58 results on '"Peter J. Tummino"'
Search Results
2. Supplementary Figure 5 from A687V EZH2 Is a Driver of Histone H3 Lysine 27 (H3K27) Hypertrimethylation
- Author
-
Michael T. McCabe, Caretha L. Creasy, Peter J. Tummino, Benjamin Schwartz, Dashyant Dhanak, Ryan G. Kruger, Steven D. Knight, Sharad K. Verma, Roland Annan, Yuchen Bai, Yong Jiang, Edward Dul, Arthur Groy, Ashley M. Hughes, Wendy S. Halsey, Michael Huddleston, Melissa B. Pappalardi, Alan P. Graves, and Heidi M. Ott
- Abstract
Supplementary Figure 5: H3K27me3 enrichment across genes modulated by EZH2 inhibition
- Published
- 2023
3. Supplementary Table Legend, Figure Legends 1-2 from ALDH1A1 Is a Novel EZH2 Target Gene in Epithelial Ovarian Cancer Identified by Genome-Wide Approaches
- Author
-
Rugang Zhang, Michael J. Birrer, Paul Cairns, Peter J. Tummino, Caretha L. Creasy, Michael Slifker, Marie E. Maradeo, Vinod Vathipadiekal, Benjamin G. Bitler, and Hua Li
- Abstract
PDF 89K
- Published
- 2023
4. Supplementary Figure 6 from A687V EZH2 Is a Driver of Histone H3 Lysine 27 (H3K27) Hypertrimethylation
- Author
-
Michael T. McCabe, Caretha L. Creasy, Peter J. Tummino, Benjamin Schwartz, Dashyant Dhanak, Ryan G. Kruger, Steven D. Knight, Sharad K. Verma, Roland Annan, Yuchen Bai, Yong Jiang, Edward Dul, Arthur Groy, Ashley M. Hughes, Wendy S. Halsey, Michael Huddleston, Melissa B. Pappalardi, Alan P. Graves, and Heidi M. Ott
- Abstract
Supplementary Figure 6: Gene Set Enrichment Analysis (GSEA) for SUP-B8 cells treated with GSK126
- Published
- 2023
5. Supplementary Table 2 from A687V EZH2 Is a Driver of Histone H3 Lysine 27 (H3K27) Hypertrimethylation
- Author
-
Michael T. McCabe, Caretha L. Creasy, Peter J. Tummino, Benjamin Schwartz, Dashyant Dhanak, Ryan G. Kruger, Steven D. Knight, Sharad K. Verma, Roland Annan, Yuchen Bai, Yong Jiang, Edward Dul, Arthur Groy, Ashley M. Hughes, Wendy S. Halsey, Michael Huddleston, Melissa B. Pappalardi, Alan P. Graves, and Heidi M. Ott
- Abstract
Supplementary Table 2: Biochemical activity of WT and A687V EZH2 complexes with modified histone peptides
- Published
- 2023
6. Supplementary Table 1 from ALDH1A1 Is a Novel EZH2 Target Gene in Epithelial Ovarian Cancer Identified by Genome-Wide Approaches
- Author
-
Rugang Zhang, Michael J. Birrer, Paul Cairns, Peter J. Tummino, Caretha L. Creasy, Michael Slifker, Marie E. Maradeo, Vinod Vathipadiekal, Benjamin G. Bitler, and Hua Li
- Abstract
XLS file - 3-1MB
- Published
- 2023
7. Supplementary Figure 2 from A687V EZH2 Is a Driver of Histone H3 Lysine 27 (H3K27) Hypertrimethylation
- Author
-
Michael T. McCabe, Caretha L. Creasy, Peter J. Tummino, Benjamin Schwartz, Dashyant Dhanak, Ryan G. Kruger, Steven D. Knight, Sharad K. Verma, Roland Annan, Yuchen Bai, Yong Jiang, Edward Dul, Arthur Groy, Ashley M. Hughes, Wendy S. Halsey, Michael Huddleston, Melissa B. Pappalardi, Alan P. Graves, and Heidi M. Ott
- Abstract
Supplementary Figure 2: Alignment of all human SET domain methyltransferases
- Published
- 2023
8. Supplementary Figure 2 from ALDH1A1 Is a Novel EZH2 Target Gene in Epithelial Ovarian Cancer Identified by Genome-Wide Approaches
- Author
-
Rugang Zhang, Michael J. Birrer, Paul Cairns, Peter J. Tummino, Caretha L. Creasy, Michael Slifker, Marie E. Maradeo, Vinod Vathipadiekal, Benjamin G. Bitler, and Hua Li
- Abstract
PDF file - 138K
- Published
- 2023
9. Supplementary Table 1 from A687V EZH2 Is a Driver of Histone H3 Lysine 27 (H3K27) Hypertrimethylation
- Author
-
Michael T. McCabe, Caretha L. Creasy, Peter J. Tummino, Benjamin Schwartz, Dashyant Dhanak, Ryan G. Kruger, Steven D. Knight, Sharad K. Verma, Roland Annan, Yuchen Bai, Yong Jiang, Edward Dul, Arthur Groy, Ashley M. Hughes, Wendy S. Halsey, Michael Huddleston, Melissa B. Pappalardi, Alan P. Graves, and Heidi M. Ott
- Abstract
Supplementary Table 1: Primers utilized for A687V EZH2 mutagenesis
- Published
- 2023
10. Supplementary Table 3 from A687V EZH2 Is a Driver of Histone H3 Lysine 27 (H3K27) Hypertrimethylation
- Author
-
Michael T. McCabe, Caretha L. Creasy, Peter J. Tummino, Benjamin Schwartz, Dashyant Dhanak, Ryan G. Kruger, Steven D. Knight, Sharad K. Verma, Roland Annan, Yuchen Bai, Yong Jiang, Edward Dul, Arthur Groy, Ashley M. Hughes, Wendy S. Halsey, Michael Huddleston, Melissa B. Pappalardi, Alan P. Graves, and Heidi M. Ott
- Abstract
Supplementary Table 3: Functional analysis of genes significantly up- and down-regulated in SUP-B8 and NALM-6 cells.
- Published
- 2023
11. Supplementary Figure 4 from A687V EZH2 Is a Driver of Histone H3 Lysine 27 (H3K27) Hypertrimethylation
- Author
-
Michael T. McCabe, Caretha L. Creasy, Peter J. Tummino, Benjamin Schwartz, Dashyant Dhanak, Ryan G. Kruger, Steven D. Knight, Sharad K. Verma, Roland Annan, Yuchen Bai, Yong Jiang, Edward Dul, Arthur Groy, Ashley M. Hughes, Wendy S. Halsey, Michael Huddleston, Melissa B. Pappalardi, Alan P. Graves, and Heidi M. Ott
- Abstract
Supplementary Figure 4: Sanger sequencing chromatograms of EZH2 mutations in acute lymphoblastic leukemia cell lines
- Published
- 2023
12. BET inhibition silences expression of MYCN and BCL2 and induces cytotoxicity in neuroblastoma tumor models.
- Author
-
Anastasia Wyce, Gopinath Ganji, Kimberly N Smitheman, Chun-Wa Chung, Susan Korenchuk, Yuchen Bai, Olena Barbash, BaoChau Le, Peter D Craggs, Michael T McCabe, Karen M Kennedy-Wilson, Lydia V Sanchez, Romain L Gosmini, Nigel Parr, Charles F McHugh, Dashyant Dhanak, Rab K Prinjha, Kurt R Auger, and Peter J Tummino
- Subjects
Medicine ,Science - Abstract
BET family proteins are epigenetic regulators known to control expression of genes involved in cell growth and oncogenesis. Selective inhibitors of BET proteins exhibit potent anti-proliferative activity in a number of hematologic cancer models, in part through suppression of the MYC oncogene and downstream Myc-driven pathways. However, little is currently known about the activity of BET inhibitors in solid tumor models, and whether down-regulation of MYC family genes contributes to sensitivity. Here we provide evidence for potent BET inhibitor activity in neuroblastoma, a pediatric solid tumor associated with a high frequency of MYCN amplifications. We treated a panel of neuroblastoma cell lines with a novel small molecule inhibitor of BET proteins, GSK1324726A (I-BET726), and observed potent growth inhibition and cytotoxicity in most cell lines irrespective of MYCN copy number or expression level. Gene expression analyses in neuroblastoma cell lines suggest a role of BET inhibition in apoptosis, signaling, and N-Myc-driven pathways, including the direct suppression of BCL2 and MYCN. Reversal of MYCN or BCL2 suppression reduces the potency of I-BET726-induced cytotoxicity in a cell line-specific manner; however, neither factor fully accounts for I-BET726 sensitivity. Oral administration of I-BET726 to mouse xenograft models of human neuroblastoma results in tumor growth inhibition and down-regulation MYCN and BCL2 expression, suggesting a potential role for these genes in tumor growth. Taken together, our data highlight the potential of BET inhibitors as novel therapeutics for neuroblastoma, and suggest that sensitivity is driven by pleiotropic effects on cell growth and apoptotic pathways in a context-specific manner.
- Published
- 2013
- Full Text
- View/download PDF
13. A687V EZH2 Is a Driver of Histone H3 Lysine 27 (H3K27) Hypertrimethylation
- Author
-
Ryan G. Kruger, Yuchen Bai, Yong Jiang, Caretha L. Creasy, Heidi M. Ott, Melissa B. Pappalardi, Edward Dul, Dashyant Dhanak, Michael T. McCabe, Benjamin Schwartz, Roland S. Annan, Peter J. Tummino, Wendy S. Halsey, Arthur Groy, Alan P. Graves, Steven D. Knight, Ashley M. Hughes, Michael J. Huddleston, and Sharad K. Verma
- Subjects
Models, Molecular ,Transcriptional Activation ,Heterozygote ,Cancer Research ,Methyltransferase ,Protein Conformation ,Molecular Sequence Data ,Mutant ,Gene Expression ,macromolecular substances ,Biology ,medicine.disease_cause ,Methylation ,Substrate Specificity ,Histones ,Histone H3 ,Cell Line, Tumor ,Precursor B-Cell Lymphoblastic Leukemia-Lymphoma ,medicine ,Cluster Analysis ,Humans ,Enhancer of Zeste Homolog 2 Protein ,Amino Acid Sequence ,Gene Silencing ,Regulation of gene expression ,Mutation ,Binding Sites ,Gene Expression Profiling ,Lysine ,EZH2 ,Polycomb Repressive Complex 2 ,Molecular biology ,Amino Acid Substitution ,Oncology ,Biochemistry ,Cell culture ,Sequence Alignment - Abstract
The EZH2 methyltransferase silences gene expression through methylation of histone H3 on lysine 27 (H3K27). Recently, EZH2 mutations have been reported at Y641, A677, and A687 in non-Hodgkin lymphoma. Although the Y641F/N/S/H/C and A677G mutations exhibit clearly increased activity with substrates dimethylated at lysine 27 (H3K27me2), the A687V mutant has been shown to prefer a monomethylated lysine 27 (H3K27me1) with little gain of activity toward H3K27me2. Herein, we demonstrate that despite this unique substrate preference, A687V EZH2 still drives increased H3K27me3 when transiently expressed in cells. However, unlike the previously described mutants that dramatically deplete global H3K27me2 levels, A687V EZH2 retains normal levels of H3K27me2. Sequencing of B-cell–derived cancer cell lines identified an acute lymphoblastic leukemia cell line harboring this mutation. Similar to exogenous expression of A687V EZH2, this cell line exhibited elevated H3K27me3 while possessing H3K27me2 levels higher than Y641- or A677-mutant lines. Treatment of A687V EZH2-mutant cells with GSK126, a selective EZH2 inhibitor, was associated with a global decrease in H3K27me3, robust gene activation, caspase activation, and decreased proliferation. Structural modeling of the A687V EZH2 active site suggests that the increased catalytic activity with H3K27me1 may be due to a weakened interaction with an active site water molecule that must be displaced for dimethylation to occur. These findings suggest that A687V EZH2 likely increases global H3K27me3 indirectly through increased catalytic activity with H3K27me1 and cells harboring this mutation are highly dependent on EZH2 activity for their survival. Mol Cancer Ther; 13(12); 3062–73. ©2014 AACR.
- Published
- 2014
14. Small-Molecule Targets in Immuno-Oncology
- Author
-
James P. Edwards, Dashyant Dhanak, Ancho Nguyen, and Peter J. Tummino
- Subjects
0301 basic medicine ,Combination therapy ,medicine.medical_treatment ,Clinical Biochemistry ,Programmed Cell Death 1 Receptor ,Biology ,Lymphocyte Activation ,Biochemistry ,Small Molecule Libraries ,03 medical and health sciences ,Cancer immunotherapy ,Neoplasms ,Drug Discovery ,medicine ,Humans ,Immunologic Factors ,CTLA-4 Antigen ,Molecular Biology ,Pharmacology ,Toll-Like Receptors ,Cancer ,Membrane Proteins ,Immunotherapy ,Nuclear Receptor Subfamily 1, Group F, Member 3 ,medicine.disease ,Acquired immune system ,Small molecule ,Immune checkpoint ,Blockade ,030104 developmental biology ,Immunology ,Cancer research ,Molecular Medicine - Abstract
Advances in understanding the role and molecular mechanisms underlying immune surveillance and control of (pre)malignancies is revolutionizing clinical practice in the treatment of cancer. Presently, multiple biologic drugs targeting the immune checkpoint proteins PD(L)1 or CTLA4 have been approved and/or are in advanced stages of clinical development for many cancers. In addition, combination therapy with these agents and other immunomodulators is being intensively explored with the aim of improving primary response rates or prolonging overall survival. The effectiveness of cancer immunotherapy with biologics is spurring research in alternate approaches including small-molecule-mediated targeting of intracellular pathways modulating the innate and adaptive immune response. This focus of this review is on some of the key intracellular pathways where the development of a small-molecule therapeutic is attractive, tractable, and potentially synergistic with extracellular biologic-mediated immune checkpoint blockade.
