You're using an outdated browser. Please upgrade to a modern browser for the best experience.
Submitted Successfully!
Thank you for your contribution! You can also upload a video entry or images related to this topic. For video creation, please contact our Academic Video Service.
Version Summary Created by Modification Content Size Created at Operation
1 Carmen Castro + 1944 word(s) 1944 2021-04-16 06:00:06

Video Upload Options

We provide professional Academic Video Service to translate complex research into visually appealing presentations. Would you like to try it?

Confirm

Are you sure to Delete?
Yes No
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Castro, C.; Nunez Abades, P. PKC Targeted Glioblastoma Treatment. Encyclopedia. Available online: https://encyclopedia.pub/entry/8841 (accessed on 11 November 2025).
Castro C, Nunez Abades P. PKC Targeted Glioblastoma Treatment. Encyclopedia. Available at: https://encyclopedia.pub/entry/8841. Accessed November 11, 2025.
Castro, Carmen, Pedro Nunez Abades. "PKC Targeted Glioblastoma Treatment" Encyclopedia, https://encyclopedia.pub/entry/8841 (accessed November 11, 2025).
Castro, C., & Nunez Abades, P. (2021, April 20). PKC Targeted Glioblastoma Treatment. In Encyclopedia. https://encyclopedia.pub/entry/8841
Castro, Carmen and Pedro Nunez Abades. "PKC Targeted Glioblastoma Treatment." Encyclopedia. Web. 20 April, 2021.
PKC Targeted Glioblastoma Treatment
Edit

Glioblastoma (GBM) is the most frequent and aggressive primary brain tumor and is associated with a poor prognosis. Despite the use of combined treatment approaches, recurrence is almost inevitable and survival longer than 14 or 15 months after diagnosis is low. It is therefore necessary to identify new therapeutic targets to fight GBM progression and recurrence. Some publications have pointed out the role of glioma stem cells (GSCs) as the origin of GBM. These cells, with characteristics of neural stem cells (NSC) present in physiological neurogenic niches, have been proposed as being responsible for the high resistance of GBM to current treatments such as temozolomide (TMZ). The protein Kinase C (PKC) family members play an essential role in transducing signals related with cell cycle entrance, differentiation and apoptosis in NSC and participate in distinct signaling cascades that determine NSC and GSC dynamics. Thus, PKC could be a suitable drugable target to treat recurrent GBM. Clinical trials have tested the efficacy of PKCβ inhibitors, and preclinical studies have focused on other PKC isozymes.

glioblastoma protein kinase C glioma stem cells neurogenesis neural stem cells temozolomide enzastaurin epidermal growth factor receptor neuregulin

1. Introduction

Gliomas account for 30% of all tumors of the central nervous system (CNS) and 80% of all malignant brain tumors in adults [1]. Although diffuse gliomas are considered as rare disorders, approximately 100,000 people worldwide are diagnosed with this pathology every year, with it being the second most common cancer in children and adolescents (26% of all cancers) [2][3][4]. Glioma incidence varies with age, sex, ethnicity, tumor histology, and between populations around the world [5].

Based on histopathological criteria and malignancy grade, gliomas are classified as astrocytomas and oligodendrogliomas (OD), either diffuse (grade II) or anaplastic (grade III) and glioblastomas (GBM; grade IV). Briefly, histological features used in the clinical diagnosis of gliomas include: nuclear atypia (grade III), in addition to necrosis and microvascular changes (grade IV). In addition, these latter tumors exhibit high infiltrative and proliferative capacity and increased mitotic activity compared to grade II lesions [6]. This review focuses on GBMs as they are the most common (54% of all gliomas) and the most aggressive in adults, with a median overall survival (OS) of ≈15 months. Additionally, GBM has a high incidence of recurrency (>90%), despite intensive clinical management including surgery, radiotherapy and adjuvant chemotherapy. GBM can appear at any age but the peak incidence is between 75 to 84 years [7]. The incidence of these tumors is approximately 50% higher in males compared to females [8] and it differs substantially between ethnic groups, e.g., it is higher in Caucasians as compared to black populations [7][9][10]. As regards their location, they are most commonly situated in the supratentorial region (frontal, parietal, temporal and occipital lobes), with the highest incidence in the frontal lobe [11], while they are rarely located in the cerebellum [12]. Furthermore, the incidence of GBM increases in patients with hereditary tumor syndromes, such as Turcot syndrome [13] and Li-Fraumeni syndrome [14].

