551
|
Zhao M, Bozzato E, Joudiou N, Ghiassinejad S, Danhier F, Gallez B, Préat V. Codelivery of paclitaxel and temozolomide through a photopolymerizable hydrogel prevents glioblastoma recurrence after surgical resection. J Control Release 2019; 309:72-81. [PMID: 31306678 DOI: 10.1016/j.jconrel.2019.07.015] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 07/02/2019] [Accepted: 07/11/2019] [Indexed: 01/03/2023]
Abstract
A photopolymerizable hydrogel-based local drug delivery system was developed for the postsurgical treatment of glioblastoma (GBM). We aimed for a local drug combination therapy with paclitaxel (PTX) and temozolomide (TMZ) within a hydrogel to synergistically inhibit tumor growth. The in vitro cytotoxicity of TMZ was assessed in U87MG cells. We demonstrated the synergistic effect of PTX and TMZ on U87MG cells by clonogenic assay. Treatment with TMZ did not induce O6-methylguanine-DNA methyltransferase related drug resistance in tumor-bearing mice. PTX had sustained release for at least 1 month in vivo in healthy mice brains. The drug combination was tolerable and suppressed tumor growth more efficiently than the single drugs in the U87MG orthotopic tumor model. The PTX and TMZ codelivery hydrogel showed superior antitumor effects and can be considered a promising approach for the postsurgical treatment of GBM.
Collapse
Affiliation(s)
- Mengnan Zhao
- Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier, 73, B1.73.12, 1200 Brussels, Belgium
| | - Elia Bozzato
- Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier, 73, B1.73.12, 1200 Brussels, Belgium
| | - Nicolas Joudiou
- Université catholique de Louvain, Louvain Drug Research Institute, Nuclear and Electron Spin Technologies Platform (NEST), Avenue Mounier, 73, B1.73.08, 1200 Brussels, Belgium
| | - Sina Ghiassinejad
- Université catholique de Louvain, Institute of Condensed Matter and Nanoscience (IMCN), Bio and Soft Matter, Croix du Sud 1, B-1348 Louvain-la-Neuve, Belgium
| | - Fabienne Danhier
- Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier, 73, B1.73.12, 1200 Brussels, Belgium
| | - Bernard Gallez
- Université catholique de Louvain, Louvain Drug Research Institute, Biomedical Magnetic Resonance Research Group, Avenue Mounier, 73, B1.73.08, 1200 Brussels, Belgium
| | - Véronique Préat
- Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier, 73, B1.73.12, 1200 Brussels, Belgium.
| |
Collapse
|
552
|
Affiliation(s)
- Timothy F Cloughesy
- UCLA Neuro-Oncology Program and Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Andrew B Lassman
- Department of Neurology and Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, USA
| |
Collapse
|
553
|
Increased incidence of second primary malignancy in patients with malignant astrocytoma: a population-based study. Biosci Rep 2019; 39:BSR20181968. [PMID: 31138756 PMCID: PMC6566465 DOI: 10.1042/bsr20181968] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 04/17/2019] [Accepted: 05/06/2019] [Indexed: 11/18/2022] Open
Abstract
We identified patients diagnosed with malignant astrocytoma (MA) as the first of two or more primary malignancies between 1973 and 2015 from Surveillance, Epidemiology and End Results (SEER) database. Multiple primaries-standardized incidence ratio (MP-SIR) was calculated to quantitate the risk of second primary malignancy (SPM). We further identified the risk factors of developing SPM and factors affecting overall survival (OS) in MA patients with SPM. Our results revealed that overall risk of SPM among MA patients was significantly higher than that in general population (SIR: 1.09, 95% confidence interval (CI): 1–1.18, P<0.05). Specific sites where the risk of SPM increased included salivary gland, bone and joints, soft tissue including heart, brain, cranial nerves other nervous system, thyroid, acute non-lymphocytic leukemia and acute myeloid leukemia. Overall risk of SPM in patients aged ≤29 and 30–59 years significantly increased (4.34- and 1.41-fold respectively). Whereas patients aged ≥60 years had a significantly decreased risk of SPM. Patients in the group of latency at 36–59, 60–119 and ≥120 months carried significantly increased overall risk of SPM. Multivariate analysis revealed that age, race, marital status, WHO grade, differentiated grade of cancer tissues, latency was independent predictor of OS in MA patients with SPM, which were all selected into the nomogram. The calibration curve for probability of survival showed good agreement between prediction by nomogram and actual observation. In conclusion, MA survivors should be advised of their increased risk for developing certain cancers in their lifetime. Our study had clinical implications for the surveillance of MA survivors at risk of developing SPM.
Collapse
|
554
|
Facchin F, Canaider S, Tassinari R, Zannini C, Bianconi E, Taglioli V, Olivi E, Cavallini C, Tausel M, Ventura C. Physical energies to the rescue of damaged tissues. World J Stem Cells 2019; 11:297-321. [PMID: 31293714 PMCID: PMC6600852 DOI: 10.4252/wjsc.v11.i6.297] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Revised: 04/24/2019] [Accepted: 05/29/2019] [Indexed: 02/06/2023] Open
Abstract
Rhythmic oscillatory patterns sustain cellular dynamics, driving the concerted action of regulatory molecules, microtubules, and molecular motors. We describe cellular microtubules as oscillators capable of synchronization and swarming, generating mechanical and electric patterns that impact biomolecular recognition. We consider the biological relevance of seeing the inside of cells populated by a network of molecules that behave as bioelectronic circuits and chromophores. We discuss the novel perspectives disclosed by mechanobiology, bioelectromagnetism, and photobiomodulation, both in term of fundamental basic science and in light of the biomedical implication of using physical energies to govern (stem) cell fate. We focus on the feasibility of exploiting atomic force microscopy and hyperspectral imaging to detect signatures of nanomotions and electromagnetic radiation (light), respectively, generated by the stem cells across the specification of their multilineage repertoire. The chance is reported of using these signatures and the diffusive features of physical waves to direct specifically the differentiation program of stem cells in situ, where they already are resident in all the tissues of the human body. We discuss how this strategy may pave the way to a regenerative and precision medicine without the needs for (stem) cell or tissue transplantation. We describe a novel paradigm based upon boosting our inherent ability for self-healing.
Collapse
Affiliation(s)
- Federica Facchin
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), School of Medicine, University of Bologna, Bologna 40100, Italy
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems, CNR, Bologna 40100, Italy
| | - Silvia Canaider
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), School of Medicine, University of Bologna, Bologna 40100, Italy
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems, CNR, Bologna 40100, Italy
| | - Riccardo Tassinari
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems, CNR, Bologna 40100, Italy
| | - Chiara Zannini
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems, CNR, Bologna 40100, Italy
| | - Eva Bianconi
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems, CNR, Bologna 40100, Italy
| | - Valentina Taglioli
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems, CNR, Bologna 40100, Italy
| | - Elena Olivi
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems, CNR, Bologna 40100, Italy
| | - Claudia Cavallini
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems, CNR, Bologna 40100, Italy
| | | | - Carlo Ventura
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), School of Medicine, University of Bologna, Bologna 40100, Italy
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems, CNR, Bologna 40100, Italy.
| |
Collapse
|
555
|
Radke J, Koch A, Pritsch F, Schumann E, Misch M, Hempt C, Lenz K, Löbel F, Paschereit F, Heppner FL, Vajkoczy P, Koll R, Onken J. Predictive MGMT status in a homogeneous cohort of IDH wildtype glioblastoma patients. Acta Neuropathol Commun 2019; 7:89. [PMID: 31167648 PMCID: PMC6549362 DOI: 10.1186/s40478-019-0745-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 05/20/2019] [Indexed: 12/26/2022] Open
Abstract
Methylation of the O(6)-Methylguanine-DNA methyltransferase (MGMT) promoter is predictive for treatment response in glioblastoma patients. However, precise predictive cutoff values to distinguish “MGMT methylated” from “MGMT unmethylated” patients remain highly debated in terms of pyrosequencing (PSQ) analysis. We retrospectively analyzed a clinically and molecularly very well-characterized cohort of 111 IDH wildtype glioblastoma patients, who underwent gross total tumor resection and received standard Stupp treatment. Detailed clinical parameters were obtained. Predictive cutoff values for MGMT promoter methylation were determined using ROC curve analysis and survival curve comparison using Log-rank (Mantel-Cox) test. MGMT status was analyzed using pyrosequencing (PSQ), semi-quantitative methylation specific PCR (sqMSP) and direct bisulfite sequencing (dBiSeq). Highly methylated (> 20%) MGMT correlated with significantly improved progression-free survival (PFS) and overall survival (OS) in our cohort. Median PFS was 7.2 months in the unmethylated group (UM, < 10% mean methylation), 10.4 months in the low methylated group (LM, 10-20% mean methylation) and 19.83 months in the highly methylated group (HM, > 20% mean methylation). Median OS was 13.4 months for UM, 17.9 months for LM and 29.93 months for HM. Within the LM group, correlation of PSQ and sqMSP or dBiSeq was only conclusive in 51.5% of our cases. ROC curve analysis revealed superior test precision for survival if additional sqMSP results were considered (AUC = 0.76) compared to PSQ (cutoff 10%) alone (AUC = 0.67). We therefore challenge the widely used, strict PSQ cutoff at 10% which might not fully reflect the clinical response to alkylating agents and suggest applying a second method for MGMT testing (e.g. MSP) to confirm PSQ results for patients with LM MGMT levels if therapeutically relevant.
Collapse
|
556
|
Reulen HJ, Suero Molina E, Zeidler R, Gildehaus FJ, Böning G, Gosewisch A, Stummer W. Intracavitary radioimmunotherapy of high-grade gliomas: present status and future developments. Acta Neurochir (Wien) 2019; 161:1109-1124. [PMID: 30980242 DOI: 10.1007/s00701-019-03882-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Accepted: 03/20/2019] [Indexed: 02/07/2023]
Abstract
There is a distinct need for new and second-line therapies to delay or prevent local tumor regrowth after current standard of care therapy. Intracavitary radioimmunotherapy, in combination with radiotherapy, is discussed in the present review as a therapeutic strategy of high potential. We performed a systematic literature search following the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA). The available body of literature on intracavitary radioimmunotherapy (iRIT) in glioblastoma and anaplastic astrocytomas is presented. Several past and current phase I and II clinical trials, using mostly an anti-tenascin monoclonal antibody labeled with I-131, have shown median overall survival of 19-25 months in glioblastoma, while adverse events remain low. Tenascin, followed by EGFR and variants, or smaller peptides have been used as targets, and most clinical studies were performed with I-131 or Y-90 as radionuclides while only recently Re-188, I-125, and Bi-213 were applied. The pharmacokinetics of iRIT, as well as the challenges encountered with this therapy, is comprehensively discussed. This promising approach deserves further exploration in future studies by incorporating several innovative modifications.
Collapse
Affiliation(s)
| | - Eric Suero Molina
- Department of Neurosurgery, University Hospital of Münster, Münster, Germany.
| | - Reinhard Zeidler
- Helmholtz-Zentrum Munich, German Research Center for Environmental Health, Research Group Gene Vectors, Munich, Germany
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital, LMU Munich, Munich, Germany
| | | | - Guido Böning
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Astrid Gosewisch
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Walter Stummer
- Department of Neurosurgery, University Hospital of Münster, Münster, Germany
| |
Collapse
|
557
|
Zureick AH, McFadden KA, Mody R, Koschmann C. Successful treatment of a TSC2-mutant glioblastoma with everolimus. BMJ Case Rep 2019; 12:12/5/e227734. [PMID: 31154346 DOI: 10.1136/bcr-2018-227734] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
A 14-year-old boy with familial Li-Fraumeni syndrome presented with diplopia. Brain MRI revealed a right temporoparietal rim-enhancing mass. Following surgical resection and diagnosis of a gigantocellular-type glioblastoma multiforme (GBM), his family wished to avoid cytotoxic chemotherapy given the amplified risk of secondary malignancy. As such, we performed whole exome and transcriptome sequencing, which revealed germline TP53 and somatic TSC2 mutations. On completion of adjuvant radiotherapy, he was started on maintenance therapy with everolimus per recommendations from our multi-institutional brain tumour precision medicine tumour board. He has achieved a complete remission with resolution of visual symptoms and remains on everolimus therapy with concurrent electromagnetic field therapy, now 33 months from diagnosis. Our data highlight the benefit of precision medicine in children with GBM and offer insight into a targetable pathway that may be involved in similar cases.
Collapse
Affiliation(s)
- Andrew H Zureick
- University of Michigan Medical School, Michigan Medicine, Ann Arbor, Michigan, USA.,Department of Radiation Oncology, Beaumont Health System, Royal Oak, Michigan, USA
| | | | - Rajen Mody
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Carl Koschmann
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Michigan Medicine, Ann Arbor, Michigan, USA
| |
Collapse
|
558
|
Chandramohan V, Bao X, Yu X, Parker S, McDowall C, Yu YR, Healy P, Desjardins A, Gunn MD, Gromeier M, Nair SK, Pastan IH, Bigner DD. Improved efficacy against malignant brain tumors with EGFRwt/EGFRvIII targeting immunotoxin and checkpoint inhibitor combinations. J Immunother Cancer 2019; 7:142. [PMID: 31142380 PMCID: PMC6542114 DOI: 10.1186/s40425-019-0614-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 05/08/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND D2C7-IT is a novel immunotoxin (IT) targeting wild-type epidermal growth factor receptor (EGFRwt) and mutant EGFR variant III (EGFRvIII) proteins in glioblastoma. In addition to inherent tumoricidal activity, immunotoxins induce secondary immune responses through the activation of T cells. However, glioblastoma-induced immune suppression is a major obstacle to an effective and durable immunotoxin-mediated antitumor response. We hypothesized that D2C7-IT-induced immune response could be effectively augmented in combination with αCTLA-4/αPD-1/αPD-L1 therapies in murine models of glioma. METHODS To study this, we overexpressed the D2C7-IT antigen, murine EGFRvIII (dmEGFRvIII), in established glioma lines, CT-2A and SMA560. The reactivity and therapeutic efficacy of D2C7-IT against CT-2A-dmEGFRvIII and SMA560-dmEGFRvIII cells was determined by flow cytometry and in vitro cytotoxicity assays, respectively. Antitumor efficacy of D2C7-IT was examined in immunocompetent, intracranial murine glioma models and the role of T cells was assessed by CD4+ and CD8+ T cell depletion. In vivo efficacy of D2C7-IT/αCTLA-4/αPD-1 monotherapy or D2C7-IT+αCTLA-4/αPD-1 combination therapy was evaluated in subcutaneous unilateral and bilateral CT-2A-dmEGFRvIII glioma-bearing immunocompetent mice. Further, antitumor efficacy of D2C7-IT+αCTLA-4/αPD-1/αPD-L1/αTim-3/αLag-3/αCD73 combination therapy was evaluated in intracranial CT-2A-dmEGFRvIII and SMA560-dmEGFRvIII glioma-bearing mice. Pairwise differences in survival curves were assessed using the generalized Wilcoxon test. RESULTS D2C7-IT effectively killed CT-2A-dmEGFRvIII (IC50 = 0.47 ng/mL) and SMA560-dmEGFRvIII (IC50 = 1.05 ng/mL) cells in vitro. Treatment of intracranial CT-2A-dmEGFRvIII and SMA560-dmEGFRvIII tumors with D2C7-IT prolonged survival (P = 0.0188 and P = 0.0057, respectively), which was significantly reduced by the depletion of CD4+ and CD8+ T cells. To augment antitumor immune responses, we combined D2C7-IT with αCTLA-4/αPD-1 in an in vivo subcutaneous CT-2A-dmEGFRvIII model. Tumor-bearing mice exhibited complete tumor regressions (4/10 in D2C7-IT+αCTLA-4 and 5/10 in D2C7-IT+αPD-1 treatment groups), and combination therapy-induced systemic antitumor response was effective against both dmEGFRvIII-positive and dmEGFRvIII-negative CT-2A tumors. In a subcutaneous bilateral CT-2A-dmEGFRvIII model, D2C7-IT+αCTLA-4/αPD-1 combination therapies showed dramatic regression of the treated tumors and measurable regression of untreated tumors. Notably, in CT-2A-dmEGFRvIII and SMA560-dmEGFRvIII intracranial glioma models, D2C7-IT+αPD-1/αPD-L1 combinations improved survival, and in selected cases generated cures and protection against tumor re-challenge. CONCLUSIONS These data support the development of D2C7-IT and immune checkpoint blockade combinations for patients with malignant glioma.
