32 results on '"J. Lathia"'
Search Results
2. Comparison Of Adjuvant Versus Salvage Therapy Among IDH Mutant Low-Grade Glioma
- Author
-
Jaleh Fallah, G.H. Barnett, David M. Peereboom, Glen Stevens, Manmeet Ahluwalia, Lilyana Angelov, M.C. Tom, Deborah Y. Park, Alireza M. Mohammadi, Jennifer S. Yu, Wei Wei, Erin S. Murphy, S. Tewari, John H. Suh, J. Lathia, and Samuel T. Chao
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Radiation ,business.industry ,medicine.medical_treatment ,Mutant ,Salvage therapy ,Internal medicine ,Medicine ,Radiology, Nuclear Medicine and imaging ,Low-Grade Glioma ,business ,Adjuvant - Published
- 2020
- Full Text
- View/download PDF
3. Abstract No. 522 Strategic targeting of hepatocellular carcinoma and its immune-microenvironment by a combination of Warburg reversal and mTOR inhibition
- Author
-
Gordon McLennan, D Das, E. Fagbongbe, J. Lathia, and D. Watson
- Subjects
business.industry ,Hepatocellular carcinoma ,Immune microenvironment ,Cancer research ,medicine ,Radiology, Nuclear Medicine and imaging ,Cardiology and Cardiovascular Medicine ,medicine.disease ,business ,PI3K/AKT/mTOR pathway - Published
- 2019
- Full Text
- View/download PDF
4. LAB-STEM CELLS
- Author
-
D. Kozono, M. Nitta, O. Sampetrean, N. Kimberly, D. Kushwaha, D. Merzon, K. Ligon, S. Zhu, K. Zhu, T. H. Kim, C.-H. Kwon, O. Becher, H. Saya, C. C. Chen, L. K. Donovan, S. M. Birks, V. Bosak, G. J. Pilkington, P. Mao, J. Li, K. Joshi, B. Hu, S. Cheng, R. W. Sobol, I. Nakano, M. Li, J. S. Hale, J. T. Myers, A. Y. Huang, C. Gladson, A. A. Sloan, J. N. Rich, J. D. Lathia, P. E. Hall, J. Gallagher, Q. Wu, M. Venere, E. Levy, M. S. Rani, P. Huang, E. Bae, J. Selfridge, L. Cheng, H. Guvenc, R. E. McLendon, A. E. Sloan, H. Phillips, A. Lai, M. Bredel, S. Bao, A. Hjelmeland, M. Sinyuk, P. Sathyan, J. Hale, P. Zinn, C. T. Carson, U. Naik, S. Majumder, L. A. Song, A. Vasanji, N. Tenley, A. B. Hjelmeland, P. Peruzzi, A. Bronisz, E. Antonio Chiocca, J. A. Godlewski, O. A. Guryanova, X. Fang, H.-M. C. Christel, C. Benito, G. Zoltan, B. Aline, S. Tilman, B. Josephine, M. Carolin, S. Thomas, G. Violaine, A. Unterberg, V. Capilla-Gonzalez, H. Guerrero-Cazares, A. Cebrian-Silla, J. M. Garcia-Verdugo, A. Quinones-Hinojosa, J. Man, J. Shoemake, J. Rich, J. Yu, X. He, F. DiMeco, A. L. Vescovi, J. A. Heth, K. M. Muraszko, X. Fan, S. A. Nguyen, O. D. Stechishin, H. A. Luchman, J. J. Kelly, J. G. Cairncross, S. Weiss, Y. Kim, E. Kim, O. O. Guryanova, M. Hitomi, J. Lathia, D. Serwanski, J. Robert, J. Lee, A. Nishiyama, J. K. Liu, W. A. Flavahan, N. Fernandez, M. Wu, S. Das, E. Bazzoli, T. Pulvirenti, M. C. Oberstadt, F. Perna, W. Boyoung, N. Schultz, J. T. Huse, E. I. Fomchenko, F. Voza, V. Tabar, C. W. Brennan, L. M. DeAngelis, S. D. Nimer, E. C. Holland, M. Squatrito, Y.-H. Chen, D. H. Gutmann, S.-H. Kim, M. K. Lee, Y.-J. Chwae, B. C. Yoo, K.-H. Kim, A. Soeda, A. Hara, T. Iwama, D. M. Park, A. Golebiewska, S. Bougnaud, D. Stieber, N. H. Brons, L. Vallar, F. Hertel, R. Bjerkvig, S. P. Niclou, P. Hamerlik, R. Rasmussen, D. Fricova, B. Jiri, A. Schulte, A. Kathagen, S. Zapf, H. Meissner, H. S. Phillips, M. Westphal, K. Lamszus, M. Sanzey, S. K. Singh, A. Vartanian, J. Gumin, E. P. Sulman, F. F. Lang, G. Zadeh, N. S. Bayin, A. Dietrich, T. Abel, M. V. Chao, H.-R. Song, C. J. Buchholz, D. Placantonakis, M. Esencay, D. Zagzag, I. V. Balyasnikova, M. S. Prasol, S. D. Ferguson, A. U. Ahmed, Y. Han, M. S. Lesniak, M. E. Barish, C. E. Brown, K. Herrmann, S. Argalian, M. Gutova, Y. Tang, A. Annala, R. A. Moats, L. Y. Ghoda, K. S. Aboody, S. Gadani, J. Adkins, A. Vsanji, R. McLendon, A. Chenn, D. Park, C. Dictus, S. Friauf, N. A. Valous, N. Grabe, B. Muerle, A. W. Unterberg, C. C. Herold-Mende, H. K. Lee, S. Finniss, E. Buchris, A. Ziv-Av, S. Casacu, C. Xiang, K. Bobbit, S. A. Rempel, T. Mikkelsen, S. Slavin, C. Brodie, D.-H. Woo, Y. Oh, M. Kim, D.-H. Nam, Q. Li, S. Salas, C. Pendleton, O. Wijesekera, D. Chesler, J. Wang, C. Smith, A. Levchenko, Q. LaPlant, K. Pitter, A.-M. Bleau, K. Helmy, J. Werbeck, L. Barrett, F. Shimizu, R. Benezra, E. Holland, Q. Chu, E. Bar, B. Orr, C. G. Eberhart, R. S. Schmid, R. E. Bash, A. M. Werneke, K. K. White, C. R. Miller, F. Agasse, N. Jhaveri, F. M. Hofman, T. C. Chen, A. Natsume, T. Wakabayashi, Y. Kondo, N. Chang, E. Moon, R. Kanai, S. Yip, A. Kimura, S. Tanaka, E. Rheinbay, D. Cahill, W. Curry, G. Mohapatra, J. Iafrate, A. Chi, R. Martuza, S. Rabkin, H. Wakimoto, C. Cusulin, J. A. Frank, and A. J. Annala
- Subjects
Abstracts ,Cancer Research ,Oncology ,business.industry ,Medicine ,Neurology (clinical) ,Stem cell ,business ,Cell biology - Published
- 2012
- Full Text
- View/download PDF
5. Cell Biology and Signaling
- Author
-
M. Agarwal, R. Nitta, S. Dovat, G. Li, H. Arita, Y. Narita, S. Fukushima, K. Tateishi, Y. Matsushita, A. Yoshida, Y. Miyakita, M. Ohno, V. P. Collins, N. Kawahara, S. Shibui, K. Ichimura, S. A. Kahn, S. Gholamin, M.-P. Junier, H. Chneiweiss, I. Weissman, S. Mitra, S. Cheshier, T. Avril, A. Hamlat, P.-J. Le Reste, J. Mosser, V. Quillien, C. Carrato, A. Munoz-Marmol, L. Serrano, L. Pijuan, C. Hostalot, S. l. Villa, A. Ariza, O. Etxaniz, C. Balana, E. T. Benveniste, Y. Zheng, B. McFarland, D. Drygin, S. Bellis, M. Bredel, D. Lotsch, C. Engelmaier, S. Allerstorfer, M. Grusch, J. Pichler, S. Weis, J. Hainfellner, C. Marosi, S. Spiegl-Kreinecker, W. Berger, A. Bronisz, M. O. Nowicki, Y. Wang, K. Ansari, E. A. Chiocca, J. Godlewski, K. Brown, M. Kwatra, T. Bui, S. Zhu, D. Kozono, J. Li, D. Kushwaha, B. Carter, C. Chen, J. Schulte, M. Srikanth, S. Das, J. Zhang, J. Lathia, L. Yin, J. Rich, E. Olson, J. Kessler, A. Chenn, A. Cherry, B. Haas, Y. H. Lin, S.-E. Ong, N. Stella, C. P. Cifarelli, R. J. Griffin, D. Cong, W. Zhu, Y. Shi, P. Clark, J. Kuo, S. Hu, D. Sun, M. Bookland, N. Darbinian, A. Dey, M. Robitaille, M. Remke, D. Faury, C. Maier, A. Malhotra, N. Jabado, M. Taylor, S. Angers, A. Kenney, X. Ren, H. Zhou, M. Schur, A. Baweja, M. Singh, A. Erdreich-Epstein, J. Fu, D. Koul, J. Yao, N. Saito, S. Zheng, R. Verhaak, Z. Lu, W. K. A. Yung, G. Gomez, S. Volinia, C. Croce, C. Brennan, W. Cavenee, F. Furnari, S. G. Lopez, D. Qu, C. Petritsch, M. Gonzalez-Huarriz, G. Aldave, D. Ravi, A. Rubio, R. Diez-Valle, M. Marigil, P. Jauregi, B. Vera, A. A. d. l. Rocha, S. Tejada-Solis, M. M. Alonso, U. Gopal, J. Isaacs, M. Gruber-Olipitz, S. Dabral, S. Ramkissoon, A. Kung, E. Pak, J. Chung, M. Theisen, Y. Sun, V. Monrose, Y. Franchetti, D. Shulman, N. Redjal, B. Tabak, R. Beroukhim, J. Zhao, S. Buonamici, K. Ligon, J. Kelleher, R. Segal, D. Canton, P. Diaz, J. Scott, K. Hara, T. Kageji, Y. Mizobuchi, K. Kitazato, T. Okazaki, T. Fujihara, K. Nakajima, H. Mure, K. Kuwayama, T. Hara, S. Nagahiro, L. Hill, H. Botfield, K. Hossain-Ibrahim, A. Logan, G. Cruickshank, Y. Liu, M. Gilbert, N. Kyprianou, V. Rangnekar, C. Horbinski, Y. Hu, C. Vo, Z. Li, C. Ke, N. Ru, K. R. Hess, M. E. Linskey, Y.-a. H. Zhou, F. Hu, K. Vinnakota, S. Wolf, H. Kettenmann, P. J. Jackson, J. D. Larson, D. A. Beckmann, B. S. Moriarity, D. A. Largaespada, S. Jalali, S. Agnihotri, S. Singh, K. Burrell, S. Croul, G. Zadeh, S.-H. Kang, M. O. Yu, N.-H. Song, K.-J. Park, S.-G. Chi, Y.-G. Chung, S. K. Kim, J. W. Kim, J. Y. Kim, J. E. Kim, S. H. Choi, T. M. Kim, S.-H. Lee, S.-K. Kim, S.-H. Park, I. H. Kim, C.-K. Park, H.