Since diagnosis and prognosis of gliomas based on histological features is insufficient, the 2016 World Health Organization Classification of Tumors of the Central Nervous System (2016 CNS WHO) incorporated molecular parameters to improve clinical interventions in patients with this pathology [15]. Amongst the molecular genetic alterations used to redefine glioma entities, the most common are mutations in the isocitrate dehydrogenase 1 and 2 (IDH1, IDH2) genes and the 1 p/19q co-deletion status [16][17][18]. According to the IDH condition, gliomas are divided into IDH-mutant (IDH1 R132 or IDH2 R172) and IDH-wild-type; 90% of GBMs (usually primary or “de novo” GBMs) are wild-type for IDH and have a poor prognosis (median OS of 1.2 years). Meanwhile, secondary GBM (10%) develops through progression from a low-grade lesion and is associated with a better prognosis and survival rate due to the IDH mutation (median OS of 3.6 years) [1][6][19][20][21]. This biomarker, used in combination with the loss of heterozygosity in chromosomal arms 1p/19q for the diagnosis of grade II and III oligodendrogliomas, is linked to favorable clinical behaviors [20][21]. In the case of tumors which cannot be classified into any of these groups due to lacking sufficient pathological and genetic information (i.e., absence of appropriate diagnostic molecular testing or inconclusive results), the 2016 CNS WHO assigned the NOS (Not Otherwise Specified) category, which should be the subject to future studies [15][16]. Nonetheless, other biomarkers with predictive value of the progression and response to the first-front therapeutics are also frequently used in the clinic. The loss of ATRX (alpha thalassemia/mental retardation syndrome X-linked) is a recurrent marker of astrocytoma and secondary GBM and is associated with IDH and TP53 mutations, which are linked to a good outcome [22]. Hypermethylation of the MGMT (O6-methylguanine-DNA methyltransferase) promoter is considered an important predictor of a good response to chemotherapy with temozolomide (TMZ) in glioma patients [23][24]. Telomerase reverse transcriptase (TERT) promoter mutations have been detected in more than 50% of primary adult GBM and are correlated with increased telomerase activity [24][25], having been linked to lower survival times in GBM patients [26]. However, in combination with IDH1 and MGMT mutations, these mutations are good predictors of grade II and grade III gliomas [27]. Finally, EGFR (epidermal growth factor receptor) expression, without loss of PTEN (phosphatase and tensin homolog), explains the sensitivity of gliomas to tyrosine kinase inhibitors [28]. In conclusion, the use of molecular traits is assisting with the classification of gliomas, the high biological heterogeneity of which require the use of different experimental models for their study [29] and different strategies of clinical management.

2. Clinical Trials Using PKC Targeting Drugs

Several clinical trials conducted over the past 20 years have tested the effects of PKC inhibitors in GBM, mainly classical PKC inhibitors in the treatment of recurrent GBM. Treatment of relapsing patients is still challenging, as current clinical management involves surgery, radiotherapy and TMZ treatment with no better outcomes having been found by using alternative drugs (reviewed in Finch et al., 2021 [30]).

