International Journal of Frontiers in Medicine, 2025, 7(4); doi: 10.25236/IJFM.2025.070410.
Ming Wei
University of Illinois Urbana-Champaign, Champaign, Illinois, United States
Glioblastoma (GBM) is the most common and aggressive primary malignant brain tumor that originates in the brain's glial cells. Due to its rapid growth and resilience against many therapies, the prognosis for individuals with GBM remains bleak. Taxol (paclitaxel), a widely used chemotherapy treatment for various types of cancer, is also effective in killing glioblastoma cells, according to multiple studies. Here, we report through a comprehensive screening of 1,001 FDA-approved drugs, 37 drugs were identified to sensitize LN-229 glioblastoma cells to Taxol treatment. Remarkably, four of these drugs have not been previously linked to GBM treatment. Further experiments showed that three of them are even efficient in inhibiting glioblastoma cell migration. These new candidates create the potential of developing novel paclitaxel-based chemotherapy combined treatments for GBM in the future.
Glioblastoma, Combining Treatment, Paclitaxel, High-Throughput Screening, Cell Death
Ming Wei. Identification of Novel Paclitaxel Sensitizers for Glioblastoma Therapy through High-Throughput Screening. International Journal of Frontiers in Medicine (2025), Vol. 7, Issue 4: 74-84. https://doi.org/10.25236/IJFM.2025.070410.
[1] Q. T. Ostrom, H. Gittleman, G. Truitt, A. Boscia, C. Kruchko, and J. S. Barnholtz-Sloan, “CBTRUS Statistical Report: Primary Brain and Other Central Nervous System Tumors Diagnosed in the United States in 2011–2015,” Neuro-Oncology, vol. 20, no. suppl_4, pp. iv1–iv86, Oct. 2018, doi: 10. 1093/neuonc/noy131.
[2] F. Hanif, K. Muzaffar, kahkashan Perveen, S. Malhi, and S. Simjee, “Glioblastoma Multiforme: A Review of its Epidemiology and Pathogenesis through Clinical Presentation and Treatment,” APJCP, vol. 18, no. 1, Jan. 2017, doi: 10.22034/APJCP.2017.18.1.3.
[3] D. Y. Zhang et al., “Ultrasound-mediated Delivery of Paclitaxel for Glioma: A Comparative Study of Distribution, Toxicity, and Efficacy of Albumin-bound Versus Cremophor Formulations,” Clinical Cancer Research, vol. 26, no. 2, pp. 477–486, Jan. 2020, doi: 10.1158/1078-0432.CCR-19-2182.
[4] B. A. Weaver, “How Taxol/paclitaxel kills cancer cells,” MBoC, vol. 25, no. 18, pp. 2677–2681, Sept. 2014, doi: 10.1091/mbc.e14-04-0916.
[5] C.-P. Yang and S. Horwitz, “Taxol®: The First Microtubule Stabilizing Agent,” IJMS, vol. 18, no. 8, p. 1733, Aug. 2017, doi: 10.3390/ijms18081733.
[6] P. Perego et al., “Characterization of an established human, malignant, glioblastoma cell line (GBM) and its response to conventional drugs,” J Cancer Res Clin Oncol, vol. 120, no. 10, pp. 585–592, Oct. 1994, doi: 10.1007/BF01212812.
[7] D. L. Silbergeld, M. R. Chicoine, and C. L. Madsen, “In vitro assessment of Taxol for human glioblastoma: chemosensitivity and cellular locomotion,” Anti-Cancer Drugs, vol. 6, no. 2, 1995, [Online]. Available: https: //journals. lww. com/anti-cancerdrugs/fulltext/1995/04000/ in_vitro _assessment_of_taxol_for_human.11.aspx
[8] J. J. Heimans et al., “Paclitaxel (TAXOL®) concentrations in brain tumor tissue,” Annals of Oncology, vol. 5, no. 10, pp. 951–953, Dec. 1994, doi: 10.1093/oxfordjournals.annonc.a058736.
