L 10.,
    16. Bens S 11.,
    17. Gröschel S 12.,
    18. Kratochwil F 6.,
    19. Wittmann A 6.,
    20. Sieber L 6.,
    21. Geörg C 12.,
    22. Wolf S 13.,
    23. Beck K 3.,
    24. Oyen F 14.,
    25. Capper D 8.,
    26. van Sluis P 15.,
    27. Volckmann R 15.,
    28. Koster J 15.,
    29. Versteeg R 15.,
    30. von Deimling A 8.,
    31. Milde T 16.,
    32. Witt O 16.,
    33. Kulozik AE 17.,
    34. Ebinger M 18.,
    35. Shalaby T 19.,
    36. Grotzer M 19.,
    37. Sumerauer D 20.,
    38. Zamecnik J 21.,
    39. Mora J 22.,
    40. Jabado N 23.,
    41. Taylor MD 24.,
    42. Huang A 24.,
    43. Aronica E 25.,
    44. Bertoni A 26.,
    45. Radlwimmer B 26.,
    46. Pietsch T 27.,
    47. Schüller U 28.,
    48. Schneppenheim R 14.,
    49. Northcott PA 6.,
    50. Korbel JO 7.,
    51. Siebert R 11.,
    52. Frühwald MC 29.,
    53. Lichter P 30.,
    54. Eils R 31.,
    55. Gajjar A 32.,
    56. Hasselblatt M 33.,
    57. Pfister SM1,
    58. Kool M
    : Atypical teratoid/rhabdoid tumors are comprised of three epigenetic subgroups with distinct enhancer landscapes. Cancer Cell 29(3): 379-93, 2016.
    OpenUrlCrossRefPubMed
  23. ↵
    1. Dickson MA,
    2. Tap WD,
    3. Keohan ML,
    4. D'Angelo SP,
    5. Gounder MM,
    6. Antonescu CR,
    7. Landa J,
    8. Qin LX,
    9. Rathbone DD,
    10. Condy MM,
    11. Ustoyev Y,
    12. Crago AM,
    13. Singer S,
    14. Schwartz GK
    : Phase II trial of the CDK4 inhibitor PD0332991 in patients with advanced CDK4-amplified well-differentiated or dedifferentiated liposarcoma. J Clin Oncol 31(16): 2024-2028, 2013.
    OpenUrlAbstract/FREE Full Text
    1. Mosse YP,
    2. Lipsitz E,
    3. Fox E,
    4. Teachey DT,
    5. Maris JM,
    6. Weigel B,
    7. Adamson PC,
    8. Ingle MA,
    9. Ahern CH,
    10. Blaney SM
    : Pediatric phase I trial and pharmacokinetic study of MLN8237, an investigational oral selective small-molecule inhibitor of Aurora kinase A: a Children's Oncology Group Phase I Consortium study. Clin Cancer Res 18(21): 6058-6064, 2012.
    OpenUrlAbstract/FREE Full Text
    1. San-Miguel JF,
    2. Richardson PG,
    3. Gunther A,
    4. Sezer O,
    5. Siegel D,
    6. Bladé J,
    7. LeBlanc R,
    8. Sutherland H,
    9. Sopala M,
    10. Mishra KK,
    11. Mu S,
    12. Bourquelot PM,
    13. Victoria Mateos M,
    14. Anderson KC
    : Phase Ib Study of Panobinostat and Bortezomib in Relapsed or Relapsed and Refractory Multiple Myeloma. J Clin Oncol 31(29): 3696-3703, 2013.
    OpenUrlAbstract/FREE Full Text
  24. ↵
    1. Watanabe T,
    2. Kato H,
    3. Kobayashi Y,
    4. Yamasaki S,
    5. Morita-Hoshi Y,
    6. Yokoyama H,
    7. Morishima Y,
    8. Ricker JL,
    9. Otsuki T,
    10. Miyagi-Maesima A,
    11. Matsuno Y,
    12. Tobinai K
    : Potential efficacy of the oral histone deacetylase inhibitor vorinostat in a phase I trial in follicular and mantle cell lymphoma. Cancer Sci 101(1): 196-200, 2010.
    OpenUrlCrossRefPubMed
PreviousNext
Back to top

In this issue

Anticancer Research: 36 (8)
Anticancer Research
Vol. 36, Issue 8
August 2016
  • Table of Contents
  • Table of Contents (PDF)
  • Index by author
  • Back Matter (PDF)
  • Ed Board (PDF)
  • Front Matter (PDF)
Print
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for your interest in spreading the word on Anticancer Research.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
Preclinical Evaluation of Combined Targeted Approaches in Malignant Rhabdoid Tumors
(Your Name) has sent you a message from Anticancer Research
(Your Name) thought you would like to see the Anticancer Research web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
6 + 14 =
Solve this simple math problem and enter the result. E.g. for 1+3, enter 4.
Citation Tools
Preclinical Evaluation of Combined Targeted Approaches in Malignant Rhabdoid Tumors
NATALIA MORENO, KORNELIUS KERL
Anticancer Research Aug 2016, 36 (8) 3883-3887;

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Reprints and Permissions
Share
Preclinical Evaluation of Combined Targeted Approaches in Malignant Rhabdoid Tumors
NATALIA MORENO, KORNELIUS KERL
Anticancer Research Aug 2016, 36 (8) 3883-3887;
Twitter logo Facebook logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Materials and Methods
    • Results
    • Discussion
    • Acknowledgements
    • References
  • Figures & Data
  • Info & Metrics
  • PDF

Related Articles

  • No related articles found.
  • PubMed
  • Google Scholar

Cited By...

  • Renal medullary carcinomas depend upon SMARCB1 loss and are sensitive to proteasome inhibition
  • Google Scholar

More in this TOC Section

  • Cytotoxic and Metalloproteinase-inhibitory Effects of Ellagic Acid Against Oral Squamous Cell Carcinoma
  • Artogomezianone Inhibits EGFR and Promotes Apoptosis in Non-small Cell Lung Cancer Cells
  • Resveratrol Derivatives Inhibit Pro-survival Akt Signaling Pathway in Lung Cancer
Show more Experimental Studies

Similar Articles

Keywords

  • Rhabdoid tumors
  • cyclin-D
  • HDAC
  • EZH2
  • aurora kinase
  • bortezomib
Anticancer Research

© 2025 Anticancer Research

Powered by HighWire