-W. Jung, M. Koldobskiy, I. Ahmed, G. Ho, A. Snowman, E. Raabe, C. Eberhart, S. Snyder, I. Gugel, A. Bornemann, G. Pantazis, S. Mack, D. Shih, N. Sabha, M. Tatagiba, B. Krischek, A. Schulte, K. Liffers, A. Kathagen, S. Riethdorf, M. Westphal, K. Lamszus, J. S. Lee, J. Xiao, P. Patel, J. Schade, J. Wang, B. Deneen, H.-R. Song, L. Leiss, C. Gjerde, H. Saed, A. Rahman, M. Lellahi, P. O. Enger, R. Leung, O. Gil, L. Lei, P. Canoll, S. Sun, D. Lee, A. S. W. Ho, J. K. S. Pu, X.-q. Zhang, N. P. Lee, P. J. R. Dat, G. K. K. Leung, D. Loetsch, E. Steiner, K. Holzmann, C. Pirker, J. Hlavaty, H. Petznek, B. Hegedus, T. Garay, T. Mohr, W. Sommergruber, W. J. Lukiw, B. M. Jones, Y. Zhao, S. Bhattacharjee, F. Culicchia, N. Magnus, D. Garnier, B. Meehan, S. McGraw, M. Hashemi, T. H. Lee, C. Milsom, N. Gerges, J. Trasler, R. Pawlinski, N. Mackman, J. Rak, Z. Maherally, A. Thorne, Q. An, E. Barbu, H. Fillmore, G. Pilkington, S. L. Tan, S. Tan, S. Choi, C. Potts, D. A. Ford, Z. Nahle, A. M. Kenney, L. Matlaf, S. Khan, A. Zider, E. Singer, C. Cobbs, L. Soroceanu, B. C. McFarland, S. W. Hong, R. Rajbhandari, G. B. Twitty, G. K. Gray, H. Yu, E. N. Benveniste, S. E. Nozell, M. Minata, S. Kim, P. Mao, J. Kaushal, I. Nakano, T. Mizowaki, T. Sasayama, K. Tanaka, K. Mizukawa, M. Nishihara, S. Nakamizo, H. Tanaka, M. Kohta, K. Hosoda, E. Kohmura, S. Moeckel, K. Meyer, P. Leukel, U. Bogdahn, M. J. Riehmenschneider, A. K. Bosserhoff, R. Spang, P. Hau, A. Mukasa, A. Watanabe, H. Ogiwara, H. Aburatani, J. Mukherjee, S. Obha, W. See, R. Pieper, R. Otsuka, D. Kung, T. Sinha, G. Meares, S. Nozell, M. Ott, U. Litzenburger, K. Rauschenbach, L. Bunse, S. Pusch, K. Ochs, F. Sahm, C. Opitz, A. von Deimling, W. Wick, M. Platten, P. Peruzzi, R. Read, T. Fenton, J. Wykosky, S. Vandenberg, I. Babic, A. Iwanami, H. Yang, P. Mischel, J. Thomas, M. W. Ronellenfitsch, A. L. Thiepold, P. N. Harter, M. Mittelbronn, J. P. Steinbach, Y. Rybakova, A. Kalen, E. Sarsour, P. Goswami, J. Silber, G. Harinath, B. Aldaz, A. W. M. Fabius, S. Turcan, T. A. Chan, J. T. Huse, A. M. Sonabend, M. Bansal, P. Guarnieri, C. Soderquist, J. Yun, B. Kennedy, J. Sisti, S. Bruce, R. Bruce, R. Shakya, T. Ludwig, S. Rosenfeld, P. A. Sims, J. N. Bruce, A. Califano, M.-T. Stockhausen, K. Kristoffersen, L. S. Olsen, H. S. Poulsen, B. Stringer, B. Day, G. Barry, M. Piper, P. Jamieson, K. Ensbey, Z. Bruce, L. Richards, A. Boyd, A. Sufit, T. Burleson, J. P. Le, A. K. Keating, T. Sundstrom, J. K. Varughese, P. Harter, L. Prestegarden, K. Petersen, F. Azuaje, C. Tepper, E. Ingham, L. Even, S. Johnson, K. O. Skaftnesmo, M. Lund-Johansen, R. Bjerkvig, K. Ferrara, F. Thorsen, H. Takeshima, S. Yamashita, K. Yokogami, S. Mizuguchi, H. Nakamura, J. Kuratsu, T. Fukushima, K. Morishita, Y. Tang, D. Vaka, S. Chen, A. Ponnuswami, Y.-J. Cho, M. Monje, T. Nakamura, D. Cahill, K. Tiemann, H. Hedman, S. P. Niclou, M. Timmer, R. Tjiong, G. Rohn, R. Goldbrunner, P. Stavrinou, M. Perrech, M. Tokita, S. Mikheev, D. Sellers, A. Mikheev, Y. Kosai, R. Rostomily, I. Tritschler, K. Seystahl, J. J. Schroeder, M. Weller, A. Wade, A. E. Robinson, J. J. Phillips, Y. Gong, Y. Ma, Z. Cheng, R. Thompson, Q.-W. Fan, C. Cheng, W. Gustafson, E. Charron, P. Zipper, R. Wong, J. Chen, J. Lau, C. Knobbe-Thosen, N. Jura, G. Reifenberger, K. Shokat, W. Weiss, S. Wu, J. Hu, T. Taylor, G. R. Villa, P. S. Mischel, S. L. Gonias, D. Yamashita, T. Kondo, H. Takahashi, A. Inoue, S. Kohno, H. Harada, S. Ohue, T. Ohnishi, P. Li, J. Ng, L. Yuelling, F. Du, T. Curran, Z.-j. Yang, D. Zhu, R. C. Castellino, E. G. Van Meir, G. Begum, Q. Wang, S.-S. Yang, S.-H. Lin, and K. Kahle
- Subjects
Cancer Research ,Tumor suppressor gene ,Central nervous system ,030204 cardiovascular system & hematology ,Biology ,urologic and male genital diseases ,Abstracts ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Progenitor cell ,Transcription factor ,030304 developmental biology ,0303 health sciences ,urogenital system ,Cell growth ,medicine.disease ,nervous system diseases ,3. Good health ,Cell biology ,medicine.anatomical_structure ,NFIB ,Oncology ,Cell culture ,Neurology (clinical) ,Glioblastoma - Abstract
The phylogenetically-conserved vertebrate transcription factor, NFIB, is an orchestrator of glial differentiation in the developing mammalian central nervous system. We found NFIB expression to be reduced in glioblastoma (GBM), the commonest and most lethal primary adult brain cancer, so investigated what effect increased expression of NFIB had on GBM. Increased expression of NFIB in primary GBM cell lines induced expression of markers of glial differentiation, inhibited cell proliferation, reduced stem/progenitor cell growth, altered cell cycle progression and inhibited tumor growth in murine models of GBM. We thus identified NFIB to be a novel tumor suppressor gene in GBM.
- Published
- 2010
- Full Text
- View/download PDF
6. SC-11DIFFERENTIAL CONNEXIN FUNCTION ENHANCES SELF-RENEWAL IN GLIOBLASTOMA
- Author
-
M. Hitomi, L. Deleyrolle, M. Sinyuk, B. Otvos, S. Brunet, A. Jarrar, W. Flavahan, W. Goan, E. Mulkearns-Hubert, A. Zhang, M. Rohaus, M. Oli, V. Vedam-Mai, J. Fortin, H. Futch, Q. Wu, M. Ahluwalia, J. Rich, B. Reynolds, and J. Lathia
- Subjects
Cancer Research ,Abstracts ,Oncology ,Neurology (clinical) ,sense organs - Abstract
The coordination of complex tumor processes requires cells to rapidly modify their phenotypes using direct cell-cell communication through gap junction channels composed of connexins. Previous reports suggest that gap junctions are tumor suppressors based on connexin 43 (Cx43), but this hypothesis fails to consider the differences in connexin-mediated intercellular communication rate and ion selectivity that drive gap junction diversity. Using patient-derived specimens, we screened connexin proteins and found that glioblastoma cancer stem cells (CSCs) expressed Cx46, while Cx43 was predominantly expressed in non-CSCs. Targeting Cx46 compromised CSC proliferation, self-renewal, and tumor initiation. Consistent with the divergent physiological nature of intercellular communication and ion selectivity between Cx46 and Cx43, CSCs with higher Cx46 had an elevated intercellular communication rate and were more depolarized than non-CSCs. The difference in connexin subtype was responsible for these phenotypic differences; Cx46 knockdown reduced the communication rate of CSCs, and Cx43 knockdown increased the depolarization of non-CSCs. The differences between the two connexins were reflected in GBM patient survival: Cx46 expression correlated with poor prognosis, while Cx43 expression was not informative. Ongoing studies are identifying differentially transported signaling molecules that are responsible for CSC maintenance based on connexin subunits. As clinically relevant gap junction inhibitors including 1-Octanol are being tested for other neurological disorders (essential tremor), we evaluated if these inhibitors were effective in glioblastoma. We confirmed that CSCs possessed functional gap junctions and that inhibitors reduced communication. These inhibitors potently inhibited proliferation and self-renewal of CSCs compared with non-CSCs and neural progenitor cells. In established xenograft tumors, gap junction inhibitors suppressed tumor growth and had an additive effect when combined with Temozolomide. Taken together, our data demonstrate a pro-tumorigenic role of gap junctions that is dependent on connexin subunit expression and provide a rationale for gap junction targeting in glioblastoma.