- Published
- 2017
15. Cancer epigenetics drug discovery and development: the challenge of hitting the mark
- Author
-
Robert M. Campbell and Peter J. Tummino
- Subjects
Antineoplastic Agents ,Biology ,Bioinformatics ,Epigenesis, Genetic ,Neoplasms ,Drug Discovery ,medicine ,Animals ,Humans ,Point Mutation ,Enhancer of Zeste Homolog 2 Protein ,Molecular Targeted Therapy ,Epigenetics ,Cancer epigenetics ,Vorinostat ,Epigenomics ,Clinical Trials as Topic ,Review Series ,Polycomb Repressive Complex 2 ,Cancer ,Histone-Lysine N-Methyltransferase ,Methyltransferases ,General Medicine ,Epigenome ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Histone ,DNA methylation ,biology.protein ,Drug Screening Assays, Antitumor ,Corrigendum ,medicine.drug - Abstract
Over the past several years, there has been rapidly expanding evidence of epigenetic dysregulation in cancer, in which histone and DNA modification play a critical role in tumor growth and survival. These findings have gained the attention of the drug discovery and development community, and offer the potential for a second generation of cancer epigenetic agents for patients following the approved “first generation” of DNA methylation (e.g., Dacogen, Vidaza) and broad-spectrum HDAC inhibitors (e.g., Vorinostat, Romidepsin). This Review provides an analysis of prospects for discovery and development of novel cancer agents that target epigenetic proteins. We will examine key examples of epigenetic dysregulation in tumors as well as challenges to epigenetic drug discovery with emerging biology and novel classes of drug targets. We will also highlight recent successes in cancer epigenetics drug discovery and consider important factors for clinical success in this burgeoning area. Epigenetic dysregulation in cancer Epigenetic information is contained in the cell in multiple forms that include DNA methylation, histone modification (methylation, acetylation, phosphorylation, etc.), nucleosome positioning, and microRNA expression, among others. This combined information constitutes the epigenome. A comprehensive understanding of epigenomic dysregulation in specific cancer types has not been elucidated yet. Currently, there is an understanding of tumor-specific types of epigenetic modifications without a full appreciation of the context of the entire cancer epigenome in the specific tumor. Cancer epigenetic dysregulation can be categorized into three types: (a) altered DNA or histone modification, (b) somatic alteration in an epigenetic protein, and (c) altered expression of an epigenetic protein. Those types of cancer epigenome dysregulation have been reviewed comprehensively elsewhere (1–3), and only will be referred to here.
- Published
- 2014
16. Inhibition of BET bromodomain proteins as a therapeutic approach in prostate cancer
- Author
-
BaoChau Le, Robert L. Vessella, Anastasia Wyce, Peter J. Tummino, Caretha L. Creasy, Susan Korenchuk, Yuchen Bai, Yan Degenhardt, Olena Barbash, Ming-Chih Crouthamel, and Charles F. McHugh
- Subjects
Male ,BRD4 ,Down-Regulation ,chemical and pharmacologic phenomena ,Apoptosis ,Cell Growth Processes ,Mice, SCID ,Biology ,Protein Serine-Threonine Kinases ,BET inhibitor ,Benzodiazepines ,Mice ,Cell Line, Tumor ,Gene expression ,bromodomain ,Animals ,Humans ,Oncogene ,Cell growth ,Gene Expression Profiling ,Nuclear Proteins ,hemic and immune systems ,BET ,prostate cancer ,Xenograft Model Antitumor Assays ,Chromatin ,Bromodomain ,Prostatic Neoplasms, Castration-Resistant ,Histone ,c-Myc ,Oncology ,Cancer research ,biology.protein ,Brd4 ,Research Paper - Abstract
BET (bromodomain and extra-terminal) proteins regulate gene expression through their ability to bind to acetylated chromatin and subsequently activate RNA PolII-driven transcriptional elongation. Small molecule BET inhibitors prevent binding of BET proteins to acetylated histones and inhibit transcriptional activation of BET target genes. BET inhibitors attenuate cell growth and survival in several hematologic cancer models, partially through the down-regulation of the critical oncogene, MYC. We hypothesized that BET inhibitors will regulate MYC expression in solid tumors that frequently over-express MYC. Here we describe the effects of the highly specific BET inhibitor, I-BET762, on MYC expression in prostate cancer models. I-BET762 potently reduced MYC expression in prostate cancer cell lines and a patient-derived tumor model with subsequent inhibition of cell growth and reduction of tumor burden in vivo. Our data suggests that I-BET762 effects are partially driven by MYC down-regulation and underlines the critical importance of additional mechanisms of I-BET762 induced phenotypes.
- Published
- 2013
17. Identification of Potent, Selective, Cell-Active Inhibitors of the Histone Lysine Methyltransferase EZH2
- Author
-
Christine Thompson, Daryl A. Scherzer, Ryan G. Kruger, Steven D. Knight, Louis V. LaFrance, Heidi M. Ott, Dashyant Dhanak, Stuart Paul Romeril, Celine Duquenne, Dominic Suarez, William H. Miller, Art Shu, Seth W. Grant, Carl A. Machutta, Joelle Lorraine Burgess, Brackley James, Caretha L. Creasy, Xinrong Tian, Yong Jiang, Sharad K. Verma, Peter J. Tummino, Alan P. Graves, Glenn S. Van Aller, Elsie Diaz, Kenneth A. Newlander, Johnson Neil W, and Michael T. McCabe
- Subjects
Genetics ,Methyltransferase ,Drug discovery ,Organic Chemistry ,EZH2 ,macromolecular substances ,Biology ,Biochemistry ,Small molecule ,Histone ,Histone methyltransferase ,Drug Discovery ,biology.protein ,Epigenetics ,PRC2 - Abstract
The histone H3-lysine 27 (H3K27) methyltransferase EZH2 plays a critical role in regulating gene expression, and its aberrant activity is linked to the onset and progression of cancer. As part of a drug discovery program targeting EZH2, we have identified highly potent, selective, SAM-competitive, and cell-active EZH2 inhibitors, including GSK926 (3) and GSK343 (6). These compounds are small molecule chemical tools that would be useful to further explore the biology of EZH2.
- Published
- 2012
18. EZH2 inhibition as a therapeutic strategy for lymphoma with EZH2-activating mutations
- Author
-
Ryan G. Kruger, William H. Miller, Louis V. LaFrance, Celine Duquenne, Alan P. Graves, Heidi M. Ott, Dashyant Dhanak, Christine Thompson, Xinrong Tian, Martin Brandt, Michael T. McCabe, Charles F. McHugh, Mellinger Mark, Susan Korenchuk, Anthony Della Pietra, Peter J. Tummino, Yan Liu, Caretha L. Creasy, Sharad K. Verma, Elsie Diaz, Gopinath Ganji, and Glenn S. Van Aller
- Subjects
Regulation of gene expression ,Multidisciplinary ,Methyltransferase ,biology ,EZH2 ,Mutant ,macromolecular substances ,medicine.disease_cause ,Histone H3 ,Histone methyltransferase ,medicine ,biology.protein ,Cancer research ,Carcinogenesis ,PRC2 - Abstract
EZH2, the catalytic subunit of the polycomb repressive complex 2 (PRC2), is involved in repressing gene expression through methylation of histone H3 on lysine 27 (H3K27). Overexpression of EZH2 is implicated in tumorigenesis, and mutations within its catalytic domain occur in lymphoma. Here, Caretha Creasy and colleagues describe a potent small-molecule inhibitor of EZH2 methyltransferase activity that decreases levels of methylated H3K27 and reactivates silenced PRC2 target genes. It also inhibits the proliferation of EZH2 mutant cell lines and the growth of EZH2 mutant xenografts in mice. Pharmacological inhibition of EZH2 activity may therefore be a viable strategy for treating EZH2 mutant lymphoma.
- Published
- 2012
19. Discovery and Characterization of a Cell-Permeable, Small-Molecule c-Abl Kinase Activator that Binds to the Myristoyl Binding Site
- Author
-
Hong Zhang, Josh Cottom, Nino Campobasso, Graham L. Simpson, Connie L. Erickson-Miller, Thau F. Ho, George Burton, Mangatt P. Biju, Sheri L. Moores, Hu Li, Paris Ward, Brett Siegfried, Xuan Hong, Ping Cao, Junya Qu, Da-Yuan Wang, Martha S. Head, Robert A. Copeland, Yoshiaki Washio, Francesca Zappacosta, Xiao-Qing Pan, Allen Oliff, Jingsong Yang, Kelly E. Fisher, Kyung O. Johanson, Sarah E. Galbraith, Sophie M. Bertrand, Peter J. Tummino, Glenn A. Hofmann, and Zhihong Lai
- Subjects
Models, Molecular ,Stereochemistry ,Molecular Sequence Data ,Clinical Biochemistry ,Allosteric regulation ,Crystallography, X-Ray ,SH2 domain ,Biochemistry ,Permeability ,hemic and lymphatic diseases ,Drug Discovery ,Humans ,Amino Acid Sequence ,Phosphorylation ,Kinase activity ,Binding site ,Proto-Oncogene Proteins c-abl ,Molecular Biology ,Myristoylation ,Pharmacology ,Binding Sites ,Chemistry ,Hydantoins ,Hep G2 Cells ,General Medicine ,Proto-Oncogene Proteins c-crk ,Small molecule ,Protein Structure, Tertiary ,Enzyme Activation ,Protein kinase domain ,Docking (molecular) ,Pyrazoles ,Molecular Medicine ,lipids (amino acids, peptides, and proteins) ,Protein Binding - Abstract
Summaryc-Abl kinase activity is regulated by a unique mechanism involving the formation of an autoinhibited conformation in which the N-terminal myristoyl group binds intramolecularly to the myristoyl binding site on the kinase domain and induces the bending of the αI helix that creates a docking surface for the SH2 domain. Here, we report a small-molecule c-Abl activator, DPH, that displays potent enzymatic and cellular activity in stimulating c-Abl activation. Structural analyses indicate that DPH binds to the myristoyl binding site and prevents the formation of the bent conformation of the αI helix through steric hindrance, a mode of action distinct from the previously identified allosteric c-Abl inhibitor, GNF-2, that also binds to the myristoyl binding site. DPH represents the first cell-permeable, small-molecule tool compound for c-Abl activation.
- Published
- 2011
20. Myelosuppression and kinase selectivity of multikinase angiogenesis inhibitors
- Author
-
King Ag, Richard R. Gontarek, Michelle Crouthamel, David H. Rominger, Peter J. Tummino, Rakesh Kumar, and Levin Ra
- Subjects
Sorafenib ,Cancer Research ,Sunitinib ,Angiogenesis ,Kinase ,Growth factor ,medicine.medical_treatment ,Autophosphorylation ,Biology ,urologic and male genital diseases ,female genital diseases and pregnancy complications ,Pazopanib ,Oncology ,Fms-Like Tyrosine Kinase 3 ,medicine ,Cancer research ,medicine.drug - Abstract
Myelosuppression has been observed with several multikinase angiogenesis inhibitors in clinical studies, although the frequency and severity varies among the different agents. Inhibitors targeting vascular endothelial growth factor receptor (VEGFR) often inhibit other kinases, which may contribute to their adverse-event profiles. Kinase selectivity of pazopanib, sorafenib, and sunitinib was evaluated in a panel of 242 kinases. Cellular potency was measured using autophosphorylation assays. Effect on human bone marrow progenitor growth in the presence of multiple growth factors was evaluated and correlated with the kinase selectivity. Sunitinib inhibited more kinases than pazopanib and sorafenib, at potencies within 10-fold of VEGFR-2. All three compounds potently inhibited VEGFR-2, platelet-derived growth factor receptor-β and c-Kit, However, pazopanib was less active against Flt-3 in both kinase and cellular assays. The inhibitory properties of pazopanib, sorafenib, and sunitinib were dependent on the growth factor used to initiate bone marrow colony formation. Addition of stem cell factor and/or Flt-3 ligand with granulocyte-macrophage colony stimulating factor resulted in significant shifts in potency for sorafenib and sunitinib but less so for pazopanib. Activity against c-kit and Flt-3 by multikinase angiogenesis inhibitors provide a potential explanation for the differences in myelosuppression observed with these agents in patients.
- Published
- 2009
21. Characterization of an exosite binding inhibitor of matrix metalloproteinase 13
- Author
-
Gregg B. Fields, Frank T. Coppo, Lusong Luo, Lata T. Gooljarsingh, Ami S. Lakdawala, Peter J. Tummino, and Richard R. Gontarek
- Subjects
Models, Molecular ,Pyrimidine ,Protein Conformation ,Stereochemistry ,Matrix metalloproteinase inhibitor ,Matrix Metalloproteinase Inhibitors ,Matrix metalloproteinase ,Biochemistry ,Article ,chemistry.chemical_compound ,Protein structure ,Matrix Metalloproteinase 13 ,medicine ,Protease Inhibitors ,Binding site ,Molecular Biology ,chemistry.chemical_classification ,Enzyme Precursors ,Binding Sites ,biology ,Chemistry ,Active site ,Tissue Inhibitor of Metalloproteinases ,Hydrogen-Ion Concentration ,Kinetics ,Enzyme ,biology.protein ,Marimastat ,medicine.drug - Abstract
Matrix metalloproteinase 13 (MMP13) is a key enzyme implicated in the degradation of the extracellular matrix in osteoarthritis. Clinical administration of broad spectrum MMP inhibitors such as marimastat has been implicated in severe musculo-skeletal side effects. Consequently, research has been focused on designing inhibitors that selectively inhibit MMP13, thereby circumventing musculo-skeletal toxicities. A series of pyrimidine dicarboxamides were recently shown to be highly selective inhibitors of MMP13 with a novel binding mode. We have applied a molecular ruler to this exosite by dual inhibition studies involving a potent dicarboxamide in the presence of two metal chelators of different sizes. A larger hydroxamate mimic overlaps and antagonizes binding of the dicarboxamide to the exosite whereas the much smaller acetohydroxamate synergizes with the dicarboxamide. These studies elucidate the steric requirement for compounds that fit exclusively into the active site, a mandate for generating highly selective MMP13 inhibitors.
