1
|
Tang Q, Ren T, Bai P, Wang X, Zhao L, Zhong R, Sun G. Novel strategies to overcome chemoresistance in human glioblastoma. Biochem Pharmacol 2024; 230:116588. [PMID: 39461382 DOI: 10.1016/j.bcp.2024.116588] [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: 06/30/2024] [Revised: 10/16/2024] [Accepted: 10/21/2024] [Indexed: 10/29/2024]
Abstract
Temozolomide (TMZ) is currently the first-line chemotherapeutic agent for the treatment of glioblastoma multiforme (GBM). However, the inherent heterogeneity of GBM often results in suboptimal outcomes, particularly due to varying degrees of resistance to TMZ. Over the past several decades, O6-methylguanine-DNA methyltransferase (MGMT)-mediated DNA repair pathway has been extensively investigated as a target to overcome TMZ resistance. Nonetheless, the combination of small molecule covalent MGMT inhibitors with TMZ and other chemotherapeutic agents has frequently led to adverse clinical effects. Recently, additional mechanisms contributing to TMZ resistance have been identified, including epidermal growth factor receptor (EGFR) mutations, overactivation of intracellular signalling pathways, energy metabolism reprogramming or survival autophagy, and changes in tumor microenvironment (TME). These findings suggest that novel therapeutic strategies targeting these mechanisms hold promise for overcoming TMZ resistance in GBM patients. In this review, we summarize the latest advancements in understanding the mechanisms underlying intrinsic and acquired TMZ resistance. Additionally, we compile various small-molecule compounds with potential to mitigate chemoresistance in GBM. These mechanism-based compounds may enhance the sensitivity of GBM to TMZ and related chemotherapeutic agents, thereby improving overall survival rates in clinical practice.
Collapse
Affiliation(s)
- Qing Tang
- Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Ting Ren
- Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Peiying Bai
- Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Xin Wang
- Department of Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100029, China
| | - Lijiao Zhao
- Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Rugang Zhong
- Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Guohui Sun
- Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China.
| |
Collapse
|
2
|
Saqib M, Zahoor A, Rahib A, Shamim A, Mumtaz H. Clinical and translational advances in primary brain tumor therapy with a focus on glioblastoma-A comprehensive review of the literature. World Neurosurg X 2024; 24:100399. [PMID: 39386927 PMCID: PMC11462364 DOI: 10.1016/j.wnsx.2024.100399] [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: 06/08/2023] [Accepted: 09/20/2024] [Indexed: 10/12/2024] Open
Abstract
This comprehensive review paper examines the most updated state of research on glioblastoma, an aggressive brain tumor with limited treatment options. By analyzing 76 recent studies, from translational and basic sciences, to clinical trials, we highlight various aspects of glioblastoma and shed light on potential therapeutic strategies. The interplay between tumor cells, neural progenitor cells, and the tumor microenvironment is explored. Targeting the PI3K-Akt-mTOR pathway through extracellular-vesicle (EV)-mediated signaling emerges as a potential therapeutic strategy. Personalized modeling approaches utilizing patient-specific MRI data offer promise for optimizing treatment strategies. The response of glioblastoma stem cells (GSCs) to different treatment modalities is examined, emphasizing the need to inhibit the transformation of proneural (PN) GSCs into resistant mesenchymal (MES) GSCs. Metabolic therapy and combination therapies show potential in reversing treatment resistance and inhibiting both PN and MES GSCs. Immunotherapy, targeted approaches, and molecular dynamics in gliomas are discussed, providing insights into early-stage diagnosis and treatment. Additionally, the potential use of Zika virus as an oncolytic agent is explored. Analysis of phase 0 to 3 clinical trials reveal promising outcomes for various experimental treatments, highlighting the importance of combination therapies, predictive signatures, and patient selection strategies. Specific compounds demonstrate potential therapeutic benefits and tolerability. Phase 3 trials indicate the efficacy of DCVax-L in improving survival rates and depatux-m in prolonging progression-free survival. These findings emphasize the importance of personalized treatment approaches and continued exploration of targeted therapies, immunotherapies, and tumor biology understanding in shaping the future of glioblastoma treatment.
