51
|
Tunc B, Hormuth D, Biros G, Yankeelov TE. Modeling of Glioma Growth with Mass Effect by Longitudinal Magnetic Resonance Imaging. IEEE Trans Biomed Eng 2021; 68:3713-3724. [PMID: 34061731 DOI: 10.1109/tbme.2021.3085523] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
It is well-known that expanding glioblastomas typically induce significant deformations of the surrounding parenchyma (i.e., the so-called ?mass effect?). In this study, we evaluate the performance of three mathematical models of tumor growth: 1) a reaction-diffusion-advection model which accounts for mass effect (RDAM), 2) a reaction-diffusion model with mass effect that is consistent only in the case of small deformations (RDM), and 3) a reaction-diffusion model that does not include the mass effect (RD). The models were calibrated with magnetic resonance imaging (MRI) data obtained during tumor development in a murine model of glioma (n = 9). We obtained T2-weighted and contrast-enhanced T1-weighted MRI at 6 time points over 10 days to determine the spatiotemporal variation in the mass effect and tumor concentration, respectively. We calibrated the three models using data 1) at the first four, 2) only at the first and fourth, and 3) only at the third and fourth time points. Each of these calibrations were run forward in time to predict the volume fraction of tumor cells at the conclusion of the experiment. The diffusion coefficient for the RDAM model (median of 10.65 ? 10-3 mm2d-1) is significantly less than those for the RD and RDM models (17.46 ? 10-3 mm2d-1 and 19.38 ? 10-3 mm2d-1, respectively). The tumor concentrations for the RD, RDM, and RDAM models have medians of 40.2%, 32.1%, and 44.7%, respectively, for the calibration using data from the first four time points. The RDM model most accurately predicts tumor growth, while the RDAM model presents the least variation in its estimates of the diffusion coefficient and proliferation rate. This study demonstrates that the mathematical models capture both tumor development and mass effect observed in experiments.
Collapse
|
52
|
Abstract
Metabolic reprogramming is an important characteristics of glioma, the most common form of malignant brain tumor. In this chapter, we aim to discuss some of the recently discovered metabolic alterations in glioma, including the dysregulated TCA cycle, amino acid, nucleotide, and lipid metabolism. We have also detailed some of the metabolomic applications in gliomas, particularly the analyses of body fluids and tissues of glioma patients. With new improvement of the technology, metabolomics will become a powerful tool to discover truly meaningful biomarkers for clinical applications in gliomas. Metabolomic studies of gliomas will also facilitate a better understanding of the molecular targets/pathways and the development of new therapeutic treatments for this devastating disease.
Collapse
|
53
|
Li Z, Langhans SA. In Vivo and Ex Vivo Pediatric Brain Tumor Models: An Overview. Front Oncol 2021; 11:620831. [PMID: 33869004 PMCID: PMC8047472 DOI: 10.3389/fonc.2021.620831] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 03/15/2021] [Indexed: 12/18/2022] Open
Abstract
After leukemia, tumors of the brain and spine are the second most common form of cancer in children. Despite advances in treatment, brain tumors remain a leading cause of death in pediatric cancer patients and survivors often suffer from life-long consequences of side effects of therapy. The 5-year survival rates, however, vary widely by tumor type, ranging from over 90% in more benign tumors to as low as 20% in the most aggressive forms such as glioblastoma. Even within historically defined tumor types such as medulloblastoma, molecular analysis identified biologically heterogeneous subgroups each with different genetic alterations, age of onset and prognosis. Besides molecularly driven patient stratification to tailor disease risk to therapy intensity, such a diversity demonstrates the need for more precise and disease-relevant pediatric brain cancer models for research and drug development. Here we give an overview of currently available in vitro and in vivo pediatric brain tumor models and discuss the opportunities that new technologies such as 3D cultures and organoids that can bridge limitations posed by the simplicity of monolayer cultures and the complexity of in vivo models, bring to accommodate better precision in drug development for pediatric brain tumors.
Collapse
Affiliation(s)
| | - Sigrid A. Langhans
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children, Wilmington, DE, United States
| |
Collapse
|
54
|
Lamirault C, Brisebard E, Patriarca A, Juchaux M, Crepin D, Labiod D, Pouzoulet F, Sebrie C, Jourdain L, Le Dudal M, Hardy D, De Marzi L, Dendale R, Jouvion G, Prezado Y. Spatially Modulated Proton Minibeams Results in the Same Increase of Lifespan as a Uniform Target Dose Coverage in F98-Glioma-Bearing Rats. Radiat Res 2021; 194:715-723. [PMID: 32991712 DOI: 10.1667/rade-19-00013.1] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 08/14/2020] [Indexed: 11/03/2022]
Abstract
Proton minibeam radiation therapy (pMBRT) is a new approach in proton radiotherapy, by which a significant increase in the therapeutic index has already been demonstrated in RG2 glioma-bearing rats. In the current study we investigated the response of other types of glioma (F98) and performed a comparative evaluation of tumor control effectiveness by pMBRT (with different levels of dose heterogeneity) versus conventional proton therapy. The results of our study showed an equivalent increase in the lifespan for all evaluated groups (conventional proton irradiation and pMBRT) and no significant differences in the histopathological analysis of the tumors or remaining brain tissue. The reduced long-term toxicity observed with pMBRT in previous evaluations at the same dose suggests a possible use of pMBRT to treat glioma with less side effects while ensuring the same tumor control achieved with standard proton therapy.
Collapse
Affiliation(s)
- Charlotte Lamirault
- Laboratoire Imagerie et Modelisation pour la Neurobiologie et la Cancerologie, CNRS-Paris 7-Paris 11, Campus d'Orsay, France
| | - Elise Brisebard
- Department of Global Health, Experimental Neuropathology Unit, Institut Pasteur, 75015 Paris, France.,Laboratoire d'Histopathologie, VetAgro-Sup, Université de Lyon, Marcy l'Etoile, Lyon, France
| | - Annalisa Patriarca
- Radiation Oncology Department, Centre de Protonthérapie d'Orsay, University Paris Saclay, Orsay, France
| | - Marjorie Juchaux
- Laboratoire Imagerie et Modelisation pour la Neurobiologie et la Cancerologie, CNRS-Paris 7-Paris 11, Campus d'Orsay, France
| | - Delphine Crepin
- Laboratoire Imagerie et Modelisation pour la Neurobiologie et la Cancerologie, CNRS-Paris 7-Paris 11, Campus d'Orsay, France
| | - Dalila Labiod
- Experimental Radiotherapy Platform Institut Curie, University Paris Saclay, Orsay, France
| | - Frederic Pouzoulet
- Experimental Radiotherapy Platform Institut Curie, University Paris Saclay, Orsay, France
| | - Catherine Sebrie
- BioMaps, Université Paris-Saclay, CEA, CNRS, Inserm,Service Hospitalier Frédéric Joliot, 91401 Orsay, France
| | - Laurene Jourdain
- BioMaps, Université Paris-Saclay, CEA, CNRS, Inserm,Service Hospitalier Frédéric Joliot, 91401 Orsay, France
| | - Marine Le Dudal
- Department of Global Health, Experimental Neuropathology Unit, Institut Pasteur, 75015 Paris, France.,Histologie, Embryologie et Anatomie Pathologique, Ecole Nationale Vétérinaire d'Alfort, Université Paris-Est, Maisons-Alfort, France
| | - David Hardy
- Department of Global Health, Experimental Neuropathology Unit, Institut Pasteur, 75015 Paris, France
| | - Ludovic De Marzi
- Radiation Oncology Department, Centre de Protonthérapie d'Orsay, University Paris Saclay, Orsay, France.,Institut Curie, University Paris Saclay, PSL Research University, Inserm U 1021-CNRS UMR 3347, Orsay, France
| | - Remi Dendale
- Radiation Oncology Department, Centre de Protonthérapie d'Orsay, University Paris Saclay, Orsay, France
| | - Gregory Jouvion
- Department of Global Health, Experimental Neuropathology Unit, Institut Pasteur, 75015 Paris, France.,Sorbonne Université, INSERM, Pathophysiology of Pediatric Genetic Diseases, Assistance Publique - Hôpitaux de Paris, Hôpital Armand-Trousseau, UF Génétique Moléculaire, Paris, France
| | - Yolanda Prezado
- Institut Curie, University Paris Saclay, PSL Research University, Inserm U 1021-CNRS UMR 3347, Orsay, France
| |
Collapse
|
55
|
Chen Z, Peng P, Zhang X, Mania-Farnell B, Xi G, Wan F. Advanced Pediatric Diffuse Pontine Glioma Murine Models Pave the Way towards Precision Medicine. Cancers (Basel) 2021; 13:cancers13051114. [PMID: 33807733 PMCID: PMC7961799 DOI: 10.3390/cancers13051114] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/01/2021] [Accepted: 03/01/2021] [Indexed: 12/14/2022] Open
Abstract
Diffuse intrinsic pontine gliomas (DIPGs) account for ~15% of pediatric brain tumors, which invariably present with poor survival regardless of treatment mode. Several seminal studies have revealed that 80% of DIPGs harbor H3K27M mutation coded by HIST1H3B, HIST1H3C and H3F3A genes. The H3K27M mutation has broad effects on gene expression and is considered a tumor driver. Determination of the effects of H3K27M on posttranslational histone modifications and gene regulations in DIPG is critical for identifying effective therapeutic targets. Advanced animal models play critical roles in translating these cutting-edge findings into clinical trial development. Here, we review current molecular research progress associated with DIPG. We also summarize DIPG animal models, highlighting novel genomic engineered mouse models (GEMMs) and innovative humanized DIPG mouse models. These models will pave the way towards personalized precision medicine for the treatment of DIPGs.
Collapse
Affiliation(s)
- Zirong Chen
- Department of Neurological Surgery, Tongji Hospital, Tongji Medical College, Huazhong University Science and Technology, Wuhan 430030, China; (Z.C.); (P.P.); (X.Z.)
| | - Peng Peng
- Department of Neurological Surgery, Tongji Hospital, Tongji Medical College, Huazhong University Science and Technology, Wuhan 430030, China; (Z.C.); (P.P.); (X.Z.)
| | - Xiaolin Zhang
- Department of Neurological Surgery, Tongji Hospital, Tongji Medical College, Huazhong University Science and Technology, Wuhan 430030, China; (Z.C.); (P.P.); (X.Z.)
| | - Barbara Mania-Farnell
- Department of Biological Science, Purdue University Northwest, Hammond, IN 46323, USA;
| | - Guifa Xi
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Correspondence: (G.X.); (F.W.); Tel.: +1-(312)5034296 (G.X.); +86-(027)-8366-5201 (F.W.)
| | - Feng Wan
- Department of Neurological Surgery, Tongji Hospital, Tongji Medical College, Huazhong University Science and Technology, Wuhan 430030, China; (Z.C.); (P.P.); (X.Z.)
- Correspondence: (G.X.); (F.W.); Tel.: +1-(312)5034296 (G.X.); +86-(027)-8366-5201 (F.W.)
| |
Collapse
|
56
|
Costa PC, Guang Z, Ledwig P, Zhang Z, Neill S, Olson JJ, Robles FE. Towards in-vivo label-free detection of brain tumor margins with epi-illumination tomographic quantitative phase imaging. BIOMEDICAL OPTICS EXPRESS 2021; 12:1621-1634. [PMID: 33796377 PMCID: PMC7984798 DOI: 10.1364/boe.416731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 02/11/2021] [Accepted: 02/20/2021] [Indexed: 05/03/2023]
Abstract
Brain tumor surgery involves a delicate balance between maximizing the extent of tumor resection while minimizing damage to healthy brain tissue that is vital for neurological function. However, differentiating between tumor, particularly infiltrative disease, and healthy brain in-vivo remains a significant clinical challenge. Here we demonstrate that quantitative oblique back illumination microscopy (qOBM)-a novel label-free optical imaging technique that achieves tomographic quantitative phase imaging in thick scattering samples-clearly differentiates between healthy brain tissue and tumor, including infiltrative disease. Data from a bulk and infiltrative brain tumor animal model show that qOBM enables quantitative phase imaging of thick fresh brain tissues with remarkable cellular and subcellular detail that closely resembles histopathology using hematoxylin and eosin (H&E) stained fixed tissue sections, the gold standard for cancer detection. Quantitative biophysical features are also extracted from qOBM which yield robust surrogate biomarkers of disease that enable (1) automated tumor and margin detection with high sensitivity and specificity and (2) facile visualization of tumor regions. Finally, we develop a low-cost, flexible, fiber-based handheld qOBM device which brings this technology one step closer to in-vivo clinical use. This work has significant implications for guiding neurosurgery by paving the way for a tool that delivers real-time, label-free, in-vivo brain tumor margin detection.
Collapse
Affiliation(s)
- Paloma Casteleiro Costa
- School of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Zhe Guang
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Patrick Ledwig
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Zhaobin Zhang
- Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Stewart Neill
- Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
- Department of Pathology & Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jeffrey J. Olson
- Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Francisco E. Robles
- School of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
- Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
57
|
Hasbum A, Quintanilla J, Jr JA, Ding MH, Levy A, Chew SA. Strategies to better treat glioblastoma: antiangiogenic agents and endothelial cell targeting agents. Future Med Chem 2021; 13:393-418. [PMID: 33399488 PMCID: PMC7888526 DOI: 10.4155/fmc-2020-0289] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 11/26/2020] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most prevalent and aggressive form of glioma, with poor prognosis and high mortality rates. As GBM is a highly vascularized cancer, antiangiogenic therapies to halt or minimize the rate of tumor growth are critical to improving treatment. In this review, antiangiogenic therapies, including small-molecule drugs, nucleic acids and proteins and peptides, are discussed. The authors further explore biomaterials that have been utilized to increase the bioavailability and bioactivity of antiangiogenic factors for better antitumor responses in GBM. Finally, the authors summarize the current status of biomaterial-based targeting moieties that target endothelial cells in GBM to more efficiently deliver therapeutics to these cells and avoid off-target cell or organ side effects.
Collapse
Affiliation(s)
- Asbiel Hasbum
- School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX 78541, USA
| | - Jaqueline Quintanilla
- Department of Health & Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX 78526, USA
| | - Juan A Amieva Jr
- Department of Health & Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX 78526, USA
| | - May-Hui Ding
- Department of Health & Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX 78526, USA
| | - Arkene Levy
- Dr Kiran C Patel College of Allopathic Medicine, Nova Southeastern University, FL 33314, USA
| | - Sue Anne Chew
- Department of Health & Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX 78526, USA
| |
Collapse
|
58
|
Wang JL, Barth RF, Cavaliere R, Puduvalli VK, Giglio P, Lonser RR, Elder JB. Phase I trial of intracerebral convection-enhanced delivery of carboplatin for treatment of recurrent high-grade gliomas. PLoS One 2020; 15:e0244383. [PMID: 33373402 PMCID: PMC7771668 DOI: 10.1371/journal.pone.0244383] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 12/07/2020] [Indexed: 12/16/2022] Open
Abstract
Background Carboplatin is a potent cytoreductive agent for a variety of solid tumors. However, when delivered systemically, clinical efficacy for the treatment of high grade gliomas is poor due to limited penetration across the blood-brain barrier (BBB). Direct intracerebral (IC) convection-enhanced delivery (CED) of carboplatin has been used to bypass the BBB and successfully treat the F98 rat glioma. Based on these studies, we initiated a Phase I clinical trial. Objective This Phase I clinical trial was conducted to establish the maximum tolerated dose and define the toxicity profile of carboplatin delivered intracerebrally via convection enhanced delivery (CED) for patients with high grade glial neoplasms. Methods Cohorts of 3 patients with recurrent WHO grade III or IV gliomas were treated with escalating doses of CED carboplatin (1–4 μg in 54mL over 72 hours) delivered via catheters placed at the time of recurrent tumor resection. The primary outcome measure was determination of the maximum tolerated dose (MTD). Secondary outcome measures included overall survival (OS), progression-free survival (PFS), and radiographic correlation. Results A total of 10 patients have completed treatment with infusion doses of carboplatin of 1μg, 2μg, and 4μg. The total planned volume of infusion was 54mL for each patient. All patients had previously received surgery and chemoradiation. Histology at treatment include GBM (n = 9) and anaplastic oligodendroglioma (n = 1). Median KPS was 90 (range, 70 to 100) at time of treatment. Median PFS and OS were 2.1 and 9.6 months after completion of CED, respectively. A single adverse event possibly related to treatment was noted (generalized seizure). Conclusions IC CED of carboplatin as a potential therapy for recurrent malignant glioma is feasible and safe at doses up to 4μg in 54mL over 72 hours. Further studies are needed to determine the maximum tolerated dose and potential efficacy.
