1
|
Barone TA, Robinson DL, Qiu J, Gurova KV, Purmal AA, Gudkov AV, Plunkett RJ. FACT inhibitor CBL0137, administered in an optimized schedule, potentiates radiation therapy for glioblastoma by suppressing DNA damage repair. J Neurooncol 2024:10.1007/s11060-024-04819-8. [PMID: 39251545 DOI: 10.1007/s11060-024-04819-8] [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: 07/30/2024] [Accepted: 08/30/2024] [Indexed: 09/11/2024]
Abstract
PURPOSE Standard-of-care for glioblastoma remains surgical debulking followed by temozolomide and radiation. However, many tumors become radio-resistant while radiation damages surrounding brain tissue. Novel therapies are needed to increase the effectiveness of radiation and reduce the required radiation dose. Drug candidate CBL0137 is efficacious against glioblastoma by inhibiting histone chaperone FACT, known to be involved in DNA damage repair. We investigated the combination of CBL0137 and radiation on glioblastoma. METHODS In vitro, we combined CBL0137 with radiation on U87MG and A1207 glioblastoma cells using the clonogenic assay to evaluate the response to several treatment regimens, and the Fast Halo Assay to examine DNA repair. In vivo, we used the optimum combination treatment regimen to evaluate the response of orthotopic tumors in nude mice. RESULTS In vitro, the combination of CBL0137 and radiation is superior to either alone and administering CBL0137 two hours prior to radiation, having the drug present during and for a prolonged period post-radiation, is an optimal schedule. CBL0137 inhibits DNA damage repair following radiation and affects the subcellular distribution of histone chaperone ATRX, a molecule involved in DNA repair. In vivo, one dose of CBL0137 is efficacious and the combination of CBL0137 with radiation increases median survival over either monotherapy. CONCLUSIONS CBL0137 is most effective with radiation for glioblastoma when present at the time of radiation, immediately after and for a prolonged period post-radiation, by inhibiting DNA repair caused by radiation. The combination leads to increased survival making it attractive as a dual therapy.
Collapse
Affiliation(s)
- Tara A Barone
- Department of Neuro-Oncology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA.
| | - Denisha L Robinson
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Jingxin Qiu
- Department of Pathology and Laboratory Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Katerina V Gurova
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | | | - Andrei V Gudkov
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Robert J Plunkett
- Department of Neuro-Oncology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA
| |
Collapse
|
2
|
Barone TA, Robinson DL, Qiu J, Gurova KV, Purmal AA, Gudkov AV, Plunkett RJ. FACT inhibitor CBL0137, administered in an optimized schedule, potentiates radiation therapy for glioblastoma by suppressing DNA damage repair. RESEARCH SQUARE 2024:rs.3.rs-4830689. [PMID: 39315270 PMCID: PMC11419263 DOI: 10.21203/rs.3.rs-4830689/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Purpose Standard-of-care for glioblastoma remains surgical debulking followed by temozolomide and radiation. However, many tumors become radio-resistant while radiation damages surrounding brain tissue. Novel therapies are needed to increase the effectiveness of radiation and reduce the required radiation dose. Drug candidate CBL0137 is efficacious against glioblastoma by inhibiting histone chaperone FACT, known to be involved in DNA damage repair. We investigated the combination of CBL0137 and radiation on glioblastoma. Methods In vitro, we combined CBL0137 with radiation on U87MG and A1207 glioblastoma cells using the clonogenic assay to evaluate the response to several treatment regimens, and the Fast Halo Assay to examine DNA repair. In vivo, we used the optimum combination treatment regimen to evaluate the response of orthotopic tumors in nude mice. Results In vitro, the combination of CBL0137 and radiation is superior to either alone and administering CBL0137 two hours prior to radiation, having the drug present during and for a prolonged period post-radiation, is an optimal schedule. CBL0137 inhibits DNA damage repair following radiation and affects the subcellular distribution of histone chaperone ATRX, a molecule involved in DNA repair. In vivo, one dose of CBL0137 is efficacious and the combination of CBL0137 with radiation increases median survival over either monotherapy. Conclusions CBL0137 is most effective with radiation for glioblastoma when present at the time of radiation, immediately after and for a prolonged period post-radiation, by inhibiting DNA repair caused by radiation. The combination leads to increased survival making it attractive as a dual therapy.
Collapse
|
3
|
Taylor J, Dubois F, Bergot E, Levallet G. Targeting the Hippo pathway to prevent radioresistance brain metastases from the lung (Review). Int J Oncol 2024; 65:68. [PMID: 38785155 PMCID: PMC11155713 DOI: 10.3892/ijo.2024.5656] [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: 11/21/2023] [Accepted: 03/04/2024] [Indexed: 05/25/2024] Open
Abstract
The prognosis for patients with non‑small cell lung cancer (NSCLC), a cancer type which represents 85% of all lung cancers, is poor with a 5‑year survival rate of 19%, mainly because NSCLC is diagnosed at an advanced and metastatic stage. Despite recent therapeutic advancements, ~50% of patients with NSCLC will develop brain metastases (BMs). Either surgical BM treatment alone for symptomatic patients and patients with single cerebral metastases, or in combination with stereotactic radiotherapy (RT) for patients who are not suitable for surgery or presenting with fewer than four cerebral lesions with a diameter range of 5‑30 mm, or whole‑brain RT for numerous or large BMs can be administered. However, radioresistance (RR) invariably prevents the action of RT. Several mechanisms of RR have been described including hypoxia, cellular stress, presence of cancer stem cells, dysregulation of apoptosis and/or autophagy, dysregulation of the cell cycle, changes in cellular metabolism, epithelial‑to‑mesenchymal transition, overexpression of programmed cell death‑ligand 1 and activation several signaling pathways; however, the role of the Hippo signaling pathway in RR is unclear. Dysregulation of the Hippo pathway in NSCLC confers metastatic properties, and inhibitors targeting this pathway are currently in development. It is therefore essential to evaluate the effect of inhibiting the Hippo pathway, particularly the effector yes‑associated protein‑1, on cerebral metastases originating from lung cancer.
