62 results on '"Elspeth M. Beauchamp"'
Search Results
2. OTS167 blocks FLT3 translation and synergizes with FLT3 inhibitors in FLT3 mutant acute myeloid leukemia
- Author
-
Bartholomew J. Eisfelder, Caner Saygin, Joseph Wynne, Margaret W. Colton, Mariafausta Fischietti, Elspeth M. Beauchamp, Jason X. Cheng, Olatoyosi Odenike, Gail Roboz, Houda Alachkar, and Wendy Stock
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Internal tandem duplication (-ITD) mutations of Fms-like tyrosine kinase 3 (FLT3) provide growth and pro-survival signals in the context of established driver mutations in FLT3 mutant acute myeloid leukemia (AML). Maternal embryonic leucine zipper kinase (MELK) is an aberrantly expressed gene identified as a target in AML. The MELK inhibitor OTS167 induces cell death in AML including cells with FLT3 mutations, yet the role of MELK and mechanisms of OTS167 function are not understood. OTS167 alone or in combination with tyrosine kinase inhibitors (TKIs) were used to investigate the effect of OTS167 on FLT3 signaling and expression in human FLT3 mutant AML cell lines and primary cells. We describe a mechanism whereby OTS167 blocks FLT3 expression by blocking FLT3 translation and inhibiting phosphorylation of eukaryotic initiation factor 4E–binding protein 1 (4E-BP1) and eukaryotic translation initiation factor 4B (eIF4B). OTS167 in combination with TKIs results in synergistic induction of FLT3 mutant cell death in FLT3 mutant cell lines and prolonged survival in a FLT3 mutant AML xenograft mouse model. Our findings suggest signaling through MELK is necessary for the translation and expression of FLT3-ITD, and blocking MELK with OTS167 represents a viable therapeutic strategy for patients with FLT3 mutant AML.
- Published
- 2021
- Full Text
- View/download PDF
3. Central Role of ULK1 in Type I Interferon Signaling
- Author
-
Diana Saleiro, Swarna Mehrotra, Barbara Kroczynska, Elspeth M. Beauchamp, Pawel Lisowski, Beata Majchrzak-Kita, Tushar D. Bhagat, Brady L. Stein, Brandon McMahon, Jessica K. Altman, Ewa M. Kosciuczuk, Darren P. Baker, Chunfa Jie, Nadereh Jafari, Craig B. Thompson, Ross L. Levine, Eleanor N. Fish, Amit K. Verma, and Leonidas C. Platanias
- Subjects
Biology (General) ,QH301-705.5 - Abstract
We provide evidence that the Unc-51-like kinase 1 (ULK1) is activated during engagement of the type I interferon (IFN) receptor (IFNR). Our studies demonstrate that the function of ULK1 is required for gene transcription mediated via IFN-stimulated response elements (ISRE) and IFNγ activation site (GAS) elements and controls expression of key IFN-stimulated genes (ISGs). We identify ULK1 as an upstream regulator of p38α mitogen-activated protein kinase (MAPK) and establish that the regulatory effects of ULK1 on ISG expression are mediated possibly by engagement of the p38 MAPK pathway. Importantly, we demonstrate that ULK1 is essential for antiproliferative responses and type I IFN-induced antineoplastic effects against malignant erythroid precursors from patients with myeloproliferative neoplasms. Together, these data reveal a role for ULK1 as a key mediator of type I IFNR-generated signals that control gene transcription and induction of antineoplastic responses.
- Published
- 2015
- Full Text
- View/download PDF
4. Targeting CHAF1B Enhances IFN Activity against Myeloproliferative Neoplasm Cells
- Author
-
Diana Saleiro, Ewa M. Kosciuczuk, Mariafausta Fischietti, Ricardo E. Perez, G. Sohae Yang, Frank Eckerdt, Elspeth M. Beauchamp, Ye Hou, Qixuan Wang, Rona Singer Weinberg, Eleanor N. Fish, Feng Yue, Ronald Hoffman, and Leonidas C. Platanias
- Abstract
Interferons (IFNs) are cytokines with potent antineoplastic and antiviral properties. IFNα has significant clinical activity in the treatment of myeloproliferative neoplasms (MPN), but the precise mechanisms by which it acts are not well understood. Here, we demonstrate that chromatin assembly factor 1 subunit B (CHAF1B), an Unc-51-like kinase 1 (ULK1)-interactive protein in the nuclear compartment of malignant cells, is overexpressed in patients with MPN. Remarkably, targeted silencing of CHAF1B enhances transcription of IFNα-stimulated genes and promotes IFNα-dependent antineoplastic responses in primary MPN progenitor cells. Taken together, our findings indicate that CHAF1B is a promising newly identified therapeutic target in MPN and that CHAF1B inhibition in combination with IFNα therapy might offer a novel strategy for treating patients with MPN. Significance: Our findings raise the potential for clinical development of drugs targeting CHAF1B to enhance IFN antitumor responses in the treatment of patients with MPN and should have important clinical translational implications for the treatment of MPN and possibly in other malignancies.
- Published
- 2023
- Full Text
- View/download PDF
5. FIGURE 4 from Targeting CHAF1B Enhances IFN Activity against Myeloproliferative Neoplasm Cells
- Author
-
Leonidas C. Platanias, Ronald Hoffman, Feng Yue, Eleanor N. Fish, Rona Singer Weinberg, Qixuan Wang, Ye Hou, Elspeth M. Beauchamp, Frank Eckerdt, G. Sohae Yang, Ricardo E. Perez, Mariafausta Fischietti, Ewa M. Kosciuczuk, and Diana Saleiro
- Abstract
Inhibition of CHAF1B increases IFNα-induced anticlonogenic effects in MPN. HEL cells (A) and SET-2 cells (B) were transfected with either control (Ctrl) siRNA or CHAF1B siRNA, and leukemic CFU-L colony formation was assessed in clonogenic assays in the presence or absence of IFNα, as indicated. Data are expressed as percentage of colony formation over control siRNA-transfected untreated cells (control), and the scatter dot plots represent means ± SEM of four independent experiments for each cell line. C, Clonogenic capability of primary malignant erythroid progenitors isolated from PV patients transfected with either control (Ctrl) siRNA or CHAF1B siRNA and either left untreated or treated with IFNα, as indicated. Data are expressed as percentage of colony formation over control siRNA-transfected untreated cells (control) and shown are means ± SEM of six independent experiments using cells from six different patients with PV. Percentages for the same patient are represented by the same symbol and different patients have different symbols. A–C, Statistical analyses were performed using one-way ANOVA followed by Tukey multiple comparisons test. *, P < 0.05; **, P < 0.01; ****, P < 0.0001.
- Published
- 2023
- Full Text
- View/download PDF
6. FIGURE 2 from Targeting CHAF1B Enhances IFN Activity against Myeloproliferative Neoplasm Cells
- Author
-
Leonidas C. Platanias, Ronald Hoffman, Feng Yue, Eleanor N. Fish, Rona Singer Weinberg, Qixuan Wang, Ye Hou, Elspeth M. Beauchamp, Frank Eckerdt, G. Sohae Yang, Ricardo E. Perez, Mariafausta Fischietti, Ewa M. Kosciuczuk, and Diana Saleiro
- Abstract
CHAF1B interacts with ULK1 in the nuclear compartment of JAK2V617F-positive cells and is overexpressed in patients with MPN. A and B, Left, ULK1–protein complexes were co-IP using an anti-ULK1 antibody from cytosolic and nuclear protein fractions isolated from untreated or IFNα-treated (10 or 240 minutes) HEL (A) or SET-2 (B) cells and then resolved by SDS-PAGE. As control, cytosolic and nuclear lysates isolated from cells treated with IFNα for 240 minutes were incubated with normal rabbit IgG (RIgG) antibody. Interaction between ULK1 and CHAF1B was assessed by immunoblotting with anti-ULK1 and anti-CHAF1B antibodies. Note: ǂ, unspecific band. A and B, Right, Equal amounts of cytosolic and nuclear protein lysates isolated from untreated and IFNα-treated HEL (A) and SET-2 (B) cells used for co-IPs were resolved by SDS-PAGE, transferred to PVDF membranes and then immunoblotted with antibodies against ULK1, CHAF1B, α-tubulin (cytosolic marker), and lamin A/C (nuclear marker), as indicated. A and B, Blots are representative of three independent experiments. C, Scatter dot plot of log2CHAF1B mRNA expression in neutrophils from healthy individuals (normal, n = 11) and patients with ET (n = 47), PMF (n = 18), and PV (n = 28). Data were extracted from NCBI GEO: GSE54646 study (21) and analyzed using GraphPad Prim 8. Shown are means ± SEM. Statistical analysis was performed using one-way ANOVA followed by Dunnett multiple comparisons test to assess P values between patients with MPN and healthy individuals. *, P < 0.05; ***, P < 0.001; ****, P < 0.0001. Scatter dot plots of log2CHAF1B mRNA expression in neutrophils from patients with ET (green triangles), PMF (purple diamonds), and PV (red squares) carrying wild-type (WT) or mutant (MUT) JAK2 (D), CALR (E), and TET2 (F) genes (JAK2 WT: ET n = 20, PMF n = 10, PV n = 5; JAK2 MUT: ET n = 26, PMF n = 8, PV n = 23; CALR WT: ET n = 27, PMF n = 11, PV n = 26; CALR MUT: ET n = 15, PMF n = 5, PV n = 1; TET2 WT: ET n = 45, PMF n = 17, PV n = 24; TET2 MUT: ET n = 2, PMF n = 1, PV n = 4). Data were extracted from NCBI GEO: GSE54646 study (21) and analyzed using GraphPad Prim 8. Patients for which no information was available for the mutational status for the JAK2, CALR, or TET2 genes were excluded from the analysis. Shown are means ± SEM. Statistical analyses were performed using two-sample two-tailed t test: *, P < 0.05.
- Published
- 2023
- Full Text
- View/download PDF
7. FIGURE 3 from Targeting CHAF1B Enhances IFN Activity against Myeloproliferative Neoplasm Cells
- Author
-
Leonidas C. Platanias, Ronald Hoffman, Feng Yue, Eleanor N. Fish, Rona Singer Weinberg, Qixuan Wang, Ye Hou, Elspeth M. Beauchamp, Frank Eckerdt, G. Sohae Yang, Ricardo E. Perez, Mariafausta Fischietti, Ewa M. Kosciuczuk, and Diana Saleiro
- Abstract
Gene-targeted inhibition of CHAF1B increases IFNα-inducible mRNA expression of ISGs in JAK2V617F-positive leukemic cells. qRT-PCR analyses of the indicated genes in control (Ctrl) siRNA or CHAF1B siRNA-transfected JAK2V617F-positive HEL (A) and SET-2 (B) cells, either left untreated or treated with IFNα for 6 hours. GAPDH mRNA expression was used for normalization. Data are expressed as fold change over control siRNA-transfected untreated cells (control) and represent means ± SEM of three independent experiments for each cell line. Statistical analyses were performed using one-way ANOVA followed by Tukey multiple comparisons test. *, P < 0.05; **, P < 0.01; ***, P < 0.001. C, ChIP assay was performed in untreated and IFNα-treated (for 6 hours at 5,000 IU/mL) HEL cells at the ISG15 promoter and the IFIT1 promoter for CHAF1B binding using an anti-CHAF1B antibody. IgG antibody was used for each promoter region as negative control. Scatter dot plots show data as percent enrichment relative to input ± SEM for three independent experiments. Statistical analyses were performed using one-way ANOVA followed by Tukey multiple comparisons test. ns, not significant; *, P < 0.05; **, P < 0.01; ***, P < 0.001. D, Immunoblot analysis of ULK1 expression in control siRNA (Ctrlsi) and ULK1 siRNA (ULK1si) transfected HEL cells either left untreated or treated with IFNα (for 6 hours at 5,000 IU/mL), as indicated. GAPDH levels were used as loading control. Blots are representative of three independent experiments used to perform ChIP assays shown in E. E, ChIP assay was performed in untreated (UT) and IFNα-treated control (Ctrl) siRNA- and ULK1 siRNA-transfected HEL cells at the ISG15 promoter and the IFIT1 promoter for CHAF1B binding using an anti-CHAF1B antibody. F, HEL cells were pretreated for 1 hour with either DMSO (Ctrl and IFNα groups) or SBI-0206965 (SBI; 10 μmol/L; SBI and SBI+IFNα groups) followed by 6 hours of treatment with either DMSO (Ctrl), SBI (10 μmol/L), IFNα (5,000 IU/mL) or SBI+IFNα, as indicated. ChIP assay was performed in HEL cells at the ISG15 promoter and the IFIT1 promoter for CHAF1B binding using an anti-CHAF1B antibody. E and F, IgG antibody was used for each promoter region as negative control. Scatter dot plots show data as percent enrichment relative to input ± SEM for three independent experiments. Statistical analyses were performed using two-way ANOVA followed by Tukey multiple comparisons test: *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001. Relevant statistical differences are shown for the binding of CHAF1B to ISG15 and IFIT1 promoter regions between experimental conditions.
