1
|
Garrett MC, Carnwath T, Albano R, Zhuang Y, Behrmann CA, Pemberton M, Barakat F, Lober R, Hoeprich M, Paravati A, Reed M, Spry H, Woo D, O'Brien E, VanCauwenbergh B, Perentesis J, Nasser R, Medvedovic M, Plas DR. CPI203, a BET inhibitor, down-regulates a consistent set of DNA synthesis genes across a wide array of glioblastoma lines. PLoS One 2025; 20:e0306846. [PMID: 40378113 DOI: 10.1371/journal.pone.0306846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 03/15/2025] [Indexed: 05/18/2025] Open
Abstract
INTRODUCTION Glioblastomas utilize malignant gene expression pathways to drive growth. Many of these gene pathways are not directly accessible with molecularly targeted pharmacological agents. Chromatin-modifying compounds can alter gene expression and target glioblastoma growth pathways. In this study, we utilize a systematic screen of chromatin-modifying compounds on a panel of patient-derived glioblastoma lines to identify promising compounds and their associated gene targets. METHODS Five glioblastoma cell lines were subjected to a drug screen of 106 chromatin-modifying compounds representing 36 unique drug classes to determine the twelve most promising drug classes and the best candidate inhibitors in each class. These twelve drugs were then tested with a panel of twelve patient-derived gliomasphere lines to identify growth inhibition and corresponding gene expression patterns. Overlap analysis and weighted co-expression network analysis (WCGNA) were utilized to determine potential target genes and gene pathways. RESULTS The initial drug screen identified twelve candidate pharmacologic agents for further testing. Drug sensitivity testing indicated an overall high degree of variability between gliomasphere lines. However, CPI203 was the most consistently effective compound, and the BET inhibitor class was the most consistently effective class of compounds across the gliomasphere panel. Correspondingly, most of the compounds tested had highly variable effects on gene expression between gliomasphere lines. CPI203 stood out as the only compound to induce a consistent effect on gene expression across different gliomasphere lines, specifically down-regulation of DNA-synthesis genes. Amongst the twelve tested cell lines, high expression of CDKN2A and CDKN2B distinguished more drug sensitive from more drug resistant lines. WCGNA identified two oncogenic gene modules (FBXO5 and MELK) that were effectively downregulated by CPI203 (FBXO5) and ML228 (FBXO5 and MELK). CONCLUSIONS The bromodomain inhibitor CPI203 induced relatively consistent effects on gene expression and growth across a variety of glioblastoma lines, specifically down-regulating genes associated with DNA replication. We propose that clinically effective BET inhibitors have the potential to induce consistent beneficial effects across a spectrum of glioblastomas.
Collapse
Affiliation(s)
- Matthew C Garrett
- Department of Neurosurgery, Kettering Health Network, Kettering, Ohio, United States of America
| | - Troy Carnwath
- University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Rebecca Albano
- Department of Neurosurgery, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Yonghua Zhuang
- Biostatistics and Bioinformatics Shared Resource, University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Catherine A Behrmann
- Department of Cancer Biology, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Merissa Pemberton
- Department of Cancer Biology, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Farah Barakat
- Alfaisal University, College of Medicine, Riyadh, Saudi Arabia
| | - Robert Lober
- Division of Neurosurgery, Dayton Children's Hospital, Dayton, Ohio, United States of America
| | - Mark Hoeprich
- Department of Neurosurgery, Kettering Health Network, Kettering, Ohio, United States of America
| | - Anthony Paravati
- Department of Radiation Oncology, Kettering Health Network, Kettering, Ohio, United States of America
| | - Marilyn Reed
- Department of Neurosurgery, Kettering Health Network, Kettering, Ohio, United States of America
| | - Hailey Spry
- Department of Neurosurgery, Kettering Health Network, Kettering, Ohio, United States of America
| | - Daniel Woo
- Department of Neurology, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Eric O'Brien
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Brett VanCauwenbergh
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - John Perentesis
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Rani Nasser
- Department of Neurosurgery, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Mario Medvedovic
- Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - David R Plas
- Department of Cancer Biology, University of Cincinnati, Cincinnati, Ohio, United States of America
| |
Collapse
|
2
|
Acanda de la Rocha AM, Berlow NE, Azzam DJ. Functional precision medicine: the future of cancer care. Trends Mol Med 2025; 31:404-408. [PMID: 39567286 PMCID: PMC12084147 DOI: 10.1016/j.molmed.2024.10.015] [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: 08/12/2024] [Revised: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 11/22/2024]
Abstract
Functional precision medicine (FPM), combining ex vivo drug sensitivity testing with genomic profiling to identify treatment options for recurrent/refractory cancer, is feasible and poised to accelerate. This forum explores the history of FPM, recent clinical advancements, and barriers to expanding the clinical utility and accessibility for pediatric/adolescent and adult cancers.
Collapse
Affiliation(s)
- Arlet M Acanda de la Rocha
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, USA
| | | | - Diana J Azzam
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, USA.
| |
Collapse
|
3
|
Herzog TJ, Krivak TC, Bush S, Diaz JP, Lentz S, Nair N, Zgheib NB, Gunderson-Jackson C, Barve A, Denning KL, Lirette ST, Howard CM, Valluri J, Claudio PP. ChemoID-guided therapy improves objective response rate in recurrent platinum-resistant ovarian cancer randomized clinical trial. NPJ Precis Oncol 2025; 9:86. [PMID: 40133484 PMCID: PMC11937309 DOI: 10.1038/s41698-025-00874-0] [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: 09/06/2024] [Accepted: 03/10/2025] [Indexed: 03/27/2025] Open
Abstract
Patients with recurrent platinum-resistant ovarian cancer (PROC) have poor clinical outcomes, owing mainly to the presence of therapy-resistant cancer stem cells (CSCs). The NCT03949283 randomized clinical trial enrolled patients with recurrent PROC to receive ChemoID-guided chemotherapy or the best physician-choice regimen selected from the same list of thirteen mono or combination chemotherapies. The primary outcome was objective response rate (ORR) assessed on CT scans using the RECIST 1.1 criteria at 6 months follow-up. Subjects treated with the ChemoID assay had an ORR of 55% (CI95 39% - 73%), compared to 5% (CI95 0% - 11%) for those treated with physician's choice chemotherapy (p <0.0001). Secondary endpoints of duration of response (DOR) and progression-free survival (PFS) of subjects treated with chemotherapies guided by the ChemoID assay versus physician's choice chemotherapy were a median of 8 months vs. 5.5 months (p <0.0001), and 11.0 months (CI95 8.0- NA) vs 3.0 months (CI95 2.0- 3.5) with 27% of hazard ratio (CI95, 0.15-0.49; p <0.001), respectively.