Collapse
Affiliation(s)
- Jasmine Taylor
- University of Caen Normandy, National Center for Scientific Research, Normandy University, Unit of Imaging and Therapeutic Strategies for Cancers and Cerebral Tissues (ISTCT)-UMR6030, GIP CYCERON, F-14074 Caen, France
| | - Fatéméh Dubois
- University of Caen Normandy, National Center for Scientific Research, Normandy University, Unit of Imaging and Therapeutic Strategies for Cancers and Cerebral Tissues (ISTCT)-UMR6030, GIP CYCERON, F-14074 Caen, France
- Departments of Pathology, and Thoracic Oncology, Caen University Hospital, F-14033 Caen, France
| | - Emmanuel Bergot
- University of Caen Normandy, National Center for Scientific Research, Normandy University, Unit of Imaging and Therapeutic Strategies for Cancers and Cerebral Tissues (ISTCT)-UMR6030, GIP CYCERON, F-14074 Caen, France
- Departments of Pneumology and Thoracic Oncology, Caen University Hospital, F-14033 Caen, France
| | - Guénaëlle Levallet
- University of Caen Normandy, National Center for Scientific Research, Normandy University, Unit of Imaging and Therapeutic Strategies for Cancers and Cerebral Tissues (ISTCT)-UMR6030, GIP CYCERON, F-14074 Caen, France
- Departments of Pathology, and Thoracic Oncology, Caen University Hospital, F-14033 Caen, France
| |
Collapse
|
4
|
Brennan RJ, Jenkinson S, Brown A, Delaunois A, Dumotier B, Pannirselvam M, Rao M, Ribeiro LR, Schmidt F, Sibony A, Timsit Y, Sales VT, Armstrong D, Lagrutta A, Mittlestadt SW, Naven R, Peri R, Roberts S, Vergis JM, Valentin JP. The state of the art in secondary pharmacology and its impact on the safety of new medicines. Nat Rev Drug Discov 2024; 23:525-545. [PMID: 38773351 DOI: 10.1038/s41573-024-00942-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/05/2024] [Indexed: 05/23/2024]
Abstract
Secondary pharmacology screening of investigational small-molecule drugs for potentially adverse off-target activities has become standard practice in pharmaceutical research and development, and regulatory agencies are increasingly requesting data on activity against targets with recognized adverse effect relationships. However, the screening strategies and target panels used by pharmaceutical companies may vary substantially. To help identify commonalities and differences, as well as to highlight opportunities for further optimization of secondary pharmacology assessment, we conducted a broad-ranging survey across 18 companies under the auspices of the DruSafe leadership group of the International Consortium for Innovation and Quality in Pharmaceutical Development. Based on our analysis of this survey and discussions and additional research within the group, we present here an overview of the current state of the art in secondary pharmacology screening. We discuss best practices, including additional safety-associated targets not covered by most current screening panels, and present approaches for interpreting and reporting off-target activities. We also provide an assessment of the safety impact of secondary pharmacology screening, and a perspective on opportunities and challenges in this rapidly developing field.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Mohan Rao
- Janssen Research & Development, San Diego, CA, USA
- Neurocrine Biosciences, San Diego, CA, USA
| | - Lyn Rosenbrier Ribeiro
- UCB Biopharma, Braine-l'Alleud, Belgium
- AstraZeneca, Cambridge, UK
- Grunenthal, Berkshire, UK
| | | | | | - Yoav Timsit
- Novartis Biomedical Research, Cambridge, MA, USA
- Blueprint Medicines, Cambridge, MA, USA
| | | | - Duncan Armstrong
- Novartis Biomedical Research, Cambridge, MA, USA
- Armstrong Pharmacology, Macclesfield, UK
| | | | | | - Russell Naven
- Takeda Pharmaceuticals, Cambridge, MA, USA
- Novartis Biomedical Research, Cambridge, MA, USA
| | - Ravikumar Peri
- Takeda Pharmaceuticals, Cambridge, MA, USA
- Alexion Pharmaceuticals, Wilmington, DE, USA
| | - Sonia Roberts
- Roche Pharma Research and Early Development, Roche Innovation Center, Basel, Switzerland
| | - James M Vergis
- Faegre Drinker Biddle and Reath, LLP, Washington, DC, USA
| | | |
Collapse
|
5
|
Mosca L, Pagano C, Tranchese RV, Grillo R, Cadoni F, Navarra G, Coppola L, Pagano M, Mele L, Cacciapuoti G, Laezza C, Porcelli M. Antitumoral Activity of the Universal Methyl Donor S-Adenosylmethionine in Glioblastoma Cells. Molecules 2024; 29:1708. [PMID: 38675528 PMCID: PMC11052366 DOI: 10.3390/molecules29081708] [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: 03/04/2024] [Revised: 04/03/2024] [Accepted: 04/04/2024] [Indexed: 04/28/2024] Open
Abstract
Glioblastoma (GBM), the most frequent and lethal brain cancer in adults, is characterized by short survival times and high mortality rates. Due to the resistance of GBM cells to conventional therapeutic treatments, scientific interest is focusing on the search for alternative and efficient adjuvant treatments. S-Adenosylmethionine (AdoMet), the well-studied physiological methyl donor, has emerged as a promising anticancer compound and a modulator of multiple cancer-related signaling pathways. We report here for the first time that AdoMet selectively inhibited the viability and proliferation of U87MG, U343MG, and U251MG GBM cells. In these cell lines, AdoMet induced S and G2/M cell cycle arrest and apoptosis and downregulated the expression and activation of proteins involved in homologous recombination DNA repair, including RAD51, BRCA1, and Chk1. Furthermore, AdoMet was able to maintain DNA in a damaged state, as indicated by the increased γH2AX/H2AX ratio. AdoMet promoted mitotic catastrophe through inhibiting Aurora B kinase expression, phosphorylation, and localization causing GBM cells to undergo mitotic catastrophe-induced death. Finally, AdoMet inhibited DNA repair and induced cell cycle arrest, apoptosis, and mitotic catastrophe in patient-derived GBM cells. In light of these results, AdoMet could be considered a potential adjuvant in GBM therapy.