- Published
- 2023
- Full Text
- View/download PDF
8. Figure S1 from Targeting CHAF1B Enhances IFN Activity against Myeloproliferative Neoplasm Cells
- Author
-
Leonidas C. Platanias, Ronald Hoffman, Feng Yue, Eleanor N. Fish, Rona Singer Weinberg, Qixuan Wang, Ye Hou, Elspeth M. Beauchamp, Frank Eckerdt, G. Sohae Yang, Ricardo E. Perez, Mariafausta Fischietti, Ewa M. Kosciuczuk, and Diana Saleiro
- Abstract
Biological processes in which putative ULK1-protein complexes are involved in the cytosol.
- Published
- 2023
- Full Text
- View/download PDF
9. Data from Targeting CHAF1B Enhances IFN Activity against Myeloproliferative Neoplasm Cells
- Author
-
Leonidas C. Platanias, Ronald Hoffman, Feng Yue, Eleanor N. Fish, Rona Singer Weinberg, Qixuan Wang, Ye Hou, Elspeth M. Beauchamp, Frank Eckerdt, G. Sohae Yang, Ricardo E. Perez, Mariafausta Fischietti, Ewa M. Kosciuczuk, and Diana Saleiro
- Abstract
Interferons (IFNs) are cytokines with potent antineoplastic and antiviral properties. IFNα has significant clinical activity in the treatment of myeloproliferative neoplasms (MPN), but the precise mechanisms by which it acts are not well understood. Here, we demonstrate that chromatin assembly factor 1 subunit B (CHAF1B), an Unc-51-like kinase 1 (ULK1)-interactive protein in the nuclear compartment of malignant cells, is overexpressed in patients with MPN. Remarkably, targeted silencing of CHAF1B enhances transcription of IFNα-stimulated genes and promotes IFNα-dependent antineoplastic responses in primary MPN progenitor cells. Taken together, our findings indicate that CHAF1B is a promising newly identified therapeutic target in MPN and that CHAF1B inhibition in combination with IFNα therapy might offer a novel strategy for treating patients with MPN.Significance:Our findings raise the potential for clinical development of drugs targeting CHAF1B to enhance IFN antitumor responses in the treatment of patients with MPN and should have important clinical translational implications for the treatment of MPN and possibly in other malignancies.
- Published
- 2023
- Full Text
- View/download PDF
10. Supplementary Table S4 from SLFN11 Negatively Regulates Noncanonical NFκB Signaling to Promote Glioblastoma Progression
- Author
-
Leonidas C. Platanias, C. David James, Feng Yue, Aneta H. Baran, Amy B. Heimberger, Elspeth M. Beauchamp, Lukas D. Streich, Christopher Gonzalez, Sang Ho, Candice Mazewski, Jamie N. Guillen Magaña, Ricardo E. Perez, Frank Eckerdt, and Mariafausta Fischietti
- Abstract
Supplementary Table S4 lists proteins that were found to bind SLFN11 after irradiation.
- Published
- 2023
- Full Text
- View/download PDF
11. Data from SLFN11 Negatively Regulates Noncanonical NFκB Signaling to Promote Glioblastoma Progression
- Author
-
Leonidas C. Platanias, C. David James, Feng Yue, Aneta H. Baran, Amy B. Heimberger, Elspeth M. Beauchamp, Lukas D. Streich, Christopher Gonzalez, Sang Ho, Candice Mazewski, Jamie N. Guillen Magaña, Ricardo E. Perez, Frank Eckerdt, and Mariafausta Fischietti
- Abstract
Glioblastoma (GBM) is an aggressive and incurable brain tumor in nearly all instances, whose disease progression is driven in part by the glioma stem cell (GSC) subpopulation. Here, we explored the effects of Schlafen family member 11 (SLFN11) in the molecular, cellular, and tumor biology of GBM. CRISPR/Cas9-mediated knockout of SLFN11 inhibited GBM cell proliferation and neurosphere growth and was associated with reduced expression of progenitor/stem cell marker genes, such as NES, SOX2, and CD44. Loss of SLFN11 stimulated expression of NFκB target genes, consistent with a negative regulatory role for SLFN11 on the NFκB pathway. Furthermore, our studies identify p21 as a direct transcriptional target of NFκB2 in GBM whose expression was stimulated by loss of SLFN11. Genetic disruption of SLFN11 blocked GBM growth and significantly extended survival in an orthotopic patient-derived xenograft model. Together, our results identify SLFN11 as a novel component of signaling pathways that contribute to GBM and GSC with implications for future diagnostic and therapeutic strategies.Significance:We identify a negative regulatory role for SLFN11 in noncanonical NFκB signaling that results in suppression of the cell-cycle inhibitor p21. We provide evidence that SLFN11 contributes to regulation of stem cell markers in GBM, promoting the malignant phenotype. In addition, SLFN11 targeting triggers p21 expression and antitumor responses. Our studies define a highly novel function for SLFN11 and identify it as a potential therapeutic target for GBM.
- Published
- 2023
- Full Text
- View/download PDF
12. Supplementary Figures S1-S3, Table S1 from SLFN11 Negatively Regulates Noncanonical NFκB Signaling to Promote Glioblastoma Progression
- Author
-
Leonidas C. Platanias, C. David James, Feng Yue, Aneta H. Baran, Amy B. Heimberger, Elspeth M. Beauchamp, Lukas D. Streich, Christopher Gonzalez, Sang Ho, Candice Mazewski, Jamie N. Guillen Magaña, Ricardo E. Perez, Frank Eckerdt, and Mariafausta Fischietti
- Abstract
Fig. S1: Expression of SLFN family members after SLFN11 knockout. Fig. S2: Reduced 3-D invasion after SLFN11 knockout. Fig. S3: Expression of stem/progenitor markers after SLFN11 add-back. Table S1: Key resources table.
- Published
- 2023
- Full Text
- View/download PDF
13. Data from Potent Antineoplastic Effects of Combined PI3Kα–MNK Inhibition in Medulloblastoma
- Author
-
Leonidas C. Platanias, Rintaro Hashizume, Hidayatullah G. Munshi, Stewart Goldman, Craig Horbinski, David Z. Chen, Quanhong Ma, Ewa M. Kosciuczuk, Gavin T. Blyth, Jessica Clymer, Elspeth M. Beauchamp, Jonathan B. Bell, and Frank Eckerdt
- Abstract
Medulloblastoma is a highly malignant pediatric brain tumor associated with poor outcome. Developing treatments that target the cancer stem cell (CSC) population in medulloblastoma are important to prevent tumor relapse and induce long-lasting clinical responses. We utilized medulloblastoma neurospheres that display CSC characteristics and found activation of the PI3K/AKT pathway in sphere-forming cells. Of all class IA PI3Ks, only the PI3Kα isoform was required for sphere formation by medulloblastoma cells. Knockdown of p110α, but not p110β or p110δ, significantly disrupted cancer stem cell frequencies as determined by extreme limiting dilution analysis (ELDA), indicating an essential role for the PI3Kα catalytic isoform in medulloblastoma CSCs. Importantly, pharmacologic inhibition of the MAPK-interacting kinase (MNK) enhanced the antineoplastic effects of targeted PI3Kα inhibition in medulloblastoma. This indicates that MNK signaling promotes survival in medulloblastoma, suggesting dual PI3Kα and MNK inhibition may provide a novel approach to target and eliminate medulloblastoma CSCs. We also observed a significant reduction in tumor formation in subcutaneous and intracranial mouse xenograft models, which further suggests that this combinatorial approach may represent an efficient therapeutic strategy for medulloblastoma.Implications:These findings raise the possibility of a unique therapeutic approach for medulloblastoma, involving MNK targeting to sensitize medulloblastoma CSCs to PI3Kα inhibition.
- Published
- 2023
- Full Text
- View/download PDF
14. Supplementary Table S1 from Potent Antineoplastic Effects of Combined PI3Kα–MNK Inhibition in Medulloblastoma
- Author
-
Leonidas C. Platanias, Rintaro Hashizume, Hidayatullah G. Munshi, Stewart Goldman, Craig Horbinski, David Z. Chen, Quanhong Ma, Ewa M. Kosciuczuk, Gavin T. Blyth, Jessica Clymer, Elspeth M. Beauchamp, Jonathan B. Bell, and Frank Eckerdt
- Abstract
Pairwise tests for differences in stem cell frequencies.
- Published
- 2023
- Full Text
- View/download PDF
15. Supplementary Figures S1 - S2 from Differential Response of Glioma Stem Cells to Arsenic Trioxide Therapy Is Regulated by MNK1 and mRNA Translation
- Author
-
Leonidas C. Platanias, Michael E. Berens, Jeffrey Raizer, Priya Kumthekar, Kristiina Vuori, Andrew P. Mazar, Craig Horbinski, Ichiro Nakano, C. David James, Shi-Yuan Cheng, Stewart Goldman, Jessica Clymer, Kristen Alley, Elspeth M. Beauchamp, Barbara Kroczynska, Seungchan Kim, Sen Peng, Darren Finlay, Harshil D. Dhruv, Frank Eckerdt, and Jonathan B. Bell
- Abstract
These figures show the PN and MES specific drugs identified by the follow-up cell viability screen. Also shown are the enrichment of translation gene sets in MES GSCs. Relates to Figure 1.
- Published
- 2023
- Full Text
- View/download PDF
16. Supplementary Table S1 from Differential Response of Glioma Stem Cells to Arsenic Trioxide Therapy Is Regulated by MNK1 and mRNA Translation
- Author
-
Leonidas C. Platanias, Michael E. Berens, Jeffrey Raizer, Priya Kumthekar, Kristiina Vuori, Andrew P. Mazar, Craig Horbinski, Ichiro Nakano, C. David James, Shi-Yuan Cheng, Stewart Goldman, Jessica Clymer, Kristen Alley, Elspeth M. Beauchamp, Barbara Kroczynska, Seungchan Kim, Sen Peng, Darren Finlay, Harshil D. Dhruv, Frank Eckerdt, and Jonathan B. Bell
- Abstract
This table shows the initial cell viability screen in GBM cells. Relates to Figure 1C.
