1
|
Choi SH, Jang J, Kim Y, Park CG, Lee SY, Kim H, Kim H. ID1 high/activin A high glioblastoma cells contribute to resistance to anti-angiogenesis therapy through malformed vasculature. Cell Death Dis 2024; 15:292. [PMID: 38658527 PMCID: PMC11043395 DOI: 10.1038/s41419-024-06678-7] [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: 06/26/2023] [Revised: 04/09/2024] [Accepted: 04/12/2024] [Indexed: 04/26/2024]
Abstract
Although bevacizumab (BVZ), a representative drug for anti-angiogenesis therapy (AAT), is used as a first-line treatment for patients with glioblastoma (GBM), its efficacy is notably limited. Whereas several mechanisms have been proposed to explain the acquisition of AAT resistance, the specific underlying mechanisms have yet to be sufficiently ascertained. Here, we established that inhibitor of differentiation 1 (ID1)high/activin Ahigh glioblastoma cell confers resistance to BVZ. The bipotent effect of activin A during its active phase was demonstrated to reduce vasculature dependence in tumorigenesis. In response to a temporary exposure to activin A, this cytokine was found to induce endothelial-to-mesenchymal transition via the Smad3/Slug axis, whereas prolonged exposure led to endothelial apoptosis. ID1 tumors showing resistance to BVZ were established to be characterized by a hypovascular structure, hyperpermeability, and scattered hypoxic regions. Using a GBM mouse model, we demonstrated that AAT resistance can be overcome by administering therapy based on a combination of BVZ and SB431542, a Smad2/3 inhibitor, which contributed to enhancing survival. These findings offer valuable insights that could contribute to the development of new strategies for treating AAT-resistant GBM.
Collapse
Affiliation(s)
- Sang-Hun Choi
- Department of Biotechnology, Korea University, Seoul, 02841, Republic of Korea
- Institute of Animal Molecular Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Junseok Jang
- Department of Biotechnology, Korea University, Seoul, 02841, Republic of Korea
- Institute of Animal Molecular Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Yoonji Kim
- Department of Biotechnology, Korea University, Seoul, 02841, Republic of Korea
- Institute of Animal Molecular Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Cheol Gyu Park
- MEDIFIC Inc, Hwaseong-si, Gyeonggi-do, 18469, Republic of Korea
| | - Seon Yong Lee
- Department of Biotechnology, Korea University, Seoul, 02841, Republic of Korea
- Institute of Animal Molecular Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Hyojin Kim
- Department of Biotechnology, Korea University, Seoul, 02841, Republic of Korea
- Institute of Animal Molecular Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Hyunggee Kim
- Department of Biotechnology, Korea University, Seoul, 02841, Republic of Korea.
- Institute of Animal Molecular Biotechnology, Korea University, Seoul, 02841, Republic of Korea.
| |
Collapse
|
2
|
Habashy KJ, Dmello C, Chen L, Arrieta VA, Kim KS, Gould A, Youngblood MW, Bouchoux G, Burdett KB, Zhang H, Canney M, Stupp R, Sonabend AM. Paclitaxel and Carboplatin in Combination with Low-intensity Pulsed Ultrasound for Glioblastoma. Clin Cancer Res 2024; 30:1619-1629. [PMID: 38295144 DOI: 10.1158/1078-0432.ccr-23-2367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 11/22/2023] [Accepted: 01/29/2024] [Indexed: 02/02/2024]
Abstract
PURPOSE We recently reported on clinical trials for patients with recurrent glioblastoma where low-intensity pulsed ultrasound and microbubbles (LIPU/MB) improved paclitaxel or carboplatin delivery into the brain. Here, we report variable local tumor control with paclitaxel at the maximal/target dose in our phase I trial (NCT04528680). To address this, we investigated the combination of paclitaxel with carboplatin in preclinical glioma models. EXPERIMENTAL DESIGN We performed MRI-based analysis to evaluate disease control in patients from our trial. We studied the cytotoxicity of paclitaxel and carboplatin against 11 human glioma lines as monotherapy and in combination at concentrations derived from human intraoperative studies. Synergy was assessed with the Loewe model and the survival benefit evaluated in two xenografts. We examined the effects on cell cycle progression, DNA damage, and apoptosis. RESULTS Patients treated with paclitaxel and LIPU/MB exhibited variable local tumor control, which correlated with overall survival. We observed limited cross-resistance to paclitaxel and carboplatin in glioma lines, with almost a third of them being exclusively susceptible to one drug. This combination led to susceptibility of 81% of lines and synergy in 55% of them. The combination proved more efficacious in two intracranial xenografts when administered with LIPU/MB, leading to complementary effects on cell cycle arrest. CONCLUSIONS Combining paclitaxel and carboplatin in gliomas may be more efficacious than monotherapy, as in other cancers, due to synergy and independent susceptibility to each drug. These results form the basis for an ongoing phase II trial (NCT04528680) where we investigate this combination with LIPU/MB.
Collapse
Affiliation(s)
- Karl J Habashy
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Northwestern Medicine Lou and Jean Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Crismita Dmello
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Northwestern Medicine Lou and Jean Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Li Chen
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Northwestern Medicine Lou and Jean Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Victor A Arrieta
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Northwestern Medicine Lou and Jean Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Kwang-Soo Kim
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Northwestern Medicine Lou and Jean Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Andrew Gould
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Northwestern Medicine Lou and Jean Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Mark W Youngblood
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Northwestern Medicine Lou and Jean Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | | | - Kirsten B Burdett
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Hui Zhang
- Northwestern Medicine Lou and Jean Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | | | - Roger Stupp
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Northwestern Medicine Lou and Jean Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Division of Hematology/Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Adam M Sonabend
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Northwestern Medicine Lou and Jean Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| |
Collapse
|
3
|
Shen Y, Thng DKH, Wong ALA, Toh TB. Mechanistic insights and the clinical prospects of targeted therapies for glioblastoma: a comprehensive review. Exp Hematol Oncol 2024; 13:40. [PMID: 38615034 PMCID: PMC11015656 DOI: 10.1186/s40164-024-00512-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 04/08/2024] [Indexed: 04/15/2024] Open
Abstract
Glioblastoma (GBM) is a fatal brain tumour that is traditionally diagnosed based on histological features. Recent molecular profiling studies have reshaped the World Health Organization approach in the classification of central nervous system tumours to include more pathogenetic hallmarks. These studies have revealed that multiple oncogenic pathways are dysregulated, which contributes to the aggressiveness and resistance of GBM. Such findings have shed light on the molecular vulnerability of GBM and have shifted the disease management paradigm from chemotherapy to targeted therapies. Targeted drugs have been developed to inhibit oncogenic targets in GBM, including receptors involved in the angiogenic axis, the signal transducer and activator of transcription 3 (STAT3), the PI3K/AKT/mTOR signalling pathway, the ubiquitination-proteasome pathway, as well as IDH1/2 pathway. While certain targeted drugs showed promising results in vivo, the translatability of such preclinical achievements in GBM remains a barrier. We also discuss the recent developments and clinical assessments of targeted drugs, as well as the prospects of cell-based therapies and combinatorial therapy as novel ways to target GBM. Targeted treatments have demonstrated preclinical efficacy over chemotherapy as an alternative or adjuvant to the current standard of care for GBM, but their clinical efficacy remains hindered by challenges such as blood-brain barrier penetrance of the drugs. The development of combinatorial targeted therapies is expected to improve therapeutic efficacy and overcome drug resistance.
Collapse
Affiliation(s)
- Yating Shen
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Dexter Kai Hao Thng
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Andrea Li Ann Wong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Department of Haematology-Oncology, National University Hospital, Singapore, Singapore
| | - Tan Boon Toh
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, Singapore.
- The Institute for Digital Medicine (WisDM), National University of Singapore, Singapore, Singapore.
| |
Collapse
|
4
|
Laing BR, Prah MA, Best BJ, Krucoff MO, Mueller WM, Schmainda KM. Application of Delta T1 maps for quantitative and objective assessment of extent of resection and survival prediction in glioblastoma. NEUROSURGERY PRACTICE 2024; 5:e00077. [PMID: 38919518 PMCID: PMC11198967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/27/2024]
Abstract
Background and Objectives Gross-total resection (GTR) and low residual tumor volume (RTV) have been associated with increased survival in glioblastoma. Largely due to the subjectivity involved, the determination of GTR and RTV remains difficult in the postoperative setting. In response, the objective of this study is to evaluate the clinical efficacy of an easy-to-use MRI metric, called delta T1 (dT1), to quantify extent of resection (EOR) and RTV, in comparison to radiologist impression, to predict overall survival (OS) in glioblastoma patients. Methods 59 patients who underwent resection of glioblastoma were retrospectively identified. Delta T1 (dT1) images, automatically created from the difference between calibrated post- and pre-contrast T1-weighted images, were used to quantify EOR and RTV. Kaplan-Meier survival estimates were determined for EOR categories, an RTV cutoff of 5cm3 and radiologist interpretation of EOR. Multivariate Cox proportional hazard regression analysis was used to evaluate RTV and EOR along with effects related to sex, KPS, MGMT, and age on OS. Results Kaplan-Meier analysis revealed a statistically significant difference in median OS for a dT1-determined RTV cutoff of 5 cm3 (P=.0024, HR=2.18 (1.232-3.856)), but not for radiological impression (P=0.666) or dT1-determined EOR (P=0.0803), which was limited to a comparison between partial and subtotal resections. Furthermore, when covariates were accounted for in multivariate Cox regression, significant differences in OS were retained for dT1-determined RTV. Additionally, a significantly strong yet short-term effect of MGMT methylation status on OS was revealed for each RTV and EOR model. Conclusion The utility of dT1 maps to quantify EOR and RTV in glioblastoma and predict survival, suggests an emerging role for dT1s with relevance for intraoperative MRI, neuro-navigation and postoperative disease surveillance.