[9] I. Ullah et al., “Nose-to-Brain Delivery of Cancer-Targeting Paclitaxel-Loaded Nanoparticles Potentiates Antitumor Effects in Malignant Glioblastoma,” Mol. Pharmaceutics, vol. 17, no. 4, pp. 1193–1204, Apr. 2020, doi: 10.1021/acs.molpharmaceut.9b01215.
[10] H. Xin et al., “Enhanced anti-glioblastoma efficacy by PTX-loaded PEGylated poly(ɛ-caprolactone) nanoparticles: In vitro and in vivo evaluation,” International Journal of Pharmaceutics, vol. 402, no. 1–2, pp. 238–247, Dec. 2010, doi: 10.1016/j.ijpharm.2010.10.005.
[11] Xiaoxiao Chen, Mingqing Yuan, Qianyu Zhang, Yu Ting Yang, Huile Gao, and Qin He, “Synergistic Combination of Doxorubicin and Paclitaxel Delivered by Blood Brain Barrier and Glioma Cells Dual Targeting Liposomes for Chemotherapy of Brain Glioma,” Current Pharmaceutical Biotechnology, vol. 17, no. 7, pp. 636–650, 2016, doi: 10.2174/1389201017666160401144440.
[12] H. West et al., “Atezolizumab in combination with carboplatin plus nab-paclitaxel chemotherapy compared with chemotherapy alone as first-line treatment for metastatic non-squamous non-small-cell lung cancer (IMpower130): a multicentre, randomised, open-label, phase 3 trial,” The Lancet Oncology, vol. 20, no. 7, pp. 924–937, July 2019, doi: 10.1016/S1470-2045(19)30167-6.
[13] M. Fukuoka et al., “Biomarker Analyses and Final Overall Survival Results From a Phase III, Randomized, Open-Label, First-Line Study of Gefitinib Versus Carboplatin/Paclitaxel in Clinically Selected Patients With Advanced Non–Small-Cell Lung Cancer in Asia (IPASS),” JCO, vol. 29, no. 21, pp. 2866–2874, July 2011, doi: 10.1200/JCO.2010.33.4235.
[14] A. Du Bois et al., “Standard first-line chemotherapy with or without nintedanib for advanced ovarian cancer (AGO-OVAR 12): a randomised, double-blind, placebo-controlled phase 3 trial,” The Lancet Oncology, vol. 17, no. 1, pp. 78–89, Jan. 2016, doi: 10.1016/S1470-2045(15)00366-6.
[15] M. D. Pegram et al., “PF-05280014 (a trastuzumab biosimilar) plus paclitaxel compared with reference trastuzumab plus paclitaxel for HER2-positive metastatic breast cancer: a randomised, double-blind study,” Br J Cancer, vol. 120, no. 2, pp. 172–182, Jan. 2019, doi: 10.1038/s41416-018-0340-2.
[16] N. J. Barrows et al., “A Screen of FDA-Approved Drugs for Inhibitors of Zika Virus Infection,” Cell Host & Microbe, vol. 20, no. 2, pp. 259–270, Aug. 2016, doi: 10.1016/j.chom.2016.07.004.
[17] I. G. Stavrovskaya et al., “Clinically Approved Heterocyclics Act on a Mitochondrial Target and Reduce Stroke-induced Pathology,” The Journal of Experimental Medicine, vol. 200, no. 2, pp. 211–222, July 2004, doi: 10.1084/jem.20032053.
[18] C.-C. Liang, A. Y. Park, and J.-L. Guan, “In vitro scratch assay: a convenient and inexpensive method for analysis of cell migration in vitro,” Nat Protoc, vol. 2, no. 2, pp. 329–333, Feb. 2007, doi: 10.1038/nprot.2007.30.
[19] W. Meng et al., “Anti‐miR‐155 oligonucleotide enhances chemosensitivity of U251 cell to taxol by inducing apoptosis,” Cell Biology International, vol. 36, no. 7, pp. 653–659, July 2012, doi: 10.1042/CBI20100918.
[20] Y. Ren et al., “MicroRNA-21 inhibitor sensitizes human glioblastoma cells U251 (PTEN-mutant) and LN229 (PTEN-wild type) to taxol,” BMC Cancer, vol. 10, no. 1, p. 27, Dec. 2010, doi: 10.1186/1471-2407-10-27.