- Published
- 2023
- Full Text
- View/download PDF
17. Supplementary Figure Legends and Supplementary Figures 1 and 2 from Direct Binding of Arsenic Trioxide to AMPK and Generation of Inhibitory Effects on Acute Myeloid Leukemia Precursors
- Author
-
Leonidas C. Platanias, Jessica K. Altman, Thomas V. O'Halloran, Benoit Viollet, Elden P. Swindell, Dhaval Nanavati, Ruth Serrano, Ewa M. Kosciuczuk, and Elspeth M. Beauchamp
- Abstract
Supplementary Figure Legends. Supplementary Figure 1: Targeted disruption of AMPKα1/2 does not affect arsenicinducible phosphorylation of AKT at Ser473. Supplementary Figure 2: As- Biotin binds to AMPKα as measured by mass spectroscopy
- Published
- 2023
- Full Text
- View/download PDF
18. Data from Differential Response of Glioma Stem Cells to Arsenic Trioxide Therapy Is Regulated by MNK1 and mRNA Translation
- Author
-
Leonidas C. Platanias, Michael E. Berens, Jeffrey Raizer, Priya Kumthekar, Kristiina Vuori, Andrew P. Mazar, Craig Horbinski, Ichiro Nakano, C. David James, Shi-Yuan Cheng, Stewart Goldman, Jessica Clymer, Kristen Alley, Elspeth M. Beauchamp, Barbara Kroczynska, Seungchan Kim, Sen Peng, Darren Finlay, Harshil D. Dhruv, Frank Eckerdt, and Jonathan B. Bell
- Abstract
Mesenchymal (MES) and proneural (PN) are two distinct glioma stem cell (GSC) populations that drive therapeutic resistance in glioblastoma (GBM). We screened a panel of 650 small molecules against patient-derived GBM cells to discover compounds targeting specific GBM subtypes. Arsenic trioxide (ATO), an FDA-approved drug that crosses the blood–brain barrier, was identified as a potent PN-specific compound in the initial screen and follow-up validation studies. Furthermore, MES and PN GSCs exhibited differential sensitivity to ATO. As ATO has been shown to activate the MAPK-interacting kinase 1 (MNK1)-eukaryotic translation initiation factor 4E (eIF4E) pathway and subsequent mRNA translation in a negative regulatory feedback manner, the mechanistic role of ATO resistance in MES GBM was explored. In GBM cells, ATO-activated translation initiation cellular events via the MNK1–eIF4E signaling axis. Furthermore, resistance to ATO in intracranial PDX tumors correlated with high eIF4E phosphorylation. Polysomal fractionation and microarray analysis of GBM cells were performed to identify ATO's effect on mRNA translation and enrichment of anti-apoptotic mRNAs in the ATO-induced translatome was found. Additionally, it was determined that MNK inhibition sensitized MES GSCs to ATO in neurosphere and apoptosis assays. Finally, examination of the effect of ATO on patients from a phase I/II clinical trial of ATO revealed that PN GBM patients responded better to ATO than other subtypes as demonstrated by longer overall and progression-free survival.Implications: These findings raise the possibility of a unique therapeutic approach for GBM, involving MNK1 targeting to sensitize MES GSCs to drugs like arsenic trioxide. Mol Cancer Res; 16(1); 32–46. ©2017 AACR.
- Published
- 2023
- Full Text
- View/download PDF
19. Supplementary Figure S1 from Potent Antineoplastic Effects of Combined PI3Kα–MNK Inhibition in Medulloblastoma
- Author
-
Leonidas C. Platanias, Rintaro Hashizume, Hidayatullah G. Munshi, Stewart Goldman, Craig Horbinski, David Z. Chen, Quanhong Ma, Ewa M. Kosciuczuk, Gavin T. Blyth, Jessica Clymer, Elspeth M. Beauchamp, Jonathan B. Bell, and Frank Eckerdt
- Abstract
Body weight of mice from flank tumor xenograft experiment
- Published
- 2023
- Full Text
- View/download PDF
20. Supplementary Methods from Differential Response of Glioma Stem Cells to Arsenic Trioxide Therapy Is Regulated by MNK1 and mRNA Translation
- Author
-
Leonidas C. Platanias, Michael E. Berens, Jeffrey Raizer, Priya Kumthekar, Kristiina Vuori, Andrew P. Mazar, Craig Horbinski, Ichiro Nakano, C. David James, Shi-Yuan Cheng, Stewart Goldman, Jessica Clymer, Kristen Alley, Elspeth M. Beauchamp, Barbara Kroczynska, Seungchan Kim, Sen Peng, Darren Finlay, Harshil D. Dhruv, Frank Eckerdt, and Jonathan B. Bell
- Abstract
Relates to materials & methods.
- Published
- 2023
- Full Text
- View/download PDF
21. SLFN11 negatively regulates non-canonical NFkB signaling to promote glioblastoma progression
- Author
-
Mariafausta Fischietti, Frank Eckerdt, Ricardo E. Perez, Jamie N. Guillen Magaña, Candice Mazewski, Sang Ho, Christopher Gonzalez, Lukas D. Streich, Elspeth M. Beauchamp, Amy B. Heimberger, Aneta H. Baran, Feng Yue, C. David James, and Leonidas C. Platanias
- Subjects
Article - Abstract
Glioblastoma (GBM) is an aggressive and incurable brain tumor in nearly all instances, whose disease progression is driven in part by the glioma stem cell (GSC) subpopulation. Here, we explored the effects of Schlafen family member 11 (SLFN11) in the molecular, cellular, and tumor biology of GBM. CRISPR/Cas9-mediated knockout of SLFN11 inhibited GBM cell proliferation and neurosphere growth and was associated with reduced expression of progenitor/stem cell marker genes, such as NES, SOX2, and CD44. Loss of SLFN11 stimulated expression of NFκB target genes, consistent with a negative regulatory role for SLFN11 on the NFκB pathway. Furthermore, our studies identify p21 as a direct transcriptional target of NFκB2 in GBM whose expression was stimulated by loss of SLFN11. Genetic disruption of SLFN11 blocked GBM growth and significantly extended survival in an orthotopic patient-derived xenograft model. Together, our results identify SLFN11 as a novel component of signaling pathways that contribute to GBM and GSC with implications for future diagnostic and therapeutic strategies. Significance: We identify a negative regulatory role for SLFN11 in noncanonical NFκB signaling that results in suppression of the cell-cycle inhibitor p21. We provide evidence that SLFN11 contributes to regulation of stem cell markers in GBM, promoting the malignant phenotype. In addition, SLFN11 targeting triggers p21 expression and antitumor responses. Our studies define a highly novel function for SLFN11 and identify it as a potential therapeutic target for GBM.
- Published
- 2022
22. Inhibitory effects of Tomivosertib in acute myeloid leukemia
- Author
-
Milagros Suarez, Leonidas C. Platanias, Shira Dinner, Elizabeth A. Eklund, Alain Mina, Ewa M. Kosciuczuk, Jessica K. Altman, Gavin T. Blyth, Elspeth M. Beauchamp, Blazej Dolniak, and Diana Saleiro
- Subjects
0301 basic medicine ,Venetoclax ,Kinase ,MNK ,EIF4E ,Myeloid leukemia ,acute myeloid leukemia ,Serine ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,Oncology ,chemistry ,Cell culture ,030220 oncology & carcinogenesis ,hemic and lymphatic diseases ,eIF4E ,Cancer research ,Phosphorylation ,Tomivosertib ,PI3K/AKT/mTOR pathway ,Research Paper - Abstract
The MAPK-interacting kinases 1 and 2 (MNK1/2) have generated increasing interest as therapeutic targets for acute myeloid leukemia (AML). We evaluated the therapeutic potential of the highly-selective MNK1/2 inhibitor Tomivosertib on AML cells. Tomivosertib was highly effective at blocking eIF4E phosphorylation on serine 209 in AML cells. Such inhibitory effects correlated with dose-dependent suppression of cellular viability and leukemic progenitor colony formation. Moreover, combination of Tomivosertib and Venetoclax resulted in synergistic anti-leukemic responses in AML cell lines. Mass spectrometry studies identified novel putative MNK1/2 interactors, while in parallel studies we demonstrated that MNK2 - RAPTOR - mTOR complexes are not disrupted by Tomivosertib. Overall, these findings demonstrate that Tomivosertib exhibits potent anti-leukemic properties on AML cells and support the development of clinical translational efforts involving the use of this drug, alone or in combination with other therapies for the treatment of AML.
- Published
- 2021
23. Abstract A39: Role of LARP1 in the leukemogenesis of Acute Myeloid Leukemia
- Author
-
Dominik A Nahotko, Aneta H Baran, Mariafausta Fischietti, Elspeth M Beauchamp, and Leonidas C Platanias
- Subjects
General Medicine - Abstract
Mammalian target of rapamycin (mTOR) is a strong driver of tumorigenesis in multiple types of tumors, however targeting mTOR for cancer therapy has yielded only limited success. This may be explained in part by signaling redundancy with other pathways, such as CDK9 mTOR-Like (CTORC) complexes, recently described by our group. One of the binders common to mTORC1 and CTORC2 is La related protein 1 (LARP1). LARP1 influences ribosome biogenesis by regulating translation of 5'terminal oligopyrimidine tract (5’ TOP) containing mRNAs, which often encode ribosomal proteins and translation factors. Here, we validate LARP1 as common target of mTORC1 and CTORC2 as well as investigate its influence on leukemogenesis. To explore LARP1’s role in leukemogenesis we utilized CRISPR/CAS9 technology to create OCI-AML5 and U937 LARP1 knockout (KO) cells. Loss of LARP1 expression significantly diminished proliferation potential of AML cell lines both in vitro as well as in vivo when implanted into the flank of athymic nude mice as xenografts. Additionally, primary CD34+ leukemia cells were transfected with LARP1 targeting or scrambled siRNA and leukemic progenitor growth was assessed in methocellulose colony formation assays. We observed suppression of leukemic progenitors colony forming ability in LARP1 siRNA transfected cells, compared to the Ctrl siRNA transfected cells. We also determined enhanced sensitivity of Larp1 KO cell lines to standard chemotherapy agents such Azacitidine, Cytarabine and Venetoclax. We employed polysome profiling to compare global translation levels between Larp1 KO and control U937 cells. Our polysomal profiling results show that KO of LARP1 in U937 cells reduces global translation and ribosome biogenesis as expected as we observed a suppression of both monosomal and polysomal peaks. In summary, Larp1 is important for leukemogenesis and influences their response to treatment with currently used chemotherapy agents. These observations together suggest that LARP1 is a promising target for development of novel antileukemia approaches. Citation Format: Dominik A Nahotko, Aneta H Baran, Mariafausta Fischietti, Elspeth M Beauchamp, Leonidas C Platanias. Role of LARP1 in the leukemogenesis of Acute Myeloid Leukemia [abstract]. In: Proceedings of the AACR Special Conference: Acute Myeloid Leukemia and Myelodysplastic Syndrome; 2023 Jan 23-25; Austin, TX. Philadelphia (PA): AACR; Blood Cancer Discov 2023;4(3_Suppl):Abstract nr A39.
- Published
- 2023
- Full Text
- View/download PDF
24. OTS167 blocks FLT3 translation and synergizes with FLT3 inhibitors in FLT3 mutant acute myeloid leukemia
- Author
-
Houda Alachkar, Mariafausta Fischietti, Margaret W Colton, Elspeth M. Beauchamp, Caner Saygin, Bartholomew J. Eisfelder, Gail J. Roboz, Olatoyosi Odenike, Jason X. Cheng, Joseph Wynne, and Wendy Stock
- Subjects
MELK Inhibitor OTS167 ,Mutant ,Biology ,lcsh:RC254-282 ,Article ,Acute myeloid leukaemia ,Maternal embryonic leucine zipper kinase ,Mice ,fluids and secretions ,Eukaryotic initiation factor ,Cell Line, Tumor ,hemic and lymphatic diseases ,Initiation factor ,Animals ,Humans ,EIF4B ,Naphthyridines ,Protein Kinase Inhibitors ,Myeloid leukemia ,Drug Synergism ,hemic and immune systems ,Hematology ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Leukemia, Myeloid, Acute ,Oncology ,fms-Like Tyrosine Kinase 3 ,Protein Biosynthesis ,Mutation ,embryonic structures ,Cancer research ,Tyrosine kinase ,Cell signalling - Abstract
Internal tandem duplication (-ITD) mutations of Fms-like tyrosine kinase 3 (FLT3) provide growth and pro-survival signals in the context of established driver mutations in FLT3 mutant acute myeloid leukemia (AML). Maternal embryonic leucine zipper kinase (MELK) is an aberrantly expressed gene identified as a target in AML. The MELK inhibitor OTS167 induces cell death in AML including cells with FLT3 mutations, yet the role of MELK and mechanisms of OTS167 function are not understood. OTS167 alone or in combination with tyrosine kinase inhibitors (TKIs) were used to investigate the effect of OTS167 on FLT3 signaling and expression in human FLT3 mutant AML cell lines and primary cells. We describe a mechanism whereby OTS167 blocks FLT3 expression by blocking FLT3 translation and inhibiting phosphorylation of eukaryotic initiation factor 4E–binding protein 1 (4E-BP1) and eukaryotic translation initiation factor 4B (eIF4B). OTS167 in combination with TKIs results in synergistic induction of FLT3 mutant cell death in FLT3 mutant cell lines and prolonged survival in a FLT3 mutant AML xenograft mouse model. Our findings suggest signaling through MELK is necessary for the translation and expression of FLT3-ITD, and blocking MELK with OTS167 represents a viable therapeutic strategy for patients with FLT3 mutant AML.
