全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...

Current Trends in Targeted Therapies for Glioblastoma Multiforme

DOI: 10.1155/2012/878425

Full-Text   Cite this paper   Add to My Lib

Abstract:

Glioblastoma multiforme (GBM) is one of the most frequently occurring tumors in the central nervous system and the most malignant tumor among gliomas. Despite aggressive treatment including surgery, adjuvant TMZ-based chemotherapy, and radiotherapy, GBM still has a dismal prognosis: the median survival is 14.6 months from diagnosis. To date, many studies report several determinants of resistance to this aggressive therapy: (1) O6-methylguanine-DNA methyltransferase (MGMT), (2) the complexity of several altered signaling pathways in GBM, (3) the existence of glioma stem-like cells (GSCs), and (4) the blood-brain barrier. Many studies aim to overcome these determinants of resistance to conventional therapy by using various approaches to improve the dismal prognosis of GBM such as modifying TMZ administration and combining TMZ with other agents, developing novel molecular-targeting agents, and novel strategies targeting GSCs. In this paper, we review up-to-date clinical trials of GBM treatments in order to overcome these 4 hurdles and to aim at more therapeutical effect than conventional therapies that are ongoing or are about to launch in clinical settings and discuss future perspectives. 1. Introduction Glioblastoma multiforme (GBM) is one of the most frequently occurring tumors in the central nervous system and the most malignant tumor among gliomas. A subanalysis in an international randomized trial by the European Organization for Research and Treatment of Cancer/National Cancer Institute of Canada (EORTC/NCIC) compared the results of radiotherapy (RT) alone with those of concomitant RT and temozolomide (TMZ) and found that the addition of TMZ to radiotherapy for newly diagnosed GBM resulted significant survival benefit [1], additionally the subgroup analysis of the 5-year survival data of the EORTC/NCIC trial also revealed its benefit [2]. Since then, TMZ has been the current first-line chemotherapeutic agent for GBM. However, despite aggressive treatment including surgery, adjuvant TMZ-based chemotherapy, and RT, GBM still has a dismal prognosis: the median survival is 14.6 months from diagnosis. Many studies aim to overcome several determinants of resistance to conventional therapy by using various approaches to improve the dismal prognosis of GBM such as modifying TMZ administration and combining TMZ with other agents, developing novel molecular-targeting agents, and novel strategies targeting GSCs. In this paper, we review up-to-date clinical trials of GBM treatments in order to overcome determinants and to aim at more therapeutical effect than

References

[1]  R. Stupp, W. P. Mason, M. J. van den Bent et al., “Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma,” The New England Journal of Medicine, vol. 352, no. 10, pp. 987–996, 2005.
[2]  R. Stupp, M. E. Hegi, W. P. Mason et al., “Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase IIII study: 5-year analysis of the EORTC-NCIC trial,” The Lancet Oncology, vol. 10, no. 5, pp. 459–466, 2009.
[3]  J. P. Fruehauf, H. Brem, S. Brem et al., “In vitro drug response and molecular markers associated with drug resistance in malignant gliomas,” Clinical Cancer Research, vol. 12, no. 15, pp. 4523–4532, 2006.
[4]  J. Ma, M. Murphy, P. J. O'Dwyer, E. Berman, K. Reed, and J. M. Gallo, “Biochemical changes associated with a multidrug-resistant phenotype of a human glioma cell line with temozolomide-acquired resistance,” Biochemical Pharmacology, vol. 63, no. 7, pp. 1219–1228, 2002.
[5]  M. Hermisson, A. Klumpp, W. Wick et al., “O6-methylguanine DNA methyltransferase and p53 status predict temozolomide sensitivity in human malignant glioma cells,” Journal of Neurochemistry, vol. 96, no. 3, pp. 766–776, 2006.
[6]  D. M. Kokkinakis, D. B. Bocangel, S. C. Schold, R. C. Moschel, and A. E. Pegg, “Thresholds of O6-alkylguanine-DNA alkyltransferase which significant resistance of human glial tumor xenografts to treatment with 1,3-bis(2-chloroethyl)-1-nitrosourea or temozolomide,” Clinical Cancer Research, vol. 7, no. 2, pp. 421–428, 2001.