- Published
- 2014
7. 552 Combined autophagy and connexin inhibition: A novel therapy for intermediate stage melanomas
- Author
-
Barbara Bedogni, J. Sha, Joshua Arbesman, and J. Lathia
- Subjects
Chemistry ,Autophagy ,Cancer research ,Connexin ,Cell Biology ,Dermatology ,Molecular Biology ,Biochemistry ,Intermediate stage - Published
- 2016
- Full Text
- View/download PDF
8. STEM CELLS
- Author
-
L. Cheng, Z. Huang, W. Zhou, Q. Wu, J. Rich, S. Bao, P. Baxter, H. Mao, X. Zhao, Z. Liu, Y. Huang, H. Voicu, S. Gurusiddappa, J. M. Su, L. Perlaky, R. Dauser, H.-c. E. Leung, K. M. Muraszko, J. A. Heth, X. Fan, C. C. Lau, T.-K. Man, M. Chintagumpala, X.-N. Li, P. Clark, M. Zorniak, Y. Cho, X. Zhang, D. Walden, E. Shusta, J. Kuo, S. Sengupta, S. Goel-Bhattacharya, S. Kulkarni, B. Cochran, C. Cusulin, A. Luchman, S. Weiss, M. Wu, N. Fernandez, S. Agnihotri, R. Diaz, J. Rutka, M. Bredel, J. Karamchandani, S. Das, B. Day, B. Stringer, F. Al-Ejeh, M. Ting, J. Wilson, K. Ensbey, P. Jamieson, Z. Bruce, Y. C. Lim, C. Offenhauser, S. Charmsaz, L. Cooper, J. Ellacott, A. Harding, J. Lickliter, P. Inglis, B. Reynolds, D. Walker, M. Lackmann, A. Boyd, A. Berezovsky, L. Poisson, L. Hasselbach, S. Irtenkauf, A. Transou, T. Mikkelsen, A. C. deCarvalho, D. Emlet, C. Del Vecchio, P. Gupta, G. Li, S. Skirboll, A. Wong, J. Figueroa, T. Shahar, A. Hossain, F. Lang, S. Fouse, J. Nakamura, C. D. James, S. Chang, J. Costello, J. M. Frerich, S. Rahimpour, Z. Zhuang, J. D. Heiss, A. Golebiewska, D. Stieber, L. Evers, E. Lenkiewicz, N. H. C. Brons, N. Nicot, A. Oudin, S. Bougnaud, F. Hertel, R. Bjerkvig, M. Barrett, L. Vallar, S. P. Niclou, X. Hao, J. Rahn, E. Ujack, X. Lun, G. Cairncross, D. Senger, S. Robbins, J. Harness, R. Lerner, Y. Ihara, R. Santos, J. D. L. Torre, A. Lu, T. Ozawa, T. Nicolaides, D. James, C. Petritsch, D. Higgins, M. Schroeder, B. Ball, B. Milligan, F. Meyer, J. Sarkaria, J. Henley, W. Flavahan, M. Hitomi, N. Rahim, Y. Kim, A. Sloan, R. Weil, I. Nakano, M. Li, J. Lathia, A. Hjelmeland, M. Kaluzova, S. Platt, M. Kent, A. Bouras, R. Machaidze, C. Hadjipanayis, S.-G. Kang, S.-H. Kim, Y.-M. Huh, E.-H. Kim, E.-K. Park, J. H. Chang, S. H. Kim, Y. K. Hong, D. S. Kim, S.-J. Lee, E. H. Kim, S. G. Kang, L. Deleyrolle, M. Sinyuk, W. Goan, B. Otvos, M. Rohaus, M. Oli, V. Vedam-Mai, D. Schonberg, S.-T. Lee, K. Chu, S. K. Lee, M. Kim, J.-K. Roh, A. Griveau, B. Reichholf, M. McMahon, D. Rowitch, R. Nitta, S. Mitra, M. Agarwal, T. Bui, J. Lin, C. Adamson, J. Martinez-Quintanilla, S.-H. Choi, D. Bhere, P. Heidari, D. He, U. Mahmood, K. Shah, S. Gholamin, A. Feroze, A. Achrol, S. Kahn, I. Weissman, S. Cheshier, E. P. Sulman, Q. Wang, E. Mostovenko, H. Liu, C. F. Lichti, A. Shavkunov, R. A. Kroes, J. R. Moskal, C. A. Conrad, F. F. Lang, M. R. Emmett, C. L. Nilsson, S. Osuka, O. Sampetrean, T. Shimizu, I. Saga, N. Onishi, E. Sugihara, J. Okubo, S. Fujita, S. Takano, A. Matsumura, H. Saya, N. Saito, J. Fu, S. Wang, W. K. A. Yung, D. Koul, R. S. Schmid, D. M. Irvin, M. Vitucci, R. E. Bash, A. M. Werneke, C. R. Miller, N. Shinojima, T. Takezaki, J. Fueyo, J. Gumin, F. Gao, F. Nwajei, F. C. Marini, M. Andreeff, J.-I. Kuratsu, S. Singh, K. Burrell, E. Koch, S. Jalali, A. Vartanian, E. Sulman, B. Wouters, G. Zadeh, R. Spelat, E. Singer, L. Matlaf, S. McAllister, L. Soroceanu, S. Spiegl-Kreinecker, D. Loetsch, M. Laaber, C. Schrangl, A. Wohrer, J. Hainfellner, C. Marosi, J. Pichler, S. Weis, G. Wurm, G. Widhalm, E. Knosp, W. Berger, J.-i. Kuratsu, Q. Tam, S. Tanaka, M. Nakada, D. Yamada, T. Todo, Y. Hayashi, J.-i. Hamada, A. Hirao, J. Tilghman, M. Ying, J. Laterra, M. Venere, C. Chang, M. Summers, S. Rosenfeld, S. Luk, J. Iafrate, D. Cahill, R. Martuza, S. Rabkin, A. Chi, H. Wakimoto, H.-G. Wirsching, S. Krishnan, K. Frei, N. Krayenbuhl, G. Reifenberger, M. Weller, G. Tabatabai, J. Man, J. Shoemake, and J. Yu
- Subjects
Cancer Research ,Abstracts ,Oncology ,Neurology (clinical) - Published
- 2011
9. Sleep disorders associated with cranial radiation-A systematic review.
- Author
-
Pascoe M, Byrne E, King A, Cooper D, Foldvary-Schaefer N, Mehra R, Lathia J, Gilbert MR, and Armstrong TS
- Subjects
- Humans, Sleep Wake Disorders etiology, Brain Neoplasms radiotherapy, Cranial Irradiation adverse effects
- Abstract
Background: Radiation is the standard-of-care treatment for primary brain tumors (PBTs) but may have profound effects on sleep that have not yet been fully characterized. This systematic review aims to further our understanding of radiation therapy on the risk of development of sleep disorders in patients with PBTs, as well as potential opportunities for prevention and treatment., Methods: A systematic search of PubMed, Embase, and Web of Science was performed (last Jan 2024) with predefined inclusion (PBT patients, radiation therapy, somnolence/circadian disruption) and exclusion (reviews/abstracts/cases/chapters, non-PBT cancer, lack of radiation) criteria, yielding 267 papers initially and 38 studies included. Data extraction and analysis (descriptive statistics, individual study summary) focused on the incidence of sleep disturbances, radiation types/doses, and pharmacologic interventions. Risk of bias assessment was conducted with the Effective Public Health Practice Project's Quality Assessment Tool for Quantitative Studies., Results: The included 38 studies (n = 2948 patients) demonstrated a high incidence of sleep disturbances in patients with PBTs throughout radiation therapy, but primarily from the end of radiation to 6 months after. Sleep symptoms were associated with radiation (dose-dependent), and pharmacotherapies were helpful in patients with formal sleep disorder diagnoses. Terminology and incidence reporting of sleep symptoms are inconsistent, and many studies had a high risk of bias., Conclusions: This systematic review highlights the ongoing challenges with sleep symptoms/disorders related to cranial irradiation treatment in the primary brain tumor population. Further investigations on the interconnectedness of sleep disturbance constructs and possible pharmacotherapies to alleviate symptoms are warranted., (Published by Oxford University Press on behalf of the Society for Neuro-Oncology 2024.)