- Published
- 2021
25. Inhibitory effects of SEL201 in acute myeloid leukemia
- Author
-
Mariafausta Fischietti, Aroop K. Kar, Frank Eckerdt, Leonidas C. Platanias, Elspeth M. Beauchamp, Alain Mina, Diana Saleiro, Elizabeth A. Eklund, Rebekah Siliezar, Krzysztof Brzózka, Ewa M. Kosciuczuk, Gavin T. Blyth, Sameem Abedin, and Tomasz Rzymski
- Subjects
0301 basic medicine ,MAPK/ERK pathway ,kinase inhibitor ,Kinase ,Chemistry ,Cell growth ,MNK ,EIF4E ,Myeloid leukemia ,acute myeloid leukemia ,SEL201 ,3. Good health ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,Cell culture ,eIF4E ,hemic and lymphatic diseases ,030220 oncology & carcinogenesis ,Cancer research ,Phosphorylation ,Protein kinase A ,Research Paper - Abstract
MAPK interacting kinase (MNK), a downstream effector of mitogen-activated protein kinase (MAPK) pathways, activates eukaryotic translation initiation factor 4E (eIF4E) and plays a key role in the mRNA translation of mitogenic and antiapoptotic genes in acute myeloid leukemia (AML) cells. We examined the antileukemic properties of a novel MNK inhibitor, SEL201. Our studies provide evidence that SEL201 suppresses eIF4E phosphorylation on Ser209 in AML cell lines and in primary patient-derived AML cells. Such effects lead to growth inhibitory effects and leukemic cell apoptosis, as well as suppression of leukemic progenitor colony formation. Combination of SEL201 with 5’-azacytidine or rapamycin results in synergistic inhibition of AML cell growth. Collectively, these results suggest that SEL201 has significant antileukemic activity and further underscore the relevance of the MNK pathway in leukemogenesis.
- Published
- 2019
- Full Text
- View/download PDF
26. Discovery of novel Mnk inhibitors using mutation‐based induced‐fit virtual high‐throughput screening
- Author
-
Leonidas C. Platanias, Ewa M. Kosciuczuk, Matthew R. Clutter, Gavin T. Blyth, Elspeth M. Beauchamp, Amy Z. Blackburn, Gary E. Schiltz, and Rama K. Mishra
- Subjects
High-throughput screening ,In silico ,Computational biology ,Protein Serine-Threonine Kinases ,01 natural sciences ,Biochemistry ,Article ,Drug Discovery ,Humans ,Protein Kinase Inhibitors ,Pharmacology ,biology ,010405 organic chemistry ,Chemistry ,Kinase ,Organic Chemistry ,EIF4E ,Intracellular Signaling Peptides and Proteins ,Myeloid leukemia ,Neoplasm Proteins ,0104 chemical sciences ,Molecular Docking Simulation ,Leukemia, Myeloid, Acute ,010404 medicinal & biomolecular chemistry ,Docking (molecular) ,Mitogen-activated protein kinase ,biology.protein ,Molecular Medicine ,Phosphorylation - Abstract
Mnk kinases (Mnk1 and 2) are downstream effectors of Map kinase pathways and regulate phosphorylation of eukaryotic initiation factor 4E (eIF4E). Engagement of the Mnk pathway is critical in acute myeloid leukemia (AML) leukemogenesis and Mnk inhibitors have potent antileukemic properties in vitro and in vivo, suggesting that targeting Mnk kinases may provide a novel approach for treating AML. Here, we report the development and application of a mutation-based induced-fit (M-IFD) in silico screen to identify novel Mnk inhibitors. The Mnk1 structure was modeled by temporarily mutating an amino acid that obstructs the ATP-binding site in the Mnk1 crystal structure while carrying out docking simulations of known inhibitors. The hit compounds display activity in Mnk biochemical and cellular assays, including acute myeloid leukemia progenitors. This approach will enable further rational structure-based drug design of new Mnk inhibitors and potentially novel ways of therapeutically targeting this kinase.
- Published
- 2019
- Full Text
- View/download PDF
27. Identification and targeting of novel CDK9 complexes in acute myeloid leukemia
- Author
-
Olga Frankfurt, Gavin T. Blyth, Connor Lantz, Eleanor N. Fish, Leonidas C. Platanias, Ahmet Dirim Arslan, Mariafausta Fischietti, Elspeth M. Beauchamp, Young Ah Goo, Elizabeth A. Eklund, Jessica K. Altman, Paul M. Thomas, Angela Yang, Imo Akpan, Sameem Abedin, Alissa Nelson, and Sara G. Radecki
- Subjects
0301 basic medicine ,Scaffold protein ,Antimetabolites, Antineoplastic ,Proteome ,Carcinogenesis ,Immunology ,Mice, Nude ,Apoptosis ,Mechanistic Target of Rapamycin Complex 2 ,Mechanistic Target of Rapamycin Complex 1 ,Biochemistry ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Biomarkers, Tumor ,Tumor Cells, Cultured ,Protein biosynthesis ,Animals ,Humans ,RNA, Messenger ,Phosphorylation ,MLST8 ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,Chemistry ,TOR Serine-Threonine Kinases ,Cytarabine ,Myeloid leukemia ,Cell Biology ,Hematology ,Cyclin-Dependent Kinase 9 ,Xenograft Model Antitumor Assays ,Leukemia, Myeloid, Acute ,030104 developmental biology ,Protein Biosynthesis ,030220 oncology & carcinogenesis ,Cancer research ,Cyclin-dependent kinase 9 ,Signal transduction ,BLOOD Commentary ,Signal Transduction - Abstract
Aberrant activation of mTOR signaling in acute myeloid leukemia (AML) results in a survival advantage that promotes the malignant phenotype. To improve our understanding of factors that contribute to mammalian target of rapamycin (mTOR) signaling activation and identify novel therapeutic targets, we searched for unique interactors of mTOR complexes through proteomics analyses. We identify cyclin dependent kinase 9 (CDK9) as a novel binding partner of the mTOR complex scaffold protein, mLST8. Our studies demonstrate that CDK9 is present in distinct mTOR-like (CTOR) complexes in the cytoplasm and nucleus. In the nucleus, CDK9 binds to RAPTOR and mLST8, forming CTORC1, to promote transcription of genes important for leukemogenesis. In the cytoplasm, CDK9 binds to RICTOR, SIN1, and mLST8, forming CTORC2, and controls messenger RNA (mRNA) translation through phosphorylation of LARP1 and rpS6. Pharmacological targeting of CTORC complexes results in suppression of growth of primitive human AML progenitors in vitro and elicits strong antileukemic responses in AML xenografts in vivo.
- Published
- 2019
- Full Text
- View/download PDF
28. Pharmacological mTOR targeting enhances the antineoplastic effects of selective PI3Kα inhibition in medulloblastoma
- Author
-
Jonathan B. Bell, Stewart Goldman, Frank Eckerdt, Jessica Clymer, Elspeth M. Beauchamp, Leonidas C. Platanias, and Gavin T. Blyth
- Subjects
lcsh:Medicine ,0302 clinical medicine ,lcsh:Science ,0303 health sciences ,education.field_of_study ,Multidisciplinary ,biology ,Cancer stem cells ,Brain Neoplasms ,Triazines ,Kinase ,TOR Serine-Threonine Kinases ,Imidazoles ,3. Good health ,030220 oncology & carcinogenesis ,Female ,Signal Transduction ,Class I Phosphatidylinositol 3-Kinases ,Population ,Mice, Nude ,Antineoplastic Agents ,Sarcoma, Ewing ,Zinc Finger Protein GLI1 ,Article ,Paediatric cancer ,03 medical and health sciences ,Cancer stem cell ,GLI1 ,Cell Line, Tumor ,medicine ,Animals ,Humans ,education ,Protein Kinase Inhibitors ,PI3K/AKT/mTOR pathway ,030304 developmental biology ,Cell Nucleus ,Medulloblastoma ,business.industry ,lcsh:R ,medicine.disease ,Xenograft Model Antitumor Assays ,CNS cancer ,Thiazoles ,Preclinical research ,Apoptosis ,Cancer cell ,Cancer research ,biology.protein ,lcsh:Q ,business - Abstract
Despite recent advances in the treatment of medulloblastoma, patients in high-risk categories still face very poor outcomes. Evidence indicates that a subpopulation of cancer stem cells contributes to therapy resistance and tumour relapse in these patients. To prevent resistance and relapse, the development of treatment strategies tailored to target subgroup specific signalling circuits in high-risk medulloblastomas might be similarly important as targeting the cancer stem cell population. We have previously demonstrated potent antineoplastic effects for the PI3Kα selective inhibitor alpelisib in medulloblastoma. Here, we performed studies aimed to enhance the anti-medulloblastoma effects of alpelisib by simultaneous catalytic targeting of the mTOR kinase. Pharmacological mTOR inhibition potently enhanced the suppressive effects of alpelisib on cancer cell proliferation, colony formation and apoptosis and additionally blocked sphere-forming ability of medulloblastoma stem-like cancer cells in vitro. We identified the HH effector GLI1 as a target for dual PI3Kα and mTOR inhibition in SHH-type medulloblastoma and confirmed these results in HH-driven Ewing sarcoma cells. Importantly, pharmacologic mTOR inhibition greatly enhanced the inhibitory effects of alpelisib on medulloblastoma tumour growth in vivo. In summary, these findings highlight a key role for PI3K/mTOR signalling in GLI1 regulation in HH-driven cancers and suggest that combined PI3Kα/mTOR inhibition may be particularly interesting for the development of effective treatment strategies in high-risk medulloblastomas.
- Published
- 2019
- Full Text
- View/download PDF
29. Potent Antineoplastic Effects of Combined PI3Kα-MNK Inhibition in Medulloblastoma
- Author
-
Ewa M. Kosciuczuk, Leonidas C. Platanias, Quanhong Ma, Frank Eckerdt, Gavin T. Blyth, Jessica Clymer, Hidayatullah G. Munshi, Elspeth M. Beauchamp, David Z. Chen, Jonathan B. Bell, Stewart Goldman, Rintaro Hashizume, and Craig Horbinski
- Subjects
0301 basic medicine ,Cancer Research ,Class I Phosphatidylinositol 3-Kinases ,Population ,Mice, Nude ,Antineoplastic Agents ,P110α ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Cancer stem cell ,Neurosphere ,Cell Line, Tumor ,Medicine ,Animals ,Humans ,education ,Cerebellar Neoplasms ,Molecular Biology ,neoplasms ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,Medulloblastoma ,Gene knockdown ,education.field_of_study ,business.industry ,medicine.disease ,Xenograft Model Antitumor Assays ,nervous system diseases ,stomatognathic diseases ,030104 developmental biology ,Oncology ,P110δ ,Copper-Transporting ATPases ,030220 oncology & carcinogenesis ,Cancer research ,Neoplastic Stem Cells ,Female ,business ,Signal Transduction - Abstract
Medulloblastoma is a highly malignant pediatric brain tumor associated with poor outcome. Developing treatments that target the cancer stem cell (CSC) population in medulloblastoma are important to prevent tumor relapse and induce long-lasting clinical responses. We utilized medulloblastoma neurospheres that display CSC characteristics and found activation of the PI3K/AKT pathway in sphere-forming cells. Of all class IA PI3Ks, only the PI3Kα isoform was required for sphere formation by medulloblastoma cells. Knockdown of p110α, but not p110β or p110δ, significantly disrupted cancer stem cell frequencies as determined by extreme limiting dilution analysis (ELDA), indicating an essential role for the PI3Kα catalytic isoform in medulloblastoma CSCs. Importantly, pharmacologic inhibition of the MAPK-interacting kinase (MNK) enhanced the antineoplastic effects of targeted PI3Kα inhibition in medulloblastoma. This indicates that MNK signaling promotes survival in medulloblastoma, suggesting dual PI3Kα and MNK inhibition may provide a novel approach to target and eliminate medulloblastoma CSCs. We also observed a significant reduction in tumor formation in subcutaneous and intracranial mouse xenograft models, which further suggests that this combinatorial approach may represent an efficient therapeutic strategy for medulloblastoma. Implications: These findings raise the possibility of a unique therapeutic approach for medulloblastoma, involving MNK targeting to sensitize medulloblastoma CSCs to PI3Kα inhibition.