Collapse
Affiliation(s)
- Brandon R. Laing
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Melissa A. Prah
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Benjamin J. Best
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Max O. Krucoff
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Wade M. Mueller
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | | |
Collapse
|
5
|
Carpentier A, Stupp R, Sonabend AM, Dufour H, Chinot O, Mathon B, Ducray F, Guyotat J, Baize N, Menei P, de Groot J, Weinberg JS, Liu BP, Guemas E, Desseaux C, Schmitt C, Bouchoux G, Canney M, Idbaih A. Repeated blood-brain barrier opening with a nine-emitter implantable ultrasound device in combination with carboplatin in recurrent glioblastoma: a phase I/II clinical trial. Nat Commun 2024; 15:1650. [PMID: 38396134 PMCID: PMC10891097 DOI: 10.1038/s41467-024-45818-7] [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: 02/23/2023] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Here, the results of a phase 1/2 single-arm trial (NCT03744026) assessing the safety and efficacy of blood-brain barrier (BBB) disruption with an implantable ultrasound system in recurrent glioblastoma patients receiving carboplatin are reported. A nine-emitter ultrasound implant was placed at the end of tumor resection replacing the bone flap. After surgery, activation to disrupt the BBB was performed every four weeks either before or after carboplatin infusion. The primary objective of the Phase 1 was to evaluate the safety of escalating numbers of ultrasound emitters using a standard 3 + 3 dose escalation. The primary objective of the Phase 2 was to evaluate the efficacy of BBB opening using magnetic resonance imaging (MRI). The secondary objectives included safety and clinical efficacy. Thirty-three patients received a total of 90 monthly sonications with carboplatin administration and up to nine emitters activated without observed DLT. Grade 3 procedure-related adverse events consisted of pre syncope (n = 3), fatigue (n = 1), wound infection (n = 2), and pain at time of device connection (n = 7). BBB opening endpoint was met with 90% of emitters showing BBB disruption on MRI after sonication. In the 12 patients who received carboplatin just prior to sonication, the progression-free survival was 3.1 months, the 1-year overall survival rate was 58% and median overall survival was 14.0 months from surgery.
Collapse
Affiliation(s)
- Alexandre Carpentier
- Sorbonne Université, AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, Service de Neurochirurgie, Paris, France.
| | - Roger Stupp
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Adam M Sonabend
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Henry Dufour
- Aix-Marseille Univ, APHM, CNRS, INP, Inst Neurophysiopathol, CHU Timone, Service de Neuro-Oncologie, Marseille, France
| | - Olivier Chinot
- Aix-Marseille Univ, APHM, CNRS, INP, Inst Neurophysiopathol, CHU Timone, Service de Neuro-Oncologie, Marseille, France
| | - Bertrand Mathon
- Sorbonne Université, AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, Service de Neurochirurgie, Paris, France
| | - François Ducray
- Hospices Civils de Lyon, Université Claude Bernard Lyon 1, Service de Neuro-Oncologie, Hospices Civils de Lyon, Cancer Research Center of Lyon, INSERM U1052, CNRS UMR 5286, Cancer Cell Plasticity Department, Lyon, France
| | - Jacques Guyotat
- Hospices Civils de Lyon, Université Claude Bernard Lyon 1, Service de Neuro-Oncologie, Hospices Civils de Lyon, Cancer Research Center of Lyon, INSERM U1052, CNRS UMR 5286, Cancer Cell Plasticity Department, Lyon, France
| | | | | | - John de Groot
- Departments of Neurology and Neurosurgery, University of California, San Francisco, CA, USA
| | - Jeffrey S Weinberg
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Benjamin P Liu
- Departments of Radiology and Radiation Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | | | | | | | | | | - Ahmed Idbaih
- Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau, ICM, AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, Service de Neuro-Oncologie, Paris, France
| |
Collapse
|
6
|
Navone SE, Guarnaccia L, Rizzaro MD, Begani L, Barilla E, Alotta G, Garzia E, Caroli M, Ampollini A, Violetti A, Gervasi N, Campanella R, Riboni L, Locatelli M, Marfia G. Role of Luteolin as Potential New Therapeutic Option for Patients with Glioblastoma through Regulation of Sphingolipid Rheostat. Int J Mol Sci 2023; 25:130. [PMID: 38203299 PMCID: PMC10779390 DOI: 10.3390/ijms25010130] [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: 10/25/2023] [Revised: 12/15/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024] Open
Abstract
Glioblastoma (GBM) is the most aggressive brain tumor, still considered incurable. In this study, conducted on primary GBM stem cells (GSCs), specifically selected as the most therapy-resistant, we examined the efficacy of luteolin, a natural flavonoid, as an anti-tumoral compound. Luteolin is known to impact the sphingolipid rheostat, a pathway regulated by the proliferative sphingosine-1-phosphate (S1P) and the proapoptotic ceramide (Cer), and implicated in numerous oncopromoter biological processes. Here, we report that luteolin is able to inhibit the expression of SphK1/2, the two kinases implicated in S1P formation, and to increase the expression of both SGPL1, the lyase responsible for S1P degradation, and CERS1, the ceramide synthase 1, thus shifting the balance toward the production of ceramide. In addition, luteolin proved to decrease the expression of protumoral signaling as MAPK, RAS/MEK/ERK and PI3K/AKT/mTOR and cyclins involved in cell cycle progression. In parallel, luteolin succeeded in upregulation of proapoptotic mediators as caspases and Bcl-2 family and cell cycle controllers as p53 and p27. Furthermore, luteolin determined the shutdown of autophagy contributing to cell survival. Overall, our data support the use of luteolin as add-on therapy, having demonstrated a good ability in impairing GSC viability and survival and increasing cell sensitivity to TMZ.
Collapse
Affiliation(s)
- Stefania Elena Navone
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (S.E.N.); (L.G.); (M.D.R.); (L.B.); (M.C.); (A.A.); (M.L.)
| | - Laura Guarnaccia
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (S.E.N.); (L.G.); (M.D.R.); (L.B.); (M.C.); (A.A.); (M.L.)
| | - Massimiliano D. Rizzaro
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (S.E.N.); (L.G.); (M.D.R.); (L.B.); (M.C.); (A.A.); (M.L.)
| | - Laura Begani
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (S.E.N.); (L.G.); (M.D.R.); (L.B.); (M.C.); (A.A.); (M.L.)
| | - Emanuela Barilla
- Andremacon Biotech Srl, Viale Ortles, 22/4, 20141 Milan, Italy; (E.B.); (G.A.); (R.C.); (L.R.)
| | - Giovanni Alotta
- Andremacon Biotech Srl, Viale Ortles, 22/4, 20141 Milan, Italy; (E.B.); (G.A.); (R.C.); (L.R.)
| | - Emanuele Garzia
- Reproductive Medicine Unit, Department of Mother and Child, San Paolo Hospital Medical School, ASST Santi Paolo e Carlo, 20142 Milan, Italy;
- Aerospace Medicine Institute “A. Mosso”, Italian Air Force, 20138 Milan, Italy
| | - Manuela Caroli
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (S.E.N.); (L.G.); (M.D.R.); (L.B.); (M.C.); (A.A.); (M.L.)
| | - Antonella Ampollini
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (S.E.N.); (L.G.); (M.D.R.); (L.B.); (M.C.); (A.A.); (M.L.)
| | - Aniello Violetti
- Space Attache’, Embassy of Italy in Washington DC, Washington, DC 20008, USA
| | - Noreen Gervasi
- Alcamena Stem Cell Therapeutics, 1450 South Rolling Road, Suite 4.069, Halethorpe, MD 21227, USA
| | - Rolando Campanella
- Andremacon Biotech Srl, Viale Ortles, 22/4, 20141 Milan, Italy; (E.B.); (G.A.); (R.C.); (L.R.)
| | - Laura Riboni
- Andremacon Biotech Srl, Viale Ortles, 22/4, 20141 Milan, Italy; (E.B.); (G.A.); (R.C.); (L.R.)
| | - Marco Locatelli
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (S.E.N.); (L.G.); (M.D.R.); (L.B.); (M.C.); (A.A.); (M.L.)
