1
|
Xing P, Pu X, Zhou Y, Liu Z, Yao Y, Liu J, Wang S, Hu Y, Li J, Wu L. Efficacy and safety of dacomitinib in treatment-naïve patients with advanced NSCLC and brain metastasis: a multicenter cohort study. Oncologist 2025; 30:oyaf079. [PMID: 40349136 PMCID: PMC12065940 DOI: 10.1093/oncolo/oyaf079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 02/26/2024] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND The data for dacomitinib, a second-generation EGFR-TKI, treating patients with advanced non-small cell lung cancer (NSCLC) and brain metastasis was lacking. This study aimed to explore the efficacy and safety of dacomitinib in treating EGFR-mutated advanced NSCLC with brain metastasis in first-line settings. METHODS Eligible patients were treatment-naïve advanced NSCLC patients with ≥1 brain metastasis no less than 5 mm treated with dacomitinib. The primary endpoint was intracranial objective response rate (ORR). Secondary endpoints included intracranial and extracranial progression-free survival (PFS), overall survival (OS), intracranial and extracranial ORR, disease control rate (DCR), and safety. The response was evaluated per modified Response Evaluation Criteria in Solid Tumors (mRECIST) and RANO-BM (Response Assessment in Neuro-Oncology Brain Metastases) criteria. RESULT Between July 2nd, 2019, and September 30th, 2022, a total of 87 treatment-naïve patients with advanced NSCLC and brain metastasis treated with dacomitinib from four hospitals were included. The data cutoff date was March 24th, 2023, and the median duration of follow-up time was 17.5 months (range 1.6-34.7 months). Based on mRECIST criteria, for all the 87 patients with evaluable brain metastasis, the iORR was 89.7% (95%CI, 81.3%-95.2%) and iDCR was 97.7% (95%CI, 91.9-99.7%), with 42 patients achieving CR, 36 patients achieving PR, and 7 patients maintaining SD. Based on RANO-BM criteria, the iORR was 71.3% (62/87, 95%CI 60.6%-80.5%) and iDCR was 97.7% (85/87, 95%CI, 91.9%-99.7%), with 42 patients achieving CR, 20 patients achieving PR, and 23 patients maintaining SD (Table). Median iPFS was 26.0 (95%CI, 20.7-31.4) months, and the 1-year and 2-year iPFS rate were 68.9% and 51.5%, respectively. Of 75 patients with evaluable extracranial lesions, 2 patients achieved CR (2.7%), the systemic ORR was 73.8% (95%CI 63.1%-82.8%) and DCR was 96.4% (89.9%-99.3%) (Table). Systemic median PFS was 14.0 (95%CI 11.1-16.9) months and median OS was 34.0 (95%CI 28.0-39.9) months. Overall, 86 of 87 (98.9%) patients experienced adverse events (AEs) of any grade. The most common (≥20%) AEs including rash (89.7%), oral ulcer (74.2%), diarrhea (67.8%), and paronychia (59.8%). Most of the AEs were grade 1 or grade 2 and no patients died due to severe AEs. CONCLUSIONS Dacomitinib showed promising efficacy and a manageable safety profile for advanced NSCLC with brain metastasis harboring EGFR mutation in the first-line treatment.
Collapse
Affiliation(s)
- Puyuan Xing
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, People’s Republic of China
| | - Xingxiang Pu
- The Second Department of Thoracic Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, 410013,People’s Republic of China
| | - Yu Zhou
- The Second Department of Thoracic Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, 410013,People’s Republic of China
| | - Ziling Liu
- Department of Oncology, the First Hospital of Jilin University, Changchun, 130031, People’s Republic of China
| | - Yu Yao
- Department of Oncology, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061, People’s Republic of China
| | - Jiayu Liu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, People’s Republic of China
| | - Shouzheng Wang
- The Department of Oncology, Beijing Chest Hospital, Beijing, 101149, People’s Republic of China
| | - Ying Hu
- The Department of Oncology, Beijing Chest Hospital, Beijing, 101149, People’s Republic of China
| | - Junling Li
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, People’s Republic of China
| | - Lin Wu
- The Second Department of Thoracic Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, 410013,People’s Republic of China
| |
Collapse
|
2
|
Mahdi A, Aittaleb M, Tissir F. Targeting Glioma Stem Cells: Therapeutic Opportunities and Challenges. Cells 2025; 14:675. [PMID: 40358199 PMCID: PMC12072158 DOI: 10.3390/cells14090675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2025] [Revised: 04/25/2025] [Accepted: 05/03/2025] [Indexed: 05/15/2025] Open
Abstract
Glioblastoma (GBM), or grade 4 glioma, is the most common and aggressive primary brain tumor in adults with a median survival of 15 months. Increasing evidence suggests that GBM's aggressiveness, invasiveness, and therapy resistance are driven by glioma stem cells (GSCs), a subpopulation of tumor cells that share molecular and functional characteristics with neural stem cells (NSCs). GSCs are heterogeneous and highly plastic. They evade conventional treatments by shifting their state and entering in quiescence, where they become metabolically inactive and resistant to radiotherapy and chemotherapy. GSCs can exit quiescence and be reactivated to divide into highly proliferative tumor cells which contributes to recurrence. Understanding the molecular mechanisms regulating the biology of GSCs, their plasticity, and the switch between quiescence and mitotic activity is essential to shape new therapeutic strategies. This review examines the latest evidence on GSC biology, their role in glioblastoma progression and recurrence, emerging therapeutic approaches aimed at disrupting their proliferation and survival, and the mechanisms underlying their resistance to therapy.
Collapse
Affiliation(s)
| | | | - Fadel Tissir
- College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Education City, Doha P.O. Box 5825, Qatar; (A.M.); (M.A.)
| |
Collapse
|
3
|
Zhu F, Qiu J, Ye H, Su W, Wang R, Fu Y. The Prognostic Significance of Epidermal Growth Factor Receptor Amplification and Epidermal Growth Factor Receptor Variant III Mutation in Glioblastoma: A Systematic Review and Meta-Analysis with Implications for Targeted Therapy. Int J Mol Sci 2025; 26:3539. [PMID: 40331985 PMCID: PMC12027172 DOI: 10.3390/ijms26083539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 04/01/2025] [Accepted: 04/07/2025] [Indexed: 05/08/2025] Open
Abstract
Glioblastoma (GBM) is the most aggressive and heterogeneous neoplasm among central nervous system tumors, with a dismal prognosis and a high recurrence rate. Among the various genetic alterations found in GBM, the amplification of epidermal growth factor receptor (EGFR) and the EGFR variant III (EGFRvIII) mutation are among the most common, though their prognostic value remains controversial. This systematic review and meta-analysis aimed to provide a comprehensive evaluation of the diagnostic and prognostic significance of EGFR amplification and the EGFRvIII mutation in GBM patients, incorporating recent studies published in the past few years to offer a more complete and up-to-date analysis. An extensive search of the PubMed, Web of Science, and Scopus databases was conducted, including studies that reported on EGFR and/or the EGFRvIII mutation status with detailed survival data. A total of 32 studies with 4208 GBM patients were included. The results indicated that EGFR amplification significantly correlated with worse OS (Overall survival) (HR = 1.27, 95% CI: 1.03-1.57), suggesting that EGFR amplification is an independent prognostic marker. The prognostic value of EGFRvIII was inconclusive, with a pooled hazard ratio for overall survival of 1.13 (95% CI: 0.94-1.36), indicating no significant effect on survival in the general population. However, a subgroup analysis suggested that EGFRvIII may be associated with poorer outcomes, particularly in recurrent GBM patients, where its prognostic significance became more evident. Furthermore, subgroup analyses based on geographic region revealed significant heterogeneity in the prognostic impact of EGFR amplification across different populations. In American cohorts, EGFR amplification was strongly associated with an increased risk of mortality (HR = 1.53, 95% CI: 1.28-1.84, p = 0.001), suggesting that it serves as a more reliable prognostic marker in this region. In contrast, no significant prognostic impact of EGFR amplification was observed in Asian (HR = 0.64, 95% CI: 0.35-1.17) or European (HR = 0.98, 95% CI: 0.80-1.19) populations. Overall, this study underscores the potential of EGFR amplification as a prognostic marker in GBM, while further research is needed to fully elucidate the role of the EGFRvIII mutation, particularly in specific patient subgroups. Clarifying these associations could offer important insights for targeted treatment strategies, improving patient outcomes.
Collapse
Affiliation(s)
| | | | | | | | - Renxi Wang
- Laboratory of Brain Disorders, Beijing Institute of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, No. 10 Xitoutiao, Beijing 100069, China; (F.Z.); (J.Q.); (H.Y.); (W.S.)
| | - Yi Fu
- Laboratory of Brain Disorders, Beijing Institute of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, No. 10 Xitoutiao, Beijing 100069, China; (F.Z.); (J.Q.); (H.Y.); (W.S.)
| |
Collapse
|
4
|
Singh G, Rohit, Kumar P, Aran KR. Targeting EGFR and PI3K/mTOR pathways in glioblastoma: innovative therapeutic approaches. Med Oncol 2025; 42:97. [PMID: 40064710 DOI: 10.1007/s12032-025-02652-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 02/24/2025] [Indexed: 03/29/2025]
Abstract
Glioblastoma (GBM) stands as the most aggressive form of primary brain cancer in adults, characterized by its rapid growth, invasive nature, and a robust propensity to induce angiogenesis, forming new blood vessels to sustain its expansion. GBM arises from astrocytes, star-shaped glial cells, and despite significant progress in understanding its molecular mechanisms, its prognosis remains grim. It is frequently associated with mutations or overexpression of the epidermal growth factor receptor (EGFR), which initiates several downstream signaling pathways. Dysregulation of key signaling pathways, such as EGFR/PTEN/AKT/mTOR, drives tumorigenesis, promotes metastasis and leads to treatment resistance. The modest survival benefits of the conventional treatment of surgical resection followed by radiation and chemotherapy underscore the pressing need for innovative therapeutic approaches. In most the tumor, overexpression of EGFR is found associated with GBM and mutations in its several variants are important for promoting ongoing mitogenic signaling and tumor growth. This receptor inhibits apoptosis and promotes cell survival and proliferation by activating downstream PI3K/AKT/mTOR pathways. This route is typically blocked by PTEN, a crucial tumor suppressor, however, GBM frequently results in abnormalities in this protein. The aim of this review is to explore the molecular foundations of GBM, with a focus on the EGFR and PI3K/mTOR pathways and their impact on tumor behavior. Additionally, this review highlights EGFR and PI3K/AKT/mTOR inhibitors currently in clinical and preclinical trials, addressing treatment resistance, challenges, and future directions.
Collapse
Affiliation(s)
- Gursimran Singh
- Department of Pharmacy Practice, ISF College of Pharmacy (an Autonomous College), Moga, Punjab, 142001, India
| | - Rohit
- Research Scholar, I.K. Gujral Punjab Technical University, Kapurthala, Punjab, 144603, India
| | - Pankaj Kumar
- Department of Pharmacology, Himachal Institute of Pharmaceutical Education and Research (HIPER), Tehsil-Nadaun, Hamirpur, Himachal Pradesh, 177033, India
| | - Khadga Raj Aran
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy (an Autonomous College), Moga, Punjab, 142001, India.
| |
Collapse
|
5
|
Wang J, Luo J, Yang S, Deng Y, Chen P, Tan Y, Liu Y. Development and validation of disulfidptosis-related genes signature for patients with glioma. Discov Oncol 2024; 15:758. [PMID: 39692962 DOI: 10.1007/s12672-024-01664-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 12/03/2024] [Indexed: 12/19/2024] Open
Abstract
BACKGROUND Disulfidptosis has recently emerged as a novel form of regulated cell death (RCD). Evasion of cell death is a hallmark of cancer, and the resistance of many tumors to apoptosis-inducing therapies has heightened interest in exploring alternative RCD mechanisms. METHODS Transcriptomic and clinical data were obtained from The Cancer Genome Atlas (TCGA), Genotype-Tissue Expression (GTEx), and Chinese Glioma Genome Atlas (CGGA). Glioma samples were classified using non-negative matrix factorization (NMF). A predictive model was constructed using Lasso regression analysis, and its performance was evaluated through receiver operating characteristic (ROC) and Kaplan-Meier survival analyses. The relationship between the model and the tumor immune microenvironment (TIME) as well as treatment sensitivity was also assessed. Finally, we validated the expression of key signature genes in glioma. RESULTS Glioma samples were categorized into two distinct subtypes based on disulfidptosis-related genes, showing significant differences in overall survival (OS) and progression-free survival (PFS) between the subtypes. A genetic risk score model was then developed using these genes. A nomogram predicting OS was constructed using the risk score and clinical variables. Patients were stratified into low- and high-risk groups based on the median risk score from the TCGA cohort. Low-risk patients had significantly better outcomes compared to high-risk patients (TCGA cohort, OS: p < 0.001; PFS: p < 0.001; CGGA cohort, OS: p < 0.001). The risk score was associated with HLA expression, immune checkpoint genes, immune cell infiltration, immune function, tumor mutation burden, tumor stemness score, and drug sensitivity. Lastly, the expression of 11 signature genes was confirmed in glioma tissues. CONCLUSIONS The disulfidptosis-related gene-based risk score model effectively predicted glioma outcomes and highlighted the role of disulfidptosis-related genes in tumor immunity. This study offers potential new avenues for glioma treatment by targeting disulfidptosis.
Collapse
Affiliation(s)
- Jia Wang
- Department of Neurosurgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing, China
| | - Junchi Luo
- Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Sha Yang
- Guizhou University Medical College, Guiyang, 550025, Guizhou Province, China
| | - Yongbing Deng
- Department of Neurosurgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing, China
| | - Peng Chen
- Department of Neurosurgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing, China
| | - Ying Tan
- Zunyi Medical University, Zunyi, Guizhou Province, China
- Department of Neurosurgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Yang Liu
- Department of Neurosurgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing, China.
| |
Collapse
|
6
|
Sharma A, Raut SS, Shukla A, Gupta S, Singh A, Mishra A. DDX3X dynamics, glioblastoma's genetic landscape, therapeutic advances, and autophagic interplay. Med Oncol 2024; 41:258. [PMID: 39368002 DOI: 10.1007/s12032-024-02525-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 09/23/2024] [Indexed: 10/07/2024]
Abstract
Glioblastoma is one of the most aggressive and deadly forms of cancer, posing significant challenges for the medical community. This review focuses on key aspects of Glioblastoma, including its genetic differences between primary and secondary types. Temozolomide is a major first-line treatment for Glioblastoma, and this article explores its development, how it works, and the issue of resistance that limits its effectiveness, prompting the need for new treatment strategies. Gene expression profiling has greatly advanced cancer research by revealing the molecular mechanisms of tumors, which is essential for creating targeted therapies for Glioblastoma. One important protein in this context is DDX3X, which plays various roles in cancer, sometimes promoting it or otherwise suppressing it. Additionally, autophagy, a process that maintains cellular balance, has complex implications in cancer treatment. Understanding autophagy helps to identify resistance mechanisms and potential treatments, with Chloroquine showing promise in treating Glioblastoma. This review covers the interplay between Glioblastoma, DDX3X, and autophagy, highlighting the challenges and potential strategies in treating this severe disease.
Collapse
Affiliation(s)
- Arpit Sharma
- Biomolecular Engineering Laboratory, School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi, 221005, India
| | - Shruti S Raut
- Biomolecular Engineering Laboratory, School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi, 221005, India
| | - Alok Shukla
- Biomolecular Engineering Laboratory, School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi, 221005, India
| | - Shivani Gupta
- Biomolecular Engineering Laboratory, School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi, 221005, India
| | - Amit Singh
- Department of Pharmacology, IMS-Banaras Hindu University, Varanasi, 221005, India.
| | - Abha Mishra
- Biomolecular Engineering Laboratory, School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi, 221005, India.
| |
Collapse
|
7
|
Feng F, Shen J, Qi Q, Zhang Y, Ni S. Empowering brain tumor management: chimeric antigen receptor macrophage therapy. Theranostics 2024; 14:5725-5742. [PMID: 39310093 PMCID: PMC11413779 DOI: 10.7150/thno.98290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 08/17/2024] [Indexed: 09/25/2024] Open
Abstract
Brain tumors pose formidable challenges in oncology due to the intricate biology and the scarcity of effective treatment modalities. The emergence of immunotherapy has opened new avenues for innovative therapeutic strategies. Chimeric antigen receptor, originally investigated in T cell-based therapy, has now expanded to encompass macrophages, presenting a compelling avenue for augmenting anti-tumor immune surveillance. This emerging frontier holds promise for advancing the repertoire of therapeutic options against brain tumors, offering potential breakthroughs in combating the formidable malignancies of the central nervous system. Tumor-associated macrophages constitute a substantial portion, ranging from 30% to 50%, of the tumor tissue and exhibit tumor-promoting phenotypes within the immune-compromised microenvironment. Constructing CAR-macrophages can effectively repolarize M2-type macrophages towards an M1-type phenotype, thereby eliciting potent anti-tumor effects. CAR-macrophages can recruit T cells to the brain tumor site, thereby orchestrating a remodeling of the immune niche to effectively inhibit tumor growth. In this review, we explore the potential limitations as well as strategies for optimizing CAR-M therapy, offering insights into the future direction of this innovative therapeutic approach.