The first clinical trials using PKC targeting drugs tested the efficacy of tamoxifen. This nonsteroidal agent with high lipid solubility is able to cross the blood–brain barrier (BBB) and reach the tumor. Tamoxifen elicits the association of PKC to the membrane, followed by an irreversible activation, and subsequent down-regulation of the enzyme, leading to cell growth inhibition [31], cellular apoptosis, and at high doses, chemoresistance reversion [32][33]. For the treatment of GBM, Couldwell and colleagues and Brandes and colleagues were the first to use high-doses of tamoxifen to inhibit PKC, based on in vitro assays that evaluated apoptosis in GBM cells either alone, or in combination with procarbazine in phase II clinical trials. The most relevant finding was an increase in radiosensitivity [34][35]. However, the OS rates shown in these studies were 6.8 and 7.2 months, and the time to progression was 3.3 months. More promising results were observed in a more recent study in which tamoxifen was tested in combination with TMZ. In this study the observed median time to progression was 9.5 months and the OS was 17.5 months [36]. Additional clinical trials have tested tamoxifen in combination with other agents such as procarbazine or TMZ to affect PKC functionality and other targets [37][38][39] (see Table 1 for further details).

Table 1. Summary of protein kinase C-related clinical trials for the treatment of glioblastoma.

  Target Authors and Year Trial Phase Nº Patients Dose PFS OS
Tamoxifen PKC Couldwell et al 1996 [34] Phase II trial 32 200 mg/day (100 mg twice daily) of tamoxifen was administered to males 160 mg/day (80 mg twice daily) of tamoxifen was administered to females n.d. 7.2 months
Tamoxifen + Procarbazine PKC + DNA Brandes et al 1999 [38] Phase II trial 53 100 mg/day of tamoxifen + 100 mg/m2/day of procarbazine were administered for 30 days with 30-day intervals between cycles 3 months (median) 6.2 months
Tamoxifen + TMZ PKC + DNA Spence et al. 2004 [39], Cristofori et al. 2013 [36] Phase II trial 16 40 mg twice daily of tamoxifen for 1 week and was escalated to 60 mg, 80 mg then 100 mg + 75 mg/m2/day of TMZ for 6 weeks, repeated every 10 weeks, with a maximum of 5 cycles n.d. 6 months
  PKC + DNA Cristofori et al. 2013 [36] Phase II trial 32 80 mg/m2/day of tamoxifen + 75–150 mg/m2/day of TMZ was administered for one week on/one week off 9.5 months (median) 17.5 months
Tamoxifen + Radiation PKC Robins et al. 2006 [35] Phase II trial 75 80 mg/m2/day of tamoxifen, divided in 4 doses of 20 mg/m2 every 6 h, was administered during and after of 60 Gy in 30 fractions × 2 Gy of radiotherapy 2.9 months (median) 11.3 months
Enzastaurin PKCβ Kreisl et al. 2009 [40] Kreisl et al. 2010 [41] Phase I trialPhase I/II trial 2215 (Phase I) 103 (Phase II) 800 mg/day of enzastaurin and 400 mg twice daily and 500 mg/day and 250 mg twice daily for patients not taking EIAEDs and 1000 mg/day and 500 mg twice daily for patients taking EIAEDs in phase I, patients who were taking EIAEDs, received 525, 700 and 900 mg/day of enzastaurin and patients in phase II, who were not taking EIAEDs, received 500 or 525 mg/day of enzastaurin 1.4 months (median) 1.3 months (median) 7% (at 6-month) 5.7 months 4.6 months
Enzastaurin vs. Lomustine PKCβ vs. DNA/Stathmin-4 Wick et al. 2010 [42] Phase III trial 266 500 mg/day of enzastaurin vs. 100 to 130 mg/m2 of lomustine on day 1 with cycles of 6 weeks Enzastaurin: 1.5 months, 11.1% (median, at 6-month); Lomustine: 1.6 months, 19% (median, at 6-month) Enzastaurin: 6.6 months Lomustine: 7.1 months
Enzastaurin + TMZ PKC β + DNA Rampling et al. 2012 [43] Phase I trial 28 250 mg/day (once daily); 500 mg/day (once daily); 500 mg/day (250 mg twice daily) of enzastaurine. 150–200 mg/m2 TMZ 5.5 months (median) 11.7 months
Enzastaurin + TMZ with radiation PKC β + DNA Butowski et al. 2010 [44] Phase I trial 12 Radiation therapy 1.8–2.0 Gy × 30 fractions 5 days a week for 6 weeks + Enzastaurin 250–500 mg/daily + TMZ 75 mg/m2 n.d. n.d.
Enzastaurin + Bevazizumab PKC β + VEGF Odia et al. 2016 [45] Phase II trial 40 Enzastaurin 500 or 875 mg/day + bevacizumab 10 mg/kg intravenously biweekly 2.0 months 7.5 months
Aprinocarsen PKC α Grossman et al. 2005 [46] Phase II trial 21 2 mg/kg/day of aprinocarsen was administered for 21 days per month 1.2 months (median) 3.4 months
PFS, progression-free survival; OS, overall survival; EIAEDs: enzyme-inducing anti-epileptic drugs; Gy: gray; n.d: not determined; OS: overall survival; PFS: progression-free survival; PKC: protein kinase C; TMZ: temozolomide.