Collapse
Affiliation(s)
- Vidyalakshmi Chandramohan
- Department of Neurosurgery and the Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Medical Sciences Research Building, Rm 181c, Box 3156, Durham, NC, 27710, USA.
| | - Xuhui Bao
- Department of Surgery, Duke University Medical Center, Durham, NC, 27710, USA
| | - Xin Yu
- Department of Neurosurgery and the Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Medical Sciences Research Building, Rm 181c, Box 3156, Durham, NC, 27710, USA
| | - Scott Parker
- Department of Neurosurgery and the Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Medical Sciences Research Building, Rm 181c, Box 3156, Durham, NC, 27710, USA
| | - Charlotte McDowall
- Department of Neurosurgery and the Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Medical Sciences Research Building, Rm 181c, Box 3156, Durham, NC, 27710, USA
| | - Yen-Rei Yu
- Department of Medicine, Duke University Medical Center, Durham, NC, 27710, USA
| | - Patrick Healy
- Duke Cancer Institute Biostatistics, Duke University Medical Center, Durham, NC, 27710, USA
| | - Annick Desjardins
- Department of Neurosurgery and the Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Medical Sciences Research Building, Rm 181c, Box 3156, Durham, NC, 27710, USA
| | - Michael D Gunn
- Department of Medicine, Duke University Medical Center, Durham, NC, 27710, USA
| | - Matthias Gromeier
- Department of Neurosurgery and the Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Medical Sciences Research Building, Rm 181c, Box 3156, Durham, NC, 27710, USA
| | - Smita K Nair
- Department of Surgery, Duke University Medical Center, Durham, NC, 27710, USA
| | - Ira H Pastan
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Darell D Bigner
- Department of Neurosurgery and the Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Medical Sciences Research Building, Rm 181c, Box 3156, Durham, NC, 27710, USA
| |
Collapse
|
559
|
Upregulation of the NLRC4 inflammasome contributes to poor prognosis in glioma patients. Sci Rep 2019; 9:7895. [PMID: 31133717 PMCID: PMC6536517 DOI: 10.1038/s41598-019-44261-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 05/08/2019] [Indexed: 02/06/2023] Open
Abstract
Inflammation in tumor microenvironments is implicated in the pathogenesis of tumor development. In particular, inflammasomes, which modulate innate immune functions, are linked to tumor growth and anticancer responses. However, the role of the NLRC4 inflammasome in gliomas remains unclear. Here, we investigated whether the upregulation of the NLRC4 inflammasome is associated with the clinical prognosis of gliomas. We analyzed the protein expression and localization of NLRC4 in glioma tissues from 11 patients by immunohistochemistry. We examined the interaction between the expression of NLRC4 and clinical prognosis via a Kaplan-Meier survival analysis. The level of NLRC4 protein was increased in brain tissues, specifically, in astrocytes, from glioma patients. NLRC4 expression was associated with a poor prognosis in glioma patients, and the upregulation of NLRC4 in astrocytomas was associated with poor survival. Furthermore, hierarchical clustering of data from the Cancer Genome Atlas dataset showed that NLRC4 was highly expressed in gliomas relative to that in a normal healthy group. Our results suggest that the upregulation of the NLRC4 inflammasome contributes to a poor prognosis for gliomas and presents a potential therapeutic target and diagnostic marker.
Collapse
|
560
|
Carpenter CD, Alnahhas I, Gonzalez J, Giglio P, Puduvalli VK. Changing paradigms for targeted therapies against diffuse infiltrative gliomas: tackling a moving target. Expert Rev Neurother 2019; 19:663-677. [PMID: 31106606 DOI: 10.1080/14737175.2019.1621169] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Introduction: Gliomas are highly heterogeneous primary brain tumors which result in a disproportionately high degree of morbidity and mortality despite their locoregional occurrence. Advances in the understanding of the biological makeup of these malignancies have yielded a number of potential tumor-driving pathways which have been identified as rational targets for therapy. However, early trials of agents that target these pathways have uniformly failed to yield improvement in outcomes in patients with malignant gliomas. Areas covered: This review provides an overview of the most common biological features of gliomas and the strategies to target the same; in addition, the current status of immunotherapy and biological therapies are outlined and the future directions to tackle the challenges of therapy for gliomas are examined. Expert opinion: The limitations of current treatments are attributed to the inability of most of these agents to cross the blood-brain barrier and to the intrinsic heterogeneity of the tumors that result in treatment resistance. The recent emergence of immune-mediated and biological therapies and of agents that target metabolic pathways in gliomas have provided strategies that may overcome tumor heterogeneity and ongoing trials of such agents are anticipated to yield improved outcomes.
Collapse
Affiliation(s)
- Candice D Carpenter
- a Department of Neurosurgery , The Ohio State University Wexner Medical Center , Columbus , OH , USA
| | - Iyad Alnahhas
- b Division of Neurooncology , The Ohio State University Wexner Medical Center , Columbus , OH , USA
| | - Javier Gonzalez
- a Department of Neurosurgery , The Ohio State University Wexner Medical Center , Columbus , OH , USA.,b Division of Neurooncology , The Ohio State University Wexner Medical Center , Columbus , OH , USA
| | - Pierre Giglio
- a Department of Neurosurgery , The Ohio State University Wexner Medical Center , Columbus , OH , USA.,b Division of Neurooncology , The Ohio State University Wexner Medical Center , Columbus , OH , USA
| | - Vinay K Puduvalli
- a Department of Neurosurgery , The Ohio State University Wexner Medical Center , Columbus , OH , USA.,b Division of Neurooncology , The Ohio State University Wexner Medical Center , Columbus , OH , USA
| |
Collapse
|
561
|
Connock M, Auguste P, Dussart C, Guyotat J, Armoiry X. Cost-effectiveness of tumor-treating fields added to maintenance temozolomide in patients with glioblastoma: an updated evaluation using a partitioned survival model. J Neurooncol 2019; 143:605-611. [PMID: 31127507 DOI: 10.1007/s11060-019-03197-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 05/13/2019] [Accepted: 05/16/2019] [Indexed: 01/13/2023]
Abstract
PURPOSE A first cost-effectiveness analysis has raised a strong concern regarding the cost of tumor treatment fields (TTF) added to maintenance temozolomide for patients with glioblastoma. This evaluation was based on effectiveness outcomes from an interim analysis of the pivotal trial, moreover it used a "standard" Markov model. Our objective was to update the cost-effectiveness evaluation using the more flexible potential of the "partitioned survival" model design and using the latest effectiveness data. METHODS We developed the model with three mutually exclusive health states: stable disease, progressive disease, and dead. Good fit parametric models were developed for overall survival and progression free survival and these generated clinically plausible extrapolations beyond the observed data. We adopted the perspective of the French national health insurance and used a 20-year time horizon. Results were expressed as cost/life-years (LY) gained (LYG). RESULTS The base case model generated incremental benefit of 0.604 LY at a cost of €453,848 which, after 4% annual discounting of benefits and costs, yielded an incremental cost effectiveness ratio (ICER) of €510,273/LYG. Using sensitivity analyses and bootstrapping methods results were found to be relatively robust and were only sensitive to TTF device costs and the modelling of overall survival. To achieve an ICER below €100,000/LYG would require a reduction in TTF device cost of approximately 85%. CONCLUSIONS Using a different type of model and updated survival outcomes, our results show TTF remains an intervention that is not cost-effective, which greatly restrains its diffusion to potentially eligible patients.
Collapse
Affiliation(s)
- Martin Connock
- Warwick Medical School, University of Warwick, Gibbet Hill Road, Coventry, CV47AL, UK
| | - Peter Auguste
- Warwick Medical School, University of Warwick, Gibbet Hill Road, Coventry, CV47AL, UK
| | - Claude Dussart
- University of Lyon, EA 4129 P2S (Parcours, Santé, Systémique), Lyon, France
| | - Jacques Guyotat
- Pierre Wertheimer Hospital, Neurosurgery Department, Lyon, France
| | - Xavier Armoiry
- Warwick Medical School, University of Warwick, Gibbet Hill Road, Coventry, CV47AL, UK.
- Pharmacy Department, Edouard Herriot Hospital, Lyon, France.
- Public Health Department, School of Pharmacy (ISPB)/ UMR CNRS MATEIS, Claude Bernard University Lyon 1, University of Lyon, 8 Avenue Rockefeller, 69008, Lyon, France.
| |
Collapse
|
562
|
Overall Survival in Malignant Glioma Is Significantly Prolonged by Neurosurgical Delivery of Etoposide and Temozolomide from a Thermo-Responsive Biodegradable Paste. Clin Cancer Res 2019; 25:5094-5106. [DOI: 10.1158/1078-0432.ccr-18-3850] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 03/11/2019] [Accepted: 05/17/2019] [Indexed: 11/16/2022]
|
563
|
Farooqi A, Yang J, Sharin V, Ezhilarasan R, Danussi C, Alvarez C, Dharmaiah S, Irvin D, Huse J, Sulman EP. Identification of patient-derived glioblastoma stem cell (GSC) lines with the alternative lengthening of telomeres phenotype. Acta Neuropathol Commun 2019; 7:76. [PMID: 31097032 PMCID: PMC6521362 DOI: 10.1186/s40478-019-0732-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 05/03/2019] [Indexed: 11/28/2022] Open
|
564
|
Yin AA, He YL, Etcheverry A, Liu YH, Aubry M, Barnholtz-Sloan J, Liu BL, Mosser J, Lu ZF, Zhang X. Novel predictive epigenetic signature for temozolomide in non-G-CIMP glioblastomas. Clin Epigenetics 2019; 11:76. [PMID: 31088577 PMCID: PMC6515684 DOI: 10.1186/s13148-019-0670-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 04/25/2019] [Indexed: 11/25/2022] Open
Abstract
Objective To identify novel epigenetic signatures that could provide predictive information that is complementary to promoter methylation status of the O-6-methylguanine-DNA methyltransferase (MGMT) gene for predicting temozolomide (TMZ) response, among glioblastomas (GBMs) without glioma-CpGs island methylator phenotype (G-CIMP) Methods Different cohorts of primary non-G-CIMP GBMs with genome-wide DNA methylation microarray data were included for discovery and validation of a multimarker signature, combined using a RISK score model. Different statistical analyses and functional experiments were performed for clinical and biological validation. Results By employing discovery cohorts with radiotherapy (RT) and TMZ versus RT alone and a strict multistep selection strategy, we identified seven CpGs, each of which was significantly correlated with overall survival (OS) of non-G-CIMP GBMs with RT/TMZ, independent of age, MGMT promoter methylation status, and other identified CpGs. A RISK score signature of the 7 CpGs was developed and validated to distinguish non-G-CIMP GBMs with differential survival outcomes to RT/TMZ, but not to RT alone. The interaction analyses also showed differential outcomes to RT/TMZ versus RT alone within the RISK score-based subgroups. The signature could also improve the risk classification by age and MGMT promoter methylation status. Functional experiments showed that HSBP2 appeared to be epigenetically regulated by one identified CpG and was associated with TMZ resistance, but it was not associated with cell proliferation or apoptosis in GBM cell lines. The predictive value of the single CpG methylation of HSBP2 by pyrosequencing was observed in a local cohort of isocitrate dehydrogenase 1 (IDH1) R132H wild-type GBMs. Conclusions This novel epigenetic signature might be a promising predictive (but not a general prognostic) biomarker and be helpful for refining the MGMT-based guiding approach to TMZ usage in non-G-CIMP GBMs. Electronic supplementary material The online version of this article (10.1186/s13148-019-0670-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- An-An Yin
- Department of Neurosurgery, Xijing Institute of Clinical Neuroscience, Xijing Hospital, Air Force Medical University, West Road, No. 169 Xi'an, Changle, 710032, Shaanxi Province, China.,State Key Laboratory of Cancer Biology, Department of Pharmacogenomics, School of Pharmacy, Air Force Medical University, Xi'an, Shaanxi Province, China.,Department of Neurosurgery, the 960th Hospital of the People's Liberation Army, Taian, Shandong Province, China
| | - Ya-Long He
- Department of Neurosurgery, Xijing Institute of Clinical Neuroscience, Xijing Hospital, Air Force Medical University, West Road, No. 169 Xi'an, Changle, 710032, Shaanxi Province, China
| | - Amandine Etcheverry
- CNRS, UMR 6290, Institut de Génétique et Développement de Rennes (IGdR), 35043, Rennes, France.,UEB, UMS 3480 Biosit, Faculté de Médecine, Université Rennes 1, 35043, Rennes, France.,CHU Rennes, Service de Génétique Moléculaire et Génomique, 35033, Rennes, France
| | - Yu-He Liu
- Department of Neurosurgery, the 960th Hospital of the People's Liberation Army, Taian, Shandong Province, China
| | - Marc Aubry
- CNRS, UMR 6290, Institut de Génétique et Développement de Rennes (IGdR), 35043, Rennes, France.,CHU Rennes, Service de Génétique Moléculaire et Génomique, 35033, Rennes, France
| | - Jill Barnholtz-Sloan
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - Bo-Lin Liu
- Department of Neurosurgery, Tangdu Hospital, Air Force Military Medical University, Xi'an, Shaanxi Province, China
| | - Jean Mosser
- CNRS, UMR 6290, Institut de Génétique et Développement de Rennes (IGdR), 35043, Rennes, France.,UEB, UMS 3480 Biosit, Faculté de Médecine, Université Rennes 1, 35043, Rennes, France.,CHU Rennes, Service de Génétique Moléculaire et Génomique, 35033, Rennes, France.,Plate-forme Génomique Santé Biosit, Université Rennes 1, 35043, Rennes, France
| | - Zi-Fan Lu
- State Key Laboratory of Cancer Biology, Department of Pharmacogenomics, School of Pharmacy, Air Force Medical University, Xi'an, Shaanxi Province, China.
