127 results on '"Samid D"'
Search Results
2. Pharmacologic Activation of Expression of Immunodominant Viral Antigens: A New Strategy for the Treatment of Epstein-Barr-Virus-Associated Malignancies
- Author
-
Robertson, K. D., Barletta, J., Samid, D., Ambinder, R. F., Capron, A., editor, Compans, R. W., editor, Cooper, M., editor, Koprowski, H., editor, McConnell, I., editor, Melchers, F., editor, Oldstone, M., editor, Olsnes, S., editor, Potter, Michael, editor, Saedler, H., editor, Vogt, P. K., editor, Wagner, H., editor, Wilson, I., editor, and Melchers, Fritz, editor
- Published
- 1995
- Full Text
- View/download PDF
3. Abstracts
- Author
-
Derlon J. M., Petit-taboué M. C., Dauphin F., Courtheoux P., Chapon F., Creissard P., Darcel F., Houtteville J. P., Kaschten, B., Sadzot, B., Stevenaert, A., Tjuvajev, Juri G., Macapinlac, Homer A., Daghighian, Farhad, Ginos, James Z., Finn, Ronald D., Jiaju Zhang, M. S., Beattie, Bradley, Graham, Martin, Larson, Steven M., Blasberg, Ronald G., Levivier, M., Goldman, S., Pirotte, B., Brucher, J. M., Balériaux, D., Luxen, A., Hildebrand, J., Brotchi, J., Go K. G., Kamman R. L., Mooyaart E. L., Heesters M. A. A. M., Sijens, P. E., Oudksrk, M., van Dijk, P., Levendag, P. C., Vecht, Ch. J., Metz, R. J., Kennedy, D. N., Rosen, B. R., Hochberg, F. H., Fishman, A. J., Filipek, P. A., Caviness, V. S., Gross, M. W., Weinzierl, F. X., Trappe, A. E., Goebel, W. E., Frank, A. M., Becker, Georg, Krone, Andreas, Schmidt, Karsten, Hofmann, Erich, Bogdahn, Ulrich, Bencsch, H., Fclber, S., Finkenstedt, G., Kremser, C., Sfockhammer, G., Aichner, F., Bogdahn U., Fröhlich T., Becker G., Krone A., Schlief R., Schürmann J., Jachimczak P., Hofmann E., Roggendorf W., Roosen K., Carapella, C. M., Carpinelli, G., Passalacqua, R., Raus, L., Giannini, M., Mastrostefano, R., Podo, F., Tofani, A., Maslrostefano, R., Mottoles, M., Ferraironi, A., Scelsa, M. G., Oppido, P., Riccio, A., Maini, C. L., Collombier, L., Taillandier, L., Dcbouverie, M., Laurens, M. H., Thouvenot, P., Weber, M., Bertrand, A., Cruickshank G. S., Patterson J., Hadley D., De Witte, Olivier, Hildebrand, Jerzy, Luxen, André, Goldman, Serge, Ernestus, R. -I., Bockhorst, K., Eis, M., Els, T., Hoehn-Berlage, M., Gliese, M., Fründ, R., Geissler, A., Woertgen, C., Holzschuh, M., Goldman, Serge, Levivier, M., Pirotte, B., Brucher, J. M., Luxen, A., Brotchi, J., Hildebrand, J., Hausmann, O., Merlo, A., Jerrnann, E., Uirich, J., Chiquet-Ehrismann, R., Müller, J., Mäcke, H., Gratzl, O., Herholz, K., Ghaemi, M., Würker, M., Pietrzyk, U., Heiss, W. -D., Kotitschke, K., Brandl, M., Tonn, J. C., Haase, A., Bogdahn, U., Kotitschke, K., Muigg, S., Felber, S., Aichner, F., Haase, A., Bogdahn, U., Krone A., Becker G., Woydt M., Roggendorf W., Hofmann E., Bogdahn U., Roosen K., Lanfermann, Heinrich, Heindel, Walter, Kugel, Harald, Erneslus, Ralf -Ingo, Röhn, Gabricle, Lackner, Klaus, Metz, R. J., Kennedy, D. N., Pardo, F. S., Kutke, S., Sorensen, A. G., Hochberg, F. H., Fishman, A. J., Filipek, P. A., Rosen, B. R., Caviness, V. S., Mechtler, L. L., Withiam-Lench, S., Shin, K., Klnkel, W. R., Patel, M., Truax, B., Kinkel, P., Shin, K., Mechtler, L., Ricci M., Pantano P., Maleci A., Pierallini S., Di Stefano D., Bozzao L., Cantore G. P., Röhn, Gabriele, Els, T., Schröder, R., Hoehn-Berlage, M., Ernestus, R. -I., Ruda, R., Mocellini, C., Soffietti, R., Campana, M., Ropolo, R., Riva, A., de Filippi, P. G., Schiffer, D., Salgado D., Rodrigues M., Salgado L., Fonseca A. T., Vieira M. R., Bravo Marques J. M., Satoh, H., Uozumi, T., Kiya, K., Kurisu, K., Arita, K., Sumida, M., Ikawa, F., Tzuk-Shina, Tz., Gomori, J. M., Rubinstein, R., Lossos, A., Siegal, T., Vaalburg, W., Paans, A. M. J., Willemsen, A. T. M., van Waarde, A., Pruim, J., Visser, G. M., Go, K. G., Valentini, S., Ting, Y. L. T., De Rose, R., Chidichimo, G., Corricro, G., van Lcycn-Pilgram, Karin, Erncslus, Ralf -Ingo, Klug, Norfried, van Leyen-Pilgram, K., Ernestus, R. -I., Schröder, R., Klug, N., Woydt M., Krone A., Tonn J. C., Becker G., Neumann U., Roggendorf W., Roosen K., Plate, Karl H., Breier, Georg, Millaucr, Birgit, Weich, Herbert A., Ullrich, Axel, Risau, Werner, Roosen N., Chopra R. K., Mikkelsen T., Rosenblum S. D., Yan P. S., Knight R., Windham J., Rosenblum M. L., Schiffer, D., Attanasio, A., Cavalla, P., Chio, A., Giordana, M. T., Migheli, A., Amberger, V., Hensel, T., Schwab, M. E., Cervoni, Luigi, Celli, Paolo, Tarantino, Roberto, Huettner, C., Tonn, J. C., Berweiler, U., Roggendorf, W., Salmon, I., Rorive, S., Rombaut, K., Pirotte, B., Haot, J., Brotchi, J., Kiss, R., Maugard-Louboutin C., Charrier J., Fayet G., Sagan C., Cuillioere P., Ricolleau G., Martin S., Menegalli-Bogeelli D., Lajat Y., Resche F., Molnàr, Péter, Bárdos, Helga, Ádány, Róza, Rogers, J. P., Pilkington, G. J., Pollo, B., Giaccone, G., Allegranza, A., Bugiani, O., Prim, J., Badia, J., Ribas, E., Coello, F., Shezen, E., Lossos, A., Abramsky, O., Siegal, T., Scerrati M., Roselli R., Iacoangeli M., Pompucci A., Rossi G. F., Deeb, Saleh M. Al., Koreich, Osama, Yaqub, Basim, Moutaery, Khalaf R. Al., Giordana, M. T., Cavalla, P., Chio, A., Marino, S., Vigliani, M. C., Schiffer, D., Deburghgraeve, V., Darcel, F., Gedouin, D., Hassel, M. Ben, Guegan, Y., Jeremic, B., Grujicic, D., Antunovic, V., Matovic, M., Shibamoto, Y., Kallio, Merja, Huhmar, Helena, Kudoh, Ch., Detta, A., Sugiura, K., Hitchcock, E. R., Mastrostefano, R., Di Russo, R., Cipriani§, M., Occhipinti, E. M., Conti, E. M. S., Clowegeser A., Ortler M., Seiwald M., Kostron H., Rajan B., Ross G., Lim C., Ashlcy S., Goode D., Traish D., Brada M., Sanden, G. A. C. vd, Schouten, L. J., Coebergh, J. W. W., Razenberg, P. P. A., Twijnstra, A., Snilders-Keilholz, A., Voormolen, J. H. C., Hermans, J., Leer, J. W. H., Taillandier, L., Baylac, F., Dcbouvcrie, M., Anxionnal, R., Bracard, S., Vignand, J. M., Duprcz, A., Weber, M., Winking, M., Böker, D. K., Simmet, T., Rothbart, David, Strugar, John, Balledux, Jeroen, Criscuolo, Gregory R., Jachimczak, Piotr, Blesch, Armin, Heβdörfer, Birgit, Bogdahn, Ulrich, Ernestus, Ralf -Ingo, Schröder, Roland, Klug, Norfrid, Krouwer, H. G. J., Duinen, S. G. v., Algra, A., Zentner, J., Wolf, H. K., Ostertun, B., Hufnagel, A., Campos, M. G., Solymosi, L., Schramm, J., Newlands, E. S., O'Reilly, S. M., Brampton, M., Soffietti, R., Chio, A., Mocellini, C., Ruda, R., Vigliani, M. C., Schiffer, D., Sciolla, R., Seliak, D., Henriksson, R., Bergenheim, A. T., Björk, P., Gunnarsson, P. -O., Hariz, Ml., Grant, R., Collie, D., Gregor A., Ebmeier K. P., Jarvis G., Lander F., Cull A., Sellar R., Brada, M., Thomas, C., Elyan, S., Hines, F., Ashley, S., Stenning, S., Bernstein J. J., Goldberg W. J., Roelcke U., Von Ammon K., Hausmann O., Radu E. W., Kaech D., Leenders K. L., Fitzek, II, M. M., Aronen J. Efird, Hochberg, F., Gruber, M., Schmidt, E., Rosen, B., Flschman, A., Pardo, P., Afra U. M. U., Sipos, L., Slouik, F., Boiardi A., Salmaggi A., Pozzi A., Farinotti L., Fariselli L., Silvani A., Brandes, A., Scelzi, E., Rigon, A., Zampieri, P., Pignataro, M., Amanzo, P. D'., Amista, P., Rotilio, A., Fiorentino, M. V., Thomas, R., Brazil, L., O'Connor, A. M., Ashley, S., Brada, M., Salvati, Maurizio, Cervoni, Luigi, Puzzilli, Fabrizio, Cervoni, Luigi, Salvati, Maurizio, Raguso, Michele, Cruickshank G. S., Duckworth R., Rumpling R., Rottuci M., Fariselli L., Boiardi A., Broggi G., Plrint, N. G., Sabattini, E., Manetto, V., Gambacorta, H., Poggi, S., Pileri, S., Ferracini, R., Grant, R., Plev D. V., Hopf N. J., Knosp E., Bohl J., Perncczky A., Kiss, R., Salmon, I., Catnby, I., Dewitte, O., Brotchi, J., Pasteels, J. L., Camby, I., Salmon, I., Darro, F., Danguy, A., Brotchi, J., Pasteels, J. L., Kiss, R., Kiu, M. C., Lai, G. M., Yang, T. S., Ng, K. T., Chen, J. S., Chang, C. N., Leung, W. M., Ho, Y. S., Rychter, M. Deblec, Klimek, A., Liberski, P. P., Karpinaka, A., Krauseneck P., Schöffel V., Müller B., Kreth, F. W., Faist, M., Warnke, P. C., Ostertag, C. B., Nielen, K. M. B. v., Visscr, M. C., Lebrun C., Lonjon M., Desjardin T., Michiels J. F., Chanalet Sa. Lagrange J. L., Roche J. L., Chatel M., Mastronardi L., Puzzilli F., Osman Farah J., Lunardi P., Matsutani, M., Ushio, Y., Takakura, K., Menten, Johan, Hamers, Han, Ribot, Jacques, Dom, René, Tcepen, Hans, Müller B., Weidner N., Krauseneck P., Naujocks, G., van