301
|
Ghiaseddin A, Hoang Minh LB, Janiszewska M, Shin D, Wick W, Mitchell DA, Wen PY, Grossman SA. Adult precision medicine: learning from the past to enhance the future. Neurooncol Adv 2021; 3:vdaa145. [PMID: 33543142 PMCID: PMC7846182 DOI: 10.1093/noajnl/vdaa145] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Despite therapeutic advances for other malignancies, gliomas remain challenging solid tumors to treat. Complete surgical resection is nearly impossible due to gliomas’ diffuse infiltrative nature, and treatment is hampered by restricted access to the tumors due to limited transport across the blood–brain barrier. Recent advances in genomic studies and next-generation sequencing techniques have led to a better understanding of gliomas and identification of potential aberrant signaling pathways. Targeting the specific genomic abnormalities via novel molecular therapies has opened a new avenue in the management of gliomas, with encouraging results in preclinical studies and early clinical trials. However, molecular characterization of gliomas revealed significant heterogeneity, which poses a challenge for targeted therapeutic approaches. In this context, leading neuro-oncology researchers and clinicians, industry innovators, and patient advocates convened at the inaugural annual Remission Summit held in Orlando, FL in February 2019 to discuss the latest advances in immunotherapy and precision medicine approaches for the treatment of adult and pediatric brain tumors and outline the unanswered questions, challenges, and opportunities that lay ahead for advancing the duration and quality of life for patients with brain tumors. Here, we provide historical context for precision medicine in other cancers, present emerging approaches for gliomas, discuss their limitations, and outline the steps necessary for future success. We focus on the advances in small molecule targeted therapy, as the use of immunotherapy as an emerging precision medicine modality for glioma treatment has recently been reviewed by our colleagues.
Collapse
Affiliation(s)
- Ashley Ghiaseddin
- Department of Neurosurgery, University of Florida, Gainesville, Florida, USA
| | - Lan B Hoang Minh
- Department of Neurosurgery, University of Florida, Gainesville, Florida, USA
| | | | - David Shin
- Department of Neurosurgery, University of Florida, Gainesville, Florida, USA
| | - Wolfgang Wick
- Neurology Clinic, Heidelberg University Medical Center, Heidelberg, Germany
| | - Duane A Mitchell
- Department of Neurosurgery, University of Florida, Gainesville, Florida, USA
| | - Patrick Y Wen
- Brigham and Women's Hospital, Boston, Massachusetts, USA.,Department of Neurology, Harvard Medical School, Boston, Massachusetts, USA.,Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Stuart A Grossman
- Department of Oncology, Johns Hopkins Kimmel Cancer Center, Baltimore, Maryland, USA
| |
Collapse
|
302
|
Chatterjee K, Atay N, Abler D, Bhargava S, Sahoo P, Rockne RC, Munson JM. Utilizing Dynamic Contrast-Enhanced Magnetic Resonance Imaging (DCE-MRI) to Analyze Interstitial Fluid Flow and Transport in Glioblastoma and the Surrounding Parenchyma in Human Patients. Pharmaceutics 2021; 13:pharmaceutics13020212. [PMID: 33557069 PMCID: PMC7913790 DOI: 10.3390/pharmaceutics13020212] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/22/2021] [Accepted: 01/23/2021] [Indexed: 01/04/2023] Open
Abstract
Background: Glioblastoma (GBM) is the deadliest and most common brain tumor in adults, with poor survival and response to aggressive therapy. Limited access of drugs to tumor cells is one reason for such grim clinical outcomes. A driving force for therapeutic delivery is interstitial fluid flow (IFF), both within the tumor and in the surrounding brain parenchyma. However, convective and diffusive transport mechanisms are understudied. In this study, we examined the application of a novel image analysis method to measure fluid flow and diffusion in GBM patients. Methods: Here, we applied an imaging methodology that had been previously tested and validated in vitro, in silico, and in preclinical models of disease to archival patient data from the Ivy Glioblastoma Atlas Project (GAP) dataset. The analysis required the use of dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI), which is readily available in the database. The analysis results, which consisted of IFF flow velocity and diffusion coefficients, were then compared to patient outcomes such as survival. Results: We characterized IFF and diffusion patterns in patients. We found strong correlations between flow rates measured within tumors and in the surrounding parenchymal space, where we hypothesized that velocities would be higher. Analyzing overall magnitudes indicated a significant correlation with both age and survival in this patient cohort. Additionally, we found that neither tumor size nor resection significantly altered the velocity magnitude. Lastly, we mapped the flow pathways in patient tumors and found a variability in the degree of directionality that we hypothesize may lead to information concerning treatment, invasive spread, and progression in future studies. Conclusions: An analysis of standard DCE-MRI in patients with GBM offers more information regarding IFF and transport within and around the tumor, shows that IFF is still detected post-resection, and indicates that velocity magnitudes correlate with patient prognosis.
Collapse
Affiliation(s)
- Krishnashis Chatterjee
- Department of Biomedical Engineering & Mechanics, Fralin Biomedical Research Institute, Virginia Tech, Roanoke, VA 24016, USA; (K.C.); (N.A.); (S.B.)
| | - Naciye Atay
- Department of Biomedical Engineering & Mechanics, Fralin Biomedical Research Institute, Virginia Tech, Roanoke, VA 24016, USA; (K.C.); (N.A.); (S.B.)
| | - Daniel Abler
- Department of Computational and Quantitative Medicine, Division of Mathematical Oncology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA; (D.A.); (P.S.); (R.C.R.)
- ARTORG Center for Biomedical Engineering Research, University of Bern, 3008 Bern, Switzerland
| | - Saloni Bhargava
- Department of Biomedical Engineering & Mechanics, Fralin Biomedical Research Institute, Virginia Tech, Roanoke, VA 24016, USA; (K.C.); (N.A.); (S.B.)
| | - Prativa Sahoo
- Department of Computational and Quantitative Medicine, Division of Mathematical Oncology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA; (D.A.); (P.S.); (R.C.R.)
| | - Russell C. Rockne
- Department of Computational and Quantitative Medicine, Division of Mathematical Oncology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA; (D.A.); (P.S.); (R.C.R.)
| | - Jennifer M. Munson
- Department of Biomedical Engineering & Mechanics, Fralin Biomedical Research Institute, Virginia Tech, Roanoke, VA 24016, USA; (K.C.); (N.A.); (S.B.)
- Correspondence: ; Tel.: +1-(540)-532-6392
| |
Collapse
|
303
|
Traylor JI, Pernik MN, Sternisha AC, McBrayer SK, Abdullah KG. Molecular and Metabolic Mechanisms Underlying Selective 5-Aminolevulinic Acid-Induced Fluorescence in Gliomas. Cancers (Basel) 2021; 13:cancers13030580. [PMID: 33540759 PMCID: PMC7867275 DOI: 10.3390/cancers13030580] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 01/22/2021] [Accepted: 01/26/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary 5-aminolevulinic acid (5-ALA) is a medication that produces fluorescence in certain cancers, which enables surgeons to visualize tumor margins during surgery. Gliomas are brain tumors that can be difficult to fully resect due to their infiltrative nature. In this review we explored what is known about the mechanism of 5-ALA, recent discoveries that increase our understanding of that mechanism, and potential targets to increase fluorescence in lower grade gliomas. Abstract 5-aminolevulinic acid (5-ALA) is a porphyrin precursor in the heme synthesis pathway. When supplied exogenously, certain cancers consume 5-ALA and convert it to the fluorogenic metabolite protoporphyrin IX (PpIX), causing tumor-specific tissue fluorescence. Preoperative administration of 5-ALA is used to aid neurosurgical resection of high-grade gliomas such as glioblastoma, allowing for increased extent of resection and progression free survival for these patients. A subset of gliomas, especially low-grade tumors, do not accumulate PpIX intracellularly or readily fluoresce upon 5-ALA administration, making gross total resection difficult to achieve in diffuse lesions. We review existing literature on 5-ALA metabolism and PpIX accumulation to explore potential mechanisms of 5-ALA-induced glioma tissue fluorescence. Targeting the heme synthesis pathway and understanding its dysregulation in malignant tissues could aid the development of adjunct therapies to increase intraoperative fluorescence after 5-ALA treatment.
Collapse
Affiliation(s)
- Jeffrey I. Traylor
- Department of Neurological Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA; (J.I.T.); (M.N.P.)
| | - Mark N. Pernik
- Department of Neurological Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA; (J.I.T.); (M.N.P.)
| | - Alex C. Sternisha
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
| | - Samuel K. McBrayer
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
- Correspondence: (S.K.M.); (K.G.A.); Tel.: +1-(214)-648-3730 (S.K.M.); +1-(214)-645-2300 (K.G.A.)
| | - Kalil G. Abdullah
- Department of Neurological Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA; (J.I.T.); (M.N.P.)
- Correspondence: (S.K.M.); (K.G.A.); Tel.: +1-(214)-648-3730 (S.K.M.); +1-(214)-645-2300 (K.G.A.)
| |
Collapse
|
304
|
Tang M, Rich JN, Chen S. Biomaterials and 3D Bioprinting Strategies to Model Glioblastoma and the Blood-Brain Barrier. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2004776. [PMID: 33326131 PMCID: PMC7854518 DOI: 10.1002/adma.202004776] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 09/06/2020] [Indexed: 05/13/2023]
Abstract
Glioblastoma (GBM) is the most prevalent and lethal adult primary central nervous system cancer. An immunosuppresive and highly heterogeneous tumor microenvironment, restricted delivery of chemotherapy or immunotherapy through the blood-brain barrier (BBB), together with the brain's unique biochemical and anatomical features result in its universal recurrence and poor prognosis. As conventional models fail to predict therapeutic efficacy in GBM, in vitro 3D models of GBM and BBB leveraging patient- or healthy-individual-derived cells and biomaterials through 3D bioprinting technologies potentially mimic essential physiological and pathological features of GBM and BBB. 3D-bioprinted constructs enable investigation of cellular and cell-extracellular matrix interactions in a species-matched, high-throughput, and reproducible manner, serving as screening or drug delivery platforms. Here, an overview of current 3D-bioprinted GBM and BBB models is provided, elaborating on the microenvironmental compositions of GBM and BBB, relevant biomaterials to mimic the native tissues, and bioprinting strategies to implement the model fabrication. Collectively, 3D-bioprinted GBM and BBB models are promising systems and biomimetic alternatives to traditional models for more reliable mechanistic studies and preclinical drug screenings that may eventually accelerate the drug development process for GBM.
Collapse
Affiliation(s)
- Min Tang
- Department of NanoEngineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Jeremy N. Rich
- Division of Regenerative Medicine, Department of Medicine, Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92093, USA
| | - Shaochen Chen
- Department of NanoEngineering, University of California San Diego, La Jolla, CA, 92093, USA
- Department of Bioengineering, Materials Science and Engineering Program, Chemical Engineering Program, University of California San Diego, La Jolla, CA, 92093, USA
| |
Collapse
|
305
|
Vaidya KS, Mitten MJ, Zelaya-Lazo AL, Oleksijew A, Alvey C, Falls HD, Mishra S, Palma J, Ansell P, Phillips AC, Reilly EB, Anderson M, Boghaert ER. Synergistic therapeutic benefit by combining the antibody drug conjugate, depatux-m with temozolomide in pre-clinical models of glioblastoma with overexpression of EGFR. J Neurooncol 2021; 152:233-243. [PMID: 33517558 DOI: 10.1007/s11060-021-03703-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 01/16/2021] [Indexed: 12/11/2022]
Abstract
PURPOSE Depatux-m is an antibody drug conjugate (ADC) that targets and inhibits growth of cancer cells overexpressing the epidermal growth factor receptor (EGFR) or the 2-7 deletion mutant (EGFRvIII) in tumor models in vitro and in vivo. Treatment of patients suffering from relapsed/refractory glioblastoma (GBM) with a combination of depatux-m and temozolomide (TMZ) tended to increase overall survival. As a first step to understand the nature of the interaction between the two drugs, we investigated whether the interaction was synergistic, additive or antagonistic. METHODS The efficacy of ADCs, antibodies, TMZ and radiation was tested in xenograft models of GBM, U-87MG and U-87MG EGFRvIII. Both models express EGFR. U-87MG EGFRvIII was transduced to express EGFRvIII. Changes in tumor volume, biomarkers of cell death and apoptosis after treatment were used to measure efficacy of the various treatments. Synergism of depatux-m and TMZ was verified in three-dimensional cultures of U-87MG and U-87MG EGFRvIII by the method of Chou and Talalay. RESULTS Combined with TMZ and radiotherapy (RT), depatux-m inhibited xenograft growth of U-87MG and U-87MG EGFRvIII more than either treatment with depatux-m or TMZ + RT. Durability of the response to depatux-m + TMZ + RT or depatux-m + TMZ was more pronounced in U-87MG EGFRvIII than in U-87MG. Efficacy of depatux-m + TMZ was synergistic in U-87MG EGFRvIII and additive in U-87MG. CONCLUSION Adding depatux-m enhances the efficacy of standard of care therapy in preclinical models of GBM. Durability of response to depatux-m + TMZ in vivo and synergy of the drug-drug interaction correlates with the amount of antigen expressed by the tumor cells.