Collapse
Affiliation(s)
- Thomas J Herzog
- Department of Obstetrics and Gynecology, University of Cincinnati Cancer Center, Cincinnati, USA
| | - Thomas C Krivak
- Division of Gynecologic Oncology, Allegheny Health Network Cancer Institute, Pittsburgh, USA
| | - Stephen Bush
- Department of Obstetrics and Gynecology, Charleston Area Medical Center Vandalia Health, Charleston, USA
| | - John P Diaz
- Gynecologic Oncology, Baptist Health South Florida, Miami Cancer Institute, Miami, USA
| | - Scott Lentz
- Gynecology Oncology Department, Kaiser Permanente Los Angeles Medical Center, Los Angeles, USA
| | - Navya Nair
- Department of Obstetrics and Gynecology, Section of Gynecologic Oncology, Louisiana State University, New Orleans, USA
- Currently, Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Miami Sylvester Cancer Center, Miami, USA
| | - Nadim Bou Zgheib
- Edwards Comprehensive Cancer Center, Joan C. Edwards School of Medicine, Marshall University, Huntington, USA
| | - Camille Gunderson-Jackson
- Department of Obstetrics and Gynecology, Section of Gynecologic Oncology, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, USA
- Currently, Mercy Clinic Gynecologic Oncology, Oklahoma City, USA
| | - Abhijit Barve
- Clinical Development & Medical Affairs, Viatris Inc, Canonsburg, USA
| | - Krista L Denning
- Department of Pathology, Joan C. Edwards School of Medicine, Marshall University, Huntington, USA
| | - Seth T Lirette
- Department of Data Science, University of Mississippi Medical Center, Translational Research Center, Jackson, USA
| | - Candace M Howard
- Department of Radiology, University of Mississippi Medical Center, Jackson, USA
| | - Jagan Valluri
- Cordgenics, LLC, Huntington, USA
- Department of Biological Sciences, Marshall University, Huntington, USA
| | - Pier Paolo Claudio
- Cordgenics, LLC, Huntington, USA.
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, USA.
| |
Collapse
|
4
|
Yuan Z, Guo M, Zhang Y, Deng Y, Sun B, Hou Y, Wang X, Jin X, Liu Y, Shi B, Yin J. MAGL targeted PROTAC degrader simultaneously enhances P53 for synergistic treatment of glioblastoma stem cell. Cell Death Discov 2025; 11:109. [PMID: 40113745 PMCID: PMC11926070 DOI: 10.1038/s41420-025-02392-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 02/13/2025] [Accepted: 03/10/2025] [Indexed: 03/22/2025] Open
Abstract
Glioblastoma (GBM) stands as the most fatal brain tumor due to limited therapeutic options and high rates of drug resistance. Current surgical and pharmacological interventions usually fail to eradicate the aggressive GBM stem cells (GSCs), which leads to the deadly GBM occurrence. Although proteolysis-targeting chimeras (PROTACs) are prosperous in drug development for tumors, their application in GBM, particularly for GSC-sensitive drug candidates remains in its nascent stages. In this regard, we designed a monoacylglycerol lipase (MAGL) targeting PROTAC, where MAGL was identified as a novel target for GSCs in our previous study. The MAGL inhibitor JZL184 was redesigned by leveraging computational chemistry analysis, and an active unit was engaged for conjugation. E3 ligand for MAGL targeted warhead conjugation was screened with bioinformatics analyses, which revealed heightened activity of the E3 ligase MDM2 in GBM, a classic negative regulator of the tumor suppressor P53, which correlates with patient prognosis. Then the PROTAC was conjugated with JZL184 analog and the MDM2 inhibitor Nutlin-3 analog. Experimental results validated that the designed JN-PROTAC effectively induced MAGL targeted degradation and concomitantly enhanced P53 activation via MDM2 inhibition and is capable of inhibiting the progression of patient-derived GSCs in vivo. This work presents a proof-of-concept PROTAC design tailored for GSCs, potentially addressing the occurrence challenges for GBM.
Collapse
Affiliation(s)
- Zheng Yuan
- Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan, China
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, China
| | - Meixia Guo
- Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan, China
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, China
| | - Yue Zhang
- Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan, China
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, China
| | - Yilin Deng
- Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan, China
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, China
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Republic of Korea
| | - Biao Sun
- Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan, China
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, China
| | - Yaning Hou
- Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan, China
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, China
| | - Xin Wang
- Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan, China
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, China
| | - Xiong Jin
- Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan, China
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, China
| | - Yang Liu
- Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan, China.
- Huaihe Hospital of Henan University, Kaifeng, China.
| | - Bingyang Shi
- Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan, China.
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, China.
| | - Jinlong Yin
- Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan, China.
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, China.
- Huaihe Hospital of Henan University, Kaifeng, China.
| |
Collapse
|
5
|
Emir SM, Karaoğlan BS, Kaşmer R, Şirin HB, Sarıyıldız B, Karakaş N. Hunting glioblastoma recurrence: glioma stem cells as retrospective targets. Am J Physiol Cell Physiol 2025; 328:C1045-C1061. [PMID: 39818986 DOI: 10.1152/ajpcell.00344.2024] [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: 05/21/2024] [Revised: 06/11/2024] [Accepted: 01/07/2025] [Indexed: 01/19/2025]
Abstract
Glioblastoma (GBM) remains one of the most aggressive and treatment-resistant brain malignancies in adults. Standard approaches, including surgical resection followed by adjuvant radio- and chemotherapy with temozolomide (TMZ), provide only transient control, as GBM frequently recurs due to its infiltrative nature and the presence of therapy-resistant subpopulations such as glioma stem cells (GSCs). GSCs, with their quiescent state and robust resistance mechanisms, evade conventional therapies, contributing significantly to relapse. Consequently, current treatment methods for GBM face significant limitations in effectively targeting GSCs. In this review, we emphasize the relationship between GBM recurrence and GSCs, discuss the current limitations, and provide future perspectives to overwhelm the challenges associated with targeting GSCs. Eliminating GSCs may suppress recurrence, achieve durable responses, and improve therapeutic outcomes for patients with GBM.
Collapse
Affiliation(s)
- Sümeyra Mengüç Emir
- Cancer Research Center, Research Institute for Health Sciences and Technologies (SABITA), İstanbul Medipol University, Istanbul, Türkiye
| | - Birnur Sinem Karaoğlan
- Cancer Research Center, Research Institute for Health Sciences and Technologies (SABITA), İstanbul Medipol University, Istanbul, Türkiye
| | - Ramazan Kaşmer
- Cancer Research Center, Research Institute for Health Sciences and Technologies (SABITA), İstanbul Medipol University, Istanbul, Türkiye
| | - Hilal Buse Şirin
- Cancer Research Center, Research Institute for Health Sciences and Technologies (SABITA), İstanbul Medipol University, Istanbul, Türkiye
| | - Batuhan Sarıyıldız
- Cancer Research Center, Research Institute for Health Sciences and Technologies (SABITA), İstanbul Medipol University, Istanbul, Türkiye
| | - Nihal Karakaş
- Cancer Research Center, Research Institute for Health Sciences and Technologies (SABITA), İstanbul Medipol University, Istanbul, Türkiye
- Department of Medical Biology, International School of Medicine, İstanbul Medipol University, Istanbul, Türkiye
| |
Collapse
|
6
|
Connor K, Golebiewska A, Byrne AT. Challenging the status quo to improve the translational potential of preclinical oncology studies. Nat Rev Cancer 2025; 25:3-4. [PMID: 39375534 DOI: 10.1038/s41568-024-00756-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/09/2024]
Affiliation(s)
- Kate Connor
- Discipline of Physiology, School of Medicine, Trinity College Dublin, Dublin, Ireland
- RCSI Precision Cancer Medicine Group, Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
- National Preclinical Imaging Centre (NPIC), Dublin, Ireland
| | - Anna Golebiewska
- NORLUX Neuro-Oncology Laboratory, Department of Cancer Research, Luxembourg Institute of Health (LIH), Luxembourg, Luxembourg
| | - Annette T Byrne
- RCSI Precision Cancer Medicine Group, Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland.