Collapse
Affiliation(s)
- Laura Mosca
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via Luigi De Crecchio 7, 80138 Naples, Italy; (L.M.); (R.V.T.); (R.G.); (F.C.); (M.P.); (M.P.)
| | - Cristina Pagano
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy; (C.P.); (G.N.); (L.C.)
| | - Roberta Veglia Tranchese
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via Luigi De Crecchio 7, 80138 Naples, Italy; (L.M.); (R.V.T.); (R.G.); (F.C.); (M.P.); (M.P.)
| | - Roberta Grillo
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via Luigi De Crecchio 7, 80138 Naples, Italy; (L.M.); (R.V.T.); (R.G.); (F.C.); (M.P.); (M.P.)
| | - Francesca Cadoni
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via Luigi De Crecchio 7, 80138 Naples, Italy; (L.M.); (R.V.T.); (R.G.); (F.C.); (M.P.); (M.P.)
| | - Giovanna Navarra
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy; (C.P.); (G.N.); (L.C.)
| | - Laura Coppola
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy; (C.P.); (G.N.); (L.C.)
| | - Martina Pagano
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via Luigi De Crecchio 7, 80138 Naples, Italy; (L.M.); (R.V.T.); (R.G.); (F.C.); (M.P.); (M.P.)
| | - Luigi Mele
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Via Luciano Armanni 5, 80138 Naples, Italy;
| | - Giovanna Cacciapuoti
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via Luigi De Crecchio 7, 80138 Naples, Italy; (L.M.); (R.V.T.); (R.G.); (F.C.); (M.P.); (M.P.)
| | - Chiara Laezza
- Institute of Endocrinology and Experimental Oncology (IEOS), National Research Council (CNR), Via Pansini 5, 80131 Naples, Italy;
| | - Marina Porcelli
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via Luigi De Crecchio 7, 80138 Naples, Italy; (L.M.); (R.V.T.); (R.G.); (F.C.); (M.P.); (M.P.)
| |
Collapse
|
6
|
Rahban M, Joushi S, Bashiri H, Saso L, Sheibani V. Characterization of prevalent tyrosine kinase inhibitors and their challenges in glioblastoma treatment. Front Chem 2024; 11:1325214. [PMID: 38264122 PMCID: PMC10804459 DOI: 10.3389/fchem.2023.1325214] [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: 10/20/2023] [Accepted: 12/21/2023] [Indexed: 01/25/2024] Open
Abstract
Glioblastoma multiforme (GBM) is a highly aggressive malignant primary tumor in the central nervous system. Despite extensive efforts in radiotherapy, chemotherapy, and neurosurgery, there remains an inadequate level of improvement in treatment outcomes. The development of large-scale genomic and proteomic analysis suggests that GBMs are characterized by transcriptional heterogeneity, which is responsible for therapy resistance. Hence, knowledge about the genetic and epigenetic heterogeneity of GBM is crucial for developing effective treatments for this aggressive form of brain cancer. Tyrosine kinases (TKs) can act as signal transducers, regulate important cellular processes like differentiation, proliferation, apoptosis and metabolism. Therefore, TK inhibitors (TKIs) have been developed to specifically target these kinases. TKIs are categorized into allosteric and non-allosteric inhibitors. Irreversible inhibitors form covalent bonds, which can lead to longer-lasting effects. However, this can also increase the risk of off-target effects and toxicity. The development of TKIs as therapeutics through computer-aided drug design (CADD) and bioinformatic techniques enhance the potential to improve patients' survival rates. Therefore, the continued exploration of TKIs as drug targets is expected to lead to even more effective and specific therapeutics in the future.
