Drug clinical trials on high-grade gliomas: challenges and hopes ================================================================ * Mengqian Huang * Shenglan Li * Parker Li * Zhuang Kang * Botao Zhang * Wenbin Li Gliomas are the most common group of malignant central nervous system tumors across all ages and have high heterogeneity. According to histopathologic criteria and molecular pathology, gliomas are classified as grades 1–4. High-grade gliomas (HGG), including the most aggressive subtype, glioblastoma (GBM), belong to grade 3 or 4. Although a standard therapy comprising surgical resection, chemotherapy, and radiation is available, almost all patients with HGG experience recurrence within several months and a dismal prognosis1. Owing to the aggressive growth pattern of HGG, complete surgical resection is difficult to achieve, particularly for deep-seated tumors, and recurrence is inevitable. Therefore, systematic treatment approaches are urgently needed. In recent years, clinical research on glioma medications has made remarkable breakthroughs in many areas. One notable area of focus is the advancement of immunotherapies. In investigation of the molecular characteristics of glioma cells, targeted therapies have become a research focus. Various drugs targeting particular molecules or signaling pathways have been developed. These medications can accurately target the growth and spread of tumor cells while causing less harm to normal brain tissues than to the tumor cells. ## Challenges Systematic treatment approaches, which are represented by drugs, face several challenges. HGGs exhibit high inter- and intra-tumor heterogeneity with complex molecular characteristics, thus leading to difficulties in the development of safe and effective therapies. Another major obstacle is the emergence of drug resistance, which may be associated with the subpopulations of glioma stem cells. In addition, the blood–brain barrier (BBB) impedes drug absorption. Failure to deliver drugs across the BBB leads to insufficient therapeutic drug concentrations at the brain tumor site, thus hampering effective treatment2. Therefore, additional efforts should be made to penetrate the BBB to enhance drug efficacy. To date, no therapy has demonstrated curative potential. Therefore, a need exists to explore novel therapeutics. ## Hopes ### Chemotherapy Efforts have been dedicated to the development of traditional chemotherapy. The front-line drug temozolomide (TMZ) from the Stupp regimen has long played a major role in chemotherapy. Challenges with chemotherapy drugs involve increasing their BBB permeation while minimizing their toxicity to normal tissues. Some nitrosoureas have antitumor activity and high lipophilicity, thus allowing them to permeate the BBB. Hence, they have potential for treating gliomas. Lomustine (CCNU) is a member of the first-generation nitrosoureas and a key part of the PCV regimen (where P indicates procarbazine, C indicates lomustine, and V indicates vincristine) used in recurrent gliomas. CCNU has served as the control arm in several randomized phase III trials. Patients with MGMT-methylated tumors may benefit from nitrosoureas, although no survival advantages have been observed with nitrosoureas combined with bevacizumab3. Irinotecan is a topoisomerase 1 inhibitor that has shown anti-glioma efficacy in animal studies. Owing to its lipophilic characteristics, irinotecan is an attractive drug that can be combined with other therapies, such as TMZ or antivascular endothelial growth factor (VEGF) therapies. A phase II clinical study conducted in 2007 used a combination of irinotecan and bevacizumab for the treatment of gliomas and yielded favorable results4. ### Immunotherapy Several studies have highlighted the immunosuppressive characteristics of gliomas with elevated levels of immunosuppressive factors and decreased immune cell activity. Treating gliomas through immune system activation has emerged as a promising new approach for improving patient survival and quality of life. The exploration of immune checkpoints, such as programmed cell death protein 1(PD-1)/programmed death-ligand 1(PD-L1), plays a crucial role in the advancement of cancer immunotherapy. However, clinical trials on PD-1/PD-L1 in recurrent or newly diagnosed GBM have not indicated its advantages in either overall survival (OS) or progression-free survival (PFS)5. The lack of meaningful benefit is attributed to the influence of the innate immunosuppressive