Collapse
Affiliation(s)
| | | | - Qichao Qi
- Department of Neurosurgery, Qilu Hospital and Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, 107 Wenhua Xi Road, Jinan, 250012, Shandong, China
| | - Yulin Zhang
- Department of Neurosurgery, Qilu Hospital and Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, 107 Wenhua Xi Road, Jinan, 250012, Shandong, China
| | - Shilei Ni
- Department of Neurosurgery, Qilu Hospital and Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, 107 Wenhua Xi Road, Jinan, 250012, Shandong, China
| |
Collapse
|
8
|
Gettinger SN, Song Z, Reckamp KL, Moscow JA, Gray RJ, Wang V, McShane LM, Rubinstein LV, Patton DR, Williams PM, Hamilton SR, Kong XT, Tricoli JV, Conley BA, Arteaga CL, Harris LN, O'Dwyer PJ, Chen AP, Flaherty KT. Phase II Trial of Afatinib in Patients With EGFR-Mutated Solid Tumors Excluding Lung Cancer: Results From NCI-MATCH ECOG-ACRIN Trial (EAY131) Subprotocol A. JCO Precis Oncol 2024; 8:e2300725. [PMID: 38986051 DOI: 10.1200/po.23.00725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 03/07/2024] [Accepted: 05/15/2024] [Indexed: 07/12/2024] Open
Abstract
PURPOSE National Cancer Institute-Molecular Analysis for Therapy Choice (NCI-MATCH) was a multicohort phase 2 trial that assigned patients with advanced pretreated cancers to molecularly targeted therapies on the basis of tumor genomic testing. NCI-MATCH Arm A evaluated afatinib, an EGFR tyrosine kinase inhibitor (TKI) approved for advanced non-small cell lung cancer, in patients with tumors other than lung cancer harboring EGFR mutations. METHODS Patients with advanced pretreated cancers other than lung cancer found to have selected actionable EGFR mutations were offered participation in Arm A. Previous therapy with an EGFR TKI was not allowed. Patients received afatinib 40 mg once daily continuously until disease progression or unacceptable toxicity. The primary end point was objective response rate (ORR). Secondary end points included progression-free survival (PFS), 6-month PFS, and overall survival (OS). RESULTS Seventeen patients received protocol therapy. Tumor types included glioblastoma multiforme (GBM) (13), gliosarcoma (1), adenocarcinoma not otherwise specified (NOS) (2), and adenosquamous carcinoma of the breast (1). Fifty-nine percent of patients received ≥2 lines of previous therapy. The ORR was 11.8% (90% CI, 2.1 to 32.6), with one complete response lasting 16.4 months (GBM harboring a rare exon 18 EGFR-SEPT14 fusion) and one partial response lasting 12.8 months (adenocarcinoma NOS with the classic EGFR mutation, p.Glu746_Ala750del). Three patients had stable disease. The 6-month PFS was 15% (90% CI, 0 to 30.7); the median OS was 9 months (90% CI, 4.6 to 14.0). Rash and diarrhea were the most common toxicities. CONCLUSION Afatinib had modest activity in a cohort of patients with heavily pretreated cancer with advanced nonlung, EGFR-mutated tumors, but the trial's primary end point was not met. Further evaluation of afatinib in GBM with EGFR exon 18 fusions may be of interest.
Collapse
Affiliation(s)
| | - Zihe Song
- Dana Farber Cancer Institute-ECOG-ACRIN Biostatistics Center, Boston, MA
| | | | - Jeffrey A Moscow
- Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Robert J Gray
- Dana Farber Cancer Institute-ECOG-ACRIN Biostatistics Center, Boston, MA
| | - Victoria Wang
- Dana Farber Cancer Institute-ECOG-ACRIN Biostatistics Center, Boston, MA
| | - Lisa M McShane
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Larry V Rubinstein
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - David R Patton
- Center for Biomedical Informatics & Information Technology, National Cancer Institute, Bethesda, MD
| | | | | | - Xiao-Tang Kong
- UC Irvine Health/Chao Family Comprehensive Cancer Center, Orange, CA
| | - James V Tricoli
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Barbara A Conley
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | | | - Lyndsay N Harris
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | | | - Alice P Chen
- Developmental Therapeutics Clinic, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | | |
Collapse
|
9
|
Pridham KJ, Hutchings KR, Beck P, Liu M, Xu E, Saechin E, Bui V, Patel C, Solis J, Huang L, Tegge A, Kelly DF, Sheng Z. Selective regulation of chemosensitivity in glioblastoma by phosphatidylinositol 3-kinase beta. iScience 2024; 27:109921. [PMID: 38812542 PMCID: PMC11133927 DOI: 10.1016/j.isci.2024.109921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/09/2024] [Accepted: 05/03/2024] [Indexed: 05/31/2024] Open
Abstract
Resistance to chemotherapies such as temozolomide is a major hurdle to effectively treat therapy-resistant glioblastoma. This challenge arises from the activation of phosphatidylinositol 3-kinase (PI3K), which makes it an appealing therapeutic target. However, non-selectively blocking PI3K kinases PI3Kα/β/δ/γ has yielded undesired clinical outcomes. It is, therefore, imperative to investigate individual kinases in glioblastoma's chemosensitivity. Here, we report that PI3K kinases were unequally expressed in glioblastoma, with levels of PI3Kβ being the highest. Patients deficient of O6-methylguanine-DNA-methyltransferase (MGMT) and expressing elevated levels of PI3Kβ, defined as MGMT-deficient/PI3Kβ-high, were less responsive to temozolomide and experienced poor prognosis. Consistently, MGMT-deficient/PI3Kβ-high glioblastoma cells were resistant to temozolomide. Perturbation of PI3Kβ, but not other kinases, sensitized MGMT-deficient/PI3Kβ-high glioblastoma cells or tumors to temozolomide. Moreover, PI3Kβ-selective inhibitors and temozolomide synergistically mitigated the growth of glioblastoma stem cells. Our results have demonstrated an essential role of PI3Kβ in chemoresistance, making PI3Kβ-selective blockade an effective chemosensitizer for glioblastoma.
Collapse
Affiliation(s)
- Kevin J. Pridham
- Fralin Biomedical Research Institute at VTC, Roanoke, VA 24016, USA
| | - Kasen R. Hutchings
- Fralin Biomedical Research Institute at VTC, Roanoke, VA 24016, USA
- Department of Internal Medicine, Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA
| | - Patrick Beck
- Fralin Biomedical Research Institute at VTC, Roanoke, VA 24016, USA
- Department of Internal Medicine, Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA
| | - Min Liu
- Fralin Biomedical Research Institute at VTC, Roanoke, VA 24016, USA
| | - Eileen Xu
- Fralin Biomedical Research Institute at VTC, Roanoke, VA 24016, USA
- Department of Internal Medicine, Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA
| | - Erin Saechin
- Fralin Biomedical Research Institute at VTC, Roanoke, VA 24016, USA
- Department of Internal Medicine, Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA
| | - Vincent Bui
- Fralin Biomedical Research Institute at VTC, Roanoke, VA 24016, USA
| | - Chinkal Patel
- Fralin Biomedical Research Institute at VTC, Roanoke, VA 24016, USA
| | - Jamie Solis
- Fralin Biomedical Research Institute at VTC, Roanoke, VA 24016, USA
| | - Leah Huang
- Fralin Biomedical Research Institute at VTC, Roanoke, VA 24016, USA
| | - Allison Tegge
- Fralin Biomedical Research Institute at VTC, Roanoke, VA 24016, USA
| | - Deborah F. Kelly
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA 16802, USA
- Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802, USA
- Center for Structural Oncology, Pennsylvania State University, University Park, PA 16802, USA
| | - Zhi Sheng
- Fralin Biomedical Research Institute at VTC, Roanoke, VA 24016, USA
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA 16802, USA
- Faculty of Health Science, Virginia Tech, Blacksburg, VA 24061, USA
| |
Collapse
|
10
|
Sadowski K, Jażdżewska A, Kozłowski J, Zacny A, Lorenc T, Olejarz W. Revolutionizing Glioblastoma Treatment: A Comprehensive Overview of Modern Therapeutic Approaches. Int J Mol Sci 2024; 25:5774. [PMID: 38891962 PMCID: PMC11172387 DOI: 10.3390/ijms25115774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 05/22/2024] [Accepted: 05/25/2024] [Indexed: 06/21/2024] Open
Abstract
Glioblastoma is the most common malignant primary brain tumor in the adult population, with an average survival of 12.1 to 14.6 months. The standard treatment, combining surgery, radiotherapy, and chemotherapy, is not as efficient as we would like. However, the current possibilities are no longer limited to the standard therapies due to rapid advancements in biotechnology. New methods enable a more precise approach by targeting individual cells and antigens to overcome cancer. For the treatment of glioblastoma, these are gamma knife therapy, proton beam therapy, tumor-treating fields, EGFR and VEGF inhibitors, multiple RTKs inhibitors, and PI3K pathway inhibitors. In addition, the increasing understanding of the role of the immune system in tumorigenesis and the ability to identify tumor-specific antigens helped to develop immunotherapies targeting GBM and immune cells, including CAR-T, CAR-NK cells, dendritic cells, and immune checkpoint inhibitors. Each of the described methods has its advantages and disadvantages and faces problems, such as the inefficient crossing of the blood-brain barrier, various neurological and systemic side effects, and the escape mechanism of the tumor. This work aims to present the current modern treatments of glioblastoma.
Collapse
Affiliation(s)
- Karol Sadowski
- The Department of Histology and Embryology, Medical University of Warsaw, Chalubinskiego 5, 02-004 Warsaw, Poland; (K.S.)
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-091 Warsaw, Poland;
- Centre for Preclinical Research, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Adrianna Jażdżewska
- The Department of Anatomy and Neurobiology, Medical University of Gdansk, Dębinki 1, 80-211 Gdansk, Poland;
| | - Jan Kozłowski
- The Department of Histology and Embryology, Medical University of Warsaw, Chalubinskiego 5, 02-004 Warsaw, Poland; (K.S.)
| | - Aleksandra Zacny
- The Department of Histology and Embryology, Medical University of Warsaw, Chalubinskiego 5, 02-004 Warsaw, Poland; (K.S.)
| | - Tomasz Lorenc
- Department of Radiology I, The Maria Sklodowska-Curie National Research Institute of Oncology, Roentgena 5, 02-781 Warsaw, Poland
| | - Wioletta Olejarz
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-091 Warsaw, Poland;
- Centre for Preclinical Research, Medical University of Warsaw, 02-091 Warsaw, Poland
| |
Collapse
|
11
|
Shen Y, Thng DKH, Wong ALA, Toh TB. Mechanistic insights and the clinical prospects of targeted therapies for glioblastoma: a comprehensive review. Exp Hematol Oncol 2024; 13:40. [PMID: 38615034 PMCID: PMC11015656 DOI: 10.1186/s40164-024-00512-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 04/08/2024] [Indexed: 04/15/2024] Open
Abstract
Glioblastoma (GBM) is a fatal brain tumour that is traditionally diagnosed based on histological features. Recent molecular profiling studies have reshaped the World Health Organization approach in the classification of central nervous system tumours to include more pathogenetic hallmarks. These studies have revealed that multiple oncogenic pathways are dysregulated, which contributes to the aggressiveness and resistance of GBM. Such findings have shed light on the molecular vulnerability of GBM and have shifted the disease management paradigm from chemotherapy to targeted therapies. Targeted drugs have been developed to inhibit oncogenic targets in GBM, including receptors involved in the angiogenic axis, the signal transducer and activator of transcription 3 (STAT3), the PI3K/AKT/mTOR signalling pathway, the ubiquitination-proteasome pathway, as well as IDH1/2 pathway. While certain targeted drugs showed promising results in vivo, the translatability of such preclinical achievements in GBM remains a barrier. We also discuss the recent developments and clinical assessments of targeted drugs, as well as the prospects of cell-based therapies and combinatorial therapy as novel ways to target GBM. Targeted treatments have demonstrated preclinical efficacy over chemotherapy as an alternative or adjuvant to the current standard of care for GBM, but their clinical efficacy remains hindered by challenges such as blood-brain barrier penetrance of the drugs. The development of combinatorial targeted therapies is expected to improve therapeutic efficacy and overcome drug resistance.
Collapse
Affiliation(s)
- Yating Shen
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Dexter Kai Hao Thng
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Andrea Li Ann Wong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Department of Haematology-Oncology, National University Hospital, Singapore, Singapore
| | - Tan Boon Toh
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, Singapore.
- The Institute for Digital Medicine (WisDM), National University of Singapore, Singapore, Singapore.
| |
Collapse
|
12
|
Hayes TK, Aquilanti E, Persky NS, Yang X, Kim EE, Brenan L, Goodale AB, Alan D, Sharpe T, Shue RE, Westlake L, Golomb L, Silverman BR, Morris MD, Fisher TR, Beyene E, Li YY, Cherniack AD, Piccioni F, Hicks JK, Chi AS, Cahill DP, Dietrich J, Batchelor TT, Root DE, Johannessen CM, Meyerson M. Comprehensive mutational scanning of EGFR reveals TKI sensitivities of extracellular domain mutants. Nat Commun 2024; 15:2742. [PMID: 38548752 PMCID: PMC10978866 DOI: 10.1038/s41467-024-45594-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 01/30/2024] [Indexed: 04/01/2024] Open
Abstract
The epidermal growth factor receptor, EGFR, is frequently activated in lung cancer and glioblastoma by genomic alterations including missense mutations. The different mutation spectra in these diseases are reflected in divergent responses to EGFR inhibition: significant patient benefit in lung cancer, but limited in glioblastoma. Here, we report a comprehensive mutational analysis of EGFR function. We perform saturation mutagenesis of EGFR and assess function of ~22,500 variants in a human EGFR-dependent lung cancer cell line. This approach reveals enrichment of erlotinib-insensitive variants of known and unknown significance in the dimerization, transmembrane, and kinase domains. Multiple EGFR extracellular domain variants, not associated with approved targeted therapies, are sensitive to afatinib and dacomitinib in vitro. Two glioblastoma patients with somatic EGFR G598V dimerization domain mutations show responses to dacomitinib treatment followed by within-pathway resistance mutation in one case. In summary, this comprehensive screen expands the landscape of functional EGFR variants and suggests broader clinical investigation of EGFR inhibition for cancers harboring extracellular domain mutations.