- Department of Medical-Surgical Physiopathology and Transplantation, University of Milan, 20122 Milan, Italy
| | - Giovanni Marfia
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (S.E.N.); (L.G.); (M.D.R.); (L.B.); (M.C.); (A.A.); (M.L.)
- Aerospace Medicine Institute “A. Mosso”, Italian Air Force, 20138 Milan, Italy
| |
Collapse
|
7
|
Olatunji G, Aderinto N, Adefusi T, Kokori E, Akinmoju O, Yusuf I, Olusakin T, Muzammil MA. Efficacy of tumour-treating fields therapy in recurrent glioblastoma: A narrative review of current evidence. Medicine (Baltimore) 2023; 102:e36421. [PMID: 38050252 PMCID: PMC10695547 DOI: 10.1097/md.0000000000036421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 11/10/2023] [Indexed: 12/06/2023] Open
Abstract
Recurrent Glioblastoma presents a formidable challenge in oncology due to its aggressive nature and limited treatment options. Tumour-Treating Fields (TTFields) Therapy, a novel therapeutic modality, has emerged as a promising approach to address this clinical conundrum. This review synthesizes the current evidence surrounding the efficacy of TTFields Therapy in the context of recurrent Glioblastoma. Diverse academic databases were explored to identify relevant studies published within the last decade. Strategic keyword selection facilitated the inclusion of studies focusing on TTFields Therapy's efficacy, treatment outcomes, and patient-specific factors. The review reveals a growing body of evidence suggesting the potential clinical benefits of TTFields Therapy for patients with recurrent Glioblastoma. Studies consistently demonstrate its positive impact on overall survival (OS) and progression-free survival (PFS). The therapy's safety profile remains favorable, with mild to moderate skin reactions being the most commonly reported adverse events. Our analysis highlights the importance of patient selection criteria, with emerging biomarkers such as PTEN mutation status influencing therapy response. Additionally, investigations into combining TTFields Therapy with other treatments, including surgical interventions and novel approaches, offer promising avenues for enhancing therapeutic outcomes. The synthesis of diverse studies underscores the potential of TTFields Therapy as a valuable addition to the armamentarium against recurrent Glioblastoma. The narrative review comprehensively explains the therapy's mechanisms, clinical benefits, adverse events, and future directions. The insights gathered herein serve as a foundation for clinicians and researchers striving to optimize treatment strategies for patients facing the challenging landscape of recurrent Glioblastoma.
Collapse
Affiliation(s)
- Gbolahan Olatunji
- Department of Medicine and Surgery, University of Ilorin, Ilorin, Nigeria
| | - Nicholas Aderinto
- Department of Medicine and Surgery, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| | | | - Emmanuel Kokori
- Department of Medicine and Surgery, University of Ilorin, Ilorin, Nigeria
| | | | - Ismaila Yusuf
- Department of Medicine and Surgery, Obafemi Awolowo University, Ife, Nigeria
| | - Tobi Olusakin
- College of Medicine, University of Ibadan, Ibadan, Nigeria
| | | |
Collapse
|
8
|
Gardam B, Gargett T, Brown MP, Ebert LM. Targeting the dendritic cell-T cell axis to develop effective immunotherapies for glioblastoma. Front Immunol 2023; 14:1261257. [PMID: 37928547 PMCID: PMC10623138 DOI: 10.3389/fimmu.2023.1261257] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 10/09/2023] [Indexed: 11/07/2023] Open
Abstract
Glioblastoma is an aggressive primary brain tumor that has seen few advances in treatments for over 20 years. In response to this desperate clinical need, multiple immunotherapy strategies are under development, including CAR-T cells, immune checkpoint inhibitors, oncolytic viruses and dendritic cell vaccines, although these approaches are yet to yield significant clinical benefit. Potential reasons for the lack of success so far include the immunosuppressive tumor microenvironment, the blood-brain barrier, and systemic changes to the immune system driven by both the tumor and its treatment. Furthermore, while T cells are essential effector cells for tumor control, dendritic cells play an equally important role in T cell activation, and emerging evidence suggests the dendritic cell compartment may be deeply compromised in glioblastoma patients. In this review, we describe the immunotherapy approaches currently under development for glioblastoma and the challenges faced, with a particular emphasis on the critical role of the dendritic cell-T cell axis. We suggest a number of strategies that could be used to boost dendritic cell number and function and propose that the use of these in combination with T cell-targeting strategies could lead to successful tumor control.
Collapse
Affiliation(s)
- Bryan Gardam
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- Translational Oncology Laboratory, Centre for Cancer Biology, University of South Australia and South Australia (SA) Pathology, Adelaide, SA, Australia
| | - Tessa Gargett
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- Translational Oncology Laboratory, Centre for Cancer Biology, University of South Australia and South Australia (SA) Pathology, Adelaide, SA, Australia
- Cancer Clinical Trials Unit, Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Michael P. Brown
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- Translational Oncology Laboratory, Centre for Cancer Biology, University of South Australia and South Australia (SA) Pathology, Adelaide, SA, Australia
- Cancer Clinical Trials Unit, Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Lisa M. Ebert
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- Translational Oncology Laboratory, Centre for Cancer Biology, University of South Australia and South Australia (SA) Pathology, Adelaide, SA, Australia
- Cancer Clinical Trials Unit, Royal Adelaide Hospital, Adelaide, SA, Australia
| |
Collapse
|
9
|
Gabashvili AN, Chmelyuk NS, Oda VV, Leonova MK, Sarkisova VA, Lazareva PA, Semkina AS, Belyakov NA, Nizamov TR, Nikitin PI. Magnetic and Fluorescent Dual-Labeled Genetically Encoded Targeted Nanoparticles for Malignant Glioma Cell Tracking and Drug Delivery. Pharmaceutics 2023; 15:2422. [PMID: 37896182 PMCID: PMC10609955 DOI: 10.3390/pharmaceutics15102422] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/21/2023] [Accepted: 09/28/2023] [Indexed: 10/29/2023] Open
Abstract
Human glioblastoma multiforme (GBM) is a primary malignant brain tumor, a radically incurable disease characterized by rapid growth resistance to classical therapies, with a median patient survival of about 15 months. For decades, a plethora of approaches have been developed to make GBM therapy more precise and improve the diagnosis of this pathology. Targeted delivery mediated by the use of various molecules (monoclonal antibodies, ligands to overexpressed tumor receptors) is one of the promising methods to achieve this goal. Here we present a novel genetically encoded nanoscale dual-labeled system based on Quasibacillus thermotolerans (Qt) encapsulins exploiting biologically inspired designs with iron-containing nanoparticles as a cargo, conjugated with human fluorescent labeled transferrin (Tf) acting as a vector. It is known that the expression of transferrin receptors (TfR) in glioma cells is significantly higher compared to non-tumor cells, which enables the targeting of the resulting nanocarrier. The selectivity of binding of the obtained nanosystem to glioma cells was studied by qualitative and quantitative assessment of the accumulation of intracellular iron, as well as by magnetic particle quantification method and laser scanning confocal microscopy. Used approaches unambiguously demonstrated that transferrin-conjugated encapsulins were captured by glioma cells much more efficiently than by benign cells. The resulting bioinspired nanoplatform can be supplemented with a chemotherapeutic drug or genotherapeutic agent and used for targeted delivery of a therapeutic agent to malignant glioma cells. Additionally, the observed cell-assisted biosynthesis of magnetic nanoparticles could be an attractive way to achieve a narrow size distribution of particles for various applications.
Collapse
Affiliation(s)
- Anna N. Gabashvili
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov Street, 119991 Moscow, Russia; (A.N.G.)
| | - Nelly S. Chmelyuk
- Laboratory “Biomedical Nanomaterials”, National University of Science and Technology “MISIS”, Leninskiy Prospekt 4, 119049 Moscow, Russia
- Department of Medical Nanobiotechnology, Pirogov Russian National Research Medical University, 1 Ostrovityanova Street, 117997 Moscow, Russia; (P.A.L.)
| | - Vera V. Oda
- MILLAB Group Ltd., 100/2 Dmitrovskoe Highway, 127247 Moscow, Russia
| | - Maria K. Leonova
- Department of Physical Chemistry, National University of Science and Technology “MISIS”, Leninskiy Prospekt 4, 119049 Moscow, Russia
| | - Viktoria A. Sarkisova
- Biology Faculty, Lomonosov Moscow State University, 1 Leninskiy Gory, 119234 Moscow, Russia
- Cell Proliferation Laboratory, Engelhardt Institute of Molecular Biology RAS, 32 Vavilov Street, 119991 Moscow, Russia
| | - Polina A. Lazareva
- Department of Medical Nanobiotechnology, Pirogov Russian National Research Medical University, 1 Ostrovityanova Street, 117997 Moscow, Russia; (P.A.L.)
| | - Alevtina S. Semkina
- Department of Medical Nanobiotechnology, Pirogov Russian National Research Medical University, 1 Ostrovityanova Street, 117997 Moscow, Russia; (P.A.L.)