- Published
- 2007
22. A DNA Hypomethylation Signature Predicts Antitumor Activity of LSD1 Inhibitors in SCLC
- Author
-
Thau F. Ho, Yan Liu, Xinrong Tian, Patrick McDevitt, Michael Butticello, Timothy K. Hart, Kimberly N. Smitheman, David Soong, Jessica L. Schneck, Peter J. Tummino, Christine L. Hann, Glenn S. Van Aller, Yuchen Bai, Nestor O. Concha, Kasparec Jiri, Melissa B. Pappalardi, Christopher L. Carpenter, Mcnulty Kenneth C, Kelly Federowicz, Jeffrey D. Carson, William H. Miller, Johnson Neil W, Chandrashekhar D. Kamat, Rouse Meagan B, Charles F. McHugh, Michael T. McCabe, Ryan G. Kruger, Dashyant Dhanak, Helai P. Mohammad, Michelle Crouthamel, Yan Degenhardt, William G. Bonnette, and Shelby A. Gorman
- Subjects
Cyclopropanes ,Cancer Research ,animal structures ,Lung Neoplasms ,Molecular Sequence Data ,Administration, Oral ,Antineoplastic Agents ,Benzoates ,Article ,Epigenesis, Genetic ,chemistry.chemical_compound ,Mice ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Epigenetics ,Enzyme Inhibitors ,Cell Proliferation ,Histone Demethylases ,biology ,Cancer ,KDM1A ,Biological activity ,Cell Biology ,DNA Methylation ,medicine.disease ,Small Cell Lung Carcinoma ,Xenograft Model Antitumor Assays ,Gene Expression Regulation, Neoplastic ,chemistry ,Biochemistry ,Oncology ,Cell culture ,biology.protein ,Cancer research ,Demethylase ,Growth inhibition ,DNA hypomethylation - Abstract
SummaryEpigenetic dysregulation has emerged as an important mechanism in cancer. Alterations in epigenetic machinery have become a major focus for targeted therapies. The current report describes the discovery and biological activity of a cyclopropylamine containing inhibitor of Lysine Demethylase 1 (LSD1), GSK2879552. This small molecule is a potent, selective, orally bioavailable, mechanism-based irreversible inactivator of LSD1. A proliferation screen of cell lines representing a number of tumor types indicated that small cell lung carcinoma (SCLC) is sensitive to LSD1 inhibition. The subset of SCLC lines and primary samples that undergo growth inhibition in response to GSK2879552 exhibit DNA hypomethylation of a signature set of probes, suggesting this may be used as a predictive biomarker of activity.
- Published
- 2015
23. A Second p53 Binding Site in the Central Domain of Mdm2 Is Essential for p53 Ubiquitination
- Author
-
Kurt R. Auger, Zhihong Lai, Michael Grooms, Thau F. Ho, Kyung O. Johanson, Robert A. Copeland, Peter J. Tummino, Robert B. Kirkpatrick, John D. Martin, Hong Zhang, and Jianhong Ma
- Subjects
HMG-box ,Ubiquitin-Protein Ligases ,Spodoptera ,Biochemistry ,Piperazines ,Cell Line ,Domain (software engineering) ,Transactivation ,Ubiquitin ,Tetramer ,Protein Interaction Mapping ,Genetics ,Animals ,Humans ,Molecular Biology ,neoplasms ,Sequence Deletion ,Zinc finger ,Binding Sites ,biology ,Chemistry ,Imidazoles ,DHR1 domain ,Proto-Oncogene Proteins c-mdm2 ,Ubiquitin ligase ,Cell biology ,Cysteine Endopeptidases ,Kinetics ,enzymes and coenzymes (carbohydrates) ,biology.protein ,Mdm2 ,Tumor Suppressor Protein p53 ,Biotechnology ,Binding domain ,P53 binding - Abstract
Mdm2 negatively regulates p53 by inhibiting its transcriptional activity and promoting its degradation by functioning as an E3 ubiquitin ligase. The primary p53 binding site on mdm2 is located in its N-terminal domain. Through binding to p53 at its N-terminal transactivation domain, mdm2 directly blocks the transcriptional activation function of p53. We discovered that truncated mdm2 protein constructs without the N-terminal p53 binding domain are at least as active as full-length mdm2 in catalyzing p53 ubiquitination. Furthermore, the deletion of the central acidic domain significantly reduces the E3 ligase activity of mdm2 toward p53. We have also performed GST pull-down experiments to probe the direct binding of various mdm2 domain constructs toward full length p53 and found that mdm2 constructs without the N-terminal p53 binding domain retain the ability to bind to p53. Our kinetic and binding data localize the second p53 binding site between amino acids 211 and 361, including the acidic domain and the zinc finger region. Our work, consistent with other reports, suggests that the p53 tetramer interacts with at least two sites on mdm2. Although the interaction between the N-termini of mdm2 and p53 blocks the transactivation activity of p53, the interaction between the central domain of mdm2 and the core domain of p53 is critical for the ubiquitination and degradation of p53. This second mdm2-p53 interaction site represents an alternative target for small molecule modulators of the mdm2-p53 pathway.
- Published
- 2006
24. A biochemical rationale for the anticancer effects of Hsp90 inhibitors: Slow, tight binding inhibition by geldanamycin and its analogues
- Author
-
Robert A. Copeland, Zhihong Lai, Tia Lewis, Kang Yan, Peter J. Tummino, Lata T. Gooljarsingh, John J. Kerrigan, Robert H. Sinnamon, Kyung O. Johanson, Christine Fernandes, Marc R. Arnone, Michael Grooms, Hong Zhang, Alastair J. King, and Robert B. Kirkpatrick
- Subjects
Boron Compounds ,Lactams, Macrocyclic ,Context (language use) ,Plasma protein binding ,Hsp90 inhibitor ,chemistry.chemical_compound ,In vivo ,Heat shock protein ,polycyclic compounds ,Benzoquinones ,Animals ,Humans ,Potency ,HSP90 Heat-Shock Proteins ,Cells, Cultured ,Fluorescent Dyes ,Antibiotics, Antineoplastic ,Multidisciplinary ,biology ,Quinones ,Biological Sciences ,Geldanamycin ,Hsp90 ,Recombinant Proteins ,Biochemistry ,chemistry ,biology.protein ,Protein Binding - Abstract
Heat shock protein (Hsp)90 is emerging as an important therapeutic target for the treatment of cancer. Two analogues of the Hsp90 inhibitor geldanamycin are currently in clinical trials. Geldanamycin (GA) and its analogues have been reported to bind purified Hsp90 with low micromolar potency, in stark contrast to their low nanomolar antiproliferative activity in cell culture and their potent antitumor activity in animal models. Several models have been proposed to account for the ≈100-fold-greater potency in cell culture, including that GA analogues bind with greater affinity to a five-protein Hsp90 complex than to Hsp90 alone. We have determined that GA and the fluorescent analogue BODIPY-GA (BDGA) both demonstrate slow, tight binding to purified Hsp90. BDGA, used to characterize the kinetics of ligand–Hsp90 interactions, was found to bind Hsp90α with k off = 2.5 × 10 −3 min −1 , t 1/2 = 4.6 h, and K i * = 10 nM. It was found that BDGA binds to a functional multiprotein Hsp90 complex with kinetics and affinity identical to that of Hsp90 alone. Also, BDGA binds to Hsp90 from multiple cell lysates in a time-dependent manner with similar kinetics. Therefore, our results indicate that the high potency of GA in cell culture and in vivo can be accounted for by its time-dependent, tight binding to Hsp90 alone. In the broader context, these studies highlight the essentiality of detailed biochemical characterization of drug–target interactions for the effective translation of in vitro pharmacology to cellular and in vivo efficacy.
- Published
- 2006
25. Hydrolysis of Biological Peptides by Human Angiotensin-converting Enzyme-related Carboxypeptidase
- Author
-
Larry Dick, Virendar K. Kaushik, Thomas F. Parsons, Peter J. Tummino, Susan L. Acton, Michael A. Patane, Kevin Godbout, Jin Tang, Elizabeth Baronas, Chad S Vickers, Frank Y. Hsieh, James M. Gavin, Paul Hales, and Andrew J. Nichols
- Subjects
Stereochemistry ,Peptide ,Carboxypeptidases ,Peptidyl-Dipeptidase A ,Biochemistry ,Catalysis ,Humans ,Amino Acid Sequence ,Molecular Biology ,Peptide sequence ,Chromatography, High Pressure Liquid ,chemistry.chemical_classification ,biology ,Hydrolysis ,Angiotensin-converting enzyme ,Cell Biology ,Hydrogen-Ion Concentration ,Chromatography, Ion Exchange ,Carboxypeptidase ,Angiotensin II ,Recombinant Proteins ,Amino acid ,Kinetics ,Enzyme ,chemistry ,Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization ,Angiotensin-converting enzyme 2 ,biology.protein ,Spectrophotometry, Ultraviolet ,Angiotensin-Converting Enzyme 2 ,Peptides ,hormones, hormone substitutes, and hormone antagonists - Abstract
Human angiotensin-converting enzyme-related carboxypeptidase (ACE2) is a zinc metalloprotease whose closest homolog is angiotensin I-converting enzyme. To begin to elucidate the physiological role of ACE2, ACE2 was purified, and its catalytic activity was characterized. ACE2 proteolytic activity has a pH optimum of 6.5 and is enhanced by monovalent anions, which is consistent with the activity of ACE. ACE2 activity is increased approximately 10-fold by Cl(-) and F(-) but is unaffected by Br(-). ACE2 was screened for hydrolytic activity against a panel of 126 biological peptides, using liquid chromatography-mass spectrometry detection. Eleven of the peptides were hydrolyzed by ACE2, and in each case, the proteolytic activity resulted in removal of the C-terminal residue only. ACE2 hydrolyzes three of the peptides with high catalytic efficiency: angiotensin II () (k(cat)/K(m) = 1.9 x 10(6) m(-1) s(-1)), apelin-13 (k(cat)/K(m) = 2.1 x 10(6) m(-1) s(-1)), and dynorphin A 1-13 (k(cat)/K(m) = 3.1 x 10(6) m(-1) s(-1)). The ACE2 catalytic efficiency is 400-fold higher with angiotensin II () as a substrate than with angiotensin I (). ACE2 also efficiently hydrolyzes des-Arg(9)-bradykinin (k(cat)/K(m) = 1.3 x 10(5) m(-1) s(-1)), but it does not hydrolyze bradykinin. An alignment of the ACE2 peptide substrates reveals a consensus sequence of: Pro-X((1-3 residues))-Pro-Hydrophobic, where hydrolysis occurs between proline and the hydrophobic amino acid.
- Published
- 2002
26. Smyd3 regulates cancer cell phenotypes and catalyzes histone H4 lysine 5 methylation
- Author
-
Glenn S. Van Aller, Peter J. Tummino, BaoChau Le, Gloria Mas, Roland S. Annan, Ryan G. Kruger, Patrick McDevitt, Nicolas Reynoird, Olena Barbash, Michael J. Huddleston, Or Gozani, Julien Sage, Shichong Liu, Benjamin A. Garcia, Robert H. Sinnamon, Anne-Flore Zmoos, Institute for Advanced Biosciences / Institut pour l'Avancée des Biosciences (Grenoble) (IAB), and Etablissement français du sang - Auvergne-Rhône-Alpes (EFS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre Hospitalier Universitaire [Grenoble] (CHU)-Centre National de la Recherche Scientifique (CNRS)-Université Grenoble Alpes (UGA)
- Subjects
Cancer Research ,[SDV]Life Sciences [q-bio] ,Blotting, Western ,Biology ,Methylation ,Chromatin remodeling ,Substrate Specificity ,Histones ,Histone H4 ,Mice ,Histone H1 ,Peptide Library ,oncogene ,Smyd3 ,Histone methylation ,Histone H2A ,Animals ,Humans ,cancer ,Histone code ,RNA, Small Interfering ,Molecular Biology ,ComputingMilieux_MISCELLANEOUS ,Mice, Knockout ,lysine ,epigenetics ,Brief Report ,Genetic Complementation Test ,EZH2 ,Histone-Lysine N-Methyltransferase ,Fibroblasts ,Chromatin ,Recombinant Proteins ,3. Good health ,Enzyme Activation ,Gene Expression Regulation, Neoplastic ,Mice, Inbred C57BL ,Phenotype ,Biochemistry ,Histone methyltransferase ,oncology ,Mutagenesis, Site-Directed ,HeLa Cells ,Plasmids - Abstract
Smyd3 is a lysine methyltransferase implicated in chromatin and cancer regulation. Here we show that Smyd3 catalyzes histone H4 methylation at lysine 5 (H4K5me). This novel histone methylation mark is detected in diverse cell types and its formation is attenuated by depletion of Smyd3 protein. Further, Smyd3-driven cancer cell phenotypes require its enzymatic activity. Thus, Smyd3, via H4K5 methylation, provides a potential new link between chromatin dynamics and neoplastic disease.