Collapse
Affiliation(s)
- Kedar S Vaidya
- In Vivo Pharmacology, Global Pharmaceutical Research and Development, Abbvie, 1 N Waukegan Road, North Chicago, IL, 60064, USA
| | - Michael J Mitten
- In Vivo Pharmacology, Global Pharmaceutical Research and Development, Abbvie, 1 N Waukegan Road, North Chicago, IL, 60064, USA
| | - Adelyn L Zelaya-Lazo
- In Vivo Pharmacology, Global Pharmaceutical Research and Development, Abbvie, 1 N Waukegan Road, North Chicago, IL, 60064, USA
| | - Anatol Oleksijew
- In Vivo Pharmacology, Global Pharmaceutical Research and Development, Abbvie, 1 N Waukegan Road, North Chicago, IL, 60064, USA
| | - Cory Alvey
- In Vivo Pharmacology, Global Pharmaceutical Research and Development, Abbvie, 1 N Waukegan Road, North Chicago, IL, 60064, USA
| | - Hugh D Falls
- Oncology Discovery, Global Pharmaceutical Research and Development, Abbvie, 1 N Waukegan Road, North Chicago, IL, 60064, USA
| | - Sasmita Mishra
- Translational Oncology, Global Pharmaceutical Research and Development, Abbvie, 1 N Waukegan Road, North Chicago, IL, 60064, USA
| | - Joann Palma
- Translational Oncology, Global Pharmaceutical Research and Development, Abbvie, 1 N Waukegan Road, North Chicago, IL, 60064, USA
| | - Peter Ansell
- Translational Oncology, Global Pharmaceutical Research and Development, Abbvie, 1 N Waukegan Road, North Chicago, IL, 60064, USA
| | - Andrew C Phillips
- Oncology Discovery, Global Pharmaceutical Research and Development, Abbvie, 1 N Waukegan Road, North Chicago, IL, 60064, USA
| | - Edward B Reilly
- Oncology Discovery, Global Pharmaceutical Research and Development, Abbvie, 1 N Waukegan Road, North Chicago, IL, 60064, USA
| | - Mark Anderson
- Oncology Discovery, Global Pharmaceutical Research and Development, Abbvie, 1 N Waukegan Road, North Chicago, IL, 60064, USA
| | - Erwin R Boghaert
- In Vivo Pharmacology, Global Pharmaceutical Research and Development, Abbvie, 1 N Waukegan Road, North Chicago, IL, 60064, USA.
| |
Collapse
|
306
|
Abstract
Cancer treatment remains a challenge due to a high level of intra- and intertumoral heterogeneity and the rapid development of chemoresistance. In the brain, this is further hampered by the blood-brain barrier that reduces passive diffusion of drugs to a minimum. Tumors grow invasively and form new blood vessels, also in brain tissue where remodeling of pre-existing vasculature is substantial. The cancer-associated vessels in the brain are considered leaky and thus could facilitate the transport of chemotherapeutic agents. Yet, brain tumors are extremely difficult to treat, and, in this review, we will address how different aspects of the vasculature in brain tumors contribute to this.
Collapse
Affiliation(s)
- Casper Hempel
- Dept of Health Technology, Technical University of Denmark, 2800, Kgs Lyngby, Denmark.
| | - Kasper B Johnsen
- Dept of Health Technology, Technical University of Denmark, 2800, Kgs Lyngby, Denmark
| | - Serhii Kostrikov
- Dept of Health Technology, Technical University of Denmark, 2800, Kgs Lyngby, Denmark
| | - Petra Hamerlik
- Brain Tumor Biology, Danish Cancer Society Research Center, 2100, Copenhagen, Denmark
| | - Thomas L Andresen
- Dept of Health Technology, Technical University of Denmark, 2800, Kgs Lyngby, Denmark.
| |
Collapse
|
307
|
Costagliola di Polidoro A, Zambito G, Haeck J, Mezzanotte L, Lamfers M, Netti PA, Torino E. Theranostic Design of Angiopep-2 Conjugated Hyaluronic Acid Nanoparticles (Thera-ANG-cHANPs) for Dual Targeting and Boosted Imaging of Glioma Cells. Cancers (Basel) 2021; 13:cancers13030503. [PMID: 33525655 PMCID: PMC7865309 DOI: 10.3390/cancers13030503] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/08/2021] [Accepted: 01/22/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Glioblastoma multiforme is the most aggressive malignant brain tumor with poor patient prognosis. The presence of the blood-brain barrier and the complex tumor microenvironment impair the efficient accumulation of drugs and contrast agents, causing late diagnosis, inefficient treatment and monitoring. Functionalized theranostic nanoparticles are a valuable tool to modulate biodistribution of active agents, promoting their active delivery and selective accumulation for an earlier diagnosis and effective treatment, and provide simultaneous therapy and imaging for improved evaluation of treatment efficacy. In this work, we developed angiopep-2 functionalized crosslinked hyaluronic acid nanoparticles encapsulating gadolinium-diethylenetriamine pentaacetic acid (Gd-DTPA) and irinotecan (Thera-ANG-cHANPs) that were shown to boost relaxometric properties of Gd-DTPA by the effect of Hydrodenticity, improve the uptake of nanoparticles by the exploitation of angiopep-2 improved transport properties, and accelerate the therapeutic effect of Irinotecan. Abstract Glioblastoma multiforme (GBM) has a mean survival of only 15 months. Tumour heterogeneity and blood-brain barrier (BBB) mainly hinder the transport of active agents, leading to late diagnosis, ineffective therapy and inaccurate follow-up. The use of hydrogel nanoparticles, particularly hyaluronic acid as naturally occurring polymer of the extracellular matrix (ECM), has great potential in improving the transport of drug molecules and, furthermore, in facilitatating the early diagnosis by the effect of hydrodenticity enabling the T1 boosting of Gadolinium chelates for MRI. Here, crosslinked hyaluronic acid nanoparticles encapsulating gadolinium-diethylenetriamine pentaacetic acid (Gd-DTPA) and the chemotherapeutic agent irinotecan (Thera-cHANPs) are proposed as theranostic nanovectors, with improved MRI capacities. Irinotecan was selected since currently repurposed as an alternative compound to the poorly effective temozolomide (TMZ), generally approved as the gold standard in GBM clinical care. Also, active crossing and targeting are achieved by theranostic cHANPs decorated with angiopep-2 (Thera-ANG-cHANPs), a dual-targeting peptide interacting with low density lipoprotein receptor related protein-1(LRP-1) receptors overexpressed by both endothelial cells of the BBB and glioma cells. Results showed preserving the hydrodenticity effect in the advanced formulation and internalization by the active peptide-mediated uptake of Thera-cHANPs in U87 and GS-102 cells. Moreover, Thera-ANG-cHANPs proved to reduce ironotecan time response, showing a significant cytotoxic effect in 24 h instead of 48 h.
Collapse
Affiliation(s)
- Angela Costagliola di Polidoro
- Department of Chemical, Materials and Production Engineering (DICMaPI), University of Naples Federico II, 80125 Naples, Italy; (A.C.d.P.); (P.A.N.)
- Fondazione Istituto Italiano di Tecnologia, IIT, 80125 Naples, Italy
| | - Giorgia Zambito
- Department of Molecular Genetics, Erasmus Medical Center, 3015 CN Rotterdam, The Netherlands; (G.Z.); (L.M.)
- Medres Medical Research GmBH, 50931 Cologne, Germany
- Department of Radiology and Nuclear Medicine, Erasmus Medical Center, 3015 CN Rotterdam, The Netherlands
| | - Joost Haeck
- AMIE Core Facility, Erasmus Medical Center, 3015 CN Rotterdam, The Netherlands;
| | - Laura Mezzanotte
- Department of Molecular Genetics, Erasmus Medical Center, 3015 CN Rotterdam, The Netherlands; (G.Z.); (L.M.)
- Medres Medical Research GmBH, 50931 Cologne, Germany
| | - Martine Lamfers
- Department of Neurosurgery, Brain Tumor Center, Erasmus Medical Center, 3015 CN Rotterdam, The Netherlands;
| | - Paolo Antonio Netti
- Department of Chemical, Materials and Production Engineering (DICMaPI), University of Naples Federico II, 80125 Naples, Italy; (A.C.d.P.); (P.A.N.)
- Fondazione Istituto Italiano di Tecnologia, IIT, 80125 Naples, Italy
- AMIE Core Facility, Erasmus Medical Center, 3015 CN Rotterdam, The Netherlands;
| | - Enza Torino
- Department of Chemical, Materials and Production Engineering (DICMaPI), University of Naples Federico II, 80125 Naples, Italy; (A.C.d.P.); (P.A.N.)
- Interdisciplinary Research Center on Biomaterials, CRIB, University of Naples Federico II, 80125 Naples, Italy
- Correspondence:
| |
Collapse
|
308
|
Duraj T, García-Romero N, Carrión-Navarro J, Madurga R, Ortiz de Mendivil A, Prat-Acin R, Garcia-Cañamaque L, Ayuso-Sacido A. Beyond the Warburg Effect: Oxidative and Glycolytic Phenotypes Coexist within the Metabolic Heterogeneity of Glioblastoma. Cells 2021; 10:202. [PMID: 33498369 PMCID: PMC7922554 DOI: 10.3390/cells10020202] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/16/2021] [Accepted: 01/18/2021] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma (GBM) is the most aggressive primary brain tumor, with a median survival at diagnosis of 16-20 months. Metabolism represents a new attractive therapeutic target; however, due to high intratumoral heterogeneity, the application of metabolic drugs in GBM is challenging. We characterized the basal bioenergetic metabolism and antiproliferative potential of metformin (MF), dichloroacetate (DCA), sodium oxamate (SOD) and diazo-5-oxo-L-norleucine (DON) in three distinct glioma stem cells (GSCs) (GBM18, GBM27, GBM38), as well as U87MG. GBM27, a highly oxidative cell line, was the most resistant to all treatments, except DON. GBM18 and GBM38, Warburg-like GSCs, were sensitive to MF and DCA, respectively. Resistance to DON was not correlated with basal metabolic phenotypes. In combinatory experiments, radiomimetic bleomycin exhibited therapeutically relevant synergistic effects with MF, DCA and DON in GBM27 and DON in all other cell lines. MF and DCA shifted the metabolism of treated cells towards glycolysis or oxidation, respectively. DON consistently decreased total ATP production. Our study highlights the need for a better characterization of GBM from a metabolic perspective. Metabolic therapy should focus on both glycolytic and oxidative subpopulations of GSCs.
Collapse
Affiliation(s)
- Tomás Duraj
- Faculty of Medicine, Institute for Applied Molecular Medicine (IMMA), CEU San Pablo University, 28668 Madrid, Spain;
| | - Noemí García-Romero
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, 28223 Madrid, Spain; (N.G.-R.); (J.C.-N.); (R.M.)
- Brain Tumor Laboratory, Fundación Vithas, Grupo Hospitales Vithas, 28043 Madrid, Spain
| | - Josefa Carrión-Navarro
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, 28223 Madrid, Spain; (N.G.-R.); (J.C.-N.); (R.M.)
- Brain Tumor Laboratory, Fundación Vithas, Grupo Hospitales Vithas, 28043 Madrid, Spain
| | - Rodrigo Madurga
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, 28223 Madrid, Spain; (N.G.-R.); (J.C.-N.); (R.M.)
- Brain Tumor Laboratory, Fundación Vithas, Grupo Hospitales Vithas, 28043 Madrid, Spain
| | | | - Ricardo Prat-Acin
- Neurosurgery Department, Hospital Universitario La Fe, 46026 Valencia, Spain;
| | | | - Angel Ayuso-Sacido
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, 28223 Madrid, Spain; (N.G.-R.); (J.C.-N.); (R.M.)