Collapse
Affiliation(s)
- Tikvah K Hayes
- Department of Medical Oncology, Dana-Farber Cancer Institute & Harvard Medical School, Boston, MA, USA
- Cancer Program, The Broad Institute of M.I.T. and Harvard, Cambridge, MA, USA
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, USA
| | - Elisa Aquilanti
- Department of Medical Oncology, Dana-Farber Cancer Institute & Harvard Medical School, Boston, MA, USA
- Cancer Program, The Broad Institute of M.I.T. and Harvard, Cambridge, MA, USA
| | - Nicole S Persky
- Cancer Program, The Broad Institute of M.I.T. and Harvard, Cambridge, MA, USA
- Genetic Perturbation Platform, The Broad Institute of M.I.T. and Harvard, Cambridge, MA, USA
- Aera Therapeutics, Cambridge, MA, USA
| | - Xiaoping Yang
- Genetic Perturbation Platform, The Broad Institute of M.I.T. and Harvard, Cambridge, MA, USA
| | - Erica E Kim
- Department of Medical Oncology, Dana-Farber Cancer Institute & Harvard Medical School, Boston, MA, USA
| | - Lisa Brenan
- Cancer Program, The Broad Institute of M.I.T. and Harvard, Cambridge, MA, USA
| | - Amy B Goodale
- Genetic Perturbation Platform, The Broad Institute of M.I.T. and Harvard, Cambridge, MA, USA
| | - Douglas Alan
- Genetic Perturbation Platform, The Broad Institute of M.I.T. and Harvard, Cambridge, MA, USA
| | - Ted Sharpe
- Data Science Platform, The Broad Institute of M.I.T. and Harvard Cambridge, Cambridge, MA, USA
| | - Robert E Shue
- Department of Medical Oncology, Dana-Farber Cancer Institute & Harvard Medical School, Boston, MA, USA
- Cancer Program, The Broad Institute of M.I.T. and Harvard, Cambridge, MA, USA
| | - Lindsay Westlake
- Cancer Program, The Broad Institute of M.I.T. and Harvard, Cambridge, MA, USA
| | - Lior Golomb
- Department of Medical Oncology, Dana-Farber Cancer Institute & Harvard Medical School, Boston, MA, USA
- Cancer Program, The Broad Institute of M.I.T. and Harvard, Cambridge, MA, USA
| | - Brianna R Silverman
- Department of Medical Oncology, Dana-Farber Cancer Institute & Harvard Medical School, Boston, MA, USA
| | - Myshal D Morris
- Summer Honors Undergraduate Research Program, Harvard Medical School, Boston, MA, USA
| | - Ty Running Fisher
- Summer Honors Undergraduate Research Program, Harvard Medical School, Boston, MA, USA
| | - Eden Beyene
- Summer Honors Undergraduate Research Program, Harvard Medical School, Boston, MA, USA
| | - Yvonne Y Li
- Department of Medical Oncology, Dana-Farber Cancer Institute & Harvard Medical School, Boston, MA, USA
- Cancer Program, The Broad Institute of M.I.T. and Harvard, Cambridge, MA, USA
| | - Andrew D Cherniack
- Department of Medical Oncology, Dana-Farber Cancer Institute & Harvard Medical School, Boston, MA, USA
- Cancer Program, The Broad Institute of M.I.T. and Harvard, Cambridge, MA, USA
| | - Federica Piccioni
- Genetic Perturbation Platform, The Broad Institute of M.I.T. and Harvard, Cambridge, MA, USA
- Merck Research Laboratories, Cambridge, MA, USA
| | - J Kevin Hicks
- Department of Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Andrew S Chi
- Center for Neuro-Oncology, Division of Neuro-Oncology, Massachusetts General Hospital, Boston, MA, USA
| | - Daniel P Cahill
- Center for Neuro-Oncology, Division of Neuro-Oncology, Massachusetts General Hospital, Boston, MA, USA
| | - Jorg Dietrich
- Department of Neurology, Division of Neuro-Oncology, Massachusetts General Hospital, Boston, MA, USA
| | - Tracy T Batchelor
- Department of Neurology, Brigham and Women's Hospital & Harvard Medical School, Boston, MA, USA
| | - David E Root
- Genetic Perturbation Platform, The Broad Institute of M.I.T. and Harvard, Cambridge, MA, USA
| | - Cory M Johannessen
- Cancer Program, The Broad Institute of M.I.T. and Harvard, Cambridge, MA, USA
- Department of Oncology, Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | - Matthew Meyerson
- Department of Medical Oncology, Dana-Farber Cancer Institute & Harvard Medical School, Boston, MA, USA.
- Cancer Program, The Broad Institute of M.I.T. and Harvard, Cambridge, MA, USA.
| |
Collapse
|
13
|
Zhang M, Ding Y, Gao M, Lu X, Tan J, Yu F, Gu C, Gu L, Ren X, Hao C, Ming L, Xu K, Mao W, Jin Y, Zhang M, You L, Wang Z, Sun Y, Jiang J, Yang Y, Zhang D, Tang X. Discovery of Novel N-(Anthracen-9-ylmethyl) Benzamide Derivatives as ZNF207 Inhibitors Promising in Treating Glioma. J Med Chem 2024; 67:3909-3934. [PMID: 38377560 DOI: 10.1021/acs.jmedchem.3c02241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
Targeting tumor stemness is an innovative approach to cancer treatment. Zinc Finger Protein 207 (ZNF207) is a promising target for weakening the stemness of glioma cells. Here, a series of novel N-(anthracen-9-ylmethyl) benzamide derivatives against ZNF207 were rationally designed and synthesized. The inhibitory activity was evaluated, and their structure-activity relationships were summarized. Among them, C16 exhibited the most potent inhibitory activity, as evidenced by its IC50 values ranging from 0.5-2.5 μM for inhibiting sphere formation and 0.5-15 μM for cytotoxicity. Furthermore, we found that C16 could hinder tumorigenesis and migration and promote apoptosis in vitro. These effects were attributed to the downregulation of stem-related genes. The in vivo evaluation demonstrated that C16 exhibited efficient permeability across the blood-brain barrier and potent efficacy in both subcutaneous and orthotopic glioma tumor models. Hence, C16 may serve as a potential lead compound targeting ZNF207 and has promising therapeutic potential for glioma.
Collapse
Affiliation(s)
- Menghan Zhang
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China 211112
| | - Yushi Ding
- Institute of Pharmaceutical Sciences, China Pharmaceutical University, Nanjing, China 211112
| | - Mengkang Gao
- School of Pharmacy, China Pharmaceutical University, Nanjing, China 211112
| | - Xiaolin Lu
- School of Science, China Pharmaceutical University, Nanjing, China 211112
| | - Jun Tan
- School of Science, China Pharmaceutical University, Nanjing, China 211112
| | - Fei Yu
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China 211112
| | - Congying Gu
- School of Science, China Pharmaceutical University, Nanjing, China 211112
| | - Lujun Gu
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China 211112
| | - Xiameng Ren
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China 211112
| | - Chenyan Hao
- School of Science, China Pharmaceutical University, Nanjing, China 211112
| | - Liqin Ming
- School of Science, China Pharmaceutical University, Nanjing, China 211112
| | - Kang Xu
- School of Science, China Pharmaceutical University, Nanjing, China 211112
| | - Wenhao Mao
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China 211112
| | - Yuqing Jin
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China 211112
| | - Min Zhang
- Institute of Pharmaceutical Sciences, China Pharmaceutical University, Nanjing, China 211112
| | - Linjun You
- Institute of Pharmaceutical Sciences, China Pharmaceutical University, Nanjing, China 211112
- Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing, China 211112
| | - Zhanbo Wang
- Institute of Pharmaceutical Sciences, China Pharmaceutical University, Nanjing, China 211112
- Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing, China 211112
| | - Yuanyuan Sun
- Shuangyun BioMed Sci & Tech (Suzhou) Co., Ltd, Suzhou, China 215000
| | - Jingwei Jiang
- Shuangyun BioMed Sci & Tech (Suzhou) Co., Ltd, Suzhou, China 215000
| | - Yong Yang
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China 211112
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China 211112
- School of Pharmacy, Xuzhou Medical University, Xuzhou, China 221004
| | - Dayong Zhang
- School of Science, China Pharmaceutical University, Nanjing, China 211112
| | - Xinying Tang
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China 211112
| |
Collapse
|
14
|
Obrador E, Moreno-Murciano P, Oriol-Caballo M, López-Blanch R, Pineda B, Gutiérrez-Arroyo JL, Loras A, Gonzalez-Bonet LG, Martinez-Cadenas C, Estrela JM, Marqués-Torrejón MÁ. Glioblastoma Therapy: Past, Present and Future. Int J Mol Sci 2024; 25:2529. [PMID: 38473776 PMCID: PMC10931797 DOI: 10.3390/ijms25052529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 02/10/2024] [Accepted: 02/16/2024] [Indexed: 03/14/2024] Open
Abstract
Glioblastoma (GB) stands out as the most prevalent and lethal form of brain cancer. Although great efforts have been made by clinicians and researchers, no significant improvement in survival has been achieved since the Stupp protocol became the standard of care (SOC) in 2005. Despite multimodality treatments, recurrence is almost universal with survival rates under 2 years after diagnosis. Here, we discuss the recent progress in our understanding of GB pathophysiology, in particular, the importance of glioma stem cells (GSCs), the tumor microenvironment conditions, and epigenetic mechanisms involved in GB growth, aggressiveness and recurrence. The discussion on therapeutic strategies first covers the SOC treatment and targeted therapies that have been shown to interfere with different signaling pathways (pRB/CDK4/RB1/P16ink4, TP53/MDM2/P14arf, PI3k/Akt-PTEN, RAS/RAF/MEK, PARP) involved in GB tumorigenesis, pathophysiology, and treatment resistance acquisition. Below, we analyze several immunotherapeutic approaches (i.e., checkpoint inhibitors, vaccines, CAR-modified NK or T cells, oncolytic virotherapy) that have been used in an attempt to enhance the immune response against GB, and thereby avoid recidivism or increase survival of GB patients. Finally, we present treatment attempts made using nanotherapies (nanometric structures having active anti-GB agents such as antibodies, chemotherapeutic/anti-angiogenic drugs or sensitizers, radionuclides, and molecules that target GB cellular receptors or open the blood-brain barrier) and non-ionizing energies (laser interstitial thermal therapy, high/low intensity focused ultrasounds, photodynamic/sonodynamic therapies and electroporation). The aim of this review is to discuss the advances and limitations of the current therapies and to present novel approaches that are under development or following clinical trials.
Collapse
Affiliation(s)
- Elena Obrador
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Paz Moreno-Murciano
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
| | - María Oriol-Caballo
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Rafael López-Blanch
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Begoña Pineda
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Julia Lara Gutiérrez-Arroyo
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon, Spain; (J.L.G.-A.); (A.L.); (C.M.-C.)
| | - Alba Loras
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon, Spain; (J.L.G.-A.); (A.L.); (C.M.-C.)
| | - Luis G. Gonzalez-Bonet
- Department of Neurosurgery, Castellon General University Hospital, 12004 Castellon, Spain;
| | - Conrado Martinez-Cadenas
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon, Spain; (J.L.G.-A.); (A.L.); (C.M.-C.)
| | - José M. Estrela
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
- Department of Physiology, Faculty of Pharmacy, University of Valencia, 46100 Burjassot, Spain
| | | |
Collapse
|
15
|
Ezzati S, Salib S, Balasubramaniam M, Aboud O. Epidermal Growth Factor Receptor Inhibitors in Glioblastoma: Current Status and Future Possibilities. Int J Mol Sci 2024; 25:2316. [PMID: 38396993 PMCID: PMC10889328 DOI: 10.3390/ijms25042316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 02/11/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Glioblastoma, a grade 4 glioma as per the World Health Organization, poses a challenge in adult primary brain tumor management despite advanced surgical techniques and multimodal therapies. This review delves into the potential of targeting epidermal growth factor receptor (EGFR) with small-molecule inhibitors and antibodies as a treatment strategy. EGFR, a mutationally active receptor tyrosine kinase in over 50% of glioblastoma cases, features variants like EGFRvIII, EGFRvII and missense mutations, necessitating a deep understanding of their structures and signaling pathways. Although EGFR inhibitors have demonstrated efficacy in other cancers, their application in glioblastoma is hindered by blood-brain barrier penetration and intrinsic resistance. The evolving realm of nanodrugs and convection-enhanced delivery offers promise in ensuring precise drug delivery to the brain. Critical to success is the identification of glioblastoma patient populations that benefit from EGFR inhibitors. Tools like radiolabeled anti-EGFR antibody 806i facilitate the visualization of EGFR conformations, aiding in tailored treatment selection. Recognizing the synergistic potential of combination therapies with downstream targets like mTOR, PI3k, and HDACs is pivotal for enhancing EGFR inhibitor efficacy. In conclusion, the era of precision oncology holds promise for targeting EGFR in glioblastoma, contingent on tailored treatments, effective blood-brain barrier navigation, and the exploration of synergistic therapies.
Collapse
Affiliation(s)
- Shawyon Ezzati
- California Northstate University College of Medicine, Elk Grove, CA 95757, USA; (S.E.); (S.S.)
| | - Samuel Salib
- California Northstate University College of Medicine, Elk Grove, CA 95757, USA; (S.E.); (S.S.)
| | | | - Orwa Aboud
- Department of Neurology, Department of Neurological Surgery, Comprehensive Cancer Center, University of California, Davis, Sacramento, CA 95817, USA
| |
Collapse
|
16
|
Stitzlein LM, Adams JT, Stitzlein EN, Dudley RW, Chandra J. Current and future therapeutic strategies for high-grade gliomas leveraging the interplay between epigenetic regulators and kinase signaling networks. J Exp Clin Cancer Res 2024; 43:12. [PMID: 38183103 PMCID: PMC10768151 DOI: 10.1186/s13046-023-02923-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 12/05/2023] [Indexed: 01/07/2024] Open
Abstract
Targeted therapies, including small molecule inhibitors directed against aberrant kinase signaling and chromatin regulators, are emerging treatment options for high-grade gliomas (HGG). However, when translating these inhibitors into the clinic, their efficacy is generally limited to partial and transient responses. Recent studies in models of high-grade gliomas reveal a convergence of epigenetic regulators and kinase signaling networks that often cooperate to promote malignant properties and drug resistance. This review examines the interplay between five well-characterized groups of chromatin regulators, including the histone deacetylase (HDAC) family, bromodomain and extraterminal (BET)-containing proteins, protein arginine methyltransferase (PRMT) family, Enhancer of zeste homolog 2 (EZH2), and lysine-specific demethylase 1 (LSD1), and various signaling pathways essential for cancer cell growth and progression. These specific epigenetic regulators were chosen for review due to their targetability via pharmacological intervention and clinical relevance. Several studies have demonstrated improved efficacy from the dual inhibition of the epigenetic regulators and signaling kinases. Overall, the interactions between epigenetic regulators and kinase signaling pathways are likely influenced by several factors, including individual glioma subtypes, preexisting mutations, and overlapping/interdependent functions of the chromatin regulators. The insights gained by understanding how the genome and epigenome cooperate in high-grade gliomas will guide the design of future therapeutic strategies that utilize dual inhibition with improved efficacy and overall survival.
Collapse
Affiliation(s)
- Lea M Stitzlein
- Department of Pediatrics Research, The MD Anderson Cancer Center, University of Texas, Box 853, 1515 Holcombe Blvd, Houston, TX, 77030, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Jack T Adams
- Department of Pediatrics Research, The MD Anderson Cancer Center, University of Texas, Box 853, 1515 Holcombe Blvd, Houston, TX, 77030, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | | | - Richard W Dudley
- Department of Pharmaceutical Sciences, University of Findlay, Findlay, OH, USA
| | - Joya Chandra
- Department of Pediatrics Research, The MD Anderson Cancer Center, University of Texas, Box 853, 1515 Holcombe Blvd, Houston, TX, 77030, USA.
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA.
- Department of Epigenetics and Molecular Carcinogenesis, The MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
17
|
Kim T, Lee A, Ahn S, Park JS, Jeun SS, Lee YS. Comprehensive Molecular Genetic Analysis in Glioma Patients by Next Generation Sequencing. Brain Tumor Res Treat 2024; 12:23-39. [PMID: 38317486 PMCID: PMC10864139 DOI: 10.14791/btrt.2023.0036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 10/16/2023] [Accepted: 11/10/2023] [Indexed: 02/07/2024] Open
Abstract
BACKGROUND Glioma is caused by multiple genomic alterations. The evolving classification of gliomas emphasizes the significance of molecular testing. Next generation sequencing (NGS) offers the assessment of parallel combinations of multiple genetic alterations and identifying actionable mutations that guide treatment. This study comprehensively analyzed glioma patients using multi-gene NGS panels, providing powerful insights to inform diagnostic classification and targeted therapies. METHODS We conducted a targeted panel-based NGS analysis on formalin-fixed and paraffin-embedded nucleic acids extracted from a total of 147 glioma patients. These samples underwent amplicon capture-based library preparation and sequenced using the Oncomine Comprehensive Assay platform. The resulting sequencing data were then analyzed using the bioinformatics tools. RESULTS A total of 301 mutations, were found in 132 out of 147 tumors (89.8%). These mutations were in 68 different genes. In 62 tumor samples (42.2%), copy number variations (CNVs) with gene amplifications occurred in 25 genes. Moreover, 25 tumor samples (17.0%) showed gene fusions in 6 genes and intragenic deletion in a gene. Our analysis identified actionable targets in several genes, including 11 with mutations, 8 with CNVs, and 3 with gene fusions and intragenic deletion. These findings could impact FDA-approved therapies, NCCN guideline-based treatments, and clinical trials. CONCLUSION We analyzed precisely diagnosing the classification of gliomas, detailing the frequency and co-occurrence of genetic alterations and identifying genetic alterations with potential therapeutic targets by NGS-based molecular analysis. The high-throughput NGS analysis is an efficient and powerful tool to comprehensively support molecular testing in neurooncology.