- National Preclinical Imaging Centre (NPIC), Dublin, Ireland.
| |
Collapse
|
7
|
Berghoff AS, Mair MJ, Spiró Z, Abdel Malak C, El-Heliebi A, Eckert F, Furtner J, König F, Leibetseder A, Nowosielski M, Oberndorfer S, Prietl B, Pichler J, Pieber TR, Spiegl-Kreinecker S, Urbanic Purkart T, Wöhrer A, Widhalm G, Preusser M. Personalized targeted glioblastoma therapies by ex vivo drug screening: Study protocol of the Advanced brain Tumor TheRApy Clinical Trial (ATTRACT). Neurooncol Adv 2025; 7:vdaf056. [PMID: 40351832 PMCID: PMC12063086 DOI: 10.1093/noajnl/vdaf056] [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] [Indexed: 05/14/2025] Open
Abstract
Background Novel approaches to guide personalized treatment in glioblastoma are urgently needed. Given the poor predictive value of genetic biomarkers in glioblastoma, we are conducting a prospective clinical trial to investigate the novel approach of cultivated patient-derived tumor cells (PDCs) for ex vivo drug screening. Methods In this randomized phase 2 study, we are testing the ability of PDC-based ex vivo drug screening to formulate a personalized recommendation for maintenance treatment in patients with newly diagnosed glioblastoma with unmethylated MGMT promoter after combined radio-chemotherapy. Based on overall survival as the primary endpoint, we plan to include 240 patients (120 per group) to show with a power of 80% that we can increase the median survival from 12 to 17 months (hazard ratio 0.7). Patients will be randomized 1:1 to either the standard group (no drug screening) or the intervention group (drug screening and personalized recommendation for maintenance treatment). In the intervention group, automated drug screening will be performed on PDCs with 28 drugs used for the treatment of solid tumors and hematological malignancies. Based on the cytotoxic activity of these drugs, as quantified by relative viability based on adenosine triphosphate levels, a molecular tumor board will recommend a personalized treatment regimen. Results The first patient was enrolled in July 2024. Interim analysis of the ATTRACT study (NCT06512311) is expected in late 2027, and final results in 2030. Trial Registration The ATTRACT trial is registered under the ID NCT06512311 (https://clinicaltrials.gov/study/NCT06512311).
Collapse
Affiliation(s)
- Anna Sophie Berghoff
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Maximilian J Mair
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Zoltán Spiró
- Precision Medicine Technologies, CBmed GmbH, Graz, Austria
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Calvin Abdel Malak
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Amin El-Heliebi
- Gottfried Schatz Research Center, Division of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
| | - Franziska Eckert
- Department of Radiation Oncology, Medical University of Vienna, Vienna, Austria
| | - Julia Furtner
- Research Center for Medical Image Analysis and Artificial Intelligence, Faculty of Medicine and Dentistry, Danube Private University, Krems an der Donau, Austria
| | - Franz König
- Center For Medical Data Sciences, Medical University of Vienna, Vienna, Austria
| | - Annette Leibetseder
- Department of Neurology, Kepler University Hospital, Johannes Kepler University Linz, Linz, Austria
- Department of Internal Medicine and Neurooncology, Kepler University Hospital Linz, Neuromed Campus, Linz, Austria
| | - Martha Nowosielski
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Stefan Oberndorfer
- Clinical Division of Neurology, Karl Landsteiner University of Health Sciences, St. Pölten, Austria
| | - Barbara Prietl
- Precision Medicine Technologies, CBmed GmbH, Graz, Austria
| | - Josef Pichler
- Department of Internal Medicine and Neurooncology, Kepler University Hospital Linz, Neuromed Campus, Linz, Austria
| | | | - Sabine Spiegl-Kreinecker
- Clinical Research Institute for Neurosciences, Johannes Kepler University and Kepler University Hospital Linz, Linz, Austria
- Department of Neurosurgery, Johannes Kepler University, Linz, Kepler University Hospital Linz, Linz, Austria
| | - Tadeja Urbanic Purkart
- Department of Neurology, Division of General Neurology, Medical University of Graz, Graz, Austria
- Precision Medicine Technologies, CBmed GmbH, Graz, Austria
| | - Adelheid Wöhrer
- Department of Pathology, Neuropathology and Molecular Pathology, Medical University of Innsbruck, Innsbruck, Austria
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Georg Widhalm
- Department of Neurosurgery, Medical University of Vienna, Vienna, Austria
| | - Matthias Preusser
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
8
|
Yuan Z, Jing H, Deng Y, Liu M, Jiang T, Jin X, Lin W, Liu Y, Yin J. P4HB maintains Wnt-dependent stemness in glioblastoma stem cells as a precision therapeutic target and serum marker. Oncogenesis 2024; 13:42. [PMID: 39580454 PMCID: PMC11585657 DOI: 10.1038/s41389-024-00541-2] [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: 08/13/2024] [Revised: 10/27/2024] [Accepted: 11/07/2024] [Indexed: 11/25/2024] Open
Abstract
Glioblastoma stem cells (GSCs) are pivotal in the recurrence and drug resistance of glioblastoma multiforme (GBM). However, precision therapeutic and diagnostic markers for GSCs have not been fully established. Here, using bioinformatics and experimental analysis, we identified P4HB, a protein disulfide isomerase, as a serum marker that maintains stemness in GSCs through the Wnt/β-catenin signaling pathway. Transcriptional silencing of P4HB induces apoptosis and diminishes stem cell-like characteristics in GSCs. Treatments with the chemical CCF624 or the China National Medical Products Administration (NMPA)-approved securinine significantly prolonged survival in patient-derived xenograft mouse models, underscoring P4HB's potential as a therapeutic target and presenting an expedited path to clinical application through drug repurposing. Additionally, elevated P4HB levels in patient serum were found to correlate with disease progression, underscoring its utility as a biomarker and its promise for precision medicine.
Collapse
Affiliation(s)
- Zheng Yuan
- Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Hongbo Jing
- Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Yilin Deng
- Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, 10408, Republic of Korea
| | - Meichen Liu
- Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, 10408, Republic of Korea
| | - Tao Jiang
- Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, 100853, China
| | - Xiong Jin
- Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Weiwei Lin
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou, 450052, China.
| | - Yang Liu
- Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China.
- Huaihe Hospital of Henan University, Kaifeng, 475004, China.
| | - Jinlong Yin
- Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China.