Roost, D., Wiestler, O. D., Kuncz, A., Nieder, C., Setzel-Sesterhein, M., Niewald, M., Schnabel, I., O'Neill, K. S., Kitchen, N. D., Wilkins, P. R., Marsh, H. T., Pierce, E., Doshi, R., Deane, R., Previtali, S., Quattrini, A., Nemni, R., Ducati, A., Wrabetz, L., Canal, N., Punt, C. J. A., Stamatakis, L., Giroux, B., Rutten, E., Quigley, Matthew R., Beth Sargent P. A. -C., Flores, Nicholas, Simon, Sheryl, Maroon, Joseph C., Quigley, Matthew R., Beth Sargent P. A. -C., Flores, Nicholas, Maroon, Joseph C., Rocca A. A., Gervasoni C., Castagna A., Picozzi P., Giugni E., Rocca A. A., Tonnarelli G. P., Ducati A., Mangili F., Truci G., Canal N., Giovanelli M., Roelcke U., Von Ammon K., Radu E. W., Leenders K. L., Sachsenheimer, W., Bimmler, T., Seiwald M., Eiter H. Rhomberg W., Ortler M., Obwegesser A., Kostron H., Steilen H., Henn W., Moringlane J. R., Kolles H., Feiden W., Zang K. D., Sleudel W. I., Steinbrecher, Andreas, Schabet, Martin, Heb, Clemens, Bamberg, Michael, Dichgans, Johannes, Stragliotto, G., Delattre, J. Y., Poisson, M., Zampieri, P., Brandes, A., Rigon, A., Tosatto, L., D'Amanzo, P., Menicucci, N., Rotilio, A., Mingrino, S., Steudel, W. I., Feld, R., Henn, W., Zang, K. D., Maire, J. Ph., Caudry, M., Guerin, J., Celerier, D., Salem, N., Demeaux, H., Fahregat, J. F., Kusak, M. E., Bucno, A., Albisua, J., Jerez, P., Sarasa, J. L., Garefa, R., de Campos, J. M., Kusak, M. E., de Campos, J. M., Bueno, A., García-Delgado, R., Sarasa, J. L., García-Sola, R., Lantsov A. A., Shustova T. I., Lcnartz, D., Wellenreuther, R., von Deirnling, A., Köning, W., Menzel, J., Scarpa, S., Manna, A., Reale, M. G., Oppido, P. A., Carapella, C. M., Frati, L., Valery, C. A., Ichen, M., Foncin, J. P., Soubrane, C., Khayat, D., Philippon, J., Vaz, R., Cruz, C., Weis S., Protopapa D., März R., Winkler P. A., Reulen H. J., Bise K., Beuls E., Berg J., Deinsberger, W., Böker, D. K., Samii, M., Caudry, M., Darrouzet, V., Guérin, J., Trouette, R., Causse, N., Bébéar, J. P., Parker, F., Vallee, J. N., Carlier, R., Zerah, M., Lacroix-Jousselin, C., Piepmeier, Joseph M., Kveton, John, Czibulka, Agnes, Tigliev G. S., Chernov M. P., Maslova L. N., Valdueza, José M., Jänisch, Werner, Bock, Alexander, Harms, Lutz, Bessell, E. M., Graus, F., Punt, J., Firth, J., Hope, T., Koriech, Osama, Al Deeb, Saleh, Al Moutaery, Khalaf, Yaqub, B., Silvani A., Salmaggi A., Pozzi A., Franzini A., Boiardi A., Goldbrunner, R., Warmuth-Metz, M., Paulus, W., Tonn, J. -Ch., Roosen, K., Strik I. I., Müller B., Markert C., Pflughaupt K. -W., Krauseneck P., O'Neill, B. P., Dinapoli, R. P., Voges, J., Sturm, V., Deuß, U., Traud, C., Treuer, H., Lehrke, R., Kim, D. G., Müller, R. P., Alexandrov Yu. S., Moutaery, K., Aabed, M., Koreich, O., Ross, G. M., Rajan, B., Traish, D., Ashley, S., Ford, D., Brada, M., Schmeets, I. L. O., Jager, J. J., Pannebakker, M. A. G., de Jong, J. M. A., van Lindert, E., Knosp, E., Kitz, K., Blond, S., Dubois, F., Assaker, R., Baranzelli, M. C., Sleiman, M., Pruvo, J. P., Coche-Dequeant, B., Matsutani M., Takakura K., Sano K., PetriČ-Grabnar, G., Jereb, B., Župančič, N., Koršič, M., Rainov N. G., Burkert W., Ushio, Yukitaka, Kochi, Masato, Itoyama, Youichi, de Campos, J. M., Kusak, M. E., Sarasa, J. L., García, R., Bueno, A., Ferrando, L., Hoang-Xuan, K., Sanson, M., Merel, P., Delattre, J. Y., Poisson, M., Delattre, O., Thomas, G., Hoang-Xuan, K., Delattre, J. Y., Poisson, M., Thomas, G., Haritz, D., Obersen, B., Grochulla, F., Gabel, D., Haselsberger K., Radner H., Pendl G., Brada, M., Laing, R. W., Warrington, A. P., Nowak, P. J. C. M., Kolkman-Deurloo, I. K. K., Visser, A. G., Berge, Hv. d., Niël, C. G. J. H., Levendag, P. C., Bergström P., Hariz M., Löfroth P. -O., Bergenheim T., Henriksson R., Blond, S., Assaker, R., Cortet-rudelli, C., Dewailly, D., Coche-dequeant, B., Castelain, B., Dinapoli, R., Shaw, E., Coffey, R., Earle, J., Foote, R., Schomberg, P., Gorman, D., Girard N., Courel M. N., Delpech B., Haselsberger K., Friehs G. M., Schröttner O., Pendl G., Pötter, R., hawliczek, R., Sperveslage, P., Prott, F. J., Wachter, S., Dieckmann, K., Würker, M., Herholz, K., Pietrzyk, U., Voges, J., Treuer, H., Sturm, V., Bauer, B., Heiss, W. -D., Jund, R., Zimmermann, F., Feldmann, H. J., Gross, M. W., Kneschaurek, P., Molls, M., Lederman, G., Lowry, J., Wertheim, S., Voulsinas, L., Fine, M., Lederman, G., Lowry, J., Wertheim, S., Fine, M., Voutsinas, I., Qian, G., Rashid, H., Lederman, G., Lowry, J., Wertheim, S., Fine, M., Voulsinas, L., Qian, G., Rashid, H., Moutaery, K., Aabed, M., Koreich, O., Scerrati M., Montemaggi P., Iacoangeli M., Pompucci A., Roselli R., Trignani R., Rossi G. F., Shin, K., Mechtler, L., West, C., Grand, W., Shin, K., Sibata, C., West, C., Mechtler, L., Grand, W., Thomas, R., Guerrero, D., James, N., Ashley, S., Gregor, A., Brada, M., Voges, J., Sturm, V., Bramer, R., Pahlke, H., Lehrke, R., Treuer, H., Banik, N., Kim, D. G., Hövels, M., Bernsen H. J. J. A., Rijken P. F. J. W., Van der Sanden B. P. J., Hagemeier N. E. M., Van der Kogel A. J., Koehler P. J., Verbiest H., Jager J., Vecht Ch. J., Ross G. M., McIlwrath A., Brown R., Mottolesb, C., Pierre'Kahn, A., Croux, M., Roche, J. L., Marchai, J., Delhemes, P., Tremoulet, M., Stilhart, B., Chazai, J., Caillaud, P., Ravon, R., Passacha, J., Bouffet, E., Dirven C. M. F., Mooy J. J. A., Molenaar W. M., Lewandowicz, G. M., Grant, N., Harkness, W., Hayward, R., Thomas, D. G. T., Darling, J. L., Delepine, N., Subovici I. I., Cornille B., Markowska S., Alkallaf JC. Desbois, KühI, J., Niethammer, D., Spaar, H. J., Gnekow, A., Havers, W., Berthold, F., Graf, N., Lampert, F., Maass, E., Mertens, R., Schöck, V., Aguzzi, A., Boukhny, A., Smirtukov, S., Prityko, A., Hoiodov, B., Geludkova, O., Nikanorov, A., Levin, P., Rothbart, David, Balledux, Jeroen, Criscuolo, Gregory R., D'haen, B., Van Calenbergh, F., Casaer, P., Dom, R., Menten, J., Goffin, J., Plets, C., Hertel, A., Hernaiz, P., Seipp, C., Siegler, K., Baum, R. P., Maul, F. D., Schwabe, D., Jacobi, G., Kornhuber, B., Hör, G., Menten, J., Casaer, P., Pilkington, G. J., Merzak, A., Rooprai, H. K., Bullock, P., van Domburg P. H. M. F., Wesseling P., Thijssen H. O. M., Wolff, J. E. A., Boos, J., Krähling, K. H., Gressner-Brocks, V., Jürgens, H., Schlegel, J., Scherthan, H., Arens, N., Stumm, Gabi, Kiessling, Marika, Merzak, A., Koochekpour, S., Pilkington, G. J., Reifenberger, G., Reifenberger, J., Liu, L., James, C. D., Wechsler, W., Collins, V. P., Fabel-Schulte, Klaus, Jachimczak, Plotr, Heßdörfer, Birgitt, Baur, Inge, Schlingensiepen, Karl -Hermann, Brysch, Wolgang, Bogdahn, Ulrich, Blesch A., Bosserhoff A. K., Apfel R., Lottspeich F., Jachimczak P., Büttner R., Bogdahn U., Cece, R., Barajon, I., Tazzari, S., Cavaletti, G., Torri-Tarelli, L., Tredici, G., Hecht, B., Turc-Carel, C., Atllas, R., Chatel, M., Gaudray, P., Gioanni, J., Hecht, F., Balledux, Jeroen, Rothbart, David, Criscuolo, Gregory R., de Campos, J. M., Kusak, M. E., Rey, J. A., Bello, M. J., Sarasa, J. L., Dubois, F., Blond, S., Parent, M., Assaker, R., Gosselin, P., Christiaens, J. L., Feld, R., Moringlane, J. R., Steudel, W. I., Schaudies, J. R., Janka M., Tonn J. C., Fischer U., Meese E., Roosen K., Remmelink, M., Salmon, I., Cras, P., Pasteels, J. L., Brotchi, J., Kiss, R., Bensadoun R. J., Frenay M., Formento J. L., Milano G., Lagrange J. L., Grellier P., Lee, J. -Y., Ernestus, R. -I., Riese, H. -H., Cervós-Navarro, J., Reutter, W., Lippitz, B., Scheitinger, C., Scholz, M., Weis, J., Gilsbach, J. M., Füzesi, L., Koochekpour, S., Merzak, A., Pilkington, G. J., Sanson, M., Li, Y. J., Hoang-Xuan, K., Delattre, J. Y., Poisson, M., Hamelin, R., Van de Kelft, Erik, Dams, Erna, Martin, Jean -Jacques, Willems, Patrick, Lehrke R., Voges J., Treuer H., Erdmann J., Müller R. P., Sturm V., Wurm R. E., Warrington A. P., Laing R. W., Sardell S., Hines F., Graham J. D., Brada M., Ushio, Yukitaka, Kuratsu, Jun -ichi, Kochi, Masato, Kitz K., Aichholzer M., Rössler K., Alesch F., Ertl A., Sorensen, P. S., Helweg-Larsen, S., Mourldsen, H., Hansen, H. H., El Sharoum, S. Y., Berfelo, M. W., Theunissen, P. H. M. H., Jager, J. J., de Jong, J. M. A., Fedorcsák, I., Nyáry, I., Osztie, É., Horvath, Á., Kontra, G., Frenay M., Burgoni-chuzel J., Paquis P., Lagrange J. L., Helweg-Larsen, S., Hansen, SW., Sørensen, PS., Salmon, I., Kiss, R., Krauseneck P., Müller B., Morche M., Tonn J. C., Lagerwaard, F. J., Levendag, P. C., Eijkenboom, W. M. H., Schmilz, P. I. M., Lentzsch S., Weber F., Franke J., Dörken B., Lunardi P., Schettini G., Osman