- Department of Basic and Applied Neurobiology, Serbsky National Medical Research Center for Psychiatry and Narcology, 23 Kropotkinskiy Lane, 119991 Moscow, Russia
| | - Nikolai A. Belyakov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov Street, 119991 Moscow, Russia; (A.N.G.)
| | - Timur R. Nizamov
- Laboratory “Biomedical Nanomaterials”, National University of Science and Technology “MISIS”, Leninskiy Prospekt 4, 119049 Moscow, Russia
| | - Petr I. Nikitin
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov Street, 119991 Moscow, Russia; (A.N.G.)
| |
Collapse
|
10
|
Schaufler A, Sanin AY, Sandalcioglu IE, Hartmann K, Croner RS, Perrakis A, Wartmann T, Boese A, Kahlert UD, Fischer I. Concept of a fully-implantable system to monitor tumor recurrence. Sci Rep 2023; 13:16362. [PMID: 37773315 PMCID: PMC10541913 DOI: 10.1038/s41598-023-43226-3] [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: 04/12/2023] [Accepted: 09/21/2023] [Indexed: 10/01/2023] Open
Abstract
Current treatment for glioblastoma includes tumor resection followed by radiation, chemotherapy, and periodic post-operative examinations. Despite combination therapies, patients face a poor prognosis and eventual recurrence, which often occurs at the resection site. With standard MRI imaging surveillance, histologic changes may be overlooked or misinterpreted, leading to erroneous conclusions about the course of adjuvant therapy and subsequent interventions. To address these challenges, we propose an implantable system for accurate continuous recurrence monitoring that employs optical sensing of fluorescently labeled cancer cells and is implanted in the resection cavity during the final stage of tumor resection. We demonstrate the feasibility of the sensing principle using miniaturized system components, optical tissue phantoms, and porcine brain tissue in a series of experimental trials. Subsequently, the system electronics are extended to include circuitry for wireless energy transfer and power management and verified through electromagnetic field, circuit simulations and test of an evaluation board. Finally, a holistic conceptual system design is presented and visualized. This novel approach to monitor glioblastoma patients is intended to early detect recurrent cancerous tissue and enable personalization and optimization of therapy thus potentially improving overall prognosis.
Collapse
Affiliation(s)
- Anna Schaufler
- Molecular and Experimental Surgery, Clinic for General-, Visceral-, Vascular - and Transplant Surgery, Faculty of Medicine, Otto-von-Guericke University Magdeburg, 39120, Magdeburg, Germany
- Department of Neurosurgery, Otto-von-Guericke University Magdeburg, 39120, Magdeburg, Germany
- INKA Health Tech Innovation Lab., Medical Faculty, Otto-von-Guericke University Magdeburg, 39120, Magdeburg, Germany
| | - Ahmed Y Sanin
- Molecular and Experimental Surgery, Clinic for General-, Visceral-, Vascular - and Transplant Surgery, Faculty of Medicine, Otto-von-Guericke University Magdeburg, 39120, Magdeburg, Germany
- Research Campus STIMULATE, Otto-von-Guericke University Magdeburg, 39120, Magdeburg, Germany
| | - I Erol Sandalcioglu
- Department of Neurosurgery, Otto-von-Guericke University Magdeburg, 39120, Magdeburg, Germany
| | - Karl Hartmann
- Department of Neurosurgery, Otto-von-Guericke University Magdeburg, 39120, Magdeburg, Germany
| | - Roland S Croner
- Molecular and Experimental Surgery, Clinic for General-, Visceral-, Vascular - and Transplant Surgery, Faculty of Medicine, Otto-von-Guericke University Magdeburg, 39120, Magdeburg, Germany
| | - Aristotelis Perrakis
- Molecular and Experimental Surgery, Clinic for General-, Visceral-, Vascular - and Transplant Surgery, Faculty of Medicine, Otto-von-Guericke University Magdeburg, 39120, Magdeburg, Germany
| | - Thomas Wartmann
- Molecular and Experimental Surgery, Clinic for General-, Visceral-, Vascular - and Transplant Surgery, Faculty of Medicine, Otto-von-Guericke University Magdeburg, 39120, Magdeburg, Germany
| | - Axel Boese
- INKA Health Tech Innovation Lab., Medical Faculty, Otto-von-Guericke University Magdeburg, 39120, Magdeburg, Germany
| | - Ulf D Kahlert
- Molecular and Experimental Surgery, Clinic for General-, Visceral-, Vascular - and Transplant Surgery, Faculty of Medicine, Otto-von-Guericke University Magdeburg, 39120, Magdeburg, Germany
- Research Campus STIMULATE, Otto-von-Guericke University Magdeburg, 39120, Magdeburg, Germany
| | - Igor Fischer
- Department of Neurosurgery, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, 40225, Düsseldorf, Germany.
| |
Collapse
|
11
|
Alizadeh M, Broomand Lomer N, Azami M, Khalafi M, Shobeiri P, Arab Bafrani M, Sotoudeh H. Radiomics: The New Promise for Differentiating Progression, Recurrence, Pseudoprogression, and Radionecrosis in Glioma and Glioblastoma Multiforme. Cancers (Basel) 2023; 15:4429. [PMID: 37760399 PMCID: PMC10526457 DOI: 10.3390/cancers15184429] [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: 07/11/2023] [Revised: 08/29/2023] [Accepted: 08/31/2023] [Indexed: 09/29/2023] Open
Abstract
Glioma and glioblastoma multiform (GBM) remain among the most debilitating and life-threatening brain tumors. Despite advances in diagnosing approaches, patient follow-up after treatment (surgery and chemoradiation) is still challenging for differentiation between tumor progression/recurrence, pseudoprogression, and radionecrosis. Radiomics emerges as a promising tool in initial diagnosis, grading, and survival prediction in patients with glioma and can help differentiate these post-treatment scenarios. Preliminary published studies are promising about the role of radiomics in post-treatment glioma/GBM. However, this field faces significant challenges, including a lack of evidence-based solid data, scattering publication, heterogeneity of studies, and small sample sizes. The present review explores radiomics's capabilities in following patients with glioma/GBM status post-treatment and to differentiate tumor progression, recurrence, pseudoprogression, and radionecrosis.
Collapse
Affiliation(s)
- Mohammadreza Alizadeh
- Physiology Research Center, Iran University of Medical Sciences, Tehran 14496-14535, Iran;
| | - Nima Broomand Lomer
- Faculty of Medicine, Guilan University of Medical Sciences, Rasht 41937-13111, Iran;
| | - Mobin Azami
- Student Research Committee, Kurdistan University of Medical Sciences, Sanandaj 66186-34683, Iran;
| | - Mohammad Khalafi
- Radiology Department, Tabriz University of Medical Sciences, Tabriz 51656-65931, Iran;
| | - Parnian Shobeiri
- School of Medicine, Tehran University of Medical Sciences, Tehran 14167-53955, Iran; (P.S.); (M.A.B.)
| | - Melika Arab Bafrani
- School of Medicine, Tehran University of Medical Sciences, Tehran 14167-53955, Iran; (P.S.); (M.A.B.)
| | - Houman Sotoudeh
- Department of Radiology and Neurology, Heersink School of Medicine, University of Alabama at Birmingham (UAB), Birmingham, AL 35294, USA
| |
Collapse
|
12
|
Giles B, Nakhjavani M, Wiesa A, Knight T, Shigdar S, Samarasinghe RM. Unravelling the Glioblastoma Tumour Microenvironment: Can Aptamer Targeted Delivery Become Successful in Treating Brain Cancers? Cancers (Basel) 2023; 15:4376. [PMID: 37686652 PMCID: PMC10487158 DOI: 10.3390/cancers15174376] [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: 08/08/2023] [Revised: 08/28/2023] [Accepted: 08/30/2023] [Indexed: 09/10/2023] Open
Abstract
The key challenges to treating glioblastoma multiforme (GBM) are the heterogeneous and complex nature of the GBM tumour microenvironment (TME) and difficulty of drug delivery across the blood-brain barrier (BBB). The TME is composed of various neuronal and immune cells, as well as non-cellular components, including metabolic products, cellular interactions, and chemical compositions, all of which play a critical role in GBM development and therapeutic resistance. In this review, we aim to unravel the complexity of the GBM TME, evaluate current therapeutics targeting this microenvironment, and lastly identify potential targets and therapeutic delivery vehicles for the treatment of GBM. Specifically, we explore the potential of aptamer-targeted delivery as a successful approach to treating brain cancers. Aptamers have emerged as promising therapeutic drug delivery vehicles with the potential to cross the BBB and deliver payloads to GBM and brain metastases. By targeting specific ligands within the TME, aptamers could potentially improve treatment outcomes and overcome the challenges associated with larger therapies such as antibodies.
Collapse
Affiliation(s)
- Breanna Giles
- School of Medicine, Deakin University, Geelong, VIC 3220, Australia; (B.G.); (S.S.); (R.M.S.)
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC 3220, Australia
| | - Maryam Nakhjavani
- School of Medicine, Deakin University, Geelong, VIC 3220, Australia; (B.G.); (S.S.); (R.M.S.)
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC 3220, Australia
| | - Andrew Wiesa
- School of Medicine, Deakin University, Geelong, VIC 3220, Australia; (B.G.); (S.S.); (R.M.S.)