microenvironment. A rational combination of trials integrating preclinical and clinical investigations may be suitable for investigating this drug. A study has found that anti-PD-1 combined with anti-PD-L1 exhibits intracranial efficacy in patients with melanoma with brain metastases; although this activity is attributable partly to BBB dysfunction caused by tumor metastasis, this new approach may aid in development of the next generation of glioma clinical trials6. A phase I clinical trial has investigated this combination treatment, and demonstrated its safety and benefits in OS in recurrent GBM (including intravenous and intracranial administration)7, and phase II/III studies are currently in progress (ClinicalTrials.gov identifiers [NCT04145115](http://www.cancerbiomed.org/lookup/external-ref?link\_type=CLINTRIALGOV&access_num=NCT04145115&atom=%2Fcbm%2F20%2F12%2F947.atom) and [NCT04396860](http://www.cancerbiomed.org/lookup/external-ref?link_type=CLINTRIALGOV&access_num=NCT04396860&atom=%2Fcbm%2F20%2F12%2F947.atom)). Oncolytic viruses (OVs) are products of genetic engineering designed to selectively infect and kill tumor cells, while causing minimal harm to normal tissues. Beyond their direct oncolytic effects, OVs upregulate immunosuppressive metabolites in the tumor microenvironment and modulate the expression of checkpoint proteins, thus leading to a sustained immune response. Hence, OVs are highly promising therapeutic approaches that are being extensively studied preclinically and clinically8. Herpes simplex virus (HSV) is neurotropic and is ideal for neurogenic tumor targeting. HSV-1 was the first genetically engineered virus to be used in treating malignant glioma; however, during the research period, neither its safety nor its efficacy was confirmed. Since then, mutant HSV vectors have been developed, including the first generation represented by G207, with enhanced safety9; the second generation represented by T-vec, the first OV whose efficacy was confirmed in a phase III clinical trial10; and the third generation oncolytic HSV-1, represented by G47∆. With a triple mutation in the genome, G47Δ displays greater replicative ability and higher anti-tumor efficacy than G207, and is an effective human GBM stem cell killer11. After the confirmation of its safety in a phase I/II study in patients with recurrent GBM, a phase II clinical trial further demonstrated its effectiveness in 2 cases (15.4%) maintaining stable disease12. Therefore, G47Δ was designated as a SAKIGAKE (breakthrough therapy) product and approved for malignant glioma treatment in Japan. Another focus of research is delta-24-arginine-glycine-aspartate (delta-24-RGD or DNX-2401), a second-generation oncolytic adenovirus that has been demonstrated to have anti-glioma activity in preclinical studies13. The completed phase I clinical trial on DNX-2401 has yielded positive outcomes: some patients experienced tumor regression after intratumoral injection of DNX-2401. Biological evaluation of the posttreatment tumor tissues has suggested that the antitumor effect may stem from the direct oncolytic effects of viral infection and the activation of immune-mediated anti-glioma responses14. Although OVs alone have demonstrated potential, their clinical benefits remain limited. Combined treatment with OVs may enable high efficacy. A phase II clinical trial has recently been completed and has yielded encouraging outcomes15. The study evaluated the efficacy of a combination treatment of immune checkpoint inhibitors and OVs in gliomas—a therapy that had been demonstrated to be effective in other tumors—and observed durable responses15,16. On the basis of the results of ongoing and completed clinical trials, OVs have potential for clinical use with favorable safety profiles in glioma treatment. Nevertheless, several challenges must be overcome. Although intratumor injection can achieve maximum delivery to tumors with minimum systematic toxicity, a neurosurgical procedure is inevitable and carries several risks, such as intracranial infection. OH2, a promising OV derived from HSV-2, was designed to decrease toxicity and increase tumor selectivity by deleting the ICP34.5 neurovirulence gene; it has enhanced antitumor immunity through the addition of the human granulocyte macrophage colony-stimulating factor (GM-CSF) gene17. The tolerance to, and antitumor activity of, OH2 have been confirmed in a phase I/II clinical trial in patients with advanced solid