- Published
- 2025
- Full Text
- View/download PDF
10. A brave new framework for glioma drug development.
- Author
-
Hotchkiss KM, Karschnia P, Schreck KC, Geurts M, Cloughesy TF, Huse J, Duke ES, Lathia J, Ashley DM, Nduom EK, Long G, Singh K, Chalmers A, Ahluwalia MS, Heimberger A, Bagley S, Todo T, Verhaak R, Kelly PD, Hervey-Jumper S, de Groot J, Patel A, Fecci P, Parney I, Wykes V, Watts C, Burns TC, Sanai N, Preusser M, Tonn JC, Drummond KJ, Platten M, Das S, Tanner K, Vogelbaum MA, Weller M, Whittle JR, Berger MS, and Khasraw M
- Subjects
- Humans, Antineoplastic Agents therapeutic use, Brain Neoplasms drug therapy, Brain Neoplasms pathology, Glioma drug therapy, Glioma pathology, Drug Development
- Abstract
Patients with brain tumours are motivated to participate in clinical trials involving repeat tissue sampling. Normalising the use of neoadjuvant and staged surgical trials necessitates collaboration among patients, regulatory agencies, and researchers. Initial and repetitive tissue sampling plays a crucial role in enhancing our understanding of resistance mechanisms and vulnerabilities in brain tumour therapy. Standardising biopsy techniques and ensuring technical uniformity across institutions are vital for effective interinstitutional collaboration. Although liquid biopsy technologies hold promise, they are not yet ready to replace tissue analysis. Clear communication about the risks and benefits of biopsies is essential, particularly regarding potential postoperative deficits. Changes in mindset and neurosurgical culture are imperative to achieve much needed breakthroughs in the development of new, effective therapies for brain tumours., Competing Interests: Declaration of interests MG declares research grant support from Evgen Pharm. KCS declares honoraria from Springworks Therapeutics and Novartis; declares research funding to her institution from Springworks Therapeutics; and serves on a data and safety monitoring board for Advarra. TFC is cofounder, major stock holder, consultant, and board member of Katmai Pharmaceuticals; holds stock for Erasca; is a member of the board and paid consultant for the 501c3 Global Coalition for Adaptive Research; holds stock in Chimerix and receives milestone payments and possible future royalties; is a member of the scientific advisory board for Break Through Cancer foundation and Cure Brain Cancer Foundation; has provided paid consulting services to Symbio, Mundipharma, Tango BlueRock, Vida Ventures, Lista Therapeutics, Stemline, Novartis, Roche, Sonalasense, Sagimet, Clinical Care Options, Ideology Health, Servier, Jubilant, Immvira, Gan & Lee, BrainStorm, Katmai, Sapience, Inovio, Vigeo Therapeutics, DNATrix, Tyme, SDP, Kintara, Bayer, Merck, Boehringer Ingelheim, VBL, Amgen, Kiyatec, AbbVie, VBI, Deciphera, Agios, Novocure, and Medscape; and has contracts with UCLA for the Brain Tumour Programme with Roche, VBI, Merck, Novartis, and Bristol Myers Squibb. The Regents of the University of California, TFC's employer, has licensed intellectual property co-invented by TFC to Katmai Pharmaceuticals. GL is consultant advisor for Agenus, Amgen, Array Biopharma, AstraZeneca, Bayer, BioNTech, Boehringer Ingelheim, Bristol Myers Squibb, Evaxion, Hexal AG (Sandoz Company), Highlight Therapeutics, IOBiotech, Immunocore, Innovent Biologics USA, Merck Sharpe & Dohme, Novartis, PHMR, Pierre Fabre, and Regeneron. AC declares research funding and honoraria from AstraZeneca, Benevolent AI, Duke Street Bio, and Evgen Pharmaceuticals; and has received honoraria from Storm Therapeutics. MSA declares a grant from Seagen; declares consulting fees from Bayer, Kiyatec, Insightec, GSK, Xoft, Nuvation, SDP Oncology, Apollomics, Prelude, Janssen, Voyager Therapeutics, Viewray, Caris Lifesciences, Pyramid Biosciences, Varian Medical Systems, Cairn Therapeutics, Anheart Therapeutics, Theraguix, Menarini Ricerche, Sumitomo Pharma Oncology, Autem Therapeutics, GT Medical Technologies, Allovir, and Equillium Bio; is on the data and safety monitoring board for VBI Vaccines; is on the scientific advisory board of Modifi biosciences and Bugworks; and is a shareholder for Mimivax, Cytodyn, MedInnovate Advisors, and Trisalus Lifesciences. SB reports consulting fees from and is a member of the advisory boards of Telix, Servier, Bayer, and Novocure; and research grants from Incyte, GSK, Lilly, Kite, and Novocure. JdG reports being an advisory board member for Kintara Pharmaceuticals, Kazia, MundiPharma, Insightec, Monteris, Carthera, Samus, Sapience, DSP Pharma, Telix, Servier, Alpha Pharmaceuticals, Nervianos, and CapitalOne; is a data safety monitoring board member for Chimerix and VBI; and has consulted for MundiPharma, Insightec, Carthera, Kintara, Deciphera, Kazia, and Nervianos. IP reports research funding from Merck. TCB reports consulting roles for Predicine; has received financial support from AbbVie and Predicine, with consulting fees paid to Mayo Clinic; and has been a member of the advisory board for Neurametrix. MPr reports honoraria from the following for-profit companies: Bayer, Bristol Myers Squibb, Novartis, Gerson Lehrman Group, CMC Contrast, GSK, Mundipharma, Roche, BMJ Journals, MedMedia, AstraZeneca, AbbVie, Lilly, Medahead, Daiichi Sankyo, Sanofi, Merck Sharp & Dohme, Tocagen, Adastra, Gan & Lee Pharmaceuticals, Janssen, Servier, Miltenyi, Boehringer Ingelheim, Telix, and Medscape. JCT reports research grants from Novocure and Munich Surgical Imaging; being on the advisory board of Advanced Accelerator Applications, Novartis; and Servier; and has received royalties from Springer Publishing. MPl reports being a founder of Tcelltech; has received research support from Bayer, Roche, Pfizer, and Merck; and is an advisory board member of Bayer and Servier. SD is speaker for the Congress of Neurological Surgeons (CNS) and the American Association of Neurological Surgeons; receives research funding from Alkermes; is a member of the Subcortical Surgery Group advisory board; has received royalties from Oxford University Press; and is provincial lead for CNS Oncology at Ontario Health. KT reports consulting fees from Oncohereos Biosciences; and advisory board roles for Advanced Accelerator Applications, Novartis; Sage Therapeutics; FYR Diagnostics; Cordance Medical; Telo Therapeutics; and Modifi Bio. MAV reports clinical trial funding to their institution from DeNovo Pharma, Infuseon, and Oncosynergy; and received honoraria fees from Biodexa and Servier Pharma. MW reports research grants from Novartis, Quercis, and Versameb; honoraria for advisory boards from Roche and Servier; and honoraria for consultation from Bayer, Curevac, Medac, Neurosense, Novartis, Novocure, Orbus, Philogen, and Servier. JRW reports research funding from AnHeart Therapeutics to their institution; received consulting fees from AnHeart Therapeutics and Servier; being on advisory boards for Roche and Merck; is a data safety monitoring member for Telix Pharmaceuticals; and is an employee of The Walter and Eliza Hall Institute and could be eligible for milestone and royalty payments related to Venetoclax. MK reports research grants from Bristol Myers Squibb, AbbVie, BioNTech, CNS Pharmaceuticals, Daiichi Sankyo, Immorna Therapeutics, Immvira Therapeutics, and Personalis; received consulting fees from The Jackson Lab for Genomic Research, AnHeart Therapeutics, Berg Pharma, George Clinical, Manarini Stemline, and Servier; received honoraria from GSK; and is on a data safety monitoring board for BPGbio. All other authors declare no competing interests., (Copyright © 2024 Elsevier Ltd. All rights reserved, including those for text and data mining, AI training, and similar technologies.)
- Published
- 2024
- Full Text
- View/download PDF
11. LCK facilitates DNA damage repair by stabilizing RAD51 and BRCA1 in the nucleus of chemoresistant ovarian cancer.
- Author
-
Dey G, Bharti R, Braley C, Alluri R, Esakov E, Crean-Tate K, McCrae K, Joehlin-Price A, Rose PG, Lathia J, Gong Z, and Reizes O
- Subjects
- Animals, Female, Humans, Mice, BRCA1 Protein genetics, Carcinoma, Ovarian Epithelial drug therapy, Cell Line, Tumor, DNA Damage, DNA Repair, Homologous Recombination, Poly(ADP-ribose) Polymerase Inhibitors pharmacology, Poly(ADP-ribose) Polymerase Inhibitors therapeutic use, Rad51 Recombinase genetics, Rad51 Recombinase metabolism, Carcinoma, Endometrioid drug therapy, Ovarian Neoplasms drug therapy, Ovarian Neoplasms genetics, Ovarian Neoplasms pathology
- Abstract
Poly-ADP Ribose Polymerase (PARP) targeted therapy is clinically approved for the treatment of homologous recombination (HR) repair deficient tumors. The remarkable success of this therapy in the treatment of HR repair deficient cancers has not translated to HR-proficient cancers. Our studies identify the novel role of non-receptor lymphocyte-specific protein tyrosine kinase (LCK) in the regulation of HR repair in endometrioid epithelial ovarian cancer (eEOC) model. We show that DNA damage leads to direct interaction of LCK with the HR repair proteins RAD51 and BRCA1 in a kinase dependent manner RAD51 and BRCA1 stabilization. LCK expression is induced and activated in the nucleus in response to DNA damage insult. Disruption of LCK expression attenuates RAD51, BRCA1, and BRCA2 protein expression by hampering there stability and results in inhibition of HR-mediated DNA repair including suppression of RAD51 foci formation, and augmentation of γH2AX foci formation. In contrast LCK overexpression leads to increased RAD51 and BRCA1 expression with a concomitant increase in HR DNA damage repair. Importantly, attenuation of LCK sensitizes HR-proficient eEOC cells to PARP inhibitor in cells and pre-clinical mouse studies. Collectively, our findings identify a novel therapeutic strategy to expand the utility of PARP targeted therapy in HR proficient ovarian cancer., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
12. CD55 in cancer: Complementing functions in a non-canonical manner.
- Author
-
Bharti R, Dey G, Lin F, Lathia J, and Reizes O
- Subjects
- Humans, Signal Transduction, Apoptosis, CD55 Antigens metabolism, Neoplasm Recurrence, Local
- Abstract
CD55, or decay accelerating factor, is a membrane lipid microdomain-associated, GPI-anchored protein implicated in the shielding of cells from complement-mediated attack via accelerating decay of C3 and C5. Loss of CD55 is associated with a number of pathologies due to hyperactivation of the complement system. CD55 is also implicated in cancer progression thought to be driven via its role in cell shielding mechanisms. We now appreciate that CD55 can signal intracellularly to promote malignant transformation, cancer progression, cell survival, angiogenesis, and inhibition of apoptosis. Outside-in signaling via CD55 is mediated by signaling pathways including JNK, JAK/STAT, MAPK/NF-κB, and LCK. Moreover, CD55 is enriched in the cancer stem cell (CSC) niche of multiple tumors including breast, ovarian, cervical, and can be induced by chemotherapeutics and hypoxic environments. CSCs are implicated in tumor recurrence and chemoresistance. Here, we review the unexpected roles of CD55 in cancer including the roles of canonical and noncanonical pathways that CD55 orchestrates. We will highlight opportunities for therapeutic targeting CD55 and gaps in the field that require more in-depth mechanistic insights., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2022 Elsevier B.V. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
13. Plasma IL13Rα2 as a novel liquid biopsy biomarker for glioblastoma.
- Author
-
Khristov V, Nesterova D, Trifoi M, Clegg T, Daya A, Barrett T, Tufano E, Shenoy G, Pandya B, Beselia G, Smith N, Mrowczynski O, Zacharia B, Waite K, Lathia J, Barnholtz-Sloan J, and Connor J
- Subjects
- Humans, Temozolomide therapeutic use, Liquid Biopsy, Biomarkers, Disease Progression, Glioblastoma drug therapy, Interleukin-13 Receptor alpha2 Subunit metabolism, Brain Neoplasms metabolism
- Abstract
Purpose: Glioblastoma (GBM) is the most common and deadliest brain tumor with unrelenting and rapid disease progression. The standard of care for GBM is surgical excision followed by radiation with concurrent and adjuvant temozolomide-centered chemotherapy (TMZ). Treatment failure and resistance is the rule and despite advances in imaging technology, early detection of treatment failure or impending resistance remains a challenge. There is a dire, unmet, need in clinical practice for minimally-invasive diagnostic tools to enable timely understanding of disease progression and treatment response. Here, we aim to address this clinical need by leveraging a unique characteristic of GBM: the overexpression of the α2 variant of the IL-13 receptor in over 75% of GBM tumors., Methods: In this study we examined patients with primary GBM from Penn State and Cleveland Clinic compared to healthy controls., Results: IL13Rα2 was detectable in plasma of GBM patients using ELISA but detection could be optimized by PEG precipitation to enrich for extracellular vesicles (EVs). Patients with GBM had elevated levels of plasma IL13Rα2, which correlated to levels of this receptor in the tumor tissue. Elevated plasma levels of IL13Rα2 predicted longer overall survival (OS) (19.8 vs. 13.2 months). Similarly, detection of IL13Rα2 + cells in tumor tissue also predicted longer OS (22.1 vs. 12.2 months)., Conclusion: These findings strongly suggest that expression of the IL13Rα2 receptor confer survival advantage in GBM patients, which can be determined through a minimally-invasive liquid biopsy. Detection of plasma IL13Rα2 can also be used to select GBM patients for targeted tumor therapy., (© 2022. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2022
- Full Text
- View/download PDF
14. Multimodal single-cell/nucleus RNA sequencing data analysis uncovers molecular networks between disease-associated microglia and astrocytes with implications for drug repurposing in Alzheimer's disease.