| | - Xiang Zhang
- Department of Neurosurgery, Xijing Institute of Clinical Neuroscience, Xijing Hospital, Air Force Medical University, West Road, No. 169 Xi'an, Changle, 710032, Shaanxi Province, China.
| |
Collapse
|
565
|
Tumour-treating fields (TTFields): Investigations on the mechanism of action by electromagnetic exposure of cells in telophase/cytokinesis. Sci Rep 2019; 9:7362. [PMID: 31089145 PMCID: PMC6517379 DOI: 10.1038/s41598-019-43621-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 04/26/2019] [Indexed: 11/25/2022] Open
Abstract
Tumour-treating fields (TTFields) use alternating electric fields which interfere with dividing cells, thereby reducing tumour growth. Previous reports suggest that electrical forces on cell structure proteins interfered with the chromosome separation during mitosis and induced apoptosis. In the present report we evaluate electromagnetic exposure of cells in telophase/cytokinesis in order to further analyse the mechanism of action on cells. We performed numerical electromagnetic simulations to analyse the field distribution in a cell during different mitotic phases. Based thereon, we developed an electric lumped element model of the mitotic cell. Both the electromagnetic simulation and the lumped element model predict a local increase of the specific absorption rate (SAR) as a measure of the electromagnetically induced power absorption density at the mitotic furrow which may help to explain the anti-proliferative effect. In accordance with other reports, cell culture experiments confirmed that TTFields reduce the proliferation of different glioma cell lines in a field strength- and frequency-dependent manner. Furthermore, we found an additional dependence on the commutation time of the electrical fields. The report gives new insights into TTFields’ anti-proliferative effect on tumours, which could help to improve future TTFields application systems.
Collapse
|
566
|
Arevalo OD, Soto C, Rabiei P, Kamali A, Ballester LY, Esquenazi Y, Zhu JJ, Riascos RF. Assessment of Glioblastoma Response in the Era of Bevacizumab: Longstanding and Emergent Challenges in the Imaging Evaluation of Pseudoresponse. Front Neurol 2019; 10:460. [PMID: 31133966 PMCID: PMC6514158 DOI: 10.3389/fneur.2019.00460] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 04/16/2019] [Indexed: 12/17/2022] Open
Abstract
Glioblastoma is the deadliest primary malignant brain neoplasm, and despite the availability of many treatment options, its prognosis remains somber. Enhancement detected by magnetic resonance imaging (MRI) was considered the best imaging marker of tumor activity in glioblastoma for decades. However, its role as a surrogate marker of tumor viability has changed with the appearance of new treatment regimens and imaging modalities. The antiangiogenic therapy created an inflection point in the imaging assessment of glioblastoma response in clinical trials and clinical practice. Although BEV led to the improvement of enhancement, it did not necessarily mean tumor response. The decrease in the enhancement intensity represents a change in the permeability properties of the blood brain barrier, and presumably, the switch of the tumor growth pattern to an infiltrative non-enhancing phenotype. New imaging techniques for the assessment of cellularity, blood flow hemodynamics, and biochemistry have emerged to overcome this hurdle; nevertheless, designing tools to assess tumor response more accurately, and in so doing, improve the assessment of response to standard of care (SOC) therapies and to novel therapies, remains challenging.
Collapse
Affiliation(s)
- Octavio D Arevalo
- Department of Diagnostic and Interventional Radiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Carolina Soto
- Faculty of Medicine, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Pejman Rabiei
- Department of Diagnostic and Interventional Radiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Arash Kamali
- Department of Diagnostic and Interventional Radiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Leomar Y Ballester
- Department of Pathology and Laboratory Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Yoshua Esquenazi
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Jay-Jiguang Zhu
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Roy Francisco Riascos
- Department of Diagnostic and Interventional Radiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
567
|
The Role of Platelets in Cancer Pathophysiology: Focus on Malignant Glioma. Cancers (Basel) 2019; 11:cancers11040569. [PMID: 31013620 PMCID: PMC6521321 DOI: 10.3390/cancers11040569] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 04/11/2019] [Accepted: 04/18/2019] [Indexed: 12/22/2022] Open
Abstract
The link between thrombocytosis and malignancy has been well known for many years and its associations with worse outcomes have been reported mainly for solid tumors. Besides measuring platelet count, it has become popular to assess platelet function in the context of malignant diseases during the last decade. Malignant gliomas differ tremendously from malignancies outside the central nervous system because they virtually never form distant metastases. This review summarizes the current understanding of the platelet-immune cell communication and its potential role in glioma resistance and progression. Particularly, we focus on platelet-derived proinflammatory modulators, such as sphingosine-1-phosphate (S1P). The multifaceted interaction with immune cells puts the platelet into an interesting perspective regarding the recent advances in immunotherapeutic approaches in malignant glioma.
Collapse
|
568
|
Carvalho JADV, Barbosa CCDL, Feher O, Maldaun MVC, Camargo VPD, Moraes FY, Marta GN. Systemic dissemination of glioblastoma: literature review. ACTA ACUST UNITED AC 2019; 65:460-468. [PMID: 30994848 DOI: 10.1590/1806-9282.65.3.460] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 08/05/2018] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Glioblastoma (GBM) is the most frequent primary malignant tumor from the central nervous system in adults. However, the presence of systemic metastasis is an extremely rare event. The objective of this study was to review the literature, evaluating the possible biological mechanisms related to the occurrence of systemic metastasis in patients diagnosed with GBM. RESULTS The mechanisms that may be related to GBM systemic dissemination are the blood-brain barrier breach, often seen in GBM cases, by the tumor itself or by surgical procedures, gaining access to blood and lymphatic vessels, associated with the acquisition of mesenchymal features of invasiveness, resistance to the immune mechanisms of defense and hostile environment through quiescence. CONCLUSIONS Tumor cells must overcome many obstacles until the development of systemic metastasis. The physiologic mechanisms are not completely clear. Although not fully understood, the pathophysiological understanding of the mechanisms that may be associated with the systemic spread is salutary for a global understanding of the disease. In addition, this knowledge may be used as a basis for a therapy to be performed in patients diagnosed with GBM distant metastasis.
Collapse
Affiliation(s)
| | - Caroline Chaul de Lima Barbosa
- Department of Radiology and Oncology - Clinical Oncology Unit; Faculdade de Medicina da Universidade de São Paulo - Instituto do Câncer do Estado de São Paulo (Icesp); Sao Paulo, Brasil.,Department of Clinical Oncology - Hospital Sírio-Libanês; Sao Paulo, Brasil
| | - Olavo Feher
- Department of Radiology and Oncology - Clinical Oncology Unit; Faculdade de Medicina da Universidade de São Paulo - Instituto do Câncer do Estado de São Paulo (Icesp); Sao Paulo, Brasil.,Department of Clinical Oncology - Hospital Sírio-Libanês; Sao Paulo, Brasil
| | - Marcos Vinicius Calfat Maldaun
- Division of Neurosurgery, Hospital Sírio-Libanês; Sao Paulo, Brasil.,Division of Neurosurgery, Santa Paula Hospital, São Paulo, SP, Brasil
| | - Veridiana Pires de Camargo
- Department of Radiology and Oncology - Clinical Oncology Unit; Faculdade de Medicina da Universidade de São Paulo - Instituto do Câncer do Estado de São Paulo (Icesp); Sao Paulo, Brasil.,Department of Clinical Oncology - Hospital Sírio-Libanês; Sao Paulo, Brasil
| | - Fabio Y Moraes
- Department of Oncology, Division of Radiation Oncology, Queen's University - Kingston Health Science Centre, Kingston, ON, Canada.,Department of Radiation Oncology, Hospital Sírio-Libanês, Sao Paulo, Brasil
| | - Gustavo Nader Marta
- Department of Radiology and Oncology, Division of Radiation Oncology, Instituto do Câncer do Estado de São Paulo (Icesp), Faculdade de Medicina da Universidade de São Paulo, Sao Paulo, Brasil.,Department of Radiation Oncology, Hospital Sírio-Libanês, Sao Paulo, Brasil
| |
Collapse
|
569
|
Idbaih A, Canney M, Belin L, Desseaux C, Vignot A, Bouchoux G, Asquier N, Law-Ye B, Leclercq D, Bissery A, De Rycke Y, Trosch C, Capelle L, Sanson M, Hoang-Xuan K, Dehais C, Houillier C, Laigle-Donadey F, Mathon B, André A, Lafon C, Chapelon JY, Delattre JY, Carpentier A. Safety and Feasibility of Repeated and Transient Blood-Brain Barrier Disruption by Pulsed Ultrasound in Patients with Recurrent Glioblastoma. Clin Cancer Res 2019; 25:3793-3801. [PMID: 30890548 DOI: 10.1158/1078-0432.ccr-18-3643] [Citation(s) in RCA: 246] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 01/08/2019] [Accepted: 03/14/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE The blood-brain barrier (BBB) limits the efficacy of drug therapies for glioblastoma (GBM). Preclinical data indicate that low-intensity pulsed ultrasound (LIPU) can transiently disrupt the BBB and increase intracerebral drug concentrations. PATIENTS AND METHODS A first-in-man, single-arm, single-center trial (NCT02253212) was initiated to investigate the transient disruption of the BBB in patients with recurrent GBM. Patients were implanted with a 1-MHz, 11.5-mm diameter cranial ultrasound device (SonoCloud-1, CarThera). The device was activated monthly to transiently disrupt the BBB before intravenous carboplatin chemotherapy. RESULTS Between 2014 and 2016, 21 patients were registered for the study and implanted with the SonoCloud-1; 19 patients received at least one sonication. In 65 ultrasound sessions, BBB disruption was visible on T1w MRI for 52 sonications. Treatment-related adverse events observed were transient and manageable: a transient edema at H1 and at D15. No carboplatin-related neurotoxicity was observed. Patients with no or poor BBB disruption (n = 8) visible on MRI had a median progression-free survival (PFS) of 2.73 months, and a median overall survival (OS) of 8.64 months. Patients with clear BBB disruption (n = 11) had a median PFS of 4.11 months, and a median OS of 12.94 months. CONCLUSIONS SonoCloud-1 treatments were well tolerated and may increase the effectiveness of systemic drug therapies, such as carboplatin, in the brain without inducing neurotoxicity.See related commentary by Sonabend and Stupp, p. 3750.
Collapse
Affiliation(s)
- Ahmed Idbaih
- Institut du Cerveau et de la Moelle épinière, ICM, AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, Sorbonne Université, INSERM, Paris, France
| | - Michael Canney
- CarThera, Institut du Cerveau et de la Moelle épinière (ICM), Paris, France
| | - Lisa Belin
- Département Biostatistique Santé Publique et Information Médicale, Sorbonne Université, AP-HP, Hôpital Pitié-Salpêtrière, INSERM, Paris, France
| | - Carole Desseaux
- CarThera, Institut du Cerveau et de la Moelle épinière (ICM), Paris, France
| | - Alexandre Vignot
- CarThera, Institut du Cerveau et de la Moelle épinière (ICM), Paris, France
| | - Guillaume Bouchoux
- CarThera, Institut du Cerveau et de la Moelle épinière (ICM), Paris, France
| | - Nicolas Asquier
- CarThera, Institut du Cerveau et de la Moelle épinière (ICM), Paris, France.,LabTAU, INSERM, Centre Léon Bérard, Université Lyon 1, Univ Lyon, Lyon, France
| | - Bruno Law-Ye
- AP-HP, Hôpitaux Universitaires La Pitié-Salpêtrière-Charles Foix, Paris, France
| | - Delphine Leclercq
- AP-HP, Hôpitaux Universitaires La Pitié-Salpêtrière-Charles Foix, Paris, France
| | - Anne Bissery
- Clinical Research Unit, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpitaux Universitaires La Pitiè-Salpêtrière, Paris, France
| | - Yann De Rycke
- Clinical Research Unit, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpitaux Universitaires La Pitiè-Salpêtrière, Paris, France
| | - Clementine Trosch
- Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpitaux Universitaires La Pitiè-Salpêtrière, Service de Neurochirurgie, Paris, France
| | - Laurent Capelle
- Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpitaux Universitaires La Pitiè-Salpêtrière, Service de Neurochirurgie, Paris, France
| | - Marc Sanson
- Institut du Cerveau et de la Moelle épinière, ICM, AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, Sorbonne Université, INSERM, Paris, France
| | - Khe Hoang-Xuan
- Institut du Cerveau et de la Moelle épinière, ICM, AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, Sorbonne Université, INSERM, Paris, France
| | - Caroline Dehais
- Institut du Cerveau et de la Moelle épinière, ICM, AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, Sorbonne Université, INSERM, Paris, France
| | - Caroline Houillier
- Institut du Cerveau et de la Moelle épinière, ICM, AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, Sorbonne Université, INSERM, Paris, France
| | - Florence Laigle-Donadey
- Institut du Cerveau et de la Moelle épinière, ICM, AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, Sorbonne Université, INSERM, Paris, France
| | - Bertrand Mathon
- Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpitaux Universitaires La Pitiè-Salpêtrière, Service de Neurochirurgie, Paris, France
| | - Arthur André
- Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpitaux Universitaires La Pitiè-Salpêtrière, Service de Neurochirurgie, Paris, France
| | - Cyril Lafon
- LabTAU, INSERM, Centre Léon Bérard, Université Lyon 1, Univ Lyon, Lyon, France
| | - Jean-Yves Chapelon
- LabTAU, INSERM, Centre Léon Bérard, Université Lyon 1, Univ Lyon, Lyon, France
| | - Jean-Yves Delattre
- Institut du Cerveau et de la Moelle épinière, ICM, AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, Sorbonne Université, INSERM, Paris, France
| | - Alexandre Carpentier
- Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpitaux Universitaires La Pitiè-Salpêtrière, Service de Neurochirurgie, Paris, France. .,Sorbonne Université, UPMC Univ Paris 06, Paris, France
| |
Collapse
|
570
|
Marino A, Almici E, Migliorin S, Tapeinos C, Battaglini M, Cappello V, Marchetti M, de Vito G, Cicchi R, Pavone FS, Ciofani G. Piezoelectric barium titanate nanostimulators for the treatment of glioblastoma multiforme. J Colloid Interface Sci 2019; 538:449-461. [PMID: 30537658 PMCID: PMC6697537 DOI: 10.1016/j.jcis.2018.12.014] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 11/28/2018] [Accepted: 12/03/2018] [Indexed: 12/31/2022]
Abstract
Major obstacles to the successful treatment of gliolastoma multiforme are mostly related to the acquired resistance to chemotherapy drugs and, after surgery, to the cancer recurrence in correspondence of residual microscopic foci. As innovative anticancer approach, low-intensity electric stimulation represents a physical treatment able to reduce multidrug resistance of cancer and to induce remarkable anti-proliferative effects by interfering with Ca2+ and K+ homeostasis and by affecting the organization of the mitotic spindles. However, to preserve healthy cells, it is utterly important to direct the electric stimuli only to malignant cells. In this work, we propose a nanotechnological approach based on ultrasound-sensitive piezoelectric nanoparticles to remotely deliver electric stimulations to glioblastoma cells. Barium titanate nanoparticles (BTNPs) have been functionalized with an antibody against the transferrin receptor (TfR) in order to obtain the dual targeting of blood-brain barrier and of glioblastoma cells. The remote ultrasound-mediated piezo-stimulation allowed to significantly reduce in vitro the proliferation of glioblastoma cells and, when combined with a sub-toxic concentration of temozolomide, induced an increased sensitivity to the chemotherapy treatment and remarkable anti-proliferative and pro-apoptotic effects.