Collapse
Affiliation(s)
- Taeeun Kim
- Department of Hospital Pathology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ahwon Lee
- Department of Hospital Pathology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Stephan Ahn
- Department of Neurosurgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jae Sung Park
- Department of Neurosurgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sin Soo Jeun
- Department of Neurosurgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Youn Soo Lee
- Department of Hospital Pathology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea.
| |
Collapse
|
18
|
Nettnin EA, Nguyen T, Arana S, Barros Guinle MI, Garcia CA, Gibson EM, Prolo LM. Review: therapeutic approaches for circadian modulation of the glioma microenvironment. Front Oncol 2023; 13:1295030. [PMID: 38173841 PMCID: PMC10762863 DOI: 10.3389/fonc.2023.1295030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 11/28/2023] [Indexed: 01/05/2024] Open
Abstract
High-grade gliomas are malignant brain tumors that are characteristically hard to treat because of their nature; they grow quickly and invasively through the brain tissue and develop chemoradiation resistance in adults. There is also a distinct lack of targeted treatment options in the pediatric population for this tumor type to date. Several approaches to overcome therapeutic resistance have been explored, including targeted therapy to growth pathways (ie. EGFR and VEGF inhibitors), epigenetic modulators, and immunotherapies such as Chimeric Antigen Receptor T-cell and vaccine therapies. One new promising approach relies on the timing of chemotherapy administration based on intrinsic circadian rhythms. Recent work in glioblastoma has demonstrated temporal variations in chemosensitivity and, thus, improved survival based on treatment time of day. This may be due to intrinsic rhythms of the glioma cells, permeability of the blood brain barrier to chemotherapy agents, the tumor immune microenvironment, or another unknown mechanism. We review the literature to discuss chronotherapeutic approaches to high-grade glioma treatment, circadian regulation of the immune system and tumor microenvironment in gliomas. We further discuss how these two areas may be combined to temporally regulate and/or improve the effectiveness of immunotherapies.
Collapse
Affiliation(s)
- Ella A. Nettnin
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Thien Nguyen
- Division of Pediatric Hematology/Oncology, Lucile Packard Children’s Hospital, Palo Alto, CA, United States
| | - Sophia Arana
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, United States
| | | | - Cesar A. Garcia
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Erin M. Gibson
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, United States
| | - Laura M. Prolo
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, United States
- Division of Pediatric Neurosurgery, Lucile Packard Children’s Hospital, Palo Alto, CA, United States
| |
Collapse
|
19
|
Dewdney B, Jenkins MR, Best SA, Freytag S, Prasad K, Holst J, Endersby R, Johns TG. From signalling pathways to targeted therapies: unravelling glioblastoma's secrets and harnessing two decades of progress. Signal Transduct Target Ther 2023; 8:400. [PMID: 37857607 PMCID: PMC10587102 DOI: 10.1038/s41392-023-01637-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 08/29/2023] [Accepted: 09/07/2023] [Indexed: 10/21/2023] Open
Abstract
Glioblastoma, a rare, and highly lethal form of brain cancer, poses significant challenges in terms of therapeutic resistance, and poor survival rates for both adult and paediatric patients alike. Despite advancements in brain cancer research driven by a technological revolution, translating our understanding of glioblastoma pathogenesis into improved clinical outcomes remains a critical unmet need. This review emphasises the intricate role of receptor tyrosine kinase signalling pathways, epigenetic mechanisms, and metabolic functions in glioblastoma tumourigenesis and therapeutic resistance. We also discuss the extensive efforts over the past two decades that have explored targeted therapies against these pathways. Emerging therapeutic approaches, such as antibody-toxin conjugates or CAR T cell therapies, offer potential by specifically targeting proteins on the glioblastoma cell surface. Combination strategies incorporating protein-targeted therapy and immune-based therapies demonstrate great promise for future clinical research. Moreover, gaining insights into the role of cell-of-origin in glioblastoma treatment response holds the potential to advance precision medicine approaches. Addressing these challenges is crucial to improving outcomes for glioblastoma patients and moving towards more effective precision therapies.
Collapse
Affiliation(s)
- Brittany Dewdney
- Cancer Centre, Telethon Kids Institute, Nedlands, WA, 6009, Australia.
- Centre For Child Health Research, University of Western Australia, Perth, WA, 6009, Australia.
| | - Misty R Jenkins
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, 3052, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, 3010, Australia
| | - Sarah A Best
- Department of Medical Biology, University of Melbourne, Melbourne, 3010, Australia
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, 3052, Australia
| | - Saskia Freytag
- Department of Medical Biology, University of Melbourne, Melbourne, 3010, Australia
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, 3052, Australia
| | - Krishneel Prasad
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, 3052, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, 3010, Australia
| | - Jeff Holst
- School of Biomedical Sciences, University of New South Wales, Sydney, 2052, Australia
| | - Raelene Endersby
- Cancer Centre, Telethon Kids Institute, Nedlands, WA, 6009, Australia
- Centre For Child Health Research, University of Western Australia, Perth, WA, 6009, Australia
| | - Terrance G Johns
- Cancer Centre, Telethon Kids Institute, Nedlands, WA, 6009, Australia
- Centre For Child Health Research, University of Western Australia, Perth, WA, 6009, Australia
| |
Collapse
|
20
|
Pant A, Lim M. Overcoming EGFR inhibitor resistance in Glioblastoma by targeting co-amplified genes. Proc Natl Acad Sci U S A 2023; 120:e2312277120. [PMID: 37672559 PMCID: PMC10515143 DOI: 10.1073/pnas.2312277120] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/08/2023] Open
Affiliation(s)
- Ayush Pant
- Department of Neurosurgery, School of Medicine, Johns Hopkins University, Baltimore, MD21287
| | - Michael Lim
- Department of Neurosurgery, School of Medicine, Stanford University, Stanford, CA94304
| |
Collapse
|
21
|
Weiser A, Sanchez Bergman A, Machaalani C, Bennett J, Roth P, Reimann RR, Nazarian J, Guerreiro Stucklin AS. Bridging the age gap: a review of molecularly informed treatments for glioma in adolescents and young adults. Front Oncol 2023; 13:1254645. [PMID: 37781183 PMCID: PMC10533987 DOI: 10.3389/fonc.2023.1254645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 08/14/2023] [Indexed: 10/03/2023] Open
Abstract
Gliomas are the most common primary central nervous system (CNS) tumors and a major cause of cancer-related mortality in children (age <15 years), adolescents and young adults (AYA, ages 15-39 years), and adults (age >39 years). Molecular pathology has helped enhance the characterization of these tumors, revealing a heterogeneous and ever more complex group of malignancies. Recent molecular analyses have led to an increased appreciation of common genomic alterations prevalent across all ages. The 2021 World Health Organization (WHO) CNS tumor classification, 5th edition (WHO CNS5) brings forward a nomenclature distinguishing "pediatric-type" and "adult-type" gliomas. The spectrum of gliomas in AYA comprises both "pediatric-like" and "adult-like" tumor entities but remains ill-defined. With fragmentation of clinical management between pediatric and adult centers, AYAs face challenges related to gaps in medical care, lower rates of enrollment in clinical trials and additional psychosocial and economic challenges. This calls for a rethinking of diagnostic and therapeutic approaches, to improve access to appropriate testing and potentially beneficial treatments to patients of all ages.
Collapse
Affiliation(s)
- Annette Weiser
- Translational Brain Tumor Research Group, Children’s Research Center, University Children’s Hospital Zurich, Zurich, Switzerland
- Division of Oncology, University Children’s Hospital Zurich, Zurich, Switzerland
| | - Astrid Sanchez Bergman
- Translational Brain Tumor Research Group, Children’s Research Center, University Children’s Hospital Zurich, Zurich, Switzerland
| | - Charbel Machaalani
- Translational Brain Tumor Research Group, Children’s Research Center, University Children’s Hospital Zurich, Zurich, Switzerland
| | - Julie Bennett
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, ON, Canada
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Patrick Roth
- Department of Neurology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Regina R. Reimann
- Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland
| | - Javad Nazarian
- Department of Pediatrics, Diffuse Midline Glioma (DMG) / Diffuse Intrinsic Pontine Glioma (DIPG) Center, Children’s Research Center, University Children’s Hospital Zurich, Zurich, Switzerland
- Research Center for Genetic Medicine, Children's National Hospital, Washington, DC, United States
| | - Ana S. Guerreiro Stucklin
- Translational Brain Tumor Research Group, Children’s Research Center, University Children’s Hospital Zurich, Zurich, Switzerland
- Division of Oncology, University Children’s Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
22
|
Vaz-Salgado MA, Villamayor M, Albarrán V, Alía V, Sotoca P, Chamorro J, Rosero D, Barrill AM, Martín M, Fernandez E, Gutierrez JA, Rojas-Medina LM, Ley L. Recurrent Glioblastoma: A Review of the Treatment Options. Cancers (Basel) 2023; 15:4279. [PMID: 37686553 PMCID: PMC10487236 DOI: 10.3390/cancers15174279] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/14/2023] [Accepted: 08/17/2023] [Indexed: 09/10/2023] Open
Abstract
Glioblastoma is a disease with a poor prognosis. Multiple efforts have been made to improve the long-term outcome, but the 5-year survival rate is still 5-10%. Recurrence of the disease is the usual way of progression. In this situation, there is no standard treatment. Different treatment options can be considered. Among them would be reoperation or reirradiation. There are different studies that have assessed the impact on survival and the selection of patients who may benefit most from these strategies. Chemotherapy treatments have also been considered in several studies, mainly with alkylating agents, with data mostly from phase II studies. On the other hand, multiple studies have been carried out with target-directed treatments. Bevacizumab, a monoclonal antibody with anti-angiogenic activity, has demonstrated activity in several studies, and the FDA has approved it for this indication. Several other TKI drugs have been evaluated in this setting, but no clear benefit has been demonstrated. Immunotherapy treatments have been shown to be effective in other types of tumors, and several studies have evaluated their efficacy in this disease, both immune checkpoint inhibitors, oncolytic viruses, and vaccines. This paper reviews data from different studies that have evaluated the efficacy of different forms of relapsed glioblastoma.
Collapse
Affiliation(s)
- Maria Angeles Vaz-Salgado
- Medical Oncology Department, Ramon y Cajal University Hospital, 28034 Madrid, Spain; (M.V.); (V.A.); (V.A.); (P.S.); (J.C.); (D.R.); (A.M.B.)
| | - María Villamayor
- Medical Oncology Department, Ramon y Cajal University Hospital, 28034 Madrid, Spain; (M.V.); (V.A.); (V.A.); (P.S.); (J.C.); (D.R.); (A.M.B.)
| | - Víctor Albarrán
- Medical Oncology Department, Ramon y Cajal University Hospital, 28034 Madrid, Spain; (M.V.); (V.A.); (V.A.); (P.S.); (J.C.); (D.R.); (A.M.B.)
| | - Víctor Alía
- Medical Oncology Department, Ramon y Cajal University Hospital, 28034 Madrid, Spain; (M.V.); (V.A.); (V.A.); (P.S.); (J.C.); (D.R.); (A.M.B.)
| | - Pilar Sotoca
- Medical Oncology Department, Ramon y Cajal University Hospital, 28034 Madrid, Spain; (M.V.); (V.A.); (V.A.); (P.S.); (J.C.); (D.R.); (A.M.B.)
| | - Jesús Chamorro
- Medical Oncology Department, Ramon y Cajal University Hospital, 28034 Madrid, Spain; (M.V.); (V.A.); (V.A.); (P.S.); (J.C.); (D.R.); (A.M.B.)
| | - Diana Rosero
- Medical Oncology Department, Ramon y Cajal University Hospital, 28034 Madrid, Spain; (M.V.); (V.A.); (V.A.); (P.S.); (J.C.); (D.R.); (A.M.B.)
| | - Ana M. Barrill
- Medical Oncology Department, Ramon y Cajal University Hospital, 28034 Madrid, Spain; (M.V.); (V.A.); (V.A.); (P.S.); (J.C.); (D.R.); (A.M.B.)
| | - Mercedes Martín
- Radiotherapy Oncology Department, Ramon y Cajal University Hospital, 28034 Madrid, Spain; (M.M.); (E.F.)
| | - Eva Fernandez
- Radiotherapy Oncology Department, Ramon y Cajal University Hospital, 28034 Madrid, Spain; (M.M.); (E.F.)
| | - José Antonio Gutierrez
- Neurosurgery Department, Ramon y Cajal University Hospital, 28034 Madrid, Spain; (J.A.G.); (L.M.R.-M.); (L.L.)
| | - Luis Mariano Rojas-Medina
- Neurosurgery Department, Ramon y Cajal University Hospital, 28034 Madrid, Spain; (J.A.G.); (L.M.R.-M.); (L.L.)
| | - Luis Ley
- Neurosurgery Department, Ramon y Cajal University Hospital, 28034 Madrid, Spain; (J.A.G.); (L.M.R.-M.); (L.L.)
| |
Collapse
|
23
|
Li XP, Guo ZQ, Wang BF, Zhao M. EGFR alterations in glioblastoma play a role in antitumor immunity regulation. Front Oncol 2023; 13:1236246. [PMID: 37601668 PMCID: PMC10436475 DOI: 10.3389/fonc.2023.1236246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 07/20/2023] [Indexed: 08/22/2023] Open
Abstract
The epidermal growth factor receptor (EGFR) is the most frequently altered gene in glioblastoma (GBM), which plays an important role in tumor development and anti-tumor immune response. While current molecular targeted therapies against the EGFR signaling pathway and its downstream key molecules have not demonstrated favorable clinical outcomes in GBM. Whereas tumor immunotherapies, especially immune checkpoint inhibitors, have shown durable antitumor responses in many cancers. However, the clinical efficacy is limited in patients carrying EGFR alterations, indicating that EGFR signaling may involve tumor immune response. Recent studies reveal that EGFR alterations not only promote GBM cell proliferation but also influence immune components in the tumor microenvironment (TME), leading to the recruitment of immunosuppressive cells (e.g., M2-like TAMs, MDSCs, and Tregs), and inhibition of T and NK cell activation. Moreover, EGFR alterations upregulate the expression of immunosuppressive molecules or cytokines (such as PD-L1, CD73, TGF-β). This review explores the role of EGFR alterations in establishing an immunosuppressive TME and hopes to provide a theoretical basis for combining targeted EGFR inhibitors with immunotherapy for GBM.
Collapse
Affiliation(s)
| | | | - Bao-Feng Wang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Zhao
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
24
|
Boylan J, Byers E, Kelly DF. The Glioblastoma Landscape: Hallmarks of Disease, Therapeutic Resistance, and Treatment Opportunities. MEDICAL RESEARCH ARCHIVES 2023; 11:10.18103/mra.v11i6.3994. [PMID: 38107346 PMCID: PMC10723753 DOI: 10.18103/mra.v11i6.3994] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Malignant brain tumors are aggressive and difficult to treat. Glioblastoma is the most common and lethal form of primary brain tumor, often found in patients with no genetic predisposition. The median life expectancy for individuals diagnosed with this condition is 6 months to 2 years and there is no known cure. New paradigms in cancer biology implicate a small subset of tumor cells in initiating and sustaining these incurable brain tumors. Here, we discuss the heterogenous nature of glioblastoma and theories behind its capacity for therapy resistance and recurrence. Within the cancer landscape, cancer stem cells are thought to be both tumor initiators and major contributors to tumor heterogeneity and therapy evasion and such cells have been identified in glioblastoma. At the cellular level, disruptions in the delicate balance between differentiation and self-renewal spur transformation and support tumor growth. While rapidly dividing cells are more sensitive to elimination by traditional treatments, glioblastoma stem cells evade these measures through slow division and reversible exit from the cell cycle. At the molecular level, glioblastoma tumor cells exploit several signaling pathways to evade conventional therapies through improved DNA repair mechanisms and a flexible state of senescence. We examine these common evasion techniques while discussing potential molecular approaches to better target these deadly tumors. Equally important, the presented information encourages the idea of augmenting conventional treatments with novel glioblastoma stem cell-directed therapies, as eliminating these harmful progenitors holds great potential to modulate tumor recurrence.