- Huaihe Hospital of Henan University, Kaifeng, 475004, China.
| |
Collapse
|
9
|
Kim KH, Lee SJ, Kim J, Moon Y. Leveraging Xenobiotic-Responsive Cancer Stemness in Cell Line-Based Tumoroids for Evaluating Chemoresistance: A Proof-of-Concept Study on Environmental Susceptibility. Int J Mol Sci 2024; 25:11383. [PMID: 39518936 PMCID: PMC11545740 DOI: 10.3390/ijms252111383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 10/16/2024] [Accepted: 10/20/2024] [Indexed: 11/16/2024] Open
Abstract
Emerging evidence suggests that cancer stemness plays a crucial role in tumor progression, metastasis, and chemoresistance. Upon exposure to internal or external stress, ribosomes stand sentinel and facilitate diverse biological processes, including oncological responses. In the present study, ribosome-inactivating stress (RIS) was evaluated for its modulation of cancer cell stemness as a pivotal factor of tumor cell reprogramming. Based on the concept of stress-responsive cancer cell stemness, we addressed human intestinal cancer cell line-based off-the-shelf spheroid cultures. Intestinal cancer cell line-based spheroids exhibited heightened levels of CD44+CD133+ cancer stemness, which was improved by chemical-induced RIS. Further evaluations revealed the potential of these stress-imprinted spheroids as a platform for chemoresistance screening. Compared to adherent cells, stemness-improved spheroid cultures displayed reduced apoptosis in response to 5-fluorouracil (5-FU), a frontline chemotherapeutic agent against colorectal cancer. Moreover, serial subcultures with repeated RIS exposure maintained and even enhanced cancer stemness and chemoresistance patterns. In particular, isolated CD44+CD133+ cancer stem cells exhibited higher chemoresistance compared to unsorted cells. To elucidate the mechanisms underlying RIS-induced stemness, RNA-seq analysis identified Wnt signaling pathways and stemness-associated signals as notable features in spheroids exposed to RIS. Loss-of-function studies targeting connective tissue growth factor (CTGF), a negative regulator of Wnt signaling, revealed that CTGF-deficient spheroids exhibited improved cancer stemness and resistance to 5-FU, with RIS further enhancing these effects. In conclusion, this proof-of-concept study demonstrates the feasibility of leveraging stress-responsive cancer stemness for the development of spheroid-based platforms for chemoresistance evaluation and elucidation of pathophysiological processes of colorectal tumorigenesis under environmental stress.
Collapse
Affiliation(s)
- Ki-Hyung Kim
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Pusan National University, Yangsan 50612, Republic of Korea; (K.-H.K.); (S.J.L.); (J.K.)
- Department of Obstetrics and Gynecology, College of Medicine, Pusan National University, Busan 49241, Republic of Korea
- Biomedical Research Institute, Pusan National University, Busan 49241, Republic of Korea
| | - Seung Joon Lee
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Pusan National University, Yangsan 50612, Republic of Korea; (K.-H.K.); (S.J.L.); (J.K.)
| | - Juil Kim
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Pusan National University, Yangsan 50612, Republic of Korea; (K.-H.K.); (S.J.L.); (J.K.)
| | - Yuseok Moon
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Pusan National University, Yangsan 50612, Republic of Korea; (K.-H.K.); (S.J.L.); (J.K.)
- Biomedical Research Institute, Pusan National University, Busan 49241, Republic of Korea
- Graduate Program of Genomic Data Sciences, Pusan National University, Yangsan 50612, Republic of Korea
| |
Collapse
|
10
|
Dakal TC, Bhushan R, Xu C, Gadi BR, Cameotra SS, Yadav V, Maciaczyk J, Schmidt‐Wolf IGH, Kumar A, Sharma A. Intricate relationship between cancer stemness, metastasis, and drug resistance. MedComm (Beijing) 2024; 5:e710. [PMID: 39309691 PMCID: PMC11416093 DOI: 10.1002/mco2.710] [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: 03/13/2024] [Revised: 08/02/2024] [Accepted: 08/05/2024] [Indexed: 09/25/2024] Open
Abstract
Cancer stem cells (CSCs) are widely acknowledged as the drivers of tumor initiation, epithelial-mesenchymal transition (EMT) progression, and metastasis. Originating from both hematologic and solid malignancies, CSCs exhibit quiescence, pluripotency, and self-renewal akin to normal stem cells, thus orchestrating tumor heterogeneity and growth. Through a dynamic interplay with the tumor microenvironment (TME) and intricate signaling cascades, CSCs undergo transitions from differentiated cancer cells, culminating in therapy resistance and disease recurrence. This review undertakes an in-depth analysis of the multifaceted mechanisms underlying cancer stemness and CSC-mediated resistance to therapy. Intrinsic factors encompassing the TME, hypoxic conditions, and oxidative stress, alongside extrinsic processes such as drug efflux mechanisms, collectively contribute to therapeutic resistance. An exploration into key signaling pathways, including JAK/STAT, WNT, NOTCH, and HEDGEHOG, sheds light on their pivotal roles in sustaining CSCs phenotypes. Insights gleaned from preclinical and clinical studies hold promise in refining drug discovery efforts and optimizing therapeutic interventions, especially chimeric antigen receptor (CAR)-T cell therapy, cytokine-induced killer (CIK) cell therapy, natural killer (NK) cell-mediated CSC-targeting and others. Ultimately use of cell sorting and single cell sequencing approaches for elucidating the fundamental characteristics and resistance mechanisms inherent in CSCs will enhance our comprehension of CSC and intratumor heterogeneity, which ultimately would inform about tailored and personalized interventions.
Collapse
Affiliation(s)
- Tikam Chand Dakal
- Genome and Computational Biology LabDepartment of BiotechnologyMohanlal Sukhadia UniversityUdaipurRajasthanIndia
| | - Ravi Bhushan
- Department of ZoologyM.S. CollegeMotihariBiharIndia
| | - Caiming Xu
- Department of General SurgeryThe First Affiliated Hospital of Dalian Medical UniversityDalianChina
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research InstituteCity of HopeMonroviaCaliforniaUSA
| | - Bhana Ram Gadi
- Stress Physiology and Molecular Biology LaboratoryDepartment of BotanyJai Narain Vyas UniversityJodhpurRajasthanIndia
| | | | - Vikas Yadav
- School of Life SciencesJawaharlal Nehru UniversityNew DelhiIndia
| | - Jarek Maciaczyk
- Department of Stereotactic and Functional NeurosurgeryUniversity Hospital of BonnBonnGermany
| | - Ingo G. H. Schmidt‐Wolf
- Center for Integrated Oncology (CIO)Department of Integrated OncologyUniversity Hospital BonnBonnGermany
| | - Abhishek Kumar
- Manipal Academy of Higher EducationManipalKarnatakaIndia
- Institute of BioinformaticsInternational Technology ParkBangaloreIndia
| | - Amit Sharma
- Department of Stereotactic and Functional NeurosurgeryUniversity Hospital of BonnBonnGermany
- Center for Integrated Oncology (CIO)Department of Integrated OncologyUniversity Hospital BonnBonnGermany
| |
Collapse
|
11
|
Rodriguez A, Ahluwalia MS, Bettegowda C, Brem H, Carter BS, Chang S, Das S, Eberhart C, Garzon-Muvdi T, Hadjipanayis CG, Hawkins C, Jacques TS, Khalessi AA, McDermott MW, Mikkelsen T, Orr BA, Phillips JJ, Rosenblum M, Shelton WJ, Solomon DA, von Deimling A, Woodworth GF, Rutka JT. Toward standardized brain tumor tissue processing protocols in neuro-oncology: a perspective for gliomas and beyond. Front Oncol 2024; 14:1471257. [PMID: 39376983 PMCID: PMC11456923 DOI: 10.3389/fonc.2024.1471257] [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: 07/26/2024] [Accepted: 09/04/2024] [Indexed: 10/09/2024] Open
Abstract
Implementation of standardized protocols in neurooncology during the surgical resection of brain tumors is needed to advance the clinical treatment paradigms that use tissue for diagnosis, prognosis, bio-banking, and treatment. Currently recommendations on intraoperative tissue procurement only exist for diffuse gliomas but management of other brain tumor subtypes can also benefit from these protocols. Fresh tissue from surgical resection can now be used for intraoperative diagnostics and functional precision medicine assays. A multidisciplinary neuro-oncology perspective is critical to develop the best avenues for practical standardization. This perspective from the multidisciplinary Oncology Tissue Advisory Board (OTAB) discusses current advances, future directions, and the imperative of adopting standardized protocols for diverse brain tumor entities. There is a growing need for consistent operating room practices to enhance patient care, streamline research efforts, and optimize outcomes.