Collapse
Affiliation(s)
- Joshua L. Wang
- Department of Neurological Surgery, The Ohio State University College of Medicine Wexner Medical Center, Columbus, Ohio, United States of America
- * E-mail:
| | - Rolf F. Barth
- Department of Pathology, The Ohio State University College of Medicine Wexner Medical Center, Columbus, Ohio, United States of America
| | - Robert Cavaliere
- Division of Neuro-Oncology, Department of Neurology, The Ohio State University College of Medicine Wexner Medical Center, Columbus, Ohio, United States of America
| | - Vinay K. Puduvalli
- Division of Neuro-Oncology, Department of Neurology, The Ohio State University College of Medicine Wexner Medical Center, Columbus, Ohio, United States of America
| | - Pierre Giglio
- Division of Neuro-Oncology, Department of Neurology, The Ohio State University College of Medicine Wexner Medical Center, Columbus, Ohio, United States of America
| | - Russell R. Lonser
- Department of Neurological Surgery, The Ohio State University College of Medicine Wexner Medical Center, Columbus, Ohio, United States of America
| | - J. Bradley Elder
- Department of Neurological Surgery, The Ohio State University College of Medicine Wexner Medical Center, Columbus, Ohio, United States of America
| |
Collapse
|
59
|
Arru C, Serra E, Porcu C, Gadau SD. Confocal investigation on colocalization between tubulin posttranslational modifications and associated proteins in rat C6 glioma cells. J Struct Biol 2020; 213:107676. [PMID: 33279655 DOI: 10.1016/j.jsb.2020.107676] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/27/2020] [Accepted: 11/28/2020] [Indexed: 01/29/2023]
Abstract
Glioblastoma multiforme is the most lethal brain tumor. In the study of mechanisms underlying its development attention has been paid to the microtubular network of its cells, mainly on βIII tubulin, considered as a marker of malignancy. In the present work, we chose to investigate the tubulin code in glioblastoma cells, analyzing the degree of interaction between tubulin post-translational modifications and different proteins associated with them. The pattern of diverse associated proteins such as EB-1, CLIP-170 and kinesin-1 and their degree of co-distribution with the most abundant post-translational tubulin modifications (tyrosination, acetylation and polyglutamylation) were evaluated. Through immunofluorescence we have shown that EB-1, CLIP-170 and kinesin-1 were well detectable in glioblastoma cells. The double fluorescence and colocalization index between the post-translational modifications of tubulin and associated proteins showed that tyrosinated α-tubulin has significantly high affinity with EB-1, CLIP-170 and kinesin-1, while for acetylated and polyglutamylated tubulin, the degree of interaction with the three associated proteins evaluated was less apparent. Data presented in this paper underline the importance of a thorough analysis of the microtubular mechanics in glioblastoma cells. This may suggest new experimental therapeutic approaches able to act more selectively on the microtubular network of cells in this type of cancer.
Collapse
Affiliation(s)
- Caterina Arru
- Department of Veterinary Medicine, University of Sassari, Italy
| | - Elisa Serra
- Department of Veterinary Medicine, University of Sassari, Italy
| | - Cristian Porcu
- Department of Veterinary Medicine, University of Sassari, Italy
| | - Sergio D Gadau
- Department of Veterinary Medicine, University of Sassari, Italy.
| |
Collapse
|
60
|
Fukuo Y, Hattori Y, Kawabata S, Kashiwagi H, Kanemitsu T, Takeuchi K, Futamura G, Hiramatsu R, Watanabe T, Hu N, Takata T, Tanaka H, Suzuki M, Miyatake SI, Kirihata M, Wanibuchi M. The Therapeutic Effects of Dodecaborate Containing Boronophenylalanine for Boron Neutron Capture Therapy in a Rat Brain Tumor Model. BIOLOGY 2020; 9:biology9120437. [PMID: 33271972 PMCID: PMC7759915 DOI: 10.3390/biology9120437] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 11/26/2020] [Accepted: 11/29/2020] [Indexed: 11/22/2022]
Abstract
Simple Summary We have developed a new boron compound for application in boron neutron capture therapy (BNCT) named boronophenylalanine–amide alkyl dodecaborate (BADB). It is characterized by a larger amount of 10B per molecule, linking boronphenylalanine (BPA) and dodecaborate, and we conducted various experiments on its efficacy. Its high accumulation at the cellular level made it a promising novel drug, but it did not sufficiently accumulate in brain tumor tissue when intravenously administered. However, in neutron irradiation experiments, the drug showed remarkably high compound biological effectiveness and significantly prolonged the survival time in rat brain tumor models. We confirmed the antitumor efficacy of BADB in BNCT and its additional efficacy when administered in combination with BPA. Though this drug showed poor results when administered as a single agent, it was superior to BPA alone when administered in combination with BPA, making it a drug that we have been waiting for in our clinical practice. Abstract Background: The development of effective boron compounds is a major area of research in the study of boron neutron capture therapy (BNCT). We created a novel boron compound, boronophenylalanine–amide alkyl dodecaborate (BADB), for application in BNCT and focused on elucidating how it affected a rat brain tumor model. Methods: The boron concentration of F98 rat glioma cells following exposure to boronophenylalanine (BPA) (which is currently being utilized clinically) and BADB was evaluated, and the biodistributions in F98 glioma-bearing rats were assessed. In neutron irradiation studies, the in vitro cytotoxicity of each boron compound and the in vivo corresponding therapeutic effect were evaluated in terms of survival time. Results: The survival fractions of the groups irradiated with BPA and BADB were not significantly different. BADB administered for 6 h after the termination of convection-enhanced delivery ensured the highest boron concentration in the tumor (45.8 μg B/g). The median survival time in the BADB in combination with BPA group showed a more significant prolongation of survival than that of the BPA group. Conclusion: BADB is a novel boron compound for BNCT that triggers a prolonged survival effect in patients receiving BNCT.
Collapse
Affiliation(s)
- Yusuke Fukuo
- Department of Neurosurgery, Osaka Medical College, 2-7 Daigaku-machi, Takatsuki-shi, Osaka 569-8686, Japan; (Y.F.); (H.K.); (T.K.); (K.T.); (G.F.); (R.H.); (M.W.)
| | - Yoshihide Hattori
- Research Center of Boron Neutron Capture Therapy, Research Organization for the 21st Century, Osaka Prefecture University, 1-1 Gakuen-cho, Nakaku, Sakai-shi, Osaka 599-8531, Japan; (Y.H.); (M.K.)
| | - Shinji Kawabata
- Department of Neurosurgery, Osaka Medical College, 2-7 Daigaku-machi, Takatsuki-shi, Osaka 569-8686, Japan; (Y.F.); (H.K.); (T.K.); (K.T.); (G.F.); (R.H.); (M.W.)
- Correspondence: ; Tel.: +81-72-683-1221
| | - Hideki Kashiwagi
- Department of Neurosurgery, Osaka Medical College, 2-7 Daigaku-machi, Takatsuki-shi, Osaka 569-8686, Japan; (Y.F.); (H.K.); (T.K.); (K.T.); (G.F.); (R.H.); (M.W.)
| | - Takuya Kanemitsu
- Department of Neurosurgery, Osaka Medical College, 2-7 Daigaku-machi, Takatsuki-shi, Osaka 569-8686, Japan; (Y.F.); (H.K.); (T.K.); (K.T.); (G.F.); (R.H.); (M.W.)
| | - Koji Takeuchi
- Department of Neurosurgery, Osaka Medical College, 2-7 Daigaku-machi, Takatsuki-shi, Osaka 569-8686, Japan; (Y.F.); (H.K.); (T.K.); (K.T.); (G.F.); (R.H.); (M.W.)
| | - Gen Futamura
- Department of Neurosurgery, Osaka Medical College, 2-7 Daigaku-machi, Takatsuki-shi, Osaka 569-8686, Japan; (Y.F.); (H.K.); (T.K.); (K.T.); (G.F.); (R.H.); (M.W.)
| | - Ryo Hiramatsu
- Department of Neurosurgery, Osaka Medical College, 2-7 Daigaku-machi, Takatsuki-shi, Osaka 569-8686, Japan; (Y.F.); (H.K.); (T.K.); (K.T.); (G.F.); (R.H.); (M.W.)
| | - Tsubasa Watanabe
- Institute for Integrated Radiation and Nuclear Science, Kyoto University, 2 Asashiro-Nishi, Kumatori-cho, Sennan-gun, Osaka 590-0494, Japan; (T.W.); (T.T.); (H.T.); (M.S.)
| | - Naonori Hu
- Kansai BNCT Medical Center, Osaka Medical College, 2-7 Daigaku-machi, Takatsuki-shi, Osaka 569-8686, Japan; (N.H.); (S.-I.M.)
| | - Takushi Takata
- Institute for Integrated Radiation and Nuclear Science, Kyoto University, 2 Asashiro-Nishi, Kumatori-cho, Sennan-gun, Osaka 590-0494, Japan; (T.W.); (T.T.); (H.T.); (M.S.)
| | - Hiroki Tanaka
- Institute for Integrated Radiation and Nuclear Science, Kyoto University, 2 Asashiro-Nishi, Kumatori-cho, Sennan-gun, Osaka 590-0494, Japan; (T.W.); (T.T.); (H.T.); (M.S.)
| | - Minoru Suzuki
- Institute for Integrated Radiation and Nuclear Science, Kyoto University, 2 Asashiro-Nishi, Kumatori-cho, Sennan-gun, Osaka 590-0494, Japan; (T.W.); (T.T.); (H.T.); (M.S.)
| | - Shin-Ichi Miyatake
- Kansai BNCT Medical Center, Osaka Medical College, 2-7 Daigaku-machi, Takatsuki-shi, Osaka 569-8686, Japan; (N.H.); (S.-I.M.)
| | - Mitsunori Kirihata
- Research Center of Boron Neutron Capture Therapy, Research Organization for the 21st Century, Osaka Prefecture University, 1-1 Gakuen-cho, Nakaku, Sakai-shi, Osaka 599-8531, Japan; (Y.H.); (M.K.)
| | - Masahiko Wanibuchi
- Department of Neurosurgery, Osaka Medical College, 2-7 Daigaku-machi, Takatsuki-shi, Osaka 569-8686, Japan; (Y.F.); (H.K.); (T.K.); (K.T.); (G.F.); (R.H.); (M.W.)
| |
Collapse
|
61
|
Mao J, Cao M, Zhang F, Zhang J, Duan X, Lu L, Yang Z, Zhang X, Zhu W, Zhang Q, Wang Z, Shen J. Peritumoral administration of IFNβ upregulated mesenchymal stem cells inhibits tumor growth in an orthotopic, immunocompetent rat glioma model. J Immunother Cancer 2020; 8:jitc-2019-000164. [PMID: 32169868 PMCID: PMC7069318 DOI: 10.1136/jitc-2019-000164] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2020] [Indexed: 12/16/2022] Open
Abstract
Background Immunotherapy with IFNβ is a promising strategy for treating malignant glioma. However, systemic administration of IFNβ is inadequate because of low intratumoral concentration and major adverse effects. This study aimed to determine whether mesenchymal stem cells (MSCs) can be used as cellular vehicles to locally deliver IFNβ for glioma therapy by using in vivo MRI tracking. Methods A recombinant lentiviral vector encoding IFNβ and ferritin heavy chain (FTH) reporter genes was constructed to transduce MSCs. The effectiveness and safety of transduction were assessed. After the IFNβ and FTH overexpressed MSCs (IFNβ-FTH-MSCs) were transplanted into intracranial orthotopic rat F98 gliomas via peritumoral, intracerebral, intratumoral or intra-arterial injection, MRI was performed to track IFNβ-FTH-MSCs and to evaluate their therapeutic effect on glioma in vivo, as validated by histologic analysis, quantitative PCR and ELISA assays. Results MSCs were efficiently and safely transduced to upregulate their IFNβ secretion and FTH expression by the constructed lentivirus. After peritumoral injection, IFNβ-FTH-MSCs appeared as hypointense signals on MRI, which gradually diminished but remained visible until 11 days. Compared with other administration routes, only peritumoral injection of IFNβ-FTH-MSCs showed a remarkable inhibition on the glioma growth. Nearly 30% of IFNβ-FTH-MSCs survived up to 11 days after peritumoral injection, while most of IFNβ-FTH-MSCs injected via other routes died within 11 days. IFNβ-FTH-MSCs grafted peritumorally secreted IFNβ persistently, leading to pronounced Batf3+ dendritic cells and CD8+ T lymphocyte infiltration within the glioma. Conclusions MSCs can be used as cellular vehicles of IFNβ to treat malignant glioma effectively via peritumoral injection.
Collapse
Affiliation(s)
- Jiaji Mao
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Minghui Cao
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Fang Zhang
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jingzhong Zhang
- The Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu, China
| | - Xiaohui Duan
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Liejing Lu
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Zehong Yang
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Xiang Zhang
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Wangshu Zhu
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Qinyuan Zhang
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Zhe Wang
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jun Shen
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China .,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| |
Collapse
|
62
|
Lesbats C, Kelly CL, Czanner G, Poptani H. Diffusion kurtosis imaging for characterizing tumor heterogeneity in an intracranial rat glioblastoma model. NMR IN BIOMEDICINE 2020; 33:e4386. [PMID: 32729637 DOI: 10.1002/nbm.4386] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 07/14/2020] [Accepted: 07/15/2020] [Indexed: 06/11/2023]
Abstract
The utility of diffusion kurtosis imaging (DKI) for assessing intra-tumor heterogeneity was evaluated in a rat model of glioblastoma multiforme. Longitudinal MRI including T2 -weighted and diffusion-weighted MRI (DWI) was performed on six female Fischer rats 8, 11 and 14 days after intracranial transplantation of F98 cells. T2 -weighted images were used to measure the tumor volumes and DWI images were used to compute diffusion tensor imaging (DTI) and DWI based parametric maps including mean diffusivity (MD), mean kurtosis (MK), axial diffusivity (AD), axial kurtosis, radial diffusivity, radial kurtosis, fractional anisotropy (FA) and kurtosis fractional anisotropy (KFA). Median values from the segmented normal contralateral cortex, tumor and edema from the diffusion parameters were compared at the three imaging time points to assess any changes in tumor heterogeneity over time. ex vivo DKI was also performed in a representative sample and compared with histology. Significant differences were observed between normal cortex, tumor and edema in both the DTI and DKI parameters. Notably, at the earliest time point MK and KFA were significantly different between normal cortex and tumor in comparison with MD or FA. Although a decreasing trend in MD, AD and FA values of the tumor were observed as the tumor grew, no significant changes in any of the DTI or DKI parameters were observed longitudinally. While DKI was equally sensitive to DTI in differentiating tumor from edema and normal brain, it was unable to detect longitudinal increases in intra-tumoral heterogeneity in the F98 model of glioblastoma multiforme.
Collapse
Affiliation(s)
- Clémentine Lesbats
- Centre for Preclinical Imaging, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Claire Louise Kelly
- Centre for Preclinical Imaging, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Gabriela Czanner
- Department of Applied Mathematics, Liverpool John Moores University, Liverpool, UK
| | - Harish Poptani
- Centre for Preclinical Imaging, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| |
Collapse
|
63
|
Dolganova IN, Aleksandrova PV, Nikitin PV, Alekseeva AI, Chernomyrdin NV, Musina GR, Beshplav ST, Reshetov IV, Potapov AA, Kurlov VN, Tuchin VV, Zaytsev KI. Capability of physically reasonable OCT-based differentiation between intact brain tissues, human brain gliomas of different WHO grades, and glioma model 101.8 from rats. BIOMEDICAL OPTICS EXPRESS 2020; 11:6780-6798. [PMID: 33282523 PMCID: PMC7687948 DOI: 10.1364/boe.409692] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/19/2020] [Accepted: 10/22/2020] [Indexed: 05/17/2023]
Abstract
Optical coherence tomography (OCT) of the ex vivo rat and human brain tissue samples is performed. The set of samples comprises intact white and gray matter, as well as human brain gliomas of the World Health Organization (WHO) Grades I-IV and glioma model 101.8 from rats. Analysis of OCT signals is aimed at comparing the physically reasonable properties of tissues, and determining the attenuation coefficient, parameter related to effective refractive index, and their standard deviations. Data analysis is based on the linear discriminant analysis and estimation of their dispersion in a four-dimensional principal component space. The results demonstrate the distinct contrast between intact tissues and low-grade gliomas and moderate contrast between intact tissues and high-grade gliomas. Particularly, the mean values of attenuation coefficient are 7.56±0.91, 3.96±0.98, and 5.71±1.49 mm-1 for human white matter, glioma Grade I, and glioblastoma, respectively. The significant variability of optical properties of high Grades and essential differences between rat and human brain tissues are observed. The dispersion of properties enlarges with increase of the glioma WHO Grade, which can be attributed to the growing heterogeneity of pathological brain tissues. The results of this study reveal the advantages and drawbacks of OCT for the intraoperative diagnosis of brain gliomas and compare its abilities separately for different grades of malignancy. The perspective of OCT to differentiate low-grade gliomas is highlighted by the low performance of the existing intraoperational methods and instruments.