Collapse
Affiliation(s)
- Jack Boylan
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA 16802, USA
- Center for Structural Oncology, Pennsylvania State University, University Park, PA 16802, USA
- Molecular, Cellular, and Integrative Biosciences Graduate Program, Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802, USA
| | - Elizabeth Byers
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA 16802, USA
- Molecular, Cellular, and Integrative Biosciences Graduate Program, Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802, USA
| | - Deborah F. Kelly
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA 16802, USA
- Center for Structural Oncology, Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
25
|
Darabi S, Xiu J, Samec T, Kesari S, Carrillo J, Aulakh S, Walsh KM, Sengupta S, Sumrall A, Spetzler D, Glantz M, Demeure MJ. Capicua (CIC) mutations in gliomas in association with MAPK activation for exposing a potential therapeutic target. Med Oncol 2023; 40:197. [PMID: 37291277 DOI: 10.1007/s12032-023-02071-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 06/02/2023] [Indexed: 06/10/2023]
Abstract
Gliomas are the most prevalent neurological cancer in the USA and care modalities are not able to effectively combat these aggressive malignancies. Identifying new, more effective treatments require a deep understanding of the complex genetic variations and relevant pathway associations behind these cancers. Drawing connections between gene mutations with a responsive genetic target can help drive therapy selections to enhance patient survival. We have performed extensive molecular profiling of the Capicua gene (CIC), a tumor and transcriptional suppressor gene, and its mutation prevalence in reference to MAPK activation within clinical glioma tissue. CIC mutations occur far more frequently in oligodendroglioma (52.1%) than in low-grade astrocytoma or glioblastoma. CIC-associated mutations were observed across all glioma subtypes, and MAPK-associated mutations were most prevalent in CIC wild-type tissue regardless of the glioma subtype. MAPK activation, however, was enhanced in CIC-mutated oligodendroglioma. The totality of our observations reported supports the use of CIC as a relevant genetic marker for MAPK activation. Identification of CIC mutations, or lack thereof, can assist in selecting, implementing, and developing MEK/MAPK-inhibitory trials to improve patient outcomes potentially.
Collapse
Affiliation(s)
- Sourat Darabi
- Hoag Family Cancer Institute, Newport Beach, CA, USA
| | | | | | - Santosh Kesari
- Hoag Family Cancer Institute, Newport Beach, CA, USA
- Pacific Neuroscience Institute, Providence Saint John's Health Center, Santa Monica, CA, USA
| | - Jose Carrillo
- Hoag Family Cancer Institute, Newport Beach, CA, USA
- Pacific Neuroscience Institute, Providence Saint John's Health Center, Santa Monica, CA, USA
| | | | - Kyle M Walsh
- Duke University School of Medicine, Durham, NC, USA
| | - Soma Sengupta
- University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | | | | | | | - Michael J Demeure
- Hoag Family Cancer Institute, Newport Beach, CA, USA
- Translational Genomics Research Institute, Phoenix, AZ, USA
| |
Collapse
|
26
|
PRMT6-CDC20 facilitates glioblastoma progression via the degradation of CDKN1B. Oncogene 2023; 42:1088-1100. [PMID: 36792756 PMCID: PMC10063447 DOI: 10.1038/s41388-023-02624-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 01/30/2023] [Accepted: 02/01/2023] [Indexed: 02/17/2023]
Abstract
PRMT6, a type I arginine methyltransferase, di-methylates the arginine residues of both histones and non-histones asymmetrically. Increasing evidence indicates that PRMT6 plays a tumor mediator involved in human malignancies. Here, we aim to uncover the essential role and underlying mechanisms of PRMT6 in promoting glioblastoma (GBM) proliferation. Investigation of PRMT6 expression in glioma tissues demonstrated that PRMT6 is overexpressed, and elevated expression of PRMT6 is negatively correlated with poor prognosis in glioma/GBM patients. Silencing PRMT6 inhibited GBM cell proliferation and induced cell cycle arrest at the G0/G1 phase, while overexpressing PRMT6 had opposite results. Further, we found that PRMT6 attenuates the protein stability of CDKN1B by promoting its degradation. Subsequent mechanistic investigations showed that PRMT6 maintains the transcription of CDC20 by activating histone methylation mark (H3R2me2a), and CDC20 interacts with and destabilizes CDKN1B. Rescue experimental results confirmed that PRMT6 promotes the ubiquitinated degradation of CDKN1B and cell proliferation via CDC20. We also verified that the PRMT6 inhibitor (EPZ020411) could attenuate the proliferative effect of GBM cells. Our findings illustrate that PRMT6, an epigenetic mediator, promotes CDC20 transcription via H3R2me2a to mediate the degradation of CDKN1B to facilitate GBM progression. Targeting PRMT6-CDC20-CDKN1B axis might be a promising therapeutic strategy for GBM.
Collapse
|
27
|
Li S, Wang C, Chen J, Lan Y, Zhang W, Kang Z, Zheng Y, Zhang R, Yu J, Li W. Signaling pathways in brain tumors and therapeutic interventions. Signal Transduct Target Ther 2023; 8:8. [PMID: 36596785 PMCID: PMC9810702 DOI: 10.1038/s41392-022-01260-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/16/2022] [Accepted: 11/21/2022] [Indexed: 01/05/2023] Open
Abstract
Brain tumors, although rare, contribute to distinct mortality and morbidity at all ages. Although there are few therapeutic options for brain tumors, enhanced biological understanding and unexampled innovations in targeted therapies and immunotherapies have considerably improved patients' prognoses. Nonetheless, the reduced response rates and unavoidable drug resistance of currently available treatment approaches have become a barrier to further improvement in brain tumor (glioma, meningioma, CNS germ cell tumors, and CNS lymphoma) treatment. Previous literature data revealed that several different signaling pathways are dysregulated in brain tumor. Importantly, a better understanding of targeting signaling pathways that influences malignant behavior of brain tumor cells might open the way for the development of novel targeted therapies. Thus, there is an urgent need for a more comprehensive understanding of the pathogenesis of these brain tumors, which might result in greater progress in therapeutic approaches. This paper began with a brief description of the epidemiology, incidence, risk factors, as well as survival of brain tumors. Next, the major signaling pathways underlying these brain tumors' pathogenesis and current progress in therapies, including clinical trials, targeted therapies, immunotherapies, and system therapies, have been systemically reviewed and discussed. Finally, future perspective and challenges of development of novel therapeutic strategies in brain tumor were emphasized.
Collapse
Affiliation(s)
- Shenglan Li
- grid.24696.3f0000 0004 0369 153XDepartment of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Can Wang
- grid.24696.3f0000 0004 0369 153XDepartment of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jinyi Chen
- grid.24696.3f0000 0004 0369 153XDepartment of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yanjie Lan
- grid.24696.3f0000 0004 0369 153XDepartment of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Weichunbai Zhang
- grid.24696.3f0000 0004 0369 153XDepartment of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhuang Kang
- grid.24696.3f0000 0004 0369 153XDepartment of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yi Zheng
- grid.24696.3f0000 0004 0369 153XDepartment of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Rong Zhang
- grid.24696.3f0000 0004 0369 153XDepartment of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jianyu Yu
- grid.24696.3f0000 0004 0369 153XDepartment of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wenbin Li
- Department of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
28
|
Mowforth OD, Brannigan J, El Khoury M, Sarathi CIP, Bestwick H, Bhatti F, Mair R. Personalised therapeutic approaches to glioblastoma: A systematic review. Front Med (Lausanne) 2023; 10:1166104. [PMID: 37122327 PMCID: PMC10140534 DOI: 10.3389/fmed.2023.1166104] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 03/23/2023] [Indexed: 05/02/2023] Open
Abstract
Introduction Glioblastoma is the most common and malignant primary brain tumour with median survival of 14.6 months. Personalised medicine aims to improve survival by targeting individualised patient characteristics. However, a major limitation has been application of targeted therapies in a non-personalised manner without biomarker enrichment. This has risked therapies being discounted without fair and rigorous evaluation. The objective was therefore to synthesise the current evidence on survival efficacy of personalised therapies in glioblastoma. Methods Studies reporting a survival outcome in human adults with supratentorial glioblastoma were eligible. PRISMA guidelines were followed. MEDLINE, Embase, Scopus, Web of Science and the Cochrane Library were searched to 5th May 2022. Clinicaltrials.gov was searched to 25th May 2022. Reference lists were hand-searched. Duplicate title/abstract screening, data extraction and risk of bias assessments were conducted. A quantitative synthesis is presented. Results A total of 102 trials were included: 16 were randomised and 41 studied newly diagnosed patients. Of 5,527 included patients, 59.4% were male and mean age was 53.7 years. More than 20 types of personalised therapy were included: targeted molecular therapies were the most studied (33.3%, 34/102), followed by autologous dendritic cell vaccines (32.4%, 33/102) and autologous tumour vaccines (10.8%, 11/102). There was no consistent evidence for survival efficacy of any personalised therapy. Conclusion Personalised glioblastoma therapies remain of unproven survival benefit. Evidence is inconsistent with high risk of bias. Nonetheless, encouraging results in some trials provide reason for optimism. Future focus should address target-enriched trials, combination therapies, longitudinal biomarker monitoring and standardised reporting.
Collapse
Affiliation(s)
- Oliver D. Mowforth
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, England, United Kingdom
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, England, United Kingdom
| | - Jamie Brannigan
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, England, United Kingdom
| | - Marc El Khoury
- School of Clinical Medicine, University of Cambridge, Cambridge, England, United Kingdom
| | | | - Harry Bestwick
- School of Clinical Medicine, University of Cambridge, Cambridge, England, United Kingdom
| | - Faheem Bhatti
- School of Clinical Medicine, University of Cambridge, Cambridge, England, United Kingdom
| | - Richard Mair
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, England, United Kingdom
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, England, United Kingdom
- *Correspondence: Richard Mair,
| |
Collapse
|
29
|
Tyrosine Kinase Inhibitors for Glioblastoma Multiforme: Challenges and Opportunities for Drug Delivery. Pharmaceutics 2022; 15:pharmaceutics15010059. [PMID: 36678688 PMCID: PMC9863099 DOI: 10.3390/pharmaceutics15010059] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
Glioblastoma multiforme (GBM) is an aggressive brain tumor with high mortality rates. Due to its invasiveness, heterogeneity, and incomplete resection, the treatment is very challenging. Targeted therapies such as tyrosine kinase inhibitors (TKIs) have great potential for GBM treatment, however, their efficacy is primarily limited by poor brain distribution due to the presence of the blood-brain barrier (BBB). This review focuses on the potential of TKIs in GBM therapy and provides an insight into the reasons behind unsuccessful clinical trials of TKIs in GBM despite the success in treating other cancer types. The main section is dedicated to the use of promising drug delivery strategies for targeted delivery to brain tumors. Use of brain targeted delivery strategies can help enhance the efficacy of TKIs in GBM. Among various drug delivery approaches used to bypass or cross BBB, utilizing nanocarriers is a promising strategy to augment the pharmacokinetic properties of TKIs and overcome their limitations. This is because of their advantages such as the ability to cross BBB, chemical stabilization of drug in circulation, passive or active targeting of tumor, modulation of drug release from the carrier, and the possibility to be delivered via non-invasive intranasal route.
Collapse
|
30
|
Lin B, Ziebro J, Smithberger E, Skinner KR, Zhao E, Cloughesy TF, Binder ZA, O’Rourke DM, Nathanson DA, Furnari FB, Miller CR. EGFR, the Lazarus target for precision oncology in glioblastoma. Neuro Oncol 2022; 24:2035-2062. [PMID: 36125064 PMCID: PMC9713527 DOI: 10.1093/neuonc/noac204] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The Lazarus effect is a rare condition that happens when someone seemingly dead shows signs of life. The epidermal growth factor receptor (EGFR) represents a target in the fatal neoplasm glioblastoma (GBM) that through a series of negative clinical trials has prompted a vocal subset of the neuro-oncology community to declare this target dead. However, an argument can be made that the core tenets of precision oncology were overlooked in the initial clinical enthusiasm over EGFR as a therapeutic target in GBM. Namely, the wrong drugs were tested on the wrong patients at the wrong time. Furthermore, new insights into the biology of EGFR in GBM vis-à-vis other EGFR-driven neoplasms, such as non-small cell lung cancer, and development of novel GBM-specific EGFR therapeutics resurrects this target for future studies. Here, we will examine the distinct EGFR biology in GBM, how it exacerbates the challenge of treating a CNS neoplasm, how these unique challenges have influenced past and present EGFR-targeted therapeutic design and clinical trials, and what adjustments are needed to therapeutically exploit EGFR in this devastating disease.
Collapse
Affiliation(s)
- Benjamin Lin
- Department of Pathology, Division of Neuropathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Julia Ziebro
- Department of Pathology, Division of Neuropathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Erin Smithberger
- Department of Pathology, Division of Neuropathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Pathobiology and Translational Sciences Program, School of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Kasey R Skinner
- Department of Pathology, Division of Neuropathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Neurosciences Curriculum, School of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Eva Zhao
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Timothy F Cloughesy
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Zev A Binder
- Department of Neurosurgery and Glioblastoma Translational Center of Excellence, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Donald M O’Rourke
- Department of Neurosurgery and Glioblastoma Translational Center of Excellence, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - David A Nathanson
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Frank B Furnari
- Department of Medicine, Division of Regenerative Medicine, University of California, San Diego, San Diego, California, USA
- Ludwig Cancer Research, San Diego, California, USA
| | - C Ryan Miller
- Department of Pathology, Division of Neuropathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
31
|
Dewdney B, Ursich L, Fletcher EV, Johns TG. Anoctamins and Calcium Signalling: An Obstacle to EGFR Targeted Therapy in Glioblastoma? Cancers (Basel) 2022; 14:cancers14235932. [PMID: 36497413 PMCID: PMC9740065 DOI: 10.3390/cancers14235932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 11/28/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022] Open
Abstract
Glioblastoma is the most common form of high-grade glioma in adults and has a poor survival rate with very limited treatment options. There have been no significant advancements in glioblastoma treatment in over 30 years. Epidermal growth factor receptor is upregulated in most glioblastoma tumours and, therefore, has been a drug target in recent targeted therapy clinical trials. However, while many inhibitors and antibodies for epidermal growth factor receptor have demonstrated promising anti-tumour effects in preclinical models, they have failed to improve outcomes for glioblastoma patients in clinical trials. This is likely due to the highly plastic nature of glioblastoma tumours, which results in therapeutic resistance. Ion channels are instrumental in the development of many cancers and may regulate cellular plasticity in glioblastoma. This review will explore the potential involvement of a class of calcium-activated chloride channels called anoctamins in brain cancer. We will also discuss the integrated role of calcium channels and anoctamins in regulating calcium-mediated signalling pathways, such as epidermal growth factor signalling, to promote brain cancer cell growth and migration.
Collapse
Affiliation(s)
- Brittany Dewdney
- Cancer Centre, Telethon Kids Institute, Perth, WA 6009, Australia
- Centre for Child Health Research, University of Western Australia, Perth, WA 6009, Australia
- Correspondence: ; Tel.: +61-8-6319-1023
| | - Lauren Ursich
- Cancer Centre, Telethon Kids Institute, Perth, WA 6009, Australia
- School of Biomedical Sciences, University of Western Australia, Perth, WA 6009, Australia
| | - Emily V. Fletcher
- Cancer Centre, Telethon Kids Institute, Perth, WA 6009, Australia
- Centre for Child Health Research, University of Western Australia, Perth, WA 6009, Australia
| | - Terrance G. Johns
- Cancer Centre, Telethon Kids Institute, Perth, WA 6009, Australia
- Centre for Child Health Research, University of Western Australia, Perth, WA 6009, Australia
| |
Collapse
|
32
|
Hintzen G, Dulat HJ, Rajkovic E. Engaging innate immunity for targeting the epidermal growth factor receptor: Therapeutic options leveraging innate immunity versus adaptive immunity versus inhibition of signaling. Front Oncol 2022; 12:892212. [PMID: 36185288 PMCID: PMC9518002 DOI: 10.3389/fonc.2022.892212] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 07/28/2022] [Indexed: 12/15/2022] Open
Abstract
The epidermal growth factor receptor (EGFR) is a key player in the normal tissue physiology and the pathology of cancer. Therapeutic approaches have now been developed to target oncogenic genetic aberrations of EGFR, found in a subset of tumors, and to take advantage of overexpression of EGFR in tumors. The development of small-molecule inhibitors and anti-EGFR antibodies targeting EGFR activation have resulted in effective but limited treatment options for patients with mutated or wild-type EGFR-expressing cancers, while therapeutic approaches that deploy effectors of the adaptive or innate immune system are still undergoing development. This review discusses EGFR-targeting therapies acting through distinct molecular mechanisms to destroy EGFR-expressing cancer cells. The focus is on the successes and limitations of therapies targeting the activation of EGFR versus those that exploit the cytotoxic T cells and innate immune cells to target EGFR-expressing cancer cells. Moreover, we discuss alternative approaches that may have the potential to overcome limitations of current therapies; in particular the innate cell engagers are discussed. Furthermore, this review highlights the potential to combine innate cell engagers with immunotherapies, to maximize their effectiveness, or with unspecific cell therapies, to convert them into tumor-specific agents.