- Author
-
Xu J, Zhang P, Huang Y, Zhou Y, Hou Y, Bekris LM, Lathia J, Chiang CW, Li L, Pieper AA, Leverenz JB, Cummings J, and Cheng F
- Subjects
- Animals, Astrocytes metabolism, Data Analysis, Drug Repositioning, Humans, Mice, Microglia metabolism, Retrospective Studies, Sequence Analysis, RNA, Alzheimer Disease drug therapy, Alzheimer Disease genetics
- Abstract
Because disease-associated microglia (DAM) and disease-associated astrocytes (DAA) are involved in the pathophysiology of Alzheimer's disease (AD), we systematically identified molecular networks between DAM and DAA to uncover novel therapeutic targets for AD. Specifically, we develop a network-based methodology that leverages single-cell/nucleus RNA sequencing data from both transgenic mouse models and AD patient brains, as well as drug-target network, metabolite-enzyme associations, the human protein-protein interactome, and large-scale longitudinal patient data. Through this approach, we find both common and unique gene network regulators between DAM (i.e., PAK1 , MAPK14 , and CSF1R ) and DAA (i.e., NFKB1 , FOS , and JUN ) that are significantly enriched by neuro-inflammatory pathways and well-known genetic variants (i.e., BIN1 ). We identify shared immune pathways between DAM and DAA, including Th17 cell differentiation and chemokine signaling. Last, integrative metabolite-enzyme network analyses suggest that fatty acids and amino acids may trigger molecular alterations in DAM and DAA. Combining network-based prediction and retrospective case-control observations with 7.2 million individuals, we identify that usage of fluticasone (an approved glucocorticoid receptor agonist) is significantly associated with a reduced incidence of AD (hazard ratio [HR] = 0.86, 95% confidence interval [CI] 0.83-0.89, P < 1.0 × 10
-8 ). Propensity score-stratified cohort studies reveal that usage of mometasone (a stronger glucocorticoid receptor agonist) is significantly associated with a decreased risk of AD (HR = 0.74, 95% CI 0.68-0.81, P < 1.0 × 10-8 ) compared to fluticasone after adjusting age, gender, and disease comorbidities. In summary, we present a network-based, multimodal methodology for single-cell/nucleus genomics-informed drug discovery and have identified fluticasone and mometasone as potential treatments in AD., (© 2021 Xu et al.; Published by Cold Spring Harbor Laboratory Press.)- Published
- 2021
- Full Text
- View/download PDF
15. Development of an arteriolar niche and self-renewal of breast cancer stem cells by lysophosphatidic acid/protein kinase D signaling.
- Author
-
Jiang Y, Guo Y, Hao J, Guenter R, Lathia J, Beck AW, Hattaway R, Hurst D, Wang QJ, Liu Y, Cao Q, Krontiras H, Chen H, Silverstein R, and Ren B
- Subjects
- CD36 Antigens analysis, Cell Communication, Cell Differentiation, Endothelial Cells cytology, Female, Humans, Protein Kinase C analysis, Signal Transduction physiology, Tumor Microenvironment, Breast Neoplasms pathology, Lysophospholipids physiology, Neoplastic Stem Cells physiology, Protein Kinase C physiology, Stem Cell Niche physiology
- Abstract
Breast cancer stem cells (BCSCs) are essential for cancer growth, metastasis and recurrence. The regulatory mechanisms of BCSC interactions with the vascular niche within the tumor microenvironment (TME) and their self-renewal are currently under extensive investigation. We have demonstrated the existence of an arteriolar niche in the TME of human BC tissues. Intriguingly, BCSCs tend to be enriched within the arteriolar niche in human estrogen receptor positive (ER
+ ) BC and bi-directionally interact with arteriolar endothelial cells (ECs). Mechanistically, this interaction is driven by the lysophosphatidic acid (LPA)/protein kinase D (PKD-1) signaling pathway, which promotes both arteriolar differentiation of ECs and self-renewal of CSCs likely via differential regulation of CD36 transcription. This study indicates that CSCs may enjoy blood perfusion to maintain their stemness features. Targeting the LPA/PKD-1 -CD36 signaling pathway may have therapeutic potential to curb tumor progression by disrupting the arteriolar niche and effectively eliminating CSCs.- Published
- 2021
- Full Text
- View/download PDF
16. Pretreatment with LCK inhibitors chemosensitizes cisplatin-resistant endometrioid ovarian tumors.
- Author
-
Crean-Tate KK, Braley C, Dey G, Esakov E, Saygin C, Trestan A, Silver DJ, Turaga SM, Connor EV, DeBernardo R, Michener CM, Rose PG, Lathia J, and Reizes O
- Subjects
- Animals, Carcinoma, Endometrioid mortality, Cisplatin pharmacology, Female, Humans, Mice, Ovarian Neoplasms mortality, Survival Analysis, Carcinoma, Endometrioid drug therapy, Cisplatin therapeutic use, Lymphocyte Specific Protein Tyrosine Kinase p56(lck) antagonists & inhibitors, Ovarian Neoplasms drug therapy
- Abstract
Background: Ovarian cancer is the most fatal gynecologic malignancy in the United States. While chemotherapy is effective in the vast majority of ovarian cancer patients, recurrence and resistance to standard systemic therapy is nearly inevitable. We discovered that activation of the non-receptor tyrosine kinase Lymphocyte Cell-Specific Protein-Tyrosine Kinase (LCK) promoted cisplatin resistance. Here, we hypothesized that treating high grade, platinum resistant endometrioid cancer cells with an LCK inhibitor (LCKi) followed by co-treatment with cisplatin would lead to increased cisplatin efficacy. Our objective was to assess clinical outcomes associated with increased LCK expression, test our hypothesis of utilizing LCKi as pre-treatment followed by co-treatment with cisplatin in platinum resistant ovarian cancer in vitro, and evaluate our findings in vivo to assess LCKi applicability as a therapeutic agent., Results: Kaplan-Meier (KM) plotter data indicated LCK expression is associated with significantly worse median progression-free survival (HR 3.19, p = 0.02), and a trend toward decreased overall survival in endometrioid ovarian tumors with elevated LCK expression (HR 2.45, p = 0.41). In vitro, cisplatin resistant ovarian endometrioid cells treated first with LCKi followed by combination LCKi-cisplatin treatment showed decreased cell viability and increased apoptosis. Immunoblot studies revealed LCKi led to increased expression of phosphorylated H2A histone family X ([Formula: see text]-H2AX), a marker for DNA damage. In vivo results demonstrate treatment with LCKi followed by LCKi-cisplatin led to significantly slowed tumor growth., Conclusions: We identified a strategy to therapeutically target cisplatin resistant endometrioid ovarian cancer leading to chemosensitization to platinum chemotherapy via treatment with LCKi followed by co-treatment with LCKi-cisplatin.
- Published
- 2021
- Full Text
- View/download PDF
17. The Clinical Impact of Cancer Stem Cells.
- Author
-
Lathia J, Liu H, and Matei D
- Subjects
- Cell Differentiation, Humans, Neoplastic Stem Cells, Neoplasms, Oncologists
- Abstract
Patients with cancer can go though many stages in their disease, including diagnosis, recurrence, metastasis, and treatment failure. Cancer stem cells (CSCs) are a subgroup of cells within tumors that may explain the mechanism by which tumors recur and progress. CSCs can both self-renew and produce progenitor cells of more differentiated cancer cells as well as heterogeneously demonstrate resistance and the abilities to migrate and metastasize. These "stemness" characteristics are often the result of dysregulation of one or more pathways, which can be detected by various biomarkers. Although there has been considerable laboratory research conducted on CSCs, its relevance to the practicing oncologist may seem questionable. We sought to determine the clinical impact of CSCs on patients. A systematic literature search was conducted to identify analyses containing survival information based on the expression of known CSC biomarkers in any cancer. Overall, 234 survival analyses were identified, of which 82% reported that high expression of CSC biomarker(s) resulted in poor overall survival and/or disease-free survival compared with low or no expression of the biomarker. Elevated stemness biomarker levels were also associated with decreased tumor differentiation, altered TNM stage, and increased metastasis. This analysis would suggest that CSCs have a clinical impact on patients and that practicing oncologists need to start considering incorporating CSC-targeting therapies into their patients' treatment regimens. IMPLICATIONS FOR PRACTICE: Cancer stem cells (CSCs) may occur at any stage of cancer and are implicated in the occurrence of resistance, recurrence, and metastasis. A systematic literature analysis has shown that the presence of CSCs, identified via the upregulation of stemness pathway biomarkers, results in reduced survival across all cancers studied. Several CSC-targeting agents are currently approved, and several others are in clinical trials. Future treatment regimens will likely include CSC-targeting agents to enable the elimination of these holdouts to current therapies., (© AlphaMed Press 2019.)
- Published
- 2020
- Full Text
- View/download PDF
18. Gliomas display distinct sex-based differential methylation patterns based on molecular subtype.
- Author
-
Johansen ML, Stetson LC, Vadmal V, Waite K, Berens ME, Connor JR, Lathia J, Rubin JB, and Barnholtz-Sloan JS
- Abstract
Background: Gliomas are the most common type of primary brain tumor and one of many cancers where males are diagnosed with greater frequency than females. However, little is known about the sex-based molecular differences in glioblastomas (GBMs) or lower grade glioma (non-GBM) subtypes. DNA methylation is an epigenetic mechanism involved in regulating gene transcription. In glioma and other cancers, hypermethylation of specific gene promoters downregulates transcription and may have a profound effect on patient outcome. The purpose of this study was to determine if sex-based methylation differences exist in different glioma subtypes., Methods: Molecular and clinical data from glioma patients were obtained from The Cancer Genome Atlas and grouped according to tumor grade and molecular subtype ( IDH1/2 mutation and 1p/19q chromosomal deletion). Sex-specific differentially methylated probes (DMPs) were identified in each subtype and further analyzed to determine if they were part of differentially methylated regions (DMRs) or associated with differentially methylated DNA transcription regulatory binding motifs., Results: Analysis of methylation data in 4 glioma subtypes revealed unique sets of both sex-specific DMPs and DMRs in each subtype. Motif analysis based on DMP position also identified distinct sex-based sets of DNA-binding motifs that varied according to glioma subtype. Downstream targets of 2 of the GBM-specific transcription binding sites, NFAT5 and KLF6 , showed differential gene expression consistent with increased methylation mediating downregulation., Conclusion: DNA methylation differences between males and females in 4 glioma molecular subtypes suggest an important, sex-specific role for DNA methylation in epigenetic regulation of gliomagenesis., (© The Author(s) 2020. Published by Oxford University Press, the Society for Neuro-Oncology and the European Association of Neuro-Oncology.)