Collapse
Affiliation(s)
- I. N. Dolganova
- Institute of Solid State Physics of the Russian Academy of Sciences, Chernogolovka 142432, Russia
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University (Sechenov University), Moscow 119991, Russia
| | - P. V. Aleksandrova
- Institute of Solid State Physics of the Russian Academy of Sciences, Chernogolovka 142432, Russia
| | - P. V. Nikitin
- Burdenko Neurosurgery Institute, Moscow 125047, Russia
- Prokhorov General Physics Institute of the Russian Academy of Sciences, Moscow 119991, Russia
| | - A. I. Alekseeva
- Institute of Solid State Physics of the Russian Academy of Sciences, Chernogolovka 142432, Russia
- Research Institute of Human Morphology, Moscow 117418, Russia
| | - N. V. Chernomyrdin
- Prokhorov General Physics Institute of the Russian Academy of Sciences, Moscow 119991, Russia
| | - G. R. Musina
- Prokhorov General Physics Institute of the Russian Academy of Sciences, Moscow 119991, Russia
| | - S. T. Beshplav
- Burdenko Neurosurgery Institute, Moscow 125047, Russia
- Prokhorov General Physics Institute of the Russian Academy of Sciences, Moscow 119991, Russia
| | - I. V. Reshetov
- Institute for Cluster Oncology, Sechenov First Moscow State Medical University (Sechenov University), Moscow 119991, Russia
- Academy of Postgraduate Education FSCC FMBA, Moscow 125310, Russia
| | - A. A. Potapov
- Burdenko Neurosurgery Institute, Moscow 125047, Russia
| | - V. N. Kurlov
- Institute of Solid State Physics of the Russian Academy of Sciences, Chernogolovka 142432, Russia
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University (Sechenov University), Moscow 119991, Russia
| | - V. V. Tuchin
- Saratov State University, Saratov 410012, Russia
- Institute of Precision Mechanics and Control of the Russian Academy of Sciences, Saratov 410028, Russia
- Tomsk State University, Tomsk 634050, Russia
| | - K. I. Zaytsev
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University (Sechenov University), Moscow 119991, Russia
- Prokhorov General Physics Institute of the Russian Academy of Sciences, Moscow 119991, Russia
| |
Collapse
|
64
|
Kleemiss F, Justies A, Duvinage D, Watermann P, Ehrke E, Sugimoto K, Fugel M, Malaspina LA, Dittmer A, Kleemiss T, Puylaert P, King NR, Staubitz A, Tzschentke TM, Dringen R, Grabowsky S, Beckmann J. Sila-Ibuprofen. J Med Chem 2020; 63:12614-12622. [PMID: 32931274 DOI: 10.1021/acs.jmedchem.0c00813] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The synthesis, characterization, biological activity, and toxicology of sila-ibuprofen, a silicon derivative of the most common nonsteroidal anti-inflammatory drug, is reported. The key improvements compared with ibuprofen are a four times higher solubility in physiological media and a lower melting enthalpy, which are attributed to the carbon-silicon switch. The improved solubility is of interest for postsurgical intravenous administration. A potential for pain relief is rationalized via inhibition experiments of cyclooxygenases I and II (COX-I and COX-II) as well as via a set of newly developed methods that combine molecular dynamics, quantum chemistry, and quantum crystallography. The binding affinity of sila-ibuprofen to COX-I and COX-II is quantified in terms of London dispersion and electrostatic interactions in the active receptor site. This study not only shows the potential of sila-ibuprofen for medicinal application but also improves our understanding of the mechanism of action of the inhibition process.
Collapse
Affiliation(s)
- Florian Kleemiss
- University of Bremen, Institute for Inorganic Chemistry and Crystallography, Leobener Str. 3 and 7, 28359 Bremen, Germany.,University of Bern, Department of Chemistry and Biochemistry, Freiestrasse 3, 3012 Bern, Switzerland
| | - Aileen Justies
- Free University of Berlin, Institute of Chemistry and Biochemistry, Fabeckstr. 34-36, 14195 Berlin, Germany
| | - Daniel Duvinage
- University of Bremen, Institute for Inorganic Chemistry and Crystallography, Leobener Str. 3 and 7, 28359 Bremen, Germany
| | - Patrick Watermann
- University of Bremen, Center for Biomolecular Interactions Bremen and Center for Environmental Research and Sustainable Technology, Leobener Str. 5, 28359 Bremen, Germany
| | - Eric Ehrke
- University of Bremen, Center for Biomolecular Interactions Bremen and Center for Environmental Research and Sustainable Technology, Leobener Str. 5, 28359 Bremen, Germany
| | - Kunihisa Sugimoto
- Japan Synchrotron Radiation Research Institute (JASRI), Diffraction & Scattering Division, 1-1-1 Kouto, Sayo-cho, Sayo-gun, Hyogo 679-5198, Japan.,Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Yoshida-Ushinomiya-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Malte Fugel
- University of Bremen, Institute for Inorganic Chemistry and Crystallography, Leobener Str. 3 and 7, 28359 Bremen, Germany
| | - Lorraine A Malaspina
- University of Bremen, Institute for Inorganic Chemistry and Crystallography, Leobener Str. 3 and 7, 28359 Bremen, Germany.,University of Bern, Department of Chemistry and Biochemistry, Freiestrasse 3, 3012 Bern, Switzerland
| | - Anneke Dittmer
- University of Bremen, Institute for Inorganic Chemistry and Crystallography, Leobener Str. 3 and 7, 28359 Bremen, Germany
| | - Torsten Kleemiss
- University of Bremen, Institute for Inorganic Chemistry and Crystallography, Leobener Str. 3 and 7, 28359 Bremen, Germany
| | - Pim Puylaert
- University of Bremen, Institute for Inorganic Chemistry and Crystallography, Leobener Str. 3 and 7, 28359 Bremen, Germany
| | - Nelly R King
- Free University of Berlin, Institute of Chemistry and Biochemistry, Fabeckstr. 34-36, 14195 Berlin, Germany
| | - Anne Staubitz
- University of Bremen, Institute for Analytical and Organic Chemistry, Leobener Str. 7, 28359 Bremen, Germany
| | | | - Ralf Dringen
- University of Bremen, Center for Biomolecular Interactions Bremen and Center for Environmental Research and Sustainable Technology, Leobener Str. 5, 28359 Bremen, Germany
| | - Simon Grabowsky
- University of Bremen, Institute for Inorganic Chemistry and Crystallography, Leobener Str. 3 and 7, 28359 Bremen, Germany.,University of Bern, Department of Chemistry and Biochemistry, Freiestrasse 3, 3012 Bern, Switzerland
| | - Jens Beckmann
- University of Bremen, Institute for Inorganic Chemistry and Crystallography, Leobener Str. 3 and 7, 28359 Bremen, Germany.,Free University of Berlin, Institute of Chemistry and Biochemistry, Fabeckstr. 34-36, 14195 Berlin, Germany
| |
Collapse
|
65
|
Bouckaert C, Germonpré C, Verhoeven J, Chong SA, Jacquin L, Mairet-Coello G, André VM, Leclercq K, Vanhove C, De Vos F, Van den Broecke C, Goethals I, Descamps B, Donche S, Carrette E, Wadman W, Boon P, Vonck K, Raedt R. Development of a Rat Model for Glioma-Related Epilepsy. Int J Mol Sci 2020; 21:E6999. [PMID: 32977526 PMCID: PMC7582710 DOI: 10.3390/ijms21196999] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/18/2020] [Accepted: 09/21/2020] [Indexed: 12/13/2022] Open
Abstract
Seizures are common in patients with high-grade gliomas (30-60%) and approximately 15-30% of glioblastoma (GB) patients develop drug-resistant epilepsy. Reliable animal models are needed to develop adequate treatments for glioma-related epilepsy. Therefore, fifteen rats were inoculated with F98 GB cells (GB group) and four rats with vehicle only (control group) in the right entorhinal cortex. MRI was performed to visualize tumor presence. A subset of seven GB and two control rats were implanted with recording electrodes to determine the occurrence of epileptic seizures with video-EEG recording over multiple days. In a subset of rats, tumor size and expression of tumor markers were investigated with histology or mRNA in situ hybridization. Tumors were visible on MRI six days post-inoculation. Time-dependent changes in tumor morphology and size were visible on MRI. Epileptic seizures were detected in all GB rats monitored with video-EEG. Twenty-one days after inoculation, rats were euthanized based on signs of discomfort and pain. This study describes, for the first time, reproducible tumor growth and spontaneous seizures upon inoculation of F98 cells in the rat entorhinal cortex. The development of this new model of GB-related epilepsy may be valuable to design new therapies against tumor growth and associated epileptic seizures.
Collapse
Affiliation(s)
- Charlotte Bouckaert
- 4Brain, Department of Head and Skin, Ghent University, 9000 Ghent, East Flanders, Belgium; (C.B.); (C.G.); (E.C.); (W.W.); (P.B.); (K.V.)
| | - Charlotte Germonpré
- 4Brain, Department of Head and Skin, Ghent University, 9000 Ghent, East Flanders, Belgium; (C.B.); (C.G.); (E.C.); (W.W.); (P.B.); (K.V.)
| | - Jeroen Verhoeven
- Department of Pharmaceutical Analysis, Ghent University, 9000 Ghent, East Flanders, Belgium; (J.V.); (F.D.V.); (C.V.d.B.)
| | - Seon-Ah Chong
- Early Solutions, Neuroscience Therapeutic Area, UCB Pharma, 1420 Braine-l’Alleud, Brabant Wallon, Belgium; (S.-A.C.); (L.J.); (G.M.-C.); (V.M.A.); (K.L.)
| | - Lucas Jacquin
- Early Solutions, Neuroscience Therapeutic Area, UCB Pharma, 1420 Braine-l’Alleud, Brabant Wallon, Belgium; (S.-A.C.); (L.J.); (G.M.-C.); (V.M.A.); (K.L.)
| | - Georges Mairet-Coello
- Early Solutions, Neuroscience Therapeutic Area, UCB Pharma, 1420 Braine-l’Alleud, Brabant Wallon, Belgium; (S.-A.C.); (L.J.); (G.M.-C.); (V.M.A.); (K.L.)
| | - Véronique Marie André
- Early Solutions, Neuroscience Therapeutic Area, UCB Pharma, 1420 Braine-l’Alleud, Brabant Wallon, Belgium; (S.-A.C.); (L.J.); (G.M.-C.); (V.M.A.); (K.L.)
| | - Karine Leclercq
- Early Solutions, Neuroscience Therapeutic Area, UCB Pharma, 1420 Braine-l’Alleud, Brabant Wallon, Belgium; (S.-A.C.); (L.J.); (G.M.-C.); (V.M.A.); (K.L.)
| | - Christian Vanhove
- Department of Electronics and information systems, Ghent University Hospital, 9000 Ghent, East Flanders, Belgium; (C.V.); (B.D.)
| | - Filip De Vos
- Department of Pharmaceutical Analysis, Ghent University, 9000 Ghent, East Flanders, Belgium; (J.V.); (F.D.V.); (C.V.d.B.)
| | - Caroline Van den Broecke
- Department of Pharmaceutical Analysis, Ghent University, 9000 Ghent, East Flanders, Belgium; (J.V.); (F.D.V.); (C.V.d.B.)
| | - Ingeborg Goethals
- Department of Diagnostic Sciences, Ghent University Hospital, 9000 Ghent, East Flanders, Belgium; (I.G.); (S.D.)
| | - Benedicte Descamps
- Department of Electronics and information systems, Ghent University Hospital, 9000 Ghent, East Flanders, Belgium; (C.V.); (B.D.)
| | - Sam Donche
- Department of Diagnostic Sciences, Ghent University Hospital, 9000 Ghent, East Flanders, Belgium; (I.G.); (S.D.)
| | - Evelien Carrette
- 4Brain, Department of Head and Skin, Ghent University, 9000 Ghent, East Flanders, Belgium; (C.B.); (C.G.); (E.C.); (W.W.); (P.B.); (K.V.)
| | - Wytse Wadman
- 4Brain, Department of Head and Skin, Ghent University, 9000 Ghent, East Flanders, Belgium; (C.B.); (C.G.); (E.C.); (W.W.); (P.B.); (K.V.)
| | - Paul Boon
- 4Brain, Department of Head and Skin, Ghent University, 9000 Ghent, East Flanders, Belgium; (C.B.); (C.G.); (E.C.); (W.W.); (P.B.); (K.V.)
| | - Kristl Vonck
- 4Brain, Department of Head and Skin, Ghent University, 9000 Ghent, East Flanders, Belgium; (C.B.); (C.G.); (E.C.); (W.W.); (P.B.); (K.V.)
| | - Robrecht Raedt
- 4Brain, Department of Head and Skin, Ghent University, 9000 Ghent, East Flanders, Belgium; (C.B.); (C.G.); (E.C.); (W.W.); (P.B.); (K.V.)
| |
Collapse
|
66
|
Taga T, Tabu K. Glioma progression and recurrence involving maintenance and expansion strategies of glioma stem cells by organizing self-advantageous niche microenvironments. Inflamm Regen 2020; 40:33. [PMID: 32952746 PMCID: PMC7493875 DOI: 10.1186/s41232-020-00142-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 09/04/2020] [Indexed: 12/16/2022] Open
Abstract
Due to the nature of enhanced resistance to conventional chemo/radiotherapies and metastasis, highly tumorigenic cancer stem cells (CSCs) have been proposed as a promising target for cancer eradication. To tackle the therapeutic difficulties of cancers involving CSCs, extensive research efforts have been directed toward understanding the extracellular microenvironments of CSCs, i.e., CSC niche, which plays important roles in CSC maintenance and expansion. Here we review recently identified mechanisms of maintenance and expansion of glioma CSCs (GSCs) leading to glioma progression and recurrence, with particular emphasis on the reports made by studies with a unique approach using polymer microarrays screening and with a unique viewpoint of necrotic particles. The polymer-based approach identified two groups of niche components, extracellular matrices (ECMs) and iron, and uncovered that co-expression of ECM-, iron-, and macrophage-related genes is predictive of glioma patients' outcome. The study in view of a unique fraction of GSC-derived necrotic particles proposed that such particles develop GSC-supportive tumor-associated macrophages (TAMs). Taken together, these studies provide new insights into the mechanisms underlying GSC-driven niche development, i.e., organization of the self-advantageous niche microenvironments for GSC maintenance and expansion leading to glioma progression and recurrence. A series of such studies can redefine the current concept of anti-GSC niche therapy that targets ligands/receptors supporting GSCs, and have potential to accelerate cancer therapy development.
Collapse
Affiliation(s)
- Tetsuya Taga
- Department of Stem Cell Regulation, Medical Research Institute, Tokyo Medical and Dental University (TMDU) , 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8510 Japan
| | - Kouichi Tabu
- Department of Stem Cell Regulation, Medical Research Institute, Tokyo Medical and Dental University (TMDU) , 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8510 Japan
| |
Collapse
|
67
|
Lim H, Martínez-Santiesteban F, Jensen MD, Chen A, Wong E, Scholl TJ. Monitoring Early Changes in Tumor Metabolism in Response to Therapy Using Hyperpolarized 13C MRSI in a Preclinical Model of Glioma. ACTA ACUST UNITED AC 2020; 6:290-300. [PMID: 32879899 PMCID: PMC7442089 DOI: 10.18383/j.tom.2020.00024] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This study shows the use of hyperpolarized 13C magnetic resonance spectroscopic imaging (MRSI) to assess therapeutic efficacy in a preclinical tumor model. 13C-labeled pyruvate was used to monitor early changes in tumor metabolism based on the Warburg effect. High-grade malignant tumors exhibit increased glycolytic activity and lactate production to promote proliferation. A rodent glioma model was used to explore altered lactate production after therapy as an early imaging biomarker for therapeutic response. Rodents were surgically implanted with C6 glioma cells and separated into 4 groups, namely, no therapy, radiotherapy, chemotherapy and combined therapy. Animals were imaged serially at 6 different time points with magnetic resonance imaging at 3 T using hyperpolarized [1-13C]pyruvate MRSI and conventional 1H imaging. Using hyperpolarized [1-13C]pyruvate MRSI, alterations in tumor metabolism were detected as changes in the conversion of lactate to pyruvate (measured as Lac/Pyr ratio) and compared with the conventional method of detecting therapeutic response using the Response Evaluation Criteria in Solid Tumors. Moreover, each therapy group expressed different characteristic changes in tumor metabolism. The group that received no therapy showed a gradual increase of Lac/Pyr ratio within the tumor. The radiotherapy group showed large variations in tumor Lac/Pyr ratio. The chemo- and combined-therapy groups showed a statistically significant reduction in tumor Lac/Pyr ratio; however, only combined therapy was capable of suppressing tumor growth, which resulted in low endpoint mortality rate. Hyperpolarized 13C MRSI detected a prompt reduction in Lac/Pyr ratio as early as 2 days post combined chemo- and radiotherapies.