[7]  K. S. Srivenugopal, X. H. Yuan, H. S. Friedman, and F. Ali-Osman, “Ubiquitination-dependent proteolysis of O6-methylguanine-DNA methyltransferase in human and murine tumor cells following inactivation with O6-benzylguanine or 1,3-bis(2-chloroethyl)-1-nitrosourea,” Biochemistry, vol. 35, no. 4, pp. 1328–1334, 1996.
[8]  M. Gilbert, “RTOG 0525: a randomized phase IIII trial comparing standard adjuvant temozolomide (TMZ) with a dose-dense (dd) schedule in newly diagnosed glioblastoma (GBM),” Journal of Clinical Oncology, vol. 29, supplement, 2011, Abstract no. 2006.
[9]  R. S. Kerbel and B. A. Kamen, “The anti-angiogenic basis of metronomic chemotherapy,” Nature Reviews Cancer, vol. 4, no. 6, pp. 423–436, 2004.
[10]  F. Bertolini, S. Paul, P. Mancuso et al., “Maximum tolerable dose and low-dose metronomic chemotherapy have opposite effects on the mobilization and viability of circulating endothelial progenitor cells,” Cancer Research, vol. 63, no. 15, pp. 4342–4346, 2003.
[11]  U. Emmenegger, S. Man, Y. Shaked et al., “A comparative analysis of low-dose metronomic cyclophosphamide reveals absent or low-grade toxicity on tissues highly sensitive to the toxic effects of maximum tolerated dose regimens,” Cancer Research, vol. 64, no. 11, pp. 3994–4000, 2004.
[12]  G. Bocci, G. Francia, S. Man, J. Lawler, and R. S. Kerbel, “Thrombospondin 1, a mediator of the antiangiogenic effects of low-dose metronomic chemotherapy,” Proceedings of the National Academy of Sciences of the United States of America, vol. 100, no. 22, pp. 12917–12922, 2003.
[13]  J. R. Perry, K. Bélanger, W. P. Mason et al., “Phase II trial of continuous dose-intense temozolomide in recurrent malignant glioma: RESCUE study,” Journal of Clinical Oncology, vol. 28, no. 12, pp. 2051–2057, 2010.
[14]  W. Wick, “NOA-08 randomized phase IIII trial of 1-week-on/1-week-off temozolomide versus involved-field radiotherapy in elderly (older than age 65) patients with newly diagnosed anaplastic astrocytoma or glioblastoma (Methusalem),” Journal of Clinical Oncology, vol. 28, supplement, 2010, Abstract no. LBA2001.
[15]  O. Chinot, “TEMOBIC: a ANOCEF phase II study of BCNU and temozolomide (TMZ) combination prior to radiotherapy (RT) in anaplasic oligodendroglial gliomas (AOG),” Journal of Clinical Oncology, vol. 29, supplement, 2011, Abstract no. 2034.
[16]  L. Liu and S. L. Gerson, “Targeted modulation of MGMT: clinical implications,” Clinical Cancer Research, vol. 12, no. 2, pp. 328–331, 2006.
[17]  T. Kanzawa, J. Bedwell, Y. Kondo, S. Kondo, and I. M. Germano, “Inhibition of DNA repair for sensitizing resistant glioma cells to temozolomide,” Journal of Neurosurgery, vol. 99, no. 6, pp. 1047–1052, 2003.
[18]  J. A. Quinn, J. Pluda, M. E. Dolan et al., “Phase II trial of carmustine plus O6-benzylguanine for patients with nitrosourea-resistant recurrent or progressive malignant glioma,” Journal of Clinical Oncology, vol. 20, no. 9, pp. 2277–2283, 2002.
[19]  J. A. Quinn, A. Desjardins, J. Weingart et al., “Phase I trial of temozolomide plus O6-benzylguanine for patients with recurrent or progressive malignant glioma,” Journal of Clinical Oncology, vol. 23, no. 28, pp. 7178–7187, 2005.