[21] X. Hong, K. Chedid, and S. N. Kalkanis, “Glioblastoma cell line-derived spheres in serum-containing medium versus serum-free medium: A comparison of cancer stem cell properties,” International Journal of Oncology, vol. 41, no. 5, pp. 1693–1700, Nov. 2012, doi: 10.3892/ijo.2012.1592.
[22] F. Touret et al., “In vitro screening of a FDA approved chemical library reveals potential inhibitors of SARS-CoV-2 replication,” Sci Rep, vol. 10, no. 1, p. 13093, Aug. 2020, doi: 10.1038/s41598-020-70143-6.
[23] S. S. Bacus et al., “Taxol-induced apoptosis depends on MAP kinase pathways (ERK and p38) and is independent of p53,” Oncogene, vol. 20, no. 2, pp. 147–155, Jan. 2001, doi: 10.1038/sj.onc.1204062.
[24] Y. H. Choi and Y. H. Yoo, “Taxol-induced growth arrest and apoptosis is associated with the upregulation of the Cdk inhibitor, p21WAF1/CIP1, in human breast cancer cells,” Oncology Reports, vol. 28, no. 6, pp. 2163–2169, Dec. 2012, doi: 10.3892/or.2012.2060.
[25] F. Seker-Polat, N. Pinarbasi Degirmenci, I. Solaroglu, and T. Bagci-Onder, “Tumor Cell Infiltration into the Brain in Glioblastoma: From Mechanisms to Clinical Perspectives,” Cancers, vol. 14, no. 2, p. 443, Jan. 2022, doi: 10.3390/cancers14020443.
[26] W. Zhang et al., “Bone Metastases of Glioblastoma: A Case Report and Review of the Literature,” Front. Oncol., vol. 11, p. 705455, Sept. 2021, doi: 10.3389/fonc.2021.705455.
[27] A. Kita et al., “Antitumor effects of YM155, a novel survivin suppressant, against human aggressive non-Hodgkin lymphoma,” Leukemia Research, vol. 35, no. 6, pp. 787–792, June 2011, doi: 10. 1016/j. leukres.2010.11.016.
[28] A. Mehta et al., “Inhibition of Survivin with YM155 Induces Durable Tumor Response in Anaplastic Thyroid Cancer,” Clinical Cancer Research, vol. 21, no. 18, pp. 4123–4132, Sept. 2015, doi: 10. 1158/1078-0432.CCR-14-3251.
[29] R. Mir et al., “YM155 sensitizes ovarian cancer cells to cisplatin inducing apoptosis and tumor regression,” Gynecologic Oncology, vol. 132, no. 1, pp. 211–220, Jan. 2014, doi: 10.1016/j. ygyno. 2013. 11. 013.
[30] K. Yamanaka, “YM155, a selective survivin suppressant, inhibits tumor spread and prolongs survival in a spontaneous metastatic model of human triple negative breast cancer,” Int J Oncol, June 2011, doi: 10.3892/ijo.2011.1077.
[31] L. R. Molife et al., “Phase II, two-stage, single-arm trial of the histone deacetylase inhibitor (HDACi) romidepsin in metastatic castration-resistant prostate cancer (CRPC),” Annals of Oncology, vol. 21, no. 1, pp. 109–113, Jan. 2010, doi: 10.1093/annonc/mdp270.
[32] D. S. Schrump et al., “Clinical and Molecular Responses in Lung Cancer Patients Receiving Romidepsin,” Clinical Cancer Research, vol. 14, no. 1, pp. 188–198, Jan. 2008, doi: 10.1158/1078-0432. CCR-07-0135.
[33] W. Zhang et al., “Histone Deacetylase Inhibitor Romidepsin Enhances Anti-Tumor Effect of Erlotinib in Non-small Cell Lung Cancer (NSCLC) Cell Lines,” Journal of Thoracic Oncology, vol. 4, no. 2, pp. 161–166, Feb. 2009, doi: 10.1097/JTO.0b013e318194fae7.
[34] A. Markham, “Tepotinib: First Approval,” Drugs, vol. 80, no. 8, pp. 829–833, June 2020, doi: 10.1007/s40265-020-01317-9.