Collapse
|
33
|
In silico validation of RNA-Seq results can identify gene fusions with oncogenic potential in glioblastoma. Sci Rep 2022; 12:14439. [PMID: 36002559 PMCID: PMC9402576 DOI: 10.1038/s41598-022-18608-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 08/16/2022] [Indexed: 11/08/2022] Open
Abstract
RNA-Sequencing (RNA-Seq) can identify gene fusions in tumors, but not all these fusions have functional consequences. Using multiple data bases, we have performed an in silico analysis of fusions detected by RNA-Seq in tumor samples from 139 newly diagnosed glioblastoma patients to identify in-frame fusions with predictable oncogenic potential. Among 61 samples with fusions, there were 103 different fusions, involving 167 different genes, including 20 known oncogenes or tumor suppressor genes (TSGs), 16 associated with cancer but not oncogenes or TSGs, and 32 not associated with cancer but previously shown to be involved in fusions in gliomas. After selecting in-frame fusions able to produce a protein product and running Oncofuse, we identified 30 fusions with predictable oncogenic potential and classified them into four non-overlapping categories: six previously described in cancer; six involving an oncogene or TSG; four predicted by Oncofuse to have oncogenic potential; and 14 other in-frame fusions. Only 24 patients harbored one or more of these 30 fusions, and only two fusions were present in more than one patient: FGFR3::TACC3 and EGFR::SEPTIN14. This in silico study provides a good starting point for the identification of gene fusions with functional consequences in the pathogenesis or treatment of glioblastoma.
Collapse
|
34
|
Schwark K, Messinger D, Cummings JR, Bradin J, Kawakibi A, Babila CM, Lyons S, Ji S, Cartaxo RT, Kong S, Cantor E, Koschmann C, Yadav VN. Receptor tyrosine kinase (RTK) targeting in pediatric high-grade glioma and diffuse midline glioma: Pre-clinical models and precision medicine. Front Oncol 2022; 12:922928. [PMID: 35978801 PMCID: PMC9376238 DOI: 10.3389/fonc.2022.922928] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
Pediatric high-grade glioma (pHGG), including both diffuse midline glioma (DMG) and non-midline tumors, continues to be one of the deadliest oncologic diagnoses (both henceforth referred to as “pHGG”). Targeted therapy options aimed at key oncogenic receptor tyrosine kinase (RTK) drivers using small-molecule RTK inhibitors has been extensively studied, but the absence of proper in vivo modeling that recapitulate pHGG biology has historically been a research challenge. Thankfully, there have been many recent advances in animal modeling, including Cre-inducible transgenic models, as well as intra-uterine electroporation (IUE) models, which closely recapitulate the salient features of human pHGG tumors. Over 20% of pHGG have been found in sequencing studies to have alterations in platelet derived growth factor-alpha (PDGFRA), making growth factor modeling and inhibition via targeted tyrosine kinases a rich vein of interest. With commonly found alterations in other growth factors, including FGFR, EGFR, VEGFR as well as RET, MET, and ALK, it is necessary to model those receptors, as well. Here we review the recent advances in murine modeling and precision targeting of the most important RTKs in their clinical context. We additionally provide a review of current work in the field with several small molecule RTK inhibitors used in pre-clinical or clinical settings for treatment of pHGG.
Collapse
Affiliation(s)
- Kallen Schwark
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, University of Michigan School of Medicine, Ann Arbor, MI, United States
| | - Dana Messinger
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, University of Michigan School of Medicine, Ann Arbor, MI, United States
| | - Jessica R. Cummings
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, University of Michigan School of Medicine, Ann Arbor, MI, United States
| | - Joshua Bradin
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, University of Michigan School of Medicine, Ann Arbor, MI, United States
| | - Abed Kawakibi
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, University of Michigan School of Medicine, Ann Arbor, MI, United States
| | - Clarissa M. Babila
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, University of Michigan School of Medicine, Ann Arbor, MI, United States
| | - Samantha Lyons
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, University of Michigan School of Medicine, Ann Arbor, MI, United States
| | - Sunjong Ji
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, University of Michigan School of Medicine, Ann Arbor, MI, United States
| | - Rodrigo T. Cartaxo
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, University of Michigan School of Medicine, Ann Arbor, MI, United States
| | - Seongbae Kong
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, University of Michigan School of Medicine, Ann Arbor, MI, United States
| | - Evan Cantor
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, University of Michigan School of Medicine, Ann Arbor, MI, United States
| | - Carl Koschmann
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, University of Michigan School of Medicine, Ann Arbor, MI, United States
| | - Viveka Nand Yadav
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, University of Michigan School of Medicine, Ann Arbor, MI, United States
- Department of Pediatrics, Children's Mercy Research Institute (CMRI), Kansas, MO, United States
- Department of Pediatrics, University of Missouri Kansas City School of Medicine, Kansas, MO, United States
- *Correspondence: Viveka Nand Yadav,
| |
Collapse
|
35
|
Montella L, Del Gaudio N, Bove G, Cuomo M, Buonaiuto M, Costabile D, Visconti R, Facchini G, Altucci L, Chiariotti L, Della Monica R. Looking Beyond the Glioblastoma Mask: Is Genomics the Right Path? Front Oncol 2022; 12:926967. [PMID: 35875139 PMCID: PMC9306486 DOI: 10.3389/fonc.2022.926967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 06/09/2022] [Indexed: 11/15/2022] Open
Abstract
Glioblastomas are the most frequent and malignant brain tumor hallmarked by an invariably poor prognosis. They have been classically differentiated into primary isocitrate dehydrogenase 1 or 2 (IDH1 -2) wild-type (wt) glioblastoma (GBM) and secondary IDH mutant GBM, with IDH wt GBMs being commonly associated with older age and poor prognosis. Recently, genetic analyses have been integrated with epigenetic investigations, strongly implementing typing and subtyping of brain tumors, including GBMs, and leading to the new WHO 2021 classification. GBM genomic and epigenomic profile influences evolution, resistance, and therapeutic responses. However, differently from other tumors, there is a wide gap between the refined GBM profiling and the limited therapeutic opportunities. In addition, the different oncogenes and tumor suppressor genes involved in glial cell transformation, the heterogeneous nature of cancer, and the restricted access of drugs due to the blood–brain barrier have limited clinical advancements. This review will summarize the more relevant genetic alterations found in GBMs and highlight their potential role as potential therapeutic targets.
Collapse
Affiliation(s)
- Liliana Montella
- Oncology Operative Unit, "Santa Maria delle Grazie" Hospital, ASL Napoli 2 NORD-, Pozzuoli, Italy
| | - Nunzio Del Gaudio
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Napoli, Italy
| | - Guglielmo Bove
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Napoli, Italy
| | - Mariella Cuomo
- CEINGE Biotecnologie Avanzate scarl, Napoli, Italy.,Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", Napoli, Italy
| | - Michela Buonaiuto
- CEINGE Biotecnologie Avanzate scarl, Napoli, Italy.,Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", Napoli, Italy
| | - Davide Costabile
- CEINGE Biotecnologie Avanzate scarl, Napoli, Italy.,SEMM-European School of Molecular Medicine, Milano, Italy
| | - Roberta Visconti
- CEINGE Biotecnologie Avanzate scarl, Napoli, Italy.,Institute of Experimental Endocrinology and Oncology, Consiglio Nazionale delle Ricerche, Napoli, Italy
| | - Gaetano Facchini
- Oncology Operative Unit, "Santa Maria delle Grazie" Hospital, ASL Napoli 2 NORD-, Pozzuoli, Italy
| | - Lucia Altucci
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Napoli, Italy.,BIOGEM, Ariano Irpino, Italy
| | - Lorenzo Chiariotti
- CEINGE Biotecnologie Avanzate scarl, Napoli, Italy.,Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", Napoli, Italy
| | - Rosa Della Monica
- CEINGE Biotecnologie Avanzate scarl, Napoli, Italy.,Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", Napoli, Italy
| |
Collapse
|
36
|
Franco C, Kausar S, Silva MFB, Guedes RC, Falcao AO, Brito MA. Multi-Targeting Approach in Glioblastoma Using Computer-Assisted Drug Discovery Tools to Overcome the Blood–Brain Barrier and Target EGFR/PI3Kp110β Signaling. Cancers (Basel) 2022; 14:cancers14143506. [PMID: 35884571 PMCID: PMC9317902 DOI: 10.3390/cancers14143506] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 07/12/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Treatment of glioblastoma is hampered by the activation of compensatory survival mechanisms by malignant cells that lead to drug resistance. Moreover, the blood–brain barrier (BBB) precludes the brain entrance of most drugs. We hypothesized that computer-assisted drug discovery tools would reveal novel multi-targeting drug candidates with BBB-permeant and favorable ADMET properties. We aimed to discover molecules with predicted ability to inhibit the EGFR/PI3Kp110β pathway and to validate their efficacy and safety in biological assays. We used quantitative structure–activity relationship models and structure-based virtual screening, and assessed ADMET properties, to identify BBB-permeant drug candidates. Moreover, we tested their anti-tumor efficacy and BBB safety and permeation in cell models. We found two EGFR, two PI3Kp110β, and, mostly, two dual inhibitors with anti-tumor effects. Among them, one EGFR and two PI3Kp110β inhibitors were able to cross the BBB endothelium without compromising it. These studies revealed novel drug candidates for glioblastoma treatment. Abstract The epidermal growth factor receptor (EGFR) is upregulated in glioblastoma, becoming an attractive therapeutic target. However, activation of compensatory pathways generates inputs to downstream PI3Kp110β signaling, leading to anti-EGFR therapeutic resistance. Moreover, the blood–brain barrier (BBB) limits drugs’ brain penetration. We aimed to discover EGFR/PI3Kp110β pathway inhibitors for a multi-targeting approach, with favorable ADMET and BBB-permeant properties. We used quantitative structure–activity relationship models and structure-based virtual screening, and assessed ADMET properties, to identify BBB-permeant drug candidates. Predictions were validated in in vitro models of the human BBB and BBB-glioma co-cultures. The results disclosed 27 molecules (18 EGFR, 6 PI3Kp110β, and 3 dual inhibitors) for biological validation, performed in two glioblastoma cell lines (U87MG and U87MG overexpressing EGFR). Six molecules (two EGFR, two PI3Kp110β, and two dual inhibitors) decreased cell viability by 40–99%, with the greatest effect observed for the dual inhibitors. The glioma cytotoxicity was confirmed by analysis of targets’ downregulation and increased apoptosis (15–85%). Safety to BBB endothelial cells was confirmed for three of those molecules (one EGFR and two PI3Kp110β inhibitors). These molecules crossed the endothelial monolayer in the BBB in vitro model and in the BBB-glioblastoma co-culture system. These results revealed novel drug candidates for glioblastoma treatment.
Collapse
Affiliation(s)
- Catarina Franco
- LASIGE, Department of Informatics, Faculty of Sciences, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal; (C.F.); (S.K.)
- Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (M.F.B.S.); (R.C.G.)
| | - Samina Kausar
- LASIGE, Department of Informatics, Faculty of Sciences, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal; (C.F.); (S.K.)
- Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (M.F.B.S.); (R.C.G.)
| | - Margarida F. B. Silva
- Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (M.F.B.S.); (R.C.G.)
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Rita C. Guedes
- Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (M.F.B.S.); (R.C.G.)
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Andre O. Falcao
- LASIGE, Department of Informatics, Faculty of Sciences, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal; (C.F.); (S.K.)
- Correspondence: (A.O.F.); (M.A.B.); Tel.: +351-217500239 (A.O.F.); +351-217946449 (M.A.B.)
| | - Maria Alexandra Brito
- Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (M.F.B.S.); (R.C.G.)
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
- Correspondence: (A.O.F.); (M.A.B.); Tel.: +351-217500239 (A.O.F.); +351-217946449 (M.A.B.)
| |
Collapse
|
37
|
Rathi S, Griffith JI, Zhang W, Zhang W, Oh JH, Talele S, Sarkaria JN, Elmquist WF. The influence of the blood-brain barrier in the treatment of brain tumours. J Intern Med 2022; 292:3-30. [PMID: 35040235 DOI: 10.1111/joim.13440] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Brain tumours have a poor prognosis and lack effective treatments. The blood-brain barrier (BBB) represents a major hurdle to drug delivery to brain tumours. In some locations in the tumour, the BBB may be disrupted to form the blood-brain tumour barrier (BBTB). This leaky BBTB enables diagnosis of brain tumours by contrast enhanced magnetic resonance imaging; however, this disruption is heterogeneous throughout the tumour. Thus, relying on the disrupted BBTB for achieving effective drug concentrations in brain tumours has met with little clinical success. Because of this, it would be beneficial to design drugs and drug delivery strategies to overcome the 'normal' BBB to effectively treat the brain tumours. In this review, we discuss the role of BBB/BBTB in brain tumour diagnosis and treatment highlighting the heterogeneity of the BBTB. We also discuss various strategies to improve drug delivery across the BBB/BBTB to treat both primary and metastatic brain tumours. Recognizing that the BBB represents a critical determinant of drug efficacy in central nervous system tumours will allow a more rapid translation from basic science to clinical application. A more complete understanding of the factors, such as BBB-limited drug delivery, that have hindered progress in treating both primary and metastatic brain tumours, is necessary to develop more effective therapies.
Collapse
Affiliation(s)
- Sneha Rathi
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN, USA
| | - Jessica I Griffith
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN, USA
| | - Wenjuan Zhang
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN, USA
| | - Wenqiu Zhang
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN, USA
| | - Ju-Hee Oh
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN, USA
| | - Surabhi Talele
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN, USA
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, USA
| | - William F Elmquist
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
38
|
Goodman AL, Velázquez Vega JE, Glenn C, Olson JJ. Congress of neurological surgeons systematic review and evidence-based guidelines update on the role of neuropathology in the management of progressive glioblastoma in adults. J Neurooncol 2022; 158:179-224. [PMID: 35648306 DOI: 10.1007/s11060-022-04005-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Accepted: 04/04/2022] [Indexed: 12/28/2022]
Abstract
TARGET POPULATION These recommendations apply to adult patients with progressive or recurrent glioblastoma (GBM). QUESTION For adult patients with progressive glioblastoma does testing for Isocitrate Dehydrogenase (IDH) 1 or 2 mutations provide new additional management or prognostic information beyond that derived from the tumor at initial presentation? RECOMMENDATION Level III: Repeat IDH mutation testing is not necessary if the tumor is histologically similar to the primary tumor and the patient's clinical course is as expected. QUESTION For adult patients with progressive glioblastoma does repeat testing for MGMT promoter methylation provide new or additional management or prognostic information beyond that derived from the tumor at initial presentation and what methods of detection are optimal? RECOMMENDATION Level III: Repeat MGMT promoter methylation is not recommended. QUESTION For adult patients with progressive glioblastoma does EGFR amplification or mutation testing provide management or prognostic information beyond that provided by histologic analysis and if performed on previous tissue samples, does it need to be repeated? RECOMMENDATION Level III: In cases that are difficult to classify as glioblastoma on histologic features EGFR amplification testing may help in classification. If a previous EGFR amplification was detected, repeat testing is not necessary. Repeat EGFR amplification or mutational testing may be recommended in patients in which target therapy is being considered. QUESTION For adult patients with progressive glioblastoma does large panel or whole genome sequencing provide management or prognostic information beyond that derived from histologic analysis? RECOMMENDATION Level III: Primary or repeat large panel or whole genome sequencing may be considered in patients who are eligible or interested in molecularly guided therapy or clinical trials. QUESTION For adult patients with progressive glioblastoma should immune checkpoint biomarker testing be performed to provide management and prognostic information beyond that obtained from histologic analysis? RECOMMENDATION Level III: The current evidence does not support making PD-L1 or mismatch repair (MMR) enzyme activity a component of standard testing. QUESTION For adult patients with progressive glioblastoma are there meaningful biomarkers for bevacizumab responsiveness and does their assessment provide additional information for tumor management and prognosis beyond that learned by standard histologic analysis? RECOMMENDATION Level III: No established Bevacizumab biomarkers are currently available based upon the inclusion criteria of this guideline.