Farah J., Qasho R., Mocellini, C., Ruda, R., Soffietti, R., Garabello, D., Sales, S., De Lucchi, R., Vasario, E., Schiffer, D., Muracciole, X., Régis, J., Manera, L., Peragut, J. C., Juin, P., Sedan, R., Nieder, C., Niewald, M., Walter, K., Schnabel, K., Nieder, C., Niewald, N., Nestle, U., Schnabel, K., Berberich, W., Oschmann, P., Theißen R. D., Reuner K. H., Kaps M., Dorndorf W., Martin, K. K., Akinwunmi, J., Rooprai, H. K., Kennedy, A., Linke, A., Ognjenovic, N., Pilkington, G. J., Svadovsky A. I., Peresedov V. V., Bulakov A. A., Butyalko M. Y., Zhirnova I. G., Labunsky D. A., Gnazdizky V. V., Gannushkina I. V., Taphoorn, M. J. B., Potman, R., Barkhof, F., Weerts, J. G., Karim, A. B. M. F., Heimans, J. J., van de Pol, M., van Aalst, V. C., Wilmink, J. T., Twijnstra, A., van der Sande, J. J., Boogerd, W., Kröger, R., Jäger A., Wismeth C., Dekant A., Brysch W., Schlingensiepen K. H., Jachimczak P., Bogdahn U., Pirolte, B., Cool, V., Gérard, C., Levivier, M., Dargent, J. L., Goldman, S., Brotchi, J., Hildebrand, J., Velu, T., Herrlinger, U., Schabet, M., Ohneseit, P., Buchholz, R., Zhu, Jianhong, Reszka, Regina, Weber, Friedrich, Walther, Wolfgang, Zhang, L. I., Brock, Mario, Roosen N., Rock J. P., Zeng H., Feng J., Fenstermacher J. D., Rosenblum M. L., Siegal, T., Gabizon, A., Beljanski M., Crochet S., Bergenheim, A. T., Zackrisson, B., Elfverson, J., Bergström, P., Henriksson, R., Butti, G., Baetta, R., Magrassi, L., De Renzis, M. R., Soma, M. R., Davegna, C., Pezzotta, S., Paoletti, R., Fumagalli, R., Infuso, L., Sankar, A. A., Darling, J. L., Thomas, D. G. T., Defer, G. -L., Brugières, P., Gray, F., Chomienne, C., Poirier, J., Degos, L., Degos, J. D., Colombo, Bruno M., DiDonato, Stefano, Finocchiaro, Gaetano, Hebeda, K. M., Sterenborg, H. J. C. M., Saarnak, A. E., Wolbers, J. G., van Gemert, M. J. C., Kaaijk P., Troost D., Leenstra S., Das P. K., Bosch D. A., Kostron H., Hochleitner B. W., Obwegeser A., Ortler M., Seiwald M., Vooys, W., Krouwer, H. G. J., de Gast, G. C., Marx, J. J. M., Osman Farah J., Lunardi P., Puzzilli F., Menovsky, T., Beek, J. F., Wolbers, J. G., van Gemert, M. J. C., Naujocks, G., Wiestler, O. D., Schirrmacher, V., Schramm, J., Schmitz, A., Eis-Hübinger, A. M., Piepmeier, p. h., Pedersen, Patricia, Greer, Charles, Quigley, Matthew R., Shih, Tommy, Elrifal, Amr, Rothfus, William, Maroon, Joseph C., Rohertson, L., Rampling, R., Whoteley, T. L., Piumb, J. A., Kerr, D. J., Falina, P. A., Crossan, I. M., Roosen N., Rock J. P., Feng J., Zeng H., Ho K. L., Fenstermacher J. D., Rosenblum M. L., Ruchoux, M. M., Vincent, S., Jonca, F., Plouet, J., Lecomte, M., Samid, D., Thibault, A., Ram, Z., Oldfield, E. H., Myers, C. E., Reed, E., Schabet, M., Herrlinger, U., Buchholz, R., Shoshan, Y., Siegal, T., Siegal, T., Shezen, E., Siegal, Tz., Stockhammer, G., Rosenblum, M., Samid, D., Lieberman, F., Terzis, A. J. A., Bjerkvig, R., Laerum, O. D., Arnold, H., Thibault, A., Samid, D., Figg, W. D., Myers, C. E., Reed, E., Thomas, R., Flux, G., Chittenden, S., Doshi, P., Brazil, L., Thomas, D. G. T., Bignor, D., Zalutsky, M., Brada, M., Tjuvajev, Juri, Kaplitt, Michael, Desai, Revathi, Bradley, M. S., Bettie B. S., Gansbacher, Bernd, Blasberg, Ronald, Haugland, H. K., Saraste, J., Rooseni, K., Laerum, O. D., Vincent, A. J. P. E., Avezaat, C. J. J., Bout, A., Noteboom, J. L., Vecht, C. h., Valerio, D., Hoogerbrugge, P. M., Weber, F., Reszka, R., Zhu, J., Walther, W., List, J., Schulz, W., Wolbers, J. G., Sterenborg, I. I. J. C. M., Kamphorst, W., van Gemert, M. J. C., van Alplien, H. A. M., Salander P., Bergenheim T., Henriksson R., Grant, R., Brazil, L., Thomas, R., Guerrero, D., Laing, R., Ashley, S., Brada, M., Schmidt B., Bauer B., Grau G., Bohnstedt, T., Frydrych A., Franz K., Lorenz R., Brandes, A., Amanzo, P. D'., Zampieri, P., Rigon, A., Scelzi, E., Rotilio, A., Berti, F., Paccagnella, A., Fiorentino, M. V., Müller B., Krauseneck P., van Deventer, P. L., Dellemijn, P. L. I., van den Bent, M. J., Vecht, Ch. J., Kansen, P. J., Tredici, G., Petruccioli, N. G., Cavaletti, G., Cavalletti, E., Kiburg, B., Müller, L. J., Moorer-van Delft, C. M., Heimans, J. J., Boer, H. H., Pace A., Bove L., Pietrangeli A., Innocenti P., Aloe A., Nardi M., Jandolo B., Kellie S. J., De Graaf S. S. N., Bloemhof H., Roebuck D., Dalla Pozza L., Uges D. D. R., Johnston I., Besser M., Chaseling R. A., Koeppen, S., Gründemann, S., Lossos, A., Siegal, T., Nitschke M., Vieregge P., Reusche E., Rob P., Kömpf D., Postma, T. J., Vermorken, J. B., Heimans, J. J., Rampling R. P., Dunlop D. J., Steward M. S., Campbell S. M., Roy S., Hilkens, P. H. E., Verweij, J., van Putten, W. L. J., Vecht, Ch. J., van den Bent, M. J., Hilkens, P. H. E., Moll, J. W. B., van der Burg, M. E. L., Planting, A. S. T., van Putten, W. L. J., Vecht, Ch. J., van den Bent, M. J., Wondrusch E., Zifko U., Drlicek M., Liszka U., Grisold W., Zifko U., Fazeny B., Dittrich Ch., Wondrusch E., Grisold W., Verschuuren, Jan J., Meneses, Patricio I., Rosenfeld, Myrna R., Kaplitt, Michael G., Posner, Jerome B., Dalmau, Josep, Sillevis Smitt P. A. E., Manley G., Posner J. B., Cavaletti, G., Bogliun, G., Margorati, L., Bianchi, G., Drlicek, M., Liska, U., Casati, B., Kolig, C., Grisold, H., Graus, F., Reñe, R., Uchuya, M., Valldeoriola, F., Delattre, J. Y., Benedetti de Cosentiro C., Ortale D., Martinez R., Lambre J., Cagnolati S., Vinai C., Salmaggi A., Nemni R., Silvani A., Forno M. G., Luksch R., Confalonieri P., Boiardi A., Nitschke M., Scholz J., Vieregge P., Kömpf D., Hochberg F. H., Pfeiffer, G., Netzer, J., Hansen, Ch., Eggers, Ch., Hagel Ch., Kunze, K., Verschuuren, Jan J., Rosenblum, Marc K., Lieberman, Frank S., Posner, Jerome B., and Dalmau, Josep
- Published
- 1994
- Full Text
- View/download PDF
4. Phenylbutyrate-induced G1 arrest and apoptosis in myeloid leukemia cells: structure–function analysis
- Author
-
DiGiuseppe, JA, Weng, L-J, Yu, KH, Fu, S, Kastan, MB, Samid, D, and Gore, SD
- Published
- 1999
- Full Text
- View/download PDF
5. RADIATION-INDUCED MICRONUCLEI YIELDS IN HUMAN OSTEOSARCOMA CELLS EXPRESSING THE ras ONCOGENE
- Author
-
MILLER, A.C., primary, SAMID, D., additional, and CLARK, E.P., additional
- Published
- 1991
- Full Text
- View/download PDF
6. Phenylbutyrate-induced G1 arrest and apoptosis in myeloid leukemia cells: structure-function analysis
- Author
-
Steven D. Gore, Shin-Huei Fu, K H Yu, Li Jun Weng, Joseph A. DiGiuseppe, Michael B. Kastan, and Samid D
- Subjects
Cancer Research ,Cellular differentiation ,Antineoplastic Agents ,Apoptosis ,Butyrate ,Biology ,Phenylbutyrate ,Structure-Activity Relationship ,Differentiation therapy ,hemic and lymphatic diseases ,Tumor Cells, Cultured ,Humans ,Dose-Response Relationship, Drug ,G1 Phase ,Myeloid leukemia ,Cell Differentiation ,Hematology ,Cell cycle ,Phenylbutyrates ,In vitro ,Oncology ,Leukemia, Myeloid ,Immunology ,Cancer research ,Cell Division - Abstract
The aromatic fatty acid phenylbutyrate (PB) induces cytostasis, differentiation, and apoptosis in primary myeloid leukemic cells at clinically achievable concentrations. In the present study, we have investigated the structural and cellular basis for PB-induced cytostasis, using the ML-1 human myeloid leukemia cell line as a model system. PB induced a dose-dependent increase in cells in G1 with a corresponding decrease in cells in S-phase of the cell cycle. At comparable doses, PB induced expression of CD11b, indicating myeloid differentiation. At higher doses, the drug induced apoptosis. The antitumor activity was independent of the aromatic ring, as butyric acid (BA) was of equal or greater potency at producing these biological changes. In contrast, shortening of the fatty acid carbon chain length, as demonstrated with phenylacetate (PA), significantly diminished drug potency. Consistent with their effects on cell cycle, PB and BA, but not PA, induced the cyclin-dependent kinase inhibitor, p21(WAF1/CIP1), and led to the appearance of hypophosphorylated Rb, suggesting a role for p21(WAF1/CIP1) in PB-induced cytostasis. Therefore, it appears that the fatty acid moiety of PB, rather than its aromatic ring, is critical for its activity in myeloid leukemic cells. These data provide a potential mechanistic basis for the increased potency of PB over PA previously demonstrated in primary leukemic samples, and support the further clinical development of PB in the treatment of hematologic malignancies.