[20]  D. A. Reardon, K. L. Fink, T. Mikkelsen et al., “Randomized phase II study of cilengitide, an integrin-targeting arginine-glycine-aspartic acid peptide, in recurrent glioblastoma multiforme,” Journal of Clinical Oncology, vol. 26, no. 34, pp. 5610–5617, 2008.
[21]  B. Nabors, “Cilengitide in patients with newly diagnosed patients with glioblastoma multiforme and unmethylated MGMG gene promoter: safety run-in results from a randomized controlled phase II study (CORE),” Journal of Neuro-Oncology, vol. 12, p. iv75, 2010, Abstract no. OT-26.
[22]  M. Chawla-Sarkar, D. J. Lindner, Y. F. Liu et al., “Apoptosis and interferons: role of interferon-stimulated genes as mediators of apoptosis,” Apoptosis, vol. 8, no. 3, pp. 237–249, 2003.
[23]  T. Wakabayashi, N. Hatano, Y. Kajita et al., “Initial and maintenance combination treatment with interferon-β, MCNU (Ranimustine), and radiotherapy for patients with previously untreated malignant glioma,” Journal of Neuro-Oncology, vol. 49, no. 1, pp. 57–62, 2000.
[24]  A. Natsume, D. IshII, T. Wakabayashi et al., “IFN-β down-regulates the expression of DNA repair gene MGMT and sensitizes resistant glioma cells to temozolomide,” Cancer Research, vol. 65, no. 17, pp. 7573–7579, 2005.
[25]  A. Natsume, T. Wakabayashi, D. IshII et al., “A combination of IFN-β and temozolomide in human glioma xenograft models: implication of p53-mediated MGMT downregulation,” Cancer Chemotherapy and Pharmacology, vol. 61, no. 4, pp. 653–659, 2008.
[26]  J. J. Vredenburgh, A. Desjardins, J. E. Herndon et al., “Bevacizumab plus irinotecan in recurrent glioblastoma multiforme,” Journal of Clinical Oncology, vol. 25, no. 30, pp. 4722–4729, 2007.
[27]  H. S. Friedman, M. D. Prados, P. Y. Wen et al., “Bevacizumab alone and in combination with irinotecan in recurrent glioblastoma,” Journal of Clinical Oncology, vol. 27, no. 28, pp. 4733–4740, 2009.
[28]  M. C. Chamberlain, “Emerging clinical principles on the use of bevacizumab for the treatment of malignant gliomas,” Cancer, vol. 116, no. 17, pp. 3988–3999, 2010.
[29]  A. D. Norden, J. Drappatz, and P. Y. Wen, “Novel anti-angiogenic therapies for malignant gliomas,” The Lancet Neurology, vol. 7, no. 12, pp. 1152–1160, 2008.
[30]  M. Gruber, “Bevacizumab in combination with radiotherapy plus concomitant and adjuvant temozolomide for newly diagnosed glioblastoma: update progression-free survival, overall survival, and toxicity,” Journal of Clinical Oncology, vol. 27, supplement, p. 15s, 2009, abstract no. 2017.
[31]  O. L. Chinot, T. de La Motte Rouge, N. Moore et al., “AVAglio: phase 3 trial of bevacizumab plus temozolomide and radiotherapy in newly diagnosed glioblastoma multiforme,” Advances in Therapy, vol. 28, no. 4, pp. 334–340, 2011.
[32]  O. Keunen, M. Johansson, A. Oudin et al., “Anti-VEGF treatment reduces blood supply and increases tumor cell invasion in glioblastoma,” Proceedings of the National Academy of Sciences of the United States of America, vol. 108, no. 9, pp. 3749–3754, 2011.
[33]  T. T. Batchelor, D. G. Duda, E. di Tomaso et al., “Phase II study of cediranib, an oral pan-vascular endothelial growth factor receptor tyrosine kinase inhibitor, in patients with recurrent glioblastoma,” Journal of Clinical Oncology, vol. 28, no. 17, pp. 2817–2823, 2010.