- Brain Tumor Laboratory, Fundación Vithas, Grupo Hospitales Vithas, 28043 Madrid, Spain
| |
Collapse
|
309
|
de Gooijer MC, Kemper EM, Buil LCM, Çitirikkaya CH, Buckle T, Beijnen JH, van Tellingen O. ATP-binding cassette transporters restrict drug delivery and efficacy against brain tumors even when blood-brain barrier integrity is lost. CELL REPORTS MEDICINE 2021; 2:100184. [PMID: 33521698 PMCID: PMC7817868 DOI: 10.1016/j.xcrm.2020.100184] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 07/21/2020] [Accepted: 12/16/2020] [Indexed: 12/22/2022]
Abstract
The impact of a compromised blood-brain barrier (BBB) on the drug treatment of intracranial tumors remains controversial. We characterize the BBB integrity in several intracranial tumor models using magnetic resonance imaging, fluorescent dyes, and autoradiography and determine the distribution and efficacy of docetaxel in brain tumors grafted in Abcb1-proficient and Abcb1-deficient mice. Leakiness of the tumor vasculature varies from extensive to absent. Regardless of the extent of leakiness, tumor blood vessels express ATP-binding cassette transporters (Abcb1 and Abcg2). A leaky vasculature results in higher docetaxel tumor levels compared to normal brain. Nevertheless, Abcb1 can reduce drug distribution and efficacy even in leaky models. Thus, BBB leakiness does not ensure the unimpeded access of ATP-binding cassette transporter substrate drugs. Therapeutic responses may be observed, but the full potential of such therapeutics may still be attenuated. Consequently, BBB-penetrable drugs with little to no affinity for efflux transporters are preferred for the treatment of intracranial tumors. Blood-brain barrier integrity in brain tumor models varies from intact to absent Brain tumor vessels express drug efflux transporters Drug transporters can impede drug entry and efficacy, even in leaky tumors Low-affinity ABC transporter drugs are favored candidates for treating brain tumors
Collapse
Affiliation(s)
- Mark C de Gooijer
- Division of Pharmacology, the Netherlands Cancer Institute, Amsterdam, the Netherlands.,Mouse Cancer Clinic, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - E Marleen Kemper
- Department of Hospital Pharmacy, Academic Medical Center, Amsterdam, the Netherlands
| | - Levi C M Buil
- Division of Pharmacology, the Netherlands Cancer Institute, Amsterdam, the Netherlands.,Mouse Cancer Clinic, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Ceren H Çitirikkaya
- Division of Pharmacology, the Netherlands Cancer Institute, Amsterdam, the Netherlands.,Mouse Cancer Clinic, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Tessa Buckle
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Jos H Beijnen
- Division of Pharmacology, the Netherlands Cancer Institute, Amsterdam, the Netherlands.,Department of Pharmacy and Pharmacology, the Netherlands Cancer Institute, Amsterdam, the Netherlands.,Division of Pharmacoepidemiology and Clinical Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | - Olaf van Tellingen
- Division of Pharmacology, the Netherlands Cancer Institute, Amsterdam, the Netherlands.,Mouse Cancer Clinic, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| |
Collapse
|
310
|
Finch A, Solomou G, Wykes V, Pohl U, Bardella C, Watts C. Advances in Research of Adult Gliomas. Int J Mol Sci 2021; 22:ijms22020924. [PMID: 33477674 PMCID: PMC7831916 DOI: 10.3390/ijms22020924] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/08/2021] [Accepted: 01/11/2021] [Indexed: 01/03/2023] Open
Abstract
Diffuse gliomas are the most frequent brain tumours, representing 75% of all primary malignant brain tumours in adults. Because of their locally aggressive behaviour and the fact that they cannot be cured by current therapies, they represent one of the most devastating cancers. The present review summarises recent advances in our understanding of glioma development and progression by use of various in vitro and in vivo models, as well as more complex techniques including cultures of 3D organoids and organotypic slices. We discuss the progress that has been made in understanding glioma heterogeneity, alteration in gene expression and DNA methylation, as well as advances in various in silico models. Lastly current treatment options and future clinical trials, which aim to improve early diagnosis and disease monitoring, are also discussed.
Collapse
Affiliation(s)
- Alina Finch
- Institute of Cancer Genomic Sciences, University of Birmingham, Birmingham B15 2TT, UK; (A.F.); (G.S.); (V.W.)
| | - Georgios Solomou
- Institute of Cancer Genomic Sciences, University of Birmingham, Birmingham B15 2TT, UK; (A.F.); (G.S.); (V.W.)
- School of Medicine, Keele University, Staffordshire ST5 5NL, UK
| | - Victoria Wykes
- Institute of Cancer Genomic Sciences, University of Birmingham, Birmingham B15 2TT, UK; (A.F.); (G.S.); (V.W.)
- Department of Neurosurgery, University Hospital Birmingham, Birmingham B15 2WB, UK
| | - Ute Pohl
- Department of Cellular Pathology, University Hospital Birmingham, Birmingham B15 2WB, UK;
| | - Chiara Bardella
- Institute of Cancer Genomic Sciences, University of Birmingham, Birmingham B15 2TT, UK; (A.F.); (G.S.); (V.W.)
- Correspondence: (C.B.); (C.W.)
| | - Colin Watts
- Institute of Cancer Genomic Sciences, University of Birmingham, Birmingham B15 2TT, UK; (A.F.); (G.S.); (V.W.)
- Department of Neurosurgery, University Hospital Birmingham, Birmingham B15 2WB, UK
- Correspondence: (C.B.); (C.W.)
| |
Collapse
|
311
|
Abstract
For a chemotherapeutic agent to be effective, it must conquer the presence of blood-brain barrier (BBB), which limits the penetration of drugs into the brain. Tumours in the brain compromise the integrity of BBB and result in a highly heterogeneous vasculature, known as blood-brain tumour barrier (BBTB). In this chapter, we firstly highlight the cellular and molecular characteristics of the BBB and BBTB as well as the challenges aroused by BBB/BBTB for drug delivery. Secondly, we discuss the current strategies overcoming the challenges in invasive and non-invasive manners. Finally, we highlight the emerging strategy using focused ultrasound (FUS) with systemic microbubbles to transiently and reversibly enhance the permeability of these barriers for drug delivery.
Collapse
|
312
|
Segura-Collar B, Mata-Martínez P, Hernández-Laín A, Sánchez-Gómez P, Gargini R. Blood-Brain Barrier Disruption: A Common Driver of Central Nervous System Diseases. Neuroscientist 2021; 28:222-237. [PMID: 33446074 DOI: 10.1177/1073858420985838] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The brain is endowed with a unique cellular composition and organization, embedded within a vascular network and isolated from the circulating blood by a specialized frontier, the so-called blood-brain barrier (BBB), which is necessary for its proper function. Recent reports have shown that increments in the permeability of the blood vessels facilitates the entry of toxic components and immune cells to the brain parenchyma and alters the phenotype of the supporting astrocytes. All of these might contribute to the progression of different pathologies such as brain cancers or neurodegenerative diseases. Although it is well known that BBB breakdown occurs due to pericyte malfunctioning or to the lack of stability of the blood vessels, its participation in the diverse neural diseases needs further elucidation. This review summarizes what it is known about BBB structure and function and how its instability might trigger or promote neuronal degeneration and glioma progression, with a special focus on the role of pericytes as key modulators of the vasculature. Moreover, we will discuss some recent reports that highlights the participation of the BBB alterations in glioma growth. This pan-disease analysis might shed some light into these otherwise untreatable diseases and help to design better therapeutic approaches.
Collapse
Affiliation(s)
| | | | | | | | - Ricardo Gargini
- Neurooncology Unit, Instituto de Salud Carlos III-UFIEC, Madrid, Spain
| |
Collapse
|
313
|
Jovito BL, Paterno LG, Sales MJA, Gross MA, Silva LP, de Souza P, Báo SN. Graphene Oxide/Zinc Oxide Nanocomposite Displaying Selective Toxicity to Glioblastoma Cell Lines. ACS APPLIED BIO MATERIALS 2021. [DOI: 10.1021/acsabm.0c01369] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- Bárbara L. Jovito
- Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil
- Graduate Program in Animal Biology, University of Brasilia, Brasilia 70910-900, Brazil
| | | | - Maria J. A. Sales
- Institute of Chemistry, University of Brasilia, Brasilia 70910-900, Brazil
| | - Marcos A. Gross
- Institute of Chemistry, University of Brasilia, Brasilia 70910-900, Brazil
| | - Luciano P. Silva
- Embrapa Genetic Resources and Biotechnology, Brasilia 70770-917, Brazil
| | - Paulo de Souza
- Institute of Physics, University of Brasilia, Brasilia 70910-900, Brazil
| | - Sônia N. Báo
- Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil
| |
Collapse
|
314
|
Piantino M, Figarol A, Matsusaki M. Three-Dimensional in vitro Models of Healthy and Tumor Brain Microvasculature for Drug and Toxicity Screening. FRONTIERS IN TOXICOLOGY 2021; 3:656254. [PMID: 35295158 PMCID: PMC8915870 DOI: 10.3389/ftox.2021.656254] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 03/31/2021] [Indexed: 12/16/2022] Open
Abstract
Tissue vascularization is essential for its oxygenation and the homogenous diffusion of nutrients. Cutting-edge studies are focusing on the vascularization of three-dimensional (3D) in vitro models of human tissues. The reproduction of the brain vasculature is particularly challenging as numerous cell types are involved. Moreover, the blood-brain barrier, which acts as a selective filter between the vascular system and the brain, is a complex structure to replicate. Nevertheless, tremendous advances have been made in recent years, and several works have proposed promising 3D in vitro models of the brain microvasculature. They incorporate cell co-cultures organized in 3D scaffolds, often consisting of components of the native extracellular matrix (ECM), to obtain a micro-environment similar to the in vivo physiological state. These models are particularly useful for studying adverse effects on the healthy brain vasculature. They provide insights into the molecular and cellular events involved in the pathological evolutions of this vasculature, such as those supporting the appearance of brain cancers. Glioblastoma multiform (GBM) is the most common form of brain cancer and one of the most vascularized solid tumors. It is characterized by a high aggressiveness and therapy resistance. Current conventional therapies are unable to prevent the high risk of recurrence of the disease. Most of the new drug candidates fail to pass clinical trials, despite the promising results shown in vitro. The conventional in vitro models are unable to efficiently reproduce the specific features of GBM tumors. Recent studies have indeed suggested a high heterogeneity of the tumor brain vasculature, with the coexistence of intact and leaky regions resulting from the constant remodeling of the ECM by glioma cells. In this review paper, after summarizing the advances in 3D in vitro brain vasculature models, we focus on the latest achievements in vascularized GBM modeling, and the potential applications for both healthy and pathological models as platforms for drug screening and toxicological assays. Particular attention will be paid to discuss the relevance of these models in terms of cell-cell, cell-ECM interactions, vascularization and permeability properties, which are crucial parameters for improving in vitro testing accuracy.
Collapse
Affiliation(s)
- Marie Piantino
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Osaka, Japan
| | - Agathe Figarol
- Institut Jean Lamour, UMR 7198 CNRS, Université de Lorraine, Nancy, France
| | - Michiya Matsusaki
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Osaka, Japan
- *Correspondence: Michiya Matsusaki
| |
Collapse
|
315
|
Riva M, Lopci E, Gay LG, Nibali MC, Rossi M, Sciortino T, Castellano A, Bello L. Advancing Imaging to Enhance Surgery: From Image to Information Guidance. Neurosurg Clin N Am 2021; 32:31-46. [PMID: 33223024 DOI: 10.1016/j.nec.2020.08.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Conventional magnetic resonance imaging (cMRI) has an established role as a crucial disease parameter in the multidisciplinary management of glioblastoma, guiding diagnosis, treatment planning, assessment, and follow-up. Yet, cMRI cannot provide adequate information regarding tissue heterogeneity and the infiltrative extent beyond the contrast enhancement. Advanced magnetic resonance imaging and PET and newer analytical methods are transforming images into data (radiomics) and providing noninvasive biomarkers of molecular features (radiogenomics), conveying enhanced information for improving decision making in surgery. This review analyzes the shift from image guidance to information guidance that is relevant for the surgical treatment of glioblastoma.
