102 results on '"Keilhack H"'
Search Results
2. The PARP14 inhibitor RBN-3143 suppresses lung inflammation in preclinical models
- Author
-
Niepel, M, primary, Vasbinder, M, additional, Molina, J, additional, Gui, B, additional, Kunii, K, additional, Blackwell, D, additional, Mateer, E, additional, Mo, J, additional, Coutts, C, additional, Perl, N, additional, Kuplast-Barr, K, additional, Church, W, additional, Majer, C, additional, Schenkel, L B, additional, Swinger, K, additional, Abo, R, additional, Bamberg, E, additional, Ren, Y, additional, Utley, L, additional, Wigle, T, additional, Reik, C, additional, Lu, A, additional, Cheung, A, additional, Minissale, E, additional, Mceachern, K, additional, Richon, V, additional, Parasuraman, S, additional, Kuntz, K, additional, and Keilhack, H, additional
- Published
- 2022
- Full Text
- View/download PDF
3. Cytogenetic examination of sorted CD34-positive cells as a possible tool to differentiate hypoplastic MDS from aplastic anemia. A case report: P679
- Author
-
Pönisch, W., Leiblein, S., Al-Ali, H. K., Friedrich, T., Lange, T., Müller, C., Pless, M., Hennig, E., Keilhack, H., Krahl, R., and Niederwieser, D.
- Published
- 2003
4. Das Verhalten der Serumeiweißkörper unter verschiedenen Stoffwechselbedingungen
- Author
-
Keilhack, H.
- Published
- 1935
- Full Text
- View/download PDF
5. Morphologie
- Author
-
Arzt, Hellner, Schlüter, Annelise, Rintelen, K., Finckh, E., Schütz, Gutzeit, R., Schröpl, E., Dietel, F., Beyer, A., Ostertag, Krepuska, G., St. Krepuska, Gaus, Böhmig, Zint, Camillo, Nissen, Karl, Klages, F., Dykstra, Jacob, Noetzel, Peters, Gerd, Matzdorff, Klauber, F., Sørensen, Biondi, Liguori-Hohenauer, Jaensch, P. A., Schultz, Werner, Rolleri, Felicitas, Wirth, Erich, Sørensen, Franz, Krah, Müller, Barbara, Merten, Orzechowski, Wüsthoff, Bansi, Reinhardt, Lüdin, Otto, Videbech, Fenster, Schmitz, A., Mark, Robert E., Zavka, Pena, A., Krieg, Eberhard, v. Knorre, Beyer, Ruth, Iljin, Ickert, Lossen, Heinz, de Rochemont, du Mesnil, Junghanns, Krah, Norpoth, L., Biedermann, W., Keilhack, H., Sandera, Isola, Stodtmeister, Kornmann, J., Bergerhoff, Reiser, B., Goebel, Eggert, Pohlner, Peiper, A., Breuer, H., Brückmann, Herold, Winkler, H., Huber, Volk, Dietrich, W., Strakosch, Froboese, Wobker, Nothdurft, Malcovati, Gál, Felix, Müller, H. D., and Heidler, Hans
- Published
- 1940
- Full Text
- View/download PDF
6. Strittige geschlechtliche Verhältnisse. Sexualpathologie. Sexualdelikte
- Author
-
van Oordt, Dubitscher, Manz, Brugger, Günther, Fritz-Hölder, Helene, Keilhack, H., v. Neureiter, Kresiment, and Ganter
- Published
- 1939
- Full Text
- View/download PDF
7. Pathologische Anatomie (Sektionstechnik) und Physiologie
- Author
-
Fischer, W., Peters, Gerd, Eversbusch, Jacob, Tobler, Tilk, Keilhack, H., Vogeler, Oeser, H., Steffel, Weimann, Merkel, Warstadt, Arno, Koch, Rudolf, Schiersmann, Janzen, R., Magnussen, Ostertag, König, Voss, Pohlandt, Demuth, Bremner, Pönitz, Schlossberger, Reinhardt, and Gross, H.
- Published
- 1942
- Full Text
- View/download PDF
8. Pathologische Anatomie (Sektionstechnik) und Physiologie
- Author
-
Koch, Rudolf, Inouye, T., Gerstel, Kluge, Meixner, Kinzl, Merkel, H., Wasmuht, Klaus, Fresen, Förster, Opitz, Erich, Holtzmann, Matzdorff, Walcher, Gregora, Keilhack, H., Haagen, Oeser, Naegeli, Th., Geller, Wiethold, Mueller, B., Spiecker, and Nohara
- Published
- 1941
- Full Text
- View/download PDF
9. A Medium-Throughput Single Cell CRISPR-Cas9 Assay to Assess Gene Essentiality
- Author
-
Grassian, A. R., primary, Scales, T. M. E., additional, Knutson, S. K., additional, Kuntz, K. W., additional, McCarthy, N. J., additional, Lowe, C. E., additional, Moore, J. D., additional, Copeland, R. A., additional, Keilhack, H., additional, Smith, J. J., additional, Wickenden, J. A., additional, and Ribich, S., additional
- Published
- 2015
- Full Text
- View/download PDF
10. 302 A phase 1 study of EPZ-6438 (E7438), an Enhancer of Zeste-Homolog 2 (EZH2) inhibitor: Preliminary activity in INI1-negative tumors
- Author
-
Italiano, A., primary, Keilhack, H., additional, Toulmonde, M., additional, Coindre, J.M., additional, Michot, J.M., additional, Massard, C., additional, Ottesen, L., additional, Reyderman, L., additional, Blakemore, S.J., additional, Kraljevic, S., additional, Thomson, B., additional, McDonald, A., additional, Ho, P.T., additional, and Ribrag, V., additional
- Published
- 2015
- Full Text
- View/download PDF
11. Phase 1 first-in-human study of the enhancer of zeste-homolog 2 (EZH2) histone methyl transferase inhibitor E7438
- Author
-
Ribrag, V., primary, Soria, J.-C., additional, Reyderman, L., additional, Chen, R., additional, Salazar, P., additional, Kumar, N., additional, Kuznetsov, G., additional, Keilhack, H., additional, Ottesen, L.H., additional, and Italiano, A., additional
- Published
- 2015
- Full Text
- View/download PDF
12. Sonstige Fernwirkungen
- Author
-
Bauke, E. -E., Fresen, Müller, H. D., Keysselitz, Keilhack, H., and Regele
- Published
- 1944
- Full Text
- View/download PDF
13. Magen
- Author
-
Keilhack, H., Hummel, R., Beyer, A., Velde, G., Sponholz, and Schlüter, Annelise
- Published
- 1941
- Full Text
- View/download PDF
14. Reticuloendotheliomatosen: Morbus Hodgkin, Leukaemie usw
- Author
-
Carere-Comes, O., Mende, Stodtmeister, Schultz, Werner, Keilhack, H., and Scholz, H.