tumors18. A clinical trial in gliomas is ongoing (ClinicalTrials.gov identifier [NCT05235074](http://www.cancerbiomed.org/lookup/external-ref?link_type=CLINTRIALGOV&access_num=NCT05235074&atom=%2Fcbm%2F20%2F12%2F947.atom)). In contrast to other OV clinical trials, the OH2 trial used a protocol including an Ommaya reservoir placement surgery, with the goal of decreasing potential risks during subsequent drug administration. This innovative approach has provided remarkable inspiration for OV clinical trials that require intratumoral injection in patients. Cellular vaccines are a viable option for glioma immunotherapy. The dendritic cell vaccine (DCV) is composed of dendritic cells loaded with tumor antigens capable of provoking immune responses. The relative safety and potential efficacy of DC-based vaccinations have been observed in phase I/II clinical trials19. A completed large phase III non-randomized controlled trial in 331 GBM patients has demonstrated the feasibility and potency of tumor lysate-loaded DCV (DCVax-L)20. The study has established the safety profile and demonstrated a survival benefit of the combination therapy, on the basis of an OS surpassing that of patients receiving the standard of care (SOC). Patients receiving the combined therapy exhibited a prolonged median OS (22.3 months after surgery in patients with newly diagnosed GBM and 13.2 months after relapse in patients with recurrent GBM); therefore, DCVax-L has substantial potential in the treatment in GBM. Some studies have focused on small molecular compounds with antitumor potential. One of these compounds is chlorogenic acid (5-caffeoylquinic acid, CGA), a phenolic compound isolated from fruits and plants, such as Eucommia ulmoides or Flos Lonicera. In a study on depression, the observation of CGA’s ability to cross the BBB has led to its application in brain tumor treatment21. Preclinical research has also investigated its anti-tumor mechanism, focusing primarily on its effect on immunoregulation. A study on macrophage polarization has indicated that CGA leads to the conversion of M2 macrophages, which predominate in the tumor microenvironment, into M1 macrophages, which induce tumor dissolution, thereby limiting glioma growth22. These promising results supported a phase I clinical trial evaluating the application of CGA in recurrent GBM. This completed trial has established the safety profile of CGA and its potential OS benefit23. A phase II trial is currently underway to further investigate its efficacy (ClinicalTrials.gov identifier [NCT03758014](http://www.cancerbiomed.org/lookup/external-ref?link_type=CLINTRIALGOV&access_num=NCT03758014&atom=%2Fcbm%2F20%2F12%2F947.atom)). ### Targeted therapy Extensive research has been conducted on targeted therapy, which offers major advantages, such as specificity and selectivity. A compound of interest is procaspase-activating compound-1 (PAC-1), which specifically targets procaspase-3 (PC-3) by chelating inhibitory labile zinc ions. PC-3 is overexpressed in GBM and shows potential oncogenic effects; moreover, its cleavage into caspase-3 plays a critical role in apoptosis. Therefore, researchers are investigating PC-3 activation as a novel anticancer target. A phase I study of PAC-1 for advanced malignancy treatment has established the maximum tolerated dose of PAC-1 and identified its clinical activity, particularly in neuroendocrine tumors24. On the basis of that study and hypothesis-generating preclinical data regarding gliomas, including the ability of PAC-1 to cross the BBB in a mouse model, a phase I study of PAC-1 in combination with TMZ was conducted on patients with HGG. The results have demonstrated that the combination is safe and well tolerated, and thus warrants further investigation25. Small molecular inhibitors targeting cyclin-dependent kinases 4 and 6 (CDK4/6) are also under investigation. CDK4/6 deregulation has been observed in gliomas, and inhibition of CDK4/6 arrests the cell cycle in the G1 phase, thus inhibiting tumor cell proliferation. CDK4/6 inhibitors have shown efficacy in certain tumors, and 3 inhibitors (ribociclib, palbociclib, and abemaciclib) have been approved by the U.S. Food & Drug Administration (FDA). A phase I/II study has found that ribociclib is safe and has promising primary efficacy when used after radiotherapy in children with newly diagnosed diffuse intrinsic pontine glioma26. Some trials have been performed on novel CDK4/6 inhibitors in the early phase (registration number ACTRN12621000479808). Resistance to CDK4/6 inhibitors remains a hurdle to be overcome, and a recent study has suggested that combining these inhibitors with tyrosine kinase inhibitors (TKIs) may yield consistent tumor responses and serve as a potential new strategy27. TKIs are a prevalent research topic, particularly in relation to epidermal growth factor receptor (EGFR). EGFR mutation is a potential marker for glioma treatment and is associated with tumor proliferation and migration. Osimertinib, which has better BBB penetration ability than first- and second-generation EGFR inhibitors, shows potential in anti-GBM therapy28. Bevacizumab targets VEGF, thereby inhibiting angiogenesis. Although a clear survival benefit is lacking, this treatment prolongs PFS, decreases peritumoral edema, and enables lower steroid use. Regorafenib is a multitargeted TKI drug that has shown an encouraging OS benefit in recurrent GBM; the AGILE trial (ClinicalTrials.gov identifier [NCT03970447](http://www.cancerbiomed.org/lookup/external-ref?link_type=CLINTRIALGOV&access_num=NCT03970447&atom=%2Fcbm%2F20%2F12%2F947.atom)) will further investigate the role of this treatment in patients with newly diagnosed GBM29. Another reliable marker for targeted therapy is the V-RAF murine sarcoma viral oncogene homolog B1 (BRAF) mutation. The efficacy of BRAF-targeted therapy, such as dabrafenib, has been established in other cancers30. Despite the relatively low incidence of BRAF mutation in HGG, clinical trials have supported further investigation in patients with HGG with BRAF mutation31. Isocitrate dehydrogenase (IDH) is a common molecular marker in tumors. Inhibitors such as ivosidenib and vorasidenib have exhibited promising efficacy in treating low-grade glioma with mutant IDH32,33. Notably, whereas drugs that target mutant forms of IDH are focused primarily on low-grade glioma, the key clinical trials started before the WHO 2021 classification. Therefore, patients whose diagnoses previously met the eligibility criteria may now carry an updated classification of HGG by histology and molecular analyses. On the basis of the results of these studies, clinical trials for IDH-mutated HGGs are ongoing ([NCT05484622](http://www.cancerbiomed.org/lookup/external-ref?link_type=CLINTRIALGOV&access_num=NCT05484622&atom=%2Fcbm%2F20%2F12%2F947.atom) and CTR20211073). Mutations in the PI3K/AKT/mTOR pathway are common among patients with IDH wild-type GBM34. Nevertheless, the efficacy and safety of mTOR inhibitors, used either alone or in combination with other treatments, have not met expectations, possibly as a result of the complex molecular regulation of this pathway35,36. View this table: [Table 1](http://www.cancerbiomed.org/content/20/12/947/T1) Table 1 Selected drug clinical trials in patients with HGG ## Discussion Before the classification of gliomas based on molecular and genomic features, gliomas were classified into 4 grades based solely on morphological diagnoses. For instance, the diagnosis of GBM included both IDH mutation and wild type. However, diffuse gliomas are heterogeneous tumors, both histologically and molecularly. The grading has changed because of continued improvements in molecular profiling. The WHO 2021 classification restricts the term “glioblastoma” to IDH wild-type tumors, and the changes in the definition may potentially affect outcomes, thus obscuring the true effectiveness of treatments. Assessing and analyzing the heterogeneity of clinical trials is crucial to obtain reliable results. Clinical glioma drug research faces hopes and challenges. Immunotherapy and targeted therapy offer hope to patients with glioma, but substantial barriers must still be overcome, including the challenges of BBB penetration and the need for personalized treatments based on complex molecular phenotypes. Well-designed preclinical studies and clinical trials with standardized processes and reliable data are essential for the successful development of new therapies that will ultimately benefit a broad range of patients. Another important aspect of glioma drug research is exploring the potential of combined and systematic therapies to overcome drug resistance and maximize clinical benefits. With continuing advancements in molecular biology, genomics, and drug development technologies, progress is expected to be achieved for glioma treatment, thus offering patients improved survival through innovative systematic therapies. Although the