- Published
- 2014
27. SMYD3 links lysine methylation of MAP3K2 to Ras-driven cancer
- Author
-
Michiel Vermeulen, Nicolas Reynoird, Julien Sage, Olena Barbash, Or Gozani, Atul J. Butte, Shichong Liu, Michael J. Huddleston, Ryan G. Kruger, Dashyant Dhanak, Purvesh Khatri, Alex W. Wilkinson, Pascal W. T. C. Jansen, Benjamin A. Garcia, Pawel K. Mazur, Peter J. Tummino, Glenn S. Van Aller, Institute for Advanced Biosciences / Institut pour l'Avancée des Biosciences (Grenoble) (IAB), and Etablissement français du sang - Auvergne-Rhône-Alpes (EFS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre Hospitalier Universitaire [Grenoble] (CHU)-Centre National de la Recherche Scientifique (CNRS)-Université Grenoble Alpes (UGA)
- Subjects
MAPK/ERK pathway ,Methyltransferase ,Lung Neoplasms ,[SDV]Life Sciences [q-bio] ,Adenocarcinoma of Lung ,MAP3K2 ,Biology ,Adenocarcinoma ,MAP Kinase Kinase Kinase 2 ,Oncogene Protein p21(ras) ,Methylation ,Proto-Oncogene Proteins A-raf ,Mice ,Cell Line, Tumor ,Animals ,Humans ,Protein Phosphatase 2 ,ComputingMilieux_MISCELLANEOUS ,Multidisciplinary ,MAP kinase kinase kinase ,Proteomics and Chromatin Biology ,MEK inhibitor ,Lysine ,Protein phosphatase 2 ,Histone-Lysine N-Methyltransferase ,MAP Kinase Kinase Kinases ,Research Highlight ,3. Good health ,Pancreatic Neoplasms ,Disease Models, Animal ,Cell Transformation, Neoplastic ,Cancer research ,Mitogen-Activated Protein Kinases ,Signal Transduction - Abstract
Deregulation of lysine methylation signalling has emerged as a common aetiological factor in cancer pathogenesis, with inhibitors of several histone lysine methyltransferases (KMTs) being developed as chemotherapeutics. The largely cytoplasmic KMT SMYD3 (SET and MYND domain containing protein 3) is overexpressed in numerous human tumours. However, the molecular mechanism by which SMYD3 regulates cancer pathways and its relationship to tumorigenesis in vivo are largely unknown. Here we show that methylation of MAP3K2 by SMYD3 increases MAP kinase signalling and promotes the formation of Ras-driven carcinomas. Using mouse models for pancreatic ductal adenocarcinoma and lung adenocarcinoma, we found that abrogating SMYD3 catalytic activity inhibits tumour development in response to oncogenic Ras. We used protein array technology to identify the MAP3K2 kinase as a target of SMYD3. In cancer cell lines, SMYD3-mediated methylation of MAP3K2 at lysine 260 potentiates activation of the Ras/Raf/MEK/ERK signalling module and SMYD3 depletion synergizes with a MEK inhibitor to block Ras-driven tumorigenesis. Finally, the PP2A phosphatase complex, a key negative regulator of the MAP kinase pathway, binds to MAP3K2 and this interaction is blocked by methylation. Together, our results elucidate a new role for lysine methylation in integrating cytoplasmic kinase-signalling cascades and establish a pivotal role for SMYD3 in the regulation of oncogenic Ras signalling.
- Published
- 2014
28. Potent antimyeloma activity of the novel bromodomain inhibitors I-BET151 and I-BET762
- Author
-
Anna K. Bassil, Binbin Liu, Valentina S. Caputo, Nigel J. Parr, Rab K. Prinjha, BaoChau Le, Trevor D. Chapman, Irene Roberts, Nicola Harker, Jason Witherington, Leanne Cutler, Niam Al-Mahdi, Anastasios Karadimitris, Antonia Rotolo, Nicholas Smithers, Ilaria Marigo, David F. Tough, Amin Rahemtulla, Maurits F. Kleijnen, Katerina Gouvedenou, Aristeidis Chaidos, Peter J. Tummino, Mohammed Suhail Chaudhry, Olena Barbash, and Andrea C. Haynes
- Subjects
Transcriptional Activation ,Immunology ,Down-Regulation ,Antineoplastic Agents ,Apoptosis ,Biology ,Animals ,Benzodiazepines ,Cell Cycle Checkpoints ,Heterocyclic Compounds, 4 or More Rings ,Humans ,Mice ,Multiple Myeloma ,Proto-Oncogene Proteins c-myc ,RNA-Binding Proteins ,Transcription Factors ,Tumor Cells, Cultured ,Biochemistry ,Downregulation and upregulation ,In vivo ,Heterocyclic Compounds ,medicine ,Multiple myeloma ,Cultured ,Cell Biology ,Hematology ,4 or More Rings ,medicine.disease ,In vitro ,Chromatin ,Bromodomain ,Tumor Cells ,Cancer research ,Cyclin-dependent kinase 9 - Abstract
The bromodomain and extraterminal (BET) protein BRD2-4 inhibitors hold therapeutic promise in preclinical models of hematologic malignancies. However, translation of these data to molecules suitable for clinical development has yet to be accomplished. Herein we expand the mechanistic understanding of BET inhibitors in multiple myeloma by using the chemical probe molecule I-BET151. I-BET151 induces apoptosis and exerts strong antiproliferative effect in vitro and in vivo. This is associated with contrasting effects on oncogenic MYC and HEXIM1, an inhibitor of the transcriptional activator P-TEFb. I-BET151 causes transcriptional repression of MYC and MYC-dependent programs by abrogating recruitment to the chromatin of the P-TEFb component CDK9 in a BRD2-4-dependent manner. In contrast, transcriptional upregulation of HEXIM1 is BRD2-4 independent. Finally, preclinical studies show that I-BET762 has a favorable pharmacologic profile as an oral agent and that it inhibits myeloma cell proliferation, resulting in survival advantage in a systemic myeloma xenograft model. These data provide a strong rationale for extending the clinical testing of the novel antimyeloma agent I-BET762 and reveal insights into biologic pathways required for myeloma cell proliferation.
- Published
- 2013
29. Helicobacter pylori Physiology Predicted from Genomic Comparison of Two Strains
- Author
-
Peter Doig, Trevor J. Trust, Gilles Carmel, Peter J. Tummino, Boudewijn L. M. de Jonge, Scott D. Mills, Brian Noonan, Eric D. Brown, Maria Uria-Nickelsen, Braydon C. Guild, Richard A. Alm, Gerald F. Vovis, and Donald T. Moir
- Subjects
Genetics ,Phase variation ,Helicobacter pylori ,biology ,Sulfur metabolism ,Physiology ,Metabolism ,biology.organism_classification ,Microbiology ,Article ,Helicobacter Infections ,Transformation (genetics) ,Infectious Diseases ,Bacterial Proteins ,Genes, Bacterial ,Humans ,Energy Metabolism ,Bacterial outer membrane ,Molecular Biology ,Genome, Bacterial ,Bacteria ,Genomic organization - Abstract
SUMMARY Helicobacter pylori is a gram-negative bacteria which colonizes the gastric mucosa of humans and is implicated in a wide range of gastroduodenal diseases. This paper reviews the physiology of this bacterium as predicted from the sequenced genomes of two unrelated strains and reconciles these predictions with the literature. In general, the predicted capabilities are in good agreement with reported experimental observations. H. pylori is limited in carbohydrate utilization and will use amino acids, for which it has transporter systems, as sources of carbon. Energy can be generated by fermentation, and the bacterium possesses components necessary for both aerobic and anaerobic respiration. Sulfur metabolism is limited, whereas nitrogen metabolism is extensive. There is active uptake of DNA via transformation and ample restriction-modification activities. The cell contains numerous outer membrane proteins, some of which are porins or involved in iron uptake. Some of these outer membrane proteins and the lipopolysaccharide may be regulated by a slipped-strand repair mechanism which probably results in phase variation and plays a role in colonization. In contrast to a commonly held belief that H. pylori is a very diverse species, few differences were predicted in the physiology of these two unrelated strains, indicating that host and environmental factors probably play a significant role in the outocme of H. pylori-related disease.
- Published
- 1999
30. Genomic-sequence comparison of two unrelated isolates of the human gastric pathogen Helicobacter pylori
- Author
-
Brian Noonan, Trevor J. Trust, Qin Jiang, Gerald F. Vovis, Cameron Ives, Peter Doig, Donald T. Moir, Maria Uria-Nickelsen, Rene Gibson, Douglas R. Smith, Scott D. Mills, Diane E. Taylor, Benjamin L. King, Peter J. Tummino, Debra M. Mills, Braydon C. Guild, Anthony Caruso, Boudewijn L. deJonge, Lo-See L. Ling, Gilles Carmel, Alm Richard A, David Merberg, and Eric D. Brown
- Subjects
Genetics ,Multidisciplinary ,Helicobacter pylori ,Sequence analysis ,Molecular Sequence Data ,Gene Expression Regulation, Bacterial ,Sequence Analysis, DNA ,Biology ,biology.organism_classification ,Genome ,Helicobacter Infections ,Hypervariable region ,Microbiology ,medicine.anatomical_structure ,Species Specificity ,Duodenal Ulcer ,Gastric mucosa ,medicine ,Humans ,Sequence Alignment ,Pathogen ,Gene ,Genome, Bacterial ,Genomic organization - Abstract
Helicobacter pylori, one of the most common bacterial pathogens of humans, colonizes the gastric mucosa, where it appears to persist throughout the host's life unless the patient is treated. Colonization induces chronic gastric inflammation which can progress to a variety of diseases, ranging in severity from superficial gastritis and peptic ulcer to gastric cancer and mucosal-associated lymphoma. Strain-specific genetic diversity has been proposed to be involved in the organism's ability to cause different diseases or even be beneficial to the infected host and to participate in the lifelong chronicity of infection. Here we compare the complete genomic sequences of two unrelated H. pylori isolates. This is, to our knowledge, the first such genomic comparison. H. pylori was believed to exhibit a large degree of genomic and allelic diversity, but we find that the overall genomic organization, gene order and predicted proteomes (sets of proteins encoded by the genomes) of the two strains are quite similar. Between 6 to 7% of the genes are specific to each strain, with almost half of these genes being clustered in a single hypervariable region.
- Published
- 1999
31. BET inhibition silences expression of MYCN and BCL2 and induces cytotoxicity in neuroblastoma tumor models
- Author
-
Dashyant Dhanak, Romain Luc Marie Gosmini, Olena Barbash, Rab K. Prinjha, BaoChau Le, Gopinath Ganji, Kimberly N. Smitheman, Chun-wa Chung, Karen M. Kennedy-Wilson, Nigel J. Parr, Peter D. Craggs, Charles F. McHugh, Lydia V. Sanchez, Peter J. Tummino, Yuchen Bai, Anastasia Wyce, Susan Korenchuk, Kurt R. Auger, and Michael T. McCabe
- Subjects
Models, Molecular ,Molecular Conformation ,Apoptosis ,Cell Cycle Proteins ,medicine.disease_cause ,Benzodiazepines ,Mice ,Neuroblastoma ,Cluster Analysis ,Gene Regulatory Networks ,Oncogene Proteins ,N-Myc Proto-Oncogene Protein ,Multidisciplinary ,Nuclear Proteins ,RNA-Binding Proteins ,Tumor Burden ,Gene Expression Regulation, Neoplastic ,Proto-Oncogene Proteins c-bcl-2 ,Medicine ,Female ,Signal transduction ,Protein Binding ,Signal Transduction ,Research Article ,Science ,Antineoplastic Agents ,Protein Serine-Threonine Kinases ,Biology ,BET inhibitor ,medicine ,Animals ,Humans ,Gene silencing ,Gene Silencing ,neoplasms ,Cell Proliferation ,Oncogene ,Cell growth ,Gene Expression Profiling ,medicine.disease ,Xenograft Model Antitumor Assays ,Molecular biology ,Disease Models, Animal ,Kinetics ,Cancer research ,Carcinogenesis ,Transcription Factors - Abstract
BET family proteins are epigenetic regulators known to control expression of genes involved in cell growth and oncogenesis. Selective inhibitors of BET proteins exhibit potent anti-proliferative activity in a number of hematologic cancer models, in part through suppression of the MYC oncogene and downstream Myc-driven pathways. However, little is currently known about the activity of BET inhibitors in solid tumor models, and whether down-regulation of MYC family genes contributes to sensitivity. Here we provide evidence for potent BET inhibitor activity in neuroblastoma, a pediatric solid tumor associated with a high frequency of MYCN amplifications. We treated a panel of neuroblastoma cell lines with a novel small molecule inhibitor of BET proteins, GSK1324726A (I-BET726), and observed potent growth inhibition and cytotoxicity in most cell lines irrespective of MYCN copy number or expression level. Gene expression analyses in neuroblastoma cell lines suggest a role of BET inhibition in apoptosis, signaling, and N-Myc-driven pathways, including the direct suppression of BCL2 and MYCN. Reversal of MYCN or BCL2 suppression reduces the potency of I-BET726-induced cytotoxicity in a cell line-specific manner; however, neither factor fully accounts for I-BET726 sensitivity. Oral administration of I-BET726 to mouse xenograft models of human neuroblastoma results in tumor growth inhibition and down-regulation MYCN and BCL2 expression, suggesting a potential role for these genes in tumor growth. Taken together, our data highlight the potential of BET inhibitors as novel therapeutics for neuroblastoma, and suggest that sensitivity is driven by pleiotropic effects on cell growth and apoptotic pathways in a context-specific manner.