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC 3220, Australia
| | - Tareeque Knight
- School of Medicine, Deakin University, Geelong, VIC 3220, Australia; (B.G.); (S.S.); (R.M.S.)
| | - Sarah Shigdar
- School of Medicine, Deakin University, Geelong, VIC 3220, Australia; (B.G.); (S.S.); (R.M.S.)
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC 3220, Australia
| | - Rasika M. Samarasinghe
- School of Medicine, Deakin University, Geelong, VIC 3220, Australia; (B.G.); (S.S.); (R.M.S.)
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC 3220, Australia
| |
Collapse
|
13
|
Pu Y, Zhou G, Zhao K, Chen Y, Shen S. Immunotherapy for Recurrent Glioma-From Bench to Bedside. Cancers (Basel) 2023; 15:3421. [PMID: 37444531 DOI: 10.3390/cancers15133421] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/12/2023] [Accepted: 06/16/2023] [Indexed: 07/15/2023] Open
Abstract
Glioma is the most aggressive malignant tumor of the central nervous system, and most patients suffer from a recurrence. Unfortunately, recurrent glioma often becomes resistant to established chemotherapy and radiotherapy treatments. Immunotherapy, a rapidly developing anti-tumor therapy, has shown a potential value in treating recurrent glioma. Multiple immune strategies have been explored. The most-used ones are immune checkpoint blockade (ICB) antibodies, which are barely effective in monotherapy. However, when combined with other immunotherapy, especially with anti-angiogenesis antibodies, ICB has shown encouraging efficacy and enhanced anti-tumor immune response. Oncolytic viruses and CAR-T therapies have shown promising results in recurrent glioma through multiple mechanisms. Vaccination strategies and immune-cell-based immunotherapies are promising in some subgroups of patients, and multiple new tumor antigenic targets have been discovered. In this review, we discuss current applicable immunotherapies and related mechanisms for recurrent glioma, focusing on multiple preclinical models and clinical trials in the last 5 years. Through reviewing the current combination of immune strategies, we would like to provide substantive thoughts for further novel therapeutic regimes treating recurrent glioma.
Collapse
Affiliation(s)
- Yi Pu
- Laboratory of Mitochondria and Metabolism, Department of Burn and Reconstructive Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
- Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Guanyu Zhou
- Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu 610041, China
- Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Kejia Zhao
- Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu 610041, China
- Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yaohui Chen
- Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shensi Shen
- Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
- National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
14
|
Miki K, Yagi M, Noguchi N, Do Y, Otsuji R, Kuga D, Kang D, Yoshimoto K, Uchiumi T. Induction of glioblastoma cell ferroptosis using combined treatment with chloramphenicol and 2-deoxy-D-glucose. Sci Rep 2023; 13:10497. [PMID: 37380755 DOI: 10.1038/s41598-023-37483-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 06/22/2023] [Indexed: 06/30/2023] Open
Abstract
Glioblastoma, a malignant tumor, has no curative treatment. Recently, mitochondria have been considered a potential target for treating glioblastoma. Previously, we reported that agents initiating mitochondrial dysfunction were effective under glucose-starved conditions. Therefore, this study aimed to develop a mitochondria-targeted treatment to achieve normal glucose conditions. This study used U87MG (U87), U373, and patient-derived stem-like cells as well as chloramphenicol (CAP) and 2-deoxy-D-glucose (2-DG). We investigated whether CAP and 2-DG inhibited the growth of cells under normal and high glucose concentrations. In U87 cells, 2-DG and long-term CAP administration were more effective under normal glucose than high-glucose conditions. In addition, combined CAP and 2-DG treatment was significantly effective under normal glucose concentration in both normal oxygen and hypoxic conditions; this was validated in U373 and patient-derived stem-like cells. 2-DG and CAP acted by influencing iron dynamics; however, deferoxamine inhibited the efficacy of these agents. Thus, ferroptosis could be the underlying mechanism through which 2-DG and CAP act. In conclusion, combined treatment of CAP and 2-DG drastically inhibits cell growth of glioblastoma cell lines even under normal glucose conditions; therefore, this treatment could be effective for glioblastoma patients.
Collapse
Affiliation(s)
- Kenji Miki
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Higashi-Ku, Fukuoka, 812-8582, Japan
- Department of Neurosurgery, Graduate School of Medical Sciences, Kyushu University, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Mikako Yagi
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Higashi-Ku, Fukuoka, 812-8582, Japan
- Department of Health Sciences, Graduate School of Medical Sciences, Kyushu University, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Naoki Noguchi
- Department of Neurosurgery, Graduate School of Medical Sciences, Kyushu University, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Yura Do
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Ryosuke Otsuji
- Department of Neurosurgery, Graduate School of Medical Sciences, Kyushu University, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Daisuke Kuga
- Department of Neurosurgery, Graduate School of Medical Sciences, Kyushu University, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Dongchon Kang
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Koji Yoshimoto
- Department of Neurosurgery, Graduate School of Medical Sciences, Kyushu University, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Takeshi Uchiumi
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Higashi-Ku, Fukuoka, 812-8582, Japan.
- Department of Health Sciences, Graduate School of Medical Sciences, Kyushu University, Higashi-Ku, Fukuoka, 812-8582, Japan.
| |
Collapse
|
15
|
Petković M, Henis M, Heese O, Relógio A. Chronotherapy in Glioblastoma: state of the art and future perspectives. EBioMedicine 2023; 89:104470. [PMID: 36796229 PMCID: PMC9958380 DOI: 10.1016/j.ebiom.2023.104470] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 01/15/2023] [Accepted: 01/25/2023] [Indexed: 02/16/2023] Open
Abstract
Circadian rhythms regulate various processes in the human body, including drug metabolism. Chronotherapy optimizes treatment timing based on the circadian rhythm of the individual patient, such that the treatment efficacy is maximized, and adverse effects are minimized. It has been explored in different cancers with varying conclusions. Glioblastoma multiforme (GBM) is the most aggressive type of brain tumour with a very dismal prognosis. In recent years, there has been very little success in designing successful therapies to fight this disease. Chronotherapy offers the opportunity to leverage existing treatments to extend patient survival and to increase their quality of life. Here, we discuss recent advances in using chronotherapy regimens in the treatment of GMB, such as radiotherapy, temozolomide (TMZ) and bortezomib, as well as discuss novel treatments with drugs of short half-life or circadian phase specific activity, and examine the therapeutic potential of new approaches that target elements of the core circadian clock.
Collapse
Affiliation(s)
- Marina Petković
- Institute for Theoretical Biology (ITB), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin 10117, Germany
| | - Melad Henis
- Institute for Systems Medicine and Faculty of Human Medicine, MSH Medical School Hamburg, Hamburg 20457, Germany
| | - Oliver Heese
- Department of Neurosurgery and Spinal Surgery, HELIOS Medical Center Schwerin, University Campus of MSH Medical School Hamburg, Hamburg 20457, Germany
| | - Angela Relógio
- Institute for Theoretical Biology (ITB), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin 10117, Germany; Institute for Systems Medicine and Faculty of Human Medicine, MSH Medical School Hamburg, Hamburg 20457, Germany; Medical Department of Hematology, Oncology, and Tumour Immunology, Molecular Cancer Research Center (MKFZ), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin 10117, Germany.
| |
Collapse
|
16
|
Anti-Vimentin Nanobody Decreases Glioblastoma Cell Invasion In Vitro and In Vivo. Cancers (Basel) 2023; 15:cancers15030573. [PMID: 36765531 PMCID: PMC9913279 DOI: 10.3390/cancers15030573] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/06/2023] [Accepted: 01/12/2023] [Indexed: 01/20/2023] Open
Abstract
PURPOSE Glioblastoma (GBM) is the most common primary brain tumour and one of the deadliest cancers. In addition to late diagnosis and inadequate treatment, the extremely low survival rate is also due to the lack of appropriate therapeutic biomarkers and corresponding therapeutic agents. One of the potential therapeutic biomarkers is the intermediate filament vimentin, which is associated with epithelial-mesenchymal transition (EMT). The purpose of this study was to analyse the effect of the anti-vimentin nanobody Nb79 on cell invasion in vitro and in vivo. To further our understanding of the mechanism of action, we investigated the association between Nb79 and EMT in GBM and GBM stem cells by analysing the expression levels of key EMT-related proteins. METHODS The expression of vimentin in glioma tissues and cells was determined by RT-qPCR. An invasion assay was performed on differentiated glioblastoma cell line U-87 MG and stem cell line NCH421k in vitro as well as in vivo in zebrafish embryos. The effect of Nb79 on expression of EMT biomarkers beta-catenin, vimentin, ZEB-1 and ZO1 was determined by Western blot and immunocytochemistry. RESULTS Our study shows that vimentin is upregulated in glioblastoma tissue compared to lower grade glioma and non-tumour brain tissue. We demonstrated that treatment with Nb79 reduced glioblastoma cell invasion by up to 64% in vitro and up to 21% in vivo. In addition, we found that the tight junction protein ZO-1 had higher expression on the cell membrane, when treated with inhibitory anti-vimentin Nb79 compared to control. CONCLUSION In conclusion, our results suggest that anti-vimentin nanobody Nb79 is a promising tool to target glioblastoma cell invasion.