Collapse
Affiliation(s)
- Attilio Marino
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, 56025 Pontedera, Italy.
| | - Enrico Almici
- Politecnico di Torino, Department of Mechanical and Aerospace Engineering, Corso Duca degli Abruzzi 24, 10129 Torino, Italy
| | - Simone Migliorin
- Politecnico di Torino, Department of Mechanical and Aerospace Engineering, Corso Duca degli Abruzzi 24, 10129 Torino, Italy
| | - Christos Tapeinos
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, 56025 Pontedera, Italy
| | - Matteo Battaglini
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, 56025 Pontedera, Italy; Scuola Superiore Sant'Anna, The Biorobotics Institute, Viale Rinaldo Piaggio 34, 56025 Pontedera, Italy
| | - Valentina Cappello
- Istituto Italiano di Tecnologia, Center for Nanotechnology Innovation, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Marco Marchetti
- European Laboratory for Nonlinear Spectroscopy (LENS), Via Nello Carrara 1, 50019 Sesto Fiorentino, Italy; Università di Firenze, Department of Physics and Astronomy, Via Giovanni Sansone 1, 50019 Sesto Fiorentino, Italy
| | - Giuseppe de Vito
- European Laboratory for Nonlinear Spectroscopy (LENS), Via Nello Carrara 1, 50019 Sesto Fiorentino, Italy; National Institute of Optics, National Research Council (INO-CNR), Largo Enrico Fermi 6, 50125 Firenze, Italy
| | - Riccardo Cicchi
- European Laboratory for Nonlinear Spectroscopy (LENS), Via Nello Carrara 1, 50019 Sesto Fiorentino, Italy; National Institute of Optics, National Research Council (INO-CNR), Largo Enrico Fermi 6, 50125 Firenze, Italy
| | - Francesco Saverio Pavone
- European Laboratory for Nonlinear Spectroscopy (LENS), Via Nello Carrara 1, 50019 Sesto Fiorentino, Italy; Università di Firenze, Department of Physics and Astronomy, Via Giovanni Sansone 1, 50019 Sesto Fiorentino, Italy; National Institute of Optics, National Research Council (INO-CNR), Largo Enrico Fermi 6, 50125 Firenze, Italy
| | - Gianni Ciofani
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, 56025 Pontedera, Italy; Politecnico di Torino, Department of Mechanical and Aerospace Engineering, Corso Duca degli Abruzzi 24, 10129 Torino, Italy.
| |
Collapse
|
571
|
Evaluating the decisions of glioma patients regarding clinical trial participation: a retrospective single provider review. Med Oncol 2019; 36:34. [DOI: 10.1007/s12032-019-1259-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 02/26/2019] [Indexed: 01/19/2023]
|
572
|
High density is a property of slow-cycling and treatment-resistant human glioblastoma cells. Exp Cell Res 2019; 378:76-86. [PMID: 30844389 DOI: 10.1016/j.yexcr.2019.03.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 03/01/2019] [Accepted: 03/02/2019] [Indexed: 12/16/2022]
Abstract
Slow-cycling and treatment-resistant cancer cells escape therapy, providing a rationale for regrowth and recurrence in patients. Much interest has focused on identifying the properties of slow-cycling tumor cells in glioblastoma (GBM), the most common and lethal primary brain tumor. Despite aggressive ionizing radiation (IR) and treatment with the alkylating agent temozolomide (TMZ), GBM patients invariably relapse and ultimately succumb to the disease. In patient biopsies, we demonstrated that GBM cells expressing the proliferation markers Ki67 and MCM2 displayed a larger cell volume compared to rare slow-cycling tumor cells. In optimized density gradients, we isolated a minor fraction of slow-cycling GBM cells in patient biopsies and tumorsphere cultures. Transcriptional profiling, self-renewal, and tumorigenicity assays reflected the slow-cycling state of high-density GBM cells (HDGCs) compared to the tumor bulk of low-density GBM cells (LDGCs). Slow-cycling HDGCs enriched for stem cell antigens proliferated a few days after isolation to generate LDGCs. Both in vitro and in vivo, we demonstrated that HDGCs show increased treatment-resistance to IR and TMZ treatment compared to LDGCs. In conclusion, density gradients represent a non-marker based approach to isolate slow-cycling and treatment-resistant GBM cells across GBM subgroups.
Collapse
|
573
|
Caramia M, Sforna L, Franciolini F, Catacuzzeno L. The Volume-Regulated Anion Channel in Glioblastoma. Cancers (Basel) 2019; 11:cancers11030307. [PMID: 30841564 PMCID: PMC6468384 DOI: 10.3390/cancers11030307] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 02/22/2019] [Accepted: 02/26/2019] [Indexed: 01/02/2023] Open
Abstract
Malignancy of glioblastoma multiforme (GBM), the most common and aggressive form of human brain tumor, strongly depends on its enhanced cell invasion and death evasion which make surgery and accompanying therapies highly ineffective. Several ion channels that regulate membrane potential, cytosolic Ca2+ concentration and cell volume in GBM cells play significant roles in sustaining these processes. Among them, the volume-regulated anion channel (VRAC), which mediates the swelling-activated chloride current (IClswell) and is highly expressed in GBM cells, arguably plays a major role. VRAC is primarily involved in reestablishing the original cell volume that may be lost under several physiopathological conditions, but also in sustaining the shape and cell volume changes needed for cell migration and proliferation. While experimentally VRAC is activated by exposing cells to hypotonic solutions that cause the increase of cell volume, in vivo it is thought to be controlled by several different stimuli and modulators. In this review we focus on our recent work showing that two conditions normally occurring in pathological GBM tissues, namely high serum levels and severe hypoxia, were both able to activate VRAC, and their activation was found to promote cell migration and resistance to cell death, both features enhancing GBM malignancy. Also, the fact that the signal transduction pathway leading to VRAC activation appears to involve GBM specific intracellular components, such as diacylglicerol kinase and phosphatidic acid, reportedly not involved in the activation of VRAC in healthy tissues, is a relevant finding. Based on these observations and the impact of VRAC in the physiopathology of GBM, targeting this channel or its intracellular regulators may represent an effective strategy to contrast this lethal tumor.
Collapse
Affiliation(s)
- Martino Caramia
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia 06123, Italy.
| | - Luigi Sforna
- Department of Experimental Medicine, University of Perugia, Perugia 06132, Italy.
| | - Fabio Franciolini
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia 06123, Italy.
| | - Luigi Catacuzzeno
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia 06123, Italy.
| |
Collapse
|
574
|
Huang CH, Lei KF, Tsang NM. Apoptosis and cell cycle arrest of hepatocellular carcinoma spheroids treated by an alternating electric field. Biotechnol Prog 2019; 35:e2787. [DOI: 10.1002/btpr.2787] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 12/17/2018] [Accepted: 02/05/2019] [Indexed: 12/22/2022]
Affiliation(s)
- Chun-Hao Huang
- Program in Biomedical Engineering; College of Engineering, Chang Gung University; Taoyuan Taiwan
| | - Kin Fong Lei
- Graduate Institute of Biomedical Engineering, Chang Gung University; Taoyuan Taiwan
- Department of Radiation Oncology; Chang Gung Memorial Hospital; Linkou Taiwan
| | - Ngan-Ming Tsang
- Department of Radiation Oncology; Chang Gung Memorial Hospital; Linkou Taiwan
- Department of Traditional Chinese Medicine; Chang Gung University; Taoyuan Taiwan
| |
Collapse
|
575
|
|
576
|
Chukwueke UN, Wen PY. Use of the Response Assessment in Neuro-Oncology (RANO) criteria in clinical trials and clinical practice. CNS Oncol 2019; 8:CNS28. [PMID: 30806082 PMCID: PMC6499019 DOI: 10.2217/cns-2018-0007] [Citation(s) in RCA: 163] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 08/15/2018] [Indexed: 11/21/2022] Open
Affiliation(s)
- Ugonma N Chukwueke
- Department of Medical Oncology, Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Department of Neurology, Harvard Medical School, 450 Brookline Avenue, Boston, MA, 02215, USA
| | - Patrick Y Wen
- Department of Medical Oncology, Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Department of Neurology, Harvard Medical School, 450 Brookline Avenue, Boston, MA, 02215, USA
| |
Collapse
|
577
|
Alieva M, Leidgens V, Riemenschneider MJ, Klein CA, Hau P, van Rheenen J. Intravital imaging of glioma border morphology reveals distinctive cellular dynamics and contribution to tumor cell invasion. Sci Rep 2019; 9:2054. [PMID: 30765850 PMCID: PMC6375955 DOI: 10.1038/s41598-019-38625-4] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 12/18/2018] [Indexed: 01/08/2023] Open
Abstract
The pathogenesis of glioblastoma (GBM) is characterized by highly invasive behavior allowing dissemination and progression. A conclusive image of the invasive process is not available. The aim of this work was to study invasion dynamics in GBM using an innovative in vivo imaging approach. Primary brain tumor initiating cell lines from IDH-wild type GBM stably expressing H2B-Dendra2 were implanted orthotopically in the brains of SCID mice. Using high-resolution time-lapse intravital imaging, tumor cell migration in the tumor core, border and invasive front was recorded. Tumor cell dynamics at different border configurations were analyzed and multivariate linear modelling of tumor cell spreading was performed. We found tumor border configurations, recapitulating human tumor border morphologies. Not only tumor borders but also the tumor core was composed of highly dynamic cells, with no clear correlation to the ability to spread into the brain. Two types of border configurations contributed to tumor cell spreading through distinct invasion patterns: an invasive margin that executes slow but directed invasion, and a diffuse infiltration margin with fast but less directed movement. By providing a more detailed view on glioma invasion patterns, our study may improve accuracy of prognosis and serve as a basis for personalized therapeutic approaches.
Collapse
Affiliation(s)
- Maria Alieva
- Hubrecht Institute-KNAW & University Medical Center Utrecht, Uppsalalaan 8, 3584 CT, Utrecht, The Netherlands.
- Prinses Máxima Center for Pediatric Oncology, Uppsalalaan 8, 3584CT, Utrecht, The Netherlands.
| | - Verena Leidgens
- Hubrecht Institute-KNAW & University Medical Center Utrecht, Uppsalalaan 8, 3584 CT, Utrecht, The Netherlands
- Department of Neurology and Wilhelm Sander-NeuroOncology Unit, University Hospital Regensburg, Regensburg, Germany
| | | | - Christoph A Klein
- Department of Experimental Medicine, University of Regensburg, Regensburg, Germany
| | - Peter Hau
- Department of Neurology and Wilhelm Sander-NeuroOncology Unit, University Hospital Regensburg, Regensburg, Germany.
| | - Jacco van Rheenen
- Hubrecht Institute-KNAW & University Medical Center Utrecht, Uppsalalaan 8, 3584 CT, Utrecht, The Netherlands
- Department of Molecular Pathology, Oncode Institute, Netherlands Cancer Institute, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands
| |
Collapse
|
578
|
Decker M, Sacks P, Abbatematteo J, De Leo E, Brennan M, Rahman M. The effects of hyperglycemia on outcomes in surgical high-grade glioma patients. Clin Neurol Neurosurg 2019; 179:9-13. [PMID: 30784896 DOI: 10.1016/j.clineuro.2019.02.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 02/05/2019] [Accepted: 02/10/2019] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To define the glucose values associated with an increase in complication rates in post-operative brain tumor patients. PATIENTS AND METHODS Patients who underwent craniotomy for resection of WHO Grade III or IV glioma from 2011 to 2014 were retrospectively reviewed. Post-operative blood glucose values were recorded for post-operative day #0, #1, and #2. Medians were obtained and assessed for significance. Multivariate analysis was performed to assess patient demographics, pre-operative findings, steroid use, and blood glucose values with respect to post-operative complications and to 30-day readmission. RESULTS 108 patients underwent craniotomy for resection of high-grade glioma and had postoperative blood glucose values documented. Median blood glucose values greater than 167 mg/dL were associated with increased serious post-operative complications, and values greater than 163 mg/dL were associated with increased 30-day readmissions. CONCLUSION Post-operative hyperglycemia in patients with high-grade gliomas places this vulnerable patient population to undue post-operative complications and readmissions, potentially delaying further treatment of their disease.