[34]  T. T. Batchelor, “The efficacy of cediranib as monotherapy and in combination with lomustine compared to lomustine alone in patients with recurrent glioblastoma: a phase IIII randomized study,” Neuro-Oncology, vol. 12, p. iv75, 2010, Abstract no. OT-25.
[35]  M. Katoh and M. Katoh, “WNT signaling pathway and stem cell signaling network,” Clinical Cancer Research, vol. 13, no. 14, pp. 4042–4045, 2007.
[36]  C. K. Cheng, Q. W. Fan, and W. A. Weiss, “PI3K signaling in glioma—animal models and therapeutic challenges,” Brain Pathology, vol. 19, no. 1, pp. 112–120, 2009.
[37]  H. Ohgaki and P. Kleihues, “Genetic alterations and signaling pathways in the evolution of gliomas,” Cancer Science, vol. 100, no. 12, pp. 2235–2241, 2009.
[38]  Q. W. Fan and W. A. Weiss, “Targeting the RTK-PI3K-mTOR axis in malignant glioma: overcoming resistance,” Current Topics in Microbiology and Immunology, vol. 347, pp. 279–296, 2010.
[39]  H. Ohgaki, P. Dessen, B. Jourde et al., “Genetic pathways to glioblastoma: a population-based study,” Cancer Research, vol. 64, no. 19, pp. 6892–6899, 2004.
[40]  L. Frederick, X. Y. Wang, G. Eley, and C. D. James, “Diversity and frequency of epidermal growth factor receptor mutations in human glioblastomas,” Cancer Research, vol. 60, no. 5, pp. 1383–1387, 2000.
[41]  A. M. Martelli, C. Evangelisti, F. Chiarini, and J. A. McCubrey, “The phosphatidylinositol 3-kinase/Akt/mTOR signaling network as a therapeutic target in acute myelogenous leukemia patients,” Oncotarget, vol. 1, no. 2, pp. 89–103, 2010.
[42]  T. L. Yuan and L. C. Cantley, “PI3K pathway alterations in cancer: variations on a theme,” Oncogene, vol. 27, no. 41, pp. 5497–5510, 2008.
[43]  P. C. De Witt Hamer, “Small molecule kinase inhibitors in glioblastoma: a systematic review of clinical studies,” Neuro-Oncology, vol. 12, no. 3, pp. 304–316, 2010.
[44]  J. N. Rich, D. A. Reardon, T. Peery et al., “Phase II trial of gefitinib in recurrent glioblastoma,” Journal of Clinical Oncology, vol. 22, no. 1, pp. 133–142, 2004.
[45]  M. J. van den Bent, A. A. Brandes, R. Rampling et al., “Randomized phase II trial of erlotinib versus temozolomide or carmustine in recurrent glioblastoma: EORTC brain tumor group study 26034,” Journal of Clinical Oncology, vol. 27, no. 8, pp. 1268–1274, 2009.
[46]  D. Strumberg, B. Schultheis, M. E. Scheulen et al., “Phase II study of nimotuzumab, a humanized monoclonal anti-epidermal growth factor receptor (EGFR) antibody, in patients with locally advanced or metastatic pancreatic cancer,” Investigational New Drugs. In press.
[47]  J. H. Uhm, K. V. Ballman, W. Wu et al., “Phase II evaluation of gefitinib in patients with newly diagnosed Grade 4 astrocytoma: Mayo/North Central Cancer Treatment Group Study N0074,” International Journal of Radiation Oncology, Biology, Physics, vol. 80, pp. 347–353, 2010.
[48]  D. A. Reardon, A. Desjardins, J. J. Vredenburgh et al., “Phase 2 trial of erlotinib plus sirolimus in adults with recurrent glioblastoma,” Journal of Neuro-Oncology, vol. 96, no. 2, pp. 219–230, 2010.
[49]  F. Bach, “Current status of a phase III trial of nimotuzumab (ti-EGF-R) in newly diagnosed glioblastoma,” Journal of Clinical Oncology, vol. 29, supplement, 2011, Abstract no. 2059.