- Published
- 2019
30. Pre-clinical evidence of PIM kinase inhibitor activity in BCR-ABL1 unmutated and mutated Philadelphia chromosome-positive (Ph+) leukemias
- Author
-
Nicholas J. Donato, Leonidas C. Platanias, Francis J. Giles, Gavin T. Blyth, Elspeth M. Beauchamp, Ahmet Dirim Arslan, Dany A. Curi, and Jessica K. Altman
- Subjects
Proto-Oncogene Proteins c-bcr ,philadelphia chromosome-positive leukemia ,Fusion Proteins, bcr-abl ,Genes, abl ,Philadelphia chromosome ,Proto-Oncogene Proteins c-pim-1 ,hemic and lymphatic diseases ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,medicine ,Tumor Cells, Cultured ,Humans ,Philadelphia Chromosome ,CML ,Protein Kinase Inhibitors ,PI3K/AKT/mTOR pathway ,PIM kinase ,Dose-Response Relationship, Drug ,Cell growth ,Kinase ,Chemistry ,Imidazoles ,medicine.disease ,3. Good health ,Pyridazines ,Leukemia ,Imatinib mesylate ,Oncology ,Immunology ,Mutation ,Cancer research ,mTOR signaling ,Imatinib Mesylate ,K562 Cells ,K562 cells ,Research Paper - Abstract
We investigated the efficacy of targeting the PIM kinase pathway in Philadelphia chromosome-positive (Ph+) leukemias. We provide evidence that inhibition of PIM, with the pan-PIM inhibitor SGI-1776, results in suppression of classic PIM effectors and also elements of the mTOR pathway, suggesting interplay between PIM and mTOR signals. Our data demonstrate that PIM inhibition enhances the effects of imatinib mesylate on Ph+ leukemia cells. We also found that PIM inhibition results in suppression of leukemic cell proliferation and induction of apoptosis of Ph+ leukemia cells, including those resistant to imatinib mesylate. Importantly, inhibition of PIM results in enhanced suppression of primary leukemic progenitors from patients with CML. Altogether these findings suggest that pharmacological PIM targeting may provide a unique therapeutic approach for the treatment of Ph+ leukemias.
- Published
- 2015
31. Central Role of ULK1 in Type I Interferon Signaling
- Author
-
Eleanor N. Fish, Craig B. Thompson, Diana Saleiro, Ross L. Levine, Ewa M. Kosciuczuk, Brandon McMahon, Pawel Lisowski, Jessica K. Altman, Tushar D. Bhagat, Leonidas C. Platanias, Darren P. Baker, Beata Majchrzak-Kita, Chunfa Jie, Barbara Kroczynska, Swarna Mehrotra, Amit Verma, Elspeth M. Beauchamp, Nadereh Jafari, and Brady L. Stein
- Subjects
MAPK/ERK pathway ,MAP Kinase Signaling System ,Protein Serine-Threonine Kinases ,Biology ,Response Elements ,p38 Mitogen-Activated Protein Kinases ,Article ,General Biochemistry, Genetics and Molecular Biology ,Mediator ,Erythroid Cells ,Interferon ,Cell Line, Tumor ,medicine ,Autophagy-Related Protein-1 Homolog ,Humans ,Protein kinase A ,lcsh:QH301-705.5 ,Cells, Cultured ,Regulation of gene expression ,Myeloproliferative Disorders ,Intracellular Signaling Peptides and Proteins ,ULK1 ,Gene Expression Regulation, Neoplastic ,lcsh:Biology (General) ,Interferon Regulatory Factors ,Interferon Type I ,Cancer research ,Interferon type I ,Interferon regulatory factors ,medicine.drug - Abstract
SummaryWe provide evidence that the Unc-51-like kinase 1 (ULK1) is activated during engagement of the type I interferon (IFN) receptor (IFNR). Our studies demonstrate that the function of ULK1 is required for gene transcription mediated via IFN-stimulated response elements (ISRE) and IFNγ activation site (GAS) elements and controls expression of key IFN-stimulated genes (ISGs). We identify ULK1 as an upstream regulator of p38α mitogen-activated protein kinase (MAPK) and establish that the regulatory effects of ULK1 on ISG expression are mediated possibly by engagement of the p38 MAPK pathway. Importantly, we demonstrate that ULK1 is essential for antiproliferative responses and type I IFN-induced antineoplastic effects against malignant erythroid precursors from patients with myeloproliferative neoplasms. Together, these data reveal a role for ULK1 as a key mediator of type I IFNR-generated signals that control gene transcription and induction of antineoplastic responses.
- Published
- 2015
- Full Text
- View/download PDF
32. Dual targeting of acute myeloid leukemia progenitors by catalytic mTOR inhibition and blockade of the p110α subunit of PI3 kinase
- Author
-
Leonidas C. Platanias, Francis J. Giles, Gavin T. Blyth, Jessica K. Altman, Elspeth M. Beauchamp, Marco Colamonici, Meghan Bliss-Moreau, Amy Szilard, and Diana Saleiro
- Subjects
Class I Phosphatidylinositol 3-Kinases ,Apoptosis ,Biology ,P110α ,Phosphatidylinositol 3-Kinases ,AML ,PI3 kinase ,Cell Line, Tumor ,hemic and lymphatic diseases ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Animals ,Humans ,Enzyme Inhibitors ,PI3K/AKT/mTOR pathway ,Phosphoinositide-3 Kinase Inhibitors ,Triazines ,Kinase ,TOR Serine-Threonine Kinases ,RPTOR ,Imidazoles ,Myeloid leukemia ,U937 Cells ,medicine.disease ,Leukemia, Myeloid, Acute ,Thiazoles ,Leukemia ,Oncology ,mTOR signaling ,Cancer research ,Signal transduction ,Research Paper ,Signal Transduction - Abstract
The mammalian target of rapamycin (mTOR) and phosphoinositide-3-kinase (PI3K) pathways are often aberrantly activated in acute myeloid leukemia (AML) and play critical roles in proliferation and survival of leukemia cells. We provide evidence that simultaneous targeting of mTOR complexes with the catalytic mTOR inhibitor OSI-027 and of the p110α subunit of PI3K with the specific inhibitor BYL-719 results in efficient suppression of effector pathways and enhanced induction of apoptosis of leukemia cells. Importantly, such a combined targeting approach results in enhanced suppression of primitive leukemic progenitors from patients with AML. Taken together, these findings raise the possibility of combination treatments of mTOR and p110α inhibitors as a unique approach to enhance responses in refractory AML.
- Published
- 2015
- Full Text
- View/download PDF
33. Differential Response of Glioma Stem Cells to Arsenic Trioxide Therapy Is Regulated by MNK1 and mRNA Translation
- Author
-
Ichiro Nakano, Craig Horbinski, Jeffrey Raizer, Kristiina Vuori, Leonidas C. Platanias, Barbara Kroczynska, Shi Yuan Cheng, Stewart Goldman, Kristen Alley, Darren Finlay, Frank Eckerdt, Andrew P. Mazar, Michael E. Berens, Seungchan Kim, C. David James, Priya Kumthekar, Jonathan B. Bell, Sen Peng, Harshil Dhruv, Jessica Clymer, and Elspeth M. Beauchamp
- Subjects
0301 basic medicine ,Cancer Research ,Antineoplastic Agents ,Biology ,Protein Serine-Threonine Kinases ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,Arsenic Trioxide ,Glioma ,Neurosphere ,Cell Line, Tumor ,medicine ,Animals ,Humans ,RNA, Messenger ,Arsenic trioxide ,Molecular Biology ,Cell Proliferation ,Cell growth ,Kinase ,EIF4E ,Intracellular Signaling Peptides and Proteins ,medicine.disease ,Xenograft Model Antitumor Assays ,030104 developmental biology ,Oncology ,chemistry ,Apoptosis ,Cancer research ,Neoplastic Stem Cells ,Stem cell ,Signal Transduction - Abstract
Mesenchymal (MES) and proneural (PN) are two distinct glioma stem cell (GSC) populations that drive therapeutic resistance in glioblastoma (GBM). We screened a panel of 650 small molecules against patient-derived GBM cells to discover compounds targeting specific GBM subtypes. Arsenic trioxide (ATO), an FDA-approved drug that crosses the blood–brain barrier, was identified as a potent PN-specific compound in the initial screen and follow-up validation studies. Furthermore, MES and PN GSCs exhibited differential sensitivity to ATO. As ATO has been shown to activate the MAPK-interacting kinase 1 (MNK1)-eukaryotic translation initiation factor 4E (eIF4E) pathway and subsequent mRNA translation in a negative regulatory feedback manner, the mechanistic role of ATO resistance in MES GBM was explored. In GBM cells, ATO-activated translation initiation cellular events via the MNK1–eIF4E signaling axis. Furthermore, resistance to ATO in intracranial PDX tumors correlated with high eIF4E phosphorylation. Polysomal fractionation and microarray analysis of GBM cells were performed to identify ATO's effect on mRNA translation and enrichment of anti-apoptotic mRNAs in the ATO-induced translatome was found. Additionally, it was determined that MNK inhibition sensitized MES GSCs to ATO in neurosphere and apoptosis assays. Finally, examination of the effect of ATO on patients from a phase I/II clinical trial of ATO revealed that PN GBM patients responded better to ATO than other subtypes as demonstrated by longer overall and progression-free survival. Implications: These findings raise the possibility of a unique therapeutic approach for GBM, involving MNK1 targeting to sensitize MES GSCs to drugs like arsenic trioxide. Mol Cancer Res; 16(1); 32–46. ©2017 AACR.
- Published
- 2017
34. Merestinib blocks Mnk kinase activity in acute myeloid leukemia progenitors and exhibits antileukemic effects in vitro and in vivo
- Author
-
Sameem Abedin, Diana Saleiro, Ewa M. Kosciuczuk, Jessica K. Altman, Francis J. Giles, Barbara Kroczynska, Leonidas C. Platanias, Frank Eckerdt, Elspeth M. Beauchamp, and Gavin T. Blyth
- Subjects
0301 basic medicine ,Myeloid ,Immunology ,Merestinib ,Biochemistry ,03 medical and health sciences ,Mice ,0302 clinical medicine ,hemic and lymphatic diseases ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Kinase activity ,Enzyme Inhibitors ,Protein kinase A ,neoplasms ,Cation Transport Proteins ,Adenosine Triphosphatases ,Myeloid Neoplasia ,Chemistry ,Kinase ,RUNX1T1 ,Myeloid leukemia ,Cell Biology ,Hematology ,medicine.disease ,Xenograft Model Antitumor Assays ,Neoplasm Proteins ,Leukemia ,Leukemia, Myeloid, Acute ,030104 developmental biology ,medicine.anatomical_structure ,Eukaryotic Initiation Factor-4E ,Copper-Transporting ATPases ,030220 oncology & carcinogenesis ,Cancer research - Abstract
Mitogen-activated protein kinase interacting protein kinases (Mnks) play important roles in the development and progression of acute myeloid leukemia (AML) by regulating eukaryotic translation initiation factor 4E (eIF4E) activation. Inhibiting Mnk1/2-induced phosphorylation of eIF4E may represent a unique approach for the treatment of AML. We provide evidence for antileukemic effects of merestinib, an orally bioavailable multikinase inhibitor with suppressive effects on Mnk activity. Our studies show that merestinib effectively blocks eIF4E phosphorylation in AML cells and suppresses primitive leukemic progenitors from AML patients in vitro and in an AML xenograft model in vivo. Our findings provide evidence for potent preclinical antileukemic properties of merestinib and support its clinical development for the treatment of patients with AML.