Stupp regimen has provided survival benefits for patients with HGG, particularly those with recurrent or deep-seated lesions that are difficult to operate on, disease management remains unclear and is influenced by multiple factors. Hence, NCCN guidelines encourage patients with HGG to participate in clinical trials if they are eligible. The increasing number of clinical trials is expected to lead to a wide range of therapeutic options for this lethal disease in the future. ## Conflict of interest statement No potential conflicts of interest are disclosed. ## Author contributions M.H. searched the literature and drafted the manuscript. S.L. and P.L. drafted and proofread the manuscript. Z.K. guided the writing of the paper. B.Z. assisted in writing the manuscript. W.L. was responsible for selecting the topic and critically revising important intellectual content. All authors contributed to the article and approved the submitted version. * Received September 18, 2023. * Accepted November 24, 2023. * Copyright: © 2023, Cancer Biology & Medicine [https://creativecommons.org/licenses/by/4.0/](https://creativecommons.org/licenses/by/4.0/) This is an open access article distributed under the terms of the [Creative Commons Attribution License (CC BY) 4.0](https://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution and reproduction in any medium, provided the original author and source are credited. ## References 1. 1. Tan AC, Ashley DM, López GY, Malinzak M, Friedman HS, Khasraw M. Management of glioblastoma: state of the art and future directions. CA Cancer J Clin. 2020; 70: 299–312. [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fcbm%2F20%2F12%2F947.atom) 2. 2. van Tellingen O, Yetkin-Arik B, de Gooijer M, Wesseling P, Wurdinger T, de Vries HE. Overcoming the blood–brain tumor barrier for effective glioblastoma treatment. Drug Resist Updat. 2015; 19: 1–12. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.drup.2015.02.002&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=25791797&link_type=MED&atom=%2Fcbm%2F20%2F12%2F947.atom) 3. 3. Wick W, Gorlia T, Bendszus M, Taphoorn M, Sahm F, Harting I, et al. Lomustine and bevacizumab in progressive glioblastoma. N Engl J Med. 2017; 377: 1954–63. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1056/NEJMoa1707358&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=29141164&link_type=MED&atom=%2Fcbm%2F20%2F12%2F947.atom) 4. 4. Vredenburgh JJ, Desjardins A, Herndon JE 2nd., Dowell JM, Reardon DA, Quinn JA, et al. Phase II trial of bevacizumab and irinotecan in recurrent malignant glioma. Clin Cancer Res. 2007; 13: 1253–9. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6ImNsaW5jYW5yZXMiO3M6NToicmVzaWQiO3M6OToiMTMvNC8xMjUzIjtzOjQ6ImF0b20iO3M6MTk6Ii9jYm0vMjAvMTIvOTQ3LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 5. 5. de Groot J, Penas-Prado M, Alfaro-Munoz K, Hunter K, Pei BL, O’Brien B, et al. Window-of-opportunity clinical trial of pembrolizumab in patients with recurrent glioblastoma reveals predominance of immune-suppressive macrophages. Neuro Oncol. 2020; 22: 539–49. [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fcbm%2F20%2F12%2F947.atom) 6. 6. Tawbi HA, Forsyth PA, Algazi A, Hamid O, Hodi FS, Moschos SJ, et al. Combined nivolumab and ipilimumab in melanoma metastatic to the brain. N Engl J Med. 2018; 379: 722–30. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1056/NEJMoa1805453&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fcbm%2F20%2F12%2F947.atom) 7. 7. Duerinck J, Schwarze JK, Awada G, Tijtgat J, Vaeyens F, Bertels C, et al. Intracerebral administration of CTLA-4 and PD-1 immune checkpoint blocking monoclonal antibodies in patients with recurrent glioblastoma: a phase I clinical trial. J Immunother Cancer. 2021; 9: e002296. 8. 8. Friedman GK, Johnston JM, Bag AK, Bernstock JD, Li R, Aban I, et al. Oncolytic HSV-1 G207 immunovirotherapy for pediatric high-grade gliomas. N Engl J Med. 2021; 384: 1613–22. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1056/NEJMoa2024947&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fcbm%2F20%2F12%2F947.atom) 9. 9. Mineta T, Rabkin SD, Yazaki T, Hunter WD, Martuza RL. Attenuated multi-mutated herpes simplex virus-1 for the treatment of malignant gliomas. Nat Med. 1995; 1: 938–43. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1038/nm0995-938&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=7585221&link_type=MED&atom=%2Fcbm%2F20%2F12%2F947.