Collapse
Affiliation(s)
- Marco Riva
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Via Festa del Perdono 7, Milan 20122, Italy; IRCCS Istituto Ortopedico Galeazzi, U.O. Neurochirurgia Oncologica, Milan, Italy.
| | - Egesta Lopci
- Unit of Nuclear Medicine, Humanitas Clinical and Research Center - IRCCS, Via Manzoni 56, Rozzano, Milan 20089, Italy. https://twitter.com/LopciEgesta
| | - Lorenzo G Gay
- IRCCS Istituto Ortopedico Galeazzi, U.O. Neurochirurgia Oncologica, Milan, Italy; Department of Oncology and Hemato-Oncology, Via Festa del Perdono 7, Milan 20122, Italy
| | - Marco Conti Nibali
- IRCCS Istituto Ortopedico Galeazzi, U.O. Neurochirurgia Oncologica, Milan, Italy; Department of Oncology and Hemato-Oncology, Via Festa del Perdono 7, Milan 20122, Italy. https://twitter.com/dr_mcn
| | - Marco Rossi
- IRCCS Istituto Ortopedico Galeazzi, U.O. Neurochirurgia Oncologica, Milan, Italy; Department of Oncology and Hemato-Oncology, Via Festa del Perdono 7, Milan 20122, Italy
| | - Tommaso Sciortino
- IRCCS Istituto Ortopedico Galeazzi, U.O. Neurochirurgia Oncologica, Milan, Italy; Department of Oncology and Hemato-Oncology, Via Festa del Perdono 7, Milan 20122, Italy
| | - Antonella Castellano
- Neuroradiology Unit and CERMAC, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, Milan 20123, Italy. https://twitter.com/antocastella
| | - Lorenzo Bello
- IRCCS Istituto Ortopedico Galeazzi, U.O. Neurochirurgia Oncologica, Milan, Italy; Department of Oncology and Hemato-Oncology, Via Festa del Perdono 7, Milan 20122, Italy
| |
Collapse
|
316
|
Ibarra LE, Vilchez ML, Caverzán MD, Milla Sanabria LN. Understanding the glioblastoma tumor biology to optimize photodynamic therapy: From molecular to cellular events. J Neurosci Res 2020; 99:1024-1047. [PMID: 33370846 DOI: 10.1002/jnr.24776] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 11/29/2020] [Indexed: 12/19/2022]
Abstract
Photodynamic therapy (PDT) has recently gained attention as an alternative treatment of malignant gliomas. Glioblastoma (GBM) is the most prevalent within tumors of the central nervous system (CNS). Conventional treatments for this CNS tumor include surgery, radiation, and chemotherapy. Surgery is still being considered as the treatment of choice. Even so, the poor prognosis and/or recurrence of the disease after applying any of these treatments highlight the urgency of exploring new therapies and/or improving existing ones to achieve the definitive eradication of tumor masses and remaining cells. PDT is a therapeutic modality that involves the destruction of tumor cells by reactive oxygen species induced by light, which were previously treated with a photosensitizing agent. However, in recent years, its experimental application has expanded to other effects that could improve overall performance against GBM. In the current review, we revisit the main advances of PDT for GBM management and also, the recent mechanistic insights about cellular and molecular aspects related to tumoral resistance to PDT of GBM.
Collapse
Affiliation(s)
- Luis Exequiel Ibarra
- Instituto de Biotecnología Ambiental y Salud (INBIAS), Universidad Nacional de Río Cuarto (UNRC) y Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Río Cuarto, Argentina.,Departamento de Biología Molecular, Facultad de Ciencias Exactas Físico-Químicas y Naturales, UNRC, Río Cuarto, Argentina
| | - María Laura Vilchez
- Instituto de Biotecnología Ambiental y Salud (INBIAS), Universidad Nacional de Río Cuarto (UNRC) y Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Río Cuarto, Argentina.,Departamento de Biología Molecular, Facultad de Ciencias Exactas Físico-Químicas y Naturales, UNRC, Río Cuarto, Argentina
| | - Matías Daniel Caverzán
- Departamento de Biología Molecular, Facultad de Ciencias Exactas Físico-Químicas y Naturales, UNRC, Río Cuarto, Argentina
| | - Laura Natalia Milla Sanabria
- Instituto de Biotecnología Ambiental y Salud (INBIAS), Universidad Nacional de Río Cuarto (UNRC) y Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Río Cuarto, Argentina.,Departamento de Biología Molecular, Facultad de Ciencias Exactas Físico-Químicas y Naturales, UNRC, Río Cuarto, Argentina
| |
Collapse
|
317
|
Affiliation(s)
- Ranjit S Bindra
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
318
|
Wei HJ, Upadhyayula PS, Pouliopoulos AN, Englander ZK, Zhang X, Jan CI, Guo J, Mela A, Zhang Z, Wang TJC, Bruce JN, Canoll PD, Feldstein NA, Zacharoulis S, Konofagou EE, Wu CC. Focused Ultrasound-Mediated Blood-Brain Barrier Opening Increases Delivery and Efficacy of Etoposide for Glioblastoma Treatment. Int J Radiat Oncol Biol Phys 2020; 110:539-550. [PMID: 33346092 DOI: 10.1016/j.ijrobp.2020.12.019] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 11/22/2020] [Accepted: 12/13/2020] [Indexed: 12/12/2022]
Abstract
PURPOSE Glioblastoma (GBM) is a devastating disease. With the current treatment of surgery followed by chemoradiation, outcomes remain poor, with median survival of only 15 months and a 5-year survival rate of 6.8%. A challenge in treating GBM is the heterogeneous integrity of the blood-brain barrier (BBB), which limits the bioavailability of systemic therapies to the brain. There is a growing interest in enhancing drug delivery by opening the BBB with the use of focused ultrasound (FUS). We hypothesize that an FUS-mediated BBB opening can enhance the delivery of etoposide for a therapeutic benefit in GBM. METHODS AND MATERIALS A murine glioma cell line (Pdgf+, Pten-/-, P53-/-) was orthotopically injected into B6(Cg)-Tyrc-2J/J mice to establish the syngeneic GBM model for this study. Animals were treated with FUS and microbubbles to open the BBB to enhance the delivery of systemic etoposide. Magnetic resonance (MR) imaging was used to evaluate the BBB opening and tumor progression. Liquid chromatography tandem mass spectrometry was used to measure etoposide concentrations in the intracranial tumors. RESULTS The murine glioma cell line is sensitive to etoposide in vitro. MR imaging and passive cavitation detection demonstrate the safe and successful BBB opening with FUS. The combined treatment of an FUS-mediated BBB opening and etoposide decreased tumor growth by 45% and prolonged median overall survival by 6 days: an approximately 30% increase. The FUS-mediated BBB opening increased the brain tumor-to-serum ratio of etoposide by 3.5-fold and increased the etoposide concentration in brain tumor tissue by 8-fold compared with treatment without ultrasound. CONCLUSIONS The current study demonstrates that BBB opening with FUS increases intratumoral delivery of etoposide in the brain, resulting in local control and overall survival benefits.
Collapse
Affiliation(s)
- Hong-Jian Wei
- Department of Radiation Oncology, Columbia University Irving Medical Center, New York, New York
| | - Pavan S Upadhyayula
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, New York
| | | | - Zachary K Englander
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, New York
| | - Xu Zhang
- Institute for Cancer Genetics, Columbia University Irving Medical Center, New York, New York; Department of Pediatrics, Columbia University Irving Medical Center, New York, New York
| | - Chia-Ing Jan
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York; Division of Molecular Pathology, Department of Pathology, China Medical University and Hospital, Taichung, Taiwan; Department of Medicine, China Medical University, Taichung, Taiwan; Translational Cell Therapy Center, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Jia Guo
- Department of Psychiatry, Columbia University, New York, New York
| | - Angeliki Mela
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York
| | - Zhiguo Zhang
- Institute for Cancer Genetics, Columbia University Irving Medical Center, New York, New York; Department of Pediatrics, Columbia University Irving Medical Center, New York, New York
| | - Tony J C Wang
- Department of Radiation Oncology, Columbia University Irving Medical Center, New York, New York; Department of Neurological Surgery, Columbia University Irving Medical Center, New York, New York; Herbert Irving Comprehensive Cancer Center, New York, New York
| | - Jeffrey N Bruce
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, New York; Herbert Irving Comprehensive Cancer Center, New York, New York
| | - Peter D Canoll
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York; Herbert Irving Comprehensive Cancer Center, New York, New York
| | - Neil A Feldstein
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, New York
| | - Stergios Zacharoulis
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York
| | - Elisa E Konofagou
- Department of Biomedical Engineering, Columbia University, New York, New York
| | - Cheng-Chia Wu
- Department of Radiation Oncology, Columbia University Irving Medical Center, New York, New York; Herbert Irving Comprehensive Cancer Center, New York, New York.
| |
Collapse
|
319
|
Hu LS, Brat DJ, Bloch O, Ramkissoon S, Lesser GJ. The Practical Application of Emerging Technologies Influencing the Diagnosis and Care of Patients With Primary Brain Tumors. Am Soc Clin Oncol Educ Book 2020; 40:1-12. [PMID: 32324425 DOI: 10.1200/edbk_280955] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Over the past decade, a variety of new and innovative technologies has led to important advances in the diagnosis and management of patients with primary malignant brain tumors. New approaches to surgical navigation and tumor localization, advanced imaging to define tumor biology and treatment response, and the widespread adoption of a molecularly defined integrated diagnostic paradigm that complements traditional histopathologic diagnosis continue to impact the day-to-day care of these patients. In the neuro-oncology clinic, discussions with patients about the role of tumor treating fields (TTFields) and the incorporation of next-generation sequencing (NGS) data into therapeutic decision-making are now a standard practice. This article summarizes newer applications of technology influencing the pathologic, neuroimaging, neurosurgical, and medical management of patients with malignant primary brain tumors.
Collapse
Affiliation(s)
- Leland S Hu
- Neuroradiology Section, Department of Radiology, Mayo Clinic, Phoenix, AZ
| | - Daniel J Brat
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Orin Bloch
- Department of Neurologic Surgery, UC Davis Comprehensive Cancer Center, Sacramento, CA
| | - Shakti Ramkissoon
- Foundation Medicine, Inc., Morrisville, NC.,Comprehensive Cancer Center, Wake Forest Baptist Health, Winston-Salem, NC.,Department of Pathology, Wake Forest School of Medicine, Winston-Salem, NC
| | - Glenn J Lesser
- Comprehensive Cancer Center, Wake Forest Baptist Health, Winston-Salem, NC
| |
Collapse
|
320
|
Griffith JI, Rathi S, Zhang W, Zhang W, Drewes LR, Sarkaria JN, Elmquist WF. Addressing BBB Heterogeneity: A New Paradigm for Drug Delivery to Brain Tumors. Pharmaceutics 2020; 12:E1205. [PMID: 33322488 PMCID: PMC7763839 DOI: 10.3390/pharmaceutics12121205] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/08/2020] [Accepted: 12/10/2020] [Indexed: 12/11/2022] Open
Abstract
Effective treatments for brain tumors remain one of the most urgent and unmet needs in modern oncology. This is due not only to the presence of the neurovascular unit/blood-brain barrier (NVU/BBB) but also to the heterogeneity of barrier alteration in the case of brain tumors, which results in what is referred to as the blood-tumor barrier (BTB). Herein, we discuss this heterogeneity, how it contributes to the failure of novel pharmaceutical treatment strategies, and why a "whole brain" approach to the treatment of brain tumors might be beneficial. We discuss various methods by which these obstacles might be overcome and assess how these strategies are progressing in the clinic. We believe that by approaching brain tumor treatment from this perspective, a new paradigm for drug delivery to brain tumors might be established.
Collapse
Affiliation(s)
- Jessica I. Griffith
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN 55455, USA; (S.R.); (W.Z.); (W.Z.)
| | - Sneha Rathi
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN 55455, USA; (S.R.); (W.Z.); (W.Z.)
| | - Wenqiu Zhang
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN 55455, USA; (S.R.); (W.Z.); (W.Z.)
| | - Wenjuan Zhang
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN 55455, USA; (S.R.); (W.Z.); (W.Z.)
| | - Lester R. Drewes
- Department of Biomedical Sciences, University of Minnesota Medical School—Duluth, Duluth, MN 55812, USA;
| | - Jann N. Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN 55902, USA;
| | - William F. Elmquist
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN 55455, USA; (S.R.); (W.Z.); (W.Z.)
| |
Collapse
|
321
|
Borah BM, Cacaccio J, Durrani FA, Bshara W, Turowski SG, Spernyak JA, Pandey RK. Sonodynamic therapy in combination with photodynamic therapy shows enhanced long-term cure of brain tumor. Sci Rep 2020; 10:21791. [PMID: 33311561 PMCID: PMC7732989 DOI: 10.1038/s41598-020-78153-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 11/20/2020] [Indexed: 11/09/2022] Open
Abstract
This article presents the construction of a multimodality platform that can be used for efficient destruction of brain tumor by a combination of photodynamic and sonodynamic therapy. For in vivo studies, U87 patient-derived xenograft tumors were implanted subcutaneously in SCID mice. For the first time, it has been shown that the cell-death mechanism by both treatment modalities follows two different pathways. For example, exposing the U87 cells after 24 h incubation with HPPH [3-(1'-hexyloxy)ethyl-3-devinyl-pyropheophorbide-a) by ultrasound participate in an electron-transfer process with the surrounding biological substrates to form radicals and radical ions (Type I reaction); whereas in photodynamic therapy, the tumor destruction is mainly caused by highly reactive singlet oxygen (Type II reaction). The combination of photodynamic therapy and sonodynamic therapy both in vitro and in vivo have shown an improved cell kill/tumor response, that could be attributed to an additive and/or synergetic effect(s). Our results also indicate that the delivery of the HPPH to tumors can further be enhanced by using cationic polyacrylamide nanoparticles as a delivery vehicle. Exposing the nano-formulation with ultrasound also triggered the release of photosensitizer. The combination of photodynamic therapy and sonodynamic therapy strongly affects tumor vasculature as determined by dynamic contrast enhanced imaging using HSA-Gd(III)DTPA.