- Published
- 1944
- Full Text
- View/download PDF
15. Sonstige Fernwirkungen
- Author
-
Keilhack, H., Isola, Pendl, E., Sörensen, Franz, Schlomka, and Jores
- Published
- 1944
- Full Text
- View/download PDF
16. 6LBA Phase 1 first-in-human study of the enhancer of zeste-homolog 2 (EZH2) histone methyl transferase inhibitor E7438 as a single agent in patients with advanced solid tumors or B cell lymphoma
- Author
-
Ribrag, V., primary, Soria, J.C., additional, Reyderman, L., additional, Chen, R., additional, Salazar, P., additional, Kumar, N., additional, Kuznetsov, G., additional, Keilhack, H., additional, Ottesen, L.H., additional, and Italiano, A., additional
- Published
- 2014
- Full Text
- View/download PDF
17. 277 The discovery and pre-clinical development of the first clinical stage EZH2-inhibitor, EPZ-6438 (E7438)
- Author
-
Kuntz, K., primary, Keilhack, H., additional, Pollock, R., additional, Knutson, S., additional, Warholic, N., additional, Richon, V., additional, Chesworth, R., additional, Copeland, R., additional, Porter-Scott, M., additional, Sneeringer, C., additional, and Wigle, T., additional
- Published
- 2014
- Full Text
- View/download PDF
18. Negative regulation of Ros receptor tyrosine kinase signaling: An epithelial function of the SH2 domain protein tyrosine phosphatase SHP-1
- Author
-
Keilhack, H., Mueller, M., Boehmer, S.A., Frank, C., Weidner, K.M., Birchmeier, W., Ligensa, T., Berndt, A., Kosmehl, H., Gunther, B., Mueller, T., Birchmeier, C., and Boehmer, F.D.
- Subjects
Cancer Research ,animal structures ,embryonic structures ,chemical and pharmacologic phenomena ,hemic and immune systems ,biological phenomena, cell phenomena, and immunity - Abstract
Male "viable motheaten" (me(v)) mice, with a naturally occurring mutation in the gene of the SH2 domain protein tyrosine phosphatase SHP-1, are sterile. Known defects in sperm maturation in these mice correlate with an impaired differentiation of the epididymis, which has similarities to the phenotype of mice with a targeted inactivation of the Ros receptor tyrosine kinase. Ros and SHP-1 are coexpressed in epididymal epithelium, and elevated phosphorylation of Ros in the epididymis of me(v) mice suggests that Ros signaling is under control of SHP-1 in vivo. Phosphorylated Ros strongly and directly associates with SHP-1 in yeast two-hybrid, glutathione S-transferase pull-down, and coimmunoprecipitation experiments. Strong binding of SHP-1 to Ros is selective compared to six other receptor tyrosine kinases. The interaction is mediated by the SHP-1 NH(2)-terminal SH2 domain and Ros phosphotyrosine 2267. Overexpression of SHP-1 results in Ros dephosphorylation and effectively downregulates Ros-dependent proliferation and transformation. We propose that SHP-1 is an important downstream regulator of Ros signaling.
- Published
- 2001
19. Preclinical Characterization of E7438, a Potent, Selective EZH2 Inhibitor with Robust Antitumor Activity Against EZH2 Mutated NHL Xenografts in Mice
- Author
-
Keilhack, H., primary
- Published
- 2013
- Full Text
- View/download PDF
20. Lectures
- Author
-
Chen, D. S., primary, Feltquate, D. M., additional, Smothers, F., additional, Hoos, A., additional, Langermann, S., additional, Marshall, S., additional, May, R., additional, Fleming, M., additional, Hodi, F. S., additional, Senderowicz, A., additional, Wiman, K. G., additional, de Dosso, S., additional, Fiedler, W., additional, Gianni, L., additional, Cresta, S., additional, Schulze-Bergkamen, H. B., additional, Gurrieri, L., additional, Salzberg, M., additional, Dietrich, B., additional, Danielczyk, A., additional, Baumeister, H., additional, Goletz, S., additional, Sessa, C., additional, Strumberg, D., additional, Schultheis, B., additional, Santel, A., additional, Gebhardt, F., additional, Meyer-Sabellek, W., additional, Keil, O., additional, Giese, K., additional, Kaufmann, J., additional, Maio, M., additional, Choy, G., additional, Covre, A., additional, Parisi, G., additional, Nicolay, H., additional, Fratta, E., additional, Fonsatti, E., additional, Sigalotti, L., additional, Coral, S., additional, Taverna, P., additional, Azab, M., additional, Deutsch, E., additional, Lepechoux, C., additional, Pignon, J. P., additional, Tao, Y. T., additional, Rivera, S., additional, Bourgier, B. C., additional, Angokai, M., additional, Bahleda, R., additional, Slimane, K., additional, Angevin, E., additional, Besse, B. B., additional, Soria, J. C., additional, Dragnev, K., additional, Beumer, J. H., additional, Anyang, B., additional, Ma, T., additional, Galimberti, F., additional, Erkmen, C. P., additional, Nugent, W., additional, Rigas, J., additional, Abraham, K., additional, Johnstone, D., additional, Memoli, V., additional, Dmitrovsky, E., additional, Voest, E. E., additional, Siu, L., additional, Janku, F., additional, Tsimberidou, A., additional, Kurzrock, R., additional, Tabernero, J., additional, Rodon, J., additional, Berger, R., additional, Onn, A., additional, Batist, G., additional, Bresson, C., additional, Lazar, V., additional, Molenaar, J. J., additional, Koster, J., additional, Ebus, M., additional, Zwijnenburg, D. A., additional, van Sluis, P., additional, Lamers, F., additional, Schild, L., additional, van der Ploeg, I., additional, Caron, H. N., additional, Versteeg, R., additional, Pouyssegur, J., additional, Marchiq, I., additional, Chiche, J., additional, Roux, D., additional, Le Floch, R., additional, Critchlow, S. E., additional, Wooster, R. F., additional, Agresta, S., additional, Yen, K. E., additional, Janne, P. A., additional, Plummer, E. R., additional, Trinchieri, G., additional, Ellis, L., additional, Chan, S. L., additional, Yeo, W., additional, Chan, A. T., additional, Mouliere, F., additional, El Messaoudi, S., additional, Gongora, C., additional, Lamy, P. J., additional, del Rio, M., additional, Lopez-Crapez, E., additional, Gillet, B., additional, Mathonnet, M., additional, Pezet, D., additional, Ychou, M., additional, Thierry, A. R., additional, Ribrag, V., additional, Vainchenker, W., additional, Constantinescu, S., additional, Keilhack, H., additional, Umelo, I. A., additional, Noeparast, A., additional, Chen, G., additional, Renard, M., additional, Geers, C., additional, Vansteenkiste, J., additional, Teugels, E., additional, de Greve, J., additional, Rixe, O., additional, Qi, X., additional, Chu, Z., additional, Celerier, J., additional, Leconte, L., additional, Minet, N., additional, Pakradouni, J., additional, Kaur, B., additional, Cuttitta, F., additional, Wagner, A. J., additional, Zhang, Y. X., additional, Sicinska, E., additional, Czaplinski, J. T., additional, Remillard, S. P., additional, Demetri, G. D., additional, Weng, S., additional, Debussche, L., additional, Agoni, L., additional, Reddy, E. P., additional, Guha, C., additional, Silence, K., additional, Thibault, A., additional, de Haard, H., additional, Dreier, T., additional, Ulrichts, P., additional, Moshir, M., additional, Gabriels, S., additional, Luo, J., additional, Carter, C., additional, Rajan, A., additional, Khozin, S., additional, Thomas, A., additional, Lopez-Chavez, A., additional, Brzezniak, C., additional, Doyle, L., additional, Keen, C., additional, Manu, M., additional, Raffeld, M., additional, Giaccone, G., additional, Lutzker, S., additional, Melief, J. M., additional, Eckhardt, S. G., additional, Trusolino, L., additional, Migliardi, G., additional, Zanella, E. R., additional, Cottino, F., additional, Galimi, F., additional, Sassi, F., additional, Marsoni, S., additional, Comoglio, P. M., additional, Bertotti, A., additional, Hidalgo, M., additional, Weroha, S. J., additional, Haluska, P., additional, Becker, M. A., additional, Harrington, S. C., additional, Goodman, K. M., additional, Gonzalez, S. E., additional, al Hilli, M., additional, Butler, K. A., additional, Kalli, K. R., additional, Oberg, A. L., additional, Huijbers, I. J., additional, Bin Ali, R., additional, Pritchard, C., additional, Cozijnsen, M., additional, Proost, N., additional, Song, J. Y., additional, Krimpenfort, P., additional, Michalak, E., additional, Jonkers, J., additional, Berns, A., additional, Banerji, U., additional, Stewart, A., additional, Thavasu, P., additional, Banerjee, S., additional, and Kaye, S. B., additional
- Published
- 2013
- Full Text
- View/download PDF
21. Final results of a translational phase l study assessing a QOD schedule of the potent AKT inhibitor MK-2206 incorporating predictive, pharmacodynamic (PD), and functional imaging biomarkers.