 

Undoubtedly, the most relevant clinical trials implicating PKC in GBM so far have analyzed the effects of enzastaurine. This small molecule is an inhibitor of PKC β that has been used for the treatment of a variety of tumors and, similarly to tamoxifen, is a lipid soluble compound that can cross the BBB. Enzastaurin was originally developed as an anti-angiogenic agent based on the role of PKC β in angiogenesis [47][48][49][50]. However, its specificity is not very high for PKC β since, at higher concentrations, the drug can inhibit other PKC isoforms including PKCα [48][49]. Despite this, enzastaurin has shown a longer half-life than TMZ (12–40 h vs. 1.8 h) and remarkable radiographic response rates in recurrent high-grade gliomas [43][44]. Although a phase III clinical trial failed to demonstrate such efficacy after comparing the monotherapies of enzastaurin and lomustine in recurrent GBM, the PFS was 1.5 months and OS was 6.6 months [42]. Therefore, these trials did not improve the effectiveness of current treatments, even in combination with lomustine, a nitrosourea that interacts with DNA, commonly used as a chemoterapeutic agent [42][40][41]. Other strategies have been explored—such as a combination with bevacizumab—with no further improvement [45]. Therefore, clinical trials using the PKC inhibitor enzastaurin have not succeeded at limiting GBM progression and invasion, either alone, or in combination with TMZ or other compounds. In addition to tamoxifen and enzastaurin, a trial to test the PKCα inhibitor aprinocarsen has been carried out without success [46].

As stated above, the tumor microenvironment and intra-tumor heterogeneity should be considered as responsible for the lack of improvement found in phase III trials with enzastaurin. Alteration of PKC expression and activity among the different cell types within the tumor needs also to be considered. Alternatively, enzastaurin itself might modify PKC expression or alter the signaling cascades that interact with PKC activity, creating resistance. Thus, identification of signaling cascades associated with PKC and compounds that target these molecules might help in the design of additional therapies that overcome intra-tumor heterogeneity and TME induced evolution.