Collapse
Affiliation(s)
- Analiz Rodriguez
- Department of Neurosurgery, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Manmeet S. Ahluwalia
- Department of Medical Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL, United States
| | - Chetan Bettegowda
- Department of Neurosurgery, Johns Hopkins Hospital, Baltimore, MD, United States
| | - Henry Brem
- Department of Neurosurgery, Johns Hopkins Hospital, Baltimore, MD, United States
| | - Bob S. Carter
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Susan Chang
- Division of Neuro-Oncology, Department of Neurosurgery, University of California San Francisco, San Francisco, CA, United States
| | - Sunit Das
- Division of Neurosurgery, St. Michael’s Hospital, University of Toronto, Toronto, ON, Canada
| | - Charles Eberhart
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Tomas Garzon-Muvdi
- Department of Neurosurgery, Emory University, Atlanta, GA, United States
| | - Costas G. Hadjipanayis
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Cynthia Hawkins
- Division of Pathology, Hospital for Sick Children, Toronto, ON, Canada
| | - Thomas S. Jacques
- Developmental Biology and Cancer Programme, UCL GOS Institute of Child Health and Department of Histopathology, Great Ormond Street Hospital NHS Foundation Trust, London, United Kingdom
| | - Alexander A. Khalessi
- Department of Radiology and Neurosciences, Don and Karen Cohn Chancellor’s Endowed Chair of Neurological Surgery, University of California, San Diego, San Diego, CA, United States
| | - Michael W. McDermott
- Division of Neurosurgery, Miami Neuroscience Institute, Miami, FL, United States
| | - Tom Mikkelsen
- Department of Neurosurgery, Hermelin Brain Tumor Center, Henry Ford Health System, Detroit, MI, United States
| | - Brent A. Orr
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Joanna J. Phillips
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, United States
- Neuropathology Division, Department of Pathology, University of California, San Francisco, San Francisco, CA, United States
| | - Mark Rosenblum
- Department of Neurosurgery, Omics Laboratory, Hermelin Brain Tumor Center, Henry Ford Health System, Detroit, MI, United States
| | - William J. Shelton
- Department of Neurosurgery, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - David A. Solomon
- Division of Neuropathology, Department of Pathology and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, United States
| | - Andreas von Deimling
- Department of Neuropathology, Institute of Pathology, Ruprecht-Karls-University of Heidelberg, Heidelberg, Germany
| | - Graeme F. Woodworth
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, United States
| | - James T. Rutka
- Division of Neurosurgery, Chair Emeritus, Hospital for Sick Children, Toronto, ON, Canada
| |
Collapse
|
12
|
Ledford A, Rodriguez A, Lipinski L, Abad A, Fenstermaker R, Edenfield J, Kanos C, Redjal N, Mansouri A, Zacharia B, Butowski N, Liu J, Han SJ, Ziu M, Cohen AL, Fabiano AJ, Miles K, Rayner M, Thompson J, Tollison K, Azimzadeh P, Holmes L, Gevaert M, DesRochers TM. Functional prediction of response to therapy prior to therapeutic intervention is associated with improved survival in patients with high-grade glioma. Sci Rep 2024; 14:19474. [PMID: 39198514 PMCID: PMC11358395 DOI: 10.1038/s41598-024-68801-0] [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: 11/09/2023] [Accepted: 07/29/2024] [Indexed: 09/01/2024] Open
Abstract
Patients with high-grade glioma (HGG) have an extremely poor prognosis compounded by a lack of advancement in clinical care over the past few decades. Regardless of classification, most newly diagnosed patients receive the same treatment, radiation and temozolomide (RT/TMZ). We developed a functional precision oncology test that prospectively identifies individual patient's response to this treatment regimen. Tumor tissues isolated from patients with newly diagnosed HGG enrolled in 3D PREDICT REGISTRY were evaluated for response to chemotherapeutic agents using the 3D Predict™ Glioma test. Patients receiving RT/TMZ were followed for 2 years. Clinical outcomes including imaging, assessments, and biomarker measurements were compared to patient matched test-predicted therapy response. Median survival between test-predicted temozolomide responders and test-predicted temozolomide non-responders revealed a statistically significant increase in progression-free survival when using the test to predict response across multiple subgroups including HGG (5.8 months), glioblastoma (4.7 months), and MGMT unmethylated glioblastoma (4.7 months). Overall survival was also positively separated across the subgroups at 7.6, 5.1, and 6.3 months respectively. The strong correlation of 3D Predict Glioma test results with clinical outcomes demonstrates that this functional test is prognostic in patients treated with RT/TMZ and supports aligning clinical treatment to test-predicted response across varying HGG subgroups.
Collapse
Affiliation(s)
| | - Analiz Rodriguez
- Department of Neurosurgery, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Lindsay Lipinski
- Department of Neurosurgery, Department of Neuro-Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14203, USA
| | - Ajay Abad
- Department of Neurosurgery, Department of Neuro-Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14203, USA
| | - Robert Fenstermaker
- Department of Neurosurgery, Department of Neuro-Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14203, USA
| | - Jeffrey Edenfield
- Institute for Translational Oncology Research, Prisma Health Cancer Institute, Greenville, SC, 29605, USA
| | - Charles Kanos
- Department of Neurosurgery, Prisma Health Southeastern Neurosurgical and Spine Institute, Greenville, SC, 29605, USA
| | - Navid Redjal
- Department of Neurosurgical Oncology, Capital Health Institute for Neurosciences, Pennington, NJ, 08534, USA
| | - Alireza Mansouri
- Department of Neurosurgery, Penn State Hershey Medical Center, Hershey, PA, 17033, USA
| | - Brad Zacharia
- Department of Neurosurgery, Penn State Hershey Medical Center, Hershey, PA, 17033, USA
| | - Nicholas Butowski
- Department of Neuro-Oncology, University of California, San Francisco, CA, 94143, USA
| | - Jesse Liu
- Department of Neurologic Surgery, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Seunggu J Han
- Department of Neurologic Surgery, Oregon Health and Science University, Portland, OR, 97239, USA
- Department of Neurological Surgery, Stanford Medicine, Palo Alto, CA, USA
| | - Mateo Ziu
- Department of Neurosurgery, Inova Healthcare System, Falls Church, VA, 22042, USA
| | - Adam L Cohen
- Department of Medical Oncology, Inova Schar Cancer Institute, Fairfax, VA, 22031, USA
| | - Andrew J Fabiano
- Department of Neurosurgery, Department of Neuro-Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14203, USA
| | | | | | - Jayla Thompson
- Kiyatec, Inc, 2 N. Main St, Greenville, SC, 29601, USA
- IQVIA, Durham, NC, 27703, USA
| | | | | | - Lillia Holmes
- Kiyatec, Inc, 2 N. Main St, Greenville, SC, 29601, USA
| | | | | |
Collapse
|
13
|
Bizzoca ME, Caponio VCA, Lo Muzio L, Claudio PP, Cortese A. Methods for Overcoming Chemoresistance in Head and Neck Squamous Cell Carcinoma: Keeping the Focus on Cancer Stem Cells, a Systematic Review. Cancers (Basel) 2024; 16:3004. [PMID: 39272862 PMCID: PMC11394389 DOI: 10.3390/cancers16173004] [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: 07/29/2024] [Revised: 08/15/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024] Open
Abstract
According to the "cancer stem cell" (CSCs) theory, tumors are a diverse and expanding group of malignant cells that originate from a small number of CSCs. Despite treatment, these cells can still become active and proliferate, which can result in distant metastasis and local recurrences. A new paradigm in cancer treatment involves targeting both CSCs and the cancer cells in a tumor. This review aims to examine the literature on methods published to overcome chemoresistance due to the presence of CSCs in head and neck cancers. The review was registered with PROSPERO (ID# CRD42024512809). After Pub Med, Scopus, and WoS database searches, 31 relevant articles on oral squamous cell carcinoma (OSCC) were selected. Compounds that increased chemosensitivity by targeting CSCs in head and neck squamous cell carcinoma (HNSCC) were divided into (1) natural products, (2) adjuvant molecules to traditional chemotherapy, and (3) CSCs targeting patient-specific fresh biopsies for functional precision medicine.