[50]  R. Lai, “Final analysis of act IIII: a phase II trial of PF04948568 (CDX-110) in combination with temozolomide (TMZ) in patients (PTS) with newly diagnosed glioblastoma (GBM),” Neuro-Oncology, vol. 12, p. iv76, 2010, Abstract no. OT-31.
[51]  D. D. Eisenstat, “A phase II study of daily afatinib (BIBW 2992) with or without temozolomide (21/28 days) in the treatment of patients with recurrent glioblastoma,” Journal of Clinical Oncology, vol. 29, supplement, 2011, Abstract no. 2010.
[52]  B. J. Druker, M. Talpaz, D. J. Resta et al., “Efficacy and safety of a specific inhibitor of the BCR-ABL tyrosine kinase in chronic myeloid leukemia,” The New England Journal of Medicine, vol. 344, no. 14, pp. 1031–1037, 2001.
[53]  J. Verweij, A. Van Oosterom, J. Y. Blay et al., “Imatinib mesylate (STI-571 Glivec, Gleevec) is an active agent for gastrointestinal stromal tumours, but does not yield responses in other soft-tissue sarcomas that are unselected for a molecular target: results from an EORTC Soft Tissue and Bone Sarcoma Group phase II study,” European Journal of Cancer, vol. 39, no. 14, pp. 2006–2011, 2003.
[54]  G. D. Demetri, M. Von Mehren, C. D. Blanke et al., “Efficacy and safety of imatinib mesylate in advanced gastrointestinal stromal tumors,” The New England Journal of Medicine, vol. 347, no. 7, pp. 472–480, 2002.
[55]  P. Y. Wen, W. K. A. Yung, K. R. Lamborn et al., “Phase I/II study of imatinib mesylate for recurrent malignant gliomas: North American Brain Tumor Consortium Study 99-08,” Clinical Cancer Research, vol. 12, no. 16, pp. 4899–4907, 2006.
[56]  E. Razis, P. Selviaridis, S. Labropoulos et al., “Phase II study of neoadjuvant imatinib in glioblastoma: evaluation of clinical and molecular effects of the treatment,” Clinical Cancer Research, vol. 15, no. 19, pp. 6258–6266, 2009.
[57]  T. Kreisl, “A phase II trial of sunitinib in the treatment of recurrent glioblastoma (GBM),” Neuro-Oncology, vol. 12, p. iv71, 2010, Abstract no. OT-12.
[58]  C. Lu-Emerson, A. D. Norden, J. Drappatz et al., “Retrospective study of dasatinib for recurrent glioblastoma after bevacizumab failure,” Journal of Neuro-Oncology, vol. 104, no. 1, pp. 287–291, 2011.
[59]  P. Workman, P. A. Clarke, F. I. Raynaud, and R. L. M. Van Montfort, “Drugging the PI3 kinome: from chemical tools to drugs in the clinic,” Cancer Research, vol. 70, no. 6, pp. 2146–2157, 2010.
[60]  B. M. Slomovitz, K. H. Lu, T. Johnston et al., “A phase 2 study of the oral mammalian target of rapamycin inhibitor, everolimus, in patients with recurrent endometrial carcinoma,” Cancer, vol. 116, no. 23, pp. 5415–5419, 2010.
[61]  E. Calvo, M. V. Bolós, and E. Grande, “Multiple roles and therapeutic implications of Akt signaling in cancer,” OncoTargets and Therapy, vol. 2, pp. 135–150, 2009.
[62]  H. Azim, H. A. Azim, and B. Escudier, “Targeting mTOR in cancer: renal cell is just a beginning,” Targeted Oncology, vol. 5, pp. 269–280, 2010.
[63]  S. A. Grossman, X. Ye, M. Chamberlain et al., “Talampanel with standard radiation and temozolomide in patients with newly diagnosed glioblastoma: a multicenter phase II trial,” Journal of Clinical Oncology, vol. 27, no. 25, pp. 4155–4161, 2009.
[64]  F. M. Iwamoto, T. N. Kreisl, L. Kim et al., “Phase 2 trial of talampanel, a glutamate receptor inhibitor, for adults with recurrent malignant gliomas,” Cancer, vol. 116, no. 7, pp. 1776–1782, 2010.