Collapse
Affiliation(s)
- Heeseung Lim
- Department of Medical Biophysics, Western University, London, ON, Canada
| | | | - Michael D Jensen
- Department of Medical Biophysics, Western University, London, ON, Canada
| | - Albert Chen
- General Electric Healthcare, Toronto, ON, Canada
| | - Eugene Wong
- Department of Medical Biophysics, Western University, London, ON, Canada.,Departments of Physics and Astronomy; Oncology; and Robarts Research Institute, Western University, London, ON, Canada, and.,Departments of Physics and Astronomy; Oncology; and Robarts Research Institute, Western University, London, ON, Canada, and
| | - Timothy J Scholl
- Department of Medical Biophysics, Western University, London, ON, Canada.,Departments of Physics and Astronomy; Oncology; and Robarts Research Institute, Western University, London, ON, Canada, and.,Ontario Institute for Cancer Research, Toronto, ON, Canada
| |
Collapse
|
68
|
Elleaume H, Barth RF, Rousseau J, Bobyk L, Balosso J, Yang W, Huo T, Nakkula R. Radiation therapy combined with intracerebral convection-enhanced delivery of cisplatin or carboplatin for treatment of the F98 rat glioma. J Neurooncol 2020; 149:193-208. [PMID: 32809095 DOI: 10.1007/s11060-020-03600-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 08/08/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND The purpose of this review is to summarize our own experimental studies carried out over a 13-year period of time using the F98 rat glioma as model for high grade gliomas. We evaluated a binary chemo-radiotherapeutic modality that combines either cisplatin (CDDP) or carboplatin, administered intracerebrally (i.c.) by means of convection-enhanced delivery (CED) or osmotic pumps, in combination with either synchrotron or conventional X-irradiation. METHODS F98 glioma cells were implanted stereotactically into the brains of syngeneic Fischer rats. Approximately 14 days later, either CDDP or carboplatin was administered i.c. by CED, followed 24 h later by radiotherapy using either a synchrotron or, subsequently, megavoltage linear accelerators (LINAC). RESULTS CDDP was administered at a dose of 3 µg in 5 µL, followed 24 h later with an irradiation dose of 15 Gy or carboplatin at a dose of 20 µg in 10 µL, followed 24 h later with 3 fractions of 8 Gy each, at the source at the European Synchrotron Radiation Facility (ESRF). This resulted in a median survival time (MeST) > 180 days with 33% long term survivors (LTS) for CDDP and a MeST > 60 days with 8 to 22% LTS, for carboplatin. Subsequently it became apparent that comparable survival data could be obtained with megavoltage X-irradiation using a LINAC source. The best survival data were obtained with a dose of 72 µg of carboplatin administered by means of Alzet® osmotic pumps over 7 days. This resulted in a MeST of > 180 days, with 55% LTS. Histopathologic examination of all the brains of the surviving rats revealed no residual tumor cells or evidence of significant radiation related effects. CONCLUSIONS The results obtained using this combination therapy has, to the best of our knowledge, yielded the most promising survival data ever reported using the F98 glioma model.
Collapse
Affiliation(s)
- Hélène Elleaume
- INSERM UA07 Team STROBE, ESRF, 71 Avenue des Martyrs, 38000, Grenoble, France.
- European Synchrotron Radiation Facility, ID17 Medical Beamline, 71 Avenue Martyrs, 38000, Grenoble, France.
| | - Rolf F Barth
- Department of Pathology, The Ohio State University, 4132 Graves Hall - 333 W. 10th Avenue, Columbus, OH, 43210, USA.
| | - Julia Rousseau
- INSERM UA07 Team STROBE, ESRF, 71 Avenue des Martyrs, 38000, Grenoble, France
- European Synchrotron Radiation Facility, ID17 Medical Beamline, 71 Avenue Martyrs, 38000, Grenoble, France
| | - Laure Bobyk
- INSERM UA07 Team STROBE, ESRF, 71 Avenue des Martyrs, 38000, Grenoble, France
- European Synchrotron Radiation Facility, ID17 Medical Beamline, 71 Avenue Martyrs, 38000, Grenoble, France
| | - Jacques Balosso
- INSERM UA07 Team STROBE, ESRF, 71 Avenue des Martyrs, 38000, Grenoble, France
- Service de Radiothérapie, Centre Hospitalier Universitaire Grenoble-Alpes, 38700, La Tronche, France
- Centre de lutte contre le Cancer F. Baclesse, 3 avenue du général Harris, 14000, Caen, France
| | - Weilian Yang
- Department of Pathology, The Ohio State University, 4132 Graves Hall - 333 W. 10th Avenue, Columbus, OH, 43210, USA
- Department of Neurosurgery, Suzhou Medical College, Suzhou, China
| | - Tianyao Huo
- Department of Pathology, The Ohio State University, 4132 Graves Hall - 333 W. 10th Avenue, Columbus, OH, 43210, USA
- Department of Health Outcomes and Policy, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Robin Nakkula
- Department of Pathology, The Ohio State University, 4132 Graves Hall - 333 W. 10th Avenue, Columbus, OH, 43210, USA
- Research Institute, Nationwide Children's Hospital, Columbus, OH, 43205, USA
| |
Collapse
|
69
|
Synthesis and Evaluation of Dodecaboranethiol Containing Kojic Acid (KA-BSH) as a Novel Agent for Boron Neutron Capture Therapy. Cells 2020; 9:cells9061551. [PMID: 32630612 PMCID: PMC7349888 DOI: 10.3390/cells9061551] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 06/18/2020] [Accepted: 06/23/2020] [Indexed: 11/17/2022] Open
Abstract
Boron neutron capture therapy (BNCT) is a form of tumor-cell selective particle irradiation using low-energy neutron irradiation of boron-10 (10B) to produce high-linear energy transfer (LET) alpha particles and recoiling 7Li nuclei (10B [n, alpha] 7Li) in tumor cells. Therefore, it is important to achieve the selective delivery of large amounts of 10B to tumor cells, with only small amounts of 10B to normal tissues. To develop practical materials utilizing 10B carriers, we designed and synthesized novel dodecaboranethiol (BSH)-containing kojic acid (KA-BSH). In the present study, we evaluated the effects of this novel 10B carrier on cytotoxicity, 10B concentrations in F98 rat glioma cells, and micro-distribution of KA-BSH in vitro. Furthermore, biodistribution studies were performed in a rat brain tumor model. The tumor boron concentrations showed the highest concentrations at 1 h after the termination of administration. Based on these results, neutron irradiation was evaluated at the Kyoto University Research Reactor Institute (KURRI) with KA-BSH. Median survival times (MSTs) of untreated and irradiated control rats were 29.5 and 30.5 days, respectively, while animals that received KA-BSH, followed by neutron irradiation, had an MST of 36.0 days (p = 0.0027, 0.0053). Based on these findings, further studies are warranted in using KA-BSH as a new B compound for malignant glioma.
Collapse
|
70
|
Nery de Albuquerque Rego G, da Hora Alves A, Penteado Nucci M, Bustamante Mamani J, Anselmo de Oliveira F, Gamarra LF. Antiangiogenic Targets for Glioblastoma Therapy from a Pre-Clinical Approach, Using Nanoformulations. Int J Mol Sci 2020; 21:ijms21124490. [PMID: 32599834 PMCID: PMC7349965 DOI: 10.3390/ijms21124490] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 06/12/2020] [Accepted: 06/18/2020] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma (GBM) is the most aggressive tumor type whose resistance to conventional treatment is mediated, in part, by the angiogenic process. New treatments involving the application of nanoformulations composed of encapsulated drugs coupled to peptide motifs that direct drugs to specific targets triggered in angiogenesis have been developed to reach and modulate different phases of this process. We performed a systematic review with the search criterion (Glioblastoma OR Glioma) AND (Therapy OR Therapeutic) AND (Nanoparticle) AND (Antiangiogenic OR Angiogenesis OR Anti-angiogenic) in Pubmed, Scopus, and Cochrane databases, in which 312 articles were identified; of these, only 27 articles were included after selection and analysis of eligibility according to the inclusion and exclusion criteria. The data of the articles were analyzed in five contexts: the characteristics of the tumor cells; the animal models used to induce GBM for antiangiogenic treatment; the composition of nanoformulations and their physical and chemical characteristics; the therapeutic anti-angiogenic process; and methods for assessing the effects on antiangiogenic markers caused by therapies. The articles included in the review were heterogeneous and varied in practically all aspects related to nanoformulations and models. However, there was slight variance in the antiangiogenic effect analysis. CD31 was extensively used as a marker, which does not provide a view of the effects on the most diverse aspects involved in angiogenesis. Therefore, the present review highlighted the need for standardization between the different approaches of antiangiogenic therapy for the GBM model that allows a more effective meta-analysis and that helps in future translational studies.
Collapse
Affiliation(s)
| | - Arielly da Hora Alves
- Hospital Israelita Albert Einstein, São Paulo 05652-900, Brazil; (G.N.d.A.R.); (A.d.H.A.); (J.B.M.); (F.A.d.O.)
| | - Mariana Penteado Nucci
- LIM44-Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo 01246-903, Brazil;
| | - Javier Bustamante Mamani
- Hospital Israelita Albert Einstein, São Paulo 05652-900, Brazil; (G.N.d.A.R.); (A.d.H.A.); (J.B.M.); (F.A.d.O.)
| | | | - Lionel Fernel Gamarra
- Hospital Israelita Albert Einstein, São Paulo 05652-900, Brazil; (G.N.d.A.R.); (A.d.H.A.); (J.B.M.); (F.A.d.O.)
- Correspondence: ; Tel.: +55-11-2151-0243
| |
Collapse
|
71
|
Use of a Luciferase-Expressing Orthotopic Rat Brain Tumor Model to Optimize a Targeted Irradiation Strategy for Efficacy Testing with Temozolomide. Cancers (Basel) 2020; 12:cancers12061585. [PMID: 32549357 PMCID: PMC7352586 DOI: 10.3390/cancers12061585] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 05/29/2020] [Accepted: 06/11/2020] [Indexed: 01/04/2023] Open
Abstract
Glioblastoma multiforme (GBM) is a common and aggressive malignant brain cancer with a mean survival time of approximately 15 months after initial diagnosis. Currently, the standard-of-care (SOC) treatment for this disease consists of radiotherapy (RT) with concomitant and adjuvant temozolomide (TMZ). We sought to develop an orthotopic preclinical model of GBM and to optimize a protocol for non-invasive monitoring of tumor growth, allowing for determination of the efficacy of SOC therapy using a targeted RT strategy combined with TMZ. A strong correlation (r = 0.80) was observed between contrast-enhanced (CE)-CT-based volume quantification and bioluminescent (BLI)-integrated image intensity when monitoring tumor growth, allowing for BLI imaging as a substitute for CE-CT. An optimized parallel-opposed single-angle RT beam plan delivered on average 96% of the expected RT dose (20, 30 or 60 Gy) to the tumor. Normal tissue on the ipsilateral and contralateral sides of the brain were spared 84% and 99% of the expected dose, respectively. An increase in median survival time was demonstrated for all SOC regimens compared to untreated controls (average 5.2 days, p < 0.05), but treatment was not curative, suggesting the need for novel treatment options to increase therapeutic efficacy.
Collapse
|
72
|
Herbener VJ, Burster T, Goreth A, Pruss M, von Bandemer H, Baisch T, Fitzel R, Siegelin MD, Karpel-Massler G, Debatin KM, Westhoff MA, Strobel H. Considering the Experimental use of Temozolomide in Glioblastoma Research. Biomedicines 2020; 8:E151. [PMID: 32512726 PMCID: PMC7344626 DOI: 10.3390/biomedicines8060151] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 05/30/2020] [Accepted: 05/31/2020] [Indexed: 12/17/2022] Open
Abstract
Temozolomide (TMZ) currently remains the only chemotherapeutic component in the approved treatment scheme for Glioblastoma (GB), the most common primary brain tumour with a dismal patient's survival prognosis of only ~15 months. While frequently described as an alkylating agent that causes DNA damage and thus-ultimately-cell death, a recent debate has been initiated to re-evaluate the therapeutic role of TMZ in GB. Here, we discuss the experimental use of TMZ and highlight how it differs from its clinical role. Four areas could be identified in which the experimental data is particularly limited in its translational potential: 1. transferring clinical dosing and scheduling to an experimental system and vice versa; 2. the different use of (non-inert) solvent in clinic and laboratory; 3. the limitations of established GB cell lines which only poorly mimic GB tumours; and 4. the limitations of animal models lacking an immune response. Discussing these limitations in a broader biomedical context, we offer suggestions as to how to improve transferability of data. Finally, we highlight an underexplored function of TMZ in modulating the immune system, as an example of where the aforementioned limitations impede the progression of our knowledge.
Collapse
Affiliation(s)
- Verena J. Herbener
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany; (V.J.H.); (A.G.); (H.v.B.); (T.B.); (R.F.); (K.-M.D.); (H.S.)
| | - Timo Burster
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, Nur-Sultan 010000, Kazakhstan;
| | - Alicia Goreth
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany; (V.J.H.); (A.G.); (H.v.B.); (T.B.); (R.F.); (K.-M.D.); (H.S.)
| | - Maximilian Pruss
- Department of Gynecology and Obstetrics, Medical Faculty, University Hospital of the Heinrich-Heine-University Duesseldorf, D-40225 Duesseldorf, Germany;
- Department of Neurosurgery, University Medical Center Ulm, D-89081 Ulm, Germany;
| | - Hélène von Bandemer
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany; (V.J.H.); (A.G.); (H.v.B.); (T.B.); (R.F.); (K.-M.D.); (H.S.)
| | - Tim Baisch
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany; (V.J.H.); (A.G.); (H.v.B.); (T.B.); (R.F.); (K.-M.D.); (H.S.)
| | - Rahel Fitzel
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany; (V.J.H.); (A.G.); (H.v.B.); (T.B.); (R.F.); (K.-M.D.); (H.S.)
| | - Markus D. Siegelin
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA;
| | - Georg Karpel-Massler
- Department of Neurosurgery, University Medical Center Ulm, D-89081 Ulm, Germany;
| | - Klaus-Michael Debatin
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany; (V.J.H.); (A.G.); (H.v.B.); (T.B.); (R.F.); (K.-M.D.); (H.S.)
| | - Mike-Andrew Westhoff
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany; (V.J.H.); (A.G.); (H.v.B.); (T.B.); (R.F.); (K.-M.D.); (H.S.)
| | - Hannah Strobel
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany; (V.J.H.); (A.G.); (H.v.B.); (T.B.); (R.F.); (K.-M.D.); (H.S.)
| |
Collapse
|
73
|
Prada F, Sheybani N, Franzini A, Moore D, Cordeiro D, Sheehan J, Timbie K, Xu Z. Fluorescein-mediated sonodynamic therapy in a rat glioma model. J Neurooncol 2020; 148:445-454. [PMID: 32500440 DOI: 10.1007/s11060-020-03536-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 05/12/2020] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Malignant gliomas have a dismal prognosis and significant efforts are being made to develop more effective treatments. Sonodynamic therapy (SDT) is an emerging modality for cancer treatment which combines ultrasound with sonosensitizers to produce a localized cytotoxic effect. The aim of this study is to demonstrate the efficacy of SDT with fluorescein (FL) and low-intensity focused ultrasound in inhibiting the growth of ectopic gliomas implanted in the rat's subcutaneous tissue. METHODS In vivo cytotoxicity of FL-SDT was evaluated in C6 rat glioma cells which were inoculated subcutaneously. Tumor specific extracellular FL extravasation and accumulation was assessed with IVIS imaging in rats receiving systemic FL. Effects of FL-SDT with focused low-intensity ultrasound on tumor growth, and histological features of the rat's tumors were investigated. Treatment related apoptosis and necrosis were analyzed using hematoxylin & eosin, and apoptosis-specific staining. RESULTS IVIS imaging revealed a high degree of FL accumulation within the tumor, with a nearly threefold increase in tumoral epifluorescence signal over background. SDT significantly inhibited outgrowth of ectopic C6 gliomas across all three FUS exposure conditions. TUNEL and active caspase-3 staining did not reveal conclusive trends across control and SDT condition for apoptosis. CONCLUSION Our results suggest that SDT with FL and low-intensity FUS is effective in inhibiting the growth of ectopic malignant gliomas in rats. The selective FL extravasation and accumulation in the tumor areas where the blood-brain barrier is damaged suggests the tumor-specificity of the treatment. The possibility to use this treatment in intracranial models and in human gliomas will have to be explored in further studies.