Collapse
Affiliation(s)
- Ballav M Borah
- Photolitec, LLC, 73 High Street, Buffalo, NY, 14203, USA
| | - Joseph Cacaccio
- Department of Cell Stress Biology, Photodynamic Therapy Center, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Farukh A Durrani
- Department of Cell Stress Biology, Photodynamic Therapy Center, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Wiam Bshara
- Department of Pathology, Pathology Network Shared Resource, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Steven G Turowski
- Translational Imaging Shared Resource, Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | | | - Ravindra K Pandey
- Department of Cell Stress Biology, Photodynamic Therapy Center, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA.
| |
Collapse
|
322
|
Brachi G, Ruiz-Ramírez J, Dogra P, Wang Z, Cristini V, Ciardelli G, Rostomily RC, Ferrari M, Mikheev AM, Blanco E, Mattu C. Intratumoral injection of hydrogel-embedded nanoparticles enhances retention in glioblastoma. NANOSCALE 2020; 12:23838-23850. [PMID: 33237080 PMCID: PMC8062960 DOI: 10.1039/d0nr05053a] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 10/24/2020] [Indexed: 05/07/2023]
Abstract
Intratumoral drug delivery is a promising approach for the treatment of glioblastoma multiforme (GBM). However, drug washout remains a major challenge in GBM therapy. Our strategy, aimed at reducing drug clearance and enhancing site-specific residence time, involves the local administration of a multi-component system comprised of nanoparticles (NPs) embedded within a thermosensitive hydrogel (HG). Herein, our objective was to examine the distribution of NPs and their cargo following intratumoral administration of this system in GBM. We hypothesized that the HG matrix, which undergoes rapid gelation upon increases in temperature, would contribute towards heightened site-specific retention and permanence of NPs in tumors. BODIPY-containing, infrared dye-labeled polymeric NPs embedded in a thermosensitive HG (HG-NPs) were fabricated and characterized. Retention and distribution dynamics were subsequently examined over time in orthotopic GBM-bearing mice. Results demonstrate that the HG-NPs system significantly improved site-specific, long-term retention of both NPs and BODIPY, with co-localization analyses showing that HG-NPs covered larger areas of the tumor and the peri-tumor region at later time points. Moreover, NPs released from the HG were shown to undergo uptake by surrounding GBM cells. Findings suggest that intratumoral delivery with HG-NPs has immense potential for GBM treatment, as well as other strategies where site-specific, long-term retention of therapeutic agents is warranted.
Collapse
Affiliation(s)
- Giulia Brachi
- Politecnico di Torino
, DIMEAS
,
C.so Duca degli Abruzzi 24
, 10129 Torino
, Italy
.
; Tel: +390110906792
- Department of Nanomedicine
, Houston Methodist Research Institute
,
6670 Bertner Ave
, Houston
, TX 77030
, USA
| | - Javier Ruiz-Ramírez
- Mathematics in Medicine Program
, Houston Methodist Research Institute
,
6670 Bertner Ave
, Houston
, TX 77030
, USA
| | - Prashant Dogra
- Mathematics in Medicine Program
, Houston Methodist Research Institute
,
6670 Bertner Ave
, Houston
, TX 77030
, USA
| | - Zhihui Wang
- Mathematics in Medicine Program
, Houston Methodist Research Institute
,
6670 Bertner Ave
, Houston
, TX 77030
, USA
| | - Vittorio Cristini
- Mathematics in Medicine Program
, Houston Methodist Research Institute
,
6670 Bertner Ave
, Houston
, TX 77030
, USA
| | - Gianluca Ciardelli
- Politecnico di Torino
, DIMEAS
,
C.so Duca degli Abruzzi 24
, 10129 Torino
, Italy
.
; Tel: +390110906792
| | - Robert C. Rostomily
- Department of Neurosurgery
, Houston Methodist Research Institute
,
6670 Bertner Ave
, Houston
, TX 77030
, USA
| | - Mauro Ferrari
- Department of Nanomedicine
, Houston Methodist Research Institute
,
6670 Bertner Ave
, Houston
, TX 77030
, USA
| | - Andrei M. Mikheev
- Department of Neurosurgery
, Houston Methodist Research Institute
,
6670 Bertner Ave
, Houston
, TX 77030
, USA
| | - Elvin Blanco
- Department of Nanomedicine
, Houston Methodist Research Institute
,
6670 Bertner Ave
, Houston
, TX 77030
, USA
| | - Clara Mattu
- Politecnico di Torino
, DIMEAS
,
C.so Duca degli Abruzzi 24
, 10129 Torino
, Italy
.
; Tel: +390110906792
- Department of Nanomedicine
, Houston Methodist Research Institute
,
6670 Bertner Ave
, Houston
, TX 77030
, USA
| |
Collapse
|
323
|
Yekula A, Muralidharan K, Rosh Z, Youngkin AE, Kang KM, Balaj L, Carter BS. Liquid Biopsy Strategies to Distinguish Progression from Pseudoprogression and Radiation Necrosis in Glioblastomas. ADVANCED BIOSYSTEMS 2020; 4:e2000029. [PMID: 32484293 PMCID: PMC7708392 DOI: 10.1002/adbi.202000029] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 04/20/2020] [Indexed: 12/13/2022]
Abstract
Liquid biopsy for the detection and monitoring of central nervous system tumors is of significant clinical interest. At initial diagnosis, the majority of patients with central nervous system tumors undergo magnetic resonance imaging (MRI), followed by invasive brain biopsy to determine the molecular diagnosis of the WHO 2016 classification paradigm. Despite the importance of MRI for long-term treatment monitoring, in the majority of patients who receive chemoradiation therapy for glioblastoma, it can be challenging to distinguish between radiation treatment effects including pseudoprogression, radiation necrosis, and recurrent/progressive disease based on imaging alone. Tissue biopsy-based monitoring is high risk and not always feasible. However, distinguishing these entities is of critical importance for the management of patients and can significantly affect survival. Liquid biopsy strategies including circulating tumor cells, circulating free DNA, and extracellular vesicles have the potential to afford significant useful molecular information at both the stage of diagnosis and monitoring for these tumors. Here, current liquid biopsy-based approaches in the context of tumor monitoring to differentiate progressive disease from pseudoprogression and radiation necrosis are reviewed.
Collapse
Affiliation(s)
- Anudeep Yekula
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA
| | | | - Zachary Rosh
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA
| | - Anna E. Youngkin
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA
- Trinity College of Arts and Sciences, Duke University, Durham, NC, USA
| | - Keiko M. Kang
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA
- School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Leonora Balaj
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA
| | - Bob S. Carter
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
324
|
Pardridge WM. Brain Delivery of Nanomedicines: Trojan Horse Liposomes for Plasmid DNA Gene Therapy of the Brain. FRONTIERS IN MEDICAL TECHNOLOGY 2020; 2:602236. [PMID: 35047884 PMCID: PMC8757841 DOI: 10.3389/fmedt.2020.602236] [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: 09/02/2020] [Accepted: 10/06/2020] [Indexed: 12/14/2022] Open
Abstract
Non-viral gene therapy of the brain is enabled by the development of plasmid DNA brain delivery technology, which requires the engineering and manufacturing of nanomedicines that cross the blood-brain barrier (BBB). The development of such nanomedicines is a multi-faceted problem that requires progress at multiple levels. First, the type of nanocontainer, e.g., nanoparticle or liposome, which encapsulates the plasmid DNA, must be developed. Second, the type of molecular Trojan horse, e.g., peptide or receptor-specific monoclonal antibody (MAb), must be selected for incorporation on the surface of the nanomedicine, as this Trojan horse engages specific receptors expressed on the BBB, and the brain cell membrane, to trigger transport of the nanomedicine from blood into brain cells beyond the BBB. Third, the plasmid DNA must be engineered without bacterial elements, such as antibiotic resistance genes, to enable administration to humans; the plasmid DNA must also be engineered with tissue-specific gene promoters upstream of the therapeutic gene, to insure gene expression in the target organ with minimal off-target expression. Fourth, upstream manufacturing of the nanomedicine must be developed and scalable so as to meet market demand for the target disease, e.g., annual long-term treatment of 1,000 patients with an orphan disease, short term treatment of 10,000 patients with malignant glioma, or 100,000 patients with new onset Parkinson's disease. Fifth, downstream manufacturing problems, such as nanomedicine lyophilization, must be solved to ensure the nanomedicine has a commercially viable shelf-life for treatment of CNS disease in humans.
Collapse
Affiliation(s)
- William M Pardridge
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
325
|
Affiliation(s)
- Gaetano Finocchiaro
- Molecular Neuro-Oncology Unit, Carlo Besta Neurological Institute, Milan, Italy
| |
Collapse
|
326
|
Yoo JY, Yeh M, Kaur B, Lee TJ. Targeted delivery of small noncoding RNA for glioblastoma. Cancer Lett 2020; 500:274-280. [PMID: 33176185 DOI: 10.1016/j.canlet.2020.11.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 10/16/2020] [Accepted: 11/05/2020] [Indexed: 12/12/2022]
Abstract
Aberrant expression of certain genes and microRNAs (miRNAs) has been shown to drive cancer development and progression, thus the modification of aberrant gene and miRNA expression presents an opportunity for therapeutic targeting. Ectopic modulation of a single dysregulated miRNA has the potential to revert therapeutically unfavorable gene expression in cancer cells by targeting multiple genes simultaneously. Although the use of noncoding RNA-based cancer therapy is a promising approach, the lack of a feasible delivery platform for small noncoding RNAs has hindered the development of this therapeutic modality. Recently, however, there has been an evolution in RNA nanotechnology, in which small noncoding RNA is loaded onto nanoparticles derived from the pRNA-3WJ viral RNA motif of the bacteriophage phi29. Preclinical studies have shown the capacity of this technology to specifically target tumor cells by conjugating these nanoparticles with ligands specific for cancer cells and resulting in the endocytic delivery of siRNA and miRNA inhibitors directly into the cell. Here we provide a systematic review of the various strategies, which have been utilized for miRNA delivery with a specific focus on the preclinical evaluation of promising RNA nanoparticles for glioblastoma (GBM) targeted therapy.
Collapse
Affiliation(s)
- Ji Young Yoo
- Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Margaret Yeh
- Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Balveen Kaur
- Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Tae Jin Lee
- Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
| |
Collapse
|
327
|
Lal S, Snape TJ. A therapeutic update on PARP inhibitors: implications in the treatment of glioma. Drug Discov Today 2020; 26:532-541. [PMID: 33157194 DOI: 10.1016/j.drudis.2020.10.029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 10/07/2020] [Accepted: 10/29/2020] [Indexed: 01/01/2023]
Abstract
Central nervous system (CNS) cancers are among the most aggressive and devastating. Further, due to unavailability of neuro-oncologists and neurosurgeons, the specialized treatment options of CNS cancers are still not completely available in most parts of the world. Among various strategies of inducing death in cancer cells, inhibition of poly(ADP-ribose) polymerase (PARP) has emerged as a beneficial therapy when combined with other anticancer agents. In this review, we provide a detailed therapeutic update of PARP inhibitors that have shown clinical activity against glioma.
Collapse
Affiliation(s)
- Samridhi Lal
- Amity Institute of Pharmacy, Amity University, Gurugram, 122413, Haryana, India.
| | - Timothy J Snape
- Leicester School of Pharmacy, De Montfort University, Leicester, LE1 9BH, UK
| |
Collapse
|
328
|
Kunadis E, Lakiotaki E, Korkolopoulou P, Piperi C. Targeting post-translational histone modifying enzymes in glioblastoma. Pharmacol Ther 2020; 220:107721. [PMID: 33144118 DOI: 10.1016/j.pharmthera.2020.107721] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/08/2020] [Accepted: 10/27/2020] [Indexed: 12/30/2022]
Abstract
Glioblastoma (GBM) is the most common primary brain tumor in adults, and the most lethal form of glioma, characterized by variable histopathology, aggressiveness and poor clinical outcome and prognosis. GBMs constitute a challenge for oncologists because of their molecular heterogeneity, extensive invasion, and tendency to relapse. Glioma cells demonstrate a variety of deregulated genomic pathways and extensive interplay with epigenetic alterations. Epigenetic modifications have emerged as essential players in GBM research, with biomarker potential for tumor classification and prognosis and for drug targeting. Histone posttranslational modifications (PTMs) are crucial regulators of chromatin architecture and gene expression, playing a pivotal role in malignant transformation, tumor development and progression. Alteration in the expression of genes coding for lysine and arginine methyltransferases (G9a, SUV39H1 and SETDB1) and acetyltransferases and deacetylases (KAT6A, SIRT2, SIRT7, HDAC4, 6, 9) contribute to GBM pathogenesis. In addition, proteins of the sumoylation pathway are upregulated in GBM cell lines, including E1 (SAE1), E2 (Ubc9) components, and a SUMO-specific protease (SENP1). Preclinical and clinical studies are currently in progress targeting epigenetic enzymes in gliomas, including a new generation of histone deacetylase (HDAC), protein arginine methyltransferase (PRMT) and bromodomain (BRD) inhibitors. Herein, we provide an update on recent advances in glioma epigenetic research, focusing on the role of histone modifications and the use of epigenetic therapy as a valid treatment option for glioblastoma.