Collapse
Affiliation(s)
- Mahdie Rahban
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Sara Joushi
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Hamideh Bashiri
- Physiology Research Center, Institute of Neuropharmacology, Department of Physiology and Pharmacology, Medical School, Kerman University of Medical Sciences, Kerman, Iran
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University, Rome, Italy
| | - Vahid Sheibani
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
7
|
Alizzi Z, Roxburgh P, Cartwright D, McLaren A, Park S, Jones R, Greening S, Hudson E, Green C, Gray S, Khalique S, Karteris E, Hall M. Description of a Retrospective Cohort of Epithelial Ovarian Cancer Patients with Brain Metastases: Evaluation of the Role of PARP Inhibitors in this Setting. J Clin Med 2023; 12:jcm12072497. [PMID: 37048581 PMCID: PMC10095324 DOI: 10.3390/jcm12072497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 02/28/2023] [Accepted: 03/15/2023] [Indexed: 03/29/2023] Open
Abstract
Background: The incidence of brain metastases (BM) in patients with epithelial ovarian cancer (EOC) is low: 0.3–11%. The onset of BM has been regarded as a late event with limited treatment options and poor prognosis. This retrospective case series aims to explore the current management strategies with particular emphasis on the use of PARP inhibitors and outcomes, as well as identification of other prognostic indicators. Methods: A total of 39 ovarian cancer patients with brain metastases were identified from eight cancer centres in the UK. Clinical characteristics, details of management, and survival data were collected. Results: A total of 14/39 had BM as their first site of relapse. The majority (29 patients) received systemic treatments in addition to local radiotherapy (RT)/surgery. Nineteen patients had BRCA mutations (one somatic), one had a RAD51C mutation, and eighteen were BRCA wild type; one was unknown. A total of 14/39 patients received maintenance PARP inhibitors. As is well known, patients who received PARPi had consistently better outcomes. This was no different for those who received PARPi as part of the management of their BM. Platinum sensitivity and receiving more than one modality of therapy (e.g., radiation +/− chemotherapy and PARPi) for BM were also good prognostic indicators. Median PFS/OS for those treated with chemotherapy and either RT or surgery, then PARP inhibitor maintenance, have not been reached after a median of 33 months follow up. Conclusions: As with abdominal relapse, maintenance treatment with PARP inhibitors also has a valuable role in managing BMs in EOC patients.
Collapse
Affiliation(s)
- Zena Alizzi
- Mount Vernon Cancer Centre, Rickmansworth Road, Northwood HA6 2RN, UK
| | - Patricia Roxburgh
- Beatson West of Scotland Cancer Centre, University of Glasgow, Glasgow G12 0YN, UK
| | - Douglas Cartwright
- Beatson West of Scotland Cancer Centre, University of Glasgow, Glasgow G12 0YN, UK
| | - Alistair McLaren
- Beatson West of Scotland Cancer Centre, University of Glasgow, Glasgow G12 0YN, UK
| | - Sarah Park
- Royal Cornwall Hospitals NHS Trust, Truro TR1 3LJ, UK
| | - Rachel Jones
- South West Wales Cancer Centre, Swansea SA2 8QA, UK
| | | | - Emma Hudson
- Velindre Cancer Centre, Cardiff CF14 2TL, UK
| | - Clare Green
- University Hospital Southampton, Southampton SO16 6YD, UK
| | - Simon Gray
- Lancashire Teaching Hospitals NHS Foundation Trust, Preston PR2 9HT, UK
| | - Saira Khalique
- Mount Vernon Cancer Centre, Rickmansworth Road, Northwood HA6 2RN, UK
- College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK
| | - Emmanouil Karteris
- College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK
| | - Marcia Hall
- Mount Vernon Cancer Centre, Rickmansworth Road, Northwood HA6 2RN, UK
- College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK
- Correspondence:
| |
Collapse
|
8
|
Wu C, Shen Y, Shi L, Zhang J, Guo T, Zhou L, Wang W, Zhang X, Yu R, Liu X. UBA1 inhibition contributes radiosensitization of glioblastoma cells via blocking DNA damage repair. Front Pharmacol 2023; 14:1073929. [PMID: 36959858 PMCID: PMC10027716 DOI: 10.3389/fphar.2023.1073929] [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: 10/19/2022] [Accepted: 02/24/2023] [Indexed: 03/09/2023] Open
Abstract
Glioblastoma multiforme (GBM) is a brain tumor with high mortality and recurrence rate. Radiotherapy and chemotherapy after surgery are the main treatment options available for GBM. However, patients with glioblastoma have a grave prognosis. The major reason is that most GBM patients are resistant to radiotherapy. UBA1 is considered an attractive potential anti-tumor therapeutic target and a key regulator of DNA double-strand break repair and genome replication in human cells. Therefore, we hypothesized that TAK-243, the first-in-class UBA1 inhibitor, might increase GBM sensitivity to radiation. The combined effect of TAK-243 and ionizing radiation on GBM cell proliferation, and colony formation ability was detected using CCK-8, colony formation, and EdU assays. The efficacy of TAK-243 combined with ionizing radiation for GBM was further evaluated in vivo, and the mechanism of TAK-243 sensitizing radiotherapy was preliminarily discussed. The results showed that TAK-243, in combination with ionizing radiation, significantly inhibited GBM cell proliferation, colony formation, cell cycle arrest in the G2/M phase, and increased the proportion of apoptosis. In addition, UBA1 inhibition by TAK-243 substantially increased the radiation-induced γ-H2AX expression and impaired the recruitment of the downstream effector molecule 53BP1. Therefore, TAK-243 inhibited the radiation-induced DNA double-strand break repair and thus inhibited the growth of GBM cells. Our results provided a new therapeutic strategy for improving the radiation sensitivity of GBM and laid a theoretical foundation and experimental basis for further clinical trials.