[65]  E. Galanis, K. A. Jaeckle, M. J. Maurer et al., “Phase II trial of Vorinostat in recurrent glioblastoma multiforme: a north central cancer treatment group study,” Journal of Clinical Oncology, vol. 27, no. 12, pp. 2052–2058, 2009.
[66]  P. Y. Wen, “Phase I study of vorinostat in combination with temozolomide in patients with malignant gliomas,” Journal of Clinical Oncology, vol. 29, supplement, 2011, Abstract no. 2032.
[67]  S. K. Singh, C. Hawkins, I. D. Clarke et al., “Identification of human brain tumour initiating cells,” Nature, vol. 432, no. 7015, pp. 396–401, 2004.
[68]  J. Lee, M. J. Son, K. Woolard et al., “Epigenetic-mediated dysfunction of the bone morphogenetic protein pathway inhibits differentiation of glioblastoma-initiating cells,” Cancer Cell, vol. 13, no. 1, pp. 69–80, 2008.
[69]  A. Eramo, L. Ricci-Vitiani, A. Zeuner et al., “Chemotherapy resistance of glioblastoma stem cells,” Cell Death and Differentiation, vol. 13, no. 7, pp. 1238–1241, 2006.
[70]  A. Natsume, S. Kinjo, K. Yuki et al., “Glioma-initiating cells and molecular pathology: implications for therapy,” Brain Tumor Pathology, vol. 28, no. 1, pp. 1–12, 2011.
[71]  M. Groszer, R. Erickson, D. D. Scripture-Adams et al., “Negative regulation of neural stem/progenitor cell proliferation by the Pten tumor suppressor gene in vivo,” Science, vol. 294, no. 5549, pp. 2186–2189, 2001.
[72]  A. M. Bleau, D. Hambardzumyan, T. Ozawa et al., “PTEN/PI3K/Akt pathway regulates the side population phenotype and ABCG2 activity in glioma tumor stem-like cells,” Cell Stem Cell, vol. 4, no. 3, pp. 226–235, 2009.
[73]  C. Gregorian, J. Nakashima, J. L. Belle et al., “Pten deletion in adult neural stem/progenitor cells enhances constitutive neurogenesis,” Journal of Neuroscience, vol. 29, no. 6, pp. 1874–1886, 2009.
[74]  C. H. Kwon, D. Zhao, J. Chen et al., “Pten haploinsufficiency accelerates formation of high-grade astrocytomas,” Cancer Research, vol. 68, no. 9, pp. 3286–3294, 2008.
[75]  A. Xiao, H. Wu, P. P. Pandolfi, D. N. Louis, and T. Van Dyke, “Astrocyte inactivation of the pRb pathway predisposes mice to malignant astrocytoma development that is accelerated by PTEN mutation,” Cancer Cell, vol. 1, no. 2, pp. 157–168, 2002.
[76]  V. Clement, P. Sanchez, N. de Tribolet, I. Radovanovic, and A. Ruiz i Altaba, “HEDGEHOG-GLI1 signaling regulates human glioma growth, cancer stem cell self-renewal, and tumorigenicity,” Current Biology, vol. 17, no. 2, pp. 165–172, 2007.
[77]  P. M. LoRusso, C. M. Rudin, J. C. Reddy et al., “Phase I trial of hedgehog pathway inhibitor vismodegib (GDC-0449) in patients with refractory, locally advanced or metastatic solid tumors,” Clinical Cancer Research, vol. 17, no. 8, pp. 2502–2511, 2011.
[78]  X. Fan, L. Khaki, T. S. Zhu et al., “NOTCH pathway blockade depletes CD133-positive glioblastoma cells and inhibits growth of tumor neurospheres and xenografts,” Stem Cells, vol. 28, no. 1, pp. 5–16, 2010.
[79]  M. M. Lino, A. Merlo, and J. L. Boulay, “Notch signaling in glioblastoma: a developmental drug target?” BMC Medicine, vol. 8, article 72, 2010.