Collapse
Affiliation(s)
| | | | - Ahmed Rahib
- Nowshera Medical College, Nowshera, Pakistan
| | - Amna Shamim
- King Edward Medical University, Lahore, Pakistan
| | | |
Collapse
|
3
|
Zhou L, Xiang H, Liu S, Chen H, Yang Y, Zhang J, Cai W. Folic Acid Functionalized AQ4N/Gd@PDA Nanoplatform with Real-Time Monitoring of Hypoxia Relief and Enhanced Synergistic Chemo/Photothermal Therapy in Glioma. Int J Nanomedicine 2024; 19:3367-3386. [PMID: 38617794 PMCID: PMC11012807 DOI: 10.2147/ijn.s451921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 03/27/2024] [Indexed: 04/16/2024] Open
Abstract
Purpose Hypoxia is often associated with glioma chemoresistance, and alleviating hypoxia is also crucial for improving treatment efficacy. However, although there are already some methods that can improve efficacy by alleviating hypoxia, real-time monitoring that can truly achieve hypoxia relief and efficacy feedback still needs to be explored. Methods AQ4N/Gd@PDA-FA nanoparticles (AGPF NPs) were synthesized using a one-pot method and were characterized. The effects of AGPF NPs on cell viability, cellular uptake, and apoptosis were investigated using the U87 cell line. Moreover, the effectiveness of AGPF NPs in alleviating hypoxia was explored in tumor-bearing mice through photoacoustic imaging. In addition, the diagnosis and treatment effect of AGPF NPs were evaluated by magnetic resonance imaging (MRI) and bioluminescent imaging (BLI) on orthotopic glioma mice respectively. Results In vitro experiments showed that AGPF NPs had good dispersion, stability, and controlled release. AGPF NPs were internalized by cells through endocytosis, and could significantly reduce the survival rate of U87 cells and increase apoptosis under irradiation. In addition, we monitored blood oxygen saturation at the tumor site in real-time through photoacoustic imaging (PAI), and the results showed that synergistic mild-photothermal therapy/chemotherapy effectively alleviated tumor hypoxia. Finally, in vivo anti-tumor experiments have shown that synergistic therapy can effectively alleviate hypoxia and inhibit the growth of orthotopic gliomas. Conclusion This work not only provides an effective means for real-time monitoring of the dynamic feedback between tumor hypoxia relief and therapeutic efficacy, but also offers a potential approach for the clinical treatment of gliomas.
Collapse
Affiliation(s)
- Longjiang Zhou
- Department of Neurology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225012, People’s Republic of China
| | - Haitao Xiang
- Suzhou Kowloon Hospital, Shanghai Jiao Tong University School of Medicine, Suzhou, 215028, People’s Republic of China
| | - Susu Liu
- School of Life Science and Technology, Xidian University and Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, Xi’an, 710126, People’s Republic of China
| | - Honglin Chen
- Department of Neurosurgery, Suqian First Hospital, Suqian, 223800, People’s Republic of China
| | - Yuanwei Yang
- Department of Neurosurgery, Suqian First Hospital, Suqian, 223800, People’s Republic of China
| | - Jianyong Zhang
- Department of Neurosurgery, Suqian First Hospital, Suqian, 223800, People’s Republic of China
| | - Wei Cai
- Department of Neurosurgery, Suqian First Hospital, Suqian, 223800, People’s Republic of China
| |
Collapse
|
4
|
Ge M, Zhu Y, Wei M, Piao H, He M. Improving the efficacy of anti-EGFR drugs in GBM: Where we are going? Biochim Biophys Acta Rev Cancer 2023; 1878:188996. [PMID: 37805108 DOI: 10.1016/j.bbcan.2023.188996] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 09/11/2023] [Accepted: 09/29/2023] [Indexed: 10/09/2023]
Abstract
The therapies targeting mutations of driver genes in cancer have advanced into clinical trials for a variety of tumors. In glioblastoma (GBM), epidermal growth factor receptor (EGFR) is the most commonly mutated oncogene, and targeting EGFR has been widely investigated as a promising direction. However, the results of EGFR pathway inhibitors have not been satisfactory. Limited blood-brain barrier (BBB) permeability, drug resistance, and pathway compensation mechanisms contribute to the failure of anti-EGFR therapies. This review summarizes recent research advances in EGFR-targeted therapy for GBM and provides insight into the reasons for the unsatisfactory results of EGFR-targeted therapy. By combining the results of preclinical studies with those of clinical trials, we discuss that improved drug penetration across the BBB, the use of multi-target combinations, and the development of peptidomimetic drugs under the premise of precision medicine may be promising strategies to overcome drug resistance in GBM.