Collapse
Affiliation(s)
- Changyong Wu
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yang Shen
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Lin Shi
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of general surgery, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Junhao Zhang
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Tongxuan Guo
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Lingni Zhou
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Wanzhou Wang
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xu Zhang
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Rutong Yu
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- *Correspondence: Rutong Yu, ; Xuejiao Liu,
| | - Xuejiao Liu
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- *Correspondence: Rutong Yu, ; Xuejiao Liu,
| |
Collapse
|
9
|
Peng X, Zhang S, Wang Y, Zhou Z, Yu Z, Zhong Z, Zhang L, Chen Z, Claret FX, Elkabets M, Wang F, Sun F, Wang R, Liang H, Lin H, Kong D. Stellettin B Sensitizes Glioblastoma to DNA-Damaging Treatments by Suppressing PI3K-Mediated Homologous Recombination Repair. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205529. [PMID: 36453577 PMCID: PMC9875605 DOI: 10.1002/advs.202205529] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/29/2022] [Indexed: 06/02/2023]
Abstract
Glioblastoma (GBM) is the most aggressive type of cancer. Its current first-line postsurgery regimens are radiotherapy and temozolomide (TMZ) chemotherapy, both of which are DNA damage-inducing therapies but show very limited efficacy and a high risk of resistance. There is an urgent need to develop novel agents to sensitize GBM to DNA-damaging treatments. Here it is found that the triterpene compound stellettin B (STELB) greatly enhances the sensitivity of GBM to ionizing radiation and TMZ in vitro and in vivo. Mechanistically, STELB inhibits the expression of homologous recombination repair (HR) factors BRCA1/2 and RAD51 by promoting the degradation of PI3Kα through the ubiquitin-proteasome pathway; and the induced HR deficiency then leads to augmented DNA damage and cell death. It is further demonstrated that STELB has the potential to rapidly penetrate the blood-brain barrier to exert anti-GBM effects in the brain, based on zebrafish and nude mouse orthotopic xenograft tumor models. The study provides strong evidence that STELB represents a promising drug candidate to improve GBM therapy in combination with DNA-damaging treatments.
Collapse
Affiliation(s)
- Xin Peng
- Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and DiagnosticsSchool of PharmacyTianjin Medical UniversityTianjin300070China
- Key Laboratory of Immune Microenvironment and Diseases (Ministry of Education)Tianjin Medical UniversityTianjin300070China
- Department of Bioinformatics and Computational BiologyThe University of Texas MD Anderson Cancer CenterHoustonTX77030USA
- Department of Systems Biologythe University of Texas MD Anderson Cancer CenterHoustonTX77030USA
| | - Shaolu Zhang
- Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and DiagnosticsSchool of PharmacyTianjin Medical UniversityTianjin300070China
- Key Laboratory of Immune Microenvironment and Diseases (Ministry of Education)Tianjin Medical UniversityTianjin300070China
| | - Yingying Wang
- Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and DiagnosticsSchool of PharmacyTianjin Medical UniversityTianjin300070China
- Key Laboratory of Immune Microenvironment and Diseases (Ministry of Education)Tianjin Medical UniversityTianjin300070China
| | - Zhicheng Zhou
- Department of Bioinformatics and Computational BiologyThe University of Texas MD Anderson Cancer CenterHoustonTX77030USA
- Department of Systems Biologythe University of Texas MD Anderson Cancer CenterHoustonTX77030USA
| | - Zixiang Yu
- Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and DiagnosticsSchool of PharmacyTianjin Medical UniversityTianjin300070China
- Key Laboratory of Immune Microenvironment and Diseases (Ministry of Education)Tianjin Medical UniversityTianjin300070China
| | - Zhenxing Zhong
- Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and DiagnosticsSchool of PharmacyTianjin Medical UniversityTianjin300070China
- Key Laboratory of Immune Microenvironment and Diseases (Ministry of Education)Tianjin Medical UniversityTianjin300070China
| | - Liang Zhang
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Zhe‐Sheng Chen
- Department of Pharmaceutical SciencesCollege of Pharmacy and Health SciencesSt. John's UniversityQueensNY11439USA
| | - Francois X. Claret
- Department of Bioinformatics and Computational BiologyThe University of Texas MD Anderson Cancer CenterHoustonTX77030USA
| | - Moshe Elkabets
- The Shraga Segal Department of MicrobiologyImmunology and GeneticsFaculty of Health SciencesBen‐Gurion University of the NegevBeer‐Sheva84105Israel
| | - Feng Wang
- Department of GeneticsSchool of Basic Medical SciencesTianjin Medical UniversityTianjin300070China
| | - Fan Sun
- Research Center for Marine DrugsState Key Laboratory of Oncogenes and Related GenesDepartment of PharmacyRenji HospitalSchool of MedicineShanghai Jiaotong UniversityShanghai200127China
| | - Ran Wang
- Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and DiagnosticsSchool of PharmacyTianjin Medical UniversityTianjin300070China
- Key Laboratory of Immune Microenvironment and Diseases (Ministry of Education)Tianjin Medical UniversityTianjin300070China
| | - Han Liang
- Department of Bioinformatics and Computational BiologyThe University of Texas MD Anderson Cancer CenterHoustonTX77030USA