[35] J. Albers et al., “The Preclinical Pharmacology of Tepotinib—A Highly Selective MET Inhibitor with Activity in Tumors Harboring MET Alterations,” Molecular Cancer Therapeutics, vol. 22, no. 7, pp. 833–843, July 2023, doi: 10.1158/1535-7163.MCT-22-0537.
[36] F. Cheng and D. Guo, “MET in glioma: signaling pathways and targeted therapies,” J Exp Clin Cancer Res, vol. 38, no. 1, p. 270, Dec. 2019, doi: 10.1186/s13046-019-1269-x.
[37] P. K. Paik et al., “Tepotinib in Non–Small-Cell Lung Cancer with MET Exon 14 Skipping Mutations,” N Engl J Med, vol. 383, no. 10, pp. 931–943, Sept. 2020, doi: 10.1056/NEJMoa2004407.
[38] L. D. Cripe et al., “Zosuquidar, a novel modulator of P-glycoprotein, does not improve the outcome of older patients with newly diagnosed acute myeloid leukemia: a randomized, placebo-controlled trial of the Eastern Cooperative Oncology Group 3999,” Blood, vol. 116, no. 20, pp. 4077–4085, Nov. 2010, doi: 10.1182/blood-2010-04-277269.
[39] P. M. Chaudhary and I. B. Roninson, “Expression and activity of P-glycoprotein, a multidrug efflux pump, in human hematopoietic stem cells,” Cell, vol. 66, no. 1, pp. 85–94, July 1991, doi: 10.1016/0092-8674(91)90141-K.
[40] Ignazio de Trizio, Mariella Errede, Antonio d’Amati, Francesco Girolamo, and Daniela Virgintino, “Expression of P-gp in Glioblastoma: What we can Learn from Brain Development,” Current Pharmaceutical Design, vol. 26, no. 13, pp. 1428–1437, 2020, doi: 10.2174/1381612826666200318130625.
[41] E. Ito et al., “Potential Use of Cetrimonium Bromide as an Apoptosis-Promoting Anticancer Agent for Head and Neck Cancer,” Molecular Pharmacology, vol. 76, no. 5, pp. 969–983, Nov. 2009, doi: 10. 1124/mol.109.055277.
[42] Y. Pan, Y. Zhang, Q. Chen, X. Tao, J. Liu, and G. G. Xiao, “CTAB Enhances Chemo-Sensitivity Through Activation of AMPK Signaling Cascades in Breast Cancer,” Front. Pharmacol., vol. 10, p. 843, July 2019, doi: 10.3389/fphar.2019.00843.
[43] N. M. Leli and C. Koumenis, “Pro-tumorigenic AMPK in glioblastoma,” Nat Cell Biol, vol. 20, no. 7, pp. 736–737, July 2018, doi: 10.1038/s41556-018-0129-9.
[44] M. Friese-Hamim et al., “Brain penetration and efficacy of tepotinib in orthotopic patient-derived xenograft models of MET-driven non-small cell lung cancer brain metastases,” Lung Cancer, vol. 163, pp. 77–86, Jan. 2022, doi: 10.1016/j.lungcan.2021.11.020.
[45] E. M. Kemper, C. Cleypool, W. Boogerd, J. H. Beijnen, and O. Van Tellingen, “The influence of the P-glycoprotein inhibitor zosuquidar trihydrochloride (LY335979) on the brain penetration of paclitaxel in mice,” Cancer Chemother Pharmacol, vol. 53, no. 2, pp. 173–178, Feb. 2004, doi: 10.1007/s00280-003-0720-y.
[46] S. A. A. Rizvi and A. M. Saleh, “Applications of nanoparticle systems in drug delivery technology,” Saudi Pharmaceutical Journal, vol. 26, no. 1, pp. 64–70, Jan. 2018, doi: 10.1016/j.jsps.2017.10.012.
[47] H. Nsairat, D. Khater, U. Sayed, F. Odeh, A. Al Bawab, and W. Alshaer, “Liposomes: structure, composition, types, and clinical applications,” Heliyon, vol. 8, no. 5, p. e09394, May 2022, doi: 10.1016/j.heliyon.2022.e09394.