Collapse
Affiliation(s)
- Manxi Ge
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China; Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Shenyang, China
| | - Yan Zhu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China; Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Shenyang, China
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China; Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Shenyang, China; Liaoning Medical Diagnosis and Treatment Center, Shenyang, China.
| | - Haozhe Piao
- Department of Neurosurgery, Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University, Shenyang, China.
| | - Miao He
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China; Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Shenyang, China.
| |
Collapse
|
5
|
Geoerger B, Marshall LV, Nysom K, Makin G, Bouffet E, Defachelles AS, Amoroso L, Aerts I, Leblond P, Barahona P, Van-Vlerken K, Fu E, Solca F, Lorence RM, Ziegler DS. Afatinib in paediatric patients with recurrent/refractory ErbB-dysregulated tumours: Results of a phase I/expansion trial. Eur J Cancer 2023; 188:8-19. [PMID: 37178647 DOI: 10.1016/j.ejca.2023.04.007] [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: 03/02/2023] [Revised: 04/13/2023] [Accepted: 04/15/2023] [Indexed: 05/15/2023]
Abstract
AIM This phase I/expansion study assessed the safety, pharmacokinetics and preliminary antitumor activity of afatinib in paediatric patients with cancer. METHODS The dose-finding part enroled patients (2-<18 years) with recurrent/refractory tumours. Patients received 18 or 23 mg/m2/d afatinib orally (tablet or solution) in 28-d cycles. In the maximum tolerated dose (MTD) expansion, eligible patients (1-<18 years) had tumours fulfilling ≥2 of the following criteria in the pre-screening: EGFR amplification; HER2 amplification; EGFR membrane staining (H-score>150); HER2 membrane staining (H-score>0). The primary end-points were dose-limiting toxicities (DLTs), afatinib exposure, and objective response. RESULTS Of 564 patients pre-screened, 536 patients had biomarker data and 63 (12%) fulfilled ≥2 EGFR/HER2 criteria required for inclusion in the expansion part. A total of 56 patients were treated (17 in the dose-finding and 39 in the expansion part). DLTs were observed in one of six MTD-evaluable patients receiving 18 mg/m²/d and in two of five MTD-evaluable patients receiving 23 mg/m²/d; 18 mg/m²/d was defined as the MTD. There were no new safety signals. Pharmacokinetics confirmed exposure consistent with the approved dose in adults. One partial response (-81% per Response Assessment in Neuro-Oncology) was observed in a patient with a glioneuronal tumour harbouring a CLIP2::EGFR fusion; unconfirmed partial responses were observed in two patients. In total, 25% of patients experienced objective response or stable disease (95% confidence interval: 14-38). CONCLUSION Targetable EGFR/HER2 drivers are rare in paediatric cancers. Treatment with afatinib led to a durable response (>3 years) in one patient with a glioneuronal tumour with CLIP2::EGFR fusion.
Collapse
Affiliation(s)
- Birgit Geoerger
- Gustave Roussy Cancer Campus, Department of Pediatric and Adolescent Oncology, INSERM U1015, Université Paris-Saclay, Villejuif, France.