- Published
- 1999
7. 463 A multicenter phase II study of capecitabine plus paclitaxel in metastatic breast cancer: Survival update
- Author
-
Gradishar, W., primary, Meza, L., additional, Hill, T., additional, Samid, D., additional, Chen, Y.-M., additional, and Amin, B., additional
- Published
- 2003
- Full Text
- View/download PDF
8. Re: Therapeutic Targeting of Transcription in Acute Promyelocytic Leukemia by Use of an Inhibitor of Histone Deacetylase RESPONSE
- Author
-
Samid, D., primary and Warrell, R. P., additional
- Published
- 1999
- Full Text
- View/download PDF
9. Transcriptional activation of the Epstein-Barr virus latency C promoter after 5-azacytidine treatment: evidence that demethylation at a single CpG site is crucial
- Author
-
Robertson, K D, primary, Hayward, S D, additional, Ling, P D, additional, Samid, D, additional, and Ambinder, R F, additional
- Published
- 1995
- Full Text
- View/download PDF
10. Phenylacetate inhibits protein prenylation in LNCaP cells transfected with the T24 H-ras gene
- Author
-
DANESI, R, primary, NARDINI, D, additional, SAMID, D, additional, AGEN, C, additional, DIPAOLO, A, additional, MYERS, C, additional, and DELTACCA, M, additional
- Published
- 1995
- Full Text
- View/download PDF
11. Silencing of the VHL tumor-suppressor gene by DNA methylation in renal carcinoma.
- Author
-
Herman, J G, primary, Latif, F, additional, Weng, Y, additional, Lerman, M I, additional, Zbar, B, additional, Liu, S, additional, Samid, D, additional, Duan, D S, additional, Gnarra, J R, additional, and Linehan, W M, additional
- Published
- 1994
- Full Text
- View/download PDF
12. Enhanced fetal hemoglobin production by phenylacetate and 4- phenylbutyrate in erythroid precursors derived from normal donors and patients with sickle cell anemia and beta-thalassemia
- Author
-
Fibach, E, primary, Prasanna, P, additional, Rodgers, GP, additional, and Samid, D, additional
- Published
- 1993
- Full Text
- View/download PDF
13. Posttranscriptional down-regulation of ras oncogene expression by inhibitors of cellular glutathione.
- Author
-
Miller, A C, primary, Gafner, J, additional, Clark, E P, additional, and Samid, D, additional
- Published
- 1993
- Full Text
- View/download PDF
14. Selective growth arrest and phenotypic reversion of prostate cancer cells in vitro by nontoxic pharmacological concentrations of phenylacetate.
- Author
-
Samid, D, primary, Shack, S, additional, and Myers, C E, additional
- Published
- 1993
- Full Text
- View/download PDF
15. Differences in Radiation-induced Micronuclei Yields of Human Cells: Influence ofRasGene Expression and Protein Localization
- Author
-
Miller, A.C., primary, Gafner, J., additional, Clark, E.P., additional, and Samid, D., additional
- Published
- 1993
- Full Text
- View/download PDF
16. Induction of erythroid differentiation and fetal hemoglobin production in human leukemic cells treated with phenylacetate
- Author
-
Samid, D, primary, Yeh, A, additional, and Prasanna, P, additional
- Published
- 1992
- Full Text
- View/download PDF
17. 145. Phenylacetate — A novel nontoxic inducer of tumor cell maturation
- Author
-
Samid, D, primary
- Published
- 1992
- Full Text
- View/download PDF
18. Modulation of radiation response of human tumour cells by the differentiation inducers, phenylacetate and phenylbutyrate.
- Author
-
Miller, A. C., Whittaker, T., Thibault, A., and Samid, D.
- Subjects
PHENYLACETATES ,FATTY acids ,CANCER cells - Abstract
Abstract. The aromatic fatty acids phenylacetate (PA) and phenylbutyrate (PB) are novel antitumour agents currently under clinical evaluation. Their ability to induce tumour differentiation in laboratory models and their low clinical toxicity profile makes them promising candidates for combination with conventional therapies. In the present studies, we characterized the interactions between these aromatic fatty acids and radiation, using as a model cell lines derived from cancers of the prostate, breast, brain and colon. Analysis of the radiation response of the tumour lines using the linear-quadratic model, demonstrated that cellular exposure to pharmacological, non-toxic concentrations of either PA or PB resulted in time-dependent and contrasting changes in radiation response. While drug pretreatment for 24 h reduced radiation sensitivity (significant alterations in both alpha and beta parameters), pre treatment for 72 h significantly increased radiosensitivity (significant alterations in alpha and beta parameters). In replicating tumour cells, these changes were accompanied by a gradual Gl-phase arrest. Cytostasis alone, however, could not explain radiosensitization, as similar alterations in radiation response were documented also in non-cycling cells. Modulation of tumour radiobiology by PA and PB was tightly correlated with early rise followed by decline in intracellular glutathione levels and the activity of antioxidant enzymes such as catalase, superoxide dismutase, glutathione reductase, glutathione peroxidase and glutathione S -transferase. Although in vitro findings identify the aromatic fatty acids PA and PB as a new class of non-toxic modulators of radiation response, the antagonistic effect of these compounds on radiation response needs further examination. Our data strongly suggest that for PA or PB to have a role in clinical radiotherapy, appropriate scheduling of combination therapies must take into account their timedependent effects in order to achieve clinical radiosensitization. [ABSTRACT FROM AUTHOR]
- Published
- 1997
- Full Text
- View/download PDF
19. Differences in Radiation-induced Micronuclei Yields of Human Cells: Influence of Ras Gene Expression and Protein Localization.
- Author
-
Miller, A. C., Gafner, J., Clark, E. P., and Samid, D.
- Published
- 1993
- Full Text
- View/download PDF
20. Phase I study of phenylacetate administered twice daily to patients with cancer.
- Author
-
Thibault, Alain, Samid, Dvorit, Cooper, Michael R., Figg, William D., Tompkins, Anne C., Patronas, Nicholas, Headlee, Donna J., Kohler, David R., Venzon, David J., Myers, Charles E., Thibault, A, Samid, D, Cooper, M R, Figg, W D, Tompkins, A C, Patronas, N, Headlee, D J, Kohler, D R, Venzon, D J, and Myers, C E
- Published
- 1995
- Full Text
- View/download PDF
21. Relationship Between EJras Expression And Radiation Resistance In Human Osteosarcoma Cells.
- Author
-
Miller, A.C., Samid, D., Kariko, K., and Clark, E.P.
- Published
- 1991
- Full Text
- View/download PDF
22. Transcriptional upregulation of gamma-globin by phenylbutyrate and analogous aromatic fatty acids
- Author
-
Hudgins, W. R., Fibach, E., Safaya, S., Rieder, R. F., Miller, A. C., and Samid, D.
- Published
- 1996
- Full Text
- View/download PDF
23. Activation of a human peroxisome proliferator-activated receptor by the antitumor agent phenylacetate and its analogs
- Author
-
Pineau, T., Hudgins, W. Robert, Liu, L., Chen, L.-C., Sher, T., Gonzalez, F. J., and Samid, D.
- Published
- 1996
- Full Text
- View/download PDF
24. Phenylacetate inhibits protein isoprenylation and growth of the androgen-independent LNCaP prostate cancer cells transfected with the T24 Ha-ras oncogene.
- Author
-
Danesi, R, Nardini, D, Basolo, F, Del Tacca, M, Samid, D, and Myers, C E
- Abstract
The refractoriness of prostate cancer to androgen suppression is the landmark of clinically aggressive disease. In this study, the androgen-dependent LNCaP prostate cancer cells were transfected with the mutated c-Ha-ras gene from the T24 human bladder cancer. The derivative clone overexpressing T24-ras (LNCaP(T24-ras)) proliferated in androgen-depleted medium and showed increased growth. Protein isoprenylation and p21ras farnesylation in LNCaP(T24-ras) cells were tested in the presence of phenylacetate to document a possible relationship with the drug-induced inhibition of cell proliferation. Phenylacetate is a differentiation inducer that down-regulates in vitro the expression of the myc oncogene and activates the human peroxisome proliferator-activated nuclear receptor involved in cell growth regulation. The drug inhibited protein isoprenylation and p21ras farnesylation in LNCaP(T24-ras) cells; IC50 values were 3.1 and 3.3 mM, respectively, compared with controls. The drug reduced the cellular levels of endogenous farnesyl-PP (mean IC50 = 3.5 mM) and inhibited activation of the p21ras downstream target, p42(MAPK)/ERK2. LNCaP(T24-ras) was more sensitive than the parental line to both growth inhibition (mean IC50 = 3.01 and 7.1 mM, respectively) and apoptosis by phenylacetate. Exogenous farnesyl- and geranylgeranyl-PP indeed reduced the effects of the drug on proliferation and apoptosis in LNCaP(T24-ras) cells. In conclusion, the inhibition of protein isoprenylation and p21ras farnesylation by phenylacetate resulted in increased chemosensitivity of the androgen-independent LNCaP(T24-ras) cells compared with LNCaP, and this effect might contribute to the pharmacological activity of the drug.