Collapse
|
17
|
Saleh HA, Mitwasi N, Ullrich M, Kubeil M, Toussaint M, Deuther-Conrad W, Neuber C, Arndt C, R. Loureiro L, Kegler A, González Soto KE, Belter B, Rössig C, Pietzsch J, Frenz M, Bachmann M, Feldmann A. Specific and safe targeting of glioblastoma using switchable and logic-gated RevCAR T cells. Front Immunol 2023; 14:1166169. [PMID: 37122703 PMCID: PMC10145173 DOI: 10.3389/fimmu.2023.1166169] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 03/30/2023] [Indexed: 05/02/2023] Open
Abstract
Glioblastoma (GBM) is still an incurable tumor that is associated with high recurrence rate and poor survival despite the current treatment regimes. With the urgent need for novel therapeutic strategies, immunotherapies, especially chimeric antigen receptor (CAR)-expressing T cells, represent a promising approach for specific and effective targeting of GBM. However, CAR T cells can be associated with serious side effects. To overcome such limitation, we applied our switchable RevCAR system to target both the epidermal growth factor receptor (EGFR) and the disialoganglioside GD2, which are expressed in GBM. The RevCAR system is a modular platform that enables controllability, improves safety, specificity and flexibility. Briefly, it consists of RevCAR T cells having a peptide epitope as extracellular domain, and a bispecific target module (RevTM). The RevTM acts as a switch key that recognizes the RevCAR epitope and the tumor-associated antigen, and thereby activating the RevCAR T cells to kill the tumor cells. However, in the absence of the RevTM, the RevCAR T cells are switched off. In this study, we show that the novel EGFR/GD2-specific RevTMs can selectively activate RevCAR T cells to kill GBM cells. Moreover, we show that gated targeting of GBM is possible with our Dual-RevCAR T cells, which have their internal activation and co-stimulatory domains separated into two receptors. Therefore, a full activation of Dual-RevCAR T cells can only be achieved when both receptors recognize EGFR and GD2 simultaneously via RevTMs, leading to a significant killing of GBM cells both in vitro and in vivo.
Collapse
Affiliation(s)
- Haidy A. Saleh
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Nicola Mitwasi
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Martin Ullrich
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Manja Kubeil
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Magali Toussaint
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Winnie Deuther-Conrad
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Christin Neuber
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Claudia Arndt
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
- Faculty of Medicine Carl Gustav Carus, Mildred Scheel Early Career Center, Technische Universität Dresden, Dresden, Germany
| | - Liliana R. Loureiro
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Alexandra Kegler
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | | | - Birgit Belter
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Claudia Rössig
- Department of Pediatric Hematology and Oncology, University Children’s Hospital Münster, Münster, Germany
| | - Jens Pietzsch
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
- Faculty of Chemistry and Food Chemistry, School of Science, Technische Universität Dresden, Dresden, Germany
| | - Marcus Frenz
- Faculty Informatik and Wirtschaftsinformatik, Provadis School of International Management and Technology AG, Frankfurt, Germany
| | - Michael Bachmann
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
- National Center for Tumor Diseases Dresden (NCT/UCC), German Cancer Research Center (DKFZ), Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site, Dresden, Germany
- *Correspondence: Michael Bachmann,
| | - Anja Feldmann
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
- National Center for Tumor Diseases Dresden (NCT/UCC), German Cancer Research Center (DKFZ), Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site, Dresden, Germany
| |
Collapse
|
18
|
Thang M, Mellows C, Mercer-Smith A, Nguyen P, Hingtgen S. Current approaches in enhancing TRAIL therapies in glioblastoma. Neurooncol Adv 2023; 5:vdad047. [PMID: 37215952 PMCID: PMC10195206 DOI: 10.1093/noajnl/vdad047] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2023] Open
Abstract
Glioblastoma (GBM) is the most prevalent, aggressive, primary brain cancer in adults and continues to pose major medical challenges due in part to its high rate of recurrence. Extensive research is underway to discover new therapies that target GBM cells and prevent the inevitable recurrence in patients. The pro-apoptotic protein tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has attracted attention as an ideal anticancer agent due to its ability to selectively kill cancer cells with minimal toxicity in normal cells. Although initial clinical evaluations of TRAIL therapies in several cancers were promising, later stages of clinical trial results indicated that TRAIL and TRAIL-based therapies failed to demonstrate robust efficacies due to poor pharmacokinetics, resulting in insufficient concentrations of TRAIL at the therapeutic site. However, recent studies have developed novel ways to prolong TRAIL bioavailability at the tumor site and efficiently deliver TRAIL and TRAIL-based therapies using cellular and nanoparticle vehicles as drug loading cargos. Additionally, novel techniques have been developed to address monotherapy resistance, including modulating biomarkers associated with TRAIL resistance in GBM cells. This review highlights the promising work to overcome the challenges of TRAIL-based therapies with the aim to facilitate improved TRAIL efficacy against GBM.
Collapse
Affiliation(s)
- Morrent Thang
- Neuroscience Center, University of North Carolina—Chapel Hill School of Medicine, Chapel Hill, North Carolina, USA
- Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina—Chapel Hill School of Pharmacy, Chapel Hill, North Carolina, USA
| | - Clara Mellows
- Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina—Chapel Hill School of Pharmacy, Chapel Hill, North Carolina, USA
| | - Alison Mercer-Smith
- Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina—Chapel Hill School of Pharmacy, Chapel Hill, North Carolina, USA
| | - Phuong Nguyen
- Michigan State University School of Medicine, East Lansing, Michigan, USA
| | - Shawn Hingtgen
- Corresponding Author: Shawn Hingtgen, PhD, Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina at Chapel Hill, Eshelman School of Pharmacy, 125 Mason Farm Road, Chapel Hill, NC 27599-7363, USA ()
| |
Collapse
|
19
|
Miki K, Yagi M, Yoshimoto K, Kang D, Uchiumi T. Mitochondrial dysfunction and impaired growth of glioblastoma cell lines caused by antimicrobial agents inducing ferroptosis under glucose starvation. Oncogenesis 2022; 11:59. [PMID: 36195584 PMCID: PMC9532440 DOI: 10.1038/s41389-022-00437-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 09/23/2022] [Accepted: 09/26/2022] [Indexed: 11/09/2022] Open
Abstract
Glioblastoma is a difficult-to-cure disease owing to its malignancy. Under normal circumstances, cancer is dependent on the glycolytic system for growth, and mitochondrial oxidative phosphorylation (OXPHOS) is not well utilized. Here, we investigated the efficacy of mitochondria-targeted glioblastoma therapy in cell lines including U87MG, LN229, U373, T98G, and two patient-derived stem-like cells. When glioblastoma cells were exposed to a glucose-starved condition (100 mg/l), they rely on mitochondrial OXPHOS for growth, and mitochondrial translation product production is enhanced. Under these circumstances, drugs that inhibit mitochondrial translation, called antimicrobial agents, can cause mitochondrial dysfunction and thus can serve as a therapeutic option for glioblastoma. Antimicrobial agents activated the nuclear factor erythroid 2-related factor 2–Kelch-like ECH-associated protein 1 pathway, resulting in increased expression of heme oxygenase-1. Accumulation of lipid peroxides resulted from the accumulation of divalent iron, and cell death occurred via ferroptosis. In conclusion, mitochondrial OXPHOS is upregulated in glioblastoma upon glucose starvation. Under this condition, antimicrobial agents cause cell death via ferroptosis. The findings hold promise for the treatment of glioblastoma.
Collapse
Affiliation(s)
- Kenji Miki
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka, 812-8582, Japan.,Department of Neurosurgery, Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Mikako Yagi
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka, 812-8582, Japan.,Department of Health Sciences, Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Koji Yoshimoto
- Department of Neurosurgery, Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Dongchon Kang
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Takeshi Uchiumi
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka, 812-8582, Japan. .,Department of Health Sciences, Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka, 812-8582, Japan.
| |
Collapse
|
20
|
Protein Quality Control in Glioblastoma: A Review of the Current Literature with New Perspectives on Therapeutic Targets. Int J Mol Sci 2022; 23:ijms23179734. [PMID: 36077131 PMCID: PMC9456419 DOI: 10.3390/ijms23179734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/12/2022] [Accepted: 08/24/2022] [Indexed: 11/17/2022] Open
Abstract
Protein quality control allows eukaryotes to maintain proteostasis under the stress of constantly changing conditions. In this review, we discuss the current literature on PQC, highlighting flaws that must exist for malignancy to occur. At the nidus of PQC, the expression of BAG1-6 reflects the cell environment; each isoform directs proteins toward different, parallel branches of the quality control cascade. The sum of these branches creates a net shift toward either homeostasis or apoptosis. With an established role in ALP, Bag3 is necessary for cell survival in stress conditions including those of the cancerous niche (i.e., hypoxia, hypermutation). Evidence suggests that excessive Bag3–HSP70 activity not only sustains, but also propagates cancers. Its role is anti-apoptotic—which allows malignant cells to persist—and intercellular—with the production of infectious ‘oncosomes’ enabling cancer expansion and recurrence. While Bag3 has been identified as a key prognostic indicator in several cancer types, its investigation is limited regarding glioblastoma. The cochaperone HSP70 has been strongly linked with GBM, while ALP inhibitors have been shown to improve GBM susceptibility to chemotherapeutics. Given the highly resilient, frequently recurrent nature of GBM, the targeting of Bag3 is a necessary consideration for the successful and definitive treatment of GBM.