| | - Lynley V Marshall
- The Royal Marsden NHS Foundation Trust and The Institute of Cancer Research, London, UK
| | - Karsten Nysom
- Department of Paediatrics and Adolescent Medicine, Rigshospitalet, Copenhagen, Denmark
| | - Guy Makin
- Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK; Royal Manchester Children's Hospital, Manchester, UK
| | - Eric Bouffet
- The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | | | | | - Isabelle Aerts
- Institut Curie, PSL Research University, Oncology Center SIREDO, Paris, France
| | - Pierre Leblond
- Institute of Pediatric Hematology and Oncology, Centre Léon Bérard, Lyon, France
| | | | | | - Eric Fu
- Boehringer Ingelheim Pharmaceuticals, Inc. Ridgefield, CT, USA
| | - Flavio Solca
- Boehringer Ingelheim RCV GmbH & Co.KG Vienna, Austria
| | | | - David S Ziegler
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW, Australia; School of Clinical Medicine, UNSW Medicine & Health, UNSW Sydney, Sydney, NSW, Australia; Children's Cancer Institute, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
6
|
Khairnar S, Sonawane A, Cheke RS, Kharkar PS, Gaikwad V, Patil S, Aware V. Hit discovery of novel 2-phenyl-substituted 4-amino-6,7-dihydro-5H-cyclopenta[d]pyrimidines as potential anti-glioblastoma therapeutics: Design, synthesis, biological evaluation, and computational screening. Drug Dev Res 2023; 84:561-578. [PMID: 36823756 DOI: 10.1002/ddr.22046] [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: 12/11/2022] [Revised: 01/15/2023] [Accepted: 01/28/2023] [Indexed: 02/25/2023]
Abstract
Glioblastoma multiforme (GBM) is a highly-aggressive, dreadful disease with poor prognosis and disappointing clinical success. There is an unmet medical need of molecularly-targeted therapeutics for GBM treatment. In the present work, a series of novel 2-phenyl-substituted 4-amino-6,7-dihydro-5H-cyclopenta[d]pyrimidines was designed, synthesized, purified, characterized, and evaluated for cytotoxicity against glioblastoma cell line U87-MG. The design process (virtual library enumeration around the core, physicochemical and molecular property prediction/calculation of the designs, filtering the undesirable ones, and the diversity analyses of the lead-like designs), was carefully curated so as to obtain a set of structurally-diverse, novel molecules (total 20), with a particular focus on the relatively unexplored core structure, 6,7-dihydro-5H-cyclopenta[d]pyrimidine. The preliminary screening was done using MTT assay at 10 and 100 μM concentrations of the title compounds F1 -F20 and positive control cisplatin, which yielded six hits (% inhibition at 10 μM: ~50%)-F2 , F3 , F5 , F7 , F15 , and F20 , which were taken up for IC50 determination. The top hits F2 and F7 (IC50 < 10 μM) were further used for computational studies such as target prediction, followed by their molecular docking in the binding sites of the top-3 predicted targets (epidermal growth factor receptor kinase domain, cyclin-dependent kinase 2 [CDK2]) /cyclin E, and anaplastic lymphoma kinase [ALK]). The docking pose analyses revealed interesting trends. The relatively planar core structure, presence of favorable hinge-binding substructures, basic groups, all added up, and culminated in appreciable cytotoxicity against GBM cell line.
Collapse
Affiliation(s)
- Sanjay Khairnar
- Department of Chemistry, SNJB's KKHA Arts, SMGL Commerce and SPHJ Science College, Chandwad, India.,Department of Chemistry, Organic Chemistry Research Centre, K. R. T. Arts, B. H. Commerce and A. M. Science College, Nashik, India
| | - Anjali Sonawane
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, India
| | - Rameshwar S Cheke
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, India
| | - Prashant S Kharkar
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, India
| | - Vishwas Gaikwad
- Department of Chemistry, Organic Chemistry Research Centre, K. R. T. Arts, B. H. Commerce and A. M. Science College, Nashik, India
| | - Sambhaji Patil
- Department of Chemistry, Organic Chemistry Research Centre, K. R. T. Arts, B. H. Commerce and A. M. Science College, Nashik, India
| | - Valmik Aware
- Department of Chemistry, SNJB's KKHA Arts, SMGL Commerce and SPHJ Science College, Chandwad, India
| |
Collapse
|
7
|
Frosina G. Most recent update of preclinical and clinical data on radioresistance and radiosensitivity of high-grade gliomas-a radiation oncologist's perspective. Strahlenther Onkol 2023; 199:1-21. [PMID: 36445383 DOI: 10.1007/s00066-022-02020-2] [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: 06/07/2022] [Accepted: 10/09/2022] [Indexed: 12/03/2022]
Abstract
PURPOSE This review article discusses the studies concerning advances in radiotherapy of high-grade gliomas published in the second half of 2021. METHODS A literature search was performed in PubMed using the terms ("gliom* and radio*") and time limits 1 July 2021-31 December 2021. The articles were then manually selected for relevance to the analyzed topics. RESULTS Considerable progress has been made in the preclinical field on the mechanisms of radioresistance and radiosensitization of high-grade gliomas (HGG). However, fewer early-phase (I/II) clinical trials have been performed and, of the latter, even fewer have produced results that justify moving to phase III. In the 6‑month period under consideration, no studies were published that would lead to a change in clinical practice and the overall survival (OS) of patients remained similar to that of 2005, the year in which it increased significantly for the last time thanks to introduction of the alkylating agent temozolomide. CONCLUSION After 17 years of stalemate in improving the OS of patients with HGG, an in-depth analysis of the causes should be carried out in order to identify whether the research efforts conducted so far, including in the radiotherapeutic field, have been the most effective or require improvement. In our opinion, in addition to the therapeutic difficulties related to the biology of HGG tumors (e.g., high infiltrating capacity, multiple resistance mechanisms, blood-brain barrier), some public research policy choices may also play a role, especially in consideration of the limited interest of the pharmaceutical industry in the field of rare cancers.