- Published
- 1996
25. Interferon-induced revertants of ras-transformed cells: resistance to transformation by specific oncogenes and retransformation by 5-azacytidine
- Author
-
Samid, D, Flessate, D M, and Friedman, R M
- Abstract
Prolonged alpha/beta interferon (IFN-alpha/beta) treatment of NIH 3T3 cells transformed by a long terminal repeat-activated Ha-ras proto-oncogene resulted in revertants that maintained a nontransformed phenotype long after IFN treatment had been discontinued. Cloned persistent revertants (PRs) produced large amounts of the ras-encoded p21 and were refractile to transformation by EJras DNA and by transforming retroviruses which carried the v-Ha-ras, v-Ki-ras, v-abl, or v-fes oncogene. Transient treatment either in vitro or in vivo with cytidine analogs that alter gene expression by inhibiting DNA methylation resulted in transformation of PR, but not of NIH 3T3, cells. The PR retransformants reverted again with IFN, suggesting that DNA methylation is involved in IFN-induced persistent reversion.
- Published
- 1987
- Full Text
- View/download PDF
26. Phenylacetate inhibits protein isoprenylation and growth of the androgen-independent LNCaP prostate cancer cells transfected with the T24 Ha-ras oncogene
- Author
-
Danesi R, Nardini D, FULVIO BASOLO, Del Tacca M, Samid D, and Ce, Myers
- Subjects
Male ,Antimetabolites, Antineoplastic ,Protein Prenylation ,Prostatic Neoplasms ,Phenylacetate ,Apoptosis ,prostate cancer ,Transfection ,Proto-Oncogene Proteins p21(ras) ,Genes, ras ,Humans ,Cell Division ,Phenylacetates - Abstract
The refractoriness of prostate cancer to androgen suppression is the landmark of clinically aggressive disease. In this study, the androgen-dependent LNCaP prostate cancer cells were transfected with the mutated c-Ha-ras gene from the T24 human bladder cancer. The derivative clone overexpressing T24-ras (LNCaP(T24-ras)) proliferated in androgen-depleted medium and showed increased growth. Protein isoprenylation and p21ras farnesylation in LNCaP(T24-ras) cells were tested in the presence of phenylacetate to document a possible relationship with the drug-induced inhibition of cell proliferation. Phenylacetate is a differentiation inducer that down-regulates in vitro the expression of the myc oncogene and activates the human peroxisome proliferator-activated nuclear receptor involved in cell growth regulation. The drug inhibited protein isoprenylation and p21ras farnesylation in LNCaP(T24-ras) cells; IC50 values were 3.1 and 3.3 mM, respectively, compared with controls. The drug reduced the cellular levels of endogenous farnesyl-PP (mean IC50 = 3.5 mM) and inhibited activation of the p21ras downstream target, p42(MAPK)/ERK2. LNCaP(T24-ras) was more sensitive than the parental line to both growth inhibition (mean IC50 = 3.01 and 7.1 mM, respectively) and apoptosis by phenylacetate. Exogenous farnesyl- and geranylgeranyl-PP indeed reduced the effects of the drug on proliferation and apoptosis in LNCaP(T24-ras) cells. In conclusion, the inhibition of protein isoprenylation and p21ras farnesylation by phenylacetate resulted in increased chemosensitivity of the androgen-independent LNCaP(T24-ras) cells compared with LNCaP, and this effect might contribute to the pharmacological activity of the drug.
27. Growth inhibition, tumor maturation, and extended survival in experimental brain tumors in rats treated with phenylacetate
- Author
-
Ram, Z., Samid, D., Walbridge, S., Oshiro, E.M., Viola, J.J., Taocheng, J.H., Shack, S., Thibault, A., Myers, C.E., and Oldfield, E.H.
- Subjects
Brain tumors -- Growth ,Cancer -- Differentiation therapy ,Business ,Health care industry - Abstract
SOURCE: Cancer Research, June 1, 1994;54(11):2923-2927. According to the authors' abstract of an article published in Cancer Research, 'Phenylacetate is a naturally occurring plasma component that suppresses the growth of [...]
- Published
- 1994
28. Increased resistance to oxidative stress in human osteosarcoma cells transformed by EJras oncogene
- Author
-
Miller, A.C., primary, Gafher, J., additional, Clark, E.P., additional, and Samid, D., additional
- Full Text
- View/download PDF
29. Relationship Between EJras Expression And Radiation Resistance In Human Osteosarcoma Cells
- Author
-
Miller, A.C., primary, Samid, D., additional, Kariko, K., additional, and Clark, E.P., additional
- Full Text
- View/download PDF
30. A phase-I and pharmacokinetic study of intravenous phenylacetate in patients with cancer
- Author
-
Thibault, A., Cooper, M.R., Figg, W.D., Venzon, D.J., Sartor, A.O., Tompkins, A.C., Weinberger, M.S., Headlee, D.J., Mccall, N.A., Samid, D., and Myers, C.E.
- Subjects
Cancer ,Chemotherapy -- Evaluation ,Intravenous therapy -- Health aspects ,Business ,Health care industry - Abstract
SOURCE: Cancer Research, April 1, 1994;54(7):1690-1694. According to the authors' abstract of an article published in Cancer Research, 'Phenylacetate has recently been shown to suppress tumor growth and promote differentiation [...]
- Published
- 1994
31. Increased resistance to oxidative stress in human osteosarcoma cells transformed by EJras oncogene.
- Author
-
Miller, A.C., Gafher, J., Clark, E.P., and Samid, D.
- Published
- 1993
- Full Text
- View/download PDF
32. P14 04 MP - RADIATION-INDUCED MICRONUCLEI YIELDS IN HUMAN OSTEOSARCOMA CELLS EXPRESSING THE ras ONCOGENE
- Author
-
MILLER, A.C., SAMID, D., and CLARK, E.P.
- Published
- 1991
- Full Text
- View/download PDF
33. Increases in NMR-visible lipid and glycerophosphocholine during phenylbutyrate-induced apoptosis in human prostate cancer cells.
- Author
-
Milkevitch M, Shim H, Pilatus U, Pickup S, Wehrle JP, Samid D, Poptani H, Glickson JD, and Delikatny EJ
- Subjects
- Cell Line, Tumor, Cytoplasm metabolism, G1 Phase drug effects, Humans, Magnetic Resonance Spectroscopy, Male, Phospholipases metabolism, Prostatic Neoplasms pathology, Apoptosis drug effects, Glycerylphosphorylcholine metabolism, Phenylbutyrates pharmacology, Prostatic Neoplasms metabolism
- Abstract
DU145 human prostatic carcinoma cells were treated with the differentiating agents phenylacetate (PA) and phenylbutyrate (PB) and examined in perfused cultures by diffusion-weighted 1H and 31P nuclear magnetic resonance spectroscopy (NMR). PA and PB (10 mM) induced significant (>3-fold) time-dependent increases in the level of NMR-visible lipids and total choline in 1H spectra, and glycerophosphocholine levels in the 31P spectra, with the increases being greater for PB. These effects were accompanied by significant increases in cytoplasmic lipid droplets and intracellular lipid volume fraction as observed by morphometric analysis of Oil Red O-stained cells. PB treatment caused cell cycle arrest in the G1 phase and induction of apoptosis. In contrast, PA-treated DU145 cells showed an accumulation of cells in G2/M and no evidence of apoptosis. These results demonstrate that significant differences exist in the mechanism of PA and PB activity, although both compounds cause similar, but graded alterations in lipid metabolism. The simultaneous accumulation of mobile lipid and glycerophosphocholine suggests that PB and PA induce phospholipid catabolism via a phospholipase-mediated pathway. The mobile lipid accumulation following the induction of either apoptosis and cytostasis by related differentiating agents indicate that the presence of NMR-visible lipids may not be a specific event causally resulting from the induction of apoptosis.
- Published
- 2005
- Full Text
- View/download PDF
34. Capecitabine plus paclitaxel as front-line combination therapy for metastatic breast cancer: a multicenter phase II study.
- Author
-
Gradishar WJ, Meza LA, Amin B, Samid D, Hill T, Chen YM, Lower EE, and Marcom PK
- Subjects
- Adult, Aged, Antineoplastic Combined Chemotherapy Protocols adverse effects, Breast Neoplasms pathology, Capecitabine, Deoxycytidine administration & dosage, Disease Progression, Female, Fluorouracil analogs & derivatives, Humans, Middle Aged, Neoplasm Recurrence, Local, Paclitaxel administration & dosage, Paclitaxel adverse effects, Survival Analysis, Time Factors, Treatment Outcome, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Breast Neoplasms drug therapy, Deoxycytidine analogs & derivatives, Deoxycytidine therapeutic use, Paclitaxel therapeutic use
- Abstract
Purpose: The goal of this multicenter, open-label phase II study was the clinical evaluation of combination therapy with the oral fluoropyrimidine capecitabine and the taxane paclitaxel in patients with metastatic breast cancer (MBC)., Patients and Methods: Forty-seven patients with MBC received oral capecitabine at 1650 mg/m(2)/d (825 mg/m(2) twice daily) on days 1 through 14, and intravenous infusion of paclitaxel at 175 mg/m(2) on day 1 of each 21-day treatment cycle. Treatment continued until disease progression, intolerable toxicity, or patient' s decision to discontinue. Patients (35 to 76 years old) had a median Karnofsky performance status of 90%. Forty-four patients (94%) received study treatment as first-line therapy for metastatic disease., Results: Objective responses occurred in 24 (51%) patients; seven (15%) complete responses and 17 (36%) partial responses. Stable disease lasting 180 days or more was observed in nine (19%); the clinical response rate was 70%. Median duration of response was 12.6 months, median time to disease progression was 10.6 months, and median overall survival time was 29.9 months. The most common treatment-related adverse events, regardless of severity, were alopecia, hand-foot syndrome, nausea, and fatigue. Neutropenia (15%), alopecia (13%), and hand-foot syndrome (11%) were the only grade 3 or 4 treatment-related adverse events that occurred in more than 10% of patients., Conclusion: The combination of capecitabine plus paclitaxel is a highly active and generally well-tolerated regimen for first-line treatment of MBC.
- Published
- 2004
- Full Text
- View/download PDF
35. Phase II study of capecitabine in patients with fluorouracil-resistant metastatic colorectal carcinoma.
- Author
-
Hoff PM, Pazdur R, Lassere Y, Carter S, Samid D, Polito D, and Abbruzzese JL
- Subjects
- Adult, Aged, Antimetabolites, Antineoplastic adverse effects, Capecitabine, Colorectal Neoplasms mortality, Deoxycytidine adverse effects, Disease-Free Survival, Drug Resistance, Neoplasm, Female, Fluorouracil pharmacology, Humans, Male, Middle Aged, Neoplasm Metastasis, Survival Rate, United States epidemiology, Antimetabolites, Antineoplastic therapeutic use, Colorectal Neoplasms drug therapy, Deoxycytidine analogs & derivatives, Deoxycytidine therapeutic use
- Abstract
Purpose: Capecitabine is an oral fluoropyrimidine converted to fluourouracil (FU) preferentially in tumor tissue. It has proven clinical activity against colorectal cancer when used as first-line therapy. The objectives of this study were to assess the safety and efficacy of capecitabine in patients with metastatic colorectal carcinoma who progressed despite previous FU therapy., Patients and Methods: According to the group sequential analysis design of this study, accrual would stop if no responses were observed in the first 20 patients treated. If one or more objective responses were confirmed, the trial would be expanded. Patients received capecitabine 1,250 mg/m(2) twice a day for 14 days, every 3 weeks. Tumor lesions were assessed every 6 weeks, and patients were followed for survival every 3 months after completing treatment., Results: Twenty-three patients were enrolled onto the study; 22 fulfilled all the eligibility criteria. No objective responses were observed among the 22 eligible patients; 11 patients (50%) had stable disease for a median duration of 141 days (range, 88-289 days). The Kaplan-Meier estimate of median time to disease progression was 64 days (95% CI, 41 to 134 days). The median survival time estimate was 389 days (95% CI, 267 to 637 days). The most frequent treatment-related adverse events were hand-foot syndrome, diarrhea, and nausea or vomiting. There were no grade 4 toxicities and no treatment-related deaths., Conclusion: Single-agent capecitabine in patients with metastatic colorectal carcinoma refractory to FU showed no objective responses and clinical benefit that was, at best, modest. The use of capecitabine in combination with other treatments in this patient population is under investigation.