Collapse
Affiliation(s)
- Francesco Prada
- Department of Neurological Surgery, University of Virginia Health Science Center, Charlottesville, VA, USA. .,Department of Neurosurgery, Fondazione IRCCS Istituto Neurologico C. Besta, Milan, Italy. .,Focused Ultrasound Foundation, Charlottesville, VA, USA.
| | - Natasha Sheybani
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Andrea Franzini
- Department of Neurological Surgery, University of Virginia Health Science Center, Charlottesville, VA, USA.,Department of Neurosurgery, Fondazione IRCCS Istituto Neurologico C. Besta, Milan, Italy
| | - David Moore
- Focused Ultrasound Foundation, Charlottesville, VA, USA
| | - Diogo Cordeiro
- Department of Neurological Surgery, University of Virginia Health Science Center, Charlottesville, VA, USA
| | - Jason Sheehan
- Department of Neurological Surgery, University of Virginia Health Science Center, Charlottesville, VA, USA
| | - Kelsie Timbie
- Focused Ultrasound Foundation, Charlottesville, VA, USA
| | - Zhiyuan Xu
- Department of Neurological Surgery, University of Virginia Health Science Center, Charlottesville, VA, USA
| |
Collapse
|
74
|
Blood-brain barrier disruption and delivery of irinotecan in a rat model using a clinical transcranial MRI-guided focused ultrasound system. Sci Rep 2020; 10:8766. [PMID: 32472017 PMCID: PMC7260193 DOI: 10.1038/s41598-020-65617-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 03/17/2020] [Indexed: 11/09/2022] Open
Abstract
We investigated controlled blood-brain barrier (BBB) disruption using a low-frequency clinical transcranial MRI-guided focused ultrasound (TcMRgFUS) device and evaluated enhanced delivery of irinotecan chemotherapy to the brain and a rat glioma model. Animals received three weekly sessions of FUS, FUS and 10 mg/kg irinotecan, or irinotecan alone. In each session, four volumetric sonications targeted 36 locations in one hemisphere. With feedback control based on recordings of acoustic emissions, 98% of the sonication targets (1045/1071) reached a pre-defined level of acoustic emission, while the probability of wideband emission (a signature for inertial cavitation) was than 1%. BBB disruption, evaluated by mapping the R1 relaxation rate after administration of an MRI contrast agent, was significantly higher in the sonicated hemisphere (P < 0.01). Histological evaluation found minimal tissue effects. Irinotecan concentrations in the brain were significantly higher (P < 0.001) with BBB disruption, but SN-38 was only detected in <50% of the samples and only with an excessive irinotecan dose. Irinotecan with BBB disruption did not impede tumor growth or increase survival. Overall these results demonstrate safe and controlled BBB disruption with a low-frequency clinical TcMRgFUS device. While irinotecan delivery to the brain was not neurotoxic, it did not improve outcomes in the F98 glioma model.
Collapse
|
75
|
Liaw K, Zhang F, Mangraviti A, Kannan S, Tyler B, Kannan RM. Dendrimer size effects on the selective brain tumor targeting in orthotopic tumor models upon systemic administration. Bioeng Transl Med 2020; 5:e10160. [PMID: 32440565 PMCID: PMC7237147 DOI: 10.1002/btm2.10160] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 03/26/2020] [Accepted: 04/03/2020] [Indexed: 12/14/2022] Open
Abstract
Malignant gliomas are the most common and aggressive form of primary brain tumors, with a median survival of 15-20 months for patients receiving maximal interventions. Advances in nanomedicine have provided tumor-specific delivery of chemotherapeutics to potentially overcome their off-target toxicities. Recent advances in dendrimer-based nanomedicines have established that hydroxyl-terminated poly(amidoamine) dendrimers can intrinsically target neuroinflammation and brain tumors from systemic administration without the need for targeting moieties. The size of nanocarriers is a critical parameter that determines their tumor-targeting efficiency, intratumor distribution, and clearance mechanism. In this study, we explore the dendrimer size effects on brain tumor targeting capability in two clinically relevant orthotopic brain tumor models, the 9L rat and GL261 mouse models, which capture differing aspects of gliomas. We show that increasing dendrimers from Generation 4 to Generation 6 significantly enhances their tumor accumulation (~10-fold greater at 24 hr), tumor specificity (~2-3 fold higher), and tumor retention. The superior tumor targeting effect of G6 dendrimers is associated with its reduced renal clearance rate, resulting in longer circulation time compared to G4 dendrimers. Additionally, the increase in dendrimer generation does not compromise its homogeneous tumor distribution and intrinsic targeting of tumor-associated macrophages. These results validate the potential for these dendrimers as an effective, clinically translatable platform for effectively targeting tumor-associated macrophages in malignant gliomas.
Collapse
Affiliation(s)
- Kevin Liaw
- Center for NanomedicineWilmer Eye Institute, Johns Hopkins School of MedicineBaltimoreMarylandUSA
- Department of Chemical and Biomolecular EngineeringJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Fan Zhang
- Center for NanomedicineWilmer Eye Institute, Johns Hopkins School of MedicineBaltimoreMarylandUSA
- Department of Materials Science and EngineeringJohns Hopkins UniversityBaltimoreMarylandUSA
| | | | - Sujatha Kannan
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins School of MedicineBaltimoreMarylandUSA
| | - Betty Tyler
- Department of NeurosurgeryJohns Hopkins School of MedicineBaltimoreMarylandUSA
| | - Rangaramanujam M. Kannan
- Center for NanomedicineWilmer Eye Institute, Johns Hopkins School of MedicineBaltimoreMarylandUSA
- Department of Chemical and Biomolecular EngineeringJohns Hopkins UniversityBaltimoreMarylandUSA
- Department of Materials Science and EngineeringJohns Hopkins UniversityBaltimoreMarylandUSA
| |
Collapse
|
76
|
Mang D, Roy SR, Hoh HH, Wu X, Zhang J, Jin C, Zhang Y. Self-Assembly of Integrin Ligands on the Apical Membrane Inhibits the Migration of Glioma Cells. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2020; 36:3750-3757. [PMID: 32191038 DOI: 10.1021/acs.langmuir.0c00291] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Malignant brain cancer remains challenging in treatment due to the highly invasive quality of gliomas. Inspired by the upregulated expression of integrin β1 subunits in tumors, we designed and synthesized an integrin-targeting self-assembling ligand based on a laminin-derived peptide that selectively forms nanofibrous microdomains on the apical membrane of glioma cells, inhibiting their migration and invasion.
Collapse
Affiliation(s)
- Dingze Mang
- Bioinspired Soft Matter Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna son, Okinawa 904-0495, Japan
| | - Sona Rani Roy
- Bioinspired Soft Matter Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna son, Okinawa 904-0495, Japan
| | - Hong Huat Hoh
- Bioinspired Soft Matter Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna son, Okinawa 904-0495, Japan
| | - Xia Wu
- Bioinspired Soft Matter Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna son, Okinawa 904-0495, Japan
| | - Jiahao Zhang
- Bioinspired Soft Matter Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna son, Okinawa 904-0495, Japan
| | - Chengzhi Jin
- Bioinspired Soft Matter Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna son, Okinawa 904-0495, Japan
| | - Ye Zhang
- Bioinspired Soft Matter Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna son, Okinawa 904-0495, Japan
| |
Collapse
|
77
|
Nivajärvi R, Olsson V, Hyppönen V, Bowen S, Leinonen HM, Lesch HP, Ardenkjaer-Larsen JH, Gröhn OHJ, Ylä-Herttuala S, Kettunen MI. Detection of lentiviral suicide gene therapy in C6 rat glioma using hyperpolarised [1- 13 C]pyruvate. NMR IN BIOMEDICINE 2020; 33:e4250. [PMID: 31909530 DOI: 10.1002/nbm.4250] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 12/06/2019] [Accepted: 12/06/2019] [Indexed: 06/10/2023]
Abstract
Hyperpolarised [1-13 C]pyruvate MRI has shown promise in monitoring therapeutic efficacy in a number of cancers including glioma. In this study, we assessed the pyruvate response to the lentiviral suicide gene therapy of herpes simplex virus-1 thymidine kinase with the prodrug ganciclovir (HSV-TK/GCV) in C6 rat glioma and compared it with traditional MR therapy markers. Female Wistar rats were inoculated with 106 C6 glioma cells. Treated animals received intratumoural lentiviral HSV-TK gene transfers on days 7 and 8 followed by 2-week GCV therapy starting on day 10. Animals were repeatedly imaged during therapy using volumetric MRI, diffusion and relaxation mapping, as well as metabolic [1-13 C]pyruvate MRS imaging. Survival (measured as time before animals reached a humane endpoint and were euthanised) was assessed up to day 30 posttherapy. HSV-TK/GCV gene therapy lengthened the median survival time from 12 to 25 days. This was accompanied by an apparent tumour growth arrest, but no changes in diffusion or relaxation parameters in treated animals. The metabolic response was more evident in the case-by-case analysis than in the group-level analysis. Treated animals also showed a 37 ± 15% decrease (P < 0.05, n = 5) in lactate-to-pyruvate ratio between therapy weeks, whereas a 44 ± 18% increase (P < 0.05, n = 6) was observed in control animals. Hyperpolarised [1-13 C]pyruvate MRI can offer complementary metabolic information to traditional MR methods to give a more comprehensive picture of the slowly developing gene therapy response. This may benefit the detection of the successful therapy response in patients.
Collapse
Affiliation(s)
- Riikka Nivajärvi
- Kuopio Biomedical Imaging Unit, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Venla Olsson
- Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Viivi Hyppönen
- Kuopio Biomedical Imaging Unit, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Sean Bowen
- Center for Hyperpolarization in Magnetic Resonance, Department of Electrical Engineering, Technical University of Denmark, Lyngby, Denmark
| | - Hanna M Leinonen
- FinVector Oy, Kuopio, Finland
- Kuopio Center for Gene and Cell Therapy, Kuopio, Finland
| | - Hanna P Lesch
- FinVector Oy, Kuopio, Finland
- Kuopio Center for Gene and Cell Therapy, Kuopio, Finland
| | - Jan Henrik Ardenkjaer-Larsen
- Center for Hyperpolarization in Magnetic Resonance, Department of Electrical Engineering, Technical University of Denmark, Lyngby, Denmark
| | - Olli H J Gröhn
- Kuopio Biomedical Imaging Unit, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Seppo Ylä-Herttuala
- Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Mikko I Kettunen
- Kuopio Biomedical Imaging Unit, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
78
|
High-Resolution Confocal Fluorescence Imaging of Serine Hydrolase Activity in Cryosections - Application to Glioma Brain Unveils Activity Hotspots Originating from Tumor-Associated Neutrophils. Biol Proced Online 2020; 22:6. [PMID: 32190011 PMCID: PMC7073015 DOI: 10.1186/s12575-020-00118-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 02/19/2020] [Indexed: 12/18/2022] Open
Abstract
Background Serine hydrolases (SHs) are a functionally diverse family of enzymes playing pivotal roles in health and disease and have emerged as important therapeutic targets in many clinical conditions. Activity-based protein profiling (ABPP) using fluorophosphonate (FP) probes has been a powerful chemoproteomic approach in studies unveiling roles of SHs in various biological systems. ABPP utilizes cell/tissue proteomes and features the FP-warhead, linked to a fluorescent reporter for in-gel fluorescence imaging or a biotin tag for streptavidin enrichment and LC-MS/MS-based target identification. Existing ABPP approaches characterize global SH activity based on mobility in gel or MS-based target identification and cannot reveal the identity of the cell-type responsible for an individual SH activity originating from complex proteomes. Results Here, by using an activity probe with broad reactivity towards the SH family, we advance the ABPP methodology to glioma brain cryosections, enabling for the first time high-resolution confocal fluorescence imaging of global SH activity in the tumor microenvironment. Tumor-associated cell types were identified by extensive immunohistochemistry on activity probe-labeled sections. Tissue-ABPP indicated heightened SH activity in glioma vs. normal brain and unveiled activity hotspots originating from tumor-associated neutrophils (TANs), rather than tumor-associated macrophages (TAMs). Thorough optimization and validation was provided by parallel gel-based ABPP combined with LC-MS/MS-based target verification. Conclusions Our study advances the ABPP methodology to tissue sections, enabling high-resolution confocal fluorescence imaging of global SH activity in anatomically preserved complex native cellular environment. To achieve global portrait of SH activity throughout the section, a probe with broad reactivity towards the SH family members was employed. As ABPP requires no a priori knowledge of the identity of the target, we envisage no imaginable reason why the presently described approach would not work for sections regardless of species and tissue source.
Collapse
|
79
|
Han Y, Sun Y, Zhang Y, Xia Q. High DPP4 expression predicts poor prognosis in patients with low-grade glioma. Mol Biol Rep 2020; 47:2189-2196. [PMID: 32076999 DOI: 10.1007/s11033-020-05321-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 02/13/2020] [Indexed: 11/25/2022]
Abstract
Dipeptidyl peptidase-IV (DPP4) plays a key role in tumor development; however, its role in glioma pathogenesis has not been determined. Here, we aimed to investigate the expression pattern of DPP4 and explore the association between expression and patient prognosis in glioma. DPP4 levels were investigated using qRT-PCR, immunohistochemistry and western blot in a rat model of glioma and also in patient samples. The relationship between DPP4 levels, WHO pathological grade gliomas, and isocitrate dehydrogenase 1 and 2 (IDH1/2) status was assessed in patient samples. Our data indicated that DPP4 levels were markedly increased in a rat model of glioma (p < 0.05, p < 0.01) and aslo in patient samples. Furthermore, the elevation of DPP4 levels in the samples obtained from pateints was associated with the pathogical grade of glioma and the IDH1/2 status (p < 0.01, p < 0.001). High DPP4 levels decreased the survival probability of patients with low-grade glioma (LGG). The data from patient samples showed that DPP4 expression increased with the pathological grade. Increased expression of DPP4 could be a promising index for determining the prognosis of glioma.
Collapse
Affiliation(s)
- Yadi Han
- Department of Clinical Laboratory Science, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, 450008, China
- Zhengzhou Key Laboratory of Digestive Tumor Markers, No. 127 Dongming Road, Zhengzhou, 450008, China
| | - Yuxue Sun
- Department of Neurosurgery, Renmin Hospital of Zhengzhou University, Zhengzhou, 450008, China
| | - Yusong Zhang
- Department of Pathology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, 450008, China.
| | - Qingxin Xia
- Department of Pathology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, 450008, China.
| |
Collapse
|
80
|
Ahlstedt J, Förnvik K, Helms G, Salford LG, Ceberg C, Skagerberg G, Redebrandt HN. Growth pattern of experimental glioblastoma. Histol Histopathol 2020; 35:871-886. [PMID: 32022242 DOI: 10.14670/hh-18-207] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Glioblastoma multiforme (GBM) is an aggressive primary brain malignancy with a very poor prognosis. Researchers employ animal models to develop potential therapies. It is important that these models have clinical relevance. This means that old models, propagated for decades in cultures, should be questioned. Parameters to be evaluated include whether animals are immune competent or not, the infiltrative growth pattern of the tumor, tumor volume resulting in symptoms and growth rate. We here describe the growth pattern of an experimental glioblastoma model in detail with GFP positive glioblastoma cells in fully immune competent animals and study tumor growth rate and tumor mass as a function of time from inoculation. We were able to correlate findings made with classical immunohistochemistry and MR findings. The tumor growth rate was fitted by a Gompertz function. The model predicted the time until onset of symptoms for 5000 inoculated cells to 18.7±0.4 days, and the tumor mass at days 10 and 14, which are commonly used as the start of treatment in therapeutic studies, were 5.97±0.62 mg and 29.1±3.0 mg, respectively. We want to raise the question regarding the clinical relevance of the outline of glioblastoma experiments, where treatment is often initiated at a very early stage. The approach presented here could potentially be modified to gain information also from other tumor models.