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=A1995RT30300049&link_type=ISI) 10. 10. Andtbacka RH, Kaufman HL, Collichio F, Amatruda T, Senzer N, Chesney J, et al. Talimogene laherparepvec improves durable response rate in patients with advanced melanoma. J Clin Oncol. 2015; 33: 2780–8. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamNvIjtzOjU6InJlc2lkIjtzOjEwOiIzMy8yNS8yNzgwIjtzOjQ6ImF0b20iO3M6MTk6Ii9jYm0vMjAvMTIvOTQ3LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 11. 11. Cheema TA, Wakimoto H, Fecci PE, Ning J, Kuroda T, Jeyaretna DS, et al. Multifaceted oncolytic virus therapy for glioblastoma in an immunocompetent cancer stem cell model. Proc Natl Acad Sci U S A. 2013; 110: 12006–11. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czoxMjoiMTEwLzI5LzEyMDA2IjtzOjQ6ImF0b20iO3M6MTk6Ii9jYm0vMjAvMTIvOTQ3LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 12. 12. Todo T, Ino Y, Ohtsu H, Shibahara J, Tanaka M. A phase I/II study of triple-mutated oncolytic herpes virus G47∆ in patients with progressive glioblastoma. Nat Commun. 2022; 13: 4119. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1038/s41467-022-31262-y&link_type=DOI) 13. 13. Fueyo J, Alemany R, Gomez-Manzano C, Fuller GN, Khan A, Conrad CA, et al. Preclinical characterization of the antiglioma activity of a tropism-enhanced adenovirus targeted to the retinoblastoma pathway. J Natl Cancer Inst. 2003; 95: 652–60. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1093/jnci/95.9.652&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=12734316&link_type=MED&atom=%2Fcbm%2F20%2F12%2F947.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000182672800008&link_type=ISI) 14. 14. Lang FF, Conrad C, Gomez-Manzano C, Yung WKA, Sawaya R, Weinberg JS, et al. Phase I study of DNX-2401 (Delta-24-RGD) oncolytic adenovirus: replication and immunotherapeutic effects in recurrent malignant glioma. J Clin Oncol. 2018; 36: 1419–27. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1200/JCO.2017.75.8219&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fcbm%2F20%2F12%2F947.atom) 15. 15. Nassiri F, Patil V, Yefet LS, Singh O, Liu J, Dang RMA, et al. Oncolytic DNX-2401 virotherapy plus pembrolizumab in recurrent gliobla stoma: a phase 1/2 trial. Nat Med. 2023; 29: 1370–8. 16. 16. Ribas A, Dummer R, Puzanov I, VanderWalde A, Andtbacka RHI, Michielin O, et al. Oncolytic virotherapy promotes intratumoral T cell infiltration and improves anti-PD-1 immunotherapy. Cell. 2017; 170: 1109–19.e10. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.cell.2017.08.027&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fcbm%2F20%2F12%2F947.atom) 17. 17. Zhao Q, Zhang W, Ning Z, Zhuang X, Lu H, Liang J, et al. A novel oncolytic herpes simplex virus type 2 has potent anti-tumor activity. PLoS One. 2014; 9: e93103. 18. 18. Zhang B, Huang J, Tang J, Hu S, Luo S, Luo Z, et al. Intratumoral OH2, an oncolytic herpes simplex virus 2, in patients with advanced solid tumors: a multicenter, phase I/II clinical trial. J Immunother Cancer. 2021; 9: e002224. 19. 19. De Vleeschouwer S, Fieuws S, Rutkowski S, van Calenbergh F, van Loon J, Goffin J, et al. Postoperative adjuvant dendritic cell-based immunotherapy in patients with relapsed glioblastoma multiforme. Clin Cancer Res. 2008; 14: 3098–104. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6ImNsaW5jYW5yZXMiO3M6NToicmVzaWQiO3M6MTA6IjE0LzEwLzMwOTgiO3M6NDoiYXRvbSI7czoxOToiL2NibS8yMC8xMi85NDcuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 20. 20. Liau LM, Ashkan K, Brem S, Campian JL, Trusheim JE, Iwamoto FM, et al. Association of autologous tumor lysate-loaded dendritic cell vaccination with extension of survival among patients with newly diagnosed and recurrent glioblastoma: a phase 3 prospective externally controlled cohort trial. JAMA Oncol. 2023; 9: 112–21. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1001/jamaoncol.2022.5370&link_type=DOI) 21. 21. Wu J, Chen H, Li H, Tang Y, Yang L, Cao S, et al. Antidepressant potential of chlorogenic acid-enriched extract from eucommia ulmoides oliver bark with neuron protection and promotion of serotonin release through enhancing synapsin I expression. Molecules. 2016; 21: 260. 22. 22. Xue N, Zhou Q, Ji M, Jin J, Lai F, Chen J, et al. Chlorogenic acid inhibits glioblastoma growth through repolarizating macrophage from M2 to M1 phenotype. Sci Rep. 2017; 7: 39011. 23. 23. Kang Z, Li S, Kang X, Deng J, Yang H, Chen F, et al. Phase I study of chlorogenic acid injection for recurrent high-grade glioma with long-term follow-up. Cancer Biol Med. 2023; 20: 465–76. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiY2JtIjtzOjU6InJlc2lkIjtzOjg6IjIwLzYvNDY1IjtzOjQ6ImF0b20iO3M6MTk6Ii9jYm0vMjAvMTIvOTQ3LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 24. 24. Danciu OC, Holdhoff M, Peterson RA, Fischer JH, Liu LC, Wang H, et al. Phase I study of procaspase-activating compound-1 (PAC-1) in the treatment of advanced malignancies. Br J Cancer. 2023; 128: 783–92. 25. 25. Holdhoff M, Nicholas MK, Peterson RA, Maraka S, Liu LC, Fischer JH, et al. Phase I dose-escalation study of procaspase-activating compound-1 in combination with temozolomide in patients with recurrent high-grade astrocytomas. Neurooncol Adv. 2023; 5: vdad087. 26. 26. DeWire M, Fuller C, Hummel TR, Chow LML, Salloum R, de Blank P, et al. A phase I/II study of ribociclib following radiation therapy in children with newly diagnosed diffuse intrinsic pontine glioma (DIPG). J Neuro Oncol. 2020; 149: 511–22. 27. 27. Olmez I, Zhang Y, Manigat L, Benamar M, Brenneman B, Nakano I, et al. Combined c-Met/Trk inhibition overcomes resistance to CDK4/6 inhibitors in glioblastoma. Cancer Res. 2018; 78: 4360–9. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiY2FucmVzIjtzOjU6InJlc2lkIjtzOjEwOiI3OC8xNS80MzYwIjtzOjQ6ImF0b20iO3M6MTk6Ii9jYm0vMjAvMTIvOTQ3LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 28. 28. Liu X, Chen X, Shi L, Shan Q, Cao Q, Yue C, et al. The third-generation EGFR inhibitor AZD9291 overcomes primary resistance by continuously blocking ERK signaling in glioblastoma. J Exp Clin Cancer Res. 2019; 38: 219. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1186/s13046-019-1235-7&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=31122294&link_type=MED&atom=%2Fcbm%2F20%2F12%2F947.atom) 29. 29. Lombardi G, De Salvo GL, Brandes AA, Eoli M, Rudà R, Faedi M, et al. Regorafenib compared with lomustine in patients with relapsed glioblastoma (REGOMA): a multicentre, open-label, randomised, controlled, phase 2 trial. Lancet Oncol. 2019; 20: 110–9. 30. 30. Hauschild A, Grob J-J, Demidov LV, Jouary T, Gutzmer R, Millward M, et al. Dabrafenib in BRAF-mutated metastatic melanoma: a multicentre, open-label, phase 3 randomised controlled trial. Lancet. 2012; 380: 358–65. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/S0140-6736(12)60868-X&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=22735384&link_type=MED&atom=%2Fcbm%2F20%2F12%2F947.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000306842800033&link_type=ISI) 31. 31. Wen PY, Stein A, van den Bent M, De Greve J, Wick A, de Vos F, et al. Dabrafenib plus trametinib in patients with BRAF(V600E)-mutant low-grade and high-grade glioma (ROAR): a multicentre, open-label, single-arm, phase 2, basket trial. Lancet Oncol. 2022; 23: 53–64. 32. 32. Mellinghoff IK, Lu M, Wen PY, Taylor JW, Maher EA, Arrillaga-Romany I, et al. Vorasidenib and ivosidenib in IDH1-mutant low-grade glioma: a randomized, perioperative phase 1 trial. Nat Med. 2023; 29: 615–22. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1038/s41591-022-02141-2&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=36823302&link_type=MED&atom=%2Fcbm%2F20%2F12%2F947.atom) 33. 33. Mellinghoff IK, van den Bent MJ, Blumenthal DT, Touat M, Peters KB, Clarke J, et al. Vorasidenib in IDH1- or IDH2-mutant low-grade glioma. N Engl J Med. 2023; 389: 589–601. 34. 34. Brennan CW, Verhaak RG, McKenna A, Campos B, Noushmehr H, Salama SR, et al. The somatic genomic landscape of glioblastoma. Cell. 2013; 155: 462–77. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.cell.2013.09.034&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=24120142&link_type=MED&atom=%2Fcbm%2F20%2F12%2F947.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000325719800021&link_type=ISI) 35. 35. Wen PY, Touat M, Alexander BM, Mellinghoff IK, Ramkissoon S, McCluskey CS, et al. Buparlisib in patients with recurrent glioblastoma harboring phosphatidylinositol 3-kinase pathway activation: an open-label, multicenter, multi-arm, phase II trial. J Clin Oncol. 2019; 37: 741–50. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1200/JCO.18.01207&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fcbm%2F20%2F12%2F947.atom) 36. 36. Rosenthal M, Clement PM, Campone M, Gil-Gil MJ, DeGroot J, Chinot O, et al. Buparlisib plus carboplatin or lomustine in patients with recurrent glioblastoma: a phase Ib/II, open-label, multicentre, randomised study. ESMO open. 2020; 5: e000672.