- Author
-
Yap, T. A., primary, Yan, L., additional, Patnaik, A., additional, Olmos, D., additional, Fearen, I., additional, Baird, R. D., additional, Papadopoulos, K. P., additional, Tunariu, N., additional, Biondo, A., additional, Keilhack, H., additional, Delgado, L. M., additional, Taylor, A., additional, Blackman, S. C., additional, Carpenter, C. L., additional, Decordova, S., additional, Heaton, S., additional, Garrett, M. D., additional, Sullivan, D., additional, De Bono, J. S., additional, and Tolcher, A. W., additional
- Published
- 2011
- Full Text
- View/download PDF
22. Metastasen
- Author
-
Schmidt, Hans W., Keilhack, H., and Wolfram
- Published
- 1944
- Full Text
- View/download PDF
23. Buchbesprechungen
- Author
-
von Lanz, Lehnartz, Ulrici, Keilhack, H., Fischer, A. W., Curschmann, H., Schulten, and Mader
- Published
- 1940
- Full Text
- View/download PDF
24. Untersuchungen über eine rationelle Dauertherapie der perniziösen Anämie.
- Author
-
Henning, N. and Keilhack, H.
- Published
- 1936
- Full Text
- View/download PDF
25. Strittige geschlechtliche Verhältnisse. Sexualpathologie. Sexualdelikte.
- Author
-
Oordt, Dubitscher, Manz, Brugger, Günther, Fritz-Hölder, Helene, Keilhack, H., Neureiter, Kresiment, and Ganter
- Published
- 1940
- Full Text
- View/download PDF
26. Identification of Src phosphorylation sites in the catenin p120ctn.
- Author
-
Mariner, D J, Anastasiadis, P, Keilhack, H, Böhmer, F D, Wang, J, and Reynolds, A B
- Abstract
p120-catenin (p120(ctn)) interacts with the cytoplasmic tail of cadherins and is thought to regulate cadherin clustering during formation of adherens junctions. Several observations suggest that p120 can both positively and negatively regulate cadherin adhesiveness depending on signals that so far remain unidentified. Although p120 tyrosine phosphorylation is a leading candidate, the role of this modification in normal and Src-transformed cells remains unknown. Here, as a first step toward pinpointing this role, we have employed two-dimensional tryptic mapping to directly identify the major sites of Src-induced p120 phosphorylation. Eight sites were identified by direct mutation of candidate tyrosines to phenylalanine and elimination of the accompanying spots on the two-dimensional maps. Identical sites were observed in vitro and in vivo, strongly suggesting that the physiologically important sites have been correctly identified. Changing all of these sites to phenylalanine resulted in a p120 mutant, p120-8F, that could not be efficiently phosphorylated by Src and failed to interact with SHP-1, a tyrosine phosphatase shown previously to interact selectively with tyrosine-phosphorylated p120 in cells stimulated with epidermal growth factor. Using selected tyrosine to phenylalanine p120 mutants as dominant negative reagents, it may now be possible to selectively block events postulated to be dependent on p120 tyrosine phosphorylation.
- Published
- 2001
- Full Text
- View/download PDF
27. The protein-tyrosine phosphatase SHP-1 binds to and dephosphorylates p120 catenin.
- Author
-
Keilhack, H, Hellman, U, van Hengel, J, van Roy, F, Godovac-Zimmermann, J, and Böhmer, F D
- Abstract
A prominent tyrosine-phosphorylated protein of approximately 100 kDa (designated pp100) in epidermal growth factor (EGF)-stimulated A431 cells was found to be a main interaction partner of the protein-tyrosine phosphatase SHP-1 in pull-down experiments with a glutathione S-transferase-SHP-1 fusion protein. Binding was largely mediated by the N-terminal SH2 domain of SHP-1 and apparently direct and independent from the previously described association of SHP-1 with the activated EGF receptor. pp100 was partially purified and identified by mass spectrometric analysis of tryptic fragments, partial amino acid sequencing, and use of authentic antibodies as the 3A isoform of the Armadillo repeat protein superfamily member p120 catenin (p120(ctn)). Different p120(ctn) isoforms expressed in human embryonal kidney 293 cells, exhibited differential binding to SHP-1 that correlated partly with the extent of EGF-dependent p120(ctn) tyrosine phosphorylation. Despite strong phosphorylation, p120(ctn) isoforms 3B and 3AB bound, however, less readily to SHP-1. SHP-1 associated transiently with p120(ctn) in EGF-stimulated A431 cells stably transfected with a tetracycline-responsive SHP-1 expression construct, and p120(ctn) exhibited elevated phosphorylation upon a tetracycline-mediated decrease in the SHP-1 level. Functions of p120(ctn), which are regulated by tyrosine phosphorylation, may be modulated by the described SHP-1-p120(ctn) interaction.
- Published
- 2000
- Full Text
- View/download PDF
28. Both SH2 domains are involved in interaction of SHP-1 with the epidermal growth factor receptor but cannot confer receptor-directed activity to SHP-1/SHP-2 chimera.
- Author
-
Tenev, T, Keilhack, H, Tomic, S, Stoyanov, B, Stein-Gerlach, M, Lammers, R, Krivtsov, A V, Ullrich, A, and Böhmer, F D
- Abstract
The previously demonstrated functional and physical interaction of the SH2 domain protein-tyrosine phosphatase SHP-1 with the epidermal growth factor (EGF) receptor (Tomic, S., Greiser, U., Lammers, R., Kharitonenkov, A., Imyanitov, E., Ullrich, A., and Böhmer, F. D. (1995) J. Biol. Chem. 270, 21277-21284) was investigated with respect to the involved structural elements of SHP-1. Various mutants of SHP-1 were transiently expressed in 293 or COS-7 cells and analyzed for their capacity to associate with immobilized autophosphorylated EGF receptor in vitro and to dephosphorylate coexpressed EGF receptor in intact cells. Inactivating point mutation of the C-terminal SH2 domain reduced the association weakly, point mutation of the N-terminal SH2 domain reduced association strongly and the respective double mutation abolished association totally. The capacity of SHP-1 to dephosphorylate coexpressed EGF receptor was impaired by all point mutations. Truncation of the N-terminal or of both SH2 domains strongly reduced or abolished association, respectively, but the truncated SHP-1 derivatives still dephosphorylated coexpressed EGF receptor effectively. Various chimeric protein-tyrosine phosphatases constructed from SHP-1 and the closely homologous SHP-2 dephosphorylated the EGF receptor when they contained the catalytic domain of SHP-1. As native SHP-2, the chimera lacked activity toward the receptor when they contained the catalytic domain of SHP-2, despite their capacity to associate with the receptor and to dephosphorylate an artificial phosphopeptide. We conclude that the differential interaction of SHP-1 and SHP-2 with the EGF receptor is due to the specificity of the respective catalytic domains rather than to the specificity of the SH2 domains. Functional interaction of native SHP-1 with the EGF receptor requires association mediated by both SH2 domains.