- Published
- 2020
- Full Text
- View/download PDF
19. Sex is an important prognostic factor for glioblastoma but not for nonglioblastoma.
- Author
-
Gittleman H, Ostrom QT, Stetson LC, Waite K, Hodges TR, Wright CH, Wright J, Rubin JB, Berens ME, Lathia J, Connor JR, Kruchko C, Sloan AE, and Barnholtz-Sloan JS
- Abstract
Background: Glioblastoma (GBM) is the most common and most malignant glioma. Nonglioblastoma (non-GBM) gliomas (WHO Grades II and III) are invasive and also often fatal. The goal of this study is to determine whether sex differences exist in glioma survival., Methods: Data were obtained from the National Cancer Database (NCDB) for years 2010 to 2014. GBM (WHO Grade IV; N = 2073) and non-GBM (WHO Grades II and III; N = 2963) were defined using the histology grouping of the Central Brain Tumor Registry of the United States. Non-GBM was divided into oligodendrogliomas/mixed gliomas and astrocytomas. Sex differences in survival were analyzed using Kaplan-Meier and multivariable Cox proportional hazards models adjusted for known prognostic variables., Results: There was a female survival advantage in patients with GBM both in the unadjusted ( P = .048) and adjusted ( P = .003) models. Unadjusted, median survival was 20.1 months (95% CI: 18.7-21.3 months) for women and 17.8 months (95% CI: 16.9-18.7 months) for men. Adjusted, median survival was 20.4 months (95% CI: 18.9-21.6 months) for women and 17.5 months (95% CI: 16.7-18.3 months) for men. When stratifying by age group (18-55 vs 56+ years at diagnosis), this female survival advantage appeared only in the older group, adjusting for covariates ( P = .017). Women (44.1%) had a higher proportion of methylated MGMT (O
6 -methylguanine-DNA methyltransferase) than men (38.4%). No sex differences were found for non-GBM., Conclusions: Using the NCDB data, there was a statistically significant female survival advantage in GBM, but not in non-GBM., (© The Author(s) 2019. Published by Oxford University Press on behalf of the Society for Neuro-Oncology and the European Association of Neuro-Oncology. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)- Published
- 2019
- Full Text
- View/download PDF
20. A Systems Pharmacology Approach Uncovers Wogonoside as an Angiogenesis Inhibitor of Triple-Negative Breast Cancer by Targeting Hedgehog Signaling.
- Author
-
Huang Y, Fang J, Lu W, Wang Z, Wang Q, Hou Y, Jiang X, Reizes O, Lathia J, Nussinov R, Eng C, and Cheng F
- Subjects
- Angiogenesis Inhibitors chemistry, Angiogenesis Inhibitors isolation & purification, Animals, Antineoplastic Agents, Phytogenic chemistry, Antineoplastic Agents, Phytogenic isolation & purification, Biological Products chemistry, Biological Products isolation & purification, Cell Line, Tumor, Cell Proliferation drug effects, Drug Screening Assays, Antitumor, Female, Flavanones chemistry, Flavanones isolation & purification, Glucosides chemistry, Glucosides isolation & purification, Hedgehog Proteins metabolism, Humans, Mammary Neoplasms, Experimental drug therapy, Mammary Neoplasms, Experimental metabolism, Mammary Neoplasms, Experimental pathology, Mice, Mice, Inbred BALB C, Mice, Nude, Neovascularization, Pathologic metabolism, Neovascularization, Pathologic pathology, Scutellaria baicalensis chemistry, Signal Transduction drug effects, Triple Negative Breast Neoplasms metabolism, Triple Negative Breast Neoplasms pathology, Angiogenesis Inhibitors pharmacology, Antineoplastic Agents, Phytogenic pharmacology, Biological Products pharmacology, Flavanones pharmacology, Glucosides pharmacology, Hedgehog Proteins antagonists & inhibitors, Neovascularization, Pathologic drug therapy, Triple Negative Breast Neoplasms drug therapy
- Abstract
Triple-negative breast cancer (TNBC) is an aggressive and heterogeneous disease that lacks clinically actionable genetic alterations that limit targeted therapies. Here we explore a systems pharmacology approach that integrates drug-target networks and large-scale genomic profiles of TNBC and identify wogonoside, one of the major active flavonoids, as a potent angiogenesis inhibitor. We validate that wogonoside attenuates cell migration, tube formation, and rat aorta microvessel outgrowth, and reduces formation of blood vessels in chicken chorioallantoic membrane and TNBC cell-induced Matrigel plugs. In addition, wogonoside inhibits growth and angiogenesis in TNBC cell xenograft models. This network-based approach predicts, and we empirically validate, wogonoside's antiangiogenic effects resulting from vascular endothelial growth factor secretion. Mechanistically, wogonoside inhibits Gli1 nuclear translocation and transcriptional activities associated with Hedgehog signaling, by promoting Smoothened degradation in a proteasome-dependent mechanism. This study offers a powerful, integrated, systems pharmacology-based strategy for oncological drug discovery and identifies wogonoside as a potential TNBC angiogenesis inhibitor., (Copyright © 2019 Elsevier Ltd. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
21. Therapeutic strategies to induce ERα in luminal breast cancer to enhance tamoxifen efficacy.
- Author
-
Esakov EL, Hale J, Richards EG, Torre-Healy L, Gullapalli K, Trivedi D, Chumakova A, Wessely O, Jensen J, Lathia J, and Reizes O
- Subjects
- Antineoplastic Agents, Hormonal pharmacology, Breast Neoplasms pathology, Cell Line, Tumor, Cell Proliferation drug effects, Drug Resistance, Neoplasm, Estradiol pharmacology, Estrogen Receptor alpha metabolism, Everolimus administration & dosage, Female, Humans, Hydroxamic Acids administration & dosage, Indazoles administration & dosage, MCF-7 Cells, Paclitaxel administration & dosage, Sulfonamides administration & dosage, Tamoxifen analogs & derivatives, Tumor Cells, Cultured, Antineoplastic Combined Chemotherapy Protocols pharmacology, Breast Neoplasms drug therapy, Breast Neoplasms metabolism, Estrogen Receptor alpha biosynthesis, Tamoxifen pharmacology
- Abstract
Breast cancer is the most prevalent malignancy and second leading cause of death in women worldwide, with hormone receptor-positive luminal breast cancers being the most widespread subtype. While these tumors are generally amenable to endocrine therapy, cellular heterogeneity and acquired ability of tumor cells to undergo cell state switching makes these populations difficult to be fully targeted and eradicated through conventional methods. We have leveraged a quality-by-design (QbD) approach that integrates biological responses with predictive mathematical modeling to identify key combinations of commercially available drugs to induce estrogen receptor expression for therapeutic targeting. This technology utilizes a high level of automation through a custom-built platform to reduce bias as well as design-of-experiments methodology to minimize the experimental iterations required. Utilizing this approach, we identified a combination of clinical compounds, each at concentrations well below their efficacious dose, able to induce the expression of estrogen receptor alpha (ESR1) in hormone-positive breast cancer cells. Induction of ESR1 in luminal cells leads to chemosensitization. These findings provide proof of concept for the utility of the QbD strategy and identify a unique drug cocktail able to sensitize breast cancer cells to tamoxifen.
- Published
- 2019
- Full Text
- View/download PDF
22. Poly(ADP-Ribose) Polymerase Inhibition Sensitizes Colorectal Cancer-Initiating Cells to Chemotherapy.
- Author
-
Jarrar A, Lotti F, DeVecchio J, Ferrandon S, Gantt G, Mace A, Karagkounis G, Orloff M, Venere M, Hitomi M, Lathia J, Rich JN, and Kalady MF
- Subjects
- Animals, Antineoplastic Agents pharmacology, Cell Line, Tumor, Colorectal Neoplasms genetics, Colorectal Neoplasms pathology, Humans, Mice, Poly(ADP-ribose) Polymerase Inhibitors pharmacology, Antineoplastic Agents therapeutic use, Colorectal Neoplasms drug therapy, DNA Repair genetics, Poly(ADP-ribose) Polymerase Inhibitors therapeutic use, Tumor Microenvironment genetics
- Abstract
Colorectal cancer (CRC) remains a leading killer in the U.S. with resistance to treatment as the largest hurdle to cure. Colorectal cancer-initiating cells (CICs) are a self-renewing tumor population that contribute to tumor relapse. Here, we report that patient-derived CICs display relative chemoresistance compared with differentiated progeny. In contrast, conventional cell lines failed model therapeutic resistance. CICs preferentially repaired chemotherapy-induced DNA breaks, prompting us to interrogate DNA damage pathways against which pharmacologic inhibitors have been developed. We found that CICs critically depended on the key single-strand break repair mediator, poly(ADP-ribose) polymerase (PARP), to survive treatment with standard-of-care chemotherapy. Small molecule PARP inhibitors (PARPi) sensitized CICs to chemotherapy and reduced chemotherapy-treated CIC viability, self-renewal, and DNA damage repair. Although PARPi monotherapy failed to kill CICs, combined PARPi therapy with chemotherapy attenuated tumor growth in vivo. Clinical significance of PARPi for CRC patients was supported by elevated PARP levels in colorectal tumors compared with normal colon, with further increases in metastases. Collectively, our results suggest that PARP inhibition serves as a point of fragility for CICs by augmenting therapeutic efficacy of chemotherapy. Stem Cells 2019;37:42-53., (© AlphaMed Press 2018.)