References

  1. Goodenberger, M.L.; Jenkins, R.B. Genetics of adult glioma. Cancer Genet. 2012, 205, 613–621.
  2. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. Ca Cancer J. Clin. 2018, 68, 394–424.
  3. Molinaro, A.M.; Taylor, J.W.; Wiencke, J.K.; Wrensch, M.R. Genetic and molecular epidemiology of adult diffuse glioma. Nat. Rev. Neurol. 2019, 15, 405–417.
  4. Suresh, S.G.; Srinivasan, A.; Scott, J.X.; Rao, S.M.; Chidambaram, B.; Chandrasekar, S. Profile and Outcome of Pediatric Brain Tumors—Experience from a Tertiary Care Pediatric Oncology Unit in South India. J. Pediatr. Neurosci. 2017, 12, 237–244.
  5. Miranda-Filho, A.; Pineros, M.; Soerjomataram, I.; Deltour, I.; Bray, F. Cancers of the brain and CNS: Global patterns and trends in incidence. Neuro. Oncol. 2017, 19, 270–280.
  6. Ohgaki, H.; Kleihues, P. The definition of primary and secondary glioblastoma. Clin. Cancer Res. 2013, 19, 764–772.
  7. Tamimi, A.F.; Juweid, M. Epidemiology and Outcome of Glioblastoma. In Glioblastoma; De Vleeschouwer, S., Ed.; Exon Publications: Brisbane, Australia, 2017.
  8. Ostrom, Q.T.; Kinnersley, B.; Wrensch, M.R.; Eckel-Passow, J.E.; Armstrong, G.; Rice, T.; Chen, Y.; Wiencke, J.K.; McCoy, L.S.; Hansen, H.M.; et al. Sex-specific glioma genome-wide association study identifies new risk locus at 3p21.31 in females, and finds sex-differences in risk at 8q24.21. Sci. Rep. 2018, 8, 7352.
  9. Hanif, F.; Muzaffar, K.; Perveen, K.; Malhi, S.M.; Simjee Sh, U. Glioblastoma Multiforme: A Review of its Epidemiology and Pathogenesis through Clinical Presentation and Treatment. Asian Pac. J. Cancer Prev. 2017, 18, 3–9.
  10. Iacob, G.; Dinca, E.B. Current data and strategy in glioblastoma multiforme. J. Med. Life 2009, 2, 386–393.
  11. Chakrabarti, I.; Cockburn, M.; Cozen, W.; Wang, Y.P.; Preston-Martin, S. A population-based description of glioblastoma multiforme in Los Angeles County, 1974–1999. Cancer 2005, 104, 2798–2806.
  12. Adams, H.; Chaichana, K.L.; Avendano, J.; Liu, B.; Raza, S.M.; Quinones-Hinojosa, A. Adult cerebellar glioblastoma: Understanding survival and prognostic factors using a population-based database from 1973 to 2009. World Neurosurg. 2013, 80, e237–e243.
  13. Hamilton, S.R.; Liu, B.; Parsons, R.E.; Papadopoulos, N.; Jen, J.; Powell, S.M.; Krush, A.J.; Berk, T.; Cohen, Z.; Tetu, B.; et al. The molecular basis of Turcot’s syndrome. N. Engl. J. Med. 1995, 332, 839–847.
  14. Kleihues, P.; Ohgaki, H. Primary and secondary glioblastomas: From concept to clinical diagnosis. Neuro Oncol. 1999, 1, 44–51.
  15. Louis, D.N.; Perry, A.; Reifenberger, G.; von Deimling, A.; Figarella-Branger, D.; Cavenee, W.K.; Ohgaki, H.; Wiestler, O.D.; Kleihues, P.; Ellison, D.W. The 2016 World Health Organization Classification of Tumors of the Central Nervous System: A summary. Acta Neuropathol. 2016, 131, 803–820.
  16. Cancer Genome Atlas Research Network; Brat, D.J.; Verhaak, R.G.; Aldape, K.D.; Yung, W.K.; Salama, S.R.; Cooper, L.A.; Rheinbay, E.; Miller, C.R.; Vitucci, M.; et al. Comprehensive, Integrative Genomic Analysis of Diffuse Lower-Grade Gliomas. N. Engl. J. Med. 2015, 372, 2481–2498.