- Department of Systems Biologythe University of Texas MD Anderson Cancer CenterHoustonTX77030USA
| | - Hou‐Wen Lin
- Research Center for Marine DrugsState Key Laboratory of Oncogenes and Related GenesDepartment of PharmacyRenji HospitalSchool of MedicineShanghai Jiaotong UniversityShanghai200127China
| | - Dexin Kong
- Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and DiagnosticsSchool of PharmacyTianjin Medical UniversityTianjin300070China
- Key Laboratory of Immune Microenvironment and Diseases (Ministry of Education)Tianjin Medical UniversityTianjin300070China
| |
Collapse
|
10
|
Daddacha W, Monroe D, Carver K, Usoro ER, Alptekin A, Xu H, Osuka S, Arbab AS, Sakamuro D. Viral Particle-Mediated SAMHD1 Depletion Sensitizes Refractory Glioblastoma to DNA-Damaging Therapeutics by Impairing Homologous Recombination. Cancers (Basel) 2022; 14:4490. [PMID: 36139652 PMCID: PMC9497202 DOI: 10.3390/cancers14184490] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/09/2022] [Accepted: 09/13/2022] [Indexed: 12/04/2022] Open
Abstract
The current standard-of-care treatment for glioblastoma includes DNA damaging agents, γ-irradiation (IR) and temozolomide (TMZ). These treatments fail frequently and there is limited alternative strategy. Therefore, identifying a new therapeutic target is urgently needed to develop a strategy that improves the efficacy of the existing treatments. Here, we report that tumor samples from GBM patients express a high level of SAMHD1, emphasizing SAMHD1's importance. The depletion of SAMHD1 using virus-like particles containing Vpx, VLP(+Vpx), sensitized two independent GBM cell lines (LN-229 and U-87) to veliparib, a well-established PARP inhibitor, and slowed cell growth in a dose-dependent manner. In the mouse GBM xenograft model, Vpx-mediated SAMHD1 depletion reduced tumor growth and SAMHD1 knockout (KO) improved survival. In combination with IR or TMZ, SAMHD1 KO and exposure to 50% growth inhibitory dose (gID50) of VLP(+Vpx) displayed a synergistic effect, resulting in impaired HR, and improved LN-229 cells' sensitivity to TMZ and IR. In conclusion, our finding demonstrates that SAMHD1 promotes GBM resistance to treatment, and it is a plausible therapeutic target to improve the efficacy of TMZ and IR in GBM. Furthermore, we show that Vpx could be a potential therapeutic tool that can be utilized to deplete SAMHD1 in GBM.
Collapse
Affiliation(s)
- Waaqo Daddacha
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA
| | - Dominique Monroe
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA
| | - Kristen Carver
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Department of Population Health Sciences, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Edidiong R. Usoro
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA
| | - Ahmet Alptekin
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA
| | - Hongyan Xu
- Department of Population Health Sciences, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Satoru Osuka
- Department of Neurosurgery, Heersink School of Medicine, The University of Alabama, Birmingham, AL 35233, USA
| | - Ali S. Arbab
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA
| | - Daitoku Sakamuro
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
11
|
Wang Y, Sun Y, Geng N, Zheng M, Zou Y, Shi B. A Biomimetic Nanomedicine Targets Orthotopic Glioblastoma by Combinatorial Co‐delivery of Temozolomide and a Methylguanine‐DNA Methyltransferase Inhibitor. ADVANCED THERAPEUTICS 2022. [DOI: 10.1002/adtp.202200095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Yibin Wang
- Henan‐Macquarie Uni Joint Centre for Biomedical Innovation School of Life Sciences Henan University Kaifeng Henan 475004 China
- Henan Key Laboratory of Brain Targeted Bio‐nanomedicine School of Life Sciences Henan University Kaifeng Henan 475004 China
| | - Yajing Sun
- Henan‐Macquarie Uni Joint Centre for Biomedical Innovation School of Life Sciences Henan University Kaifeng Henan 475004 China
- Henan Key Laboratory of Brain Targeted Bio‐nanomedicine School of Life Sciences Henan University Kaifeng Henan 475004 China
| | - Nan Geng
- Henan‐Macquarie Uni Joint Centre for Biomedical Innovation School of Life Sciences Henan University Kaifeng Henan 475004 China
- Henan Key Laboratory of Brain Targeted Bio‐nanomedicine School of Life Sciences Henan University Kaifeng Henan 475004 China
| | - Meng Zheng
- Henan‐Macquarie Uni Joint Centre for Biomedical Innovation School of Life Sciences Henan University Kaifeng Henan 475004 China
- Henan Key Laboratory of Brain Targeted Bio‐nanomedicine School of Life Sciences Henan University Kaifeng Henan 475004 China
| | - Yan Zou
- Henan‐Macquarie Uni Joint Centre for Biomedical Innovation School of Life Sciences Henan University Kaifeng Henan 475004 China
- Henan Key Laboratory of Brain Targeted Bio‐nanomedicine School of Life Sciences Henan University Kaifeng Henan 475004 China
- Macquarie Medical School Faculty of Medicine Health and Human Sciences Macquarie University Sydney NSW 2109 Australia
| | - Bingyang Shi
- Henan‐Macquarie Uni Joint Centre for Biomedical Innovation School of Life Sciences Henan University Kaifeng Henan 475004 China
- Henan Key Laboratory of Brain Targeted Bio‐nanomedicine School of Life Sciences Henan University Kaifeng Henan 475004 China
- Macquarie Medical School Faculty of Medicine Health and Human Sciences Macquarie University Sydney NSW 2109 Australia
| |
Collapse
|
12
|