- Published
- 1997
29. Phosphotyrosine 1173 mediates binding of the protein-tyrosine phosphatase SHP-1 to the epidermal growth factor receptor and attenuation of receptor signaling.
- Author
-
Keilhack, H, Tenev, T, Nyakatura, E, Godovac-Zimmermann, J, Nielsen, L, Seedorf, K, and Böhmer, F D
- Abstract
The protein-tyrosine phosphatase SHP-1 binds to and dephosphorylates the epidermal growth factor receptor (EGFR), and both SH2 domains of SHP-1 are important for this interaction (Tenev, T., Keilhack, H., Tomic, S., Stoyanov, B., Stein-Gerlach, M., Lammers, R., Krivtsov, A. V., Ullrich, A., and Böhmer, F. D. (1997) J. Biol. Chem. 272, 5966-5973). We mapped the EGFR phosphotyrosine 1173 as the major binding site for SHP-1 by a combination of phosphopeptide activation, phosphopeptide competition, and receptor YF mutant analysis. Mutational conversion of the EGFR sequence 1171-1176 AEYLRV into the high affinity SHP-1 binding sequence LEYLYL of the erythropoietin receptor (EpoR) led to a highly elevated SHP-1 binding to the mutant EGFR (EGFR1171-1176EpoR) and in turn to an enhanced dephosphorylation of the receptor. SHP-1 expression interfered with EGF-dependent mitogen-activated protein kinase stimulation, and this effect was more pronounced in case of EGFR1171-1176EpoR. Reduced SHP-1 binding to the EGFR Y1173F mutant resulted in a reduced receptor dephosphorylation by coexpressed SHP-1 and less interference with EGF-dependent mitogen-activated protein kinase stimulation. The effects of receptor mutations on SHP-1 binding were, however, stronger than those on receptor dephosphorylation by SHP-1. Therefore, receptor dephosphorylation may be the result of the combined activity of receptor-bound SHP-1 and SHP-1 bound to an auxiliary docking protein.
- Published
- 1998
30. Strittige geschlechtliche Verhältnisse. Sexualpathologie. Sexualdelikte
- Author
-
Oordt, van, Dubitscher, Manz, Brugger, Günther, Fritz-Hölder, Helene, Keilhack, H., Neureiter, v., Kresiment, and Ganter
- Published
- 1939
- Full Text
- View/download PDF
31. Reticuloendotheliomatosen.
- Author
-
Carere-Comes, O., Mende, Stodtmeister, Schultz, Werner, Keilhack, H., and Scholz, H.
- Published
- 1944
- Full Text
- View/download PDF
32. Untersuchungen über eine rationelle Dauertherapie der perniziösen Anämie
- Author
-
Henning, N., primary and Keilhack, H., additional
- Published
- 1936
- Full Text
- View/download PDF
33. Über den Nachweis von basophilgetüpfelten Erythrozyten im Sternalpunktat bei Bleivergiftung
- Author
-
Henning, N., primary and Keilhack, H., additional
- Published
- 1940
- Full Text
- View/download PDF
34. Sonstige Fernwirkungen
- Author
-
Bauke, E. -E., Fresen, Müller, H. D., Keysselitz, Keilhack, H., and Regele
- Published
- 1943
- Full Text
- View/download PDF
35. Buchbesprechungen
- Author
-
Lanz, von, Lehnartz, Ulrici, Keilhack, H., Fischer, A. W., Curschmann, H., Schulten, and Mader
- Published
- 1940
- Full Text
- View/download PDF
36. Buchbesprechungen.
- Author
-
Lanz, Lehnartz, Ulrici, Keilhack, H., Fischer, A., Curschmann, H., Schulten, and Mader
- Published
- 1940
- Full Text
- View/download PDF
37. O7:2 PHASE 1 FIRST-IN-HUMAN STUDY OF THE ENHANCER OF ZESTE-HOMOLOG 2 (EZH2) HISTONE METHYL TRANSFERASE INHIBITOR E7438
- Author
-
Ribrag, V., Soria, J.-C., Reyderman, L., Chen, R., Salazar, P., Kumar, N., Kuznetsov, G., Keilhack, H., Ottesen, L. H., and Italiano, A.
- Published
- 2015
- Full Text
- View/download PDF
38. Selective Pharmaceutical Inhibition of PARP14 Mitigates Allergen-Induced IgE and Mucus Overproduction in a Mouse Model of Pulmonary Allergic Response.
- Author
-
Eddie AM, Chen KW, Schenkel LB, Swinger KK, Molina JR, Kunii K, Raybuck AL, Keilhack H, Gibson-Corley KN, Niepel M, Peebles RS, Boothby MR, and Cho SH
- Subjects
- Animals, Disease Models, Animal, Immunoglobulin E, Mice, Mucus metabolism, Pharmaceutical Preparations metabolism, Poly(ADP-ribose) Polymerase Inhibitors metabolism, Poly(ADP-ribose) Polymerase Inhibitors pharmacology, Poly(ADP-ribose) Polymerase Inhibitors therapeutic use, Poly(ADP-ribose) Polymerases genetics, Poly(ADP-ribose) Polymerases metabolism, Poly(ADP-ribose) Polymerases therapeutic use, Allergens, Asthma drug therapy
- Abstract
The type 2 cytokines IL-4 and IL-13, which share use of an IL-4 receptor α-chain and its nuclear induction of the transcription factor STAT6, are crucial in elicitation and maintenance of allergic conditions including asthma. STAT6 binds poly(ADP-ribose) polymerase (PARP)14, an ADP-ribosyl monotransferase. Elimination of PARP14 by gene targeting led to attenuation of OVA-specific allergic lung inflammation. However, PARP14 has multiple functional domains apart from the portion that catalyzes ADP-ribosylation, and it is not clear whether inhibition of the catalytic function has any biological consequence. Using BALB/c mice sensitized to the allergen Alternaria alternata, we show that peroral administration of RBN012759, a highly selective inhibitor of ADP-ribosylation by PARP14 with negligible impact on other members of the PARP gene family, achieved biologically active plasma concentrations and altered several responses to the Ag. Specifically, the pharmaceutical compound decreased mucus after allergen challenge, blunted the induced increases in circulating IgE, and prevented suppression of IgG2a. We conclude that PARP14 catalytic activity can contribute to pathogenesis in allergic or atopic processes and propose that other biological endpoints dependent on ADP-ribosylation by PARP14 can be targeted using selective inhibition., (Copyright © 2022 The Authors.)
- Published
- 2022
- Full Text
- View/download PDF
39. PARP7 negatively regulates the type I interferon response in cancer cells and its inhibition triggers antitumor immunity.