- Published
- 2013
32. The in vitro ejection of zinc from human immunodeficiency virus (HIV) type 1 nucleocapsid protein by disulfide benzamides with cellular anti-HIV activity
- Author
-
Tod P. Holler, Lisa Maloney, Rocco D. Gogliotti, Jeffrey D. Scholten, John M. Domagala, Patricia J. Harvey, Donald Hupe, and Peter J. Tummino
- Subjects
Molecular Sequence Data ,chemistry.chemical_element ,Zinc ,Biology ,medicine.disease_cause ,Antiviral Agents ,Tosyl Compounds ,Structure-Activity Relationship ,Capsid ,medicine ,Humans ,Structure–activity relationship ,Amino Acid Sequence ,Disulfides ,Cloning, Molecular ,Fluorescent Dyes ,Zinc finger ,Multidisciplinary ,Viral Core Proteins ,Tryptophan ,RNA ,Simian immunodeficiency virus ,Virology ,Recombinant Proteins ,In vitro ,Kinetics ,Biochemistry ,chemistry ,Mechanism of action ,Benzamides ,Aminoquinolines ,HIV-1 ,medicine.symptom ,Research Article - Abstract
Several disulfide benzamides have been shown to possess wide-spectrum antiretroviral activity in cell culture at low micromolar to submicromolar concentrations, inhibiting human immunodeficiency virus (HIV) type 1 (HIV-1) clinical and drug-resistant strains along with HIV-2 and simian immunodeficiency virus [Rice, W. G., Supko, J. G., Malspeis, L., Buckheit, R. W., Jr., Clanton, D., Bu, M., Graham, L., Schaeffer, C. A., Turpin, J. A., Domagala, J., Gogliotti, R., Bader, J. P., Halliday, S. M., Coren, L., Sowder, R. C., II, Arthur, L. O. & Henderson, L. E. (1995) Science 270, 1194-1197]. Rice and coworkers have proposed that the compounds act by "attacking" the two zinc fingers of HIV nucleocapsid protein. Shown here is evidence that low micromolar concentrations of the anti-HIV disulfide benzamides eject zinc from HIV nucleocapsid protein (NCp7) in vitro, as monitored by the zinc-specific fluorescent probe N-(6-methoxy-8-quinoyl)-p-toluenesulfonamide (TSQ). Structurally similar disulfide benzamides that do not inhibit HIV-1 in culture do not eject zinc, nor do analogs of the antiviral compounds with the disulfide replaced with a methylene sulfide. The kinetics of NCp7 zinc ejection by disulfide benzamides were found to be nonsaturable and biexponential, with the rate of ejection from the C-terminal zinc finger 7-fold faster than that from the N-terminal. The antiviral compounds were found to inhibit the zinc-dependent binding of NCp7 to HIV psi RNA, as studied by gel-shift assays, and the data correlated well with the zinc ejection data. Anti-HIV disulfide benzamides specifically eject NCp7 zinc and abolish the protein's ability to bind psi RNA in vitro, providing evidence for a possible antiretroviral mechanism of action of these compounds. Congeners of this class are under advanced preclinical evaluation as a potential chemotherapy for acquired immunodeficiency syndrome.
- Published
- 1996
33. Abstract 4709: Broad activity for the combination of BET and MEK inhibitors across solid and hematologic cancers
- Author
-
Victoria Ortiz, Melissa C. Musso, Olena Barbash, Christopher L. Carpenter, Lijoy K. Mathew, Xi-Ping Zhang, Charles F. McHugh, Anastasia Wyce, Peter J. Tummino, Susan Korenchuk, Daniel J. Felitsky, Melissa Stern, Ramona Plant, Jeanne J. Matteo, Yan Degenhardt, Sakina Khaku, and Kathryn Keenan
- Subjects
Cancer Research ,MEK inhibitor ,Biology ,medicine.disease_cause ,Bromodomain ,chemistry.chemical_compound ,Oncology ,chemistry ,Apoptosis ,Cell culture ,Cancer research ,medicine ,Epigenetics ,Growth inhibition ,Signal transduction ,Carcinogenesis - Abstract
BET (bromodomain and extra-terminal) family proteins are epigenetic readers that modulate expression genes involved in cell growth and oncogenesis. Selective small molecule BET inhibitors, such as the GSK I-BETs (I-BET762, I-BET151), abrogate binding of BET proteins to acetylated chromatin and inhibit transcription of BET target genes. We and others have previously demonstrated single agent activity for BET inhibitors in a number of pre-clinical solid and hematologic tumor models. Transcriptomics and mechanistic studies from several of these tumor types indicate that BET inhibitors influence numerous signaling pathways at the transcriptional level, including RAS/RAF/MEK signaling. Here we describe the synergistic effects of combining BET and MEK inhibitors in various solid and hematologic cancer models. We observe synergistic growth inhibition and apoptosis in a subset of cell lines representing multiple tumor types, as well as patient-derived xenograft models treated with the combination ex vivo. Additionally, combination of BET and MEK inhibitors results in improved tumor growth inhibition in cell line xenograft models compared to either single agent therapy. Further exploration of the combination in sensitive tumor types highlights multiple mechanisms potentially driving synergy, and suggests possible markers associated with sensitivity to the combination. Taken together, our data highlight the potential of BET/MEK inhibitor combinations to improve upon the efficacy observed for these agents as monotherapies in a wide variety of preclinical cancer models. All studies were conducted in accordance with the GSK Policy on the Care, Welfare and Treatment of Laboratory Animals and were reviewed by the Institutional Animal Care and Use Committee either at GSK or by the ethical review process at the institution where the work was performed. Human biological samples were sourced ethically and their research use was in accord with the terms of the informed consents. Citation Format: Anastasia Wyce, Daniel J. Felitsky, Xi-Ping Zhang, Jeanne J. Matteo, Susan Korenchuk, Lijoy K. Mathew, Melissa Musso, Sakina Khaku, Victoria Ortiz, Kathryn Keenan, Melissa Stern, Yan Degenhardt, Ramona Plant, Charles F. McHugh, Peter J. Tummino, Christopher Carpenter, Olena Barbash. Broad activity for the combination of BET and MEK inhibitors across solid and hematologic cancers. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 4709.
- Published
- 2016
34. Development and validation of reagents and assays for EZH2 peptide and nucleosome high-throughput screens
- Author
-
Stephanie Chen, Mehul Patel, Louis V. LaFrance, Glenn S. Van Aller, BaoChau Le, Sharon Sweitzer, Yong Jiang, Andrew J. Pope, Danielle Key, Zining Wu, Christopher J. Nixon, Ryan G. Kruger, Melissa B. Pappalardi, Caretha L. Creasy, Carl A. Machutta, Michael T. McCabe, Sara H. Thrall, Benjamin Schwartz, Martin Brandt, Elsie Diaz, Sharad K. Verma, Glenn A. Hofmann, and Peter J. Tummino
- Subjects
Methyltransferase ,macromolecular substances ,Biology ,Biochemistry ,Analytical Chemistry ,Histone H3 ,Histone methylation ,Histone H2A ,Histone code ,Humans ,Enhancer of Zeste Homolog 2 Protein ,Cancer epigenetics ,EZH2 ,Polycomb Repressive Complex 2 ,Reproducibility of Results ,Molecular biology ,High-Throughput Screening Assays ,Nucleosomes ,Kinetics ,Histone methyltransferase ,Molecular Medicine ,Indicators and Reagents ,Drug Screening Assays, Antitumor ,Peptides ,Biotechnology - Abstract
Histone methyltransferases (HMT) catalyze the methylation of histone tail lysines, resulting in changes in gene transcription. Misregulation of these enzymes has been associated with various forms of cancer, making this target class a potential new area for the development of novel chemotherapeutics. EZH2 is the catalytic component of the polycomb group repressive complex (PRC2), which selectively methylates histone H3 lysine 27 (H3K27). EZH2 is overexpressed in prostate, breast, bladder, brain, and other tumor types and is recognized as a molecular marker for cancer progression and aggressiveness. Several new reagents and assays were developed to aid in the identification of EZH2 inhibitors, and these were used to execute two high-throughput screening campaigns. Activity assays using either an H3K27 peptide or nucleosomes as substrates for methylation are described. The strategy to screen EZH2 with either a surrogate peptide or a natural substrate led to the identification of the same tractable series. Compounds from this series are reversible, are [(3)H]-S-adenosyl-L-methionine competitive, and display biochemical inhibition of H3K27 methylation.
- Published
- 2012
35. Does the proliferation of human lymphoma cells depend on the mutation of EZH2 and consequential epigenetic modification of H3K27?
- Author
-
Michael T. McCabe, Caretha L. Creasy, Ashley M. Hughes, Kimberly N. Smitheman, Anthony Della Pietra, Kimberly E. Allen, Sara H. Thrall, Martin Brandt, Gopinath Ganji, Elsie Diaz, Edward Dul, Melissa B. Pappalardi, Ryan G. Kruger, Peter J. Tummino, Heidi M. Ott, Wendy S. Halsey, Yong Jiang, Alan P. Graves, Seth A. Gilbert, Stephanie Chen, and Benjamin J. Schwartz
- Subjects
Heterozygote ,Lymphoma, B-Cell ,Mutant ,DNA Mutational Analysis ,Molecular Sequence Data ,Glycine ,macromolecular substances ,Biology ,medicine.disease_cause ,Methylation ,Substrate Specificity ,Histones ,Histone H3 ,Germline mutation ,Cell Line, Tumor ,medicine ,Humans ,Enhancer of Zeste Homolog 2 Protein ,Amino Acid Sequence ,Mutation ,Multidisciplinary ,Alanine ,Binding Sites ,Base Sequence ,Lysine ,EZH2 ,Polycomb Repressive Complex 2 ,Histone-Lysine N-Methyltransferase ,Biological Sciences ,Molecular biology ,DNA-Binding Proteins ,Gene Expression Regulation, Neoplastic ,Histone ,Histone methyltransferase ,biology.protein ,Histone Methyltransferases ,Mutant Proteins ,Transcription Factors - Abstract
Trimethylation of histone H3 on lysine 27 (H3K27me3) is a repressive posttranslational modification mediated by the histone methyltransferase EZH2. EZH2 is a component of the polycomb repressive complex 2 and is overexpressed in many cancers. In B-cell lymphomas, its substrate preference is frequently altered through somatic mutation of the EZH2 Y641 residue. Herein, we identify mutation of EZH2 A677 to a glycine (A677G) among lymphoma cell lines and primary tumor specimens. Similar to Y641 mutant cell lines, an A677G mutant cell line revealed aberrantly elevated H3K27me3 and decreased monomethylated H3K27 (H3K27me1) and dimethylated H3K27 (H3K27me2). A677G EZH2 possessed catalytic activity with a substrate specificity that was distinct from those of both WT EZH2 and Y641 mutants. Whereas WT EZH2 displayed a preference for substrates with less methylation [unmethylated H3K27 (H3K27me0):me1:me2 k cat / K m ratio = 9:6:1] and Y641 mutants preferred substrates with greater methylation (H3K27me0:me1:me2 k cat / K m ratio = 1:2:13), the A677G EZH2 demonstrated nearly equal efficiency for all three substrates (H3K27me0:me1:me2 k cat / K m ratio = 1.1:0.6:1). When transiently expressed in cells, A677G EZH2, but not WT EZH2, increased global H3K27me3 and decreased H3K27me2. Structural modeling of WT and mutant EZH2 suggested that the A677G mutation acquires the ability to methylate H3K27me2 through enlargement of the lysine tunnel while preserving activity with H3K27me0/me1 substrates through retention of the Y641 residue that is crucial for orientation of these smaller substrates. This mutation highlights the interplay between Y641 and A677 residues in the substrate specificity of EZH2 and identifies another lymphoma patient population that harbors an activating mutation of EZH2.
- Published
- 2012
36. ALDH1A1 is a novel EZH2 target gene in epithelial ovarian cancer identified by genome-wide approaches
- Author
-
Caretha L. Creasy, Marie E. Maradeo, Peter J. Tummino, Rugang Zhang, Vinod Vathipadiekal, Hua Li, Michael Slifker, Michael J. Birrer, Benjamin G. Bitler, and Paul Cairns
- Subjects
Cancer Research ,Chromatin Immunoprecipitation ,endocrine system diseases ,Blotting, Western ,macromolecular substances ,Biology ,Real-Time Polymerase Chain Reaction ,Article ,Aldehyde Dehydrogenase 1 Family ,Histone H3 ,Cell Line, Tumor ,Gene expression ,Biomarkers, Tumor ,Humans ,Enhancer of Zeste Homolog 2 Protein ,RNA, Messenger ,RNA, Small Interfering ,Gene ,Oligonucleotide Array Sequence Analysis ,Regulation of gene expression ,Ovarian Neoplasms ,Gene knockdown ,Gene Expression Profiling ,EZH2 ,Polycomb Repressive Complex 2 ,Retinal Dehydrogenase ,Aldehyde Dehydrogenase ,Molecular biology ,female genital diseases and pregnancy complications ,Gene expression profiling ,DNA-Binding Proteins ,Gene Expression Regulation, Neoplastic ,Oncology ,Cancer research ,Female ,Chromatin immunoprecipitation ,Transcription Factors - Abstract
Epithelial ovarian cancer (EOC) remains the most lethal gynecologic malignancy in the United States. EZH2 silences gene expression through trimethylating lysine 27 on histone H3 (H3K27Me3). EZH2 is often overexpressed in EOC and has been suggested as a target for EOC intervention. However, EZH2 target genes in EOC remain poorly understood. Here, we mapped the genomic loci occupied by EZH2/H3K27Me3 using chromatin immunoprecipitation followed by next-generation sequencing (ChIP-seq) and globally profiled gene expression in EZH2-knockdown EOC cells. Cross-examination of gene expression and ChIP-seq revealed a list of 60 EZH2 direct target genes whose expression was upregulated more than 1.5-fold upon EZH2 knockdown. For three selected genes (ALDH1A1, SSTR1, and DACT3), we validated their upregulation upon EZH2 knockdown and confirmed the binding of EZH2/H3K27Me3 to their genomic loci. Furthermore, the presence of H3K27Me3 at the genomic loci of these EZH2 target genes was dependent upon EZH2. Interestingly, expression of ALDH1A1, a putative marker for EOC stem cells, was significantly downregulated in high-grade serous EOC (n = 53) compared with ovarian surface epithelial cells (n = 10, P < 0.001). Notably, expression of ALDH1A1 negatively correlated with expression of EZH2 (n = 63, Spearman r = −0.41, P < 0.001). Thus, we identified a list of 60 EZH2 target genes and established that ALDH1A1 is a novel EZH2 target gene in EOC cells. Our results suggest a role for EZH2 in regulating EOC stem cell equilibrium via regulation of ALDH1A1 expression. Cancer Prev Res; 5(3); 484–91. ©2011 AACR.