  17. Sahm, F.; Reuss, D.; Koelsche, C.; Capper, D.; Schittenhelm, J.; Heim, S.; Jones, D.T.; Pfister, S.M.; Herold-Mende, C.; Wick, W.; et al. Farewell to oligoastrocytoma: In situ molecular genetics favor classification as either oligodendroglioma or astrocytoma. Acta Neuropathol. 2014, 128, 551–559.
  18. Wiestler, B.; Capper, D.; Sill, M.; Jones, D.T.; Hovestadt, V.; Sturm, D.; Koelsche, C.; Bertoni, A.; Schweizer, L.; Korshunov, A.; et al. Integrated DNA methylation and copy-number profiling identify three clinically and biologically relevant groups of anaplastic glioma. Acta Neuropathol. 2014, 128, 561–571.
  19. Ostrom, Q.T.; Gittleman, H.; Truitt, G.; Boscia, A.; Kruchko, C.; Barnholtz-Sloan, J.S. CBTRUS Statistical Report: Primary Brain and Other Central Nervous System Tumors Diagnosed in the United States in 2011-2015. Neuro Oncol. 2018, 20, iv1–iv86.
  20. Yip, S.; Butterfield, Y.S.; Morozova, O.; Chittaranjan, S.; Blough, M.D.; An, J.; Birol, I.; Chesnelong, C.; Chiu, R.; Chuah, E.; et al. Concurrent CIC mutations, IDH mutations, and 1p/19q loss distinguish oligodendrogliomas from other cancers. J. Pathol. 2012, 226, 7–16.
  21. Buckner, J.; Giannini, C.; Eckel-Passow, J.; Lachance, D.; Parney, I.; Laack, N.; Jenkins, R. Management of diffuse low-grade gliomas in adults—Use of molecular diagnostics. Nat. Rev. Neurol. 2017, 13, 340–351.
  22. Wiestler, B.; Capper, D.; Holland-Letz, T.; Korshunov, A.; von Deimling, A.; Pfister, S.M.; Platten, M.; Weller, M.; Wick, W. ATRX loss refines the classification of anaplastic gliomas and identifies a subgroup of IDH mutant astrocytic tumors with better prognosis. Acta Neuropathol. 2013, 126, 443–451.
  23. Dahlrot, R.H.; Dowsett, J.; Fosmark, S.; Malmstrom, A.; Henriksson, R.; Boldt, H.; de Stricker, K.; Sorensen, M.D.; Poulsen, H.S.; Lysiak, M.; et al. Prognostic value of O-6-methylguanine-DNA methyltransferase (MGMT) protein expression in glioblastoma excluding nontumour cells from the analysis. Neuropathol. Appl. Neurobiol. 2018, 44, 172–184.
  24. Brennan, C.W.; Verhaak, R.G.; McKenna, A.; Campos, B.; Noushmehr, H.; Salama, S.R.; Zheng, S.; Chakravarty, D.; Sanborn, J.Z.; Berman, S.H.; et al. The somatic genomic landscape of glioblastoma. Cell 2013, 155, 462–477.
  25. Vinagre, J.; Almeida, A.; Populo, H.; Batista, R.; Lyra, J.; Pinto, V.; Coelho, R.; Celestino, R.; Prazeres, H.; Lima, L.; et al. Frequency of TERT promoter mutations in human cancers. Nat. Commun. 2013, 4, 2185.
  26. Killela, P.J.; Reitman, Z.J.; Jiao, Y.; Bettegowda, C.; Agrawal, N.; Diaz, L.A.; Friedman, A.H.; Friedman, H.; Gallia, G.L.; Giovanella, B.C.; et al. TERT promoter mutations occur frequently in gliomas and a subset of tumors derived from cells with low rates of self-renewal. Proc. Natl. Acad. Sci. USA 2013, 110, 6021–6026.
  27. Lee, Y.; Koh, J.; Kim, S.I.; Won, J.K.; Park, C.K.; Choi, S.H.; Park, S.H. The frequency and prognostic effect of TERT promoter mutation in diffuse gliomas. Acta Neuropathol. Commun. 2017, 5, 62.