- Published
- 2016
35. Wnt signaling promotes proliferation and stemness regulation of spermatogonial stem/progenitor cells
- Author
-
Maria Kokkinaki, Elspeth M. Beauchamp, Aykut Üren, Shannon Fallen, Martin Dym, and Nady Golestaneh
- Subjects
Male ,endocrine system ,Embryology ,Cellular differentiation ,Cell ,Dishevelled Proteins ,Biology ,Cell fate determination ,Transfection ,Mice ,Endocrinology ,Cell Movement ,Genes, Reporter ,Wnt3A Protein ,medicine ,Animals ,Humans ,Phosphorylation ,Progenitor cell ,Cell Shape ,Cells, Cultured ,beta Catenin ,Adaptor Proteins, Signal Transducing ,Cell Proliferation ,Mice, Inbred BALB C ,Cell growth ,Stem Cells ,Wnt signaling pathway ,Obstetrics and Gynecology ,Cell Differentiation ,Cell migration ,Cell Biology ,Phosphoproteins ,Embryonic stem cell ,Frizzled Receptors ,Spermatogonia ,Cell biology ,Wnt Proteins ,medicine.anatomical_structure ,Reproductive Medicine ,Culture Media, Conditioned ,Signal Transduction - Abstract
Spermatogonial stem cells (SSCs) self-renew throughout life to produce progenitor cells that are able to differentiate into spermatozoa. However, the mechanisms underlying the cell fate determination between self-renewal and differentiation have not yet been delineated. Culture conditions and growth factors essential for self-renewal and proliferation of mouse SSCs have been investigated, but no information is available related to growth factors that affect fate determination of human spermatogonia. Wnts form a large family of secreted glycoproteins, the members of which are involved in cell proliferation, differentiation, organogenesis, and cell migration. Here, we show that Wnts and their receptors Fzs are expressed in mouse spermatogonia and in the C18-4 SSC line. We demonstrate that WNT3A induces cell proliferation, morphological changes, and cell migration in C18-4 cells. Furthermore, we show that β-catenin is activated during testis development in 21-day-old mice. In addition, our study demonstrates that WNT3A sustained adult human embryonic stem (ES)-like cells derived from human germ cells in an undifferentiated stage, expressing essential human ES cell transcription factors. These results demonstrate for the first time that Wnt/β-catenin pathways, especially WNT3A, may play an important role in the regulation of mouse and human spermatogonia.
- Published
- 2009
- Full Text
- View/download PDF
36. Human Schlafen 5 (SLFN5) Is a Regulator of Motility and Invasiveness of Renal Cell Carcinoma Cells
- Author
-
Amit Verma, Elspeth M. Beauchamp, Leonidas C. Platanias, Barbara Kroczynska, Satya Khuon, Evangelos Mavrommatis, Hidayatullah G. Munshi, Kathleen J. Green, Ahmet Dirim Arslan, Antonella Sassano, and Ten Leong Chew
- Subjects
Regulator ,Alpha interferon ,Motility ,Cell Cycle Proteins ,Matrix metalloproteinase ,Biology ,urologic and male genital diseases ,Interferon ,RNA interference ,Renal cell carcinoma ,Cell Movement ,Cell Line, Tumor ,Matrix Metalloproteinase 13 ,medicine ,Humans ,Neoplasm Invasiveness ,RNA, Messenger ,RNA, Small Interfering ,Molecular Biology ,Carcinoma, Renal Cell ,Cell Proliferation ,Cell growth ,Interferon-alpha ,Cell Biology ,medicine.disease ,Kidney Neoplasms ,Cell biology ,Gene Expression Regulation, Neoplastic ,RNA Interference ,Matrix Metalloproteinase 1 ,medicine.drug - Abstract
We provide evidence that human SLFN5, an interferon (IFN)-inducible member of the Schlafen (SLFN) family of proteins, exhibits key roles in controlling motility and invasiveness of renal cell carcinoma (RCC) cells. Our studies define the mechanism by which this occurs, demonstrating that SLFN5 negatively controls expression of the matrix metalloproteinase 1 gene (MMP-1), MMP-13, and several other genes involved in the control of malignant cell motility. Importantly, our data establish that SLFN5 expression correlates with a better overall survival in a large cohort of patients with RCC. The inverse relationship between SLFN5 expression and RCC aggressiveness raises the possibility of developing unique therapeutic approaches in the treatment of RCC, by modulating SLFN5 expression.
- Published
- 2015
37. Direct binding of arsenic trioxide to AMPK and generation of inhibitory effects on acute myeloid leukemia precursors
- Author
-
Elden P. Swindell, Ewa M. Kosciuczuk, Elspeth M. Beauchamp, Benoit Viollet, Leonidas C. Platanias, Jessica K. Altman, Dhaval Nanavati, Thomas V. O'Halloran, and Ruth Serrano
- Subjects
MAPK/ERK pathway ,Cancer Research ,Antineoplastic Agents ,Biology ,AMP-Activated Protein Kinases ,Ribosomal Protein S6 Kinases, 90-kDa ,Arsenicals ,Article ,chemistry.chemical_compound ,Mice ,Arsenic Trioxide ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Arsenic trioxide ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,Kinase ,TOR Serine-Threonine Kinases ,Myeloid leukemia ,AMPK ,Oxides ,medicine.disease ,Xenograft Model Antitumor Assays ,Leukemia ,Leukemia, Myeloid, Acute ,Oncology ,chemistry ,Biochemistry ,Cancer research ,Phosphorylation ,Signal Transduction - Abstract
Arsenic trioxide (As2O3) exhibits potent antineoplastic effects and is used extensively in clinical oncology for the treatment of a subset of patients with acute myeloid leukemia (AML). Although As2O3 is known to regulate activation of several signaling cascades, the key events, accounting for its antileukemic properties, remain to be defined. We provide evidence that arsenic can directly bind to cysteine 299 in AMPKα and inhibit its activity. This inhibition of AMPK by arsenic is required in part for its cytotoxic effects on primitive leukemic progenitors from patients with AML, while concomitant treatment with an AMPK activator antagonizes in vivo the arsenic-induced antileukemic effects in a xenograft AML mouse model. A consequence of AMPK inhibition is activation of the mTOR pathway as a negative regulatory feedback loop. However, when AMPK expression is lost, arsenic-dependent activation of the kinase RSK downstream of MAPK activity compensates the generation of regulatory feedback signals through phosphorylation of downstream mTOR targets. Thus, therapeutic regimens with As2O3 will need to include inhibitors of both the mTOR and RSK pathways in combination to prevent engagement of negative feedback loops and maximize antineoplastic responses. Mol Cancer Ther; 14(1); 202–12. ©2014 AACR.
- Published
- 2014
38. Autophagy is a survival mechanism of acute myeloid leukemia precursors during dual mTORC2/mTORC1 targeting
- Author
-
Antonella Sassano, Amy Szilard, Francis J. Giles, Elias Gounaris, Dennis J. Goussetis, Marco Colamonici, Leonidas C. Platanias, Elizabeth A. Eklund, Jessica K. Altman, Elspeth M. Beauchamp, and Olga Frankfurt
- Subjects
Cancer Research ,Myeloid ,Cell Survival ,mTORC1 ,Mechanistic Target of Rapamycin Complex 2 ,Biology ,Mechanistic Target of Rapamycin Complex 1 ,mTORC2 ,Article ,Myelogenous ,Cell Line, Tumor ,medicine ,Autophagy ,Humans ,Progenitor cell ,Clonogenic assay ,Protein Kinase Inhibitors ,TOR Serine-Threonine Kinases ,medicine.disease ,Cell biology ,Leukemia ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,Oncology ,Multiprotein Complexes ,Cancer research ,Neoplastic Stem Cells - Abstract
Purpose: To examine whether induction of autophagy is a mechanism of leukemic cell resistance to dual mTORC1/mTORC2 inhibitors in acute myelogenous leukemia (AML) leukemic progenitors. Experimental Design: Combinations of different experimental approaches were used to assess induction of autophagy, including immunoblotting to detect effects on LC3II and p62/SQTM1 expression and on ULK1 phosphorylation, immunofluorescence, and electron microscopy. Functional responses were assessed using cell viability and apoptosis assays, and clonogenic leukemic progenitor assays in methylcellulose. Results: We provide evidence that treatment of AML cells with catalytic mTOR inhibitors results in induction of autophagy, which acts as a regulatory mechanism to promote leukemic cell survival. Such induction of autophagy by dual mTORC1/mTORC2 inhibitors partially protects primitive leukemic precursors from the inhibitory effects of such agents and limits their activities. Simultaneous blockade of the autophagic process using chloroquine or by knockdown of ULK1 results in enhanced antileukemic responses. Conclusions: Dual targeting of mTORC2 and mTORC1 results in induction of autophagy in AML cells. Combinations of catalytic mTOR targeting agents and autophagy inhibitors may provide a unique approach to target primitive leukemic precursors in AML. Clin Cancer Res; 20(9); 2400–9. ©2014 AACR.
- Published
- 2014
39. Resveratrol enhances the suppressive effects of arsenic trioxide on primitive leukemic progenitors
- Author
-
Edward J. Wu, Dennis J. Goussetis, Leonidas C. Platanias, Jessica K. Altman, Ewa M. Kosciuczuk, Elspeth M. Beauchamp, and Elizabeth A. Eklund
- Subjects
Cancer Research ,Antineoplastic Agents ,Apoptosis ,Resveratrol ,Pharmacology ,Arsenicals ,chemistry.chemical_compound ,Arsenic Trioxide ,hemic and lymphatic diseases ,Cell Line, Tumor ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,Stilbenes ,medicine ,Autophagy ,Humans ,Progenitor cell ,Arsenic trioxide ,Clonogenic assay ,Myeloid leukemia ,Drug Synergism ,Oxides ,medicine.disease ,Leukemia ,Leukemia, Myeloid, Acute ,Oncology ,chemistry ,Neoplastic Stem Cells ,Molecular Medicine ,Stem cell ,Research Paper - Abstract
Efforts to enhance the antileukemic properties of arsenic trioxide are clinically relevant and may lead to the development of new therapeutic approaches for the management of certain hematological malignancies. We provide evidence that concomitant treatment of acute myeloid leukemia (AML) cells or chronic myeloid leukemia (CML) cells with resveratrol potentiates arsenic trioxide-dependent induction of apoptosis. Importantly, clonogenic assays in methylcellulose demonstrate potent suppressive effects of the combination of these agents on primitive leukemic progenitors derived from patients with AML or CML. Taken together, these findings suggest that combinations of arsenic trioxide with resveratrol may provide an approach for targeting of early leukemic precursors and, possibly, leukemia initiating stem cells.
- Published
- 2014
40. Regulatory Effects of Arsenic on Cellular Signaling Pathways: Biological Effects and Therapeutic Implications
- Author
-
Leonidas C. Platanias, Elspeth M. Beauchamp, and Ruth Serrano
- Subjects
MAPK/ERK pathway ,Acute promyelocytic leukemia ,Cell signaling ,chemistry.chemical_element ,medicine.disease ,Fusion protein ,chemistry.chemical_compound ,chemistry ,Cancer cell ,Cancer research ,medicine ,Arsenic trioxide ,PI3K/AKT/mTOR pathway ,Arsenic - Abstract
Arsenic compounds exert important biological effects and arsenic trioxide has been approved by the Food and Drug Administration (FDA) for the treatment of patients with acute promyelocytic leukemia (APL). Much of arsenic’s actions in cells reflect its ability to bind thiol groups in cellular proteins or to affect the production of reactive oxygen species (ROS), leading to the engagement and regulation of several cellular signaling pathways. Arsenic has been also shown to degrade abnormal fusion proteins found in myeloid leukemias. It has also been shown to effect NFκB, MAPK, mTOR and Hedgehog pathways which can modulate the viability of cancer cells. Many clinical trials have been performed to examine the clinical efficacy of arsenic trioxide alone or in combination with other agents in the treatment of various hematological malignancies. The continuous advances in basic and translational research and the better understanding of the mechanisms of action of arsenic should lead to more effective combinations with other agents that could result in better clinical outcomes.