Collapse
Affiliation(s)
- Guido Frosina
- Mutagenesis & Cancer Prevention Unit, IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132, Genova, Italy.
| |
Collapse
|
8
|
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: 6] [Impact Index Per Article: 2.0] [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
|
9
|
Lange F, Venus J, Shams Esfand Abady D, Porath K, Einsle A, Sellmann T, Neubert V, Reichart G, Linnebacher M, Köhling R, Kirschstein T. Galvanotactic Migration of Glioblastoma and Brain Metastases Cells. Life (Basel) 2022; 12:life12040580. [PMID: 35455071 PMCID: PMC9027426 DOI: 10.3390/life12040580] [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: 03/22/2022] [Revised: 04/11/2022] [Accepted: 04/11/2022] [Indexed: 12/27/2022] Open
Abstract
Galvanotaxis, the migration along direct current electrical fields, may contribute to the invasion of brain cancer cells in the tumor-surrounding tissue. We hypothesized that pharmacological perturbation of the epidermal growth factor (EGF) receptor and downstream phosphatidylinositol 3-kinase (PI3K)/AKT pathway prevent galvanotactic migration. In our study, patient-derived glioblastoma and brain metastases cells were exposed to direct current electrical field conditions. Velocity and direction of migration were estimated. To determine the effects of EGF receptor antagonist afatinib and AKT inhibitor capivasertib, assays of cell proliferation, apoptosis and immunoblot analyses were performed. Both inhibitors attenuated cell proliferation in a dose-dependent manner and induced apoptosis. We found that most of the glioblastoma cells migrated preferentially in an anodal direction, while brain metastases cells were unaffected by direct current stimulations. Afatinib presented only a mild attenuation of galvanotaxis. In contrast, capivasertib abolished the migration of glioblastoma cells without genetic alterations in the PI3K/AKT pathway, but not in cells harboring PTEN mutation. In these cells, an increase in the activation of ERK1/2 may in part substitute the inhibition of the AKT pathway. Overall, our data demonstrate that glioblastoma cells migrate in the electrical field and the PI3K/AKT pathway was found to be highly involved in galvanotaxis.
Collapse
Affiliation(s)
- Falko Lange
- Oscar-Langendorff-Institute of Physiology, Rostock University Medical Center, 18057 Rostock, Germany; (J.V.); (D.S.E.A.); (K.P.); (A.E.); (T.S.); (V.N.); (G.R.); (R.K.); (T.K.)
- Center for Transdisciplinary Neurosciences Rostock, University of Rostock, 18147 Rostock, Germany
- Correspondence:
| | - Jakob Venus
- Oscar-Langendorff-Institute of Physiology, Rostock University Medical Center, 18057 Rostock, Germany; (J.V.); (D.S.E.A.); (K.P.); (A.E.); (T.S.); (V.N.); (G.R.); (R.K.); (T.K.)
| | - Daria Shams Esfand Abady
- Oscar-Langendorff-Institute of Physiology, Rostock University Medical Center, 18057 Rostock, Germany; (J.V.); (D.S.E.A.); (K.P.); (A.E.); (T.S.); (V.N.); (G.R.); (R.K.); (T.K.)