  28. Mellinghoff, I.K.; Wang, M.Y.; Vivanco, I.; Haas-Kogan, D.A.; Zhu, S.; Dia, E.Q.; Lu, K.V.; Yoshimoto, K.; Huang, J.H.; Chute, D.J.; et al. Molecular determinants of the response of glioblastomas to EGFR kinase inhibitors. N. Engl. J. Med. 2005, 353, 2012–2024.
  29. Gómez-Oliva, R.; Domínguez-García, S.; Carrascal, L.; Abalos-Martínez, J.; Pardillo-Díaz, R.; Verástegui, C.; Castro, C.; Nunez-Abades, P.; Geribaldi-Doldán, N. Evolution of Experimental Models in the Study of Glioblastoma: Toward Finding Efficient Treatments. Front. Oncol. 2021, 10, 3245.
  30. Finch, A.; Solomou, G.; Wykes, V.; Pohl, U.; Bardella, C.; Watts, C. Advances in Research of Adult Gliomas. Int. J. Mol. Sci 2021, 22, 924.
  31. Gundimeda, U.; Chen, Z.H.; Gopalakrishna, R. Tamoxifen modulates protein kinase C via oxidative stress in estrogen receptor-negative breast cancer cells. J. Biol. Chem. 1996, 271, 13504–13514.
  32. Millward, M.J.; Cantwell, B.M.; Lien, E.A.; Carmichael, J.; Harris, A.L. Intermittent high-dose tamoxifen as a potential modifier of multidrug resistance. Eur. J. Cancer 1992, 28, 805–810.
  33. Pollack, I.F.; Randall, M.S.; Kristofik, M.P.; Kelly, R.H.; Selker, R.G.; Vertosick, F.T., Jr. Effect of tamoxifen on DNA synthesis and proliferation of human malignant glioma lines in vitro. Cancer Res. 1990, 50, 7134–7138.
  34. Couldwell, W.T.; Hinton, D.R.; Surnock, A.A.; DeGiorgio, C.M.; Weiner, L.P.; Apuzzo, M.L.; Masri, L.; Law, R.E.; Weiss, M.H. Treatment of recurrent malignant gliomas with chronic oral high-dose tamoxifen. Clin. Cancer Res. 1996, 2, 619–622.
  35. Robins, H.I.; Won, M.; Seiferheld, W.F.; Schultz, C.J.; Choucair, A.K.; Brachman, D.G.; Demas, W.F.; Mehta, M.P. Phase 2 trial of radiation plus high-dose tamoxifen for glioblastoma multiforme: RTOG protocol BR-0021. Neuro Oncol. 2006, 8, 47–52.
  36. Cristofori, A.; Carrabba, G.; Lanfranchi, G.; Menghetti, C.; Rampini, P.; Caroli, M. Continuous tamoxifen and dose-dense temozolomide in recurrent glioblastoma. Anticancer Res. 2013, 33, 3383–3389.
  37. Gao, J.; Aksoy, B.A.; Dogrusoz, U.; Dresdner, G.; Gross, B.; Sumer, S.O.; Sun, Y.; Jacobsen, A.; Sinha, R.; Larsson, E.; et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci. Signal. 2013, 6, pl1.
  38. Brandes, A.A.; Ermani, M.; Turazzi, S.; Scelzi, E.; Berti, F.; Amista, P.; Rotilio, A.; Licata, C.; Fiorentino, M.V. Procarbazine and high-dose tamoxifen as a second-line regimen in recurrent high-grade gliomas: A phase II study. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 1999, 17, 645–650.
  39. Spence, A.M.; Peterson, R.A.; Scharnhorst, J.D.; Silbergeld, D.L.; Rostomily, R.C. Phase II study of concurrent continuous Temozolomide (TMZ) and Tamoxifen (TMX) for recurrent malignant astrocytic gliomas. J. Neuro Oncol. 2004, 70, 91–95.
  40. Kreisl, T.N.; Kim, L.; Moore, K.; Duic, P.; Kotliarova, S.; Walling, J.; Musib, L.; Thornton, D.; Albert, P.S.; Fine, H.A. A phase I trial of enzastaurin in patients with recurrent gliomas. Clin. Cancer Res. 2009, 15, 3617–3623.
  41. Kreisl, T.N.; Kotliarova, S.; Butman, J.A.; Albert, P.S.; Kim, L.; Musib, L.; Thornton, D.; Fine, H.A. A phase I/II trial of enzastaurin in patients with recurrent high-grade gliomas. Neuro Oncol. 2010, 12, 181–189.