Collapse
Affiliation(s)
- Abigail L Goodman
- Carolinas Pathology, Atrium Health Carolinas Medical Center, Charlotte, NC, USA.
| | - José E Velázquez Vega
- Department of Pathology and Laboratory Medicine, Children's Healthcare of Atlanta and Emory University, Atlanta, GA, USA
| | - Chad Glenn
- Department of Neurosurgery, Stephenson Cancer Center, The University of Oklahoma, Oklahoma City, OK, USA
| | - Jeffrey J Olson
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
39
|
Gatto L, Di Nunno V, Franceschi E, Tosoni A, Bartolini S, Brandes AA. Pharmacotherapeutic Treatment of Glioblastoma: Where Are We to Date? Drugs 2022; 82:491-510. [PMID: 35397073 DOI: 10.1007/s40265-022-01702-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/08/2022] [Indexed: 12/30/2022]
Abstract
The clinical management of glioblastoma (GBM) is still bereft of treatments able to significantly improve the poor prognosis of the disease. Despite the extreme clinical need for novel therapeutic drugs, only a small percentage of patients with GBM benefit from inclusion in a clinical trial. Moreover, often clinical studies do not lead to final interpretable conclusions. From the mistakes and negative results obtained in the last years, we are now able to plan a novel generation of clinical studies for patients with GBM, allowing the testing of multiple anticancer agents at the same time. This assumes critical importance, considering that, thanks to improved knowledge of altered molecular mechanisms related to the disease, we are now able to propose several potential effective compounds in patients with both newly diagnosed and recurrent GBM. Among the novel compounds assessed, the initially great enthusiasm toward trials employing immune checkpoint inhibitors (ICIs) was disappointing due to the negative results that emerged in three randomized phase III trials. However, novel biological insights into the disease suggest that immunotherapy can be a convincing and effective treatment in GBM even if ICIs failed to prolong the survival of these patients. In this regard, the most promising approach consists of engineered immune cells such as chimeric antigen receptor (CAR) T, CAR M, and CAR NK alone or in combination with other treatments. In this review, we discuss several issues related to systemic treatments in GBM patients. First, we assess critical issues toward the planning of clinical trials and the strategies employed to overcome these obstacles. We then move on to the most relevant interventional studies carried out on patients with previously untreated (newly diagnosed) GBM and those with recurrent and pretreated disease. Finally, we investigate novel immunotherapeutic approaches with special emphasis on preclinical and clinical data related to the administration of engineered immune cells in GBM.
Collapse
Affiliation(s)
- Lidia Gatto
- Department of Oncology, AUSL Bologna, Bologna, Italy
| | | | - Enrico Franceschi
- Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, Via Altura 3, Bologna, Italy.
| | - Alicia Tosoni
- Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, Via Altura 3, Bologna, Italy
| | - Stefania Bartolini
- Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, Via Altura 3, Bologna, Italy
| | - Alba Ariela Brandes
- Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, Via Altura 3, Bologna, Italy
| |
Collapse
|
40
|
Li S, Zhang N, Liu S, Zhang H, Liu J, Qi Y, Zhang Q, Li X. ITGA5 Is a Novel Oncogenic Biomarker and Correlates With Tumor Immune Microenvironment in Gliomas. Front Oncol 2022; 12:844144. [PMID: 35371978 PMCID: PMC8971292 DOI: 10.3389/fonc.2022.844144] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 02/17/2022] [Indexed: 12/12/2022] Open
Abstract
Gliomas are the most aggressive primary intracranial malignancies with poor overall survival. ITGA5 is one member of the integrin adhesion molecule family and is implicated in cancer metastasis and oncogenesis. However, few studies have explored the association between tumor immune microenvironment and ITGA5 expression level in gliomas. Firstly, we analyzed 3,047 glioma patient samples collected from the TCGA, the CGGA, and the GEO databases, proving that high ITGA5 expression positively related to aggressive clinicopathological features and poor survival in glioma patients. Then, based on the ITGA5 level, immunological characteristics and genomic alteration were explored through multiple algorithms. We observed that ITGA5 was involved in pivotal oncological pathways, immune-related processes, and distinct typical genomic alterations in gliomas. Notably, ITGA5 was found to engage in remolding glioma immune infiltration and immune microenvironment, manifested by higher immune cell infiltration when ITGA5 is highly expressed. We also demonstrated a strong correlation between ITGA5 and immune checkpoint molecules that may be beneficial from immune checkpoint blockade strategies. In addition, ITGA5 was found to be a robust and sensitive indicator for plenty of chemotherapy drugs through drug sensitivity prediction. Altogether, our comprehensive analyses deciphered the prognostic, immunological, and therapeutic value of ITGA5 in glioma, thus improving individual and precise therapy for combating gliomas.
Collapse
Affiliation(s)
- Shuyu Li
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Nan Zhang
- One-Third Lab, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Shiyang Liu
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hao Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Jiajing Liu
- Department of Neurology, Affiliated People’s Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yiwei Qi
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Qi Zhang
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Xingrui Li
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
41
|
Picca A, Guyon D, Santonocito OS, Baldini C, Idbaih A, Carpentier A, Naccarato AG, Caccese M, Lombardi G, Di Stefano AL. Innovating Strategies and Tailored Approaches in Neuro-Oncology. Cancers (Basel) 2022; 14:1124. [PMID: 35267432 PMCID: PMC8909701 DOI: 10.3390/cancers14051124] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/11/2022] [Accepted: 02/18/2022] [Indexed: 01/25/2023] Open
Abstract
Diffuse gliomas, the most frequent and aggressive primary central nervous system neoplasms, currently lack effective curative treatments, particularly for cases lacking the favorable prognostic marker IDH mutation. Nonetheless, advances in molecular biology allowed to identify several druggable alterations in a subset of IDH wild-type gliomas, such as NTRK and FGFR-TACC fusions, and BRAF hotspot mutations. Multi-tyrosine kinase inhibitors, such as regorafenib, also showed efficacy in the setting of recurrent glioblastoma. IDH inhibitors are currently in the advanced phase of clinical evaluation for patients with IDH-mutant gliomas. Several immunotherapeutic approaches, such as tumor vaccines or checkpoint inhibitors, failed to improve patients' outcomes. Even so, they may be still beneficial in a subset of them. New methods, such as using pulsed ultrasound to disrupt the blood-brain barrier, gene therapy, and oncolytic virotherapy, are well tolerated and may be included in the therapeutic armamentarium soon.
Collapse
Affiliation(s)
- Alberto Picca
- Institut du Cerveau-Paris Brain Institute-ICM, Sorbonne Université, Inserm, CNRS, AP-HP, Hôpital Universitaire La Pitié Salpêtrière, DMU Neurosciences, 75013 Paris, France; (A.P.); (A.I.)
| | - David Guyon
- Department of Medical Oncology, Gustave Roussy University Hospital, 94800 Villejuif, France;
| | - Orazio Santo Santonocito
- Division of Neurosurgery, Spedali Riuniti di Livorno—USL Toscana Nord-Ovest, 57124 Livorno, Italy;
| | - Capucine Baldini
- Drug Development Department (DITEP), Gustave Roussy University Hospital, 94800 Villejuif, France;
| | - Ahmed Idbaih
- Institut du Cerveau-Paris Brain Institute-ICM, Sorbonne Université, Inserm, CNRS, AP-HP, Hôpital Universitaire La Pitié Salpêtrière, DMU Neurosciences, 75013 Paris, France; (A.P.); (A.I.)
| | - Alexandre Carpentier
- Service de Neurochirurgie, Hôpital Universitaire La Pitié Salpêtrière, 75013 Paris, France;
| | - Antonio Giuseppe Naccarato
- Department of Translational Research and New Technologies in Medicine and Surgery, Division of Pathology, University of Pisa, 56100 Pisa, Italy;
- Anatomia Patologica 1, Department of Laboratory Medicine, Pisa University Hospital, 56126 Pisa, Italy
| | - Mario Caccese
- Department of Oncology, Oncology 1, Veneto Institute of Oncology IOV-IRCCS, 35128 Padua, Italy; (M.C.); (G.L.)
| | - Giuseppe Lombardi
- Department of Oncology, Oncology 1, Veneto Institute of Oncology IOV-IRCCS, 35128 Padua, Italy; (M.C.); (G.L.)
| | - Anna Luisa Di Stefano
- Division of Neurosurgery, Spedali Riuniti di Livorno—USL Toscana Nord-Ovest, 57124 Livorno, Italy;
- Department of Neurology, Foch Hospital, 92150 Suresnes, France
| |
Collapse
|
42
|
Yang K, Wu Z, Zhang H, Zhang N, Wu W, Wang Z, Dai Z, Zhang X, Zhang L, Peng Y, Ye W, Zeng W, Liu Z, Cheng Q. Glioma targeted therapy: insight into future of molecular approaches. Mol Cancer 2022; 21:39. [PMID: 35135556 PMCID: PMC8822752 DOI: 10.1186/s12943-022-01513-z] [Citation(s) in RCA: 406] [Impact Index Per Article: 135.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 01/12/2022] [Indexed: 12/13/2022] Open
Abstract
Gliomas are the common type of brain tumors originating from glial cells. Epidemiologically, gliomas occur among all ages, more often seen in adults, which males are more susceptible than females. According to the fifth edition of the WHO Classification of Tumors of the Central Nervous System (WHO CNS5), standard of care and prognosis of gliomas can be dramatically different. Generally, circumscribed gliomas are usually benign and recommended to early complete resection, with chemotherapy if necessary. Diffuse gliomas and other high-grade gliomas according to their molecule subtype are slightly intractable, with necessity of chemotherapy. However, for glioblastoma, feasible resection followed by radiotherapy plus temozolomide chemotherapy define the current standard of care. Here, we discuss novel feasible or potential targets for treatment of gliomas, especially IDH-wild type glioblastoma. Classic targets such as the p53 and retinoblastoma (RB) pathway and epidermal growth factor receptor (EGFR) gene alteration have met failure due to complex regulatory network. There is ever-increasing interest in immunotherapy (immune checkpoint molecule, tumor associated macrophage, dendritic cell vaccine, CAR-T), tumor microenvironment, and combination of several efficacious methods. With many targeted therapy options emerging, biomarkers guiding the prescription of a particular targeted therapy are also attractive. More pre-clinical and clinical trials are urgently needed to explore and evaluate the feasibility of targeted therapy with the corresponding biomarkers for effective personalized treatment options.
Collapse
Affiliation(s)
- Keyang Yang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhijing Wu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Hao Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Nan Zhang
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- One-Third Lab, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Wantao Wu
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Zeyu Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Ziyu Dai
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xun Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Liyang Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yun Peng
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
- Teaching and Research Section of Clinical Nursing, Xiangya Hospital of Central South University, Changsha, China
| | - Weijie Ye
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
| | - Wenjing Zeng
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
| | - Zhixiong Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
43
|
Law M, Grayling MJ, Mander AP. A stochastically curtailed single‐arm phase II trial design for binary outcomes. J Biopharm Stat 2022; 32:671-691. [PMID: 35077268 PMCID: PMC7614398 DOI: 10.1080/10543406.2021.2009498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Phase II clinical trials are a critical aspect of the drug development process. With drug development costs ever increasing, novel designs that can improve the efficiency of phase II trials are extremely valuable.Phase II clinical trials for cancer treatments often measure a binary outcome. The final trial decision is generally to continue or cease development. When this decision is based solely on the result of a hypothesis test, the result may be known with certainty before the planned end of the trial. Unfortunately, there is often no opportunity for early stopping when this occurs.Some existing designs do permit early stopping in this case, accordingly reducing the required sample size and potentially speeding up drug development. However, more improvements can be achieved by stopping early when the final trial decision is very likely, rather than certain, known as stochastic curtailment. While some authors have proposed approaches of this form, these approaches have various limitations.In this work we address these limitations by proposing new design approaches for single-arm phase II binary outcome trials that use stochastic curtailment. We use exact distributions, avoid simulation, consider a wider range of possible designs and permit early stopping for promising treatments. As a result, we are able to obtain trial designs that have considerably reduced sample sizes on average.
Collapse
Affiliation(s)
- Martin Law
- Hub for Trials Methodology Research, Medical Research Council Biostatistics Unit, University of Cambridge, Cambridge, UK
- Papworth Trials Unit Collaboration, Royal Papworth Hospital, Cambridge, UK
| | - Michael J. Grayling
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Adrian P. Mander
- College of Biomedical and Life Sciences, Cardiff University, Cardiff, UK
| |
Collapse
|
44
|
Qin A, Musket A, Musich PR, Schweitzer JB, Xie Q. Receptor tyrosine kinases as druggable targets in glioblastoma: Do signaling pathways matter? Neurooncol Adv 2021; 3:vdab133. [PMID: 34806012 PMCID: PMC8598918 DOI: 10.1093/noajnl/vdab133] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Glioblastoma (GBM) is the most malignant primary brain tumor without effective therapies. Since bevacizumab was FDA approved for targeting vascular endothelial growth factor receptor 2 (VEGFR2) in adult patients with recurrent GBM, targeted therapy against receptor tyrosine kinases (RTKs) has become a new avenue for GBM therapeutics. In addition to VEGFR, the epidermal growth factor receptor (EGFR), platelet-derived growth factor receptor (PDGFR), hepatocyte growth factor receptor (HGFR/MET), and fibroblast growth factor receptor (FGFR) are major RTK targets. However, results from clinical Phase II/III trials indicate that most RTK-targeting therapeutics including tyrosine kinase inhibitors (TKIs) and neutralizing antibodies lack clinical efficacy, either alone or in combination. The major challenge is to uncover the genetic RTK alterations driving GBM initiation and progression, as well as to elucidate the mechanisms toward therapeutic resistance. In this review, we will discuss the genetic alterations in these 5 commonly targeted RTKs, the clinical trial outcomes of the associated RTK-targeting therapeutics, and the potential mechanisms toward the resistance. We anticipate that future design of new clinical trials with combination strategies, based on the genetic alterations within an individual patient’s tumor and mechanisms contributing to therapeutic resistance after treatment, will achieve durable remissions and improve outcomes in GBM patients.
Collapse
Affiliation(s)
- Anna Qin
- Department of Biomedical Science, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA
| | - Anna Musket
- Department of Biomedical Science, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA
| | - Phillip R Musich
- Department of Biomedical Science, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA
| | - John B Schweitzer
- Department of Pathology, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA
| | - Qian Xie
- Department of Biomedical Science, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA
| |
Collapse
|
45
|
Domènech M, Hernández A, Plaja A, Martínez-Balibrea E, Balañà C. Hypoxia: The Cornerstone of Glioblastoma. Int J Mol Sci 2021; 22:12608. [PMID: 34830491 PMCID: PMC8620858 DOI: 10.3390/ijms222212608] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 11/19/2021] [Accepted: 11/19/2021] [Indexed: 12/16/2022] Open
Abstract
Glioblastoma is the most aggressive form of brain tumor in adults and is characterized by the presence of hypervascularization and necrosis, both caused by a hypoxic microenvironment. In this review, we highlight that hypoxia-induced factor 1 (HIF-1), the main factor activated by hypoxia, is an important driver of tumor progression in GB patients. HIF-1α is a transcription factor regulated by the presence or absence of O2. The expression of HIF-1 has been related to high-grade gliomas and aggressive tumor behavior. HIF-1 promotes tumor progression via the activation of angiogenesis, immunosuppression, and metabolic reprogramming, promoting cell invasion and survival. Moreover, in GB, HIF-1 is not solely modulated by oxygen but also by oncogenic signaling pathways, such as MAPK/ERK, p53, and PI3K/PTEN. Therefore, the inhibition of the hypoxia pathway could represent an important treatment alternative in a disease with very few therapy options. Here, we review the roles of HIF-1 in GB progression and the inhibitors that have been studied thus far, with the aim of shedding light on this devastating disease.