[80]  A. Pannuti, K. Foreman, P. Rizzo et al., “Targeting Notch to target cancer stem cells,” Clinical Cancer Research, vol. 16, no. 12, pp. 3141–3152, 2010.
[81]  W. J. Nelson and R. Nusse, “Convergence of Wnt, β-Catenin, and Cadherin pathways,” Science, vol. 303, no. 5663, pp. 1483–1487, 2004.
[82]  F. Takahashi-Yanaga and M. Kahn, “Targeting Wnt signaling: can we safely eradicate cancer stem cells?” Clinical Cancer Research, vol. 16, no. 12, pp. 3153–3162, 2010.
[83]  M. W. Pitz, A. Desai, S. A. Grossman, and J. O. Blakeley, “Tissue concentration of systemically administered antineoplastic agents in human brain tumors,” Journal of Neuro-Oncology, vol. 104, no. 3, pp. 629–638, 2011.
[84]  M. Westphal, Z. Ram, V. Riddle, D. Hilt, and E. Bortey, “Gliadel wafer in initial surgery for malignant glioma: long-term follow-up of a multicenter controlled trial,” Acta Neurochirurgica, vol. 148, no. 3, pp. 269–275, 2006.
[85]  S. Valtonen, U. Timonen, P. Toivanen et al., “Interstitial chemotherapy with carmustine-loaded polymers for high- grade gliomas: a randomized double-blind study,” Neurosurgery, vol. 41, no. 1, pp. 44–49, 1997.
[86]  J. Plowman, W. R. Waud, A. D. Koutsoukos, L. V. Rubinstein, T. D. Moore, and M. R. Grever, “Preclinical antitumor activity of temozolomide in mice: efficacy against human brain tumor xenografts and synergism with 1,3-bis(2-chloroethyl)-1- nitrosourea,” Cancer Research, vol. 54, no. 14, pp. 3793–3799, 1994.
[87]  H. C. Bock, M. J. A. Puchner, F. Lohmann et al., “First-line treatment of malignant glioma with carmustine implants followed by concomitant radiochemotherapy: a multicenter experience,” Neurosurgical Review, vol. 33, no. 4, pp. 441–449, 2010.
[88]  K. S. Bankiewicz, J. L. Eberling, M. Kohutnicka et al., “Convection-enhanced delivery of AAV vector in Parkinsonian monkeys; in vivo detection of gene expression and restoration of dopaminergic function using pro-drug approach,” Experimental Neurology, vol. 164, no. 1, pp. 2–14, 2000.
[89]  P. Hadaczek, M. Kohutnicka, M. T. Krauze et al., “Convection-enhanced delivery of adeno-associated virus type 2 (AAV2) into the striatum and transport of AAV2 within monkey brain,” Human Gene Therapy, vol. 17, no. 3, pp. 291–302, 2006.
[90]  M. T. Krauze, S. R. Vandenberg, Y. Yamashita et al., “Safety of real-time convection-enhanced delivery of liposomes to primate brain: a long-term retrospective,” Experimental Neurology, vol. 210, no. 2, pp. 638–644, 2008.
[91]  T. A. Wynn, “IL-13 effector functions,” Annual Review of Immunology, vol. 21, pp. 425–456, 2003.
[92]  W. Debinski, N. I. Obiri, S. K. Powers, I. Pastan, and R. K. Puri, “Human glioma cells overexpress receptors for interleukin 13 and are extremely sensitive to a novel chimeric protein composed of interleukin 13 and Pseudomonas exotoxin,” Clinical Cancer Research, vol. 1, no. 11, pp. 1253–1258, 1995.
[93]  M. Mut, J. H. Sherman, M. E. Shaffrey, and D. Schiff, “Cintredekin besudotox in treatment of malignant glioma,” Expert Opinion on Biological Therapy, vol. 8, no. 6, pp. 805–812, 2008.
[94]  S. Kunwar, S. M. Chang, M. D. Prados et al., “Safety of intraparenchymal convection-enhanced delivery of cintredekin besudotox in early-phase studies,” Neurosurgical Focus, vol. 20, no. 4, p. E15, 2006.