Collapse
Affiliation(s)
- Jonatan Ahlstedt
- Rausing Laboratory, Division of Neurosurgery, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Karolina Förnvik
- Rausing Laboratory, Division of Neurosurgery, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Gunther Helms
- Medical Radiation Physics, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Leif G Salford
- Rausing Laboratory, Division of Neurosurgery, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Crister Ceberg
- Medical Radiation Physics, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Gunnar Skagerberg
- Department of Neurosurgery, Skåne University Hospital in Lund, Lund, Sweden
| | - Henrietta Nittby Redebrandt
- Department of Neurosurgery, Skåne University Hospital in Lund, Lund, Sweden.,Rausing Laboratory, Division of Neurosurgery, Department of Clinical Sciences Lund, Lund University, Lund, Sweden.
| |
Collapse
|
81
|
Commisso C. The pervasiveness of macropinocytosis in oncological malignancies. Philos Trans R Soc Lond B Biol Sci 2020; 374:20180153. [PMID: 30967003 DOI: 10.1098/rstb.2018.0153] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
In tumour cells, macropinocytosis functions as an amino acid supply route and supports cancer cell survival and proliferation. Initially demonstrated in oncogenic KRAS-driven models of pancreatic cancer, macropinocytosis triggers the internalization of extracellular proteins via discrete endocytic vesicles called macropinosomes. The incoming protein cargo is targeted for lysosome-dependent degradation, causing the intracellular release of amino acids. These protein-derived amino acids support metabolic fitness by contributing to the intracellular amino acid pools, as well as to the biosynthesis of central carbon metabolites. In this way, macropinocytosis represents a novel amino acid supply route that tumour cells use to survive the nutrient-poor conditions of the tumour microenvironment. Macropinocytosis has also emerged as an entry mechanism for a variety of nanomedicines, suggesting that macropinocytosis regulation in the tumour setting can be harnessed for the delivery of anti-cancer therapeutics. A slew of recent studies point to the possibility that macropinocytosis is a pervasive feature of many different tumour types. In this review, we focus on the role of this important uptake mechanism in a variety of cancers and highlight the main molecular drivers of macropinocytosis in these malignancies. This article is part of the Theo Murphy meeting issue 'Macropinocytosis'.
Collapse
Affiliation(s)
- Cosimo Commisso
- Tumor Initiation and Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute , La Jolla, CA 92037 , USA
| |
Collapse
|
82
|
Laws MT, Bonomi RE, Gelovani DJ, Llaniguez J, Lu X, Mangner T, Gelovani JG. Noninvasive quantification of SIRT1 expression-activity and pharmacologic inhibition in a rat model of intracerebral glioma using 2-[ 18F]BzAHA PET/CT/MRI. Neurooncol Adv 2020; 2:vdaa006. [PMID: 32118205 PMCID: PMC7034639 DOI: 10.1093/noajnl/vdaa006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background Several studies demonstrated that glioblastoma multiforme progression and recurrence is linked to epigenetic regulatory mechanisms. Sirtuin 1 (SIRT1) plays an important role in glioma progression, invasion, and treatment response and is a potential therapeutic target. The aim of this study is to test the feasibility of 2-[18F]BzAHA for quantitative imaging of SIRT1 expression–activity and monitoring pharmacologic inhibition in a rat model of intracerebral glioma. Methods Sprague Dawley rats bearing 9L (N = 12) intracerebral gliomas were injected with 2-[18F]BzAHA (300–500 µCi/animal i.v.) and dynamic positron-emission tomography (PET) imaging was performed for 60 min. Then, SIRT1 expression in 9L tumors (N = 6) was studied by immunofluorescence microscopy (IF). Two days later, rats with 9L gliomas were treated either with SIRT1 specific inhibitor EX-527 (5 mg/kg, i.p.; N = 3) or with histone deacetylases class IIa specific inhibitor MC1568 (30 mg/kg, i.p.; N = 3) and 30 min later were injected i.v. with 2-[18F]BzAHA. PET-computerized tomography-magnetic resonance (PET/CT/MR) images acquired after EX-527 and MC1568 treatments were co-registered with baseline images. Results Standard uptake values (SUVs) of 2-[18F]BzAHA in 9L tumors measured at 20 min post-radiotracer administration were 1.11 ± 0.058 and had a tumor-to-brainstem SUV ratio of 2.73 ± 0.141. IF of 9L gliomas revealed heterogeneous upregulation of SIRT1, especially in hypoxic and peri-necrotic regions. Significant reduction in 2-[18F]BzAHA SUV and distribution volume in 9L tumors was observed after administration of EX-527, but not MC1568. Conclusions PET/CT/MRI with 2-[18F]BzAHA can facilitate studies to elucidate the roles of SIRT1 in gliomagenesis and progression, as well as to optimize therapeutic doses of novel SIRT1 inhibitors.
Collapse
Affiliation(s)
- Maxwell T Laws
- Department of Biomedical Engineering, College of Engineering and School of Medicine, Wayne State University, Detroit, Michigan, USA
| | - Robin E Bonomi
- Department of Biomedical Engineering, College of Engineering and School of Medicine, Wayne State University, Detroit, Michigan, USA
| | - David J Gelovani
- Department of Biomedical Engineering, College of Engineering and School of Medicine, Wayne State University, Detroit, Michigan, USA
| | - Jeremy Llaniguez
- Department of Biomedical Engineering, College of Engineering and School of Medicine, Wayne State University, Detroit, Michigan, USA
| | - Xin Lu
- Positron Emission Tomography Center, Wayne State University, Detroit, Michigan, USA
| | - Thomas Mangner
- Positron Emission Tomography Center, Wayne State University, Detroit, Michigan, USA
| | - Juri G Gelovani
- Department of Biomedical Engineering, College of Engineering and School of Medicine, Wayne State University, Detroit, Michigan, USA.,Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan, USA.,Department of Neurosurgery, Wayne State University School of Medicine, Detroit, Michigan, USA.,Molecular Imaging Program, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, USA
| |
Collapse
|
83
|
Mamani JB, Marinho BS, Rego GNDA, Nucci MP, Alvieri F, Santos RSD, Ferreira JVM, Oliveira FAD, Gamarra LF. Magnetic hyperthermia therapy in glioblastoma tumor on-a-Chip model. EINSTEIN-SAO PAULO 2020; 18:eAO4954. [PMID: 31939525 PMCID: PMC6924828 DOI: 10.31744/einstein_journal/2020ao4954] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 06/25/2019] [Indexed: 12/29/2022] Open
Abstract
OBJECTIVE To evaluate the magnetic hyperthermia therapy in glioblastoma tumor-on-a-Chip model using a microfluidics device. METHODS The magnetic nanoparticles coated with aminosilane were used for the therapy of magnetic hyperthermia, being evaluated the specific absorption rate of the magnetic nanoparticles at 300 Gauss and 305kHz. A preculture of C6 cells was performed before the 3D cells culture on the chip. The process of magnetic hyperthermia on the Chip was performed after administration of 20μL of magnetic nanoparticles (10mgFe/mL) using the parameters that generated the specific absorption rate value. The efficacy of magnetic hyperthermia therapy was evaluated by using the cell viability test through the following fluorescence staining: calcein acetoxymethyl ester (492/513nm), for live cells, and ethidium homodimer-1 (526/619nm) for dead cells dyes. RESULTS Magnetic nanoparticles when submitted to the alternating magnetic field (300 Gauss and 305kHz) produced a mean value of the specific absorption rate of 115.4±6.0W/g. The 3D culture of C6 cells evaluated by light field microscopy imaging showed the proliferation and morphology of the cells prior to the application of magnetic hyperthermia therapy. Fluorescence images showed decreased viability of cultured cells in organ-on-a-Chip by 20% and 100% after 10 and 30 minutes of the magnetic hyperthermia therapy application respectively. CONCLUSION The study showed that the therapeutic process of magnetic hyperthermia in the glioblastoma on-a-chip model was effective to produce the total cell lise after 30 minutes of therapy.
Collapse
Affiliation(s)
| | | | | | - Mariana Penteado Nucci
- Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | | | | | | | | | | |
Collapse
|
84
|
Sharma HS, Muresanu DF, Castellani RJ, Nozari A, Lafuente JV, Tian ZR, Sahib S, Bryukhovetskiy I, Bryukhovetskiy A, Buzoianu AD, Patnaik R, Wiklund L, Sharma A. Pathophysiology of blood-brain barrier in brain tumor. Novel therapeutic advances using nanomedicine. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 151:1-66. [PMID: 32448602 DOI: 10.1016/bs.irn.2020.03.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
85
|
Design, synthesis and cytotoxicity of the antitumor agent 1-azabicycles for chemoresistant glioblastoma cells. Invest New Drugs 2019; 38:1257-1271. [PMID: 31838735 DOI: 10.1007/s10637-019-00877-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 11/05/2019] [Indexed: 10/25/2022]
Abstract
Twelve multi-functional pyrrolizidinones, indolizidinones and pyrroliazepinones were prepared from formal aza-[3 + 2] and aza-[3 + 3] cycloadditions of five- to seven-membered heterocyclic enaminones as diverse ambident electrophiles. The antitumor activity of these alkaloid-like compounds was investigated through an initial screening performed on human glioblastoma multiform (GBM) cell lines (GL-15, U251), on murine glioma cells line (C6) and on normal glial cells. Of the compounds tested, the new pyrrolo[1,2a]azepinone, [ethyl (3-oxo-1,2-diphenyl-6,7,8,9-tetrahydro-3H-pyrrolo[1,2a]azepin-9a(5H)-yl)acetate] or (Compound-13) exhibited selective cytotoxic effects on GBM-temozolomide resistant cells. Compound-13 exerted dose-dependent cytotoxic activity by promoting arrest of cells in the G0/G1 phase of the cell cycle in the first 24 h. The apoptotic effect observed was in a time-dependent manner. Anti-migratory effect promoted by the treatment with compound-13 was also observed. Moreover, healthy mixed glial cell cultures from rat brain exhibited no cytotoxicity effect upon exposure to compound-13. Thus, the present study paves the way for the use of compound-13 as novel antitumor scaffold candidate for glioma cell therapy.
Collapse
|
86
|
Multi-layered core-sheath fiber membranes for controlled drug release in the local treatment of brain tumor. Sci Rep 2019; 9:17936. [PMID: 31784666 PMCID: PMC6884550 DOI: 10.1038/s41598-019-54283-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 11/07/2019] [Indexed: 12/21/2022] Open
Abstract
Interstitial chemotherapy plays a pivotal role in the treatment of glioblastoma multiforme (GBM), an aggressive form of primary brain cancer, by enhancing drug biodistribution to the tumor and avoiding systemic toxicities. The use of new polymer structures that extend the release of cytotoxic agents may therefore increase survival and prevent recurrence. A novel core-sheath fiber loaded with the drug carmustine (BCNU) was evaluated in an in vivo brain tumor model. Three-dimensional discs were formed from coaxially electrospun fiber membranes and in vitro BCNU release kinetics were measured. In vivo survival was assessed following implantation of discs made of compressed core-sheath fibers (NanoMesh) either concurrently with or five days after intracranial implantation of 9L gliosarcoma. Co-implantation of NanoMesh and 9L gliosarcoma resulted in statistically significant long-term survival (>150 days). Empty control NanoMesh confirmed the safety of these novel implants. Similarly, Day 5 studies showed significant median, overall, and long-term survival rates, suggesting optimal control of tumor growth, confirmed with histological and immunohistochemical analyses. Local chemotherapy by means of biodegradable NanoMesh implants is a new treatment paradigm for the treatment for brain tumors. Drug delivery with coaxial core-sheath structures benefits from high drug loading, controlled long-term release kinetics, and slow polymer degradation. This represents a promising evolution for the current treatment of GBM.
Collapse
|
87
|
Lorger M, Andreou T, Fife C, James F. Immune Checkpoint Blockade - How Does It Work in Brain Metastases? Front Mol Neurosci 2019; 12:282. [PMID: 31824260 PMCID: PMC6881300 DOI: 10.3389/fnmol.2019.00282] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 11/05/2019] [Indexed: 01/13/2023] Open
Abstract
Immune checkpoints restrain the immune system following its activation and their inhibition unleashes anti-tumor immune responses. Immune checkpoint inhibitors revolutionized the treatment of several cancer types, including melanoma, and immune checkpoint blockade with anti-PD-1 and anti-CTLA-4 antibodies is becoming a frontline therapy in metastatic melanoma. Notably, up to 60% of metastatic melanoma patients develop metastases in the brain. Brain metastases (BrM) are also very common in patients with lung and breast cancer, and occur in ∼20-40% of patients across different cancer types. Metastases in the brain are associated with poor prognosis due to the lack of efficient therapies. In the past, patients with BrM used to be excluded from immune-based clinical trials due to the assumption that such therapies may not work in the context of "immune-specialized" environment in the brain, or may cause harm. However, recent trials in patients with BrM demonstrated safety and intracranial activity of anti-PD-1 and anti-CTLA-4 therapy. We here discuss how immune checkpoint therapy works in BrM, with focus on T cells and the cross-talk between BrM, the immune system, and tumors growing outside the brain. We discuss major open questions in our understanding of what is required for an effective immune checkpoint inhibitor therapy in BrM.
Collapse
Affiliation(s)
- Mihaela Lorger
- Institute of Medical Research at St. James’s, School of Medicine, University of Leeds, Leeds, United Kingdom
| | - Tereza Andreou
- Institute of Medical Research at St. James’s, School of Medicine, University of Leeds, Leeds, United Kingdom
| | - Christopher Fife
- Institute of Medical Research at St. James’s, School of Medicine, University of Leeds, Leeds, United Kingdom
| | - Fiona James
- Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| |
Collapse
|
88
|
Jost G, Frenzel T, Boyken J, Pietsch H. Impact of brain tumors and radiotherapy on the presence of gadolinium in the brain after repeated administration of gadolinium-based contrast agents: an experimental study in rats. Neuroradiology 2019; 61:1273-1280. [PMID: 31297571 PMCID: PMC6817760 DOI: 10.1007/s00234-019-02256-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 07/01/2019] [Indexed: 02/04/2023]
Abstract
PURPOSE To investigate the impact of blood-brain barrier (BBB) alterations induced by an experimental tumor and radiotherapy on MRI signal intensity (SI) in deep cerebellar nuclei (DCN) and the presence of gadolinium after repeated administration of a linear gadolinium-based contrast agent in rats. METHODS Eighteen Fischer rats were divided into a tumor (gliosarcoma, GS9L model), a radiotherapy, and a control group. All animals received 5 daily injections (1.8 mmol/kg) of gadopentetate dimeglumine. For tumor-bearing animals, the BBB disruption was confirmed by contrast-enhanced MRI. Animals from the tumor and radiation group underwent radiotherapy in 6 fractions of 5 Gray. The SI ratio between DCN and brain stem was evaluated on T1-weigthed MRI at baseline and 1 week after the last administration. Subsequently, the brain was dissected for gadolinium quantification by inductively coupled plasma-mass spectrometry. Statistical analysis was done with the Kruskal-Wallis test. RESULTS An increased but similar DCN/brain stem SI ratio was found for all three groups (p = 0.14). The gadolinium tissue concentrations (median, nmol/g) were 6.7 (tumor), 6.3 (radiotherapy), and 6.8 (control) in the cerebellum (p = 0.64) and 17.8/14.6 (tumor), 20.0/18.9 (radiotherapy), and 17.8/15.9 (control) for the primary tumor (p = 0.98) and the contralateral hemisphere (p = 0.41) of the cerebrum, respectively. CONCLUSION An experimental brain tumor treated by radiotherapy or radiotherapy alone did not alter DCN signal hyperintensity and gadolinium concentration in the rat brain 1 week after repeated administration of gadopentetate. This suggests that a local BBB disruption does not affect the amount of retained gadolinium in the brain.