- Published
- 2013
- Full Text
- View/download PDF
41. The evolution of the TOR pathway and its role in cancer
- Author
-
Leonidas C. Platanias and Elspeth M. Beauchamp
- Subjects
Regulation of gene expression ,Cancer Research ,Models, Genetic ,TOR Serine-Threonine Kinases ,Autophagy ,Antineoplastic Agents ,Nutrient sensing ,Biology ,medicine.disease_cause ,Cell biology ,Cell Physiological Phenomena ,Evolution, Molecular ,Gene Expression Regulation ,Neoplasms ,Genetics ,medicine ,TOR complex ,Animals ,Humans ,Signal transduction ,Carcinogenesis ,Molecular Biology ,PI3K/AKT/mTOR pathway ,Signal Transduction - Abstract
The target of rapamycin (TOR) pathway is highly conserved among eukaryotes and has evolved to couple nutrient sensing to cellular growth. TOR is found in two distinct signaling complexes in cells, TOR complex 1 (TORC1) and TOR complex 2 (TORC2). These complexes are differentially regulated and act as effectors for the generation of signals that drive diverse cellular processes such as growth, proliferation, protein synthesis, rearrangement of the cytoskeleton, autophagy, metabolism and survival. Mammalian TOR (mTOR) is very important for development in embryos, while in adult organisms it is linked to aging and lifespan effects. In humans, the mTOR pathway is implicated in the tumorigenesis of multiple cancer types and its deregulation is associated with familial cancer syndromes. Because of its high biological relevance, different therapeutic strategies have been developed to target this signaling cascade, resulting in the emergence of unique pharmacological inhibitors that are either already approved for use in clinical oncology or currently under preclinical or clinical development. Multimodal treatment strategies that simultaneously target multiple nodes of the pathway and/or negative feedback regulatory loops may ultimately provide the best therapeutic advantage in targeting this pathway for the treatment of malignancies.
- Published
- 2012
42. A new era for an ancient drug: arsenic trioxide and Hedgehog signaling
- Author
-
Elspeth M, Beauchamp and Aykut, Uren
- Subjects
Oncogene Proteins ,Arsenic Trioxide ,Leukemia, Promyelocytic, Acute ,Humans ,Antineoplastic Agents ,Bone Neoplasms ,Hedgehog Proteins ,Oxides ,Sarcoma, Ewing ,Cerebellar Neoplasms ,Arsenicals ,Medulloblastoma - Abstract
Arsenic has been used for ages as a therapeutic agent. Currently, it is an FDA approved drug to treat acute promyelocytic leukemia where it leads to degradation of the PML-RAR fusion protein. It has been shown to have various other targets in cells such as JNK, NFκB, thioredoxin reductase, and MAPK pathways. Most of its effects in cells have been through arsenic's ability to bind to thiol groups in cysteine residues. Recent evidence has shown that arsenic can inhibit the Hedgehog pathway by inhibiting GLI proteins. The proposed mechanism of action is through direct binding. Potential binding sites include the critical cysteine residues in GLI zinc finger domains. The role of the Hedgehog pathway has been implicated in many cancers such as basal cell carcinoma, medulloblastoma, Ewing sarcoma, and rhabdoid tumors. Current Hedgehog pathway inhibitors have been fraught with resistance issues and so arsenic trioxide may provide an alternative therapy when combined with these other inhibitors or after acquired resistance.
- Published
- 2012
43. A New Era for an Ancient Drug
- Author
-
Aykut Üren and Elspeth M. Beauchamp
- Subjects
MAPK/ERK pathway ,Acute promyelocytic leukemia ,Zinc finger ,Thioredoxin reductase ,Biology ,medicine.disease ,Fusion protein ,Hedgehog signaling pathway ,chemistry.chemical_compound ,Biochemistry ,chemistry ,medicine ,Cancer research ,Arsenic trioxide ,Hedgehog - Abstract
Arsenic has been used for ages as a therapeutic agent. Currently, it is an FDA approved drug to treat acute promyelocytic leukemia where it leads to degradation of the PML-RAR fusion protein. It has been shown to have various other targets in cells such as JNK, NFκB, thioredoxin reductase, and MAPK pathways. Most of its effects in cells have been through arsenic's ability to bind to thiol groups in cysteine residues. Recent evidence has shown that arsenic can inhibit the Hedgehog pathway by inhibiting GLI proteins. The proposed mechanism of action is through direct binding. Potential binding sites include the critical cysteine residues in GLI zinc finger domains. The role of the Hedgehog pathway has been implicated in many cancers such as basal cell carcinoma, medulloblastoma, Ewing sarcoma, and rhabdoid tumors. Current Hedgehog pathway inhibitors have been fraught with resistance issues and so arsenic trioxide may provide an alternative therapy when combined with these other inhibitors or after acquired resistance.
- Published
- 2012
- Full Text
- View/download PDF
44. Beta-catenin accelerates human papilloma virus type-16 mediated cervical carcinogenesis in transgenic mice
- Author
-
Aykut Üren, Deborah L. Berry, Bhaskar Kallakury, Lauren E. Drebing, Junfeng Sun, Gülay Bulut, Elspeth M. Beauchamp, Richard Schlegel, Christopher P. Crum, Jeffrey A. Toretsky, and Shannon Fallen
- Subjects
Mouse ,Papillomavirus E7 Proteins ,Uterine Cervical Neoplasms ,lcsh:Medicine ,Cervical Cancer ,Mice ,0302 clinical medicine ,Molecular Cell Biology ,Signaling in Cellular Processes ,lcsh:Science ,Wnt Signaling Pathway ,beta Catenin ,WNT Signaling Cascade ,Cervical cancer ,0303 health sciences ,Human papillomavirus 16 ,Multidisciplinary ,integumentary system ,Wnt signaling pathway ,Animal Models ,Beta-Catenin Signaling ,Signaling Cascades ,3. Good health ,medicine.anatomical_structure ,Cell Transformation, Neoplastic ,Phenotype ,Oncology ,030220 oncology & carcinogenesis ,Disease Progression ,Medicine ,Female ,Research Article ,Signal Transduction ,Genetically modified mouse ,Beta-catenin ,medicine.drug_class ,Transgene ,Mice, Transgenic ,Biology ,Signaling Pathways ,03 medical and health sciences ,Catenin Signal Transduction ,Model Organisms ,medicine ,Animals ,Humans ,Cervix ,030304 developmental biology ,Oncogene ,lcsh:R ,Cancers and Neoplasms ,Estrogens ,medicine.disease ,Disease Models, Animal ,Estrogen ,Immunology ,biology.protein ,Cancer research ,lcsh:Q ,Gynecological Tumors - Abstract
Human papilloma virus (HPV) is the principal etiological agent of cervical cancer in women, and its DNA is present in virtually all of these tumors. However, exposure to the high-risk HPV types alone is insufficient for tumor development. Identifying specific collaborating factors that will lead to cervical cancer remains an unanswered question, especially because millions of women are exposed to HPV. Our earlier work using an in vitro model indicated that activation of the canonical Wnt pathway in HPV-positive epithelial cells was sufficient to induce anchorage independent growth. We therefore hypothesized that constitutive activation of this pathway might function as the “second hit.” To address this possibility, we developed two double-transgenic (DT) mouse models, K14-E7/ΔN87βcat and K14-HPV16/ΔN87βcat that express either the proteins encoded by the E7 oncogene or the HPV16 early region along with constitutively active β-catenin, which was expressed by linking it to the keratin-14 (K14) promoter. We initiated tumor formation by treating all groups with estrogen for six months. Invasive cervical cancer was observed in 11% of the K14-ΔN87βcat mice, expressing activated β-catenin and in 50% of the animals expressing the HPV16 E7 oncogene. In double-transgenic mice, coexpression of β-catenin and HPV16 E7 induced invasive cervical cancer at about 7 months in 94% of the cases. We did not observe cervical cancer in any group unless the mice were treated with estrogen. In the second model, K14-HPV16 mice suffered cervical dysplasias, but this phenotype was not augmented in HPV16/ΔN87βcat mice. In summary, the phenotypes of the K14-E7/ΔN87βcat mice support the hypothesis that activation of the Wnt/β-catenin pathway in HPV-associated premalignant lesions plays a functional role in accelerating cervical carcinogenesis.
- Published
- 2011
45. Overcoming treatment challenges in imatinib-resistant chronic myelogenous leukemia
- Author
-
Leonidas C. Platanias, Dany A. Curi, and Elspeth M. Beauchamp
- Subjects
Cancer Research ,business.industry ,Kinesins ,Mitosis ,Apoptosis ,Hematology ,medicine.disease ,Imatinib resistant ,Oncology ,Chromones ,Drug Resistance, Neoplasm ,Benzamides ,medicine ,Cancer research ,Humans ,business ,Chronic myelogenous leukemia - Published
- 2014
- Full Text
- View/download PDF
46. Arsenic trioxide inhibits human cancer cell growth and tumor development in mice by blocking Hedgehog/GLI pathway
- Author
-
Jeffrey A. Toretsky, Kamal P. Sajwan, Lymor Ringer, Metin Ozdemirli, Olga Rodriguez, Tobey J. MacDonald, Gülay Bulut, Michael D. Hall, Daniel Peaceman, Chris Albanese, Aykut Üren, Elspeth M. Beauchamp, and Yichien Lee
- Subjects
Antineoplastic Agents ,Sarcoma, Ewing ,Biology ,Models, Biological ,Zinc Finger Protein GLI1 ,Arsenicals ,Receptors, G-Protein-Coupled ,chemistry.chemical_compound ,Mice ,Arsenic Trioxide ,GLI1 ,Cell Line, Tumor ,Neoplasms ,Animals ,Humans ,Hedgehog Proteins ,Arsenic trioxide ,Hedgehog ,Transcription factor ,Oncogene ,integumentary system ,Cell growth ,Oxides ,General Medicine ,Smoothened Receptor ,Transplantation ,chemistry ,Immunology ,biology.protein ,Cancer research ,Signal transduction ,Research Article ,Medulloblastoma ,Signal Transduction ,Transcription Factors - Abstract
The Hedgehog (Hh) pathway is activated in some human cancers, including medulloblastoma. The glioma-associated oncogene homolog (GLI) transcription factors are critical mediators of the activated Hh pathway, and their expression may be elevated in some tumors independent of upstream Hh signaling. Thus, therapies targeting GLI transcription factors may benefit a wide spectrum of patients with mutations at different nodal points of the Hh pathway. In this study, we present evidence that arsenic trioxide (ATO) suppresses human cancer cell growth and tumor development in mice by inhibiting GLI1. Mechanistically, ATO directly bound to GLI1 protein, inhibited its transcriptional activity, and decreased expression of endogenous GLI target genes. Consistent with this, ATO inhibited the growth of human cancer cell lines that depended on upregulated GLI expression in vitro and in vivo in a xenograft model of Ewing sarcoma. Furthermore, ATO improved survival of a clinically relevant spontaneous mouse model of medulloblastoma with activated Hh pathway signaling. Our results establish ATO as a Hh pathway inhibitor acting at the level of GLI1 both in vitro and in vivo. These results warrant the clinical investigation of ATO for tumors with activated Hh/GLI signaling, in particular patients who develop resistance to current therapies targeting the Hh pathway upstream of GLI.