Collapse
Affiliation(s)
- Matthew Decker
- Lillian S. Wells Department of Neurosurgery, 1600 S.W. Archer Road, University of Florida, Gainesville, FL, 32608, United States.
| | - Patricia Sacks
- Lillian S. Wells Department of Neurosurgery, 1600 S.W. Archer Road, University of Florida, Gainesville, FL, 32608, United States
| | - Joseph Abbatematteo
- Lillian S. Wells Department of Neurosurgery, 1600 S.W. Archer Road, University of Florida, Gainesville, FL, 32608, United States
| | - Edward De Leo
- Lillian S. Wells Department of Neurosurgery, 1600 S.W. Archer Road, University of Florida, Gainesville, FL, 32608, United States
| | - Meghan Brennan
- Lillian S. Wells Department of Neurosurgery, 1600 S.W. Archer Road, University of Florida, Gainesville, FL, 32608, United States
| | - Maryam Rahman
- Lillian S. Wells Department of Neurosurgery, 1600 S.W. Archer Road, University of Florida, Gainesville, FL, 32608, United States
| |
Collapse
|
579
|
Wen PY, Touat M, Alexander BM, Mellinghoff IK, Ramkissoon S, McCluskey CS, Pelton K, Haidar S, Basu SS, Gaffey SC, Brown LE, Martinez-Ledesma JE, Wu S, Kim J, Wei W, Park MA, Huse JT, Kuhn JG, Rinne ML, Colman H, Agar NYR, Omuro AM, DeAngelis LM, Gilbert MR, de Groot JF, Cloughesy TF, Chi AS, Roberts TM, Zhao JJ, Lee EQ, Nayak L, Heath JR, Horky LL, Batchelor TT, Beroukhim R, Chang SM, Ligon AH, Dunn IF, Koul D, Young GS, Prados MD, Reardon DA, Yung WKA, Ligon KL. Buparlisib in Patients With Recurrent Glioblastoma Harboring Phosphatidylinositol 3-Kinase Pathway Activation: An Open-Label, Multicenter, Multi-Arm, Phase II Trial. J Clin Oncol 2019; 37:741-750. [PMID: 30715997 DOI: 10.1200/jco.18.01207] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
PURPOSE Phosphatidylinositol 3-kinase (PI3K) signaling is highly active in glioblastomas. We assessed pharmacokinetics, pharmacodynamics, and efficacy of the pan-PI3K inhibitor buparlisib in patients with recurrent glioblastoma with PI3K pathway activation. METHODS This study was a multicenter, open-label, multi-arm, phase II trial in patients with PI3K pathway-activated glioblastoma at first or second recurrence. In cohort 1, patients scheduled for re-operation after progression received buparlisib for 7 to 13 days before surgery to evaluate brain penetration and modulation of the PI3K pathway in resected tumor tissue. In cohort 2, patients not eligible for re-operation received buparlisib until progression or unacceptable toxicity. Once daily oral buparlisib 100 mg was administered on a continuous 28-day schedule. Primary end points were PI3K pathway inhibition in tumor tissue and buparlisib pharmacokinetics in cohort 1 and 6-month progression-free survival (PFS6) in cohort 2. RESULTS Sixty-five patients were treated (cohort 1, n = 15; cohort 2, n = 50). In cohort 1, reduction of phosphorylated AKTS473 immunohistochemistry score was achieved in six (42.8%) of 14 patients, but effects on phosphoribosomal protein S6S235/236 and proliferation were not significant. Tumor-to-plasma drug level was 1.0. In cohort 2, four (8%) of 50 patients reached 6-month PFS6, and the median PFS was 1.7 months (95% CI, 1.4 to 1.8 months). The most common grade 3 or greater adverse events related to treatment were lipase elevation (n = 7 [10.8%]), fatigue (n = 4 [6.2%]), hyperglycemia (n = 3 [4.6%]), and elevated ALT (n = 3 [4.6%]). CONCLUSION Buparlisib had minimal single-agent efficacy in patients with PI3K-activated recurrent glioblastoma. Although buparlisib achieved significant brain penetration, the lack of clinical efficacy was explained by incomplete blockade of the PI3K pathway in tumor tissue. Integrative results suggest that additional study of PI3K inhibitors that achieve more-complete pathway inhibition may still be warranted.
Collapse
Affiliation(s)
- Patrick Y Wen
- 1 Dana-Farber Cancer Institute, Boston, MA.,2 Brigham and Women's Hospital, Boston, MA
| | - Mehdi Touat
- 1 Dana-Farber Cancer Institute, Boston, MA.,2 Brigham and Women's Hospital, Boston, MA
| | - Brian M Alexander
- 1 Dana-Farber Cancer Institute, Boston, MA.,2 Brigham and Women's Hospital, Boston, MA
| | | | | | | | | | - Sam Haidar
- 1 Dana-Farber Cancer Institute, Boston, MA
| | - Sankha S Basu
- 1 Dana-Farber Cancer Institute, Boston, MA.,2 Brigham and Women's Hospital, Boston, MA
| | | | | | | | - Shaofang Wu
- 4 The University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Jungwoo Kim
- 5 California Institute of Technology, Pasadena, CA
| | - Wei Wei
- 6 David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA.,10 Institute for Systems Biology, Seattle, WA
| | - Mi-Ae Park
- 1 Dana-Farber Cancer Institute, Boston, MA
| | - Jason T Huse
- 4 The University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - John G Kuhn
- 7 The University of Texas, San Antonio, San Antonio, TX
| | - Mikael L Rinne
- 1 Dana-Farber Cancer Institute, Boston, MA.,2 Brigham and Women's Hospital, Boston, MA
| | - Howard Colman
- 8 Huntsman Cancer Institute and University of Utah, Salt Lake City, UT
| | - Nathalie Y R Agar
- 1 Dana-Farber Cancer Institute, Boston, MA.,2 Brigham and Women's Hospital, Boston, MA
| | | | | | - Mark R Gilbert
- 4 The University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - John F de Groot
- 4 The University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Timothy F Cloughesy
- 6 David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA
| | - Andrew S Chi
- 9 New York University School of Medicine, New York, NY
| | | | | | - Eudocia Q Lee
- 1 Dana-Farber Cancer Institute, Boston, MA.,2 Brigham and Women's Hospital, Boston, MA
| | - Lakshmi Nayak
- 1 Dana-Farber Cancer Institute, Boston, MA.,2 Brigham and Women's Hospital, Boston, MA
| | | | | | | | - Rameen Beroukhim
- 1 Dana-Farber Cancer Institute, Boston, MA.,2 Brigham and Women's Hospital, Boston, MA
| | - Susan M Chang
- 12 University of California, San Francisco, San Francisco, CA
| | | | - Ian F Dunn
- 2 Brigham and Women's Hospital, Boston, MA
| | - Dimpy Koul
- 4 The University of Texas M.D. Anderson Cancer Center, Houston, TX
| | | | | | - David A Reardon
- 1 Dana-Farber Cancer Institute, Boston, MA.,2 Brigham and Women's Hospital, Boston, MA
| | - W K Alfred Yung
- 4 The University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Keith L Ligon
- 1 Dana-Farber Cancer Institute, Boston, MA.,2 Brigham and Women's Hospital, Boston, MA
| |
Collapse
|
580
|
Wildes TJ, Flores CT, Mitchell DA. Concise Review: Modulating Cancer Immunity with Hematopoietic Stem and Progenitor Cells. Stem Cells 2019; 37:166-175. [PMID: 30353618 PMCID: PMC6368859 DOI: 10.1002/stem.2933] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 09/19/2018] [Accepted: 10/02/2018] [Indexed: 12/17/2022]
Abstract
Hematopoietic stem and progenitor cells (HSPCs) are the progenitor cells that can regenerate the entire blood compartment, including the immune system. Recent studies have unearthed considerable immune-modulating potential of these cells. They can migrate through chemotactic gradients, differentiate into functional immune cells, and crosstalk with immune cells during infections, autoimmune diseases, and cancers. Although the primary role of HSPCs during solid malignancies is considered immunosuppressive, recent studies have discovered immune-activating HSPCs and progeny. In this review, we will discuss the recent evidence that HSPCs act as immunomodulators during solid cancers and highlight the future directions of discovery. Stem Cells 2019;37:166-175.
Collapse
Affiliation(s)
- Tyler J. Wildes
- University of Florida Brain Tumor Immunotherapy Program, Preston A. Wells, Jr. Center for Brain Tumor Therapy, Lillian S. Wells Department of NeurosurgeryMcKnight Brain Institute, University of FloridaGainesvilleFloridaUSA
| | - Catherine T. Flores
- University of Florida Brain Tumor Immunotherapy Program, Preston A. Wells, Jr. Center for Brain Tumor Therapy, Lillian S. Wells Department of NeurosurgeryMcKnight Brain Institute, University of FloridaGainesvilleFloridaUSA
| | - Duane A. Mitchell
- University of Florida Brain Tumor Immunotherapy Program, Preston A. Wells, Jr. Center for Brain Tumor Therapy, Lillian S. Wells Department of NeurosurgeryMcKnight Brain Institute, University of FloridaGainesvilleFloridaUSA
| |
Collapse
|
581
|
Ursu R, Thomas L, Psimaras D, Chinot O, Le Rhun E, Ricard D, Charissoux M, Cuzzubbo S, Sejalon F, Quillien V, Hoang-Xuan K, Ducray F, Portal JJ, Tibi A, Mandonnet E, Levy-Piedbois C, Vicaut E, Carpentier AF. Angiotensin II receptor blockers, steroids and radiotherapy in glioblastoma-a randomised multicentre trial (ASTER trial). An ANOCEF study. Eur J Cancer 2019; 109:129-136. [PMID: 30716716 DOI: 10.1016/j.ejca.2018.12.025] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 12/18/2018] [Accepted: 12/23/2018] [Indexed: 11/20/2022]
Abstract
BACKGROUND Glioblastomas (GBMs) induce a peritumoural vasogenic oedema impairing functional status and quality of life. Steroids reduce brain tumour-related oedema but are associated with numerous side-effects. It was reported in a retrospective series that angiotensin receptor blockers might be associated with reduced peritumoural oedema. The ASTER study is a randomised, placebo-controlled trial to assess whether or not the addition of Losartan to standard of care (SOC) can reduce steroid requirement during radiotherapy (RT) in patients with newly diagnosed GBM. PATIENTS AND METHODS Patients with a histologically confirmed GBM after biopsy or partial surgical resection were randomised between Losartan or placebo in addition to SOC with RT and temozolomide (TMZ). The primary objective was to investigate the steroid dosage required to control brain oedema on the last day of RT in each arm. The secondary outcomes were steroids dosage 1 month after the end of RT, assessment of cerebral oedema on magnetic resonance imaging, tolerance and survival. RESULTS Seventy-five patients were randomly assigned to receive Losartan (37 patients) or placebo (38 patients). No difference in the steroid dosage required to control brain oedema on the last day of RT, or one month after completion of RT, was seen between both arms. The incidence of adverse events was similar in both arms. Median overall survival was similar in both arms. CONCLUSIONS Losartan, although well tolerated, does not reduce the steroid requirement in newly diagnosed GBM patients treated with concomitant RT and TMZ. Trial registration number NCT01805453 with ClinicalTrials.gov.
Collapse
Affiliation(s)
- R Ursu
- Department of Neurology, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France; University Paris Diderot, Sorbonne Paris Cité, Paris, France.
| | - L Thomas
- Department of Neuro-Oncology, Hospices Civils de Lyon, Groupe Hospitalier Est, Lyon, France
| | - D Psimaras
- Department of Neurology Mazarin, Assistance Publique-Hôpitaux de Paris, Sorbonne Université, Hôpital Pitié-Salpêtrière, Paris, France
| | - O Chinot
- Department of Neuro-Oncology, CHU Timone, Marseille, France; Aix-Marseille Université, Assistance Publique-Hôpitaux de Marseille, Marseille, France
| | - E Le Rhun
- University of Lille, Inserm, U-1192, F-59000 Lille, France; CHU Lille, General and Stereotaxic Neurosurgery Service, F-59000 Lille, France; Oscar Lambret Center, Neurology, Medical Oncology Department, F-59000 Lille, France
| | - D Ricard
- Department of Neurology, Hôpital d'Instruction des Armées Percy, Service de Santé des Armées, Paris, France
| | - M Charissoux
- Department of Radiation Oncology, Institut du Cancer de Montpellier, Montpellier cedex 5, France
| | - S Cuzzubbo
- Department of Neurology, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France; University Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - F Sejalon
- Department of Neurology, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - V Quillien
- Centre de Lutte Contre le Cancer Eugène Marquis, F-35042 Rennes, France; INSERM U1242, "Chemistry, Oncogenesis, Stress, Signaling", Université de Rennes 1, Rennes, France
| | - K Hoang-Xuan
- Department of Neurology Mazarin, Assistance Publique-Hôpitaux de Paris, Sorbonne Université, Hôpital Pitié-Salpêtrière, Paris, France
| | - F Ducray
- Department of Neuro-Oncology, Hospices Civils de Lyon, Groupe Hospitalier Est, Lyon, France; Department of Cancer Cell Plasticity, Cancer Research Centre of Lyon, INSERM U1052, CNRS UMR5286, Lyon, France; University Claude Bernard Lyon 1, Lyon, France
| | - J-J Portal
- AP-HP, Unité de Recherche Clinique, Hôpital Fernand Widal, Université Paris-Diderot, Paris, France
| | - A Tibi
- Agence Générale des Equipements et Produits de Santé (AGEPS), Paris, France
| | - E Mandonnet
- Department of Neurosurgery, Lariboisière Hospital, APHP, Paris, France; University Paris 7, Paris, France; IMNC, UMR 8165, Orsay, France
| | - C Levy-Piedbois
- Ramsey Générale de Santé, Institut de Radiothérapie des Hauts-Energies, Bobigny, France
| | - E Vicaut
- AP-HP, Unité de Recherche Clinique, Hôpital Fernand Widal, Université Paris-Diderot, Paris, France
| | - A F Carpentier
- Department of Neurology, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France; University Paris Diderot, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
582
|
Lu G, Rao M, Zhu P, Liang B, El-Nazer RT, Fonkem E, Bhattacharjee MB, Zhu JJ. Triple-drug Therapy With Bevacizumab, Irinotecan, and Temozolomide Plus Tumor Treating Fields for Recurrent Glioblastoma: A Retrospective Study. Front Neurol 2019; 10:42. [PMID: 30766509 PMCID: PMC6366009 DOI: 10.3389/fneur.2019.00042] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 01/14/2019] [Indexed: 12/11/2022] Open
Abstract
Clinical studies treating pediatric and adult solid tumors, such as glioblastoma (GBM), with a triple-drug regimen of temozolomide (TMZ), bevacizumab (BEV), and irinotecan (IRI) [TBI] have demonstrated various efficacies, but with no unexpected toxicities. The TBI regimen has never been studied in recurrent GBM (rGBM) patients. In this retrospective study, we investigated the outcomes and side effects of rGBM patients who had received the TBI regimen. We identified 48 adult rGBM patients with a median age of 56 years (range: 26-76), who received Tumor Treating Fields (TTFields) treatment for 30 days or longer, and concurrent salvage chemotherapies. The patients were classified into two groups based on chemotherapies received: TBI with TTFields (TBI+T, N = 18) vs. bevacizumab (BEV)-based chemotherapies with TTFields (BBC+T, N = 30). BBC regimens were either BEV monotherapy, BEV+IRI or BEV+CCNU. Patients in TBI+T group received on average 19 cycles of TMZ, 26 and 21 times infusions with BEV and IRI, respectively. Median overall survival (OS) and progression-free survival (PFS) for rGBM (OS-R and PFS-R) patients who received TBI+T were 18.9 and 10.7 months, respectively. In comparison, patients who received BBC+T treatment had OS-R and PFS-R of 11.8 (P > 0.05) and 4.7 (P < 0.05) months, respectively. Although the median PFS results were significantly different by 1.5 months (6.6 vs. 5.1) between TBI+T and BBC+T groups, the median OS difference of 14.7 months (32.5 vs. 17.8) was more pronounced, P < 0.05. Patients tolerated TBI+T or BBC+T treatments well and there were no unexpected toxicities. The most common side effects from TBI+T treatment included grade III hypertension (38.9%) and leukopenia (22.2%). In conclusion, the TBI regimen might play a role in the improvement of PFS-R and OS-R among rGBM patients. Prospective studies with a larger sample size are warranted to study the efficacy and toxicity of TBI+T regimen for rGBM.