- Published
- 2011
37. A comparative study of succinate-supported respiration and ATP/ADP translocation in liver mitochondria from adult and old rats
- Author
-
Ari Gafni and Peter J. Tummino
- Subjects
Male ,Aging ,medicine.medical_specialty ,Succinic Acid ,chemistry.chemical_element ,Mitochondria, Liver ,Chromosomal translocation ,Mitochondrion ,Oxygen ,Adenosine Triphosphate ,Oxygen Consumption ,Internal medicine ,Respiration ,medicine ,Animals ,Translocase ,biology ,Succinates ,Electron transport chain ,Rats, Inbred F344 ,Rats ,Adenosine Diphosphate ,Kinetics ,Endocrinology ,medicine.anatomical_structure ,chemistry ,biology.protein ,ATP–ADP translocase ,Developmental Biology ,Respiratory tract - Abstract
This study was undertaken to compare the rates of succinate-supported hepatic mitochondrial respiration between 12 months (adult) and 29 months (old) male Fischer 344 rats. Experiments were also performed to determine the activity of adenine nucleotide translocase and the effect of its inhibition on mitochondrial respiration. Succinate-supported state 3 mitochondrial respiration was found to decline 20% between 12 and 29 months of age in rat liver, along with a similar 25% decrease in the respiratory control ratio with age. Adenine nucleotide translocase activity is shown to decrease 39% from adult to old rat liver mitochondria. This decrease does not, however, account for the decline in state 3 respiration, since translocase activity is approximately 50% greater than state 3 respiration in both adult and old rats. Therefore, adenine nucleotide translocase is not rate-limiting for state 3 mitochondrial respiration. Neither the rate of succinate permeation into the mitochondrial nor the rate of electron transport is rate-limiting for state 3 respiration, indicated by the greatly increased oxygen consumption with addition of the uncoupler carbonyl cyanide m-chlorophenyl hydrazone (m-CCCP). These processes, therefore, are not responsible for the observed decline in state 3 respiration. The implications and possible cause of the age-related decrease in the maximal rate of ATP-synthesis are discussed.
- Published
- 1991
38. Effects of oncogenic p110alpha subunit mutations on the lipid kinase activity of phosphoinositide 3-kinase
- Author
-
Glenn S. Van Aller, Kurt R. Auger, Robert H. Sinnamon, Dashyant Dhanak, Ruth Lehr, Robert A. Copeland, Robert B. Kirkpatrick, Lusong Luo, Jeffrey D. Carson, Richard R. Gontarek, and Peter J. Tummino
- Subjects
Class I Phosphatidylinositol 3-Kinases ,Lipid kinase activity ,Mitogen-activated protein kinase kinase ,Biochemistry ,MAP2K7 ,Phosphatidylinositol 3-Kinases ,Adenosine Triphosphate ,Catalytic Domain ,Tumor Cells, Cultured ,Humans ,ASK1 ,Phosphorylation ,Molecular Biology ,Alleles ,Adaptor Proteins, Signal Transducing ,MAP kinase kinase kinase ,biology ,Cyclin-dependent kinase 4 ,Cyclin-dependent kinase 2 ,Cell Biology ,Oncogenes ,Molecular biology ,Recombinant Proteins ,Enzyme Activation ,Kinetics ,Mutation ,biology.protein ,Insulin Receptor Substrate Proteins ,Cyclin-dependent kinase 9 - Abstract
The PIK3CA gene, encoding the p110α catalytic subunit of Class IA PI3Ks (phosphoinositide 3-kinases), is frequently mutated in many human tumours. The three most common tumour-derived alleles of p110α, H1047R, E542K and E545K, were shown to potently activate PI3K signalling in human epithelial cells. In the present study, we examine the biochemical activity of the recombinantly purified PI3K oncogenic mutants. The kinetic characterizations of the wt (wild-type) and the three ‘hot spot’ PI3K mutants show that the mutants all have approx. 2-fold increase in lipid kinase activities. Interestingly, the phosphorylated IRS-1 (insulin receptor substrate-1) protein shows activation of the lipid kinase activity for the wt and H1047R but not E542K and E545K PI3Kα, suggesting that these mutations represent different mechanisms of lipid kinase activation and hence transforming activity in cancer cells.
- Published
- 2007
39. Abstract B34: Antitumor activity and sensitivity evaluation of novel BET inhibitors in neuroblastoma
- Author
-
Olena Barbash, John M. Maris, Ryan M. Kinsey, Maria Gagliardi, Mike R. Russell, Anastasia Wyce, Pichai Raman, Priya Khurana, Robert W. Schnepp, Peter J. Tummino, Lori S. Hart, Jason R. Healy, Louis Chesler, and Laura Danielson
- Subjects
Cancer Research ,Cancer ,Cell cycle ,Biology ,medicine.disease ,medicine.disease_cause ,Molecular biology ,Oncology ,In vivo ,Cell culture ,Neuroblastoma ,Gene expression ,medicine ,Cancer research ,Epigenetics ,Carcinogenesis ,Molecular Biology - Abstract
Children with neuroblastoma exhibit marked variability in outcome based on age at diagnosis, disease stage and tumor biologic characteristics. Genomic amplification of MYCN is a potent oncogenic driver and negative prognostic marker in neuroblastoma patients. Therefore, there is a pressing need to identify therapeutic agents that target MYCN gene expression. The bromodomain and extra-terminal (BET) family of proteins are epigenetic regulators known to control expression of genes involved in cell growth and oncogenesis. Previous reports demonstrate that BET inhibition (BETi) significantly attenuates cellular proliferation in numerous cancer models, perhaps via modulation of the MYC and/or MYCN oncogenes. Here, we show that potent BET inhibition induces anti-tumor effects in preclinical neuroblastoma models. Specifically, GSK726 and GSK762 (GlaxoSmithKline) were used for in vitro cytotoxicity and in vivo therapeutic studies, respectively. Neuroblastoma cell lines (n=22) were treated with GSK726 to calculate IC50 values in a luminescence-based cell viability assay and differential sensitivity was observed with an IC50 range of 27 nM to 9.5 µM. In sensitive cell lines, cell cycle distribution and induction of apoptosis were also measured. In these cell lines, GSK726 treatment resulted in MYCN depletion, G1 arrest within 24 hours, and apoptosis as measured by cleaved-PARP. To assess in vivo efficacy, GSK762 was subcutaneously administered in xenograft models and in genetically engineered neuroblastoma mouse models overexpressing MYCN and MYCN/ALK F1174L in the neural crest. In both models, GSK762 treatment resulted in tumor growth delay. Further assessment of results seen in vitro and in vivo indicated that MYCN amplification status did not fully predict sensitivity to GSK726 or GSK762. Thus, to determine additional biomarkers of sensitivity, we examined baseline gene expression data at the extremes of IC50 values by comparing sensitive (n=6; IC50940 nM) neuroblastoma cell lines. Gene expression data from the neuroblastoma cell lines generated on HuGene1.0ST expression microarrays (Affymetrix) were utilized, and data were analyzed using the Limma package in R/Bioconductor. Univariate analysis, with a false discovery rate less than 0.25, revealed 6 genes (PTER, PCDHB14, RFTN1, JAK2, MYCBP, and DACH1) that were differentially expressed between sensitive and resistant cell lines in the MYCN amplified setting. While all 6 genes predicted BETi sensitivity in the MYCN-amplified subset of cell lines, only DACH1 expression predicted sensitivity to BET inhibition irrespective of MYCN amplification status (p=7.92x10-7). In addition, high DACH1 expression was correlated with poor patient outcome (p=1.74x10-5). As a result, high DACH1 expression serves as a candidate biomarker for future studies with respect to sensitivity to BETi. Ultimately, these studies will help to optimize the clinical utility of BETi in neuroblastoma and perhaps other MYC-driven malignancies. Citation Format: Jason R. Healy, Robert W. Schnepp, Lori S. Hart, Pichai Raman, Laura Danielson, Michael Russell, Priya Khurana, Maria Gagliardi, Ryan M. Kinsey, Anastasia Wyce, Olena Barbash, Peter J. Tummino, Louis Chesler, John M. Maris. Antitumor activity and sensitivity evaluation of novel BET inhibitors in neuroblastoma. [abstract]. In: Proceedings of the AACR Special Conference on Myc: From Biology to Therapy; Jan 7-10, 2015; La Jolla, CA. Philadelphia (PA): AACR; Mol Cancer Res 2015;13(10 Suppl):Abstract nr B34.
- Published
- 2015
40. Abstract 2867: Inhibition of BET bromodomain proteins as a therapeutic approach in small cell lung cancer
- Author
-
Maureen R. Bleam, Jeanne J. Matteo, David Soong, Xi-Ping Zhang, Susan Korenchuk, Michael Butticello, BaoChau Le, Charles F. McHugh, Olena Barbash, Yan Degenhardt, Peter J. Tummino, Anastasia Wyce, Yuchen Bai, Christopher L. Carpenter, and Ramona Plant
- Subjects
Cancer Research ,BRD4 ,Oncogene ,Cancer ,Biology ,medicine.disease_cause ,medicine.disease ,Molecular biology ,Bromodomain ,BET inhibitor ,chemistry.chemical_compound ,Oncology ,chemistry ,Cancer research ,medicine ,Growth inhibition ,Carcinogenesis ,MYC Family Gene - Abstract
BET (bromodomain and extra-terminal) family proteins are epigenetic readers that regulate expression of genes involved in cell growth and oncogenesis. Selective small molecule BET inhibitors, such as the GSK I-BETs (I-BET762, I-BET151), abrogate binding of BET proteins to acetylated histones and inhibit transcriptional activation of BET target genes. BET inhibitors attenuate proliferation and survival in a number of hematologic cancer models, partially through down-regulation of the critical oncogene, MYC. We and others have previously shown activity for BET inhibitors in solid tumor models such as neuroblastoma and prostate cancer, with concomitant down-regulation of MYC family gene expression. However, MYC or MYCN silencing only partially accounts for the activity of BET inhibitors in these models, suggesting that transcriptional regulation of multiple pathways promotes I-BET induced phenotypes. Here we describe the activity of the selective BET inhibitor, I-BET762, in small cell lung cancer (SCLC), a highly aggressive malignancy with few treatment options. We observe potent growth inhibition and apoptosis in a subset of cell lines and patient-derived SCLC models in vitro, as well as significant tumor growth inhibition in cell line and primary mouse xenografts. These anti-proliferative effects are likely mediated by inhibition of BRD2, BRD3, and BRD4, as siRNAs individually targeting all three proteins result in partial inhibition of SCLC cell line growth. Similar to our observations in neuroblastoma and prostate cancer, I-BET762 treatment in SCLC cell lines results in transcriptional changes affecting MYC and other pathways. We explore the contribution of these changes to the anti-proliferative effects observed in SCLC models, and identify potential combination strategies to enhance the activity of I-BET762. Taken together, our data highlight the potential of BET inhibitors as novel therapeutic agents to treat small cell lung cancer driven by various oncogenic pathways. All studies were conducted in accordance with the GSK Policy on the Care, Welfare and Treatment of Laboratory Animals and were reviewed by the Institutional Animal Care and Use Committee either at GSK or by the ethical review process at the institution where the work was performed. Citation Format: Anastasia Wyce, BaoChau Le, Yuchen Bai, David Soong, Xi-Ping Zhang, Jeanne J. Matteo, Susan Korenchuk, Michael F. Butticello, Ramona Plant, Maureen R. Bleam, Yan Degenhardt, Charles F. McHugh, Christopher Carpenter, Peter J. Tummino, Olena Barbash. Inhibition of BET bromodomain proteins as a therapeutic approach in small cell lung cancer. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 2867. doi:10.1158/1538-7445.AM2015-2867
- Published
- 2015
41. Abstract A23: SMYD3 links methylation of MAP3K2 to Ras-driven tumors
- Author
-
Michiel Vermeulen, Purvesh Khatri, Alex W. Wilkinson, Julien Sage, Pascal W. T. C. Jansen, Glenn S. Van Aller, Peter J. Tummino, Pawel K. Mazur, Benjamin A. Garcia, Atul J. Butte, Nicolas Reynoird, Olena Barbash, Or Gozani, Shichong Liu, Michael J. Huddleston, and Ryan G. Kruger
- Subjects
MAPK/ERK pathway ,Genetics ,Cancer Research ,Kinase ,Cancer ,Protein phosphatase 2 ,Methylation ,MAP3K2 ,Biology ,medicine.disease ,medicine.disease_cause ,Oncology ,medicine ,Cancer research ,Phosphatase complex ,Carcinogenesis - Abstract
The Ras family of oncogenes is activated in a large fraction of human cancers. Treatment of Ras-driven tumors with inhibitors of protein kinases in the Ras signaling network, such as Raf or MEK is a promising therapeutic strategy. However, toxicity issues and the emergence of tumor cells that are resistant to these drugs underscore the need for a better understanding of the Ras pathway and for the development of novel therapeutic options to improve the survival of cancer patients. Deregulation in lysine methylation signaling has emerged as a common etiologic factor in cancer pathogenesis, with inhibitors of several histone lysine methyltransferases (KMTs) being developed as chemotherapeutics. The largely cytoplasmic KMT SMYD3 (SET and MYND domain containing protein 3) is overexpressed in numerous human tumors. However, the molecular mechanism by which SMYD3 regulates cancer pathways and its relationship to tumorigenesis in vivo are largely unknown. Here we show that methylation of MAP3K2 by SMYD3 increases MAP Kinase signaling and promotes the formation of Ras-driven carcinomas. Using mouse models for pancreatic ductal adenocarcinoma (PDAC) and lung adenocarcinoma (LAC), we found that abrogating SMYD3 catalytic activity inhibits tumor development in response to oncogenic Ras. We employed protein array technology to identify the MAP3K2 kinase as a target of SMYD3. In cancer cell lines, SMYD3-mediated methylation of MAP3K2 at lysine 260 potentiates activation of the Ras/Raf/MEK/ERK signaling module. Finally, the PP2A phosphatase complex, a key negative regulator of the MAP Kinase pathway, binds to MAP3K2 and this interaction is blocked by methylation. Together, our results elucidate a new role for lysine methylation in integrating cytoplasmic kinase-signaling cascades and establish a pivotal role for SMYD3 in the regulation of oncogenic Ras signaling. Citation Format: Pawel K. Mazur, Nicolas Reynoird, Purvesh Khatri, Atul J. Butte, Alex Wilkinson, Benjamin Garcia, Shichong Liu, Michiel Vermeulen, Pascal W.T.C. Jansen, Peter J. Tummino, Ryan G. Kruger, Glenn S. Van Aller, Olena Barbash, Michael Huddleston, Or Gozani, Julien Sage. SMYD3 links methylation of MAP3K2 to Ras-driven tumors. [abstract]. In: Proceedings of the AACR Special Conference on Pancreatic Cancer: Innovations in Research and Treatment; May 18-21, 2014; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2015;75(13 Suppl):Abstract nr A23.