Collapse
Affiliation(s)
- Maria Eleonora Bizzoca
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | | | - Lorenzo Lo Muzio
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Pier Paolo Claudio
- Department of Pharmacology and Toxicology, Cancer Center & Research Institute, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Antonio Cortese
- Unit of Maxillofacial Surgery, Department of Medicine, Surgery, and Dentistry, University of Salerno, 84084 Salerno, Italy
| |
Collapse
|
14
|
Shelton WJ, Santos Horta E, Stephen Nix J, Gokden M, Rodriguez A. Functional precision medicine assay for recurrent meningioma: a proof of principle. Illustrative case. JOURNAL OF NEUROSURGERY. CASE LESSONS 2024; 8:CASE24242. [PMID: 39074389 DOI: 10.3171/case24242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 05/20/2024] [Indexed: 07/31/2024]
Abstract
BACKGROUND Meningiomas are the most prevalent primary central nervous system tumors. Although low-grade meningiomas are considered benign tumors, a subset of these can behave aggressively, showing progression and recurrence. In such cases, functional assays could influence treatment decisions and improve patient outcomes. OBSERVATIONS A 78-year-old female presented with a long-standing history of a supratentorial meningioma that was initially resected and treated with Gamma Knife radiosurgery. Surveillance revealed progression. She began systemic therapy with everolimus and octreotide but was lost to follow-up and did not continue the treatment. She returned because of a rapid decline in her neurological status. Biopsy with advanced molecular characterization by next-generation sequencing revealed NF2 and CREBBP mutations, and a commercial functional assay was done. This assay successfully isolated cancer stem cells (CSCs) from biopsy cores and identified potential drugs based on cellular sensitivity profiles. This is the first reported case in which a commercial functional drug screen was used for a meningioma. LESSONS In cases in which meningiomas exhibit specific genetic alterations and characteristics of aggressiveness, functional assays can be a useful tool for isolating CSCs. The authors report success in obtaining drug-screen profiling for a World Health Organization grade 1 meningioma. Multimodal approaches utilizing multi-omics analyses with functional assays can improve patient outcomes. https://thejns.org/doi/10.3171/CASE24242.
Collapse
Affiliation(s)
- William J Shelton
- Departments of Neurosurgery, University of Arkansas for Medical Sciences, College of Medicine, Little Rock, Arkansas
| | - Erika Santos Horta
- Departments of Medical Oncology, University of Arkansas for Medical Sciences, College of Medicine, Little Rock, Arkansas
| | - James Stephen Nix
- Departments of Pathology, University of Arkansas for Medical Sciences, College of Medicine, Little Rock, Arkansas
| | - Murat Gokden
- Departments of Pathology, University of Arkansas for Medical Sciences, College of Medicine, Little Rock, Arkansas
| | - Analiz Rodriguez
- Departments of Neurosurgery, University of Arkansas for Medical Sciences, College of Medicine, Little Rock, Arkansas
| |
Collapse
|
15
|
Giambra M, Di Cristofori A, Raimondo F, Rigolio R, Conconi D, Chiarello G, Tabano SM, Antolini L, Nicolini G, Bua M, Ferlito D, Carrabba G, Giussani CG, Lavitrano M, Bentivegna A. Vacuolar Proton-Translocating ATPase May Take Part in the Drug Resistance Phenotype of Glioma Stem Cells. Int J Mol Sci 2024; 25:2743. [PMID: 38473989 DOI: 10.3390/ijms25052743] [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: 01/31/2024] [Revised: 02/15/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
The vacuolar proton-translocating ATPase (V-ATPase) is a transmembrane multi-protein complex fundamental in maintaining a normal intracellular pH. In the tumoral contest, its role is crucial since the metabolism underlying carcinogenesis is mainly based on anaerobic glycolytic reactions. Moreover, neoplastic cells use the V-ATPase to extrude chemotherapy drugs into the extra-cellular compartment as a drug resistance mechanism. In glioblastoma (GBM), the most malignant and incurable primary brain tumor, the expression of this pump is upregulated, making it a new possible therapeutic target. In this work, the bafilomycin A1-induced inhibition of V-ATPase in patient-derived glioma stem cell (GSC) lines was evaluated together with temozolomide, the first-line therapy against GBM. In contrast with previous published data, the proposed treatment did not overcome resistance to the standard therapy. In addition, our data showed that nanomolar dosages of bafilomycin A1 led to the blockage of the autophagy process and cellular necrosis, making the drug unusable in models which are more complex. Nevertheless, the increased expression of V-ATPase following bafilomycin A1 suggests a critical role of the proton pump in GBM stem components, encouraging the search for novel strategies to limit its activity in order to circumvent resistance to conventional therapy.