Collapse
Affiliation(s)
- Guangrong Lu
- The Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston (UTHealth), McGovern Medical School, Houston, TX, United States
| | - Mayank Rao
- The Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston (UTHealth), McGovern Medical School, Houston, TX, United States
| | - Ping Zhu
- The Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston (UTHealth), McGovern Medical School, Houston, TX, United States
- Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, UTHealth School of Public Health, Houston, TX, United States
| | - Buqing Liang
- Baylor Scott and White Health, Temple, TX, United States
| | | | - Ekokobe Fonkem
- Baylor Scott and White Health, Temple, TX, United States
| | - Meenakshi B. Bhattacharjee
- Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center at Houston (UTHealth), McGovern Medical School, Houston, TX, United States
| | - Jay-Jiguang Zhu
- The Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston (UTHealth), McGovern Medical School, Houston, TX, United States
| |
Collapse
|
583
|
Mandel JJ, Yust-Katz S, Patel AJ, Cachia D, Liu D, Park M, Yuan Y, Kent TA, de Groot JF. Inability of positive phase II clinical trials of investigational treatments to subsequently predict positive phase III clinical trials in glioblastoma. Neuro Oncol 2019; 20:113-122. [PMID: 29016865 DOI: 10.1093/neuonc/nox144] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background Glioblastoma is the most common primary malignant brain tumor in adults, but effective therapies are lacking. With the scarcity of positive phase III trials, which are increasing in cost, we examined the ability of positive phase II trials to predict statistically significant improvement in clinical outcomes of phase III trials. Methods A PubMed search was conducted to identify phase III clinical trials performed in the past 25 years for patients with newly diagnosed or recurrent glioblastoma. Trials were excluded if they did not examine an investigational chemotherapy or agent, if they were stopped early owing to toxicity, if they lacked prior phase II studies, or if a prior phase II study was negative. Results Seven phase III clinical trials in newly diagnosed glioblastoma and 4 phase III clinical trials in recurrent glioblastoma met the inclusion criteria. Only 1 (9%) phase III study documented an improvement in overall survival and changed the standard of care. Conclusion The high failure rate of phase III trials demonstrates the urgent need to increase the reliability of phase II trials of treatments for glioblastoma. Strategies such as the use of adaptive trial designs, Bayesian statistics, biomarkers, volumetric imaging, and mathematical modeling warrant testing. Additionally, it is critical to increase our expectations of phase II trials so that positive findings increase the probability that a phase III trial will be successful.
Collapse
Affiliation(s)
- Jacob J Mandel
- Baylor College of Medicine, Department of Neurology, Houston, Texas, USA
| | - Shlomit Yust-Katz
- Rabin Medical Center, Department of Neurosurgery, Petah Tikva, Israel
| | - Akash J Patel
- Baylor College of Medicine, Department of Neurology, Houston, Texas, USA
| | - David Cachia
- Medical University of South Carolina, Department of Neurosurgery, Charleston, South Carolina, USA
| | - Diane Liu
- The University of Texas MD Anderson Cancer Center, Department of Biostatistics, Houston, Texas, USA
| | - Minjeong Park
- The University of Texas MD Anderson Cancer Center, Department of Biostatistics, Houston, Texas, USA
| | - Ying Yuan
- The University of Texas MD Anderson Cancer Center, Department of Biostatistics, Houston, Texas, USA
| | - Thomas A Kent
- Baylor College of Medicine, Department of Neurology, Houston, Texas, USA
| | - John F de Groot
- The University of Texas MD Anderson Cancer Center, Department of Neuro-Oncology, Houston, Texas, USA
| |
Collapse
|
584
|
Kinzel A, Ambrogi M, Varshaver M, Kirson ED. Tumor Treating Fields for Glioblastoma Treatment: Patient Satisfaction and Compliance With the Second-Generation Optune ® System. CLINICAL MEDICINE INSIGHTS-ONCOLOGY 2019; 13:1179554918825449. [PMID: 30728735 PMCID: PMC6351720 DOI: 10.1177/1179554918825449] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 12/20/2018] [Indexed: 12/02/2022]
Abstract
Background: Tumor treating fields (TTFields) are a non-invasive antimitotic therapy that delivers alternating electric fields via the Optune® system. The Phase III EF-14 trial in newly diagnosed glioblastoma multiforme (GBM) showed significantly improved progression-free, overall and long-term survival when Optune was used together with maintenance temozolomide (TMZ) compared with TMZ alone. Compliance (average monthly use) was associated with better clinical outcome. The first-generation Optune system weighed approximately 6 pounds (~2.7 kg). The second-generation redesigned Optune system weighs 2.7 pounds (~1.2 kg). We tested and compared GBM patient experience with the second-generation system versus the first-generation system. Methods: Ten newly diagnosed and recurrent GBM patients in Germany (median age: 52.9 years [31-79]) were prospectively monitored over the first month of transitioning from the first-generation to the second-generation Optune system. Questionnaires using a numerical analog scale assessed feedback at baseline (first generation) and after 1 month of second-generation use. Results: After transitioning to the second-generation system, compliance improved by more than 10% in four patients, was maintained in five patients and decreased by more than 10% in one patient. Following transition, eight out of nine patients reported a reduction in the triggering of malfunction alarms. Self-reported patient feedback showed improved handling and portability (weight, mobility) of the second- versus the first-generation Optune system. Conclusions: This patient user survey suggests that patient satisfaction with the second-generation Optune system is improved versus the first-generation system. Improved features of the new system help patients achieve and maintain a higher rate of treatment compliance.
Collapse
|
585
|
Neal A, Moffat BA, Stein JM, Nanga RPR, Desmond P, Shinohara RT, Hariharan H, Glarin R, Drummond K, Morokoff A, Kwan P, Reddy R, O'Brien TJ, Davis KA. Glutamate weighted imaging contrast in gliomas with 7 Tesla magnetic resonance imaging. NEUROIMAGE-CLINICAL 2019; 22:101694. [PMID: 30822716 PMCID: PMC6396013 DOI: 10.1016/j.nicl.2019.101694] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 01/10/2019] [Accepted: 01/27/2019] [Indexed: 01/04/2023]
Abstract
Introduction Diffuse gliomas are incurable malignancies, which undergo inevitable progression and are associated with seizure in 50–90% of cases. Glutamate has the potential to be an important glioma biomarker of survival and local epileptogenicity if it can be accurately quantified noninvasively. Methods We applied the glutamate-weighted imaging method GluCEST (glutamate chemical exchange saturation transfer) and single voxel MRS (magnetic resonance spectroscopy) at 7 Telsa (7 T) to patients with gliomas. GluCEST contrast and MRS metabolite concentrations were quantified within the tumour region and peritumoural rim. Clinical variables of tumour aggressiveness (prior adjuvant therapy and previous radiological progression) and epilepsy (any prior seizures, seizure in last month and drug refractory epilepsy) were correlated with respective glutamate concentrations. Images were separated into post-hoc determined patterns and clinical variables were compared across patterns. Results Ten adult patients with a histo-molecular (n = 9) or radiological (n = 1) diagnosis of grade II-III diffuse glioma were recruited, 40.3 +/− 12.3 years. Increased tumour GluCEST contrast was associated with prior adjuvant therapy (p = .001), and increased peritumoural GluCEST contrast was associated with both recent seizures (p = .038) and drug refractory epilepsy (p = .029). We distinguished two unique GluCEST contrast patterns with distinct clinical and radiological features. MRS glutamate correlated with GluCEST contrast within the peritumoural voxel (R = 0.89, p = .003) and a positive trend existed in the tumour voxel (R = 0.65, p = .113). Conclusion This study supports the role of glutamate in diffuse glioma biology. It further implicates elevated peritumoural glutamate in epileptogenesis and altered tumour glutamate homeostasis in glioma aggressiveness. Given the ability to non-invasively visualise and quantify glutamate, our findings raise the prospect of 7 T GluCEST selecting patients for individualised therapies directed at the glutamate pathway. Larger studies with prospective follow-up are required. 7 T GluCEST glioma imaging is feasible, producing high quality quantifiable images. Increased peritumoural GluCEST contrast correlates with drug resistant epilepsy. Increased tumour GluCEST contrast is associated with prior adjuvant therapy. Two GluCEST patterns were identified with distinct clinico-radiological features. GluCEST contrast correlates with MRS glutamate in peritumoural regions.
Collapse
Affiliation(s)
- Andrew Neal
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Australia; Department of Neurology, Royal Melbourne Hospital, Australia.
| | - Bradford A Moffat
- Melbourne Node of the National Imaging Facility, Department of Radiology, University of Melbourne, Australia
| | - Joel M Stein
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| | - Ravi Prakash Reddy Nanga
- Center for Magnetic Resonance & Optical Imaging, Department of Radiology, University of Pennsylvania, Philadelphia, PA, United States
| | - Patricia Desmond
- Department of Radiology, Royal Melbourne Hospital, Australia; Department of Radiology and Medicine, University of Melbourne, Australia
| | - Russell T Shinohara
- Department of Biostatistics, Epidemiology, and Informatics, Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, PA, United States
| | - Hari Hariharan
- Center for Magnetic Resonance & Optical Imaging, Department of Radiology, University of Pennsylvania, Philadelphia, PA, United States
| | - Rebecca Glarin
- Department of Radiology, Royal Melbourne Hospital, Australia; Department of Radiology and Medicine, University of Melbourne, Australia
| | - Katharine Drummond
- Department of Neurosurgery, Royal Melbourne Hospital, Australia; Department of Surgery, University of Melbourne, Australia; Melbourne Brain Centre, The Royal Melbourne Hospital, Australia
| | - Andrew Morokoff
- Department of Neurosurgery, Royal Melbourne Hospital, Australia; Department of Surgery, University of Melbourne, Australia
| | - Patrick Kwan
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Australia; Department of Neurology, Royal Melbourne Hospital, Australia; Department of Neuroscience, Central Clinical School, Monash University, Australia; Department of Neurology, The Alfred Hospital Monash University, Australia
| | - Ravinder Reddy
- Center for Magnetic Resonance & Optical Imaging, Department of Radiology, University of Pennsylvania, Philadelphia, PA, United States
| | - Terence J O'Brien
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Australia; Department of Neurology, Royal Melbourne Hospital, Australia; Department of Neuroscience, Central Clinical School, Monash University, Australia; Department of Neurology, The Alfred Hospital Monash University, Australia
| | - Kathryn A Davis
- Penn Epilepsy Center, Department of Neurology, Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
586
|
Ganipineni LP, Ucakar B, Joudiou N, Riva R, Jérôme C, Gallez B, Danhier F, Préat V. Paclitaxel-loaded multifunctional nanoparticles for the targeted treatment of glioblastoma. J Drug Target 2019; 27:614-623. [PMID: 30633585 DOI: 10.1080/1061186x.2019.1567738] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION We hypothesised that the active targeting of αvβ3 integrin overexpressed in neoangiogenic blood vessels and glioblastoma (GBM) cells combined with magnetic targeting of paclitaxel- and SPIO-loaded PLGA-based nanoparticles could improve accumulation of nanoparticles in the tumour and therefore improve the treatment of GBM. METHODS PTX/SPIO PLGA nanoparticles with or without RGD-grafting were characterised. Their in vitro cellular uptake and cytotoxicity was evaluated by fluorospectroscopy and MTT assay. In vivo safety and anti-tumour efficacy of different targeting strategies were evaluated in orthotopic U87MG tumour model over multiple intravenous injections. RESULTS The nanoparticles of 250 nm were negatively charged. RGD targeted nanoparticles showed a specific and higher cellular uptake than untargeted nanoparticles by activated U87MG and HUVEC cells. In vitro IC50 of PTX after 48 h was ∼1 ng/mL for all the PTX-loaded nanoparticles. The median survival time of the mice treated with magnetic targeted nanoparticles was higher than the control (saline) mice or mice treated with other evaluated strategies. The 6 doses of PTX did not induce any detectable toxic effects on liver, kidney and heart when compared to Taxol. CONCLUSION The magnetic targeting strategy resulted in a better therapeutic effect than the other targeting strategies (passive, active).
Collapse
Affiliation(s)
- Lakshmi Pallavi Ganipineni
- a Université Catholique de Louvain, Advanced Drug Delivery and Biomaterials, Louvain Drug Research Institute , Brussels , Belgium
| | - Bernard Ucakar
- a Université Catholique de Louvain, Advanced Drug Delivery and Biomaterials, Louvain Drug Research Institute , Brussels , Belgium
| | - Nicolas Joudiou
- b Université Catholique de Louvain, Nuclear and Electron Spin Technologies Platform (NEST), Louvain Drug Research Institute , Brussels , Belgium
| | - Raphaël Riva
- c University of Liège, Center for Education and Research on Macromolecules (CERM), CESAM Research Unit , Liège , Belgium
| | - Christine Jérôme
- c University of Liège, Center for Education and Research on Macromolecules (CERM), CESAM Research Unit , Liège , Belgium
| | - Bernard Gallez
- b Université Catholique de Louvain, Nuclear and Electron Spin Technologies Platform (NEST), Louvain Drug Research Institute , Brussels , Belgium
| | - Fabienne Danhier
- a Université Catholique de Louvain, Advanced Drug Delivery and Biomaterials, Louvain Drug Research Institute , Brussels , Belgium
| | - Véronique Préat
- a Université Catholique de Louvain, Advanced Drug Delivery and Biomaterials, Louvain Drug Research Institute , Brussels , Belgium
| |
Collapse
|
587
|
Predictive markers for MGMT promoter methylation in glioblastomas. Neurosurg Rev 2019; 42:867-876. [DOI: 10.1007/s10143-018-01061-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 10/23/2018] [Accepted: 11/22/2018] [Indexed: 12/24/2022]
|
588
|
Wilke C, Grosshans D, Duman J, Brown P, Li J. Radiation-induced cognitive toxicity: pathophysiology and interventions to reduce toxicity in adults. Neuro Oncol 2019; 20:597-607. [PMID: 29045710 DOI: 10.1093/neuonc/nox195] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Radiotherapy is ubiquitous in the treatment of patients with both primary brain tumors as well as disease which is metastatic to the brain. This therapy is not without cost, however, as cognitive decline is frequently associated with cranial radiation, particularly with whole brain radiotherapy (WBRT). The precise mechanisms responsible for radiation-induced morbidity remain incompletely understood and continue to be an active area of ongoing research. In this article, we review the hypothetical means by which cranial radiation induces cognitive decline as well as potential therapeutic approaches to prevent, minimize, or reverse treatment-induced cognitive deterioration. We additionally review advances in imaging modalities that can potentially be used to identify site-specific radiation-induced anatomic or functional changes in the brain and their correlation with clinical outcomes.