- Published
- 2004
- Full Text
- View/download PDF
36. Inhibition of estrogen-dependent breast cell responses with phenylacetate.
- Author
-
Sawatsri S, Samid D, Malkapuram S, and Sidell N
- Subjects
- Antimetabolites, Antineoplastic pharmacology, Breast Neoplasms genetics, Cell Division drug effects, Cyclin D1 genetics, Dose-Response Relationship, Drug, Drug Interactions, Drug Screening Assays, Antitumor, Humans, Promoter Regions, Genetic drug effects, Tumor Cells, Cultured, Breast Neoplasms pathology, Estrogens pharmacology, Gene Expression drug effects, Phenylacetates pharmacology
- Abstract
The aromatic fatty acid phenylacetate (PA) and its analogs have come under intense investigation due to their ability to cause the growth arrest of a variety of neoplasia, including human breast cancer. We have determined that PA and its halide derivative 4-chlorophenylacetate (4-CPA) showed marked antiproliferative activity on 3 of 6 human breast cancer cell lines tested. Interestingly, the 3 cell lines that were growth inhibited by PA and 4-CPA were estrogen receptor (ER) positive (T47-D, MCF-7 and ZR-75-1) whereas those that were little affected by these compounds were ER-negative (MDA-MB-157, MDA-MB-231 and SK-Br-3). Dose response studies indicated that 4-CPA inhibited the growth of the sensitive (ER+) cell lines with a potency 3-4 times that of PA. These findings suggest that there is "cross-talk" between the PA and estrogen signaling pathways such that PA can directly inhibit estrogen-dependent events. This hypothesis was directly tested in vitro using ER+ MCF-7 cells that were stably transfected with a luciferase reporter construct driven by the full length (1745 bp) cyclin D1 promoter (MCF-7-D1). Our experiments with MCF-7-D1 cells indicated that PA and 4-CPA inhibited basal and estrogen-induced reporter gene activity by up to 90%, resulting in almost complete elimination of estrogen-dependent cyclin D1 gene activation. Using a reporter gene construct (ERE(V)-tk-Luc) containing a canonical estrogen response element that was transiently transfected into MCF-7 and MDA-MB-231 cells, we have also demonstrated inhibition of promoter activity by PA and 4-CPA that was directly mediated by blockage of activity through the ERE. Taken together, these findings indicate that PA analogs possess potent antiestrogen properties that may, at least partly, account for their antiproliferative effects on ER+ breast cancer cells. The data suggests a novel mechanism of action that might bypass some of the limitations of conventional antiestrogen therapy., (Copyright 2001 Wiley-Liss, Inc.)
- Published
- 2001
- Full Text
- View/download PDF
37. Peroxisome proliferator-activated receptor gamma as a novel target in cancer therapy: binding and activation by an aromatic fatty acid with clinical antitumor activity.
- Author
-
Samid D, Wells M, Greene ME, Shen W, Palmer CN, and Thibault A
- Subjects
- Animals, Antimetabolites, Antineoplastic metabolism, Antimetabolites, Antineoplastic pharmacology, Cell Division drug effects, Cell Line, Cyclin-Dependent Kinase Inhibitor p21, Cyclins genetics, Dose-Response Relationship, Drug, Enzyme Inhibitors pharmacology, Flavonoids pharmacology, Gene Expression Regulation, Neoplastic drug effects, Humans, Mitogen-Activated Protein Kinases antagonists & inhibitors, Mitogen-Activated Protein Kinases metabolism, Neoplasms genetics, Neoplasms metabolism, Phenylacetates metabolism, Phenylacetates pharmacology, Phosphorylation, Protein Binding, RNA, Messenger drug effects, RNA, Messenger genetics, RNA, Messenger metabolism, Radioligand Assay, Receptors, Cytoplasmic and Nuclear metabolism, Transcription Factors metabolism, Transcriptional Activation drug effects, Tumor Cells, Cultured, Up-Regulation, Neoplasms prevention & control, Receptors, Cytoplasmic and Nuclear genetics, Transcription Factors genetics
- Abstract
Aromatic fatty acids, of which phenylacetate is a prototype, constitute a class of low toxicity drugs with demonstrated antitumor activity in experimental models and in humans. Using in vitro models, we show here a tight correlation between tumor growth arrest by phenylacetate and activation of peroxisome proliferator-activated receptor gamma (PPARgamma), a member of the nuclear receptor superfamily. In support are the following observations: (a) the efficacy of phenylacetate as a cytostatic agent was correlated with pre-treatment levels of PPARgamma, as documented using established tumor lines and forced expression models; (b) in responsive tumor cells, PPARgamma expression was up-regulated within 2-9 h of treatment preceding increases in p21waf1, a marker of cell cycle arrest; (c) inhibition of mitogen-activated protein kinase, a negative regulator of PPARgamma, enhanced drug activity; and (d) phenylacetate interacted directly with the ligand-binding site of PPARgamma and activated its transcriptional function. The ability to bind and activate PPARgamma was common to biologically active analogues of phenylacetate and corresponded to their potency as antitumor agents (phenylacetate < phenylbutyrate < p-chloro-phenylacetate < p-iodo-phenylbutyrate), whereas an inactive derivative, phenylacetylglutamine, had no effect on PPARgamma. These findings point to PPARgamma as a novel target in cancer therapy and provide the first identification of ligands that have selective antitumor activity in patients.
- Published
- 2000
38. Phenylbutyrate induces cell differentiation and modulates Epstein-Barr virus gene expression in Burkitt's lymphoma cells.
- Author
-
Bar-Ner M, Thibault A, Tsokos M, Magrath IT, and Samid D
- Subjects
- Burkitt Lymphoma metabolism, Cell Differentiation drug effects, Cell Division drug effects, Cell Size drug effects, DNA, Viral biosynthesis, Dose-Response Relationship, Drug, Flow Cytometry, Gene Expression drug effects, Herpesvirus 4, Human growth & development, Histocompatibility Antigens Class I biosynthesis, Humans, Tumor Cells, Cultured, Up-Regulation, Viral Matrix Proteins biosynthesis, Virus Activation drug effects, Burkitt Lymphoma pathology, Burkitt Lymphoma virology, Gene Expression Regulation, Viral drug effects, Herpesvirus 4, Human genetics, Phenylbutyrates pharmacology
- Abstract
Although Burkitt's lymphoma (BL) is a readily treated malignancy, recurrences, as well as disease arising in immunosuppressed patients, are notoriously resistant to conventional therapeutic approaches. The EBV is associated with a significant proportion of these lymphomas that evade immune surveillance through decreased expression of both viral and cellular antigens. Increasing the immunogenicity of BL cells may, therefore, represent a potentially beneficial therapeutic maneuver. Using in vitro models of EBV-transformed lymphoblastoid as well as BL cell lines, we demonstrate increased expression of genes coding for HLA class I and EBV latent proteins by the differentiation inducer phenylbutyrate (PB). The aromatic fatty acid also caused cytostasis associated with sustained declines in c-myc expression, a direct antitumor effect that was independent of the EBV status. We conclude, therefore, that differentiation therapy of BL with PB may lead to growth arrest with increased tumor immunogenicity in vivo. The findings may have clinical relevance because the in vitro activity has been observed with PB concentrations that are well tolerated and nonimmunosuppressive in humans, a desirable feature for the different patient populations afflicted with this disease.
- Published
- 1999
39. Re: Therapeutic targeting of transcription in acute promyelocytic leukemia by use of an inhibitor of histone deacetylase.
- Author
-
Samid D
- Subjects
- DNA, Neoplasm metabolism, Drug Synergism, Histone Deacetylases genetics, Humans, Leukemia, Promyelocytic, Acute enzymology, Leukemia, Promyelocytic, Acute genetics, Methylation drug effects, Neovascularization, Pathologic prevention & control, Antineoplastic Agents pharmacology, Histone Deacetylase Inhibitors, Leukemia, Promyelocytic, Acute drug therapy, Phenylbutyrates pharmacology, Transcription, Genetic drug effects, Tretinoin pharmacology
- Published
- 1999
- Full Text
- View/download PDF
40. A phase II study of 5-aza-2'deoxycytidine (decitabine) in hormone independent metastatic (D2) prostate cancer.
- Author
-
Thibault A, Figg WD, Bergan RC, Lush RM, Myers CE, Tompkins A, Reed E, and Samid D
- Subjects
- Aged, Azacitidine therapeutic use, Decitabine, Humans, Male, Middle Aged, Predictive Value of Tests, Prostatic Neoplasms blood, Treatment Outcome, Antimetabolites, Antineoplastic therapeutic use, Azacitidine analogs & derivatives, Biomarkers, Tumor blood, DNA Modification Methylases antagonists & inhibitors, Enzyme Inhibitors therapeutic use, Fibroblast Growth Factor 2 blood, Prostatic Neoplasms drug therapy
- Abstract
Aims and Background: Decitabine (5-aza-2'-deoxycytidine) is an S-phase-specific pyrimidine analog with hypomethylation properties. In laboratory models of prostate cancer (PC-3 and DU-145), decitabine induces cellular differentiation and enhanced expression of genes involved in tumor suppression, immunogenicity, and programmed cell death., Methods: We conducted a phase II study of decitabine in 14 men with progressive, metastatic prostate cancer recurrent after total androgen blockade and flutamide withdrawal. Decitabine was administered at a dose of 75 mg/m2/dose i.v. as a 1 hour infusion every 8 hours for three doses. Cycles of therapy were repeated every 5 to 8 weeks to allow for resolution of toxicity., Results: Two of 12 patients evaluable for response had stable disease with a time to progression of more than 10 weeks. This activity was seen in 2 of 3 African-American patients. Toxicity was similar to previously reported experience. No significant changes in urinary concentrations of the angiogenic factor bFGF, a potential biomarker of tumor activity, were identified over time in 7 unselected patients with progressive disease., Conclusions: We conclude that decitabine is a well tolerated regimen with modest clinical activity against hormone-independent prostate cancer. Further investigations in patients of African-American origin may be warranted.