Collapse
Affiliation(s)
- Elena Kunadis
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, 11527 Athens, Greece
| | - Eleftheria Lakiotaki
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, 11527 Athens, Greece
| | - Penelope Korkolopoulou
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, 11527 Athens, Greece
| | - Christina Piperi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, 11527 Athens, Greece.
| |
Collapse
|
329
|
Abstract
![]()
Nanocarriers
(NCs) are promising tools to improve drug delivery
across the blood–brain barrier (BBB) for more effective treatment
of brain disorders, although there is a scarcity of clinical translation
of brain-directed NCs. In order to drive the development of brain-oriented
NCs toward clinical success, it is essential to understand the prerequisites
for nanodelivery to be successful in brain treatment. In this Perspective,
we present how pharmacokinetic/pharmacodynamic (PK/PD), formulation
and nanotoxicity factors impact the therapeutic success of brain-specific
nanodelivery. Properties including high loading efficiency, slow in vivo drug release, long systemic circulation, an increase
in unbound brain-to-plasma concentration/exposure ratio (Kp,uu,brain), high drug potency, and minimal nanotoxicity
are prerequisites that should preferably be combined to maximize the
therapeutic potential of a brain-targeted NC. The PK of brain-directed
NCs needs to be evaluated in a more therapeutically relevant manner,
focusing on the released, unbound drug. It is more crucial to increase
the Kp,uu,brain than to improve the ability
of the NC to cross the BBB in its intact form. Brain-targeted NCs,
which are mostly developed for treating brain tumors, including metastases,
should aim to enhance drug delivery not just to tumor regions with
disrupted BBB, but equally important to regions with intact BBB where
the drugs themselves have problems reaching. This article provides
critical insights into how a brain-targeted nanoformulation needs
to be designed and optimized to achieve therapeutic success in the
brain.
Collapse
Affiliation(s)
- Yang Hu
- Translational PKPD Research Group, Department of Pharmacy, Faculty of Pharmacy, Uppsala University, SE-751 23 Uppsala, Sweden
| | - Margareta Hammarlund-Udenaes
- Translational PKPD Research Group, Department of Pharmacy, Faculty of Pharmacy, Uppsala University, SE-751 23 Uppsala, Sweden
| |
Collapse
|
330
|
Amero P, Khatua S, Rodriguez-Aguayo C, Lopez-Berestein G. Aptamers: Novel Therapeutics and Potential Role in Neuro-Oncology. Cancers (Basel) 2020; 12:cancers12102889. [PMID: 33050158 PMCID: PMC7600320 DOI: 10.3390/cancers12102889] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/25/2020] [Accepted: 09/29/2020] [Indexed: 12/15/2022] Open
Abstract
A relatively new paradigm in cancer therapeutics is the use of cancer cell-specific aptamers, both as therapeutic agents and for targeted delivery of anticancer drugs. After the first therapeutic aptamer was described nearly 25 years ago, and the subsequent first aptamer drug approved, many efforts have been made to translate preclinical research into clinical oncology settings. Studies of aptamer-based technology have unveiled the vast potential of aptamers in therapeutic and diagnostic applications. Among pediatric solid cancers, brain tumors are the leading cause of death. Although a few aptamer-related translational studies have been performed in adult glioblastoma, the use of aptamers in pediatric neuro-oncology remains unexplored. This review will discuss the biology of aptamers, including mechanisms of targeting cell surface proteins, various modifications of aptamer structure to enhance therapeutic efficacy, the current state and challenges of aptamer use in neuro-oncology, and the potential therapeutic role of aptamers in pediatric brain tumors.
Collapse
Affiliation(s)
- Paola Amero
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA;
| | - Soumen Khatua
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Cristian Rodriguez-Aguayo
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA;
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
- Correspondence: (C.R.-A.); (G.L.-B.); Tel.: +1-713-563-6150 (C.R.-A.); +1-713-792-8140 (G.L.-B.)
| | - Gabriel Lopez-Berestein
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA;
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Correspondence: (C.R.-A.); (G.L.-B.); Tel.: +1-713-563-6150 (C.R.-A.); +1-713-792-8140 (G.L.-B.)
| |
Collapse
|
331
|
van Tellingen O, de Gooijer MC, Sarkaria JN, Elmquist WF. Comments on: "Synergistic activity of mTORC1/2 kinase and MEK inhibitors suppresses pediatric low-grade glioma tumorigenicity and vascularity". Neuro Oncol 2020; 22:1404-1405. [PMID: 32556220 DOI: 10.1093/neuonc/noaa112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Affiliation(s)
- Olaf van Tellingen
- Division of Pharmacology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Mark C de Gooijer
- Division of Pharmacology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | | | - William F Elmquist
- Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
332
|
Hawkins CC, Ali T, Ramanadham S, Hjelmeland AB. Sphingolipid Metabolism in Glioblastoma and Metastatic Brain Tumors: A Review of Sphingomyelinases and Sphingosine-1-Phosphate. Biomolecules 2020; 10:E1357. [PMID: 32977496 PMCID: PMC7598277 DOI: 10.3390/biom10101357] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/18/2020] [Accepted: 09/20/2020] [Indexed: 01/05/2023] Open
Abstract
Glioblastoma (GBM) is a primary malignant brain tumor with a dismal prognosis, partially due to our inability to completely remove and kill all GBM cells. Rapid tumor recurrence contributes to a median survival of only 15 months with the current standard of care which includes maximal surgical resection, radiation, and temozolomide (TMZ), a blood-brain barrier (BBB) penetrant chemotherapy. Radiation and TMZ cause sphingomyelinases (SMase) to hydrolyze sphingomyelins to generate ceramides, which induce apoptosis. However, cells can evade apoptosis by converting ceramides to sphingosine-1-phosphate (S1P). S1P has been implicated in a wide range of cancers including GBM. Upregulation of S1P has been linked to the proliferation and invasion of GBM and other cancers that display a propensity for brain metastasis. To mediate their biological effects, SMases and S1P modulate signaling via phospholipase C (PLC) and phospholipase D (PLD). In addition, both SMase and S1P may alter the integrity of the BBB leading to infiltration of tumor-promoting immune populations. SMase activity has been associated with tumor evasion of the immune system, while S1P creates a gradient for trafficking of innate and adaptive immune cells. This review will explore the role of sphingolipid metabolism and pharmacological interventions in GBM and metastatic brain tumors with a focus on SMase and S1P.
Collapse
Affiliation(s)
- Cyntanna C. Hawkins
- Department of Cell, Developmental, and Integrative Biology, University of Birmingham at Alabama, Birmingham, AL 35233, USA; (C.C.H.); (S.R.)
| | - Tomader Ali
- Research Department, Imperial College London Diabetes Centre, Abu Dhabi P.O. Box 48338, UAE;
| | - Sasanka Ramanadham
- Department of Cell, Developmental, and Integrative Biology, University of Birmingham at Alabama, Birmingham, AL 35233, USA; (C.C.H.); (S.R.)
- Comprehensive Diabetes Center, University of Birmingham at Alabama, Birmingham, AL 35294, USA
| | - Anita B. Hjelmeland
- Department of Cell, Developmental, and Integrative Biology, University of Birmingham at Alabama, Birmingham, AL 35233, USA; (C.C.H.); (S.R.)
| |
Collapse
|
333
|
Karmur BS, Philteos J, Abbasian A, Zacharia BE, Lipsman N, Levin V, Grossman S, Mansouri A. Blood-Brain Barrier Disruption in Neuro-Oncology: Strategies, Failures, and Challenges to Overcome. Front Oncol 2020; 10:563840. [PMID: 33072591 PMCID: PMC7531249 DOI: 10.3389/fonc.2020.563840] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 08/13/2020] [Indexed: 01/05/2023] Open
Abstract
The blood-brain barrier (BBB) presents a formidable challenge in the development of effective therapeutics in neuro-oncology. This has fueled several decades of efforts to develop strategies for disrupting the BBB, but progress has not been satisfactory. As such, numerous drug- and device-based methods are currently being investigated in humans. Through a focused assessment of completed, active, and pending clinical trials, our first aim in this review is to outline the scientific foundation, successes, and limitations of the BBBD strategies developed to date. Among 35 registered trials relevant to BBBD in neuro-oncology in the ClinicalTrials.gov database, mannitol was the most common drug-based method, followed by RMP-7 and regadenoson. MR-guided focused ultrasound was the most common device-based method, followed by MR-guided laser ablation, ultrasound, and transcranial magnetic stimulation. While most early-phase studies focusing on safety and tolerability have met stated objectives, advanced-phase studies focusing on survival differences and objective tumor response have been limited by heterogeneous populations and tumors, along with a lack of control arms. Based on shared challenges among all methods, our second objective is to discuss strategies for confirmation of BBBD, choice of systemic agent and drug design, alignment of BBBD method with real-world clinical workflow, and consideration of inadvertent toxicity associated with disrupting an evolutionarily-refined barrier. Finally, we conclude with a strategic proposal to approach future studies assessing BBBD.
Collapse
Affiliation(s)
- Brij S Karmur
- Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | | | - Aram Abbasian
- Faculty of Medicine, University of Toronto, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Brad E Zacharia
- Penn State Health Neurosurgery, College of Medicine, Penn State University, Hershey, PA, United States
| | - Nir Lipsman
- Division of Neurosurgery, University of Toronto, Toronto, ON, Canada
| | - Victor Levin
- Department of Neurosurgery, Medical School, University of California, San Francisco, San Francisco, CA, United States
| | - Stuart Grossman
- Department of Oncology, Johns Hopkins Medicine, Baltimore, MD, United States
| | - Alireza Mansouri
- Penn State Health Neurosurgery, College of Medicine, Penn State University, Hershey, PA, United States
| |
Collapse
|
334
|
Li J, Jiang J, Wu J, Bao X, Sanai N. Physiologically Based Pharmacokinetic Modeling of Central Nervous System Pharmacokinetics of CDK4/6 Inhibitors to Guide Selection of Drug and Dosing Regimen for Brain Cancer Treatment. Clin Pharmacol Ther 2020; 109:494-506. [PMID: 32799335 PMCID: PMC7854954 DOI: 10.1002/cpt.2021] [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: 05/04/2020] [Accepted: 08/06/2020] [Indexed: 12/21/2022]
Abstract
A better understanding of the human central nervous system (CNS) pharmacokinetics is critical to the selection of the right drug and refinement of dosing regimen for more effective treatment of primary and metastatic brain cancer. Using the physiologically‐based pharmacokinetic (PBPK) modeling approach, we systematically compared the CNS pharmacokinetics of three cyclin D‐cyclin dependent kinase 4 and 6 (CDK4/6) inhibitors (ribociclib, palbociclib, and abemaciclib) in patients with cancer. A PBPK model platform was developed and verified for predicting plasma and CNS pharmacokinetics. Target engagement ratio (TER), defined as the ratio of the average steady‐state unbound drug brain concentration to the in vitro half‐maximal inhibitory concentration (IC50) for CDK4/6 inhibition, was used as a crude predictor of efficacy. As compared with ribociclib and palbociclib, abemaciclib penetrated into the human brain to a larger extent, but at a slower rate, and was retained in the brain longer. Following the standard dosing regimens, the predicted CDK4/6 TERs were 26/5.2 for abemaciclib, 2.4/0.62 for ribociclib, and 0.36/0.27 for palbociclib. Simulations suggested that abemaciclib achieved comparable TERs following twice daily or daily dosing; ribociclib may sufficiently inhibit both CDK4 and CDK6 at the maximum tolerated dose; whereas, palbociclib achieved TERs < 0.5 even at a dose 50% higher than the standard dose. In conclusion, the PBPK modeling, supported by available preclinical and clinical evidence, suggests that abemaciclib is the best CDK4/6 inhibitor for brain cancer treatment, whereas palbociclib is not recommended. The model refined dosing regimen is 300 mg daily on a 4‐weeks‐on schedule for abemaciclib, and 900 mg daily on a 3‐weeks‐on/1‐week‐off schedule for ribociclib.