| | - Katrin Porath
- Oscar-Langendorff-Institute of Physiology, Rostock University Medical Center, 18057 Rostock, Germany; (J.V.); (D.S.E.A.); (K.P.); (A.E.); (T.S.); (V.N.); (G.R.); (R.K.); (T.K.)
| | - Anne Einsle
- Oscar-Langendorff-Institute of Physiology, Rostock University Medical Center, 18057 Rostock, Germany; (J.V.); (D.S.E.A.); (K.P.); (A.E.); (T.S.); (V.N.); (G.R.); (R.K.); (T.K.)
| | - Tina Sellmann
- Oscar-Langendorff-Institute of Physiology, Rostock University Medical Center, 18057 Rostock, Germany; (J.V.); (D.S.E.A.); (K.P.); (A.E.); (T.S.); (V.N.); (G.R.); (R.K.); (T.K.)
| | - Valentin Neubert
- Oscar-Langendorff-Institute of Physiology, Rostock University Medical Center, 18057 Rostock, Germany; (J.V.); (D.S.E.A.); (K.P.); (A.E.); (T.S.); (V.N.); (G.R.); (R.K.); (T.K.)
| | - Gesine Reichart
- Oscar-Langendorff-Institute of Physiology, Rostock University Medical Center, 18057 Rostock, Germany; (J.V.); (D.S.E.A.); (K.P.); (A.E.); (T.S.); (V.N.); (G.R.); (R.K.); (T.K.)
| | - Michael Linnebacher
- Molecular Oncology and Immunotherapy, Rostock University Medical Center, 18057 Rostock, Germany;
| | - Rüdiger Köhling
- Oscar-Langendorff-Institute of Physiology, Rostock University Medical Center, 18057 Rostock, Germany; (J.V.); (D.S.E.A.); (K.P.); (A.E.); (T.S.); (V.N.); (G.R.); (R.K.); (T.K.)
- Center for Transdisciplinary Neurosciences Rostock, University of Rostock, 18147 Rostock, Germany
| | - Timo Kirschstein
- Oscar-Langendorff-Institute of Physiology, Rostock University Medical Center, 18057 Rostock, Germany; (J.V.); (D.S.E.A.); (K.P.); (A.E.); (T.S.); (V.N.); (G.R.); (R.K.); (T.K.)
- Center for Transdisciplinary Neurosciences Rostock, University of Rostock, 18147 Rostock, Germany
| |
Collapse
|
10
|
Owen S, Alken S, Alshami J, Guiot MC, Kavan P, Reardon DA, Muanza T, Gibson N, Pemberton K, Solca F, Cseh A, Saran F. Genomic Analysis of Tumors from Patients with Glioblastoma with Long-Term Response to Afatinib. Onco Targets Ther 2022; 15:367-380. [PMID: 35422631 PMCID: PMC9005142 DOI: 10.2147/ott.s346725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 03/16/2022] [Indexed: 12/05/2022] Open
Abstract
Glioblastoma is an aggressive form of central nervous system tumor. Recurrence rates following primary therapy are high, and few second-line treatment options provide durable clinical benefit. Aberrations of the epidermal growth factor receptor (EGFR) gene are observed in up to 57% of glioblastoma cases and EGFR overexpression has been identified in approximately 60% of primary glioblastomas. In preclinical studies, afatinib, a second-generation ErbB blocker, inhibited cell proliferation in cells harboring mutations commonly found in glioblastoma. In two previous Phase I/II studies of afatinib plus temozolomide in patients with glioblastoma, limited efficacy was observed; however, there was notable benefit in patients with the EGFR variant III (EGFRvIII) mutation, EGFR amplification, and those with loss of phosphatase and tensin homolog (PTEN). This case series report details treatment histories of three long-term responders from these trials. Next-generation sequencing of tumor samples identified alterations in a number of cancer-related genes, including mutations in, and amplification of, EGFR. Tumor samples from all three patients shared favorable prognostic factors, eg O6-methylguanine-DNA methyl-transferase (MGMT) gene promoter methylation; however, negative prognostic factors were also observed, suggesting that these shared genetic features did not completely account for the favorable responses. The genetic profile of the tumor from Patient 1 showed clear differences from the other two tumors: lack of involvement of EGFR aberrations but with a mutation occurring in PTPN11. Preclinical studies showed that single-agent afatinib and temozolomide both separately inhibit the growth of tumors with a C-terminal EGFR truncation, thus providing further rationale for combining these two agents in the treatment of glioblastomas harboring EGFR aberrations. These findings suggest that afatinib may provide treatment benefit in patients with glioblastomas that harbor ErbB family aberrations and, potentially, other genetic aberrations. Further studies are needed to establish which patients with newly diagnosed/recurrent glioblastomas may potentially benefit from treatment with afatinib.