- Published
- 1998
- Full Text
- View/download PDF
41. Impact of the putative differentiating agents sodium phenylbutyrate and sodium phenylacetate on proliferation, differentiation, and apoptosis of primary neoplastic myeloid cells.
- Author
-
Gore SD, Samid D, and Weng LJ
- Subjects
- Acute Disease, Antigens, Differentiation biosynthesis, Antigens, Differentiation genetics, Antigens, Neoplasm biosynthesis, Antigens, Neoplasm genetics, Apoptosis drug effects, Bone Marrow pathology, Cell Cycle drug effects, Cell Differentiation drug effects, Cell Division drug effects, Cell Lineage, Gene Expression Regulation, Leukemic drug effects, Humans, Myelodysplastic Syndromes pathology, Tumor Cells, Cultured drug effects, Tumor Stem Cell Assay, Leukemia, Myeloid pathology, Phenylacetates pharmacology, Phenylbutyrates pharmacology
- Abstract
Sodium phenylacetate (PA) and sodium phenylbutyrate (PB) are aromatic fatty acids that can effect differentiation in a variety of cell lines at doses that may be clinically attainable. We have studied the impact of these two agents on lineage- and differentiation stage-specific antigen expression, proliferation, apoptosis, and clonogenic cell survival in primary cultures of bone marrow samples from patients with myeloid neoplasms at presentation and in remission and from normal volunteers. PB inhibited the proliferation of primary acute myeloid leukemia cells in suspension culture with an ID50 of 6.6 mM, similar to its ED50 in cell lines. At higher doses (>/=5 mM), PB also induced apoptosis. PB inhibited clonogenic leukemia cell growth with a median ID50 of less than 2 mM; however, colony-forming units-granulocyte/macrophage from patients with myelodysplasia and normal volunteers were inhibited with a similar ID50. In contrast to PB, its metabolite PA had no significant effect on either acute myeloid leukemia proliferation or apoptosis. Expression of the monocytic marker CD14 was increased in monocytic and myelomonocytic leukemias in response to PB, and to a lesser extent, PA. Surprisingly, both agents appeared to increase expression of the progenitor cell antigen CD34, as well as the DR locus of the human leukocyte antigen. These data indicate that PB, but not its metabolite PA, has significant cytostatic and differentiating activity against primary neoplastic myeloid cells at doses that may be achievable clinically.
- Published
- 1997
42. Tumor radiosensitization based on the use of inhibitors of the mevalonate pathway of cholesterol synthesis.
- Author
-
Miller AC and Samid D
- Subjects
- Adenocarcinoma, Brain Neoplasms, Breast Neoplasms, Cobalt Radioisotopes, Colonic Neoplasms, Cyclin-Dependent Kinase Inhibitor p21, Cyclins biosynthesis, Cyclohexenes, Dose-Response Relationship, Radiation, Female, Fibrosarcoma, Gamma Rays, Genes, ras drug effects, Genes, ras radiation effects, Glioblastoma, Humans, Limonene, Lung Neoplasms, Male, Mevalonic Acid antagonists & inhibitors, Prostatic Neoplasms, Tumor Cells, Cultured, Cell Survival drug effects, Cell Survival radiation effects, Cholesterol biosynthesis, Mevalonic Acid metabolism, Radiation-Sensitizing Agents pharmacology, Terpenes pharmacology
- Published
- 1997
43. Plasma protein binding of phenylacetate and phenylbutyrate, two novel antineoplastic agents.
- Author
-
Boudoulas S, Lush RM, McCall NA, Samid D, Reed E, and Figg WD
- Subjects
- Adult, Antimetabolites, Antineoplastic therapeutic use, Humans, Hydrogen-Ion Concentration, Neoplasms blood, Neoplasms drug therapy, Phenylacetates therapeutic use, Phenylbutyrates therapeutic use, Protein Binding, Serum Albumin metabolism, Antimetabolites, Antineoplastic blood, Phenylacetates blood, Phenylbutyrates blood
- Abstract
Phenylacetate and phenylbutyrate, two novel inducers of tumor cytostasis and differentiation, are currently in clinical trials for the treatment of cancer in adults. The purpose of our study was to evaluate the plasma protein-binding characteristics of phenylacetate and phenylbutyrate in the plasma of normal volunteers and that of patients with cancer. Drug plasma protein-binding analysis was examined using three separate devices: a micropartition system and two equilibrium dialysis systems, all of which exhibited similar results. Phenylacetate and phenylbutyrate concentrations were determined by high-performance liquid chromatography. Both drugs exhibited concentration-dependent binding. Our results showed sodium phenylacetate to have a higher free fraction than sodium phenylbutyrate at corresponding concentrations (> 0.442 +/- 0.008 and > 0.188 +/- 0.001, respectively). Plasma pH did not greatly affect protein binding of either drug. As albumin concentration decreased, an increase in free fraction of both drugs was observed, however alpha 1-acid glyco-protein showed no change in free fraction as its concentration increased. Patients with cancer with lower levels of albumin showed an increase in free fraction with both phenylacetate and phenylbutyrate. When phenylacetate and phenylbutyrate were added together in plasma, the free fraction of phenylacetate increased, whereas the phenylbutyrate free fraction slightly decreased. We conclude that phenylacetate and phenylbutyrate have high free fractions that change with varying albumin levels and when both phenylacetate and phenylbutyrate are present together in plasma.
- Published
- 1996
- Full Text
- View/download PDF
44. Up-regulation and functional role of p21Waf1/Cip1 during growth arrest of human breast carcinoma MCF-7 cells by phenylacetate.
- Author
-
Gorospe M, Shack S, Guyton KZ, Samid D, and Holbrook NJ
- Subjects
- Animals, Antisense Elements (Genetics), Breast Neoplasms, Calcium-Calmodulin-Dependent Protein Kinases metabolism, Cell Division drug effects, Cyclin-Dependent Kinase 2, Cyclin-Dependent Kinase Inhibitor p21, Cyclin-Dependent Kinases genetics, Cyclin-Dependent Kinases metabolism, Cyclins genetics, Female, Fibroblasts cytology, Fibroblasts drug effects, Fibroblasts enzymology, Gene Expression Regulation, Neoplastic physiology, Humans, Mice, Mice, Knockout, Phosphorylation, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases metabolism, RNA, Messenger metabolism, Retinoblastoma Protein metabolism, Signal Transduction physiology, Tumor Cells, Cultured cytology, Tumor Cells, Cultured drug effects, Tumor Cells, Cultured enzymology, Up-Regulation drug effects, Antimetabolites, Antineoplastic pharmacology, CDC2-CDC28 Kinases, Cyclins metabolism, Enzyme Inhibitors metabolism, Phenylacetates pharmacology
- Abstract
Phenylacetate (PA) and related aromatic fatty acids constitute a novel class of relatively nontoxic antineoplastic agents. These compounds induce tumor cytostasis and growth inhibition and differentiation of cancer cells, but little is known regarding the molecular events mediating these biological effects. Using human breast carcinoma MCF-7 cells as a model, we show here that PA-induced growth arrest is associated with enhanced expression of the cyclin-dependent kinase inhibitor p21Waf1/Cip1 and dephosphorylation of the retinoblastoma protein (pRB). The induction of p21WAF1/CIP1 mRNA by PA was independent of the cellular p53 status. To directly assess the contribution of p21Waf1/Cip1 to PA-mediated cytostasis, we compared the effects of PA in parental MCF-7 cells and cells expressing reduced levels of p21Waf1/Cip1 protein (clones AS.3 and AS.4), accomplished through constitutive expression of antisense p21Waf1/Cip1 transcripts. In contrast to parental cells, AS.3 and AS.4 cells did not show reduced pRB phosphorylation following PA treatment, indicating that p21Waf1/Cip1 induction by PA is required for dephosphorylation (inactivation) of pRB, a known mediator of cell cycle control. A prominent role for p21Waf1/Cip1 in mediating PA-induced growth arrest was further supported by the demonstration that embryonal fibroblasts derived from a p21WAF1/CIP1 knockout mouse (p21-/- mouse embryonal fibroblasts) did not growth arrest following PA treatment, whereas PA effectively induced p21WAF1/CIP1 mRNA and growth inhibition of the wild-type mouse embryonal fibroblasts. Taken together, our findings strongly support a role for p21Waf1/Cip1 in the PA-mediated inhibition of cell growth.
- Published
- 1996
45. The differentiating agent phenylacetate increases prostate-specific antigen production by prostate cancer cells.
- Author
-
Walls R, Thibault A, Liu L, Wood C, Kozlowski JM, Figg WD, Sampson ML, Elin RJ, and Samid D
- Subjects
- Animals, Carcinoma pathology, Cell Differentiation, Cell Division drug effects, Dose-Response Relationship, Drug, Gene Expression Regulation drug effects, Humans, Male, Mice, Mice, Inbred BALB C, Prostate-Specific Antigen genetics, Prostatic Neoplasms pathology, RNA, Messenger metabolism, Rats, Tumor Cells, Cultured, Carcinoma metabolism, Phenylacetates pharmacology, Prostate-Specific Antigen biosynthesis, Prostatic Neoplasms metabolism
- Abstract
The prostatic-specific antigen (PSA) is the tumor marker most widely relied upon for the monitoring of patients with prostate cancer. Recently, declines in the serum concentrations of PSA have been advocated as a surrogate marker of tumor response in clinical trials of investigational antitumor agents. We examined the hypothesis that this postulate may not apply to the evaluation of drugs such as phenylacetate, a differentiating agent endowed with mechanisms of action different from those of classic cytotoxic chemotherapy. Using human prostatic carcinoma LNCaP cells as a model, we show that phenylacetate induces PSA production despite inhibition of tumor cell proliferation. Incubation of LNCaP cultures with cytostatic doses of phenylacetate (3-10 mM) resulted in a three- to fourfold increase in PSA secretion per cell. This appears to result from upregulation of PSA gene expression, as indicated by elevated PSA mRNA steady-state levels in treated cells. The increase in PSA production per cell was confirmed in rats bearing subcutaneous LNCaP tumor implants that were treated systemically with phenylacetate. Further comparative studies indicate that upregulation of PSA is common to various differentiation inducers, including all-trans-retinoic acid, 1,25-dihydroxyvitamin D3, and butyrate but is not induced by other antitumor agents of clinical interest such as suramin. We conclude that declines in PSA may be treatment specific and that the exclusive use of this criterion as a marker of disease response may mislead the proper evaluation of differentiating agents in prostate cancer patients.