Collapse
|
21
|
Patil VM, Menon N, Chatterjee A, Tonse R, Choudhari A, Mahajan A, Puranik AD, Epari S, Jadhav M, Pathak S, Peelay Z, Walavalkar R, Muthuluri HK, Ravi Krishna M, Chandrasekharan A, Pande N, Gupta T, Banavali S, Jalali R. Mebendazole plus lomustine or temozolomide in patients with recurrent glioblastoma: A randomised open-label phase II trial. EClinicalMedicine 2022; 49:101449. [PMID: 35747192 PMCID: PMC9156991 DOI: 10.1016/j.eclinm.2022.101449] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 04/15/2022] [Accepted: 04/25/2022] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND Recurrent glioblastoma (GBM) has dismal outcomes and limited treatment options. Mebendazole (MBZ) has activity in glioma both in-vivo and in-vitro, and is well tolerated in combination with lomustine (CCNU) and temozolomide (TMZ). In this study, we sought to determine whether the addition of MBZ to CCNU or TMZ would improve overall survival (OS) in recurrent GBM. METHODS In this phase II randomized open-label trial, adult patients with ECOG PS 0-3, with recurrent GBM who were not eligible for re-radiation, were randomized 1:1 to the CCNU-MBZ and TMZ-MBZ arms. CCNU was administered at 110 mg/m2 every 6 weeks with MBZ 800 mg thrice daily and TMZ was administered at 200 mg/m2 once daily on days 1-5 of a 28 days cycle with MBZ 1600 mg thrice daily. The primary endpoint was OS at 9 months. A 9-month OS of 55% or more in any arm was hypothesized to warrant further evaluation and a value below 35% was too low to warrant further investigation. OS was analyzed using intention to treat (ITT) and per-protocol (PP) analyses. Per-protocol analysis was used for safety analysis. Clinical Trials Registry-India number, CTRI/2018/01/011542. FINDINGS Participants were recruited from 14th March 2019 to 18th June 2021, 44 patients were randomised on each arm. At 17.4 months, 68 events for OS analysis had occurred, 33 in the TMZ-MBZ and 35 in the CCNU-MBZ arm. The 9-month OS was 36.6% (95% CI 22.3-51.0) and 45% (95% CI 29.6-59.2) in the TMZ-MBZ and CCNU-MBZ arms respectively, in the ITT population. ECOG PS was the only independent prognostic factor impacting OS (HR-0.48, 95% CI 0.27-0.85; P = 0.012). Grade 3-5 adverse events were seen in 8 (18.6%; n = 43) and 4 (9.5%; n = 42) patients in the TMZ-MBZ and CCNU-MBZ arms respectively. There were no treatment related deaths. INTERPRETATION The addition of MBZ to TMZ or CCNU failed to achieve the pre-set benchmark of 55% 9-month OS. This was probably due to 28.6% of patients having poor PS of 2-3. FUNDING Brain Tumor Foundation (BTF) of India, Indian Cooperative Oncology Network (ICON), and India Cancer Research Consortium (ICRC) under ICMR (Indian Council of Medical Research).
Collapse
Affiliation(s)
- Vijay M. Patil
- Department of Medical Oncology, Tata Memorial Center, Mumbai, India
- Homi Bhabha National Institute (HBNI), Mumbai, India
- Corresponding author at: Department of Medical Oncology, Tata Memorial Hospital, 1110, Homi Bhabha Block, Parel, Mumbai, India.
| | - Nandini Menon
- Department of Medical Oncology, Tata Memorial Center, Mumbai, India
- Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Abhishek Chatterjee
- Department of Radiation Oncology, Tata Memorial Center, Mumbai, India
- Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Raees Tonse
- Department of Radiation Oncology, Tata Memorial Center, Mumbai, India
- Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Amit Choudhari
- Department of Radiology, Tata Memorial Center, Mumbai, India
- Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Abhishek Mahajan
- Department of Radiology, Tata Memorial Center, Mumbai, India
- Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Ameya D. Puranik
- Department of Nuclear Medicine, Tata Memorial Center, Mumbai, India
- Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Sridhar Epari
- Department of Pathology, Tata Memorial Center, Mumbai, India
- Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Monica Jadhav
- Department of Medical Oncology, Tata Memorial Center, Mumbai, India
- Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Shruti Pathak
- Department of Medical Oncology, Tata Memorial Center, Mumbai, India
- Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Zoya Peelay
- Department of Medical Oncology, Tata Memorial Center, Mumbai, India
- Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Rutuja Walavalkar
- Department of Medical Oncology, Tata Memorial Center, Mumbai, India
- Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Hemanth K. Muthuluri
- Department of Medical Oncology, Tata Memorial Center, Mumbai, India
- Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Madala Ravi Krishna
- Department of Medical Oncology, Tata Memorial Center, Mumbai, India
- Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Arun Chandrasekharan
- Department of Medical Oncology, Tata Memorial Center, Mumbai, India
- Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Nikhil Pande
- Department of Medical Oncology, Tata Memorial Center, Mumbai, India
- Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Tejpal Gupta
- Department of Radiation Oncology, Tata Memorial Center, Mumbai, India
- Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Shripad Banavali
- Department of Medical Oncology, Tata Memorial Center, Mumbai, India
- Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Rakesh Jalali
- Department of Radiation Oncology, Tata Memorial Center, Mumbai, India
- Homi Bhabha National Institute (HBNI), Mumbai, India
| |
Collapse
|
22
|
Krigers A, Pinggera D, Demetz M, Kornberger LM, Kerschbaumer J, Thomé C, Freyschlag CF. The Routine Application of Tumor-Treating Fields in the Treatment of Glioblastoma WHO° IV. Front Neurol 2022; 13:900377. [PMID: 35785334 PMCID: PMC9243748 DOI: 10.3389/fneur.2022.900377] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 04/25/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction:Tumor-treating fields (TTFs) are a specific local oncological treatment modality in glioblastoma multiforme WHO° IV (GBM). Their mechanism of action is based on the effect of electrical fields interfering with the mitotic activity of malignant cells. Prospective studies have demonstrated efficacy, but TTF benefits are still controversially discussed. This treatment was implemented in our center as the standard of care in January 2016. We thus discuss the current state of the art and our long-term experience in the routine application of TTF.MethodsThe data of 48 patients suffering from GBM and treated with TTF were assessed and compared with previously published studies. Up-to-date information from open sources was evaluated.ResultsA total of 31 males and 17 females harboring a GBM were treated with TTF, between January 2016 and August 2021, in our center. In 98% of cases, TTFs were started within 6 weeks after concomitant radiochemotherapy (Stupp protocol). Mean overall survival was 22.6 months (95% CI: 17.3–27.9). Current indications, benefits, and restrictions were evaluated. Future TTF opportunities and ongoing studies were reviewed.ConclusionTTFs are a feasible and routinely applicable specific oncological treatment option for glioblastoma multiforme WHO° IV. Further research is ongoing to extend the indications and the efficacy of TTF.
Collapse
|
23
|
Odia Y, Cavalcante L, Safran H, Powell SF, Munster PN, Ma WW, Carneiro BA, Bastos BR, Mikrut S, Mikrut W, Giles FJ, Sahebjam S. Malignant glioma subset from actuate 1801: Phase I/II study of 9-ING-41, GSK-3β inhibitor, monotherapy or combined with chemotherapy for refractory malignancies. Neurooncol Adv 2022; 4:vdac012. [PMID: 35402914 PMCID: PMC8989389 DOI: 10.1093/noajnl/vdac012] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background GSK3β serine/threonine kinase regulates metabolism and glycogen biosynthesis. GSK3β overexpression promotes progression and resistance through NF-κB and p53 apoptotic pathways. GSK3β inhibits immunomodulation by downregulating PD-L1 and LAG-3 checkpoints and increasing NK and T-cell tumor killing. 9-ING-41, a small-molecule, selective GSK3β inhibitor, showed preclinical activity in chemo-resistant PDX glioblastoma models, including enhanced lomustine antitumor effect. Methods Refractory malignancies (n = 162) were treated with 9-ING-41 monotherapy (n = 65) or combined with 8 cytotoxic regimens after prior exposure (NCT03678883). Recurrent gliomas (n = 18) were treated with 9-ING-41 IV TIW q21day cycles at 3.3, 5, 9.3, 15 mg/kg, as monotherapy or combined with lomustine 30 mg/m² PO weekly q84day cycles. Primary objective was safety. Results RP2D of 15 mg/kg IV TIW was confirmed across all 9 regimens, no accentuated chemotherapy toxicity noted. Glioma subtypes included: 13 glioblastoma, 2 anaplastic astrocytomas, 1 anaplastic oligodendroglioma, 1 astrocytoma. Median age 52 (30-69) years; 6 female, 12 male; median ECOG 1 (0-2); median recurrences 3 (1-6). All received upfront radiation/temozolomide (18/18), plus salvage nitrosoureas (15/18), bevacizumab (8/18), TTFields (6/18), or immunotherapy (4/18). IDH/mutation(3/18); 1p19q/codeletion(1/18); MGMT/methylated(1/18). Four received 9-ING-41 monotherapy, 14 concurrent with lomustine. No severe toxicities were attributed to 9-ING-41, only mild vision changes (9/18, 50%), or infusion reactions (4/18, 22%). Lomustine-related toxicities: G3/4 thrombocytopenia (3/14, 21%), G1/2 fatigue (4/14, 28%). Median days on therapy was 55 (4-305); 1 partial response (>50%) was noted. Median OS was 5.5 (95% CI: 2.8-11.4) months and PFS-6 was 16.7%. Conclusion 9-ING-41 plus/minus lomustine is safe and warrants further study in glioma patients.