- Author
-
Gozgit JM, Vasbinder MM, Abo RP, Kunii K, Kuplast-Barr KG, Gui B, Lu AZ, Molina JR, Minissale E, Swinger KK, Wigle TJ, Blackwell DJ, Majer CR, Ren Y, Niepel M, Varsamis ZA, Nayak SP, Bamberg E, Mo JR, Church WD, Mady ASA, Song J, Utley L, Rao PE, Mitchison TJ, Kuntz KW, Richon VM, and Keilhack H
- Subjects
- Adaptive Immunity drug effects, Animals, Cell Line, Tumor, Cell Proliferation drug effects, Cell Survival drug effects, HEK293 Cells, HeLa Cells, Humans, Mice, Neoplasms genetics, Neoplasms metabolism, Signal Transduction drug effects, Small Molecule Libraries pharmacology, Tumor Escape drug effects, Xenograft Model Antitumor Assays, Drug Resistance, Neoplasm drug effects, Interferon Type I metabolism, Neoplasms drug therapy, Nucleoside Transport Proteins genetics, Nucleoside Transport Proteins metabolism, Small Molecule Libraries administration & dosage
- Abstract
PARP7 is a monoPARP that catalyzes the transfer of single units of ADP-ribose onto substrates to change their function. Here, we identify PARP7 as a negative regulator of nucleic acid sensing in tumor cells. Inhibition of PARP7 restores type I interferon (IFN) signaling responses to nucleic acids in tumor models. Restored signaling can directly inhibit cell proliferation and activate the immune system, both of which contribute to tumor regression. Oral dosing of the PARP7 small-molecule inhibitor, RBN-2397, results in complete tumor regression in a lung cancer xenograft and induces tumor-specific adaptive immune memory in an immunocompetent mouse cancer model, dependent on inducing type I IFN signaling in tumor cells. PARP7 is a therapeutic target whose inhibition induces both cancer cell-autonomous and immune stimulatory effects via enhanced IFN signaling. These data support the targeting of a monoPARP in cancer and introduce a potent and selective PARP7 inhibitor to enter clinical development., Competing Interests: Declaration of interests J.M.G., M.M.V., R.P.A., K.K., K.G.K.-B., B.G., A.Z.L., J.R. Molina, E.M., K.K.S., T.J.W., D.J.B., C.R.M., Y.R., M.N., Z.A.V., S.P.N., E.B., J.-R. Mo, W.D.C., A.S.A.M., J.S., L.U., K.W.K., V.M.R., and H.K. are all employees and shareholders of Ribon Therapeutics at the time of data collection. P.E.R. served as a consultant to Ribon Therapeutics, and T.J.M. is a founder and shareholder in Ribon Therapeutics. M.M.V., K.K.S., and K.W.K. are inventors on US Patent Nos. 10550105, 10870641, and 11014913, which are related to RBN-2397 and assigned to Ribon Therapeutics., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
40. A potent and selective PARP14 inhibitor decreases protumor macrophage gene expression and elicits inflammatory responses in tumor explants.
- Author
-
Schenkel LB, Molina JR, Swinger KK, Abo R, Blackwell DJ, Lu AZ, Cheung AE, Church WD, Kunii K, Kuplast-Barr KG, Majer CR, Minissale E, Mo JR, Niepel M, Reik C, Ren Y, Vasbinder MM, Wigle TJ, Richon VM, Keilhack H, and Kuntz KW
- Subjects
- Animals, Antineoplastic Agents chemical synthesis, Antineoplastic Agents chemistry, Dose-Response Relationship, Drug, Female, Gene Expression Regulation drug effects, Gene Expression Regulation genetics, HEK293 Cells, Humans, Inflammation genetics, Inflammation metabolism, Interleukin-4 genetics, Kidney Neoplasms genetics, Kidney Neoplasms metabolism, Macrophages metabolism, Male, Mice, Mice, Inbred C57BL, Models, Molecular, Molecular Structure, Poly(ADP-ribose) Polymerases genetics, RAW 264.7 Cells, RNA, Messenger drug effects, RNA, Messenger genetics, RNA, Messenger metabolism, Antineoplastic Agents pharmacology, Inflammation drug therapy, Interleukin-4 antagonists & inhibitors, Kidney Neoplasms drug therapy, Macrophages drug effects, Poly(ADP-ribose) Polymerases metabolism
- Abstract
PARP14 has been implicated by genetic knockout studies to promote protumor macrophage polarization and suppress the antitumor inflammatory response due to its role in modulating interleukin-4 (IL-4) and interferon-γ signaling pathways. Here, we describe structure-based design efforts leading to the discovery of a potent and highly selective PARP14 chemical probe. RBN012759 inhibits PARP14 with a biochemical half-maximal inhibitory concentration of 0.003 μM, exhibits >300-fold selectivity over all PARP family members, and its profile enables further study of PARP14 biology and disease association both in vitro and in vivo. Inhibition of PARP14 with RBN012759 reverses IL-4-driven protumor gene expression in macrophages and induces an inflammatory mRNA signature similar to that induced by immune checkpoint inhibitor therapy in primary human tumor explants. These data support an immune suppressive role of PARP14 in tumors and suggest potential utility of PARP14 inhibitors in the treatment of cancer., Competing Interests: Declaration of interests L.B.S., K.K.S., M.M.V., and K.W.K. are co-inventors on pending patent application WO 2019/126443 and issued U.S. Pat. No. 10,562,891, both of which relate to quinazolinone inhibitors of PARP14 for the treatment of cancer and other diseases and are assigned to Ribon Therapeutics, Inc., of Cambridge, MA, USA., (Copyright © 2021 Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
41. Targeted Degradation of PARP14 Using a Heterobifunctional Small Molecule.
- Author
-
Wigle TJ, Ren Y, Molina JR, Blackwell DJ, Schenkel LB, Swinger KK, Kuplast-Barr K, Majer CR, Church WD, Lu AZ, Mo J, Abo R, Cheung A, Dorsey BW, Niepel M, Perl NR, Vasbinder MM, Keilhack H, and Kuntz KW
- Subjects
- Humans, Macrophages drug effects, Macrophages metabolism, Molecular Structure, Small Molecule Libraries chemical synthesis, Small Molecule Libraries chemistry, Poly(ADP-ribose) Polymerases metabolism, Small Molecule Libraries pharmacology
- Abstract
PARP14 is an interferon-stimulated gene that is overexpressed in multiple tumor types, influencing pro-tumor macrophage polarization as well as suppressing the antitumor inflammation response by modulating IFN-γ and IL-4 signaling. PARP14 is a 203 kDa protein that possesses a catalytic domain responsible for the transfer of mono-ADP-ribose to its substrates. PARP14 also contains three macrodomains and a WWE domain which are binding modules for mono-ADP-ribose and poly-ADP-ribose, respectively, in addition to two RNA recognition motifs. Catalytic inhibitors of PARP14 have been shown to reverse IL-4 driven pro-tumor gene expression in macrophages, however it is not clear what roles the non-enzymatic biomolecular recognition motifs play in PARP14-driven immunology and inflammation. To further understand this, we have discovered a heterobifunctional small molecule designed based on a catalytic inhibitor of PARP14 that binds in the enzyme's NAD
+ -binding site and recruits cereblon to ubiquitinate it and selectively target it for degradation., (© 2021 Wiley-VCH GmbH.)- Published