Collapse
Affiliation(s)
- Martina Giambra
- PhD Program in Neuroscience, University of Milano-Bicocca, 20900 Monza, Italy
- GBM-BI-TRACE (GlioBlastoMa-BIcocca-TRAnslational-CEnter), University of Milano-Bicocca, 20900 Monza, Italy
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Andrea Di Cristofori
- PhD Program in Neuroscience, University of Milano-Bicocca, 20900 Monza, Italy
- GBM-BI-TRACE (GlioBlastoMa-BIcocca-TRAnslational-CEnter), University of Milano-Bicocca, 20900 Monza, Italy
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
- Neurosurgery, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Francesca Raimondo
- GBM-BI-TRACE (GlioBlastoMa-BIcocca-TRAnslational-CEnter), University of Milano-Bicocca, 20900 Monza, Italy
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Roberta Rigolio
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Donatella Conconi
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Gaia Chiarello
- GBM-BI-TRACE (GlioBlastoMa-BIcocca-TRAnslational-CEnter), University of Milano-Bicocca, 20900 Monza, Italy
- Pathology, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Silvia Maria Tabano
- Laboratory of Medical Genetics, Ospedale Maggiore Policlinico, IRCCS Ca' Granda, 20122 Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy
| | - Laura Antolini
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Gabriella Nicolini
- GBM-BI-TRACE (GlioBlastoMa-BIcocca-TRAnslational-CEnter), University of Milano-Bicocca, 20900 Monza, Italy
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Miriam Bua
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
- Neurosurgery, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Davide Ferlito
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
- Neurosurgery, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Giorgio Carrabba
- GBM-BI-TRACE (GlioBlastoMa-BIcocca-TRAnslational-CEnter), University of Milano-Bicocca, 20900 Monza, Italy
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
- Neurosurgery, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Carlo Giorgio Giussani
- GBM-BI-TRACE (GlioBlastoMa-BIcocca-TRAnslational-CEnter), University of Milano-Bicocca, 20900 Monza, Italy
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
- Neurosurgery, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Marialuisa Lavitrano
- GBM-BI-TRACE (GlioBlastoMa-BIcocca-TRAnslational-CEnter), University of Milano-Bicocca, 20900 Monza, Italy
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Angela Bentivegna
- GBM-BI-TRACE (GlioBlastoMa-BIcocca-TRAnslational-CEnter), University of Milano-Bicocca, 20900 Monza, Italy
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| |
Collapse
|
16
|
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
|
17
|
Habeeb M, Vengateswaran HT, You HW, Saddhono K, Aher KB, Bhavar GB. Nanomedicine facilitated cell signaling blockade: difficulties and strategies to overcome glioblastoma. J Mater Chem B 2024; 12:1677-1705. [PMID: 38288615 DOI: 10.1039/d3tb02485g] [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: 02/15/2024]
Abstract
Glioblastoma (GBM) is a highly aggressive and lethal type of brain tumor with complex and diverse molecular signaling pathways involved that are in its development and progression. Despite numerous attempts to develop effective treatments, the survival rate remains low. Therefore, understanding the molecular mechanisms of these pathways can aid in the development of targeted therapies for the treatment of glioblastoma. Nanomedicines have shown potential in targeting and blocking signaling pathways involved in glioblastoma. Nanomedicines can be engineered to specifically target tumor sites, bypass the blood-brain barrier (BBB), and release drugs over an extended period. However, current nanomedicine strategies also face limitations, including poor stability, toxicity, and low therapeutic efficacy. Therefore, novel and advanced nanomedicine-based strategies must be developed for enhanced drug delivery. In this review, we highlight risk factors and chemotherapeutics for the treatment of glioblastoma. Further, we discuss different nanoformulations fabricated using synthetic and natural materials for treatment and diagnosis to selectively target signaling pathways involved in GBM. Furthermore, we discuss current clinical strategies and the role of artificial intelligence in the field of nanomedicine for targeting GBM.
Collapse
Affiliation(s)
- Mohammad Habeeb
- Department of Pharmaceutics, Crescent School of Pharmacy, B.S. Abdur Rahman Crescent Institute of Science and Technology, Chennai-600048, India.
| | - Hariharan Thirumalai Vengateswaran
- Department of Pharmaceutics, Crescent School of Pharmacy, B.S. Abdur Rahman Crescent Institute of Science and Technology, Chennai-600048, India.
| | - Huay Woon You
- Pusat PERMATA@Pintar Negara, Universiti Kebangsaan 43600, Bangi, Selangor, Malaysia
| | - Kundharu Saddhono
- Faculty of Teacher Training and Education, Universitas Sebelas Maret, 57126, Indonesia
| | - Kiran Balasaheb Aher
- Department of Pharmaceutical Quality Assurance, Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, Maharashtra, 424001, India
| | - Girija Balasaheb Bhavar
- Department of Pharmaceutical Chemistry, Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, Maharashtra, 424001, India
| |
Collapse
|
18
|
Mathews AP, Shelton WJ, Horta ES, Reddy Damalcheruvu P, Nix JS, Gokden M, Rodriguez A. Operative planning for a functional precision medicine assay of recurrent high-grade glioma: illustrative case. JOURNAL OF NEUROSURGERY. CASE LESSONS 2024; 7:CASE23679. [PMID: 38346299 PMCID: PMC10865468 DOI: 10.3171/case23679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 12/19/2023] [Indexed: 02/16/2024]
Abstract
BACKGROUND Functional precision medicine (FPM) represents a personalized and efficacious modality for treating malignant neoplasms. However, acquiring sufficient live tissue to perform FPM analyses is complicated by both difficult identification on imaging and radiation necrosis, particularly in cases of recurrence. The authors describe a case of planning biopsy trajectories for an FPM assay in a patient with recurrent high-grade glioma. OBSERVATIONS A 25-year-old male with a history of recurrent high-grade glioma was scheduled for laser ablation and biopsy with ChemoID assaying after regions of potential recurrence were identified on follow-up imaging. Preoperative magnetic resonance (MR) spectroscopy of the regions showed areas of high choline/creatine ratios within lesions of radiation necrosis, which helped in planning the biopsy trajectories to selectively target malignancies for FPM analysis. ChemoID results showed high tumor susceptibility to lomustine, which was implemented as adjuvant therapy. LESSONS FPM therapy in the setting of recurrence is complicated by radiation necrosis, which can present as malignancy on imaging and interfere with tissue acquisition during biopsy or resection. Thus, operative approaches should be carefully planned with the assistance of imaging modalities such as MR spectroscopy to better ensure effective tissue acquisition for accurate FPM analysis and to promote more definitive treatment of recurrence.
Collapse
Affiliation(s)
| | | | | | | | - J. Stephen Nix
- Pathology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Murat Gokden
- Pathology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | | |
Collapse
|
19
|
|
20
|
Marcu LG, Dell’Oro M, Bezak E. Opportunities in Cancer Therapies: Deciphering the Role of Cancer Stem Cells in Tumour Repopulation. Int J Mol Sci 2023; 24:17258. [PMID: 38139085 PMCID: PMC10744048 DOI: 10.3390/ijms242417258] [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: 11/18/2023] [Revised: 12/06/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Tumour repopulation during treatment is a well acknowledged yet still challenging aspect of cancer management. The latest research results show clear evidence towards the existence of cancer stem cells (CSCs) that are responsible for tumour repopulation, dissemination, and distant metastases in most solid cancers. Cancer stem cell quiescence and the loss of asymmetrical division are two powerful mechanisms behind repopulation. Another important aspect in the context of cancer stem cells is cell plasticity, which was shown to be triggered during fractionated radiotherapy, leading to cell dedifferentiation and thus reactivation of stem-like properties. Repopulation during treatment is not limited to radiotherapy, as there is clinical proof for repopulation mechanisms to be activated through other conventional treatment techniques, such as chemotherapy. The dynamic nature of stem-like cancer cells often elicits resistance to treatment by escaping drug-induced cell death. The aims of this scoping review are (1) to describe the main mechanisms used by cancer stem cells to initiate tumour repopulation during therapy; (2) to present clinical evidence for tumour repopulation during radio- and chemotherapy; (3) to illustrate current trends in the identification of CSCs using specific imaging techniques; and (4) to highlight novel technologies that show potential in the eradication of CSCs.