Collapse
Affiliation(s)
- Christopher Wilke
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas (C.W., D.G., J.L.); Department of Neuroscience, Baylor College of Medicine, Houston, Texas (J.D.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (P.B.)
| | - David Grosshans
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas (C.W., D.G., J.L.); Department of Neuroscience, Baylor College of Medicine, Houston, Texas (J.D.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (P.B.)
| | - Joseph Duman
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas (C.W., D.G., J.L.); Department of Neuroscience, Baylor College of Medicine, Houston, Texas (J.D.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (P.B.)
| | - Paul Brown
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas (C.W., D.G., J.L.); Department of Neuroscience, Baylor College of Medicine, Houston, Texas (J.D.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (P.B.)
| | - Jing Li
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas (C.W., D.G., J.L.); Department of Neuroscience, Baylor College of Medicine, Houston, Texas (J.D.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (P.B.)
| |
Collapse
|
589
|
Hadjipanayis CG, Stummer W. 5-ALA and FDA approval for glioma surgery. J Neurooncol 2019; 141:479-486. [PMID: 30644008 DOI: 10.1007/s11060-019-03098-y] [Citation(s) in RCA: 232] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 01/09/2019] [Indexed: 12/14/2022]
Abstract
The US Food and Drug Administration (FDA) approved 5-aminolevulinic acid (5-ALA; Gleolan®; photonamic GmbH and Co. KG) for use as an intraoperative optical imaging agent in patients with suspected high-grade gliomas (HGGs) in 2017. This was the first ever optical imaging agent approved as an adjunct for the visualization of malignant tissue during surgery for brain tumors. The approval occurred a decade after European approval and a multicenter, phase III randomized trial which confirmed that surgeons using 5-ALA fluorescence-guided surgery as a surgical adjunct could achieve more complete resections of tumors in HGG patients and better patient outcomes than with conventional microsurgery. Much of the delay in the US FDA approval of 5-ALA stemmed from its conceptualization as a therapeutic and not as an intraoperative imaging tool. We chronicle the challenges encountered during the US FDA approval process to highlight a new standard for approval of intraoperative optical imaging agents in brain tumors.
Collapse
Affiliation(s)
- Constantinos G Hadjipanayis
- Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Neurosurgery, Mount Sinai Beth Israel, New York, NY, USA.
| | - Walter Stummer
- Department of Neurosurgery, Universitätsklinikum Münster, Münster, Germany
| |
Collapse
|
590
|
Yu M, Su D, Yang Y, Qin L, Hu C, Liu R, Zhou Y, Yang C, Yang X, Wang G, Gao H. D-T7 Peptide-Modified PEGylated Bilirubin Nanoparticles Loaded with Cediranib and Paclitaxel for Antiangiogenesis and Chemotherapy of Glioma. ACS APPLIED MATERIALS & INTERFACES 2019; 11:176-186. [PMID: 30525386 DOI: 10.1021/acsami.8b16219] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The blood-brain tumor barrier (BTB) and blood-brain barrier (BBB) have always been the major barriers in glioma therapy. In this report, we proposed D-T7 peptide-modified nanoparticles actively targeted glioma by overcoming the BBB and BTB to improve the antiglioma efficacy. Glioma-targeting experiments showed that the penetration effect of the D-T7 peptide-modified nanoparticles was 7.89-fold higher than that of unmodified nanoparticles. Furthermore, cediranib (CD) and paclitaxel (PTX) were used for the combination of the antiangiogenesis and chemotherapy for glioma. PEGylated bilirubin nanoparticles (BRNPs) were selected as a suitable drug delivery system (CD&PTX@TBRBPs) owing to the antioxidant, anti-inflammatory, and reactive oxygen species-responsive ability. 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and apoptosis assays showed that CD&PTX@TBRBPs had the highest cytotoxicity and the median survival time of the CD&PTX@TBRNP group was 3.31-fold and 1.23-fold longer than that of the saline and CD&PTX@BRNP groups, respectively. All the results showed that we constructed a novel and accessible peptide-modified dual drug carrier with an enhanced antiglioma effect.
Collapse
Affiliation(s)
- Meinan Yu
- Faculty of Life Science and Technology , Kunming University of Science and Technology , 727 South Jing Ming Road , Chenggong County, Kunming 650500 , P. R. China
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy , Sichuan University , No. 17, Block 3, Southern Renmin Road , Chengdu 610041 , P. R. China
| | - Dunyan Su
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy , Sichuan University , No. 17, Block 3, Southern Renmin Road , Chengdu 610041 , P. R. China
| | - Yuanyuan Yang
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy , Sichuan University , No. 17, Block 3, Southern Renmin Road , Chengdu 610041 , P. R. China
| | - Lin Qin
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy , Sichuan University , No. 17, Block 3, Southern Renmin Road , Chengdu 610041 , P. R. China
| | - Chuan Hu
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy , Sichuan University , No. 17, Block 3, Southern Renmin Road , Chengdu 610041 , P. R. China
| | - Rui Liu
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy , Sichuan University , No. 17, Block 3, Southern Renmin Road , Chengdu 610041 , P. R. China
| | - Yang Zhou
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy , Sichuan University , No. 17, Block 3, Southern Renmin Road , Chengdu 610041 , P. R. China
| | - Chuanyao Yang
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy , Sichuan University , No. 17, Block 3, Southern Renmin Road , Chengdu 610041 , P. R. China
| | - Xiaotong Yang
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy , Sichuan University , No. 17, Block 3, Southern Renmin Road , Chengdu 610041 , P. R. China
| | - Guanlin Wang
- Faculty of Life Science and Technology , Kunming University of Science and Technology , 727 South Jing Ming Road , Chenggong County, Kunming 650500 , P. R. China
| | - Huile Gao
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy , Sichuan University , No. 17, Block 3, Southern Renmin Road , Chengdu 610041 , P. R. China
| |
Collapse
|
591
|
Moeckel S, LaFrance K, Wetsch J, Seliger C, Riemenschneider MJ, Proescholdt M, Hau P, Vollmann-Zwerenz A. ATF4 contributes to autophagy and survival in sunitinib treated brain tumor initiating cells (BTICs). Oncotarget 2019; 10:368-382. [PMID: 30719230 PMCID: PMC6349458 DOI: 10.18632/oncotarget.26569] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2017] [Accepted: 12/29/2018] [Indexed: 12/20/2022] Open
Abstract
Receptor tyrosine kinase (RTK) pathways are known to play an important role in tumor cell proliferation of glioblastoma (GBM). Cellular determinants of RTK-inhibitor sensitivity are important to optimize and tailor treatment strategies. The stress response gene activating transcription factor 4 (ATF4) is involved in homeostasis and cellular protection. However, little is known about its function in GBM. We found that the ATF4/p-eIF2α pathway is activated in response to Sunitinib in primary tumor initiating progenitor cell cultures (BTICs). Furthermore, lysosome entrapment of RTK-inhibitors (RTK-Is) leads to accumulation of autophagosomes. In case of Sunitinib treated cells, autophagy is additionally increased by ATF4 mediated upregulation of autophagy genes. Inhibition of ATF4 by small interfering RNA (siRNA) reduced autophagy and cell proliferation after Sunitinib treatment in a subset of BTIC cultures. Overall, this study suggests a pro-survival role of the ATF4/p-eIF2α pathway in a cell type and treatment specific manner.
Collapse
Affiliation(s)
- Sylvia Moeckel
- Wilhelm Sander-NeuroOncology Unit and Department of Neurology, Regensburg University Hospital, Regensburg, Germany
| | - Kelly LaFrance
- Wilhelm Sander-NeuroOncology Unit and Department of Neurology, Regensburg University Hospital, Regensburg, Germany
| | - Julia Wetsch
- Wilhelm Sander-NeuroOncology Unit and Department of Neurology, Regensburg University Hospital, Regensburg, Germany
| | - Corinna Seliger
- Wilhelm Sander-NeuroOncology Unit and Department of Neurology, Regensburg University Hospital, Regensburg, Germany
| | | | - Martin Proescholdt
- Department of Neurosurgery, Regensburg University Hospital, Regensburg, Germany
| | - Peter Hau
- Wilhelm Sander-NeuroOncology Unit and Department of Neurology, Regensburg University Hospital, Regensburg, Germany
| | - Arabel Vollmann-Zwerenz
- Wilhelm Sander-NeuroOncology Unit and Department of Neurology, Regensburg University Hospital, Regensburg, Germany
| |
Collapse
|
592
|
Epilepsy Associates with Decreased HIF-1α/STAT5b Signaling in Glioblastoma. Cancers (Basel) 2019; 11:cancers11010041. [PMID: 30621209 PMCID: PMC6356242 DOI: 10.3390/cancers11010041] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 12/21/2018] [Accepted: 12/28/2018] [Indexed: 02/06/2023] Open
Abstract
Epilepsy at presentation is an independent favorable prognostic factor in glioblastoma (GBM). In this study, we analyze the oncologic signaling pathways that associate with epilepsy in human GBMs, and that can underlie this prognostic effect. Following ethical approval and patient consent, fresh frozen GBM tissue was obtained from 76 patient surgeries. Hospital records were screened for the presence of seizures at presentation of the disease. mRNA and miRNA expression-based and gene set enrichment analyses were performed on these tissues, to uncover candidate oncologic pathways that associate with epilepsy. We performed qPCR experiments and immunohistochemistry on tissue microarrays containing 286 GBMs to further explore the association of these candidate pathways and of markers of mesenchymal transformation (NF-κB, CEBP-β, STAT3, STAT5b, VEGFA, SRF) with epilepsy. Gene sets involved in hypoxia/HIF-1α, STAT5, CEBP-β and epithelial-mesenchymal transformation signaling were significantly downregulated in epileptogenic GBMs. On confirmatory protein expression analyses, epileptogenic tumors were characterized by a significant downregulation of phospho-STAT5b, a target of HIF-1α. Epilepsy status did not associate with molecular subclassification or miRNA expression patterns of the tumors. Epileptogenic GBMs correlate with decreased hypoxia/ HIF-1α/STAT5b signaling compared to glioblastomas that do not present with epilepsy.
Collapse
|
593
|
Halle B, Mongelard K, Poulsen FR. Convection-enhanced Drug Delivery for Glioblastoma: A Systematic Review Focused on Methodological Differences in the Use of the Convection-enhanced Delivery Method. Asian J Neurosurg 2019; 14:5-14. [PMID: 30937002 PMCID: PMC6417332 DOI: 10.4103/ajns.ajns_302_17] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Glioblastoma (GBM) is a leading cause of brain cancer-related death. The blood–brain barrier (BBB) prevents the transport of most systemic delivered molecules to the brain. This constitutes a major problem in the therapy of brain tumors. In the last decade, numerous different drug-delivery approaches have been developed to overcome the BBB. The objective of this study is to provide an overview of the methodological aspects used in all preclinical and clinical studies published from 2011 to 2016 where convection-enhanced delivery (CED) was used for drug delivery in the treatment of GBM. A systematic review of English articles published in the past 5 years was undertaken using PubMed and Embase. The search terms (brain tumor [MeSH Terms]) AND (CED OR convection enhanced delivery) were used in PubMed and a similar search was carried out in Embase using their “multi-field search.” All studies using CED on an intracranial GBM model were included. The search resulted in 151 hits after duplicates were removed. In total, 30 studies were included in the review. Of these, two publications studied the technical aspects of the CED method. Furthermore, only one study was a clinical study. The research field is focused on preclinical drug development trials and less emphasis is placed on the CED technique itself. However, it is important that future studies focus on establishing optimal protocols for the use of CED in rodents as well as for big brain models to be able to use the CED method in patients with GBM.
Collapse
Affiliation(s)
- Bo Halle
- Department of Neurosurgery, Odense University Hospital and BRIDGE - Brain Research - Inter-Disciplinary Guided Excellence, Odense, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Kristian Mongelard
- Department of Neurosurgery, Odense University Hospital and BRIDGE - Brain Research - Inter-Disciplinary Guided Excellence, Odense, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Frantz Rom Poulsen
- Department of Neurosurgery, Odense University Hospital and BRIDGE - Brain Research - Inter-Disciplinary Guided Excellence, Odense, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
594
|
Central Nervous System Tumors. Radiat Oncol 2019. [DOI: 10.1007/978-3-319-97145-2_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
595
|
Toms SA, Kim CY, Nicholas G, Ram Z. Increased compliance with tumor treating fields therapy is prognostic for improved survival in the treatment of glioblastoma: a subgroup analysis of the EF-14 phase III trial. J Neurooncol 2019; 141:467-473. [PMID: 30506499 PMCID: PMC6342854 DOI: 10.1007/s11060-018-03057-z] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 11/21/2018] [Indexed: 01/22/2023]
Abstract
BACKGROUND Tumor treating fields (TTFields) is a non-invasive, antimitotic therapy. In the EF-14 phase 3 trial in newly diagnosed glioblastoma, TTFields plus temozolomide (TTFields/TMZ) improved progression free (PFS) and overall survival (OS) versus TMZ alone. Previous data indicate a ≥ 75% daily compliance improves outcomes. We analyzed compliance data from TTFields/TMZ patients in the EF-14 study to correlate TTFields compliance with PFS and OS and identify potential lower boundary for compliance with improved clinical outcomes. METHODS Compliance was assessed by usage data from the NovoTTF-100A device and calculated as percentage per month of TTFields delivery. TTFields/TMZ patients were segregated into subgroups by percent monthly compliance. A Cox proportional hazard model controlled for sex, extent of resection, MGMT methylation status, age, region, and performance status was used to investigate the effect of compliance on PFS and OS. RESULTS A threshold value of 50% compliance with TTFields/TMZ improved PFS (HR 0.70, 95% CI 0.47-1.05) and OS (HR 0.67, 95% CI 0.45-0.99) versus TMZ alone with improved outcome as compliance increased. At compliance > 90%, median survival was 24.9 months (28.7 months from diagnosis) and 5-year survival rate was 29.3%. Compliance was independent of gender, extent of resection, MGMT methylation status, age, region and performance status (HR 0.78; p = 0.031; OS at compliance ≥ 75% vs. < 75%). CONCLUSION A compliance threshold of 50% with TTFields/TMZ correlated with significantly improved OS and PFS versus TMZ alone. Patients with compliance > 90% showed extended median and 5-year survival rates. Increased compliance with TTFields therapy is independently prognostic for improved survival in glioblastoma.