- 2021
- Full Text
- View/download PDF
42. In Vitro and Cellular Probes to Study PARP Enzyme Target Engagement.
- Author
-
Wigle TJ, Blackwell DJ, Schenkel LB, Ren Y, Church WD, Desai HJ, Swinger KK, Santospago AG, Majer CR, Lu AZ, Niepel M, Perl NR, Vasbinder MM, Keilhack H, and Kuntz KW
- Subjects
- Binding, Competitive, Fluorescence Resonance Energy Transfer, Fluorescent Dyes chemical synthesis, Fluorescent Dyes metabolism, HEK293 Cells, Humans, Isoenzymes antagonists & inhibitors, Isoenzymes genetics, Isoenzymes metabolism, Kinetics, NAD chemistry, NAD metabolism, Nanoparticles chemistry, Poly(ADP-ribose) Polymerase Inhibitors chemical synthesis, Poly(ADP-ribose) Polymerase Inhibitors metabolism, Poly(ADP-ribose) Polymerases chemistry, Poly(ADP-ribose) Polymerases genetics, Protein Binding, Recombinant Proteins biosynthesis, Recombinant Proteins isolation & purification, Recombinant Proteins metabolism, Surface Plasmon Resonance, Fluorescent Dyes chemistry, Poly(ADP-ribose) Polymerase Inhibitors chemistry, Poly(ADP-ribose) Polymerases metabolism
- Abstract
Poly(ADP-ribose) polymerase (PARP) enzymes use nicotinamide adenine dinucleotide (NAD
+ ) to modify up to seven different amino acids with a single mono(ADP-ribose) unit (MARylation deposited by PARP monoenzymes) or branched poly(ADP-ribose) polymers (PARylation deposited by PARP polyenzymes). To enable the development of tool compounds for PARP monoenzymes and polyenzymes, we have developed active site probes for use in in vitro and cellular biophysical assays to characterize active site-directed inhibitors that compete for NAD+ binding. These assays are agnostic of the protein substrate for each PARP, overcoming a general lack of knowledge around the substrates for these enzymes. The in vitro assays use less enzyme than previously described activity assays, enabling discrimination of inhibitor potencies in the single-digit nanomolar range, and the cell-based assays can differentiate compounds with sub-nanomolar potencies and measure inhibitor residence time in live cells., Competing Interests: Declaration of Interests All authors are employees and shareholders of Ribon Therapeutics and a patent on this work has been granted (WO/2019/212946)., (Copyright © 2020 Elsevier Ltd. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
43. Enabling drug discovery for the PARP protein family through the detection of mono-ADP-ribosylation.
- Author
-
Lu AZ, Abo R, Ren Y, Gui B, Mo JR, Blackwell D, Wigle T, Keilhack H, and Niepel M
- Subjects
- ADP-Ribosylation drug effects, Drug Discovery trends, HeLa Cells, Humans, Poly(ADP-ribose) Polymerase Inhibitors chemistry, ADP-Ribosylation physiology, Drug Discovery methods, Poly(ADP-ribose) Polymerase Inhibitors metabolism, Poly(ADP-ribose) Polymerase Inhibitors pharmacology, Poly(ADP-ribose) Polymerases metabolism
- Abstract
Poly-ADP-ribose polymerases (PARPs) are a family of enzymes responsible for transferring individual or chains of ADP-ribose subunits to substrate targets as a type of post-translational modification. PARPs regulate a wide variety of important cellular processes, ranging from DNA damage repair to antiviral response. However, most research to date has focused primarily on the polyPARPs, which catalyze the formation of ADP-ribose polymer chains, while the monoPARPs, which transfer individual ADP-ribose monomers, have not been studied as thoroughly. This is partially due to the lack of robust assays to measure mono-ADP-ribosylation in the cell. In this study, the recently developed MAR/PAR antibody has been shown to detect mono-ADP-ribosylation in cells, enabling the field to investigate the function and therapeutic potential of monoPARPs. In this study, the antibody was used in conjunction with engineered cell lines that overexpress various PARPs to establish a panel of assays to evaluate the potencies of literature-reported PARP inhibitors. These assays should be generally applicable to other PARP family members for future compound screening efforts. A convenient and generalizable workflow to identify and validate PARP substrates has been established. As an initial demonstration, aryl hydrocarbon receptor was verified as a direct PARP7 substrate and other novel substrates for this enzyme were also identified and validated. This workflow takes advantage of commercially available detection reagents and conventional mass spectrometry instrumentation and methods. Ultimately, these assays and methods will help drive research in the PARP field and benefit future therapeutics development., (Copyright © 2019 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
44. Tazemetostat, an EZH2 inhibitor, in relapsed or refractory B-cell non-Hodgkin lymphoma and advanced solid tumours: a first-in-human, open-label, phase 1 study.
- Author
-
Italiano A, Soria JC, Toulmonde M, Michot JM, Lucchesi C, Varga A, Coindre JM, Blakemore SJ, Clawson A, Suttle B, McDonald AA, Woodruff M, Ribich S, Hedrick E, Keilhack H, Thomson B, Owa T, Copeland RA, Ho PTC, and Ribrag V
- Subjects
- Aged, Antineoplastic Agents adverse effects, Antineoplastic Agents pharmacokinetics, Benzamides adverse effects, Benzamides pharmacokinetics, Biphenyl Compounds, Dose-Response Relationship, Drug, Enhancer of Zeste Homolog 2 Protein metabolism, Enzyme Inhibitors adverse effects, Enzyme Inhibitors pharmacokinetics, Female, France, Humans, Lymphoma, B-Cell enzymology, Lymphoma, B-Cell pathology, Male, Maximum Tolerated Dose, Middle Aged, Morpholines, Pyridones adverse effects, Pyridones pharmacokinetics, Time Factors, Treatment Outcome, Antineoplastic Agents administration & dosage, Benzamides administration & dosage, Enhancer of Zeste Homolog 2 Protein antagonists & inhibitors, Enzyme Inhibitors administration & dosage, Lymphoma, B-Cell drug therapy, Pyridones administration & dosage
- Abstract
Background: Activating enhancer of zeste homolog 2 (EZH2) mutations or aberrations of the switch/sucrose non-fermentable (SWI/SNF) complex (eg, mutations or deletions of the subunits INI1 or SMARCA4) can lead to aberrant histone methylation, oncogenic transformation, and a proliferative dependency on EZH2 activity. In this first-in-human study, we aimed to investigate the safety, clinical activity, pharmacokinetics, and pharmacodynamics of tazemetostat, a first-in-class selective inhibitor of EZH2., Methods: We did an open-label, multicentre, dose-escalation, phase 1 study using a 3 + 3 design with planned cohort expansion at the two highest doses below the maximally tolerated dose. The study was done at two centres in France: Institut Gustave Roussy (Villejuif, Val de Marne) and Institut Bergonié (Bordeaux, Gironde). Eligible patients had relapsed or refractory B-cell non-Hodgkin lymphoma or an advanced solid tumour and were older than 18 years, with Eastern Cooperative Oncology Group performance status of 0 or 1, and adequate end-organ function. Tazemetostat was administered orally from 100 mg twice daily to 1600 mg twice daily in 28-day cycles. The primary endpoint was to establish the maximum tolerated dose or recommended phase 2 dose of tazemetostat, as determined by dose-limiting toxicities, laboratory values, and other safety or pharmacokinetic measures in cycle one according to local investigator assessment. Safety was assessed in patients who received at least one dose of tazemetostat; antitumour activity was assessed in the intention-to-treat population. This study is registered with ClinicalTrials.gov, number NCT01897571. The phase 1 part of the study is complete, and phase 2 is ongoing., Findings: Between June 13, 2013, and Sept 21, 2016, 64 patients (21 with B-cell non-Hodgkin lymphoma, and 43 with advanced solid tumours) received doses of tazemetostat. The most common treatment-related adverse events, regardless of attribution, were asthenia (21 [33%] of 64 treatment-related events), anaemia (nine [14%]), anorexia (four [6%]), muscle spasms (nine [14%]), nausea (13 [20%]), and vomiting (six [9%]), usually grade 1 or 2 in severity. A single dose-limiting toxicity of grade 4 thrombocytopenia was identified at the highest dose of 1600 mg twice daily. No treatment-related deaths occurred; seven (11%) patients had non-treatment-related deaths (one at 200 mg twice daily, four at 400 mg twice daily, and two at 1600 mg twice daily). The recommended phase 2 dose was determined to be 800 mg twice daily. Durable objective responses, including complete responses, were observed in eight (38%) of 21 patients with B-cell non-Hodgkin lymphoma and two (5%) of 43 patients with solid tumours., Interpretation: Tazemetostat showed a favourable safety profile and antitumour activity in patients with refractory B-cell non-Hodgkin lymphoma and advanced solid tumours, including epithelioid sarcoma. Further clinical investigation of tazemetostat monotherapy is ongoing in phase 2 studies in adults and a phase 1 study for children, which are currently enrolling patients who have B-cell non-Hodgkin lymphoma and INI1-negative or SMARCA4-negative tumours., Funding: Epizyme and Eisai., (Copyright © 2018 Elsevier Ltd. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