- Published
- 2019
- Full Text
- View/download PDF
23. Cancer Connectors: Connexins, Gap Junctions, and Communication.
- Author
-
Sinyuk M, Mulkearns-Hubert EE, Reizes O, and Lathia J
- Abstract
Despite concerted clinical and research efforts, cancer is a leading cause of death worldwide. Surgery, radiation, and chemotherapy have remained the most common standard-of-care strategies against cancer for decades. However, the side effects of these therapies demonstrate the need to investigate adjuvant novel treatment modalities that minimize the harm caused to healthy cells and tissues. Normal and cancerous cells require communication amongst themselves and with their surroundings to proliferate and drive tumor growth. It is vital to understand how intercellular and external communication impacts tumor cell malignancy. To survive and grow, tumor cells, and their normal counterparts utilize cell junction molecules including gap junctions (GJs), tight junctions, and adherens junctions to provide contact points between neighboring cells and the extracellular matrix. GJs are specialized structures composed of a family of connexin proteins that allow the free diffusion of small molecules and ions directly from the cytoplasm of adjacent cells, without encountering the extracellular milieu, which enables rapid, and coordinated cellular responses to internal and external stimuli. Importantly, connexins perform three main cellular functions. They enable direct gap junction intercellular communication (GJIC) between cells, form hemichannels to allow cell communication with the extracellular environment, and serve as a site for protein-protein interactions to regulate signaling pathways. Connexins themselves have been found to promote tumor cell growth and invasiveness, contributing to the overall tumorigenicity and have emerged as attractive anti-tumor targets due to their functional diversity. However, connexins can also serve as tumor suppressors, and therefore, a complete understanding of the roles of the connexins and GJs in physiological and pathophysiological conditions is needed before connexin targeting strategies are applied. Here, we discuss how the three aspects of connexin function, namely GJIC, hemichannel formation, and connexin-protein interactions, function in normal cells, and contribute to tumor cell growth, proliferation, and death. Finally, we discuss the current state of anti-connexin therapies and speculate which role may be most amenable for the development of targeting strategies.
- Published
- 2018
- Full Text
- View/download PDF
24. The p38 signaling pathway mediates quiescence of glioma stem cells by regulating epidermal growth factor receptor trafficking.
- Author
-
Soeda A, Lathia J, Williams BJ, Wu Q, Gallagher J, Androutsellis-Theotokis A, Giles AJ, Yang C, Zhuang Z, Gilbert MR, Rich JN, and Park DM
- Subjects
- Apoptosis, Cell Differentiation, Cell Line, Tumor, Cell Proliferation, Cell Self Renewal, ErbB Receptors genetics, Gene Expression, Glioma genetics, Glioma pathology, Humans, Ligands, Phosphorylation, Protein Binding, Protein Transport, ErbB Receptors metabolism, Glioma metabolism, Neoplastic Stem Cells metabolism, Resting Phase, Cell Cycle, Signal Transduction, p38 Mitogen-Activated Protein Kinases metabolism
- Abstract
EGFR pathway is upregulated in malignant gliomas, and its downstream signaling is important for self-renewal of glioma cancer stem-like cells (GSC). p38 mitogen-activated protein kinase (MAPK) signaling, a stress-activated signaling cascade with suppressive and permissive effects on tumorigenesis, can promote internalization and ubiquitin ligase mediated degradation of EGFR. In this study, we investigated the role of p38 MAPK signaling on the self-renewal of GSCs with the hypothesis that inhibition may lead to enhanced self-renewal capacity by retention of EGFR. Inhibition of p38 MAPK pathway led to increase in EGFR expression but surprisingly, reduced proliferation. Additional functional evaluation revealed that p38 inhibition was associated with decrease in cell death and maintenance of undifferentiated state. Further probing the effect of p38 inhibition demonstrated attenuation of EGFR downstream signaling activity in spite of prolonged surface expression of the receptor. In vitro observations were confirmed in xenograft in vivo experiments. These data suggest that p38 MAPK control of EGFR signaling activity may alter GSC cell cycle state by regulating quiescence and passage into transit amplifying state.
- Published
- 2017
- Full Text
- View/download PDF
25. Induction of HEXIM1 activities by HMBA derivative 4a1: Functional consequences and mechanism.
- Author
-
Ketchart W, Yeh IJ, Zhou H, Thiagarajan PS, Lathia J, Reizes O, Exner A, Su B, and Montano MM
- Subjects
- Acetamides chemistry, Animals, Antigens, Polyomavirus Transforming genetics, Antineoplastic Agents chemistry, Benzeneacetamides chemistry, Breast Neoplasms genetics, Breast Neoplasms metabolism, Breast Neoplasms pathology, Cell Differentiation drug effects, Cell Movement drug effects, Cyclin-Dependent Kinase 9 metabolism, Dose-Response Relationship, Drug, Drug Resistance, Neoplasm drug effects, Female, Gene Expression Regulation, Neoplastic, Humans, MCF-7 Cells, Mammary Neoplasms, Experimental genetics, Mammary Neoplasms, Experimental metabolism, Mammary Neoplasms, Experimental pathology, Mice, Transgenic, Molecular Structure, Neoplasm Metastasis, Neoplastic Stem Cells drug effects, Neoplastic Stem Cells metabolism, Neoplastic Stem Cells pathology, Phenotype, RNA Interference, RNA-Binding Proteins genetics, Signal Transduction drug effects, Structure-Activity Relationship, Tamoxifen pharmacology, Time Factors, Transcription Factors, Transfection, Up-Regulation, Acetamides pharmacology, Antineoplastic Agents pharmacology, Benzeneacetamides pharmacology, Breast Neoplasms drug therapy, Mammary Neoplasms, Experimental drug therapy, RNA-Binding Proteins biosynthesis
- Abstract
We have been studying the role of Hexamethylene bisacetamide (HMBA) Induced Protein 1 (HEXIM1) as a tumor suppressor whose expression is decreased in tamoxifen resistant and metastatic breast cancer. HMBA was considered the most potent and specific inducer for HMBA inducible protein 1 (HEXIM1) prior to our studies. Moreover, the ability of HMBA to induce differentiation is advantageous for its therapeutic use when compared to cytotoxic agents. However, HMBA induced HEXIM1 expression required at mM concentrations and induced dose limiting toxicity, thrombocytopenia. Thus we structurally optimized HMBA and identified a more potent inducer of HEXIM1 expression, 4a1. The studies reported herein tested the ability of 4a1 to induce HEXIM1 activities using a combination of biochemical, cell phenotypic, and in vivo assays. 4a1 induced breast cell differentiation, including the stem cell fraction in triple negative breast cancer cells. Clinically relevant HEXIM1 activities that are also induced by 4a1 include enhancement of the inhibitory effects of tamoxifen and inhibition of breast tumor metastasis. We also provide mechanistic basis for the phenotypic effects of 4a1. Our results support the potential of an unsymmetrical HMBA derivative, such as 4a1, as lead compound for further drug development., (Copyright © 2016 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
26. Increased cancer stem cell invasion is mediated by myosin IIB and nuclear translocation.
- Author
-
Thomas D, Thiagarajan PS, Rai V, Reizes O, Lathia J, and Egelhoff T
- Subjects
- Active Transport, Cell Nucleus, Cell Line, Tumor, Cell Movement, Humans, Neoplasm Invasiveness, Cell Nucleus metabolism, Neoplastic Stem Cells pathology, Nonmuscle Myosin Type IIB physiology
- Abstract
Despite many advances in the treatment of breast cancer, it remains one of the leading causes of death among women. One hurdle for effective therapy is the treatment of the highly invasive and tumorigenic subpopulation of tumors called cancer stem cells (CSCs). CSCs, when stimulated with EGF, migrate through a physiological 3D collagen matrix at a higher velocity than non-stem cancer cells (non-SCCs). This increased invasion is due, in part, by an enhanced nuclear translocation ability of CSCs. We observed no difference between CSC and non-SCC in cellular migration rates on a 2D surface. Furthermore, during transwell migration using large diameter transwell pores, both CSC and non-SCC populations migrated with similar efficiency. However, when challenged with more restrictive transwells, CSCs were dramatically more capable of transwell migration. These results implicate nuclear translocation as a major rate limiting factor for CSC dissemination. We further show that non-muscle myosin IIB is critical for this enhanced nuclear translocation and the ability for cancer stem cells to efficiently migrate through restrictive 3D environments. These studies suggest that cytoskeletal elements upregulated in CSCs, such as myosin IIB, may be valuable targets for intervention in cancer stem cell dispersal from tumors., Competing Interests: The authors declare no conflicts of interest with this.
- Published
- 2016
- Full Text
- View/download PDF
27. A Tumor Suppressor Function for Notch Signaling in Forebrain Tumor Subtypes.
- Author
-
Giachino C, Boulay JL, Ivanek R, Alvarado A, Tostado C, Lugert S, Tchorz J, Coban M, Mariani L, Bettler B, Lathia J, Frank S, Pfister S, Kool M, and Taylor V
- Subjects
- Animals, Basic Helix-Loop-Helix Transcription Factors genetics, Basic Helix-Loop-Helix Transcription Factors metabolism, Brain Neoplasms genetics, Brain Neoplasms mortality, Brain Neoplasms pathology, Cell Proliferation, Databases, Genetic, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Gene Transfer Techniques, Glioma genetics, Glioma mortality, Glioma pathology, Humans, Immunoglobulin J Recombination Signal Sequence-Binding Protein genetics, Immunoglobulin J Recombination Signal Sequence-Binding Protein metabolism, Infusions, Intraventricular, Kaplan-Meier Estimate, Mice, Knockout, Neoplasm Grading, Neoplastic Stem Cells pathology, Neural Stem Cells pathology, Phenotype, Platelet-Derived Growth Factor administration & dosage, Prosencephalon pathology, Proto-Oncogene Proteins c-sis genetics, Proto-Oncogene Proteins c-sis metabolism, Receptor, Notch1 genetics, Receptor, Notch1 metabolism, Receptor, Notch2 genetics, Receptor, Notch2 metabolism, Receptors, Notch genetics, Recombinant Proteins administration & dosage, Repressor Proteins genetics, Repressor Proteins metabolism, Time Factors, Tumor Burden, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism, Tumor Suppressor Proteins genetics, Brain Neoplasms metabolism, Glioma metabolism, Neoplastic Stem Cells metabolism, Neural Stem Cells metabolism, Prosencephalon metabolism, Receptors, Notch metabolism, Signal Transduction, Tumor Suppressor Proteins metabolism
- Abstract
In the brain, Notch signaling maintains normal neural stem cells, but also brain cancer stem cells, indicating an oncogenic role. Here, we identify an unexpected tumor suppressor function for Notch in forebrain tumor subtypes. Genetic inactivation of RBP-Jκ, a key Notch mediator, or Notch1 and Notch2 receptors accelerates PDGF-driven glioma growth in mice. Conversely, genetic activation of the Notch pathway reduces glioma growth and increases survival. In humans, high Notch activity strongly correlates with distinct glioma subtypes, increased patient survival, and lower tumor grade. Additionally, simultaneous inactivation of RBP-Jκ and p53 induces primitive neuroectodermal-like tumors in mice. Hence, Notch signaling cooperates with p53 to restrict cell proliferation and tumor growth in mouse models of human brain tumors., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