- Published
- 2015
42. Expression, purification, and enzymatic characterization of the dual specificity mitogen-activated protein kinase phosphatase, MKP-4
- Author
-
Thomas G. Gant, Anthony J. Petrolonis, Haiyan Xu, Thomas F. Parsons, Andrew J. Nichols, Thomas H. Lubben, Rebecca Roy, Christine M. Dolliver, Peter J. Tummino, Ping Li, James M. Trevillyan, Zhi Li, Regina M. Reilly, and Suk-Bong Hong
- Subjects
Phosphatase ,DUSP6 ,Protein tyrosine phosphatase ,Biochemistry ,p38 Mitogen-Activated Protein Kinases ,Substrate Specificity ,Inhibitory Concentration 50 ,Drug Discovery ,Dual-specificity phosphatase ,Phosphoprotein Phosphatases ,Humans ,Insulin ,c-Raf ,Protein kinase A ,Molecular Biology ,Cells, Cultured ,Mitogen-Activated Protein Kinase 3 ,biology ,Akt/PKB signaling pathway ,Chemistry ,Organic Chemistry ,Protein phosphatase 2 ,Hydrogen-Ion Concentration ,Molecular biology ,Kinetics ,Spectrometry, Fluorescence ,Gene Expression Regulation ,biology.protein ,Dual-Specificity Phosphatases ,Mitogen-Activated Protein Kinase Phosphatases ,Electrophoresis, Polyacrylamide Gel ,Protein Tyrosine Phosphatases - Abstract
Mitogen-activated protein kinase phosphatase-4 (MKP-4) is a dual specificity phosphatase, which acts as a negative regulator of insulin-stimulated pathways [1]. Here, we describe expression, purification, and biochemical characterization of MKP-4. We used the Baculovirus expression system and purification with a combination of affinity and gel filtration chromatography to generate pure MKP-4 and MKP-4/p38 complex. Both MKP-4 and the MKP-4/p38 complex exhibited moderate activity toward the surrogate substrates p-nitrophenyl phosphate, 6, 8-difluoro-4-methylumbelliferyl phosphate, and 3-O-methylfluorescein phosphate. The phosphatase activity could be inhibited by peroxovanate, a potent inhibitor of protein tyrosine phosphatases. We further determined kinetic parameters for the MKP-4 and the MKP-4/p38 by using spectrophotometric and fluorescence intensity methods. The MKP-4/p38 complex was found to provide substantially higher phosphatase activity than MKP-4 alone, similar to what has been shown for MKP-3. Our data allow the configuration of screens for modulators of MKP-4 activity.
- Published
- 2004
43. Abstract B75: Defining the antitumor activity and sensitivity profiles of BET inhibitors in neuroblastoma
- Author
-
Laura Danielson, Robert W. Schnepp, Peter J. Tummino, Maria Gagliardi, John M. Maris, Louis Chesler, Anastasia Wyce, Lori S. Hart, Pichai Raman, Ryan M. Kinsey, and Olena Barbash
- Subjects
Cancer Research ,BRD4 ,medicine.diagnostic_test ,Cell cycle ,Biology ,medicine.disease ,Pediatric cancer ,Molecular biology ,Flow cytometry ,Bromodomain ,Oncology ,Neuroblastoma ,medicine ,Gene silencing ,N-Myc - Abstract
Background: MYCN amplification is the most frequent somatically acquired genomic alteration in neuroblastoma and is a potent oncogenic driver. Targeting MYCN therapeutically has been complicated by the challenges inherent in targeting transcription factors. Recently, several groups have shown that inhibiting bromodomain and extra-terminal (BET) proteins (BRD2, BRD3, BRD4, and BRDT) and their ability to bind to acetylated lysine residues within histone tails resulted in silencing of MYC family protein expression and demonstrated therapeutic efficacy in preclinical cancer models. We hypothesized that potent inhibition of bromodomain-chromatin interactions would show anti-tumor activity in preclinical models of neuroblastoma and that reliable biomarkers of activity could be discovered. Methods: GlaxoSmithKline compounds GSK726 and GSK762 potently inhibit BET proteins (BETi) and were used for in vitro cytotoxicity and in vivo therapeutic studies, respectively. We exposed a panel of 15 cell lines to a 4-log dose range of GSK726 to determine IC50s in a luminescence-based cell viability assay. We also measured cell cycle changes by flow cytometry and apoptosis by immunoblotting for cleaved PARP. We tested the in vivo efficacy of the GSK762 in subcutaneous xenograft models (3 sensitive and 2 resistant cell lines) and in genetically engineered neuroblastoma mouse models (overexpressing MYCN and MYCN/ALK F1174L in the neural crest). For biomarker discovery, gene expression data from the neuroblastoma cell lines generated on HuGene1.0ST expression microarrays (Affymetrix) were utilized, and data were analyzed using the Limma package in R/Bioconductor. Results: Neuroblastoma cell lines (N=15) were differentially sensitive to GSK726, with a median IC50 of 128nM and a range of 17nM to >10uM. In sensitive cell lines, GSK726 treatment resulted in MYCN depletion, G1 arrest within 24 hours, and apoptosis. In subcutaneous xenograft models and genetically engineered neuroblastoma mouse models, GSK762 treatment resulted in tumor growth delay. Importantly, MYCN amplification status did not fully predict sensitivity to GSK726 or GSK762. Thus, to determine additional biomarkers of sensitivity, we examined baseline gene expression data at the extremes of IC50s by comparing sensitive (N=6; IC50940 nM) neuroblastoma cell lines. Univariate analysis, with an FDR < 0.25, revealed 6 genes (PTER, PCDHB14, RFTN1, JAK2, MYCBP, and DACH1) differentially expressed between sensitive and resistant cell lines in the MYCN amplified setting. While all 6 genes predicted BETi sensitivity in the MYCN-amplified subset of cell lines, only DACH1 expression predicted sensitivity to BET inhibition irrespective of MYCN amplification status (7.92x10-7). Moreover, DACH1 levels were heterogeneously expressed in a panel of 88 primary neuroblastoma tumors (Versteeg-88) and high expression was correlated with poor patient outcome (1.74x10-5). Conclusions: BET inhibitors demonstrated significant anti-tumor activity in a subset of neuroblastoma preclinical models. Although MYCN protein levels decreased in neuroblastoma cell lines that were sensitive to BETi, MYCN amplification status alone did not fully explain BETi sensitivity. DACH1 expression serves as a candidate biomarker for sensitivity to BET inhibition. These studies will help to optimize the clinical utility of BETi in neuroblastoma and perhaps other MYC-driven malignancies. Citation Format: Robert Schnepp, Lori Hart, Pichai Raman, Laura Danielson, Maria Gagliardi, Ryan Kinsey, Anastasia Wyce, Olena Barbash, Peter Tummino, Louis Chesler, John Maris. Defining the antitumor activity and sensitivity profiles of BET inhibitors in neuroblastoma. [abstract]. In: Proceedings of the AACR Special Conference on Pediatric Cancer at the Crossroads: Translating Discovery into Improved Outcomes; Nov 3-6, 2013; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2013;74(20 Suppl):Abstract nr B75.
- Published
- 2014
44. Abstract 5151: A687V EZH2 is a driver of histone H3 lysine 27 (H3K27) hyper-trimethylation
- Author
-
Melissa B. Pappalardi, Alan P. Graves, Ryan G. Kruger, Heidi M. Ott, Caretha L. Creasy, Michael T. McCabe, and Peter J. Tummino
- Subjects
Regulation of gene expression ,Cancer Research ,Mutation ,Methyltransferase ,Mutant ,EZH2 ,macromolecular substances ,Methylation ,Biology ,medicine.disease_cause ,Molecular biology ,Histone H3 ,Oncology ,Cell culture ,medicine - Abstract
The EZH2 methyltransferase silences gene expression through methylation of histone H3 on lysine 27 (H3K27). Recently, EZH2 mutations have been reported at Y641, A677, and A687 in non-Hodgkin's lymphoma. While the Y641F/N/S/H/C and A677G mutations exhibit clearly increased activity with substrates di-methylated at lysine 27 (H3K27me2), the A687V mutant has been shown to prefer a mono-methylated lysine 27 (H3K27me1) with little gain of activity toward H3K27me2. Herein, we demonstrate that despite this unique substrate preference, A687V EZH2 still drives increased H3K27me3 when transiently expressed in cells. However, unlike the previously described mutants which dramatically deplete global H3K27me2 levels, A687V EZH2 retains normal levels of H3K27me2. Sequencing of B-cell derived cancer cell lines identified a cell line harboring this mutation. Similar to exogenous expression of A687V EZH2, this cell line exhibited elevated H3K27me3 while possessing H3K27me2 levels higher than Y641 or A677 mutant lines. Treatment of A687V EZH2 mutant cells with GSK126, a selective EZH2 inhibitor, induced global H3K27me3 demethylation, robust gene activation, caspase activation, and decreased proliferation. These findings suggest that A687V EZH2 likely increases global H3K27me3 indirectly through increased catalytic activity with H3K27me1 and cells harboring this mutation are highly dependent on EZH2 activity for their survival. Citation Format: Heidi Ott, Alan Graves, Melissa Pappalardi, Ryan Kruger, Peter Tummino, Caretha Creasy, Michael T. McCabe. A687V EZH2 is a driver of histone H3 lysine 27 (H3K27) hyper-trimethylation. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 5151. doi:10.1158/1538-7445.AM2014-5151
- Published
- 2014
45. Abstract 382: Inhibition of BET bromodomain proteins as a therapeutic approach in prostate cancer
- Author
-
Caretha L. Creasy, Susan Korenchuk, Robert L. Vessella, Charles F. McHugh, Anastasia Wyce, BaoChau Le, Yan Degenhardt, Yuchen Bai, Ming-Chih Crouthamel, Peter J. Tummino, and Olena Barbash
- Subjects
Cancer Research ,Oncogene ,Cell growth ,Cancer ,Biology ,medicine.disease_cause ,medicine.disease ,Bromodomain ,BET inhibitor ,Prostate cancer ,Oncology ,medicine ,Cancer research ,Carcinogenesis ,MYC Family Gene - Abstract
BET (bromodomain and extra-terminal) family proteins are epigenetic regulators known to control expression of genes involved in cell growth and oncogenesis. Selective small molecule BET inhibitors prevent binding of BET proteins to acetylated histones and inhibit transcriptional activation of BET target genes. BET inhibitors attenuate cell growth and survival in a number of hematologic cancer models, partially through down-regulation of the critical oncogene, MYC. We hypothesized that BET inhibitors will similarly regulate expression of MYC family genes (MYC, MYCN, MYCL1) in solid tumor models characterized by MYC family amplification or over-expression. We and others have recently shown activity for BET inhibitors in MYCN-amplified neuroblastoma models, with concomitant down-regulation of MYCN expression. Here we describe the effects of the highly specific BET inhibitor, I-BET762, on MYC expression and cell growth in prostate cancer models. I-BET762 treatment inhibited MYC expression accompanied by growth inhibition and decreased survival in prostate cancer cell lines that over-express MYC. In addition to MYC signatures, gene expression profiling in cell lines identified numerous cell cycle-associated genes as being significantly down-regulated by I-BET762. Importantly, our data suggests that I-BET762 effects are partially driven by MYC down-regulation and underlines the critical importance of additional mechanisms of I-BET762 induced phenotypes. Consistent with our in vitro observations, BET inhibition reduces MYC expression and tumor burden in a primary model of castration resistant prostate cancer that expresses high levels of MYC. Taken together, our data highlight the potential of BET inhibitors as a novel therapeutic approach to treat prostate tumors driven by MYC over-expression. Citation Format: Anastasia Wyce, Yan Degenhardt, Yuchen Bai, BaoChau Le, Susan Korenchuk, Ming-Chih Crouthamel, Charles F. McHugh, Robert Vessella, Caretha L. Creasy, Peter J. Tummino, Olena Barbash. Inhibition of BET bromodomain proteins as a therapeutic approach in prostate cancer. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 382. doi:10.1158/1538-7445.AM2014-382
- Published
- 2014
46. The human immunodeficiency virus type 1 (HIV-1) nucleocapsid protein zinc ejection activity of disulfide benzamides and benzisothiazolones: correlation with anti-HIV and virucidal activities
- Author
-
Yuntao Song, Joseph P. Sanchez, John M. Domagala, Rocco D. Gogliotti, Peter J. Tummino, Thomas J. McQuade, Patricia J. Harvey, and Donald Hupe
- Subjects
animal structures ,Anti-HIV Agents ,chemistry.chemical_element ,Gene Products, gag ,Zinc ,gag Gene Products, Human Immunodeficiency Virus ,Dithiothreitol ,chemistry.chemical_compound ,Structure-Activity Relationship ,Viral Proteins ,Capsid ,Cytopathogenic Effect, Viral ,medicine ,Structure–activity relationship ,Humans ,Pharmacology (medical) ,Disulfides ,Benzamide ,Pharmacology ,chemistry.chemical_classification ,Zinc finger ,Biological activity ,Zinc Fingers ,Kinetics ,Thiazoles ,Infectious Diseases ,Mechanism of action ,Biochemistry ,chemistry ,Benzamides ,Thiol ,HIV-1 ,Capsid Proteins ,medicine.symptom ,Research Article - Abstract
It has been shown previously by our group and others that a series of four disulfide benzamides with cellular anti-human immunodeficiency virus (HIV) activity can eject zinc from HIV type 1 nucleocapsid protein (NCp7) in vitro while analogs without antiviral activity do not. We also found that the zinc ejection activity correlates with the loss of the ability of NCp7 to bind to HIV psi RNA in vitro. These observations indicate that the antiviral disulfide benzamides may act at a novel retroviral target of action, i.e., the nucleocapsid protein. The present studies examine the relationship among disulfide benzamide structure, in vitro NCp7 zinc ejection activity, and antiviral activity for a larger series of compounds. All of the antiviral disulfide benzamides were found to eject NCp7 zinc, while some disulfide benzamides with zinc ejection activity are not antiviral. Utilizing the thiol reagent 5,5'-dithiobis(2-nitrobenzoic acid), it was determined that the o-amido-phenyl disulfides being studied cyclize in aqueous solution to form benzisothiazolones. A series of benzisothiazolones, which are stable in solution in the absence of dithiothreitol, were found to eject NCp7 zinc at a rate similar to that of their disulfide benzamide analogs and to possess similar antiviral activity. It was also found that the relative rates of HIV inactivation by various disulfide benzamides and benzisothiazolones correlate with their relative kinetic rates of NCp7 zinc ejection, which is consistent with the nucleocapsid protein being the target of action of these compounds.