[95]  C. Kjellman, S. P. Olofsson, O. Hansson et al., “Expression of TGF-β isoforms, TGF-β receptors, and Smad molecules at different stages of human glioma,” International Journal of Cancer, vol. 89, no. 3, pp. 251–258, 2000.
[96]  P. Hau, P. Jachimczak, R. Schlingensiepen et al., “Inhibition of TGF-β2 with ap 12009 in recurrent malignant gliomas: from preclinical to phase I/II studies,” Oligonucleotides, vol. 17, no. 2, pp. 201–212, 2007.
[97]  M. D. Hjelmeland, A. B. Hjelmeland, S. Sathornsumetee et al., “SB-431542, a small molecule transforming growth factor-β-receptor antagonist, inhibits human glioma cell line proliferation and motility,” Molecular Cancer Therapeutics, vol. 3, no. 6, pp. 737–745, 2004.
[98]  U. Naumann, P. Maass, A. K. Gleske, S. Aulwurm, M. Weller, and G. Eisele, “Glioma gene therapy with soluble transforming growth factor-β receptors II and IIII,” International Journal of Oncology, vol. 33, no. 4, pp. 759–765, 2008.
[99]  T. Schneider, A. Becker, K. Ringe, A. Reinhold, R. Firsching, and B. A. Sabel, “Brain tumor therapy by combined vaccination and antisense oligonucleotide delivery with nanoparticles,” Journal of Neuroimmunology, vol. 195, no. 1-2, pp. 21–27, 2008.
[100]  T. T. Tran, M. Uhl, Y. M. Jing et al., “Inhibiting TGF-β signaling restores immune surveillance in the SMA-560 glioma model,” Neuro-Oncology, vol. 9, no. 3, pp. 259–270, 2007.
[101]  M. Uhl, S. Aulwurm, J. Wischhusen et al., “SD-208, a novel transforming growth factor β receptor I kinase inhibitor, inhibits growth and invasiveness and enhances immunogenicity of murine and human glioma cells in vitro and in vivo,” Cancer Research, vol. 64, no. 21, pp. 7954–7961, 2004.
[102]  K. H. Schlingensiepen, R. Schlingensiepen, A. Steinbrecher et al., “Targeted tumor therapy with the TGF-β2 antisense compound AP 12009,” Cytokine and Growth Factor Reviews, vol. 17, no. 1-2, pp. 129–139, 2006.
[103]  L. Valliéres, “Trabedersen, a TGFβ2-specific antisense oligonucleotide for the treatment of malignant gliomas and other tumors overexpressing TGFβ2,” IDrugs, vol. 12, no. 7, pp. 445–453, 2009.
[104]  U. Bogdahn, P. Hau, G. Stockhammer et al., “Targeted therapy for high-grade glioma with the TGF-β2 inhibitor trabedersen: results of a randomized and controlled phase IIb study,” Neuro-Oncology, vol. 13, no. 1, pp. 132–142, 2011.
[105]  R. McLendon, A. Friedman, D. Bigner et al., “Comprehensive genomic characterization defines human glioblastoma genes and core pathways,” Nature, vol. 455, no. 7216, pp. 1061–1068, 2008.
[106]  D. W. Parsons, S. Jones, X. Zhang et al., “An integrated genomic analysis of human glioblastoma multiforme,” Science, vol. 321, no. 5897, pp. 1807–1812, 2008.
[107]  H. Noushmehr, D. J. Weisenberger, K. Diefes et al., “Identification of a CpG Island Methylator Phenotype that Defines a Distinct Subgroup of Glioma,” Cancer Cell, vol. 17, no. 5, pp. 510–522, 2010.
[108]  T. M. Kim, W. Huang, R. Park, P. J. Park, and M. D. Johnson, “A developmental taxonomy of glioblastoma defined and maintained by microRNAs,” Cancer Research, vol. 71, no. 9, pp. 3387–3399, 2011.
[109]  R. G. W. Verhaak, K. A. Hoadley, E. Purdom et al., “Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1,” Cancer Cell, vol. 17, no. 1, pp. 98–110, 2010.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133

WeChat 1538708413