Blankenstein LJ, Cordes N, Kunz-Schughart LA, Vehlow A. Targeting of p21-Activated Kinase 4 Radiosensitizes Glioblastoma Cells via Impaired DNA Repair. Cells 2022; 11:cells11142133. [PMID: 35883575 PMCID: PMC9316146 DOI: 10.3390/cells11142133] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/05/2022] [Accepted: 07/06/2022] [Indexed: 01/27/2023] Open
Abstract
Glioblastoma is a devastating malignant disease with poor patient overall survival. Strong invasiveness and resistance to radiochemotherapy have challenged the identification of molecular targets that can finally improve treatment outcomes. This study evaluates the influence of all six known p21-activated kinase (PAK) protein family members on the invasion capacity and radio-response of glioblastoma cells by employing a siRNA-based screen. In a panel of human glioblastoma cell models, we identified PAK4 as the main PAK isoform regulating invasion and clonogenic survival upon irradiation and demonstrated the radiosensitizing potential of PAK4 inhibition. Mechanistically, we show that PAK4 depletion and pharmacological inhibition enhanced the number of irradiation-induced DNA double-strand breaks and reduced the expression levels of various DNA repair proteins. In conclusion, our data suggest PAK4 as a putative target for radiosensitization and impairing DNA repair in glioblastoma, deserving further scrutiny in extended combinatorial treatment testing.
Collapse
Affiliation(s)
- Leon J. Blankenstein
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, PF 41, 01307 Dresden, Germany; (L.J.B.); (N.C.); (L.A.K.-S.)
- National Center for Tumor Diseases, Partner Site Dresden: German Cancer Research Center, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Nils Cordes
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, PF 41, 01307 Dresden, Germany; (L.J.B.); (N.C.); (L.A.K.-S.)
- National Center for Tumor Diseases, Partner Site Dresden: German Cancer Research Center, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- Department of Radiotherapy and Radiation Oncology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, PF 50, 01307 Dresden, Germany
- Helmholtz-Zentrum Dresden—Rossendorf, Institute of Radiooncology—OncoRay, Bautzner Landstr. 400, 01328 Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69192 Heidelberg, Germany
| | - Leoni A. Kunz-Schughart
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, PF 41, 01307 Dresden, Germany; (L.J.B.); (N.C.); (L.A.K.-S.)
- National Center for Tumor Diseases, Partner Site Dresden: German Cancer Research Center, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Anne Vehlow
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, PF 41, 01307 Dresden, Germany; (L.J.B.); (N.C.); (L.A.K.-S.)
- National Center for Tumor Diseases, Partner Site Dresden: German Cancer Research Center, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69192 Heidelberg, Germany
- Correspondence:
| |
Collapse
|
13
|
Immunotherapy for Neuro-oncology. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1342:233-258. [PMID: 34972967 DOI: 10.1007/978-3-030-79308-1_7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Immunotherapy has changed the landscape of treatment of many solid and hematological malignancies and is at the forefront of cancer breakthroughs. Several circumstances unique to the central nervous system (CNS) such as limited space for an inflammatory response, difficulties with repeated sampling, corticosteroid use for management of cerebral edema, and immunosuppressive mechanisms within the tumor and brain parenchyma have posed challenges in clinical development of immunotherapy for intracranial tumors. Nonetheless, the success of immunotherapy in brain metastases (BMs) from solid cancers such as melanoma and non-small cell lung cancer (NSCLC) proves that the CNS is not an immune-privileged organ and is capable of initiating and regulating immune responses that lead to tumor control. However, the development of immunotherapeutics for the most malignant primary brain tumor, glioblastoma (GBM), has been challenging due to systemic and profound tumor-mediated immunosuppression unique to GBM, intratumoral and intertumoral heterogeneity, and lack of stably expressed clonal antigens. Here, we review recent advances in the field of immunotherapy for neuro-oncology with a focus on BM, GBM, and rare CNS cancers.
Collapse
|
14
|
Fougner V, Hasselbalch B, Lassen U, Weischenfeldt J, Poulsen HS, Urup T. Implementing targeted therapies in the treatment of glioblastoma: Previous shortcomings, future promises, and a multimodal strategy recommendation. Neurooncol Adv 2022; 4:vdac157. [PMID: 36325372 PMCID: PMC9616055 DOI: 10.1093/noajnl/vdac157] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2023] Open
Abstract
The introduction of targeted therapies to the field of oncology has prolonged the survival of several tumor types. Despite extensive research and numerous trials, similar outcomes have unfortunately not been realized for glioblastoma. For more than 15 years, the standard treatment of glioblastoma has been unchanged. This review walks through the elements that have challenged the success of previous trials and highlight some future promises. Concurrently, this review describes how institutions, through a multimodal and comprehensive strategy with 4 essential components, may increase the probability of finding a meaningful role for targeted therapies in the treatment of glioblastoma. These components are (1) prudent trial designs, (2) considered drug and target selection, (3) harnessed real-world clinical and molecular evidence, and (4) incorporation of translational research.