Collapse
Affiliation(s)
- Yazmin Odia
- Department of Neuro-Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, Florida, USA,Corresponding Author: Yazmin Odia, MD MS FAAN, Chief of Neuro-Oncology, Miami Cancer Institute, Baptist Health South Florida, 8900 North Kendall Drive, Miami, FL 33176, USA ()
| | | | - Howard Safran
- Department of Hematology Oncology, Cancer Center at Brown University, Lifespan Cancer Institute, Providence, Rhode Island, USA
| | | | - Pamela N Munster
- Department of Hematology Oncology, University of California San Francisco, San Francisco, California, USA
| | - Wen Wee Ma
- Department of Medical Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Benedito A Carneiro
- Department of Hematology Oncology, Cancer Center at Brown University, Lifespan Cancer Institute, Providence, Rhode Island, USA
| | - Bruno R Bastos
- Department of Neuro-Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, Florida, USA
| | | | | | | | - Solmaz Sahebjam
- Department of Neuro-Oncology, Moffitt Cancer Center & Research Institute, University of South Florida, Tampa, Florida, USA,Present affiliation: National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
24
|
Cardona AF, Jaramillo-Velásquez D, Ruiz-Patiño A, Polo C, Jiménez E, Hakim F, Gómez D, Ramón JF, Cifuentes H, Mejía JA, Salguero F, Ordoñez C, Muñoz Á, Bermúdez S, Useche N, Pineda D, Ricaurte L, Zatarain-Barrón ZL, Rodríguez J, Avila J, Rojas L, Jaller E, Sotelo C, Garcia-Robledo JE, Santoyo N, Rolfo C, Rosell R, Arrieta O. Efficacy of osimertinib plus bevacizumab in glioblastoma patients with simultaneous EGFR amplification and EGFRvIII mutation. J Neurooncol 2021; 154:353-364. [PMID: 34498213 DOI: 10.1007/s11060-021-03834-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 08/19/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Amplification of EGFR and its active mutant EGFRvIII are common in glioblastoma (GB). While EGFR and EGFRvIII play critical roles in pathogenesis, targeted therapy with EGFR-tyrosine kinase inhibitors or antibodies has shown limited efficacy. To improve the likelihood of effectiveness, we targeted adult patients with recurrent GB enriched for simultaneous EGFR amplification and EGFRvIII mutation, with osimertinib/bevacizumab at doses described for non-small cell lung cancer. METHODS We retrospectively explored whether previously described EGFRvIII mutation in association with EGFR gene amplification could predict response to osimertinib/bevacizumab combination in a subset of 15 patients treated at recurrence. The resistance pattern in a subgroup of subjects is described using a commercial next-generation sequencing panel in liquid biopsy. RESULTS There were ten males (66.7%), and the median patient's age was 56 years (range 38-70 years). After their initial diagnosis, 12 patients underwent partial (26.7%) or total resection (53.3%). Subsequently, all cases received IMRT and concurrent and adjuvant temozolomide (TMZ; the median number of cycles 9, range 6-12). The median follow-up after recurrence was 17.1 months (95% CI 12.3-22.6). All patients received osimertinib/bevacizumab as a second-line intervention with a median progression-free survival (PFS) of 5.1 months (95% CI 2.8-7.3) and overall survival of 9.0 months (95% CI 3.9-14.0). The PFS6 was 46.7%, and the overall response rate was 13.3%. After exposure to the osimertinib/bevacizumab combination, the main secondary alterations were MET amplification, STAT3, IGF1R, PTEN, and PDGFR. CONCLUSIONS While the osimertinib/bevacizumab combination was marginally effective in most GB patients with simultaneous EGFR amplification plus EGFRvIII mutation, a subgroup experienced a long-lasting meaningful benefit. The findings of this brief cohort justify the continuation of the research in a clinical trial. The pattern of resistance after exposure to osimertinib/bevacizumab includes known mechanisms in the regulation of EGFR, findings that contribute to the understanding and targeting in a stepwise rational this pathway.
Collapse
Affiliation(s)
- Andrés F Cardona
- Clinical and Translational Oncology Group, Brain Tumor Unit, Clínica del Country, Calle 116 No. 9 - 72, c. 318, Bogotá, Colombia. .,Foundation for Clinical and Applied Cancer Research - FICMAC, Bogotá, Colombia. .,Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad El Bosque, Bogotá, Colombia. .,Thoracic Oncology Unit, Clínica del Country, Bogotá, Colombia.
| | | | - Alejandro Ruiz-Patiño
- Foundation for Clinical and Applied Cancer Research - FICMAC, Bogotá, Colombia.,Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad El Bosque, Bogotá, Colombia
| | - Carolina Polo
- Foundation for Clinical and Applied Cancer Research - FICMAC, Bogotá, Colombia.,Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad El Bosque, Bogotá, Colombia
| | - Enrique Jiménez
- Neurosurgery Department, Fundación Santa Fe de Bogotá, Bogotá, Colombia
| | - Fernando Hakim
- Neurosurgery Department, Fundación Santa Fe de Bogotá, Bogotá, Colombia
| | - Diego Gómez
- Neurosurgery Department, Fundación Santa Fe de Bogotá, Bogotá, Colombia
| | | | | | | | - Fernando Salguero
- Neurosurgery Department, Fundación Santa Fe de Bogotá, Bogotá, Colombia
| | - Camila Ordoñez
- Foundation for Clinical and Applied Cancer Research - FICMAC, Bogotá, Colombia.,Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad El Bosque, Bogotá, Colombia
| | - Álvaro Muñoz
- Radio-Oncology Department, Fundación Santa Fe de Bogotá, Bogotá, Colombia
| | - Sonia Bermúdez
- Neuroradiology Section, Radiology Department, Fundación Santa Fe de Bogotá, Bogotá, Colombia
| | - Nicolas Useche
- Neuroradiology Section, Radiology Department, Fundación Santa Fe de Bogotá, Bogotá, Colombia
| | - Diego Pineda
- Neuroradiology Section, Radiology Department, Clínica del Country, Bogotá, Colombia
| | | | | | - July Rodríguez
- Foundation for Clinical and Applied Cancer Research - FICMAC, Bogotá, Colombia.,Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad El Bosque, Bogotá, Colombia
| | - Jenny Avila
- Foundation for Clinical and Applied Cancer Research - FICMAC, Bogotá, Colombia.,Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad El Bosque, Bogotá, Colombia
| | - Leonardo Rojas
- Clinical and Translational Oncology Group, Brain Tumor Unit, Clínica del Country, Calle 116 No. 9 - 72, c. 318, Bogotá, Colombia.,Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad El Bosque, Bogotá, Colombia.,Clinical Oncology Department, Clínica Colsanitas, Bogotá, Colombia
| | - Elvira Jaller
- Foundation for Clinical and Applied Cancer Research - FICMAC, Bogotá, Colombia.,Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad El Bosque, Bogotá, Colombia
| | - Carolina Sotelo
- Foundation for Clinical and Applied Cancer Research - FICMAC, Bogotá, Colombia.,Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad El Bosque, Bogotá, Colombia
| | | | - Nicolas Santoyo
- Foundation for Clinical and Applied Cancer Research - FICMAC, Bogotá, Colombia.,Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad El Bosque, Bogotá, Colombia
| | - Christian Rolfo
- Center for Thoracic Oncology, Tisch Cáncer Center, Mount Sinai Hospital System & Icahn School of Medicine, Mount Sinai, New York, NY, USA
| | - Rafael Rosell
- Cancer Biology and Precision Medicine Program, Catalan Institute of Oncology, Barcelona, Spain
| | - Oscar Arrieta
- Laboratory of Personalized Medicine, Instituto Nacional de Cancerología (INCan), Mexico City, Mexico
| |
Collapse
|