28. Cancer stem cells: targeting the roots of cancer, seeds of metastasis, and sources of therapy resistance.
- Author
-
Adorno-Cruz V, Kibria G, Liu X, Doherty M, Junk DJ, Guan D, Hubert C, Venere M, Mulkearns-Hubert E, Sinyuk M, Alvarado A, Caplan AI, Rich J, Gerson SL, Lathia J, and Liu H
- Subjects
- Drug Resistance, Neoplasm, Epigenesis, Genetic, Humans, Neoplasm Metastasis, Neoplastic Stem Cells drug effects, Tumor Microenvironment, Neoplastic Stem Cells physiology
- Abstract
With the goal to remove the roots of cancer, eliminate metastatic seeds, and overcome therapy resistance, the 2014 inaugural International Cancer Stem Cell (CSC) Conference at Cleveland, OH, convened together over 320 investigators, including 55 invited world-class speakers, 25 short oral presenters, and 100 poster presenters, to gain an in-depth understanding of CSCs and explore therapeutic opportunities targeting CSCs. The meeting enabled intriguing discussions on several topics including: genetics and epigenetics; cancer origin and evolution; microenvironment and exosomes; metabolism and inflammation; metastasis and therapy resistance; single cell and heterogeneity; plasticity and reprogramming; as well as other new concepts. Reports of clinical trials targeting CSCs emphasized the urgent need for strategically designing combinational CSC-targeting therapies against cancer., (©2015 American Association for Cancer Research.)
- Published
- 2015
- Full Text
- View/download PDF
29. Chemotherapy activates cancer-associated fibroblasts to maintain colorectal cancer-initiating cells by IL-17A.
- Author
-
Lotti F, Jarrar AM, Pai RK, Hitomi M, Lathia J, Mace A, Gantt GA Jr, Sukhdeo K, DeVecchio J, Vasanji A, Leahy P, Hjelmeland AB, Kalady MF, and Rich JN
- Subjects
- Animals, Cell Line, Tumor, Cell Separation, Chemokines metabolism, Coculture Techniques, Cytokines metabolism, Fibroblasts metabolism, Flow Cytometry, Humans, Hyaluronan Receptors metabolism, Immunohistochemistry, Mice, Neoplasm Transplantation, Signal Transduction, Time Factors, Antineoplastic Agents chemistry, Colorectal Neoplasms drug therapy, Fibroblasts cytology, Interleukin-17 metabolism
- Abstract
Many solid cancers display cellular hierarchies with self-renewing, tumorigenic stemlike cells, or cancer-initiating cells (CICs) at the apex. Whereas CICs often exhibit relative resistance to conventional cancer therapies, they also receive critical maintenance cues from supportive stromal elements that also respond to cytotoxic therapies. To interrogate the interplay between chemotherapy and CICs, we investigated cellular heterogeneity in human colorectal cancers. Colorectal CICs were resistant to conventional chemotherapy in cell-autonomous assays, but CIC chemoresistance was also increased by cancer-associated fibroblasts (CAFs). Comparative analysis of matched colorectal cancer specimens from patients before and after cytotoxic treatment revealed a significant increase in CAFs. Chemotherapy-treated human CAFs promoted CIC self-renewal and in vivo tumor growth associated with increased secretion of specific cytokines and chemokines, including interleukin-17A (IL-17A). Exogenous IL-17A increased CIC self-renewal and invasion, and targeting IL-17A signaling impaired CIC growth. Notably, IL-17A was overexpressed by colorectal CAFs in response to chemotherapy with expression validated directly in patient-derived specimens without culture. These data suggest that chemotherapy induces remodeling of the tumor microenvironment to support the tumor cellular hierarchy through secreted factors. Incorporating simultaneous disruption of CIC mechanisms and interplay with the tumor microenvironment could optimize therapeutic targeting of cancer.
- Published
- 2013
- Full Text
- View/download PDF
30. Hypoxia-inducible factors regulate tumorigenic capacity of glioma stem cells.
- Author
-
Li Z, Bao S, Wu Q, Wang H, Eyler C, Sathornsumetee S, Shi Q, Cao Y, Lathia J, McLendon RE, Hjelmeland AB, and Rich JN
- Subjects
- Animals, Biomarkers, Tumor metabolism, Brain Neoplasms blood supply, Brain Neoplasms pathology, Cell Hypoxia, Cell Proliferation, Cell Survival, Cells, Cultured, Glioblastoma blood supply, Glioblastoma pathology, Humans, Mice, Mice, Inbred BALB C, Mice, Nude, Neoplasm Transplantation, Neovascularization, Pathologic metabolism, Neovascularization, Pathologic pathology, Stem Cells physiology, Vascular Endothelial Growth Factor A metabolism, Basic Helix-Loop-Helix Transcription Factors physiology, Brain Neoplasms metabolism, Glioblastoma metabolism, Hypoxia-Inducible Factor 1, alpha Subunit physiology, Neoplastic Stem Cells physiology
- Abstract
Glioblastomas are lethal cancers characterized by florid angiogenesis promoted in part by glioma stem cells (GSCs). Because hypoxia regulates angiogenesis, we examined hypoxic responses in GSCs. We now demonstrate that hypoxia-inducible factor HIF2alpha and multiple HIF-regulated genes are preferentially expressed in GSCs in comparison to non-stem tumor cells and normal neural progenitors. In tumor specimens, HIF2alpha colocalizes with cancer stem cell markers. Targeting HIFs in GSCs inhibits self-renewal, proliferation, and survival in vitro, and attenuates tumor initiation potential of GSCs in vivo. Analysis of a molecular database reveals that HIF2A expression correlates with poor glioma patient survival. Our results demonstrate that GSCs differentially respond to hypoxia with distinct HIF induction patterns, and HIF2alpha might represent a promising target for antiglioblastoma therapies.
- Published
- 2009
- Full Text
- View/download PDF
31. SDF1alpha/CXCR4 signaling, via ERKs and the transcription factor Egr1, induces expression of a 67-kDa form of glutamic acid decarboxylase in embryonic hippocampal neurons.
- Author
-
Luo Y, Lathia J, Mughal M, and Mattson MP
- Subjects
- Animals, Chemokine CXCL12 pharmacology, Dose-Response Relationship, Drug, Embryo, Mammalian cytology, Female, Gene Expression Regulation, Developmental drug effects, Gene Expression Regulation, Enzymologic drug effects, Hippocampus embryology, MAP Kinase Signaling System drug effects, Mice, Pregnancy, RNA, Messenger biosynthesis, RNA, Small Interfering pharmacology, Rats, Rats, Sprague-Dawley, Receptors, CXCR4 agonists, Response Elements physiology, gamma-Aminobutyric Acid biosynthesis, Chemokine CXCL12 metabolism, Early Growth Response Protein 1 metabolism, Embryo, Mammalian enzymology, Extracellular Signal-Regulated MAP Kinases metabolism, Gene Expression Regulation, Developmental physiology, Gene Expression Regulation, Enzymologic physiology, Glutamate Decarboxylase biosynthesis, Hippocampus enzymology, MAP Kinase Signaling System physiology, Neurites enzymology, Receptors, CXCR4 metabolism
- Abstract
Stromal cell-derived factor alpha (SDF1alpha) and its cognate receptor CXCR4 play an important role in neuronal development in the hippocampus, but the genes directly regulated by SDF1alpha/CXCR4 signaling are unknown. To study the role of CXCR4 targeted genes in neuronal development, we used neuronal cultures established from embryonic day 18 rats. Hippocampal neurons express CXCR4 receptor proteins and are stimulated by SDF1alpha resulting in activation of extracellular signal-regulated kinase (ERK)1/2 and the transcription factor cAMP-response element-binding protein. SDF1alpha rapidly induces the expression of the early growth response gene Egr1, a transcription factor involved in activity-dependent neuronal responses, in a concentration-dependent manner. Gel-shift analysis showed that SDF1alpha enhances DNA binding activity to the Egr1-containing promoter for GAD67. Chromatin immunoprecipitation analysis using an Egr1 antibody indicated that SDF1alpha stimulation increases binding of Egr1 to a GAD67 promoter DNA sequence. SDF1alpha stimulation increases the expression of GAD67 at both the mRNA and protein levels, and increases the amount and neurite localization of gamma-aminobutyric acid (GABA) in neurons already expressing GABA. SDF1alpha-induced Egr1/GAD67 expression is mediated by the G protein-coupled CXCR4 receptor and activation of the ERK pathway. Reduction of Egr1 gene expression using small interfering RNA technology lowers the level of GAD67 transcripts and inhibits SDF1alpha-induced GABA production. Inhibition of CXCR4 activation in the developing mouse brain in utero greatly reduced Egr1 and GAD67 mRNA levels and GAD67 protein levels, suggesting a pivotal role for CXCR4 signaling in the development of GABAergic neurons in vivo. Our data suggest that SDF1alpha/CXCR4/G protein/ERK signaling induces the expression of the GAD67 system via Egr1 activation, a mechanism that may promote the maturation of GABAergic neurons during development.
- Published
- 2008
- Full Text
- View/download PDF
32. In situ monitoring of soil dissolution dynamics: a rapid and simple method for determining worst-case soils for cleaning validation.
- Author
-
Sharnez R, Lathia J, Kahlenberg D, Prabhu S, and Dekleva M
- Subjects
- Drug Contamination prevention & control, Drug Industry standards, Electric Conductivity, Injections, Models, Theoretical, Reproducibility of Results, Sensitivity and Specificity, Solutions chemistry, Technology, Pharmaceutical instrumentation, Technology, Pharmaceutical methods, Time Factors, Equipment Contamination prevention & control, Hygiene standards, Soil Pollutants analysis, Solubility
- Abstract
A common approach utilized in validating the cleaning of multi-product equipment is to challenge the cleaning cycle with the hardest-to-clean, or "worst-case", soil. The worst-case soil is often determined through bench-scale experiments. These experiments can be tricky and time-consuming, especially when the dissolution dynamics of the soils are characterized by crossovers beyond the limit of visual detection. This paper describes a simple in situ method for monitoring soil dissolution dynamics and rapidly determining worst-case soils for cleaning validation. The method utilizes the real-time response of electrical conductivity to identify crossovers and to determine the optimal time for endpoint analysis. The results address the sensitivity and reproducibility of the method and the identification of critical experimental parameters. The dissolution dynamics of several parenteral products are compared.
- Published
- 2004
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.