Collapse
Affiliation(s)
- S A Toms
- Department of Neurosurgery, Warren Alpert Medical School of Brown University, Providence, RI, USA.
| | - C Y Kim
- Seoul National University, Bundang, South Korea
| | - G Nicholas
- Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Z Ram
- Tel Aviv Medical Center, Tel Aviv, Israel
| |
Collapse
|
596
|
Geraldo LHM, Garcia C, da Fonseca ACC, Dubois LGF, de Sampaio e Spohr TCL, Matias D, de Camargo Magalhães ES, do Amaral RF, da Rosa BG, Grimaldi I, Leser FS, Janeiro JM, Macharia L, Wanjiru C, Pereira CM, Moura-Neto V, Freitas C, Lima FRS. Glioblastoma Therapy in the Age of Molecular Medicine. Trends Cancer 2019; 5:46-65. [DOI: 10.1016/j.trecan.2018.11.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 11/09/2018] [Accepted: 11/12/2018] [Indexed: 12/11/2022]
|
597
|
Dressler EV, Liu M, Garcia CR, Dolecek TA, Pittman T, Huang B, Villano JL. Patterns and disparities of care in glioblastoma. Neurooncol Pract 2019; 6:37-46. [PMID: 30740232 PMCID: PMC6352755 DOI: 10.1093/nop/npy014] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Glioblastoma is an aggressive disease with a defined standard of care offering crucial survival benefits. Disparities in care may influence treatment decisions. This study seeks to evaluate potential patterns in care delivery using the National Cancer Database (NCDB). METHODS We evaluated the NCDB from 1998 to 2011 for patients diagnosed with glioblastoma older than 20 years of age in order to describe current hospital-based demographics, rates of treatment modality by age, race, gender, likelihood of receiving treatment, and survival probabilities. RESULTS From 1998 to 2011, 100672 patients were diagnosed with glioblastoma in the United States. Of these, 54% were younger than 65 years of age, while 20% were 75 years of age or older. The most common type of treatment was surgery (73%), followed by radiation (69%) and chemotherapy (50%). Eleven percent of patients did not receive any form of therapy. Patients receiving no form of treatment were more likely to be older, female, black, or Hispanic. Tumors that did not involve brainstem, ventricles, or the cerebellum were associated with more aggressive treatment and better overall survival. The median survival was 7.5 months. The use of concomitant surgical resection, chemotherapy, and radiation demonstrated greater survival benefit. CONCLUSIONS Median survival for glioblastoma is significantly less than reported in clinical trials. Sociodemographic factors such as age, gender, race, and socioeconomic status affect treatment decisions for glioblastoma. The elderly are greatly undertreated, as many elderly patients receive no treatment or significantly less than standard of care.
Collapse
Affiliation(s)
- Emily V Dressler
- Department of Biostatistical Sciences, School of Medicine Wake Forest School of Medicine, Winston Salem, NC
| | - Meng Liu
- Division of Cancer Biostatistics, University of Kentucky, Lexington, Kentucky
| | | | - Therese A Dolecek
- University of Kentucky, Lexington, Kentucky; Division of Epidemiology and Biostatistics and Institute for Health Research and Policy, School of Public Health, University of Illinois at Chicago, Chicago, Illinois
| | - Thomas Pittman
- Department of Neurosurgery, University of Kentucky, Lexington, Kentucky
| | - Bin Huang
- Division of Cancer Biostatistics, University of Kentucky, Lexington, Kentucky
| | - John L Villano
- Department of Medicine, University of Kentucky, Lexington, Kentucky
- Department of Neurology, University of Kentucky, Lexington, Kentucky
- Department of Neurosurgery, University of Kentucky, Lexington, Kentucky
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
598
|
Yin Y, Boesteanu AC, Binder ZA, Xu C, Reid RA, Rodriguez JL, Cook DR, Thokala R, Blouch K, McGettigan-Croce B, Zhang L, Konradt C, Cogdill AP, Panjwani MK, Jiang S, Migliorini D, Dahmane N, Posey AD, June CH, Mason NJ, Lin Z, O’Rourke DM, Johnson LA. Checkpoint Blockade Reverses Anergy in IL-13Rα2 Humanized scFv-Based CAR T Cells to Treat Murine and Canine Gliomas. Mol Ther Oncolytics 2018; 11:20-38. [PMID: 30306125 PMCID: PMC6174845 DOI: 10.1016/j.omto.2018.08.002] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 08/22/2018] [Indexed: 12/31/2022] Open
Abstract
We generated two humanized interleukin-13 receptor α2 (IL-13Rα2) chimeric antigen receptors (CARs), Hu07BBz and Hu08BBz, that recognized human IL-13Rα2, but not IL-13Rα1. Hu08BBz also recognized canine IL-13Rα2. Both of these CAR T cell constructs demonstrated superior tumor inhibitory effects in a subcutaneous xenograft model of human glioma compared with a humanized EGFRvIII CAR T construct used in a recent phase 1 clinical trial (ClinicalTrials.gov: NCT02209376). The Hu08BBz demonstrated a 75% reduction in orthotopic tumor growth using low-dose CAR T cell infusion. Using combination therapy with immune checkpoint blockade, humanized IL-13Rα2 CAR T cells performed significantly better when combined with CTLA-4 blockade, and humanized EGFRvIII CAR T cells' efficacy was improved by PD-1 and TIM-3 blockade in the same mouse model, which was correlated with the levels of checkpoint molecule expression in co-cultures with the same tumor in vitro. Humanized IL-13Rα2 CAR T cells also demonstrated benefit from a self-secreted anti-CTLA-4 minibody in the same mouse model. In addition to a canine glioma cell line (J3T), canine osteosarcoma lung cancer and leukemia cell lines also express IL-13Rα2 and were recognized by Hu08BBz. Canine IL-13Rα2 CAR T cell was also generated and tested in vitro by co-culture with canine tumor cells and in vivo in an orthotopic model of canine glioma. Based on these results, we are designing a pre-clinical trial to evaluate the safety of canine IL-13Rα2 CAR T cells in dog with spontaneous IL-13Rα2-positive glioma, which will help to inform a human clinical trial design for glioblastoma using humanized scFv-based IL-13Rα2 targeting CAR T cells.
Collapse
Affiliation(s)
- Yibo Yin
- The Fourth Section of Department of Neurosurgery, The First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alina C. Boesteanu
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Zev A. Binder
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Chong Xu
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Parker Institute for Cancer Immunotherapy, Philadelphia, PA 19104, USA
| | - Reiss A. Reid
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jesse L. Rodriguez
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Danielle R. Cook
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Radhika Thokala
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kristin Blouch
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bevin McGettigan-Croce
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Logan Zhang
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christoph Konradt
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alexandria P. Cogdill
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - M. Kazim Panjwani
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Parker Institute for Cancer Immunotherapy, Philadelphia, PA 19104, USA
| | - Shuguang Jiang
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Denis Migliorini
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Parker Institute for Cancer Immunotherapy, Philadelphia, PA 19104, USA
| | - Nadia Dahmane
- Department of Neurological Surgery, Weill Cornell Medicine, New York, NY 10065, USA
| | - Avery D. Posey
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Parker Institute for Cancer Immunotherapy, Philadelphia, PA 19104, USA
| | - Carl H. June
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Parker Institute for Cancer Immunotherapy, Philadelphia, PA 19104, USA
| | - Nicola J. Mason
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Parker Institute for Cancer Immunotherapy, Philadelphia, PA 19104, USA
| | - Zhiguo Lin
- The Fourth Section of Department of Neurosurgery, The First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Donald M. O’Rourke
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Laura A. Johnson
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
599
|
Tian Y, Mi G, Chen Q, Chaurasiya B, Li Y, Shi D, Zhang Y, Webster TJ, Sun C, Shen Y. Acid-Induced Activated Cell-Penetrating Peptide-Modified Cholesterol-Conjugated Polyoxyethylene Sorbitol Oleate Mixed Micelles for pH-Triggered Drug Release and Efficient Brain Tumor Targeting Based on a Charge Reversal Mechanism. ACS APPLIED MATERIALS & INTERFACES 2018; 10:43411-43428. [PMID: 30508486 DOI: 10.1021/acsami.8b15147] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Glioblastoma multiforme is the most devastating malignant brain tumor in adults. Even with the standard care of therapy, the prognosis remains dismal due to tumor heterogeneity, tumor infiltration, and, more importantly, the restrictive nature of the blood-brain barrier (BBB). To overcome the challenge of effectively delivering therapeutic cargo into the brain, herein a "smart", multifunctional polymeric micelle was developed using a cholesterol-conjugated polyoxyethylene sorbitol oleate. A cell-penetrating peptide, arginine-glycine repeats (RG)5, was incorporated into the micelles to improve cellular uptake, while a pH-sensitive masking sequence, histidine-glutamic acid repeats (HE)5, was introduced for charge shielding to minimize nonspecific binding and uptake at physiological pH. Results demonstrated that (RG)5- and (HE)5-modified mixed micelles were optimized using this strategy to effectively mask the cationic charges of the activated cell-penetrating peptide (RG)5 at physiological pH, i.e., limiting internalization, and were selectively triggered in response to a mildly acidic microenvironment in vitro based on a charge reversal mechanism. In vivo results further confirmed that such micelles preferentially accumulated in both brain and tumor tissues in both xenograft and orthotropic glioma mouse models. Furthermore, micelles significantly inhibited tumor growth with limited toxicity to peripheral tissues. The combination of BBB penetration, tumor targeting, potent efficacy, and high tolerance of these micelles strongly suggests that they could be a promising candidate for safe and effective drug delivery to the brain.
Collapse
Affiliation(s)
- Yu Tian
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy , China Pharmaceutical University , Nanjing 210009 , China
| | - Gujie Mi
- Department of Chemical Engineering, 313 Snell Engineering Center , Northeastern University , 360 Huntington Avenue , Boston , Massachusetts 02115 , United States
| | - Qian Chen
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy , China Pharmaceutical University , Nanjing 210009 , China
| | - Birendra Chaurasiya
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy , China Pharmaceutical University , Nanjing 210009 , China
| | - Yanan Li
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy , China Pharmaceutical University , Nanjing 210009 , China
| | - Di Shi
- Department of Chemical Engineering, 313 Snell Engineering Center , Northeastern University , 360 Huntington Avenue , Boston , Massachusetts 02115 , United States
| | - Yong Zhang
- Children's Hospital of Nanjing Medical University , Nanjing 210008 , China
| | - Thomas J Webster
- Department of Chemical Engineering, 313 Snell Engineering Center , Northeastern University , 360 Huntington Avenue , Boston , Massachusetts 02115 , United States
| | - Chunmeng Sun
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy , China Pharmaceutical University , Nanjing 210009 , China
| | - Yan Shen
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy , China Pharmaceutical University , Nanjing 210009 , China
| |
Collapse
|
600
|
Zschaeck S, Wust P, Graf R, Misch M, Onken J, Ghadjar P, Badakhshi H, Florange J, Budach V, Kaul D. Locally dose-escalated radiotherapy may improve intracranial local control and overall survival among patients with glioblastoma. Radiat Oncol 2018; 13:251. [PMID: 30567592 PMCID: PMC6299982 DOI: 10.1186/s13014-018-1194-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 11/27/2018] [Indexed: 05/02/2023] Open
Abstract
Background The dismal overall survival (OS) prognosis of glioblastoma, even after trimodal therapy, can be attributed mainly to the frequent incidence of intracranial relapse (ICR), which tends to present as an in-field recurrence after a radiation dose of 60 Gray (Gy). In this study, molecular marker-based prognostic indices were used to compare the outcomes of radiation with a standard dose versus a moderate dose escalation. Methods This retrospective analysis included 156 patients treated between 2009 and 2016. All patients were medically fit for postoperative chemoradiotherapy. In the dose-escalation cohort a simultaneous integrated boost of up to 66 Gy (66 Gy RT) within small high-risk volumes was applied. All other patients received daily radiation to a total dose of 60 Gy or twice daily to a total dose of 59.2 Gy (60 Gy RT). Results A total of 133 patients received standard 60 Gy RT, while 23 received 66 Gy RT. Patients in the 66 Gy RT group were younger (p < 0.001), whereas concomitant temozolomide use was more frequent in the 60 Gy RT group (p < 0.001). Other intergroup differences in known prognostic factors were not observed. Notably, the median time to ICR was significantly prolonged in the 66 Gy RT arm versus the 60 Gy RT arm (12.2 versus 7.6 months, p = 0.011), and this translated to an improved OS (18.8 versus 15.3 months, p = 0.012). A multivariate analysis revealed a strong association of 66 Gy RT with a prolonged time to ICR (hazard ratio = 0.498, p = 0.01) and OS (hazard ratio = 0.451, p = 0.01). These differences remained significant after implementing molecular marker-based prognostic scores (ICR p = 0.008, OS p = 0.007) and propensity-scored matched pairing (ICR p = 0.099, OS p = 0.023). Conclusion Radiation dose escalation was found to correlate with an improved time to ICR and OS in this cohort of glioblastoma patients. However, further prospective validation of these results is warranted. Electronic supplementary material The online version of this article (10.1186/s13014-018-1194-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sebastian Zschaeck
- Department of Radiation Oncology, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Peter Wust
- Department of Radiation Oncology, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Reinhold Graf
- Department of Radiation Oncology, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Martin Misch
- Department of Neurosurgery, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Julia Onken
- Department of Neurosurgery, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Pirus Ghadjar
- Department of Radiation Oncology, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Harun Badakhshi
- Department of Radiation Oncology, Ernst von Bergmann Medical Center, Potsdam, Germany
| | - Julian Florange
- Department of Radiation Oncology, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Volker Budach
- Department of Radiation Oncology, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - David Kaul
- Department of Radiation Oncology, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.
| |
Collapse
|