45. EZH2 Modifies Sunitinib Resistance in Renal Cell Carcinoma by Kinome Reprogramming.
- Author
-
Adelaiye-Ogala R, Budka J, Damayanti NP, Arrington J, Ferris M, Hsu CC, Chintala S, Orillion A, Miles KM, Shen L, Elbanna M, Ciamporcero E, Arisa S, Pettazzoni P, Draetta GF, Seshadri M, Hancock B, Radovich M, Kota J, Buck M, Keilhack H, McCarthy BP, Persohn SA, Territo PR, Zang Y, Irudayaraj J, Tao WA, Hollenhorst P, and Pili R
- Subjects
- Animals, Bevacizumab pharmacology, Carcinoma, Renal Cell pathology, Cell Line, Tumor, Enhancer of Zeste Homolog 2 Protein genetics, Female, Humans, Kidney Neoplasms pathology, Lung Neoplasms drug therapy, Lung Neoplasms secondary, Mice, Mice, Inbred ICR, Mice, SCID, Neovascularization, Pathologic drug therapy, Phosphorylation, Receptor Protein-Tyrosine Kinases metabolism, Sunitinib, Vascular Endothelial Growth Factor A antagonists & inhibitors, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Carcinoma, Renal Cell drug therapy, Drug Resistance, Neoplasm physiology, Enhancer of Zeste Homolog 2 Protein antagonists & inhibitors, Enhancer of Zeste Homolog 2 Protein metabolism, Indoles pharmacology, Kidney Neoplasms drug therapy, Pyrroles pharmacology, Receptor Protein-Tyrosine Kinases antagonists & inhibitors
- Abstract
Acquired and intrinsic resistance to receptor tyrosine kinase inhibitors (RTKi) represents a major hurdle in improving the management of clear cell renal cell carcinoma (ccRCC). Recent reports suggest that drug resistance is driven by tumor adaptation via epigenetic mechanisms that activate alternative survival pathways. The histone methyl transferase EZH2 is frequently altered in many cancers, including ccRCC. To evaluate its role in ccRCC resistance to RTKi, we established and characterized a spontaneously metastatic, patient-derived xenograft model that is intrinsically resistant to the RTKi sunitinib, but not to the VEGF therapeutic antibody bevacizumab. Sunitinib maintained its antiangiogenic and antimetastatic activity but lost its direct antitumor effects due to kinome reprogramming, which resulted in suppression of proapoptotic and cell-cycle-regulatory target genes. Modulating EZH2 expression or activity suppressed phosphorylation of certain RTKs, restoring the antitumor effects of sunitinib in models of acquired or intrinsically resistant ccRCC. Overall, our results highlight EZH2 as a rational target for therapeutic intervention in sunitinib-resistant ccRCC as well as a predictive marker for RTKi response in this disease. Cancer Res; 77(23); 6651-66. ©2017 AACR ., (©2017 American Association for Cancer Research.)
- Published
- 2017
- Full Text
- View/download PDF
46. EZH2 Inhibition by Tazemetostat Results in Altered Dependency on B-cell Activation Signaling in DLBCL.
- Author
-
Brach D, Johnston-Blackwell D, Drew A, Lingaraj T, Motwani V, Warholic NM, Feldman I, Plescia C, Smith JJ, Copeland RA, Keilhack H, Chan-Penebre E, Knutson SK, Ribich SA, Raimondi A, and Thomenius MJ
- Subjects
- Adenine analogs & derivatives, Animals, B-Lymphocytes drug effects, Biphenyl Compounds, Cell Proliferation drug effects, DNA Methylation drug effects, Drug Synergism, Enhancer of Zeste Homolog 2 Protein antagonists & inhibitors, Gene Expression Regulation, Neoplastic drug effects, Humans, Lymphoma, Large B-Cell, Diffuse genetics, Lymphoma, Large B-Cell, Diffuse pathology, Mice, Morpholines, Mutation, Piperidines, Signal Transduction drug effects, Xenograft Model Antitumor Assays, Benzamides administration & dosage, Enhancer of Zeste Homolog 2 Protein genetics, Lymphoma, Large B-Cell, Diffuse drug therapy, Pyrazoles administration & dosage, Pyridones administration & dosage, Pyrimidines administration & dosage
- Abstract
The EZH2 small-molecule inhibitor tazemetostat (EPZ-6438) is currently being evaluated in phase II clinical trials for the treatment of non-Hodgkin lymphoma (NHL). We have previously shown that EZH2 inhibitors display an antiproliferative effect in multiple preclinical models of NHL, and that models bearing gain-of-function mutations in EZH2 were consistently more sensitive to EZH2 inhibition than lymphomas with wild-type (WT) EZH2 Here, we demonstrate that cell lines bearing EZH2 mutations show a cytotoxic response, while cell lines with WT- EZH2 show a cytostatic response and only tumor growth inhibition without regression in a xenograft model. Previous work has demonstrated that cotreatment with tazemetostat and glucocorticoid receptor agonists lead to a synergistic antiproliferative effect in both mutant and wild-type backgrounds, which may provide clues to the mechanism of action of EZH2 inhibition in WT- EZH2 models. Multiple agents that inhibit the B-cell receptor pathway (e.g., ibrutinib) were found to have synergistic benefit when combined with tazemetostat in both mutant and WT- EZH2 backgrounds of diffuse large B-cell lymphomas (DLBCL). The relationship between B-cell activation and EZH2 inhibition is consistent with the proposed role of EZH2 in B-cell maturation. To further support this, we observe that cell lines treated with tazemetostat show an increase in the B-cell maturation regulator, PRDM1 /BLIMP1, and gene signatures corresponding to more advanced stages of maturation. These findings suggest that EZH2 inhibition in both mutant and wild-type backgrounds leads to increased B-cell maturation and a greater dependence on B-cell activation signaling. Mol Cancer Ther; 16(11); 2586-97. ©2017 AACR ., (©2017 American Association for Cancer Research.)
- Published
- 2017
- Full Text
- View/download PDF
47. Selective Killing of SMARCA2- and SMARCA4-deficient Small Cell Carcinoma of the Ovary, Hypercalcemic Type Cells by Inhibition of EZH2: In Vitro and In Vivo Preclinical Models.