Collapse
Affiliation(s)
- Marta Domènech
- B·ARGO (Badalona Applied Research Group of Oncology) Medical Oncology Department, Catalan Institute of Oncology Badalona, 08916 Badalona, Spain; (M.D.); (A.H.); (A.P.)
| | - Ainhoa Hernández
- B·ARGO (Badalona Applied Research Group of Oncology) Medical Oncology Department, Catalan Institute of Oncology Badalona, 08916 Badalona, Spain; (M.D.); (A.H.); (A.P.)
| | - Andrea Plaja
- B·ARGO (Badalona Applied Research Group of Oncology) Medical Oncology Department, Catalan Institute of Oncology Badalona, 08916 Badalona, Spain; (M.D.); (A.H.); (A.P.)
| | - Eva Martínez-Balibrea
- Germans Trias i Pujol Research Institute (IGTP), ProCURE Program, Catalan Institute of Oncology, 08916 Badalona, Spain;
| | - Carmen Balañà
- B·ARGO (Badalona Applied Research Group of Oncology) Medical Oncology Department, Catalan Institute of Oncology Badalona, 08916 Badalona, Spain; (M.D.); (A.H.); (A.P.)
| |
Collapse
|
46
|
Di Nunno V, Franceschi E, Tosoni A, Gatto L, Lodi R, Bartolini S, Brandes AA. Glioblastoma: Emerging Treatments and Novel Trial Designs. Cancers (Basel) 2021; 13:cancers13153750. [PMID: 34359651 PMCID: PMC8345198 DOI: 10.3390/cancers13153750] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/19/2021] [Accepted: 07/21/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Nowadays, very few systemic agents have shown clinical activity in patients with glioblastoma, making the research of novel therapeutic approaches a critical issue. Fortunately, the availability of novel compounds is increasing thanks to better biological knowledge of the disease. In this review we want to investigate more promising ongoing clinical trials in both primary and recurrent GBM. Furthermore, a great interest of the present work is focused on novel trial design strategies. Abstract Management of glioblastoma is a clinical challenge since very few systemic treatments have shown clinical efficacy in recurrent disease. Thanks to an increased knowledge of the biological and molecular mechanisms related to disease progression and growth, promising novel treatment strategies are emerging. The expanding availability of innovative compounds requires the design of a new generation of clinical trials, testing experimental compounds in a short time and tailoring the sample cohort based on molecular and clinical behaviors. In this review, we focused our attention on the assessment of promising novel treatment approaches, discussing novel trial design and possible future fields of development in this setting.
Collapse
Affiliation(s)
- Vincenzo Di Nunno
- Department of Oncology, AUSL Bologna, Via Altura 3, 40139 Bologna, Italy; (E.F.); (A.T.); (L.G.); (S.B.); (A.A.B.)
- Correspondence: ; Tel.: +39-0516225697
| | - Enrico Franceschi
- Department of Oncology, AUSL Bologna, Via Altura 3, 40139 Bologna, Italy; (E.F.); (A.T.); (L.G.); (S.B.); (A.A.B.)
| | - Alicia Tosoni
- Department of Oncology, AUSL Bologna, Via Altura 3, 40139 Bologna, Italy; (E.F.); (A.T.); (L.G.); (S.B.); (A.A.B.)
| | - Lidia Gatto
- Department of Oncology, AUSL Bologna, Via Altura 3, 40139 Bologna, Italy; (E.F.); (A.T.); (L.G.); (S.B.); (A.A.B.)
| | - Raffaele Lodi
- Istituto delle Scienze Neurologiche di Bologna, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 40139 Bologna, Italy;
| | - Stefania Bartolini
- Department of Oncology, AUSL Bologna, Via Altura 3, 40139 Bologna, Italy; (E.F.); (A.T.); (L.G.); (S.B.); (A.A.B.)
| | - Alba Ariela Brandes
- Department of Oncology, AUSL Bologna, Via Altura 3, 40139 Bologna, Italy; (E.F.); (A.T.); (L.G.); (S.B.); (A.A.B.)
| |
Collapse
|
47
|
Kovalenko TF, Larionova TD, Antipova NV, Shakhparonov MI, Pavlyukov MS. The Role of Non-coding RNAs in the Pathogenesis of Glial Tumors. Acta Naturae 2021; 13:38-51. [PMID: 34707896 PMCID: PMC8526181 DOI: 10.32607/actanaturae.11270] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 03/15/2021] [Indexed: 11/25/2022] Open
Abstract
Among the many malignant neoplasms, glioblastoma (GBM) leads to one of the worst prognosis for patients and has an almost 100% recurrence rate. The only chemotherapeutic drug that is widely used for treating glioblastoma is temozolomide, a DNA alkylating agent. Its impact, however, is only minor; it increases patients' survival just by 12 to 14 months. Multiple highly selective compounds that affect specific proteins and have performed well in other types of cancer have proved ineffective against glioblastoma. Hence, there is an urgent need for novel methods that could help achieve the long-awaited progress in glioblastoma treatment. One of the potentially promising approaches is the targeting of non-coding RNAs (ncRNAs). These molecules are characterized by extremely high multifunctionality and often act as integrators by coordinating multiple key signaling pathways within the cell. Thus, the impact on ncRNAs has the potential to lead to a broader and stronger impact on cells, as opposed to the more focused action of inhibitors targeting specific proteins. In this review, we summarize the functions of long noncoding RNAs, circular RNAs, as well as microRNAs, PIWI-interacting RNAs, small nuclear and small nucleolar RNAs. We provide a classification of these transcripts and describe their role in various signaling pathways and physiological processes. We also provide examples of oncogenic and tumor suppressor ncRNAs belonging to each of these classes in the context of their involvement in the pathogenesis of gliomas and glioblastomas. In conclusion, we considered the potential use of ncRNAs as diagnostic markers and therapeutic targets for the treatment of glioblastoma.
Collapse
Affiliation(s)
- T. F. Kovalenko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry Russian Academy of Sciences, Moscow, 117997 Russia
| | - T. D. Larionova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry Russian Academy of Sciences, Moscow, 117997 Russia
| | - N. V. Antipova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry Russian Academy of Sciences, Moscow, 117997 Russia
| | - M. I. Shakhparonov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry Russian Academy of Sciences, Moscow, 117997 Russia
| | - M. S. Pavlyukov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry Russian Academy of Sciences, Moscow, 117997 Russia
| |
Collapse
|
48
|
Bolcaen J, Nair S, Driver CHS, Boshomane TMG, Ebenhan T, Vandevoorde C. Novel Receptor Tyrosine Kinase Pathway Inhibitors for Targeted Radionuclide Therapy of Glioblastoma. Pharmaceuticals (Basel) 2021; 14:626. [PMID: 34209513 PMCID: PMC8308832 DOI: 10.3390/ph14070626] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/18/2021] [Accepted: 06/21/2021] [Indexed: 12/15/2022] Open
Abstract
Glioblastoma (GB) remains the most fatal brain tumor characterized by a high infiltration rate and treatment resistance. Overexpression and/or mutation of receptor tyrosine kinases is common in GB, which subsequently leads to the activation of many downstream pathways that have a critical impact on tumor progression and therapy resistance. Therefore, receptor tyrosine kinase inhibitors (RTKIs) have been investigated to improve the dismal prognosis of GB in an effort to evolve into a personalized targeted therapy strategy with a better treatment outcome. Numerous RTKIs have been approved in the clinic and several radiopharmaceuticals are part of (pre)clinical trials as a non-invasive method to identify patients who could benefit from RTKI. The latter opens up the scope for theranostic applications. In this review, the present status of RTKIs for the treatment, nuclear imaging and targeted radionuclide therapy of GB is presented. The focus will be on seven tyrosine kinase receptors, based on their central role in GB: EGFR, VEGFR, MET, PDGFR, FGFR, Eph receptor and IGF1R. Finally, by way of analyzing structural and physiological characteristics of the TKIs with promising clinical trial results, four small molecule RTKIs were selected based on their potential to become new therapeutic GB radiopharmaceuticals.
Collapse
Affiliation(s)
- Julie Bolcaen
- Radiobiology, Radiation Biophysics Division, Nuclear Medicine Department, iThemba LABS, Cape Town 7131, South Africa;
| | - Shankari Nair
- Radiobiology, Radiation Biophysics Division, Nuclear Medicine Department, iThemba LABS, Cape Town 7131, South Africa;
| | - Cathryn H. S. Driver
- Radiochemistry, South African Nuclear Energy Corporation, Pelindaba, Brits 0240, South Africa;
- Pre-Clinical Imaging Facility, Nuclear Medicine Research Infrastructure, Pelindaba, Brits 0242, South Africa;
| | - Tebatso M. G. Boshomane
- Department of Nuclear Medicine, University of Pretoria Steve Biko Academic Hospital, Pretoria 0001, South Africa;
| | - Thomas Ebenhan
- Pre-Clinical Imaging Facility, Nuclear Medicine Research Infrastructure, Pelindaba, Brits 0242, South Africa;
- Department of Nuclear Medicine, University of Pretoria Steve Biko Academic Hospital, Pretoria 0001, South Africa;
- Preclinical Drug Development Platform, Department of Science and Technology, North West University, Potchefstroom 2520, South Africa
| | - Charlot Vandevoorde
- Radiobiology, Radiation Biophysics Division, Nuclear Medicine Department, iThemba LABS, Cape Town 7131, South Africa;
| |
Collapse
|
49
|
Halatsch ME, Kast RE, Karpel-Massler G, Mayer B, Zolk O, Schmitz B, Scheuerle A, Maier L, Bullinger L, Mayer-Steinacker R, Schmidt C, Zeiler K, Elshaer Z, Panther P, Schmelzle B, Hallmen A, Dwucet A, Siegelin MD, Westhoff MA, Beckers K, Bouche G, Heiland T. A phase Ib/IIa trial of 9 repurposed drugs combined with temozolomide for the treatment of recurrent glioblastoma: CUSP9v3. Neurooncol Adv 2021; 3:vdab075. [PMID: 34377985 PMCID: PMC8349180 DOI: 10.1093/noajnl/vdab075] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Background The dismal prognosis of glioblastoma (GBM) may be related to the ability of GBM cells to develop mechanisms of treatment resistance. We designed a protocol called Coordinated Undermining of Survival Paths combining 9 repurposed non-oncological drugs with metronomic temozolomide—version 3—(CUSP9v3) to address this issue. The aim of this phase Ib/IIa trial was to assess the safety of CUSP9v3. Methods Ten adults with histologically confirmed GBM and recurrent or progressive disease were included. Treatment consisted of aprepitant, auranofin, celecoxib, captopril, disulfiram, itraconazole, minocycline, ritonavir, and sertraline added to metronomic low-dose temozolomide. Treatment was continued until toxicity or progression. Primary endpoint was dose-limiting toxicity defined as either any unmanageable grade 3–4 toxicity or inability to receive at least 7 of the 10 drugs at ≥ 50% of the per-protocol doses at the end of the second treatment cycle. Results One patient was not evaluable for the primary endpoint (safety). All 9 evaluable patients met the primary endpoint. Ritonavir, temozolomide, captopril, and itraconazole were the drugs most frequently requiring dose modification or pausing. The most common adverse events were nausea, headache, fatigue, diarrhea, and ataxia. Progression-free survival at 12 months was 50%. Conclusions CUSP9v3 can be safely administered in patients with recurrent GBM under careful monitoring. A randomized phase II trial is in preparation to assess the efficacy of the CUSP9v3 regimen in GBM.
Collapse
Affiliation(s)
| | | | | | - Benjamin Mayer
- Institute for Epidemiology and Medical Biometry, Ulm University, Ulm, Germany
| | - Oliver Zolk
- Department of Clinical Pharmacology, Ulm University Hospital, Ulm, Germany
| | - Bernd Schmitz
- Division of Neuroradiology, Department of Diagnostic and Interventional Radiology, Ulm University Hospital, Ulm, Germany
| | - Angelika Scheuerle
- Division of Neuropathology, Department of Pathology, Ulm University Hospital, Ulm, Germany
| | - Ludwig Maier
- Central Pharmacy, Ulm University Hospital, Ulm, Germany
| | - Lars Bullinger
- Division of Hematology and Oncology, Department of Internal Medicine, Ulm University Hospital, Ulm, Germany
| | - Regine Mayer-Steinacker
- Division of Hematology and Oncology, Department of Internal Medicine, Ulm University Hospital, Ulm, Germany
| | - Carl Schmidt
- Department of Neurosurgery, Ulm University Hospital, Ulm, Germany
| | - Katharina Zeiler
- Department of Neurosurgery, Ulm University Hospital, Ulm, Germany
| | - Ziad Elshaer
- Department of Neurosurgery, Ulm University Hospital, Ulm, Germany
| | - Patricia Panther
- Department of Neurosurgery, Ulm University Hospital, Ulm, Germany
| | - Birgit Schmelzle
- Institute of Experimental Cancer Research, Ulm University Hospital, Ulm, Germany
| | - Anke Hallmen
- Division of Hematology and Oncology, Department of Internal Medicine, Ulm University Hospital, Ulm, Germany
| | - Annika Dwucet
- Department of Neurosurgery, Ulm University Hospital, Ulm, Germany
| | - Markus D Siegelin
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York, USA
| | - Mike-Andrew Westhoff
- Department of Pediatric and Adolescent Medicine, Basic Research Division, Ulm University Hospital, Ulm, Germany
| | | | | | - Tim Heiland
- Department of Neurosurgery, Ulm University Hospital, Ulm, Germany
| |
Collapse
|
50
|
Abousaud M, Faroqui NM, Lesser G, Strowd RE, Ramkissoon SH, Kwatra M, Houston KS, Hsu FC, Carter A, Petro R, DeTroye AT. Clinical Experience using Osimertinib in Patients with Recurrent Malignant Gliomas Containing EGFR Alterations. JOURNAL OF CANCER SCIENCE AND CLINICAL THERAPEUTICS 2021; 5:210-220. [PMID: 35601813 PMCID: PMC9121798 DOI: 10.26502/jcsct.5079114] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Background EGFR alterations are commonly observed in malignant gliomas (MG). Osimertinib, an irreversible EGFR-tyrosine kinase inhibitor, effectively penetrates the blood brain barrier and achieves therapeutic concentrations in brain tissue. Materials and Methods This retrospective chart review identified six patients with recurrent MG and EGFR alterations who received osimertinib. Results Four patients were assessed for response. One patient had a partial response, two patients achieved stable disease and one was refractory. One patient with an EGFR vIII rearrangement remained on treatment for 236 days and a second patient with an EGFR vIII mutation remained on treatment for 294 days and continued on treatment at the time of analysis. Thrombocytopenia occurred in two patients, one patient developed grade 1 diarrhea and pneumonia, and another patient developed grade 1 mucositis. Conclusion Osimertinib had a tolerable safety profile in this heavily pretreated brain tumor population. Osimertinib may benefit select patients with recurrent MG containing EGFR alterations.
Collapse
Affiliation(s)
- Marin Abousaud
- Department of Pharmacy, Emory Healthcare, Atlanta, GA, USA
| | - Naqeeb M Faroqui
- Department of General Surgery, Wellstar Atlanta Medical Center, Atlanta, GA, USA
| | - Glenn Lesser
- Wake Forest Baptist Health Comprehensive Cancer Center, Department of Internal Medicine, Section on Hematology and Oncology, Winston-Salem, NC, USA
| | - Roy E Strowd
- Wake Forest Baptist Health Comprehensive Cancer Center, Department of Internal Medicine, Section on Hematology and Oncology, Winston-Salem, NC, USA
| | - Shakti H Ramkissoon
- Wake Forest Baptist Health Comprehensive Cancer Center, Department of Pathology, Winston-Salem, NC, USA
- Foundation Medicine, Morrisville, NC, USA
| | - Madan Kwatra
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Kristin S Houston
- Wake Forest Baptist Health Comprehensive Cancer Center, Department of Internal Medicine, Section on Hematology and Oncology, Winston-Salem, NC, USA
| | - Fang-Chi Hsu
- Department of Biostatistics and Data Science, Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Annette Carter
- Wake Forest Baptist Health Comprehensive Cancer Center, Department of Internal Medicine, Section on Hematology and Oncology, Winston-Salem, NC, USA
| | - Robin Petro
- Wake Forest Baptist Health Comprehensive Cancer Center, Department of Internal Medicine, Section on Hematology and Oncology, Winston-Salem, NC, USA
| | - Alisha T DeTroye
- Wake Forest Baptist Health Comprehensive Cancer Center, Department of Internal Medicine, Section on Hematology and Oncology, Winston-Salem, NC, USA
| |
Collapse
|