Collapse
Affiliation(s)
- Vincent Fougner
- Department for Cancer Treatment, DCCC—Brain Tumor Center, Rigshospitalet, Copenhagen, Capitol Region of Denmark, Denmark
| | - Benedikte Hasselbalch
- Department for Cancer Treatment, DCCC—Brain Tumor Center, Rigshospitalet, Copenhagen, Capitol Region of Denmark, Denmark
| | - Ulrik Lassen
- Department for Cancer Treatment, DCCC—Brain Tumor Center, Rigshospitalet, Copenhagen, Capitol Region of Denmark, Denmark
| | - Joachim Weischenfeldt
- BRIC - Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Hans Skovgaard Poulsen
- Department for Cancer Treatment, DCCC—Brain Tumor Center, Rigshospitalet, Copenhagen, Capitol Region of Denmark, Denmark
| | - Thomas Urup
- Department for Cancer Treatment, DCCC—Brain Tumor Center, Rigshospitalet, Copenhagen, Capitol Region of Denmark, Denmark
| |
Collapse
|
15
|
Zampieri LX, Sboarina M, Cacace A, Grasso D, Thabault L, Hamelin L, Vazeille T, Dumon E, Rossignol R, Frédérick R, Sonveaux E, Lefranc F, Sonveaux P. Olaparib Is a Mitochondrial Complex I Inhibitor That Kills Temozolomide-Resistant Human Glioblastoma Cells. Int J Mol Sci 2021; 22:ijms222111938. [PMID: 34769368 PMCID: PMC8584761 DOI: 10.3390/ijms222111938] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/28/2021] [Accepted: 10/29/2021] [Indexed: 12/15/2022] Open
Abstract
Glioblastoma represents the highest grade of brain tumors. Despite maximal resection surgery associated with radiotherapy and concomitant followed by adjuvant chemotherapy with temozolomide (TMZ), patients have a very poor prognosis due to the rapid recurrence and the acquisition of resistance to TMZ. Here, initially considering that TMZ is a prodrug whose activation is pH-dependent, we explored the contribution of glioblastoma cell metabolism to TMZ resistance. Using isogenic TMZ-sensitive and TMZ-resistant human glioblastoma cells, we report that the expression of O6-methylguanine DNA methyltransferase (MGMT), which is known to repair TMZ-induced DNA methylation, does not primarily account for TMZ resistance. Rather, fitter mitochondria in TMZ-resistant glioblastoma cells are a direct cause of chemoresistance that can be targeted by inhibiting oxidative phosphorylation and/or autophagy/mitophagy. Unexpectedly, we found that PARP inhibitor olaparib, but not talazoparib, is also a mitochondrial Complex I inhibitor. Hence, we propose that the anticancer activities of olaparib in glioblastoma and other cancer types combine DNA repair inhibition and impairment of cancer cell respiration.
Collapse
Affiliation(s)
- Luca X. Zampieri
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (L.X.Z.); (M.S.); (A.C.); (D.G.); (L.T.); (L.H.); (T.V.)
| | - Martina Sboarina
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (L.X.Z.); (M.S.); (A.C.); (D.G.); (L.T.); (L.H.); (T.V.)
| | - Andrea Cacace
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (L.X.Z.); (M.S.); (A.C.); (D.G.); (L.T.); (L.H.); (T.V.)
| | - Debora Grasso
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (L.X.Z.); (M.S.); (A.C.); (D.G.); (L.T.); (L.H.); (T.V.)
| | - Léopold Thabault
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (L.X.Z.); (M.S.); (A.C.); (D.G.); (L.T.); (L.H.); (T.V.)
- Louvain Drug Research Institute (LDRI), UCLouvain, 1200 Brussels, Belgium; (R.F.); (E.S.)
| | - Loïc Hamelin
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (L.X.Z.); (M.S.); (A.C.); (D.G.); (L.T.); (L.H.); (T.V.)
| | - Thibaut Vazeille
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (L.X.Z.); (M.S.); (A.C.); (D.G.); (L.T.); (L.H.); (T.V.)
| | - Elodie Dumon
- INSERM U1211, Laboratory of Rare Diseases, Metabolism and Genetics (MRGM), Ecole des Sages Femmes, Bordeaux University, 33076 Bordeaux, France; (E.D.); (R.R.)
| | - Rodrigue Rossignol
- INSERM U1211, Laboratory of Rare Diseases, Metabolism and Genetics (MRGM), Ecole des Sages Femmes, Bordeaux University, 33076 Bordeaux, France; (E.D.); (R.R.)
| | - Raphaël Frédérick
- Louvain Drug Research Institute (LDRI), UCLouvain, 1200 Brussels, Belgium; (R.F.); (E.S.)
| | - Etienne Sonveaux
- Louvain Drug Research Institute (LDRI), UCLouvain, 1200 Brussels, Belgium; (R.F.); (E.S.)
| | - Florence Lefranc
- Service de Neurochirurgie, Hôpital Erasme, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium;
| | - Pierre Sonveaux
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (L.X.Z.); (M.S.); (A.C.); (D.G.); (L.T.); (L.H.); (T.V.)
- Correspondence:
| |
Collapse
|