- Author
-
Chan-Penebre E, Armstrong K, Drew A, Grassian AR, Feldman I, Knutson SK, Kuplast-Barr K, Roche M, Campbell J, Ho P, Copeland RA, Chesworth R, Smith JJ, Keilhack H, and Ribich SA
- Subjects
- Animals, Carcinoma, Small Cell diagnosis, Carcinoma, Small Cell genetics, Carcinoma, Small Cell pathology, Cell Line, Tumor, Chromosomal Proteins, Non-Histone genetics, Diagnosis, Differential, Enhancer of Zeste Homolog 2 Protein antagonists & inhibitors, Female, Gene Expression Regulation, Neoplastic drug effects, Histone-Lysine N-Methyltransferase genetics, Humans, Hypercalcemia diagnosis, Hypercalcemia drug therapy, Hypercalcemia genetics, Hypercalcemia pathology, Mice, Mutation, Ovarian Neoplasms diagnosis, Ovarian Neoplasms genetics, Ovarian Neoplasms pathology, Rhabdoid Tumor diagnosis, Rhabdoid Tumor genetics, Rhabdoid Tumor pathology, Xenograft Model Antitumor Assays, Carcinoma, Small Cell drug therapy, DNA Helicases genetics, Enhancer of Zeste Homolog 2 Protein genetics, Nuclear Proteins genetics, Ovarian Neoplasms drug therapy, Rhabdoid Tumor drug therapy, Transcription Factors genetics
- Abstract
The SWI/SNF complex is a major regulator of gene expression and is increasingly thought to play an important role in human cancer, as evidenced by the high frequency of subunit mutations across virtually all cancer types. We previously reported that in preclinical models, malignant rhabdoid tumors, which are deficient in the SWI/SNF core component INI1 (SMARCB1), are selectively killed by inhibitors of the H3K27 histone methyltransferase EZH2. Given the demonstrated antagonistic activities of the SWI/SNF complex and the EZH2-containing PRC2 complex, we investigated whether additional cancers with SWI/SNF mutations are sensitive to selective EZH2 inhibition. It has been recently reported that ovarian cancers with dual loss of the redundant SWI/SNF components SMARCA4 and SMARCA2 are characteristic of a rare rhabdoid-like subtype known as small-cell carcinoma of the ovary hypercalcemic type (SCCOHT). Here, we provide evidence that a subset of commonly used ovarian carcinoma cell lines were misdiagnosed and instead were derived from a SCCOHT tumor. We also demonstrate that tazemetostat, a potent and selective EZH2 inhibitor currently in phase II clinical trials, induces potent antiproliferative and antitumor effects in SCCOHT cell lines and xenografts deficient in both SMARCA2 and SMARCA4. These results exemplify an additional class of rhabdoid-like tumors that are dependent on EZH2 activity for survival. Mol Cancer Ther; 16(5); 850-60. ©2017 AACR ., (©2017 American Association for Cancer Research.)
- Published
- 2017
- Full Text
- View/download PDF
48. Initial testing (stage 1) of tazemetostat (EPZ-6438), a novel EZH2 inhibitor, by the Pediatric Preclinical Testing Program.
- Author
-
Kurmasheva RT, Sammons M, Favours E, Wu J, Kurmashev D, Cosmopoulos K, Keilhack H, Klaus CR, Houghton PJ, and Smith MA
- Subjects
- Animals, Biphenyl Compounds, Cell Proliferation drug effects, Drug Evaluation, Preclinical, Female, Humans, Mice, Mice, SCID, Morpholines, Neoplasms, Experimental pathology, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Benzamides pharmacology, Enhancer of Zeste Homolog 2 Protein antagonists & inhibitors, Neoplasms, Experimental drug therapy, Pyridones pharmacology
- Abstract
Background: Tazemetostat (EPZ-6438) is a selective inhibitor of the histone methyltransferase EZH2 and currently in clinical development for non-Hodgkin lymphoma and genetically defined tumors., Procedures: Tazemetostat was tested against the Pediatric Preclinical Testing Program (PPTP) solid tumor xenografts using a dose of 400 mg/kg administered twice daily by oral gavage for 28 days. H3K27me3:H3 ratios were determined in control and treated tumors., Results: Tazemetostat induced significant differences in event-free survival (EFS) distribution compared with control in nine of 30 (30%) of the xenografts studied. Significant differences in EFS distribution were observed in five of seven (71%) rhabdoid tumor xenograft lines compared with four of 23 (17%) nonrhabdoid xenograft lines (chi-square [χ
2 ] test P = 0.006). Tazemetostat induced tumor growth inhibition meeting criteria for intermediate and high EFS treated-to-control (T/C) activity in two of 25 (8%) and one of 25 (4%) xenografts, respectively. Intermediate and high activity for the EFS T/C metric was observed exclusively among rhabdoid tumor xenografts (three of five rhabdoid tumor vs 0 of 22 nonrhabdoid tumors (χ² test P < 0.001). One rhabdoid tumor xenograft (G401) showed stable disease. For one rhabdoid tumor (G401), delayed tumor regression to tazemetostat was noted following 1 week of tumor growth. Tazemetostat induced significant reduction of H3K27me3 levels in the majority of tumors compared with controls., Conclusions: Tazemetostat demonstrated significant antitumor activity in rhabdoid tumor models but showed no consistent activity against any other histology. Tazemetostat reduced H3K27me3 levels irrespective of tumor response. Further preclinical testing to evaluate tazemetostat in combination with other anticancer agents is warranted., (© 2016 Wiley Periodicals, Inc.)- Published
- 2017
- Full Text
- View/download PDF
49. Correction: Preclinical Evidence of Anti-Tumor Activity Induced by EZH2 Inhibition in Human Models of Synovial Sarcoma.
- Author
-
Kawano S, Grassian AR, Tsuda M, Knutson SK, Warholic NM, Kuznetsov G, Xu S, Xiao Y, Pollock RM, Smith JJ, Kuntz KW, Ribich S, Minoshima Y, Matsui J, Copeland RA, Tanaka S, and Keilhack H
- Abstract
[This corrects the article DOI: 10.1371/journal.pone.0158888.].
- Published
- 2017
- Full Text
- View/download PDF
50. In Vitro Long-Term Proliferation Assays to Study Antiproliferative Effects of PARP Inhibitors on Cancer Cells.
- Author
-
Keilhack H and Chang P
- Subjects
- Animals, Cell Cycle drug effects, Cell Differentiation drug effects, Cell Line, Tumor, Cell Proliferation drug effects, Humans, Biological Assay methods, Poly(ADP-ribose) Polymerase Inhibitors pharmacology, Poly(ADP-ribose) Polymerases metabolism
- Abstract
Cell proliferation assays are an important component of small molecule inhibitor screens for cancer therapies. An important but often overlooked variable involves the timing and timeframe of inhibitor treatment. Whereas many traditional chemotherapeutics kill or inhibit proliferation on the timeframe of hours or in a few days of treatment, more targeted therapies that affect other cancer-relevant pathways, including differentiation or cell stress responses, can take longer, often several days to weeks to impact cellular growth and survival. Many poly(ADP-ribose) polymerases (PARPs) are involved in cellular stress pathways; therefore, phenotypic effects of PARP inhibition are often only observed with long-term inhibitor treatment. Here we summarize several assays for analyzing long-term proliferation of both adherent and suspension cells, relying either on growth in two-dimensional tissue culture or on systems than enable growth in 3D.
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.