Collapse
Affiliation(s)
- Gregor Jost
- Bayer AG, MR & CT Contrast Media Research, Muellerstrasse 178, 13353, Berlin, Germany.
| | - Thomas Frenzel
- Bayer AG, MR & CT Contrast Media Research, Muellerstrasse 178, 13353, Berlin, Germany
| | - Janina Boyken
- Bayer AG, MR & CT Contrast Media Research, Muellerstrasse 178, 13353, Berlin, Germany
| | - Hubertus Pietsch
- Bayer AG, MR & CT Contrast Media Research, Muellerstrasse 178, 13353, Berlin, Germany
| |
Collapse
|
89
|
Li N, Engels E, Davis JA, Dipuglia A, Vogel S, Valceski M, Rosenfeld AB, Lerch MLF, Corde S, Tehei M. Polo-like kinase 1 inhibitor BI6727 sensitizes 9L gliosarcoma cells to ionizing irradiation. Biomed Phys Eng Express 2019. [DOI: 10.1088/2057-1976/ab4d0e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
90
|
De Meulenaere V, Bonte E, Verhoeven J, Kalala Okito JP, Pieters L, Vral A, De Wever O, Leybaert L, Goethals I, Vanhove C, Descamps B, Deblaere K. Adjuvant therapeutic potential of tonabersat in the standard treatment of glioblastoma: A preclinical F98 glioblastoma rat model study. PLoS One 2019; 14:e0224130. [PMID: 31634381 PMCID: PMC6802836 DOI: 10.1371/journal.pone.0224130] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 10/07/2019] [Indexed: 12/21/2022] Open
Abstract
Purpose Even with an optimal treatment protocol, the median survival of glioblastoma (GB) patients is only 12–15 months. Hence, there is need for novel effective therapies that improve survival outcomes. Recent evidence suggests an important role for connexin (Cx) proteins (especially Cx43) in the microenvironment of malignant glioma. Cx43-mediated gap junctional communication has been observed between tumor cells, between astrocytes and between tumor cells and astrocytes. Therefore, gap junction directed therapy using a pharmacological suppressor or modulator, such as tonabersat, could be a promising target in the treatment of GB. In this preclinical study, we evaluated the possible therapeutic potential of tonabersat in the F98 model. Procedures Female Fischer rats were inoculated with ± 25.000 F98 tumor cells in the right frontal lobe. Eight days post-inoculation contrast-enhanced T1-weighted (CE-T1w) magnetic resonance (MR) images were acquired to confirm tumor growth in the brain. After tumor confirmation, rats were randomized into a Control Group, a Connexin Modulation Group (CM), a Standard Medical Treatment Group (ST), and a Standard Medical Treatment with adjuvant Connexin Modulation Group (STCM). To evaluate therapy response, T2-weighted (T2w) and CE-T1w sequences were acquired at several time points. Tumor volume analysis was performed on CE-T1w images and statistical analysis was performed using a linear mixed model. Results Significant differences in estimated geometric mean tumor volumes were found between the ST Group and the Control Group and also between the STCM Group and the Control Group. In addition, significant differences in estimated geometric mean tumor volumes between the ST Group and the STCM Group were demonstrated. No significant differences in estimated geometric mean tumor volumes were found between the Control Group and the CM Group. Conclusion Our results demonstrate a therapeutic potential of tonabersat for the treatment of GB when used in combination with radiotherapy and temozolomide chemotherapy.
Collapse
Affiliation(s)
| | - Ellen Bonte
- Department of Radiology, Ghent University Hospital, Ghent, Belgium
| | - Jeroen Verhoeven
- Department of Pharmaceutical analysis, Ghent University, Ghent, Belgium
| | | | - Leen Pieters
- Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | - Anne Vral
- Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | - Olivier De Wever
- Department of Experimental Cancer Research, Ghent University, Ghent, Belgium
| | - Luc Leybaert
- Department of Basic Medical Sciences, Ghent University, Ghent, Belgium
| | - Ingeborg Goethals
- Department of Nuclear Medicine, Ghent University Hospital, Ghent, Belgium
| | | | | | - Karel Deblaere
- Department of Radiology, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
91
|
Monfared H, Jahangard Y, Nikkhah M, Mirnajafi-Zadeh J, Mowla SJ. Potential Therapeutic Effects of Exosomes Packed With a miR-21-Sponge Construct in a Rat Model of Glioblastoma. Front Oncol 2019; 9:782. [PMID: 31482067 PMCID: PMC6710330 DOI: 10.3389/fonc.2019.00782] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 08/01/2019] [Indexed: 01/01/2023] Open
Abstract
Glioblastoma multiforme (GBM) is a grade 4 and the most aggressive form of glioma, with a poor response to current treatments. The expression of microRNAs (miRNAs) is widely dysregulated in various cancers, including GBM. One of the overexpressed miRNAs in GBM is miR-21 which promotes proliferation, invasion and metastatic behaviors of tumor cells. With a size of 30-100 nm, the extracellular vesicles "exosomes" have emerged as a novel and powerful drug delivering systems. Recently, exosomal transfer of miRNAs or anti-miRNAs to tumor cells has introduced a new approach for therapeutic application of miRNAs to combat cancer. Here, we have tried to down-regulate miR-21 expression in glioma cell lines, U87-MG, and C6, by using engineered exosomes, packed with a miR-21-sponge construct. Our data revealed that the engineered exosomes have the potential to suppress miR-21 and consequently to upregulate miR-21 target genes, PDCD4 and RECK. Interestingly, in cells treated with miR-21-sponge exosomes we observed a decline in proliferation and also an elevation in apoptotic rates. Finally, in a rat model of glioblastoma, administrating exosomes loaded with a miR-21-sponge construct leads to a significant reduction in the volume of the tumors. In brief, our findings suggest a new therapeutic strategy to use engineered exosomes to deliver a miR-21-sponge construct to GBM cells, in order to block its malignant behavior.
Collapse
Affiliation(s)
- Hamideh Monfared
- Department of Molecular Genetics, Faculty of Biological Science, Tarbiat Modares University, Tehran, Iran
| | - Yavar Jahangard
- Department of Molecular Genetics, Faculty of Biological Science, Tarbiat Modares University, Tehran, Iran
| | - Maryam Nikkhah
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Javad Mirnajafi-Zadeh
- Department of Physiology, Faculty of Medical Science, Tarbiat Modares University, Tehran, Iran
| | - Seyed Javad Mowla
- Department of Molecular Genetics, Faculty of Biological Science, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
92
|
McDannold N, Zhang Y, Supko JG, Power C, Sun T, Peng C, Vykhodtseva N, Golby AJ, Reardon DA. Acoustic feedback enables safe and reliable carboplatin delivery across the blood-brain barrier with a clinical focused ultrasound system and improves survival in a rat glioma model. Am J Cancer Res 2019; 9:6284-6299. [PMID: 31534551 PMCID: PMC6735504 DOI: 10.7150/thno.35892] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 07/22/2019] [Indexed: 02/05/2023] Open
Abstract
The blood-brain barrier (BBB) restricts delivery of most chemotherapy agents to brain tumors. Here, we investigated a clinical focused ultrasound (FUS) device to disrupt the BBB in rats and enhance carboplatin delivery to the brain using the F98 glioma model. Methods: In each rat, 2-3 volumetric sonications (5 ms bursts at 1.1 Hz for 75s) targeted 18-27 locations in one hemisphere. Sonication was combined with Definity microbubbles (10 µl/kg) and followed by intravenous carboplatin (50 mg/kg). Closed-loop feedback control was performed based on acoustic emissions analysis. Results: Safety and reliability were established in healthy rats after three sessions with carboplatin; BBB disruption was induced in every target without significant damage evident in MRI or histology. In tumor-bearing rats, concentrations of MRI contrast agent (Gadavist) were 1.7 and 3.3 times higher in the tumor center and margin, respectively, than non-sonicated tumors (P<0.001). Tissue-to-plasma ratios of intact carboplatin concentrations were increased by 7.3 and 2.9 times in brain and tumor respectively, at one hour after FUS and 4.2 and 2.4 times at four hours. Tumor volume doubling time in rats receiving FUS and carboplatin increased by 96% and 126% compared to rats that received carboplatin alone and non-sonicated controls, respectively (P<0.05); corresponding increases in median survival were 48% and 66% (P<0.01). Conclusion: Overall, this work demonstrates that actively-controlled BBB disruption with a clinical device can enhance carboplatin delivery without neurotoxicity at level that reduces tumor growth and improves survival in an aggressive and infiltrative rat glioma model.
Collapse
|
93
|
Guequén A, Zamorano P, Córdova F, Koning T, Torres A, Ehrenfeld P, Boric MP, Salazar-Onfray F, Gavard J, Durán WN, Quezada C, Sarmiento J, Sánchez FA. Interleukin-8 Secreted by Glioblastoma Cells Induces Microvascular Hyperpermeability Through NO Signaling Involving S-Nitrosylation of VE-Cadherin and p120 in Endothelial Cells. Front Physiol 2019; 10:988. [PMID: 31440166 PMCID: PMC6694439 DOI: 10.3389/fphys.2019.00988] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 07/18/2019] [Indexed: 01/08/2023] Open
Abstract
Glioblastoma is a highly aggressive brain tumor, characterized by the formation of dysfunctional blood vessels and a permeable endothelial barrier. S-nitrosylation, a post-translational modification, has been identified as a regulator of endothelial function. In this work we explored whether S-nitrosylation induced by glioblastoma tumors regulates the endothelial function. As proof of concept, we observed that S-nitrosylation is present in the tumoral microenvironment of glioblastoma in two different animal models. Subsequently, we measured S nitrosylation and microvascular permeability in EAhy296 endothelial cells and in cremaster muscle. In vitro, conditioned medium from the human glioblastoma cell line U87 activates endothelial nitric oxide synthase, causes VE-cadherin- S-nitrosylation and induces hyperpermeability. Blocking Interleukin-8 (IL-8) in the conditioned medium inhibited S-nitrosylation of VE-cadherin and hyperpermeability. Recombinant IL-8 increased endothelial permeability by activating eNOS, S-nitrosylation of VE-cadherin and p120, internalization of VE-cadherin and disassembly of adherens junctions. In vivo, IL-8 induced S-nitrosylation of VE-cadherin and p120 and conditioned medium from U87 cells caused hyperpermeability in the mouse cremaster muscle. We conclude that eNOS signaling induced by glioma cells-secreted IL-8 regulates endothelial barrier function in the context of glioblastoma involving S-nitrosylation of VE-cadherin and p120. Our results suggest that inhibiting S-nitrosylation may be an effective way to control and/or block damage to the endothelial barrier and prevent cancer progression.
Collapse
Affiliation(s)
- Anita Guequén
- Instituto de Inmunología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
| | - Patricia Zamorano
- Instituto de Inmunología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
| | - Francisco Córdova
- Instituto de Inmunología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
| | - Tania Koning
- Instituto de Inmunología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
| | - Angelo Torres
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile
| | - Pamela Ehrenfeld
- Instituto de Histología, Anatomía y Patología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
- Centro Interdisciplinario de Estudios del Sistema Nervioso (CISNe), Universidad Austral de Chile, Valdivia, Chile
| | - Mauricio P. Boric
- Departamento de Fisiología, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Flavio Salazar-Onfray
- Instituto Milenio de Inmunología e Inmunoterapia, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Julie Gavard
- Team SOAP, Signaling in Oncogenesis, Angiogenesis and Permeability, INSERM, CNRS, Institut de Cancérologie de l’Ouest, Université de Nantes, Nantes, France
| | - Walter N. Durán
- Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, The State University of New Jersey, Newark, NJ, United States
| | - Claudia Quezada
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile
| | - José Sarmiento
- Instituto de Fisiología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
| | - Fabiola A. Sánchez
- Instituto de Inmunología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
- Centro Interdisciplinario de Estudios del Sistema Nervioso (CISNe), Universidad Austral de Chile, Valdivia, Chile
| |
Collapse
|
94
|
Rego GNDA, Mamani JB, Souza TKF, Nucci MP, Silva HRD, Gamarra LF. Therapeutic evaluation of magnetic hyperthermia using Fe3O4-aminosilane-coated iron oxide nanoparticles in glioblastoma animal model. EINSTEIN-SAO PAULO 2019; 17:eAO4786. [PMID: 31390427 PMCID: PMC6668731 DOI: 10.31744/einstein_journal/2019ao4786] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 03/28/2019] [Indexed: 11/21/2022] Open
Abstract
Objective: To evaluate the potential of magnetic hyperthermia using aminosilane-coated superparamagnetic iron oxide nanoparticles in glioblastoma tumor model. Methods: The aminosilane-coated superparamagnetic iron oxide nanoparticles were analyzed as to their stability in aqueous medium and their heating potential through specific absorption rate, when submitted to magnetic hyperthermia with different frequencies and intensities of alternating magnetic field. In magnetic hyperthermia in vitro assays, the C6 cells cultured and transduced with luciferase were analyzed by bioluminescence in the absence/presence of alternating magnetic field, and also with and without aminosilane-coated superparamagnetic iron oxide nanoparticles. In the in vivo study, the measurement of bioluminescence was performed 21 days after glioblastoma induction with C6 cells in rats. After 24 hours, the aminosilane-coated superparamagnetic iron oxide nanoparticles were implanted in animals, and magnetic hyperthermia was performed for 40 minutes, using the best conditions of frequency and intensity of alternating magnetic field tested in the in vitro study (the highest specific absorption rate value) and verified the difference of bioluminescence before and after magnetic hyperthermia. Results: The aminosilane-coated superparamagnetic iron oxide nanoparticles were stable, and their heating capacity increased along with higher frequency and intensity of alternating magnetic field. The magnetic hyperthermia application with 874kHz and 200 Gauss of alternating magnetic field determined the best value of specific absorption rate (194.917W/g). When these magnetic hyperthermia parameters were used in in vitro and in vivo analysis, resulted in cell death of 52.0% and 32.8%, respectively, detected by bioluminescence. Conclusion: The magnetic hyperthermia was promissing for the therapeutical process of glioblastoma tumors in animal model, using aminosilane-coated superparamagnetic iron oxide nanoparticles, which presented high specific absorption rate.
Collapse
Affiliation(s)
| | | | | | - Mariana Penteado Nucci
- Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | | | | |
Collapse
|
95
|
Sassi K, Nury T, Zarrouk A, Sghaier R, Khalafi-Nezhad A, Vejux A, Samadi M, Aissa-Fennira FB, Lizard G. Induction of a non-apoptotic mode of cell death associated with autophagic characteristics with steroidal maleic anhydrides and 7β-hydroxycholesterol on glioma cells. J Steroid Biochem Mol Biol 2019; 191:105371. [PMID: 31034873 DOI: 10.1016/j.jsbmb.2019.04.020] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 04/19/2019] [Accepted: 04/24/2019] [Indexed: 01/07/2023]
Abstract
Steroidal maleic anhydrides were prepared in one step: lithocholic, chenodeoxicholic, deoxicholic, ursocholic, and hyodeoxicholic acid derivatives. Their capability to induce cell death was studied on C6 rat glioma cells, and 7β-hydroxycholesterol was used as positive cytotoxic control. The highest cytotoxicity was observed with lithocholic and chenodeoxicholic acid derivatives (23-(4-methylfuran-2,5-dione)-3α-hydroxy-24-nor-5β-cholane (compound 1a), and 23-(4-methylfuran-2,5-dione)-3α,7α-dihydroxy-24-nor-5β-cholane (compound 1b), respectively), which induce a non-apoptotic mode of cell death associated with mitochondrial membrane potential loss and reactive oxygen species overproduction. No cells with condensed and/or fragmented nuclei, no PARP degradation and no cleaved-caspase-3, which are apoptotic criteria, were observed. Similar effects were found with 7β-hydroxycholesterol. The cell clonogenic survival assay showed that compound 1b was more cytotoxic than compound 1a and 7β-hydroxycholesterol. Compound 1b and 7β-hydroxycholesterol also induce cell cycle modifications. In addition, compounds 1a and 1b, and 7β-hydroxycholesterol favour the formation of large acidic vacuoles revealed by staining with acridine orange and monodansylcadaverine evocating autophagic vacuoles; they also induce an increased ratio of [LC3-II / LC3-I], and modify the expression of mTOR, Beclin-1, Atg12, and Atg5-Atg12 which is are autophagic criteria. The ratio [LC3-II / LC3-I] is also strongly modified by bafilomycin acting on the autophagic flux. Rapamycin, an autophagic inducer, and 3-methyladenine, an autophagic inhibitor, reduce and increase 7β-hydroxycholesterol-induced cell death, respectively, supporting that 7β-hydroxycholesterol induces survival autophagy. Alpha-tocopherol also strongly attenuates 7β-hydroxycholesterol-induced cell death. However, rapamycin, 3-methyladenine, and α-tocopherol have no effect on compounds 1a and 1b-induced cell death. It is concluded that these compounds trigger a non apoptotic mode of cell death, involving the mitochondria and associated with several characteristics of autophagy.