Collapse
Affiliation(s)
- Loredana G. Marcu
- UniSA Allied Health & Human Performance, University of South Australia, Adelaide, SA 5001, Australia;
- Faculty of Informatics and Science, University of Oradea, 410087 Oradea, Romania
| | - Mikaela Dell’Oro
- Australian Centre for Quantitative Imaging, School of Medicine, The University of Western Australia, Perth, WA 6009, Australia;
| | - Eva Bezak
- UniSA Allied Health & Human Performance, University of South Australia, Adelaide, SA 5001, Australia;
- Faculty of Chemistry & Physics, University of Adelaide, Adelaide, SA 5000, Australia
| |
Collapse
|
21
|
Zhou W, Yan K, Xi Q. BMP signaling in cancer stemness and differentiation. CELL REGENERATION (LONDON, ENGLAND) 2023; 12:37. [PMID: 38049682 PMCID: PMC10695912 DOI: 10.1186/s13619-023-00181-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 11/06/2023] [Indexed: 12/06/2023]
Abstract
The BMP (Bone morphogenetic protein) signaling pathway plays a central role in metazoan biology, intricately shaping embryonic development, maintaining tissue homeostasis, and influencing disease progression. In the context of cancer, BMP signaling exhibits context-dependent dynamics, spanning from tumor suppression to promotion. Cancer stem cells (CSCs), a modest subset of neoplastic cells with stem-like attributes, exert substantial influence by steering tumor growth, orchestrating therapy resistance, and contributing to relapse. A comprehensive grasp of the intricate interplay between CSCs and their microenvironment is pivotal for effective therapeutic strategies. Among the web of signaling pathways orchestrating cellular dynamics within CSCs, BMP signaling emerges as a vital conductor, overseeing CSC self-renewal, differentiation dynamics, and the intricate symphony within the tumor microenvironment. Moreover, BMP signaling's influence in cancer extends beyond CSCs, intricately regulating cellular migration, invasion, and metastasis. This multifaceted role underscores the imperative of comprehending BMP signaling's contributions to cancer, serving as the foundation for crafting precise therapies to navigate multifaceted challenges posed not only by CSCs but also by various dimensions of cancer progression. This article succinctly encapsulates the diverse roles of the BMP signaling pathway across different cancers, spanning glioblastoma multiforme (GBM), diffuse intrinsic pontine glioma (DIPG), colorectal cancer, acute myeloid leukemia (AML), lung cancer, prostate cancer, and osteosarcoma. It underscores the necessity of unraveling underlying mechanisms and molecular interactions. By delving into the intricate tapestry of BMP signaling's engagement in cancers, researchers pave the way for meticulously tailored therapies, adroitly leveraging its dualistic aspects-whether as a suppressor or promoter-to effectively counter the relentless march of tumor progression.
Collapse
Affiliation(s)
- Wei Zhou
- State Key Laboratory of Molecular Oncology, MOE Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Kun Yan
- Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Qiaoran Xi
- State Key Laboratory of Molecular Oncology, MOE Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China.
- Joint Graduate Program of Peking-Tsinghua-NIBS, Tsinghua University, Beijing, China.
| |
Collapse
|
22
|
Guo L, Li F, Liu H, Kong D, Chen C, Sun S. SIX1 amplification modulates stemness and tumorigenesis in breast cancer. J Transl Med 2023; 21:866. [PMID: 38031089 PMCID: PMC10685563 DOI: 10.1186/s12967-023-04679-2] [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: 06/23/2023] [Accepted: 10/29/2023] [Indexed: 12/01/2023] Open
Abstract
BACKGROUND Sine oculis homeobox homolog 1 (SIX1) is a transcription factor that has recently been identified as a crucial regulator of embryonic development and tumorigenesis. SIX1 is upregulated in different types of tumors, including breast cancer. However, the role and mechanism of SIX1 upregulation in breast cancer carcinogenesis remains uncertain. METHODS In this study, we utilized various databases such as UALCAN, TCGA, STRING, and Kaplan-Meier Plotter to investigate the mRNA expression, prognosis, transcriptional profile changes, signal pathway rewiring, and interaction with cancer stem cells of SIX1 in breast cancer. We also conducted both in vitro and in vivo experiments to validate its positive regulation effect on breast cancer stem cells. RESULTS Our findings demonstrated that the expression of SIX1 varies among different subtypes of breast cancer and that it upregulates breast cancer grading and lymph node metastasis. Besides, SIX1 participates in the rewiring of several cancer signaling pathways, including estrogen, WNT, MAPK, and other pathways, and interacts with cancer stem cells. SIX1 showed a significant positive correlation with breast cancer stem cell markers such as ALDH1A1, EPCAM, ITGB1, and SOX2. Moreover, our in vitro and in vivo experiments confirmed that SIX1 can promote the increase in the proportion of stem cells and tumor progression. CONCLUSIONS Altogether, our results suggest that SIX1 plays an essential regulatory role in breast cancer's occurrence, and its amplification can be utilized as a diagnostic and prognostic predictor. The interaction between SIX1 and cancer stem cells may play a critical role in regulating breast cancer's initiation and metastasis.
Collapse
Affiliation(s)
- Liantao Guo
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan, 430060, Hubei, China
| | - Faminzi Li
- Reproductive Medical Center, Renmin Hospital of Wuhan University and Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, No. 238 Jiefang Road, Wuchang District, Wuhan, 430060, Hubei, China
| | - Hanqing Liu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan, 430060, Hubei, China
| | - Deguang Kong
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan, 430060, Hubei, China.
| | - Chuang Chen
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan, 430060, Hubei, China.
| | - Shengrong Sun
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan, 430060, Hubei, China.
| |
Collapse
|
23
|
Merati A, Kotian S, Acton A, Placzek W, Smithberger E, Shelton AK, Miller CR, Stern JL. Glioma Stem Cells Are Sensitized to BCL-2 Family Inhibition by Compromising Histone Deacetylases. Int J Mol Sci 2023; 24:13688. [PMID: 37761989 PMCID: PMC10530722 DOI: 10.3390/ijms241813688] [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: 07/29/2023] [Revised: 08/14/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
Glioblastoma (GBM) remains an incurable disease with an extremely high five-year recurrence rate. We studied apoptosis in glioma stem cells (GSCs) in response to HDAC inhibition (HDACi) combined with MEK1/2 inhibition (MEKi) or BCL-2 family inhibitors. MEKi effectively combined with HDACi to suppress growth, induce cell cycle defects, and apoptosis, as well as to rescue the expression of the pro-apoptotic BH3-only proteins BIM and BMF. A RNAseq analysis of GSCs revealed that HDACi repressed the pro-survival BCL-2 family genes MCL1 and BCL-XL. We therefore replaced MEKi with BCL-2 family inhibitors and observed enhanced apoptosis. Conversely, a ligand for the cancer stem cell receptor CD44 led to reductions in BMF, BIM, and apoptosis. Our data strongly support further testing of HDACi in combination with MEKi or BCL-2 family inhibitors in glioma.
Collapse
Affiliation(s)
- Aran Merati
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Spandana Kotian
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Alexus Acton
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - William Placzek
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Erin Smithberger
- O’Neal Comprehensive Cancer Center, Birmingham, AL 35294, USA
- Department of Pathology, Division of Neuropathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Abigail K. Shelton
- O’Neal Comprehensive Cancer Center, Birmingham, AL 35294, USA
- Department of Pathology, Division of Neuropathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - C. Ryan Miller
- O’Neal Comprehensive Cancer Center, Birmingham, AL 35294, USA
- Department of Pathology, Division of Neuropathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Josh L. Stern
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- O’Neal Comprehensive Cancer Center, Birmingham, AL 35294, USA
| |
Collapse
|