- Published
- 1996
- Full Text
- View/download PDF
46. Vulnerability of multidrug-resistant tumor cells to the aromatic fatty acids phenylacetate and phenylbutyrate.
- Author
-
Shack S, Miller A, Liu L, Prasanna P, Thibault A, and Samid D
- Subjects
- ATP Binding Cassette Transporter, Subfamily B, Member 1 genetics, Catalase metabolism, Cell Differentiation drug effects, Cell Division drug effects, Doxorubicin pharmacology, Drug Resistance, Multiple, Glutathione metabolism, Humans, Tumor Cells, Cultured, Verapamil pharmacology, Antineoplastic Agents pharmacology, Drug Resistance, Neoplasm, Phenylacetates pharmacology, Phenylbutyrates pharmacology
- Abstract
Cytotoxic chemotherapies often give rise to multidrug resistance, which remains a major problem in cancer management. In pursuit of alternative treatments for chemoresistant tumor cells, we tested the response of multidrug-resistant (MDR) tumor cell lines to the aromatic fatty acids phenylacetate (PA) and phenylbutyrate (PB), two differentiation inducers currently in clinical trials. Both compounds induced cytostasis and maturation of multidrug-resistant breast, ovarian, and colon carcinoma cells with no significant effect on cell viability. In contrast to their poor response to doxorubicin, the MDR cells were generally more sensitive to growth arrest by PA and PB than their parental counterparts. The aromatic fatty acids, like the differentiation-inducing aliphatic fatty acid butyrate, up-regulated mdr-1 gene expression. However, while butyrate increased multidrug resistance, PA and PB potentiated the cytotoxic activity of doxorubicin against MDR cells. The latter was associated with time-dependent declines in glutathione levels and in the activity of superoxide dismutase, catalase, glutathione peroxidase, glutathione reductase, and glutathione S-transferase, the antioxidant enzymes implicated in cell resistance to free radical-based therapies. Taken together, our in vitro data indicate that PA and PB, differentiation inducers of the aromatic fatty acid class, may provide an alternative approach to the treatment of MDR tumors.
- Published
- 1996
47. Phenylacetate is an inhibitor of prostatic growth and development in organ culture.
- Author
-
Lipschutz JH, Samid D, and Cunha GR
- Subjects
- Animals, Animals, Newborn, Cell Differentiation drug effects, Immunohistochemistry, Male, Organ Culture Techniques, Prostate cytology, Prostatic Hyperplasia drug therapy, Rats, Rats, Sprague-Dawley, Antimetabolites, Antineoplastic pharmacology, Phenylacetates pharmacology, Prostate drug effects
- Abstract
Purpose: Benign prostatic hyperplasia (BPH) is the most common proliferative disease affecting men, and the obstructive uropathy it causes results in serious morbidity and financial cost. Phenylacetate (PA) is a small molecule that is a product of phenylalanine metabolism and is normally present in the mammalian circulation at very low levels. It has long been safely used in humans to treat the hyperammonemia resulting from urea synthesis disorders and liver failure. It has recently been investigated as an anticancer agent because it decreased growth and increased differentiation of a variety of human neoplasms, including prostate cancer in which a phase I trial has recently been completed., Materials and Methods: Because of PA's growth-inhibitory effects on a variety of cell lines and the idea that BPH is due to a reawakening of embryonic-like inductive activity in prostatic stromal cells, which then induce development of epithelial nodules, we examined the effect of PA on serum-free organ cultures of developing rat prostates., Results: We found that PA markedly decreased rat prostatic growth and ductal morphogenesis at concentrations that have previously been well tolerated in patients. In ventral prostates grown for 7 days in organ culture, histodifferentiation was inhibited as measured by a marked decrease in ductal lumen formation and ductal branching morphogenesis. This inhibition of differentiation was confirmed by using cytokeratin antibodies specific for basal and luminal cells. Synthesis of DNA was also significantly decreased per organ with PA. The growth inhibitory effects of PA were reversible, and the mechanism did not appear to be due to glutamine or glycine deprivation, or androgen receptor inhibition., Conclusions: In common with earlier studies, we found that PA inhibits prostatic growth; however, in our organ culture system, differentiation was also largely inhibited. These studies indicate that there may be a role for PA in treating BPH or in elucidating the mechanism by which it occurs since BPH apparently involves the neoformation of ductal-acinar tissue in aged men via mechanisms fundamentally similar if not identical to those in fetal prostatic development.
- Published
- 1996
48. Phase I study of lovastatin, an inhibitor of the mevalonate pathway, in patients with cancer.
- Author
-
Thibault A, Samid D, Tompkins AC, Figg WD, Cooper MR, Hohl RJ, Trepel J, Liang B, Patronas N, Venzon DJ, Reed E, and Myers CE
- Subjects
- Adult, Aged, Female, Humans, Lovastatin pharmacology, Male, Middle Aged, Neoplasms blood, Ubiquinone blood, Antineoplastic Agents adverse effects, Hydroxymethylglutaryl-CoA Reductase Inhibitors adverse effects, Lovastatin adverse effects, Neoplasms drug therapy
- Abstract
Lovastatin, an inhibitor of the enzyme 3-hydroxy-3-methylglutaryl-coenzyme A reductase (the major regulatory enzyme of the mevalonate pathway of cholesterol synthesis), displays antitumor activity in experimental models. We therefore conducted a Phase I trial to characterize the tolerability of lovastatin administered at progressively higher doses to cancer patients. From January 1992 to July 1994, 88 patients with solid tumors (median age, 57 +/- 14 years) were treated p.o. with 7-day courses of lovastatin given monthly at doses ranging from 2 to 45 mg/kg/day. The inhibitory effects of lovastatin were monitored through serum concentrations of cholesterol and ubiquinone, two end products of the mevalonate pathway. Concentrations of lovastatin and its active metabolites were also determined, by bioassay, in the serum of selected patients. Cyclical treatment with lovastatin markedly inhibited the mevalonate pathway, evidenced by reductions in both cholesterol and ubiquinone concentrations, by up to 43 and 49% of pretreatment values, respectively. The effect was transient, however, and its magnitude appeared to be dose independent. Drug concentrations reached up to 3.9 micrometer and were in the range associated with antiproliferative activity in vitro. Myopathy was the dose-limiting toxicity. Other toxicities included nausea, diarrhea, and fatigue. Treatment with ubiquinone was associated with reversal of lovastatin-induced myopathy, and its prophylactic administration prevented the development of this toxicity in a cohort of 56 patients. One minor response was documented in a patient with recurrent high-grade glioma. Lovastatin given p.o. at a dose of 25 mg/kg daily for 7 consecutive days is well tolerated. The occurrence of myopathy, the dose-limiting toxicity, can be prevented by ubiquinone supplementation. To improve on the transient inhibitory activity of this dosing regimen on the mevalonate pathway, alternative schedules based on uninterrupted administration of lovastatin should also be studied.
- Published
- 1996
49. Lipid metabolism as a target for brain cancer therapy: synergistic activity of lovastatin and sodium phenylacetate against human glioma cells.
- Author
-
Prasanna P, Thibault A, Liu L, and Samid D
- Subjects
- Antimetabolites, Antineoplastic pharmacology, Antimetabolites, Antineoplastic therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Brain Neoplasms drug therapy, Brain Neoplasms pathology, Cell Division drug effects, Drug Synergism, Glioma drug therapy, Glioma pathology, Humans, Lovastatin therapeutic use, Mevalonic Acid metabolism, Neoplasm Invasiveness, Phenylacetates therapeutic use, Tumor Cells, Cultured, Ubiquinone pharmacology, Antineoplastic Combined Chemotherapy Protocols pharmacology, Brain Neoplasms metabolism, Glioma metabolism, Lipid Metabolism, Lovastatin pharmacology, Phenylacetates pharmacology
- Abstract
Malignant gliomas, the most common form of primary brain tumors, are highly dependent on the mevalonate (MVA) pathway for the synthesis of lipid moieties critical to cell replication. Human glioblastoma cells were found to be uniquely vulnerable to growth arrest by lovastatin, a competitive inhibitor of the enzyme regulating MVA synthesis, 3-hydroxy-3-methylglutaryl coenzyme A reductase. The sodium salt of phenylacetic acid (NaPA), an inhibitor of MVA-pyrophosphate decarboxylase, the enzyme that controls MVA use, acted synergistically with lovastatin to suppress malignant growth. When used at pharmacologically attainable concentrations, the two compounds induced profound cytostasis and loss of malignant properties such as invasiveness and expression of the transforming growth factor-beta 2 gene, coding for a potent immunosuppressive cytokine. Supplementation with exogenous ubiquinone, an end product of the MVA pathway, failed to rescue the cells, suggesting that decreased synthesis of intermediary products are responsible for the antitumor effects observed. In addition to blocking the MVA pathway, lovastatin alone and in combination with NaPA increased the expression of the peroxisome proliferator-activated receptor, a transcription factor implicated in the control of lipid metabolism, cell growth, and differentiation. Our results indicate that targeting lipid metabolism with lovastatin, used alone or in combination with the aromatic fatty acid NaPA, may offer a novel approach to the treatment of malignant gliomas.
- Published
- 1996
- Full Text
- View/download PDF
50. Functional analysis of the promoter and first intron of the human lysyl oxidase gene.
- Author
-
Csiszar K, Entersz I, Trackman PC, Samid D, and Boyd CD
- Subjects
- Animals, Base Sequence, Extracellular Matrix physiology, Gene Expression Regulation, Neoplastic, Humans, Introns, Mice, Molecular Sequence Data, Osteosarcoma genetics, Proto-Oncogene Proteins p21(ras) physiology, RNA, Messenger genetics, Rats, Sequence Alignment, Sequence Homology, Nucleic Acid, Transcription, Genetic, Gene Expression Regulation, Enzymologic, Promoter Regions, Genetic, Protein-Lysine 6-Oxidase genetics
- Abstract
Alterations in the synthesis and activity of lysyl oxidase occur concomitant with developmental changes in collagen and elastin deposition and with the pathogenesis of several acquired and heritable connective tissue disorders. To begin to unravel the mechanisms that control lysyloxidase gene expression, we have previously reported the complete exon-intron structure of the human lysyl oxidase gene. We have now sequenced this entire gene, including all six introns and 4 kb of DNA 5' of exon 1. Analysis of over 13 kb of intervening sequence and 5' flanking sequence revealed a concentration of conserved consensus sequence elements within the first intron and 1 kb immediately 5' of exon 1. Analysis of intron 1 and the 5' flanking domain, using recombinant plasmids containing the chloramphenicol acetyl transferase (CAT) reporter gene, identified functional DNA sequence elements within these non-coding domains responsible for inhibition and up-regulation of CAT activity in primary cultures of human skin fibroblasts, in smooth muscle cells, revertant cells derived from an osteosarcoma cell line and malignant c-Ha-ras-transformed osteosarcoma cells. DNA sequence elements within intron 1, in particular, resulted in a marked increase in CAT reporter activity in cultured fibroblasts, smooth muscle cells and osteosarcoma cells. In c-Ha-ras-transformed osteosarcoma cells, however, no such enhancer activity of intron 1 sequence was observed. Ras-transformed osteosarcoma cells exhibited reduced steady-state levels of lysyl oxidase mRNA that was primarily controlled through reduced transcription of the lysyl oxidase gene. The lack of any up-regulation of CAT activity in these ras-transformed cells by sequence elements within intron 1 suggests a complex interaction between cis-acting domains and trans-acting transcriptional factors in the 5' promoter domain and the first intron of the lysyl oxidase gene.
- Published
- 1996
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.