Collapse
Affiliation(s)
- K Sassi
- Univ. Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270 / Inserm, Dijon, France; Univ. Tunis El Manar, Laboratory of Onco-Hematology (LR05ES05), Faculty of Medicine, Tunis, Tunisia
| | - T Nury
- Univ. Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270 / Inserm, Dijon, France
| | - A Zarrouk
- Univ. Monastir, Lab-NAFS 'Nutrition - Functional Food & Vascular Health' (LR12ES05), Monastir, & Faculty of Medicine, Laboratory of Biochemistry, Sousse, Tunisia
| | - R Sghaier
- Univ. Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270 / Inserm, Dijon, France; Univ. Monastir, Lab-NAFS 'Nutrition - Functional Food & Vascular Health' (LR12ES05), Monastir, & Faculty of Medicine, Laboratory of Biochemistry, Sousse, Tunisia; Univ. Manouba, Laboratory of Biotechnology and Valorisation of Bio-Geo Ressources, Higher Institute of Biotechnology (LR11ES31), Sidi Thabet, Tunisia
| | - A Khalafi-Nezhad
- Dept. of Chemistry, College of Sciences, Shiraz University, Shiraz, Iran
| | - A Vejux
- Univ. Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270 / Inserm, Dijon, France
| | - M Samadi
- LCPMC-A2, ICPM, Dept of Chemistry, Univ. Lorraine, Metz Technopôle, Metz, France.
| | - F Ben Aissa-Fennira
- Univ. Tunis El Manar, Laboratory of Onco-Hematology (LR05ES05), Faculty of Medicine, Tunis, Tunisia
| | - G Lizard
- Univ. Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270 / Inserm, Dijon, France.
| |
Collapse
|
96
|
Verhoeven J, Bolcaen J, De Meulenaere V, Kersemans K, Descamps B, Donche S, Van den Broecke C, Boterberg T, Kalala JP, Deblaere K, Vanhove C, De Vos F, Goethals I. Technical feasibility of [ 18F]FET and [ 18F]FAZA PET guided radiotherapy in a F98 glioblastoma rat model. Radiat Oncol 2019; 14:89. [PMID: 31146757 PMCID: PMC6543630 DOI: 10.1186/s13014-019-1290-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 05/08/2019] [Indexed: 12/21/2022] Open
Abstract
Background Glioblastoma (GB) is the most common primary malignant brain tumor. Standard medical treatment consists of a maximal safe surgical resection, subsequently radiation therapy (RT) and chemotherapy with temozolomide (TMZ). An accurate definition of the tumor volume is of utmost importance for guiding RT. In this project we investigated the feasibility and treatment response of subvolume boosting to a PET-defined tumor part. Method F98 GB cells inoculated in the rat brain were imaged using T2- and contrast-enhanced T1-weighted (T1w) MRI. A dose of 20 Gy (5 × 5 mm2) was delivered to the target volume delineated based on T1w MRI for three treatment groups. Two of those treatment groups received an additional radiation boost of 5 Gy (1 × 1 mm2) delivered to the region either with maximum [18F]FET or [18F]FAZA PET tracer uptake, respectively. All therapy groups received intraperitoneal (IP) injections of TMZ. Finally, a control group received no RT and only control IP injections. The average, minimum and maximum dose, as well as the D90-, D50- and D2- values were calculated for nine rats using both RT plans. To evaluate response to therapy, follow-up tumor volumes were delineated based on T1w MRI. Results When comparing the dose volume histograms, a significant difference was found exclusively between the D2-values. A significant difference in tumor growth was only found between active therapy and sham therapy respectively, while no significant differences were found when comparing the three treatment groups. Conclusion In this study we showed the feasibility of PET guided subvolume boosting of F98 glioblastoma in rats. No evidence was found for a beneficial effect regarding tumor response. However, improvements for dose targeting in rodents and studies investigating new targeted drugs for GB treatment are mandatory. Electronic supplementary material The online version of this article (10.1186/s13014-019-1290-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Julie Bolcaen
- Ghent University Hospital, Department of Nuclear Medicine, Ghent, Belgium.,National Research Foundation (NRF), iThemba LABS, Somerset West, South Africa
| | - Valerie De Meulenaere
- Ghent University Hospital, Department of Radiology and Medical Imaging, Ghent, Belgium
| | - Ken Kersemans
- Ghent University Hospital, Department of Nuclear Medicine, Ghent, Belgium
| | - Benedicte Descamps
- IBiTech-MEDISIP Ghent University, Department of Electronics and Information Systems, Ghent, Belgium
| | - Sam Donche
- Ghent University Hospital, Department of Nuclear Medicine, Ghent, Belgium
| | | | - Tom Boterberg
- Ghent University Hospital, Department of Radiation Oncology, Ghent, Belgium
| | | | - Karel Deblaere
- Ghent University Hospital, Department of Radiology and Medical Imaging, Ghent, Belgium
| | - Christian Vanhove
- IBiTech-MEDISIP Ghent University, Department of Electronics and Information Systems, Ghent, Belgium
| | - Filip De Vos
- Laboratory of Radiopharmacy, Ghent University, Ghent, Belgium
| | - Ingeborg Goethals
- Ghent University Hospital, Department of Nuclear Medicine, Ghent, Belgium
| |
Collapse
|
97
|
Molecular imaging HDACs class IIa expression-activity and pharmacologic inhibition in intracerebral glioma models in rats using PET/CT/(MRI) with [ 18F]TFAHA. Sci Rep 2019; 9:3595. [PMID: 30837601 PMCID: PMC6401080 DOI: 10.1038/s41598-019-40054-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 02/08/2019] [Indexed: 12/20/2022] Open
Abstract
HDAC class IIa enzymes (HDAC4, 5, 7, 9) are important for glioma progression, invasion, responses to TMZ and radiotherapy, and prognosis. In this study, we demonstrated the efficacy of PET/CT/(MRI) with [18F]TFAHA for non-invasive and quantitative imaging of HDAC class IIa expression-activity in intracerebral 9L and U87-MG gliomas in rats. Increased accumulation of [18F]TFAHA in 9L and U87-MG tumors was observed at 20 min post radiotracer administration with SUV of 1.45 ± 0.05 and 1.08 ± 0.05, respectively, and tumor-to-cortex SUV ratios of 1.74 ± 0.07 and 1.44 ± 0.03, respectively. [18F]TFAHA accumulation was also observed in normal brain structures known to overexpress HDACs class IIa: hippocampus, n.accumbens, PAG, and cerebellum. These results were confirmed by immunohistochemical staining of brain tissue sections revealing the upregulation of HDACs 4, 5, and 9, and HIF-1α, hypoacetylation of H2AK5ac, H2BK5ac, H3K9ac, H4K8ac, and downregulation of KLF4. Significant reduction in [18F]TFAHA accumulation in 9L tumors was observed after administration of HDACs class IIa specific inhibitor MC1568, but not the SIRT1 specific inhibitor EX-527. Thus, PET/CT/(MRI) with [18F]TFAHA can facilitate studies to elucidate the roles of HDAC class IIa enzymes in gliomagenesis and progression and to optimize therapeutic doses of novel HDACs class IIa inhibitors in gliomas.
Collapse
|
98
|
Fratantonio D, Molonia MS, Bashllari R, Muscarà C, Ferlazzo G, Costa G, Saija A, Cimino F, Speciale A. Curcumin potentiates the antitumor activity of Paclitaxel in rat glioma C6 cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 55:23-30. [PMID: 30668434 DOI: 10.1016/j.phymed.2018.08.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 06/06/2018] [Accepted: 08/06/2018] [Indexed: 05/20/2023]
Abstract
BACKGROUND Glioma is the most common primary cancer in central nervous system, especially in brain. Paclitaxel (PTX) is a microtubule stabilizing agent with anticancer potential, but its clinical application to brain tumours is limited by drug resistance, side effects, and lower brain penetration. PURPOSE Herein we explored the in vitro effects, in glioma C6 cells, of the combination of PTX with curcumin, a natural compound with chemotherapeutic activity, in order to improve cytotoxic effects and overcome PTX limitations. RESULTS Our data confirmed PTX antiproliferative activity that was improved by curcumin. These effects were confirmed by clonogenic assay and G0/G1 cell cycle arrest. PTX significantly promoted generation of intracellular reactive species (RS), while curcumin did not affect RS production; the combination of the two drugs resulted in a slight but significant increase in RS levels. Furthermore, we found a constitutive activation of NF-κB in C6 cell line that was inhibited by PTX and curcumin. Interestingly, combination of the drugs totally inhibited NF-κB nuclear translocation and reduced IκB phosphorylation. Our results also supported the involvement of p53-p21 axis in the anticancer effects of curcumin and PTX. The combination of the two drugs further increased p53 and p21 levels enhancing the antiproliferative effects. Furthermore, PTX plus curcumin most impressively activated caspase-3, effector of apoptosis pathways, and reduced the expression of the anti-apoptotic protein Bcl-2. CONCLUSION In conclusion, our findings demonstrated that combination of PTX and curcumin exerts a potentiated anti-glioma efficacy in vitro that may help in reducing dosage and/or minimizing side effects of cytotoxic therapy.
Collapse
Affiliation(s)
- Deborah Fratantonio
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Annunziata, 98168 Messina, Italy
| | - Maria Sofia Molonia
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Annunziata, 98168 Messina, Italy
| | - Romina Bashllari
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Annunziata, 98168 Messina, Italy
| | - Claudia Muscarà
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Annunziata, 98168 Messina, Italy
| | - Guido Ferlazzo
- Laboratory of Immunology and Biotherapy, Dept. of Human Pathology, Center of Research Cell Factory UniMe, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy
| | - Gregorio Costa
- Laboratory of Immunology and Biotherapy, Dept. of Human Pathology, Center of Research Cell Factory UniMe, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy
| | - Antonella Saija
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Annunziata, 98168 Messina, Italy
| | - Francesco Cimino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Annunziata, 98168 Messina, Italy.
| | - Antonio Speciale
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Annunziata, 98168 Messina, Italy
| |
Collapse
|
99
|
Verhoeven J, Hulpia F, Kersemans K, Bolcaen J, De Lombaerde S, Goeman J, Descamps B, Hallaert G, Van den Broecke C, Deblaere K, Vanhove C, Van der Eycken J, Van Calenbergh S, Goethals I, De Vos F. New fluoroethyl phenylalanine analogues as potential LAT1-targeting PET tracers for glioblastoma. Sci Rep 2019; 9:2878. [PMID: 30814660 PMCID: PMC6393465 DOI: 10.1038/s41598-019-40013-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 02/07/2019] [Indexed: 02/07/2023] Open
Abstract
The use of O-(2-[18F]fluoroethyl)-L-tyrosine ([18F]FET) as a positron emission tomography (PET) tracer for brain tumor imaging might have some limitations because of the relatively low affinity for the L-type amino acid transporter 1 (LAT1). To assess the stereospecificity and evaluate the influence of aromatic ring modification of phenylalanine LAT1 targeting tracers, six different fluoroalkylated phenylalanine analogues were synthesized. After in vitro Ki determination, the most promising compound, 2-[18F]-2-fluoroethyl-L-phenylalanine (2-[18F]FELP), was selected for further evaluation and in vitro comparison with [18F]FET. Subsequently, 2-[18F]FELP was assessed in vivo and compared with [18F]FET and [18F]FDG in a F98 glioblastoma rat model. 2-[18F]FELP showed improved in vitro characteristics over [18F]FET, especially when the affinity and specificity for system L is concerned. Based on our results, 2-[18F]FELP is a promising new PET tracer for brain tumor imaging.
Collapse
Affiliation(s)
| | - Fabian Hulpia
- Laboratory for Medicinal Chemistry, Ghent University, Ghent, Belgium
| | - Ken Kersemans
- Ghent University Hospital, Department of Nuclear Medicine, Ghent, Belgium
| | - Julie Bolcaen
- Ghent University Hospital, Department of Nuclear Medicine, Ghent, Belgium
| | | | - Jan Goeman
- Laboratory for Organic and Bio-organic synthesis, Ghent University, Ghent, Belgium
| | - Benedicte Descamps
- IBiTech-MEDISIP Ghent University, Department of Electronics and Information Systems, Ghent, Belgium
| | - Giorgio Hallaert
- Ghent University Hospital, Department of Neurosurgery, Ghent, Belgium
| | | | - Karel Deblaere
- Ghent University Hospital, Department of Radiology and Medical Imaging, Ghent, Belgium
| | - Christian Vanhove
- IBiTech-MEDISIP Ghent University, Department of Electronics and Information Systems, Ghent, Belgium
| | - Johan Van der Eycken
- Laboratory for Organic and Bio-organic synthesis, Ghent University, Ghent, Belgium
| | | | - Ingeborg Goethals
- Ghent University Hospital, Department of Nuclear Medicine, Ghent, Belgium
| | - Filip De Vos
- Laboratory of Radiopharmacy, Ghent University, Ghent, Belgium
| |
Collapse
|
100
|
Prezado Y, Jouvion G, Guardiola C, Gonzalez W, Juchaux M, Bergs J, Nauraye C, Labiod D, De Marzi L, Pouzoulet F, Patriarca A, Dendale R. Tumor Control in RG2 Glioma-Bearing Rats: A Comparison Between Proton Minibeam Therapy and Standard Proton Therapy. Int J Radiat Oncol Biol Phys 2019; 104:266-271. [PMID: 30703513 DOI: 10.1016/j.ijrobp.2019.01.080] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 01/15/2019] [Accepted: 01/18/2019] [Indexed: 10/27/2022]
Abstract
PURPOSE Proton minibeam radiation therapy (pMBRT) is a novel radiation therapy approach that exploits the synergies of proton therapy with the gain in normal tissue preservation observed upon irradiation with narrow, spatially fractionated, beams. The net gain in normal tissue sparing that has been shown by pMBRT may lead to the efficient treatment of very radioresistant tumors, which are currently mostly treated palliatively. The aim of this study was to perform an evaluation of the tumor effectiveness of proton minibeam radiation therapy for the treatment of RG2 glioma-bearing rats. METHODS AND MATERIALS Two groups (n = 9) of RG2 glioma-bearing rats were irradiated with either standard proton therapy or with pMBRT, with a dose prescription of 25 Gy in 1 fraction. The animals were followed up for a maximum of 6 months. At the end of the study, histopathological studies were performed to assess both the tumor presence and the possible side effects. RESULTS Tumor control was achieved in the 2 irradiated series, with superior survival in the pMBRT group compared with the standard proton therapy group. Long-term (>170 days) survival rates of 22% and 67% were obtained in the standard proton therapy and pMBRT groups, respectively. No tumor was observed in the histopathological analysis. Although animals with long-term survival in the standard radiation therapy exhibit substantial brain damage, including marked radionecrosis, less severe toxicity was observed in the pMBRT group. CONCLUSIONS pMBRT offers a significant increase in the therapeutic index of brain tumors: The majority of the glioma-bearing rats (67%) survived 6 months with less severe side effects.
Collapse
Affiliation(s)
- Yolanda Prezado
- Laboratoire d'Imagerie et Modélisation en Neurobiologie et Cancérologie (IMNC), Centre National de la Recherche Scientifique (CNRS), Universités Paris 11 and Paris 7, Campus d'Orsay, Orsay, France.
| | - Gregory Jouvion
- Institut Pasteur, Neuropathologie Experimentale, Paris, France
| | - Consuelo Guardiola
- Laboratoire d'Imagerie et Modélisation en Neurobiologie et Cancérologie (IMNC), Centre National de la Recherche Scientifique (CNRS), Universités Paris 11 and Paris 7, Campus d'Orsay, Orsay, France
| | - Wilfredo Gonzalez
- Laboratoire d'Imagerie et Modélisation en Neurobiologie et Cancérologie (IMNC), Centre National de la Recherche Scientifique (CNRS), Universités Paris 11 and Paris 7, Campus d'Orsay, Orsay, France
| | - Marjorie Juchaux
- Laboratoire d'Imagerie et Modélisation en Neurobiologie et Cancérologie (IMNC), Centre National de la Recherche Scientifique (CNRS), Universités Paris 11 and Paris 7, Campus d'Orsay, Orsay, France
| | - Judith Bergs
- Laboratoire d'Imagerie et Modélisation en Neurobiologie et Cancérologie (IMNC), Centre National de la Recherche Scientifique (CNRS), Universités Paris 11 and Paris 7, Campus d'Orsay, Orsay, France
| | - Catherine Nauraye
- Institut Curie, PSL Research University, Radiation Oncology Department, Centre de Protonthérapie d'Orsay, Orsay, France
| | - Dalila Labiod
- Institut Curie, PSL Research University, Translational Research Department, Experimental Radiotherapy Platform, Orsay, France
| | - Ludovic De Marzi
- Institut Curie, PSL Research University, Radiation Oncology Department, Centre de Protonthérapie d'Orsay, Orsay, France
| | - Frederic Pouzoulet
- Institut Curie, PSL Research University, Translational Research Department, Experimental Radiotherapy Platform, Orsay, France
| | - Annalisa Patriarca
- Institut Curie, PSL Research University, Radiation Oncology Department, Centre de Protonthérapie d'Orsay, Orsay, France
| | - Remi Dendale
- Institut Curie, PSL Research University, Radiation Oncology Department, Centre de Protonthérapie d'Orsay, Orsay, France
| |
Collapse
|