- Published
- 2010
47. Wnt-3a and Dickkopf-1 stimulate neurite outgrowth in Ewing tumor cells via a Frizzled3- and c-Jun N-terminal kinase-dependent mechanism
- Author
-
Elspeth M. Beauchamp, Jeffrey A. Toretsky, Aykut Üren, Yoshimi Endo, Jeffrey S. Rubin, David L. Woods, and William G. Taylor
- Subjects
Small interfering RNA ,Frizzled ,Neurite ,Recombinant Fusion Proteins ,Dishevelled Proteins ,Sarcoma, Ewing ,Wnt1 Protein ,Biology ,Receptors, G-Protein-Coupled ,Wnt3 Protein ,Cell Line, Tumor ,Wnt3A Protein ,Neurites ,Humans ,RNA, Small Interfering ,Molecular Biology ,LDL-Receptor Related Proteins ,Adaptor Proteins, Signal Transducing ,Wnt signaling pathway ,JNK Mitogen-Activated Protein Kinases ,LRP6 ,Membrane Proteins ,LRP5 ,Cell Biology ,Articles ,Phosphoproteins ,Molecular biology ,Frizzled Receptors ,Cell biology ,Neoplasm Proteins ,Wnt Proteins ,Low Density Lipoprotein Receptor-Related Protein-5 ,DKK1 ,Low Density Lipoprotein Receptor-Related Protein-6 ,Intercellular Signaling Peptides and Proteins - Abstract
Recombinant Wnt-3a stimulated the rapid formation of elongated processes in Ewing sarcoma family tumor (ESFT) cells that were identified as neurites. The processes stained positively for polymerized actin and microtubules as well as synapsin I and growth-associated protein 43. Inhibition of the Wnt receptor, Frizzled3 (Fzd3), with antiserum or by short interfering RNA (siRNA) markedly reduced neurite extension. Knockdown of Dishevelled-2 (Dvl-2) and Dvl-3 also suppressed neurite outgrowth. Surprisingly, disruption of the Wnt/Fzd/lipoprotein receptor-related protein (LRP) complex and the associated beta-catenin signaling by treating cells either with the Wnt antagonist Dickkopf-1 (Dkk1) or LRP5/LRP6 siRNA enhanced neuritogenesis. Neurite outgrowth induced by Dkk1 or with LRP5/LRP6 siRNA was inhibited by secreted Fzd-related protein 1, a Wnt antagonist that binds directly to Wnt. Moreover, Dkk1 stimulation of neurite outgrowth was blocked by Fzd3 siRNA. These results suggested that Dkk1 shifted endogenous Wnt activity from the beta-catenin pathway to Fzd3-mediated, noncanonical signaling that is responsible for neurite formation. In particular, c-Jun amino-terminal kinase (JNK) was important for neurite outgrowth stimulated by both Wnt-3a and Dkk1. Our data demonstrate that Fzd3, Dvl, and JNK activity mediate Wnt-dependent neurite outgrowth and that ESFT cell lines will be useful experimental models for the study of Wnt-dependent neurite extension.
- Published
- 2008
48. YK-4-279 Inhibits ERG and ETV1 Mediated Prostate Cancer Cell Invasion
- Author
-
Elspeth M. Beauchamp, Yali Kong, Said Rahim, Jeffrey A. Toretsky, Milton L. Brown, and Aykut Üren
- Subjects
Male ,Indoles ,Cancer Treatment ,lcsh:Medicine ,Biochemistry ,ETV1 ,Metastasis ,Prostate cancer ,0302 clinical medicine ,Chlorocebus aethiops ,Drug Discovery ,Basic Cancer Research ,lcsh:Science ,Transcriptional Regulator ERG ,0303 health sciences ,Multidisciplinary ,Prostate Cancer ,Prostate Diseases ,Chromoplexy ,3. Good health ,DNA-Binding Proteins ,Oncology ,030220 oncology & carcinogenesis ,FLI1 ,COS Cells ,Medicine ,Erg ,Research Article ,Chromatin Immunoprecipitation ,Drugs and Devices ,Drug Research and Development ,Urology ,Blotting, Western ,Antineoplastic Agents ,Biology ,03 medical and health sciences ,Cell Line, Tumor ,LNCaP ,medicine ,Animals ,Humans ,Neoplasm Invasiveness ,030304 developmental biology ,lcsh:R ,Prostatic Neoplasms ,Cancers and Neoplasms ,Chemotherapy and Drug Treatment ,medicine.disease ,Molecular biology ,Kinetics ,Genitourinary Tract Tumors ,Small Molecules ,Trans-Activators ,lcsh:Q ,Drug Screening Assays, Antitumor ,Transcription Factors - Abstract
Background Genomic rearrangements involving the ETS family of transcription factors occur in 40–70% of prostate cancer cases. ERG and ETV1 are the most common ETS members observed in these genetic alterations. The high prevalence of these rearrangements and their biological significance represents a novel therapeutic target for the treatment of prostate cancer. Methods and Findings We recently reported the development of YK-4-279, a small molecule inhibitor of EWS-FLI1 oncoprotein in Ewing's Sarcoma. Since ERG and ETV1 belong to the same class of ETS factors as FLI1, we tested the ability of YK-4-279 to inhibit biological functions of ERG and ETV1 proteins in prostate cancer. YK-4-279 inhibited ERG and ETV1 mediated transcriptional activity in a luciferase assay. YK-4-279 also decreased ERG and ETV1 downstream target mRNA and protein expression in ETV1-fusion positive LNCaP and ERG fusion positive VCaP cells. YK-4-279 reduced the motility of LNCaP cells in a scratch assay and the invasive phenotype of both LNCaP and VCaP cells in a HUVEC invasion assay. Fusion-negative PC3 cells were unresponsive to YK-4-279. SiRNA mediated ERG knockdown in VCaP cells resulted in a loss of drug responsiveness. Concurrently, transient ERG expression in PC-3 cells resulted in increased invasive potential, which was reduced by YK-4-279. Conclusion These data demonstrate that YK-4-279 inhibits ERG and ETV1 biological activity in fusion-positive prostate cancer cells leading to decreased motility and invasion. Therefore, YK-4-279 may have an impact on metastasis in prostate cancer and it may be further evaluated for its clinical applications in prostate cancer in addition to Ewing's sarcoma.
- Published
- 2011
- Full Text
- View/download PDF
49. Abstract 4328: Beta-catenin accelerates human papillomavirus type16 -E7 mediated cervical carcinogenesis in transgenic mice
- Author
-
Richard Schlegel, Jeffrey A. Toretsky, Lauren E. Drebing, Christopher P. Crum, Aykut Üren, Deborah L. Berry, Bhaskar Kallakury, Shannon Fallen, Gülay Bulut, Elspeth M. Beauchamp, and Junfeng Sun
- Subjects
Cervical cancer ,Cancer Research ,Beta-catenin ,integumentary system ,biology ,Oncogene ,business.industry ,Transgene ,Wnt signaling pathway ,Cancer ,medicine.disease ,Malignant transformation ,Oncology ,Catenin ,Immunology ,biology.protein ,Medicine ,business - Abstract
Human papilloma virus (HPV) is the principal etiological agent of cervical cancer in women and its DNA is present and expressed in virtually all cervical cancers. Exposure to the high-risk HPV types is not sufficient for tumor development. Approximately 80% of women will be infected with HPV in their lifetime, however only a small percentage of infected women develop cervical cancer. It is unknown who among the millions of women infected with HPV will go on to develop cervical cancer. Therefore, it remains critical to discover additional cellular changes that lead to malignant transformation. The persistence of viral infection is one critical factor in the predisposition to cancer, presumably providing genetic instability that leads to additional genetic/epigenetic changes. Our earlier work using an in vitro model indicated that activation of the canonical Wnt pathway in HPV-positive epithelial cells was sufficient to induce anchorage independent growth. Therefore, we hypothesized that constitutive activation of the Wnt pathway might function as a second hit. To address this possibility, we generated two new double transgenic (DT) mouse models. The first model, K14-HPV16/ΔN87βcat is established by crossing K14-ΔN87βcat mice with K14-HPV16 mice. These mice express wild type HPV16 early region oncoproteins including E6 and E7 and constitutively active -catenin protein in cervical epithelia under the control of tissue specific K14 promoter. The second model, K14-E7/ΔN87βcat mice were generated by crossing K14-ΔN87cat cat mice with K14-E7 mice and they express only the E7 oncoprotein and constitutively active -catenin under the same K14 promoter. All the transgenic animals in both crosses were maintained as heterozygotes. Within the K14-HPV16/ΔN87βcat model, we did not observe any cervical pathology in the wild type and K14-ΔN87βcat mice, whereas we observed CINIII in 26.7% of K14-HPV16 animals and CINIII and invasive tumors in 30% of the double transgenic animals at an average of 6 months of age. Within the K14-E7/ΔN87βcat model, wild type animals did not develop any cervical pathology. Invasive cervical cancer was observed in 10.5% of the animals expressing activated β-catenin and in 50% of the animals expressing HPV16-E7 oncogene. In double transgenic animals, expression of β-catenin and the HPV16-E7 oncogene induced invasive cervical cancer at an average age of 6 months in 93.75% of the cases. In summary, our data support the hypothesis that activation of the Wnt/β-catenin pathway in HPV pre-malignant lesions has a functional role in accelerating cervical carcinogenesis. These findings may have both preventive and therapeutic applications for cervical cancer patients. HPV positive patients may be screened for activation of Wnt signaling for early intervention. Late stage patients may benefit from novel anti-beta-catenin agents. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 4328. doi:10.1158/1538-7445.AM2011-4328
- Published
- 2011
- Full Text
- View/download PDF
50. Abstract 663: YK-4-279 inhibits ERG and ETV1 mediated prostate cancer cell invasion
- Author
-
Aykut Üren, Said Rahim, Elspeth M. Beauchamp, Yali Kong, Jeffrey A. Toretsky, and Milton L. Brown
- Subjects
Cancer Research ,Promoter ,Biology ,medicine.disease ,TMPRSS2 ,ETV1 ,Metastasis ,Prostate cancer ,medicine.anatomical_structure ,Oncology ,Prostate ,FLI1 ,Immunology ,LNCaP ,Cancer research ,medicine - Abstract
Chromosomal translocations involving the ETS family of transcription factors are found in a majority of prostate cancers, including the most clinically aggressive forms. These translocations produce a chimeric gene, which fuses the promoter region of an androgen responsive gene, such as TMPRSS2, to the coding region of ETS factors, most frequently ETV1 or ERG. Over-expression of ETS factors in prostate cancer cells results in a more invasive phenotype. The high prevalence of these rearrangements, and their biological significance represents a novel therapeutic target for the treatment of prostate cancer. We recently reported the development of YK-4-279, a small molecule inhibitor of EWS-FLI1 oncoprotein in Ewing's Sarcoma. ERG and ETV1 belong to the same class of ETS factors as FLI1. Due to the close homology of FLI1 with ERG and ETV1, we tested the ability of YK-4-279 to inhibit ETS biological activity in prostate cell-lines such as VCaP and LNCaP that demonstrate androgen dependent ERG and ETV1 expression, respectively. YK-4-279 inhibited ERG and ETV1 mediated transcription of target genes such as PLAU, PLAT, ADAM19 and MMP-13, which are involved in breakdown of the extracellular matrix and metastasis. YK-4-279 reduced the motility and invasive phenotype of ETV1-fusion positive LNCaP cells and ERG fusion-positive VCaP cells in in vitro invasion assays. However, ETS fusion-negative PC-3 cells were unresponsive to YK-4-279. ERG knockdown in VCaP cells resulted in a loss of drug responsiveness. Transient ERG expression in PC-3 cells resulted in an increased invasive phenotype, which was reduced by YK-4-279. Our results demonstrate that YK-4-279 inhibits ERG and ETV1 biological activity in fusion-positive prostate cancer cells leading to decreased motility and invasion. Therefore, YK-4-279 may have an impact on metastasis in prostate cancer, which is a leading cause of death, and it may be further evaluated for its clinical applications in prostate cancer, in addition to Ewing's sarcoma. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 663. doi:10.1158/1538-7445.AM2011-663
- Published
- 2011
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.