Collapse
Affiliation(s)
- Jing Li
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Jun Jiang
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Jianmei Wu
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Xun Bao
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Nader Sanai
- Barrow Neurological Institute, St. Joseph's Hospital & Medical Center, Phoenix, Arizona, USA
| |
Collapse
|
335
|
Cui X, Ma C, Vasudevaraja V, Serrano J, Tong J, Peng Y, Delorenzo M, Shen G, Frenster J, Morales RTT, Qian W, Tsirigos A, Chi AS, Jain R, Kurz SC, Sulman EP, Placantonakis DG, Snuderl M, Chen W. Dissecting the immunosuppressive tumor microenvironments in Glioblastoma-on-a-Chip for optimized PD-1 immunotherapy. eLife 2020; 9:52253. [PMID: 32909947 PMCID: PMC7556869 DOI: 10.7554/elife.52253] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 08/27/2020] [Indexed: 12/13/2022] Open
Abstract
Programmed cell death protein-1 (PD-1) checkpoint immunotherapy efficacy remains unpredictable in glioblastoma (GBM) patients due to the genetic heterogeneity and immunosuppressive tumor microenvironments. Here, we report a microfluidics-based, patient-specific 'GBM-on-a-Chip' microphysiological system to dissect the heterogeneity of immunosuppressive tumor microenvironments and optimize anti-PD-1 immunotherapy for different GBM subtypes. Our clinical and experimental analyses demonstrated that molecularly distinct GBM subtypes have distinct epigenetic and immune signatures that may lead to different immunosuppressive mechanisms. The real-time analysis in GBM-on-a-Chip showed that mesenchymal GBM niche attracted low number of allogeneic CD154+CD8+ T-cells but abundant CD163+ tumor-associated macrophages (TAMs), and expressed elevated PD-1/PD-L1 immune checkpoints and TGF-β1, IL-10, and CSF-1 cytokines compared to proneural GBM. To enhance PD-1 inhibitor nivolumab efficacy, we co-administered a CSF-1R inhibitor BLZ945 to ablate CD163+ M2-TAMs and strengthened CD154+CD8+ T-cell functionality and GBM apoptosis on-chip. Our ex vivo patient-specific GBM-on-a-Chip provides an avenue for a personalized screening of immunotherapies for GBM patients.
Collapse
Affiliation(s)
- Xin Cui
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, United States.,Department of Biomedical Engineering, New York University, Brooklyn, United States.,Department of Biomedical Engineering, Jinan University, Guangzhou, China
| | - Chao Ma
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, United States.,Department of Biomedical Engineering, New York University, Brooklyn, United States
| | | | - Jonathan Serrano
- Department of Pathology, NYU Langone Health, New York, United States
| | - Jie Tong
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, United States.,Department of Biomedical Engineering, New York University, Brooklyn, United States
| | - Yansong Peng
- Department of Biomedical Engineering, New York University, Brooklyn, United States
| | - Michael Delorenzo
- Department of Pathology, NYU Langone Health, New York, United States
| | - Guomiao Shen
- Department of Pathology, NYU Langone Health, New York, United States
| | - Joshua Frenster
- Stem Cell Biology Program, NYU School of Medicine, New York, United States
| | | | - Weiyi Qian
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, United States
| | | | - Andrew S Chi
- Perlmutter Cancer Center, NYU Langone Health, New York, United States.,Department of Neurology, NYU Langone Health, New York, United States
| | - Rajan Jain
- Department of Neuroradiology, NYU Langone Health, New York, United States.,Department of Neurosurgery, NYU Langone Health, New York, United States
| | - Sylvia C Kurz
- Perlmutter Cancer Center, NYU Langone Health, New York, United States.,Department of Neurosurgery, NYU Langone Health, New York, United States
| | - Erik P Sulman
- Perlmutter Cancer Center, NYU Langone Health, New York, United States.,Department of Radiation Oncology, NYU Langone Health, New York, United States
| | - Dimitris G Placantonakis
- Perlmutter Cancer Center, NYU Langone Health, New York, United States.,Department of Neurosurgery, NYU Langone Health, New York, United States
| | - Matija Snuderl
- Department of Pathology, NYU Langone Health, New York, United States.,Perlmutter Cancer Center, NYU Langone Health, New York, United States
| | - Weiqiang Chen
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, United States.,Department of Biomedical Engineering, New York University, Brooklyn, United States.,Perlmutter Cancer Center, NYU Langone Health, New York, United States
| |
Collapse
|
336
|
Pierce CF, Kwasnicki A, Lakka SS, Engelhard HH. Cerebral Microdialysis as a Tool for Assessing the Delivery of Chemotherapy in Brain Tumor Patients. World Neurosurg 2020; 145:187-196. [PMID: 32890850 DOI: 10.1016/j.wneu.2020.08.161] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 08/20/2020] [Accepted: 08/22/2020] [Indexed: 12/27/2022]
Abstract
The development of curative treatment for glioblastoma has been extremely challenging. Chemotherapeutic agents that have seemed promising have failed in clinical trials. Drugs that can successfully target cancer cells within the brain must first traverse the brain interstitial fluid. Cerebral microdialysis (CMD) is an invasive technique in which interstitial fluid can be directly sampled. CMD has primarily been used clinically in the setting of head trauma and subarachnoid hemorrhage. Our goal was to review the techniques, principles, and new data pertaining to CMD to highlight its use in neuro-oncology. We conducted a literature search using the PubMed database and selected studies in which the investigators had used CMD in either animal brain tumor models or clinical trials. The references were reviewed for additional information. Studies of CMD have shown its importance as a neurosurgical technique. CMD allows for the collection of pharmacokinetic data on drug penetrance across the blood-brain barrier and metabolic data to characterize the response to chemotherapy. Although no complications have been reported, the current CMD technique (as with any procedure) has risks and limitations, which we have described in the present report. Animal CMD experiments have been used to exclude central nervous system drug candidates from progressing to clinical trials. At present, patients undergoing CMD have been monitored in the intensive care unit, owing to the requisite tethering to the apparatus. This can be expected to change soon because of advances in microminiaturization. CMD is an extremely valuable, yet underused, technique. Future CMD applications will have central importance in assessing drug delivery to tumor cells in vivo, allowing a pathway to successful therapy for malignant brain tumors.
Collapse
Affiliation(s)
- Charles F Pierce
- Department of Neurosurgery, The University of Illinois at Chicago, Chicago, Illinois, USA
| | - Amanda Kwasnicki
- Department of Neurosurgery, The University of Illinois at Chicago, Chicago, Illinois, USA
| | - Sajani S Lakka
- Department of Medicine, The University of Illinois at Chicago, Chicago, Illinois, USA
| | - Herbert H Engelhard
- Department of Neurosurgery, The University of Illinois at Chicago, Chicago, Illinois, USA; Department of Bioengineering, The University of Illinois at Chicago, Chicago, Illinois, USA.
| |
Collapse
|
337
|
Wen PY, Weller M, Lee EQ, Alexander BM, Barnholtz-Sloan JS, Barthel FP, Batchelor TT, Bindra RS, Chang SM, Chiocca EA, Cloughesy TF, DeGroot JF, Galanis E, Gilbert MR, Hegi ME, Horbinski C, Huang RY, Lassman AB, Le Rhun E, Lim M, Mehta MP, Mellinghoff IK, Minniti G, Nathanson D, Platten M, Preusser M, Roth P, Sanson M, Schiff D, Short SC, Taphoorn MJB, Tonn JC, Tsang J, Verhaak RGW, von Deimling A, Wick W, Zadeh G, Reardon DA, Aldape KD, van den Bent MJ. Glioblastoma in adults: a Society for Neuro-Oncology (SNO) and European Society of Neuro-Oncology (EANO) consensus review on current management and future directions. Neuro Oncol 2020; 22:1073-1113. [PMID: 32328653 PMCID: PMC7594557 DOI: 10.1093/neuonc/noaa106] [Citation(s) in RCA: 598] [Impact Index Per Article: 149.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Glioblastomas are the most common form of malignant primary brain tumor and an important cause of morbidity and mortality. In recent years there have been important advances in understanding the molecular pathogenesis and biology of these tumors, but this has not translated into significantly improved outcomes for patients. In this consensus review from the Society for Neuro-Oncology (SNO) and the European Association of Neuro-Oncology (EANO), the current management of isocitrate dehydrogenase wildtype (IDHwt) glioblastomas will be discussed. In addition, novel therapies such as targeted molecular therapies, agents targeting DNA damage response and metabolism, immunotherapies, and viral therapies will be reviewed, as well as the current challenges and future directions for research.
Collapse
Affiliation(s)
- Patrick Y Wen
- Dana-Farber Cancer Institute, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Michael Weller
- Department of Neurology and Brain Tumor Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Eudocia Quant Lee
- Dana-Farber Cancer Institute, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Brian M Alexander
- Dana-Farber Cancer Institute, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Jill S Barnholtz-Sloan
- Case Western Reserve University School of Medicine and University Hospitals of Cleveland, Cleveland, Ohio, USA
| | - Floris P Barthel
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, USA
| | - Tracy T Batchelor
- Department of Neurology, Brigham and Women’s Hospital, Dana-Farber Cancer Institute and Harvard Medical School
| | - Ranjit S Bindra
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Susan M Chang
- University of California San Francisco, San Francisco, California, USA
| | - E Antonio Chiocca
- Department of Neurosurgery, Brigham and Women’s Hospital, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, USA
| | - Timothy F Cloughesy
- David Geffen School of Medicine, Department of Neurology, University of California Los Angeles, Los Angeles, California, USA
| | - John F DeGroot
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Mark R Gilbert
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Monika E Hegi
- Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Craig Horbinski
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Raymond Y Huang
- Division of Neuroradiology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Andrew B Lassman
- Department of Neurology and Herbert Irving Comprehensive Cancer Center, NewYork-Presbyterian Hospital/Columbia University Irving Medical Center, New York, New York, USA
| | - Emilie Le Rhun
- University of Lille, Inserm, Neuro-oncology, General and Stereotaxic Neurosurgery service, University Hospital of Lille, Lille, France; Breast Cancer Department, Oscar Lambret Center, Lille, France and Department of Neurology & Brain Tumor Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Michael Lim
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Ingo K Mellinghoff
- Department of Neurology and Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Giuseppe Minniti
- Radiation Oncology Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - David Nathanson
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California, USA
| | - Michael Platten
- Department of Neurology, Medical Faculty Mannheim, MCTN, Heidelberg University, Heidelberg, Germany
| | - Matthias Preusser
- Division of Oncology, Department of Medicine, Medical University of Vienna, Vienna, Austria
| | - Patrick Roth
- Department of Neurology and Brain Tumor Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Marc Sanson
- Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière – Charles Foix, Service de Neurologie 2-Mazarin, Paris, France
| | - David Schiff
- University of Virginia School of Medicine, Division of Neuro-Oncology, Department of Neurology, University of Virginia, Charlottesville, Virginia, USA
| | - Susan C Short
- Leeds Institute of Medical Research at St James’s, University of Leeds, Leeds, UK
| | - Martin J B Taphoorn
- Department of Neurology, Medical Center Haaglanden, The Hague and Department of Neurology, Leiden University Medical Center, the Netherlands
| | | | - Jonathan Tsang
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California, USA
| | - Roel G W Verhaak
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, USA
| | - Andreas von Deimling
- Neuropathology and Clinical Cooperation Unit Neuropathology, University Heidelberg and German Cancer Center, Heidelberg, Germany
| | - Wolfgang Wick
- Department of Neurology and Neuro-oncology Program, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Gelareh Zadeh
- MacFeeters Hamilton Centre for Neuro-Oncology Research, Princess Margaret Cancer Centre, Toronto, Canada
| | - David A Reardon
- Dana-Farber Cancer Institute, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Kenneth D Aldape
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | | |
Collapse
|
338
|
Intratumoral platelet aggregate formation in a murine preclinical glioma model depends on podoplanin expression on tumor cells. Blood Adv 2020; 3:1092-1102. [PMID: 30948364 DOI: 10.1182/bloodadvances.2018015966] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 02/17/2019] [Indexed: 11/20/2022] Open
Abstract
Binding of the sialomucin-like transmembrane glycoprotein podoplanin (PDPN) to the platelet receptor C-type lectin-like receptor 2 induces platelet activation and aggregation. In human high-grade gliomas, PDPN is highly expressed both in tumor cells and in tumor-associated astrocytes. In glioma patients, high expression of PDPN is associated with worse prognosis and has been shown to correlate with intratumoral platelet aggregation and an increased risk of venous thromboembolism (VTE). To functionally assess the role of PDPN in platelet aggregation in vivo, we established a syngeneic orthotopic murine glioma model in C57/Bl6 mice, based on transplantation of p53- and Pten-deficient neural stem cells. This model is characterized by the presence of intratumoral platelet aggregates and by the upregulation of PDPN both in glioma cells and in astrocytes, reflecting the characteristics of human gliomas. Deletion of PDPN either in tumor cells or in astrocytes resulted in glioma formation with similar penetrance and grade compared with control mice. Importantly, only the lack of PDPN in tumor cells, but not in astrocytes, caused a significant reduction in intratumoral platelet aggregates, whereas in vitro, both cell types have similar platelet aggregation-inducing capacities. Our results demonstrate a causative link between PDPN and platelet aggregation in gliomas and pinpoint the tumor cells as the major players in PDPN-induced platelet aggregation. Our data indicate that blocking PDPN specifically on tumor cells could represent a novel strategy to prevent platelet aggregation and thereby reduce the risk of VTE in glioma patients.