Collapse
Affiliation(s)
- Scott Owen
- Clinical Research Unit, Montreal Neurological Institute and Hospital, McGill University Health Center, Montreal, Canada
| | - Scheryll Alken
- Radiation Oncology Unit, Royal Marsden Hospital, London, UK
- St James’s Hospital, Dublin, Ireland
| | - Jad Alshami
- Clinical Research Unit, Montreal Neurological Institute and Hospital, McGill University Health Center, Montreal, Canada
| | - Marie-Christine Guiot
- Clinical Research Unit, Montreal Neurological Institute and Hospital, McGill University Health Center, Montreal, Canada
- Neuropathology Division, Montreal Neurological Institute and Hospital, McGill University Health Center, Montreal, Canada
| | - Petr Kavan
- Clinical Research Unit, Montreal Neurological Institute and Hospital, McGill University Health Center, Montreal, Canada
| | - David A Reardon
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Thierry Muanza
- Clinical Research Unit, Montreal Neurological Institute and Hospital, McGill University Health Center, Montreal, Canada
- Neuropathology Division, Montreal Neurological Institute and Hospital, McGill University Health Center, Montreal, Canada
- Radiation Oncology, Jewish General Hospital, Montreal, Canada
| | - Neil Gibson
- Drug Metabolism & Pharmacokinetics, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | | | - Flavio Solca
- Department of Pharmacology, Boehringer Ingelheim RCV GmbH & Co. KG, Vienna, Austria
| | - Agnieszka Cseh
- Department of Medical Affairs, Boehringer Ingelheim International, Ingelheim am Rhein, Germany
| | - Frank Saran
- Radiation Oncology Unit, Royal Marsden Hospital, London, UK
- Department of Blood and Cancer, Auckland City Hospital, Auckland, New Zealand
- Correspondence: Frank Saran, Auckland City Hospital, Cancer and Blood Service, Building 8, 99 Park Road, Grafton, Private Bag 92024, Auckland, 1142, New Zealand, Tel +64 09 623 6046, Email
| |
Collapse
|
11
|
Liu H, Qiu W, Sun T, Wang L, Du C, Hu Y, Liu W, Feng F, Chen Y, Sun H. Therapeutic strtegies of glioblastoma (GBM): The current advances in the molecular targets and bioactive small molecule compounds. Acta Pharm Sin B 2021; 12:1781-1804. [PMID: 35847506 PMCID: PMC9279645 DOI: 10.1016/j.apsb.2021.12.019] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 12/02/2021] [Accepted: 12/21/2021] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma (GBM) is the most common aggressive malignant tumor in brain neuroepithelial tumors and remains incurable. A variety of treatment options are currently being explored to improve patient survival, including small molecule inhibitors, viral therapies, cancer vaccines, and monoclonal antibodies. Among them, the unique advantages of small molecule inhibitors have made them a focus of attention in the drug discovery of glioblastoma. Currently, the most used chemotherapeutic agents are small molecule inhibitors that target key dysregulated signaling pathways in glioblastoma, including receptor tyrosine kinase, PI3K/AKT/mTOR pathway, DNA damage response, TP53 and cell cycle inhibitors. This review analyzes the therapeutic benefit and clinical development of novel small molecule inhibitors discovered as promising anti-glioblastoma agents by the related targets of these major pathways. Meanwhile, the recent advances in temozolomide resistance and drug combination are also reviewed. In the last part, due to the constant clinical failure of targeted therapies, this paper reviewed the research progress of other therapeutic methods for glioblastoma, to provide patients and readers with a more comprehensive understanding of the treatment landscape of glioblastoma.
Collapse
|