  42. Wick, W.; Puduvalli, V.K.; Chamberlain, M.C.; van den Bent, M.J.; Carpentier, A.F.; Cher, L.M.; Mason, W.; Weller, M.; Hong, S.; Musib, L.; et al. Phase III study of enzastaurin compared with lomustine in the treatment of recurrent intracranial glioblastoma. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2010, 28, 1168–1174.
  43. Rampling, R.; Sanson, M.; Gorlia, T.; Lacombe, D.; Lai, C.; Gharib, M.; Taal, W.; Stoffregen, C.; Decker, R.; van den Bent, M.J. A phase I study of LY317615 (enzastaurin) and temozolomide in patients with gliomas (EORTC trial 26054). Neuro Oncol. 2012, 14, 344–350.
  44. Butowski, N.; Chang, S.M.; Lamborn, K.R.; Polley, M.Y.; Parvataneni, R.; Hristova-Kazmierski, M.; Musib, L.; Nicol, S.J.; Thornton, D.E.; Prados, M.D. Enzastaurin plus temozolomide with radiation therapy in glioblastoma multiforme: A phase I study. Neuro Oncol. 2010, 12, 608–613.
  45. Odia, Y.; Iwamoto, F.M.; Moustakas, A.; Fraum, T.J.; Salgado, C.A.; Li, A.; Kreisl, T.N.; Sul, J.; Butman, J.A.; Fine, H.A. A phase II trial of enzastaurin (LY317615) in combination with bevacizumab in adults with recurrent malignant gliomas. J. Neuro Oncol. 2016, 127, 127–135.
  46. Grossman, S.A.; Alavi, J.B.; Supko, J.G.; Carson, K.A.; Priet, R.; Dorr, F.A.; Grundy, J.S.; Holmlund, J.T. Efficacy and toxicity of the antisense oligonucleotide aprinocarsen directed against protein kinase C-alpha delivered as a 21-day continuous intravenous infusion in patients with recurrent high-grade astrocytomas. Neuro Oncol. 2005, 7, 32–40.
  47. Graff, J.R.; McNulty, A.M.; Hanna, K.R.; Konicek, B.W.; Lynch, R.L.; Bailey, S.N.; Banks, C.; Capen, A.; Goode, R.; Lewis, J.E.; et al. The protein kinase Cbeta-selective inhibitor, Enzastaurin (LY317615.HCl), suppresses signaling through the AKT pathway, induces apoptosis, and suppresses growth of human colon cancer and glioblastoma xenografts. Cancer Res. 2005, 65, 7462–7469.
  48. Teicher, B.A.; Alvarez, E.; Menon, K.; Esterman, M.A.; Considine, E.; Shih, C.; Faul, M.M. Antiangiogenic effects of a protein kinase Cbeta-selective small molecule. Cancer Chemother. Pharm. 2002, 49, 69–77.
  49. Teicher, B.A.; Menon, K.; Alvarez, E.; Shih, C.; Faul, M.M. Antiangiogenic and antitumor effects of a protein kinase Cbeta inhibitor in human breast cancer and ovarian cancer xenografts. Investig. New Drugs 2002, 20, 241–251.
  50. Ma, S.; Rosen, S.T. Enzastaurin. Curr. Opin. Oncol. 2007, 19, 590–595.
More
Upload a video for this entry
Information
Subjects: Oncology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : Carmen Castro , PEDRO NUNEZ ABADES
View Times: 562
Revision: 1 time (View History)
Update Date: 20 Apr 2021
1000/1000
Hot Most Recent
Notice
You are not a member of the advisory board for this topic. If you want to update advisory board member profile, please contact office@encyclopedia.pub.
OK
Confirm
Only members of the Encyclopedia advisory board for this topic are allowed to note entries. Would you like to become an advisory board member of the Encyclopedia?
Yes
No
Academic Video Service