- Published
- 1997
47. Inhibition Of LSD1 As a Therapeutic Strategy For The Treatment Of Acute Myeloid Leukemia
- Author
-
Jessica L. Schneck, Dominic Suarez, Peter J. Tummino, Scott A. Armstrong, Yan Liu, Patrick McDevitt, Helai P. Mohammad, Glenn S. Van Aller, Xinrong Tian, William H. Miller, Kimberly N. Smitheman, Kelly Federowicz, Thau F. Ho, Jeffrey D. Carson, Melissa B. Pappalardi, Johnson Neil W, Monica Cusan, Kasparec Jiri, Rouse Meagan B, Charles F. McHugh, and Ryan G. Kruger
- Subjects
CD86 ,business.industry ,Cellular differentiation ,Immunology ,Myeloid leukemia ,Cell Biology ,Hematology ,Biochemistry ,chemistry.chemical_compound ,Haematopoiesis ,Immunophenotyping ,medicine.anatomical_structure ,chemistry ,Cancer research ,medicine ,Bone marrow ,Growth inhibition ,Stem cell ,business - Abstract
Lysine specific demethylase 1 (LSD1) is a histone H3K4me1/2 demethylase found in various transcriptional co-repressor complexes. These complexes include Histone Deacetylases (HDAC1/2) and Co-Repressor for Element-1-Silencing Transcription factor (CoREST). LSD1 mediated H3K4 demethylation can result in a repressive chromatin environment that silences gene expression. LSD1 has been shown to play a role in development in various contexts. LSD1 can interact with pluripotency factors in human embryonic stem cells and is important for decommissioning enhancers in stem cell differentiation. Beyond embryonic settings, LSD1 is also critical for hematopoietic differentiation. LSD1 is overexpressed in multiple cancer types and recent studies suggest inhibition of LSD1 reactivates the all-trans retinoic acid receptor pathway in acute myeloid leukemia (AML). These studies implicate LSD1 as a key regulator of the epigenome that modulates gene expression through post-translational modification of histones and through its presence in transcriptional complexes. The current study describes the anti-tumor effects of a novel LSD1 inhibitor (GSK2879552) in AML. GSK2879552 is a potent, selective, mechanism-based, irreversible inhibitor of LSD1. Screening of over 150 cancer cell lines revealed that AML cells have a unique requirement for LSD1. While LSD1 inhibition did not affect the global levels of H3K4me1 or H3K4me2, local changes in these histone marks were observed near transcriptional start sites of putative LSD1 target genes. This increase in the transcriptionally activating histone modification correlated with a dose dependent increase in gene expression. Treatment with GSK2879552 promoted the expression of cell surface markers, including CD11b and CD86, associated with a differentiated immunophenotype in 12 of 13 AML cell lines. For example, in SKM-1 cells, increases in cell surface expression of CD86 and CD11b occurred after as early as one day of treatment with EC50 values of 13 and 7 nM respectively. In a separate study using an MV-4-11 engraftment model, increases in CD86 and CD11b were observed as early as 8 hours post dosing. GSK2879552 treatment resulted in a potent anti-proliferative growth effect in 19 of 25 AML cell lines (average EC50 = 38 nM), representing a range of AML subtypes. Potent growth inhibition was also observed on AML blast colony forming ability in 4 out of 5 bone marrow samples derived from primary AML patient samples (average EC50 = 205 nM). The effects of LSD1 inhibition were further characterized in an in vivo mouse model of AML induced by transduction of mouse hematopoietic progenitor cells with a retrovirus encoding MLL-AF9 and GFP. Primary AML cells were transplanted into a cohort of secondary recipient mice and upon engraftment, the mice were treated for 17 days. After 17 days of treatment, control treated mice had 80% GFP+ cells in the bone marrow whereas treated mice possessed 2.8% GFP positive cells (p Together, these data demonstrate that pharmacological inhibition of LSD1 may provide a promising treatment for AML by promoting differentiation and subsequent growth inhibition of AML blasts. GSK2879552 is currently in late preclinical development and clinical trials are anticipated to start in 2014. All studies were conducted in accordance with the GSK Policy on the Care, Welfare and Treatment of Laboratory Animals and were reviewed the Institutional Animal Care and Use Committee either at GSK or by the ethical review process at the institution where the work was performed. Disclosures: Kruger: GlaxoSmithKline Pharmaceuticals: Employment. Mohammad:GlaxoSmithKline Pharmaceuticals: Employment. Smitheman:GlaxoSmithKline Pharmaceuticals: Employment. Liu:GlaxoSmithKline Pharmaceuticals: Employment. Pappalardi:GlaxoSmithKline Pharmaceuticals: Employment. Federowicz:GlaxoSmithKline Pharmaceuticals: Employment. Van Aller:GlaxoSmithKline Pharmaceuticals: Employment. Kasparec:GlaxoSmithKline Pharmaceuticals: Employment. Tian:GlaxoSmithKline Pharmaceuticals: Employment. Suarez:GlaxoSmithKline Pharmaceuticals: Employment. Rouse:GlaxoSmithKline Pharmaceuticals: Employment. Schneck:GlaxoSmithKline Pharmaceuticals: Employment. Carson:GlaxoSmithKline Pharmaceuticals: Employment. McDevitt:GlaxoSmithKline Pharmaceuticals: Employment. Ho:GlaxoSmithKline Pharmaceuticals: Employment. McHugh:GlaxoSmithKline Pharmaceuticals: Employment. Miller:GlaxoSmithKline Pharmaceuticals: Employment. Johnson:GlaxoSmithKline Pharmaceuticals: Employment. Armstrong:Epizyme Inc.: Has consulted for Epizyme Inc. Other. Tummino:GlaxoSmithKline Pharmaceuticals: Employment.
- Published
- 2013
48. Abstract 3145: Smyd3 catalyzes a novel methylation mark and regulates cancer cell proliferation
- Author
-
Roland S. Annan, Peter J. Tummino, Glenn S. VanAller, Olena Barbash, Or Gozani, Ryan G. Kruger, Nicolas Reynoird, BaoChau Le, Michael J. Huddleston, and Ben A. Garcia
- Subjects
Cancer Research ,Methyltransferase ,Chemistry ,Wild type ,Cancer ,Methylation ,medicine.disease ,Molecular biology ,Histone H4 ,Histone H3 ,Oncology ,Cancer cell ,medicine ,Nucleosome - Abstract
SET and MYND domain-containing protein 3 (Smyd3) is a lysine methyltransferase over-expressed in liver, breast and rectal carcinomas. We have shown that knockdown of Smyd3 has an anti-proliferative effect in cancer cell lines and that this effect can be rescued with wild type but not catalytically inactive Smyd3, directly linking Smyd3 catalytic activity to proliferation. Published reports indicate Smyd3 methylates lysine 4 of histone H3, lysine 20 of histone H4 and VEGFR1. However, activity testing of a peptide library and intact recombinant nucleosomes indicate that a unique methylation mark is catalyzed by Smyd3. LC-MS/MS analysis of in vitro reaction products have identified the location of this novel site of methylation. The existence of this novel mark and its dependence on Smyd3 activity was confirmed in cancer cells and mouse embryonic fibroblasts. We speculate that overexpression of Smyd3 causes aberrant methylation and plays an important role in the deregulation of gene expression leading to uncontrolled cellular proliferation in cancer making Smyd3 a promising target for anti-cancer therapy. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 3145. doi:1538-7445.AM2012-3145
- Published
- 2012
49. Abstract 2939: Discovery and synthesis of highly potent and selective small molecule inhibitors of the histone methyltransferase EZH2
- Author
-
Michael T. McCabe, Christine Thompson, Ryan G. Kruger, Caretha L. Creasy, Louis V. LaFrance, Stuart Paul Romeril, Mellinger Mark, Charles F. McHugh, Xinrong Tian, Peter J. Tummino, Dash Dhanak, Ken A. Newlander, Dominic Suarez, Alan P. Graves, Celine Duquenne, Brackley James, Sharad K. Verma, BaoChau Le, Daryl A. Scherzer, Steven D. Knight, Johnson Neil W, William H. Miller, Elsie Diaz, Seth W. Grant, Art Shu, Heidi Morgan-Ott, and Joelle Lorraine Burgess
- Subjects
Cancer Research ,EZH2 ,Cancer ,Biology ,medicine.disease ,Small molecule ,Oncology ,Structural biology ,Biochemistry ,Histone methyltransferase ,Gene expression ,Histone methylation ,Cancer research ,medicine ,Cancer epigenetics - Abstract
The histone methyltransferases are a group of enzymes which catalyze the transfer of a methyl group from the co-factor S-Adenosylmethionine (SAM) to the lysine and arginine residues of histone tails. This post-translational modification is a key event in maintaining gene expression patterns. In recent years, the relationships between aberrant histone methylation patterns and cancer progression have been recognized. These developments, along with an improved understanding of the underlying structural biology, have made histone methyltransferases highly attractive targets for therapeutic intervention. The histone lysine methyltransferase EZH2 (Enhancer of Zeste Homolog 2) is frequently over-expressed in a wide variety of cancerous tissues. There is a strong correlation between overexpression of EZH2 and aberrant transcriptional signaling in cells, ultimately resulting in poor clinical prognosis. Inhibition of EZH2 is expected to alter transcriptional expression and ultimately lead to an improved clinical outcome. This presentation will describe medicinal chemistry efforts in the development of highly potent and selective small molecule inhibitors of EZH2. The synthesis, SAR, and identification of a clinical candidate will be discussed. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 2939. doi:1538-7445.AM2012-2939
- Published
- 2012
50. Abstract 1057: Mutation of EZH2 A677 in human B-cell lymphoma promotes hyper-trimethylation of H3K27
- Author
-
Yong Jiang, Caretha L. Creasy, Edward Dul, Sharad K. Verma, Benjamin Schwartz, Ryan G. Kruger, Heidi M. Ott, Michael T. McCabe, Gopinath Ganji, Peter J. Tummino, Kimberly N. Smitheman, Wendy S. Halsey, Ashley M. Hughes, Alan P. Graves, Elsie Diaz, Stephanie Chen, Melissa B. Pappalardi, Anthony Della-Pietra, Kimberly E. Allen, Sara H. Thrall, and Martin Brandt
- Subjects
Cancer Research ,Mutation ,biology ,EZH2 ,Mutant ,macromolecular substances ,Methylation ,medicine.disease_cause ,Histone H3 ,Germline mutation ,Oncology ,Histone methyltransferase ,biology.protein ,Cancer research ,medicine ,PRC2 - Abstract
Trimethylation of histone H3 on lysine 27 (H3K27me3) is a repressive post-translational modification mediated by the histone methyltransferase EZH2. EZH2 is a component of the Polycomb Repressive Complex 2 (PRC2) and its expression and catalytic activity are dysregulated in cancers. While EZH2 may be over-expressed as a result of multiple mechanisms in tumors, only somatic mutation of the EZH2 Y641 residue has thus far been reported to alter its substrate preference and enhance its catalytic efficiency to generate H3K27me3. Herein, we report mutation of the A677 residue of EZH2 to a glycine (A677G) in a lymphoma cell line with aberrantly elevated H3K27me3 levels. Additional EZH2 sequence analysis in 41 primary lymphoma specimens identified another occurrence of this mutation. Biochemical evaluation of recombinant EZH2 complexes revealed that A677G EZH2 possesses catalytic activity with substrate specificity that is novel and distinct from those of wild-type and Y641 mutants. Whereas wild-type EZH2 displayed a preference for substrates with less methylation (i.e. H3K27me0>me1>me2), the Y641 mutants exhibited greatly decreased activity with H3K27me0 and increased activity with H3K27me2. The A677G EZH2, on the other hand, exhibited nearly equal efficiency for all three substrates. A677G EZH2, but not wild-type EZH2, was shown to be capable of significantly increasing global H3K27me3 when transiently expressed in an EZH2 wild-type cancer cell line. Finally, structural modeling suggests that the mutation results in a larger lysine tunnel capable of accommodating the H3K27me2 substrate while retaining the ability to properly orient H3K27me0 and H3K27me1 with the Y641 residue. In addition, functional and biochemical analyses are performed with reversible SAM-competitive EZH2 inhibitors. Therefore, this mutation appears to contribute to the aberrant epigenetic profile observed in certain lymphomas. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 1057. doi:1538-7445.AM2012-1057
- Published
- 2012
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.