Collapse
|
339
|
Patel NV, Khatri D, D'Amico R, Abrams M, Reichman N, Filippi CG, Anderson T, Ratzon F, Wong T, Fralin S, Li M, Faltings L, Langer DJ, Boockvar JA. Vascularized Temporoparietal Fascial Flap: A Novel Surgical Technique to Bypass the Blood-Brain Barrier in Glioblastoma. World Neurosurg 2020; 143:38-45. [PMID: 32712410 DOI: 10.1016/j.wneu.2020.07.132] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/17/2020] [Accepted: 07/18/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND The major difficulty in treating glioblastoma stems from the intrinsic privileged nature of the brain. This complicates therapy, as many traditionally potent chemotherapeutics cannot access their target sites in the brain. Several techniques have been investigated to overcome this barrier and facilitate drug delivery. However, these techniques have inherent shortcomings related to the delivery system, the drug itself, or its bioactivity. Periosteal flaps and temporoparietal fascial flaps (TPFFs) are widely used options because they have predictable vasculature and a wide rotational arc. These flaps are not restricted by the blood-brain barrier, as they derive their vascular supply from branches of the external carotid artery, which can be readily identified with Doppler ultrasound. We hypothesized that transposition of a vascularized TPFF to the walls of a resected tumor surgical cavity may bring autologous tissue not restricted by the blood-brain barrier in close vicinity of the resected tumor bed microenvironment. This offers a nonselective, long-lasting gateway to target the residual tumor cells nesting in the brain adjacent to the tumor. CASE DESCRIPTION A 47-year-old, right-handed woman with newly diagnosed multifocal glioblastoma underwent excision of the tumor and TPFF placement. This illustrative case report represents the first case of the use of this novel surgical technique with radiologic follow-up. CONCLUSIONS The blood-brain barrier is identified as a major barrier for effective drug delivery in glioblastoma. This study demonstrates the feasibility of the TPFF technique to bypass this barrier and help facilitate the goal of improving drug delivery.
Collapse
Affiliation(s)
- Nitesh V Patel
- Department of Neurosurgery, Lenox Hill Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New York, New York, USA
| | - Deepak Khatri
- Department of Neurosurgery, Lenox Hill Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New York, New York, USA.
| | - Randy D'Amico
- Department of Neurosurgery, Lenox Hill Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New York, New York, USA
| | - Madeline Abrams
- Department of Neurosurgery, Lenox Hill Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New York, New York, USA
| | - Noah Reichman
- Department of Neurosurgery, Lenox Hill Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New York, New York, USA
| | - Christopher G Filippi
- Department of Radiology, Lenox Hill Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New York, New York, USA
| | - Todd Anderson
- Department of Pathology, Lenox Hill Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New York, New York, USA
| | - Fanni Ratzon
- Department of Pathology, Lenox Hill Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New York, New York, USA
| | - Tamika Wong
- Department of Neurosurgery, Lenox Hill Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New York, New York, USA
| | - Sherese Fralin
- Department of Neurosurgery, Lenox Hill Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New York, New York, USA
| | - Mona Li
- Department of Neurosurgery, Lenox Hill Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New York, New York, USA
| | - Lukas Faltings
- Department of Neurosurgery, Lenox Hill Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New York, New York, USA
| | - David J Langer
- Department of Neurosurgery, Lenox Hill Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New York, New York, USA
| | - John A Boockvar
- Department of Neurosurgery, Lenox Hill Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New York, New York, USA
| |
Collapse
|
340
|
Qian J, Herman MG, Brinkmann DH, Laack NN, Kemp BJ, Hunt CH, Lowe V, Pafundi DH. Prediction of MGMT Status for Glioblastoma Patients Using Radiomics Feature Extraction From 18F-DOPA-PET Imaging. Int J Radiat Oncol Biol Phys 2020; 108:1339-1346. [PMID: 32634544 DOI: 10.1016/j.ijrobp.2020.06.073] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 06/15/2020] [Accepted: 06/28/2020] [Indexed: 12/13/2022]
Abstract
PURPOSE Methylation of the O6-methylguanine methyltransferase (MGMT) gene promoter is associated with improved treatment response and survival in patients with glioblastoma (GB), but the necessary pathologic specimen can be nondiagnostic. In this study, we assessed whether radiomics features from pretreatment 18F-DOPA positron emission tomography (PET) imaging could be used to predict pathologic MGMT status. METHODS AND MATERIALS This study included 86 patients with newly diagnosed GB, split into 3 groups (training, validating, and predicting). We performed a radiomics analysis on 18F-DOPA PET images by extracting features from 2 tumor-based contours: a "Gold" contour of all abnormal uptake per expert nuclear medicine physician and a high-grade glioma (HGG) contour based on a tumor-to-normal hemispheric ratio >2.0, representing the most aggressive components. Feature selection was performed by comparing the weighted feature importance and filtering with bivariate analysis. Optimization of model parameters was explored using grid search with selected features. The stability of the model with increasing input features was also investigated for model robustness. The model predictions were then applied by comparing the overall survival probability of the patients with GB and unknown MGMT status versus those with known MGMT status. RESULTS A radiomics signature was constructed to predict MGMT methylation status. Using features extracted from HGG contour alone with a random forest model, we achieved 80% ± 10% accuracy for 95% confidence level in predicting MGMT status. The prediction accuracy was not improved with the addition of the Gold contour or with more input features. The model was applied to the patients with unknown MGMT methylation status. The prediction results are consistent with what is expected using overall survival as a surrogate. CONCLUSIONS This study suggests that 3 features from radiomics modeling of 18F-DOPA PET imaging can predict MGMT methylation status with reasonable accuracy. These results could provide valuable therapeutic guidance for patients in whom MGMT testing is inconclusive or nondiagnostic.
Collapse
Affiliation(s)
- Jing Qian
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | - Michael G Herman
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | | | - Nadia N Laack
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | - Bradley J Kemp
- Department of Diagnostic Radiology, Mayo Clinic, Rochester, Minnesota
| | | | - Val Lowe
- Department of Diagnostic Radiology, Mayo Clinic, Rochester, Minnesota
| | - Deanna H Pafundi
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, Florida.
| |
Collapse
|
341
|
Ibarra LE, Beaugé L, Arias-Ramos N, Rivarola VA, Chesta CA, López-Larrubia P, Palacios RE. Trojan horse monocyte-mediated delivery of conjugated polymer nanoparticles for improved photodynamic therapy of glioblastoma. Nanomedicine (Lond) 2020; 15:1687-1707. [DOI: 10.2217/nnm-2020-0106] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Aim: To assess monocyte-based delivery of conjugated polymer nanoparticles (CPNs) for improved photodynamic therapy (PDT) in glioblastoma (GBM). Materials & methods: Human monocyte cells (THP-1) and murine monocytes isolated from bone marrow (mBMDMs) were employed as stealth CPN carriers to penetrate into GBM spheroids and an orthotopic model of the tumor. The success of PDT, using this cell-mediated targeting strategy, was determined by its effect on the spheroids. Results: CPNs did not affect monocyte viability in the absence of light and did not show nonspecific release after cell loading. Activated monocytes incorporated CPNs in a higher proportion than monocytes in their naive state, without a loss of cellular functionality. In vitro PDT efficacy using cell-mediated delivery was superior to that using non vehiculized CPNs. Conclusion: CPN-loaded monocytes could efficiently deliver CPNs into GBM spheroids and the orthotopic model. Improved PDT in spheroids was confirmed using this delivery strategy.
Collapse
Affiliation(s)
- Luis E Ibarra
- Instituto de Biotecnología Ambiental y Salud (INBIAS), Universidad Nacional de Río Cuarto (UNRC) y Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Río Cuarto, 5800, Córdoba, Argentina
- Departamento de Biología Molecular, Facultad de Ciencias Exactas Fisicoquímicas y Naturales, UNRC, Río Cuarto, 5800, Córdoba, Argentina
| | - Lucía Beaugé
- Departamento de Biología Molecular, Facultad de Ciencias Exactas Fisicoquímicas y Naturales, UNRC, Río Cuarto, 5800, Córdoba, Argentina
| | - Nuria Arias-Ramos
- Instituto de Investigaciones Biomédicas “Alberto Sols”, CSIC/UAM, Madrid, 28029, España
| | - Viviana A Rivarola
- Instituto de Biotecnología Ambiental y Salud (INBIAS), Universidad Nacional de Río Cuarto (UNRC) y Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Río Cuarto, 5800, Córdoba, Argentina
- Departamento de Biología Molecular, Facultad de Ciencias Exactas Fisicoquímicas y Naturales, UNRC, Río Cuarto, 5800, Córdoba, Argentina
| | - Carlos A Chesta
- Instituto de Investigaciones en Tecnologías Energéticas y Materiales Avanzados (IITEMA), Universidad Nacional de Río Cuarto (UNRC) y CONICET, Río Cuarto, 5800, Córdoba, Argentina
- Departamento de Química, Facultad de Ciencias Exactas Fisicoquímicas y Naturales, UNRC, Río Cuarto, 5800, Córdoba, Argentina
| | - Pilar López-Larrubia
- Instituto de Investigaciones Biomédicas “Alberto Sols”, CSIC/UAM, Madrid, 28029, España
| | - Rodrigo E Palacios
- Instituto de Investigaciones en Tecnologías Energéticas y Materiales Avanzados (IITEMA), Universidad Nacional de Río Cuarto (UNRC) y CONICET, Río Cuarto, 5800, Córdoba, Argentina
- Departamento de Química, Facultad de Ciencias Exactas Fisicoquímicas y Naturales, UNRC, Río Cuarto, 5800, Córdoba, Argentina
| |
Collapse
|
342
|
Hanif S, Muhammad P, Chesworth R, Rehman FU, Qian RJ, Zheng M, Shi BY. Nanomedicine-based immunotherapy for central nervous system disorders. Acta Pharmacol Sin 2020; 41:936-953. [PMID: 32467570 PMCID: PMC7468531 DOI: 10.1038/s41401-020-0429-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 04/22/2020] [Indexed: 12/15/2022] Open
Abstract
Central nervous system (CNS) disorders represent a broad spectrum of brain ailments with short- and long-term disabilities, and nanomedicine-based approaches provide a new therapeutic approach to treating CNS disorders. A variety of potential drugs have been discovered to treat several neuronal disorders; however, their therapeutic success can be limited by the presence of the blood-brain barrier (BBB). Furthermore, unique immune functions within the CNS provide novel target mechanisms for the amelioration of CNS diseases. Recently, various therapeutic approaches have been applied to fight brain-related disorders, with moderate outcomes. Among the various therapeutic strategies, nanomedicine-based immunotherapeutic systems represent a new era that can deliver useful cargo with promising pharmacokinetics. These approaches exploit the molecular and cellular targeting of CNS disorders for enhanced safety, efficacy, and specificity. In this review, we focus on the efficacy of nanomedicines that utilize immunotherapy to combat CNS disorders. Furthermore, we detailed summarize nanomedicine-based pathways for CNS ailments that aim to deliver drugs across the BBB by mimicking innate immune actions. Overview of how nanomedicines can utilize multiple immunotherapy pathways to combat CNS disorders. ![]()
Collapse
|
343
|
Blomquist MR, Ensign SF, D'Angelo F, Phillips JJ, Ceccarelli M, Peng S, Halperin RF, Caruso FP, Garofano L, Byron SA, Liang WS, Craig DW, Carpten JD, Prados MD, Trent JM, Berens ME, Iavarone A, Dhruv H, Tran NL. Temporospatial genomic profiling in glioblastoma identifies commonly altered core pathways underlying tumor progression. Neurooncol Adv 2020; 2:vdaa078. [PMID: 32743548 PMCID: PMC7388612 DOI: 10.1093/noajnl/vdaa078] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Background Tumor heterogeneity underlies resistance and disease progression in glioblastoma (GBM), and tumors most commonly recur adjacent to the surgical resection margins in contrast non-enhancing (NE) regions. To date, no targeted therapies have meaningfully altered overall patient survival in the up-front setting. The aim of this study was to characterize intratumoral heterogeneity in recurrent GBM using bulk samples from primary resection and recurrent samples taken from contrast-enhancing (EN) and contrast NE regions. Methods Whole exome and RNA sequencing were performed on matched bulk primary and multiple recurrent EN and NE tumor samples from 16 GBM patients who received standard of care treatment alone or in combination with investigational clinical trial regimens. Results Private mutations emerge across multi-region sampling in recurrent tumors. Genomic clonal analysis revealed increased enrichment in gene alterations regulating the G2M checkpoint, Kras signaling, Wnt signaling, and DNA repair in recurrent disease. Subsequent functional studies identified augmented PI3K/AKT transcriptional and protein activity throughout progression, validated by phospho-protein levels. Moreover, a mesenchymal transcriptional signature was observed in recurrent EN regions, which differed from the proneural signature in recurrent NE regions. Conclusions Subclonal populations observed within bulk resected primary GBMs transcriptionally evolve across tumor recurrence (EN and NE regions) and exhibit aberrant gene expression of common signaling pathways that persist despite standard or targeted therapy. Our findings provide evidence that there are both adaptive and clonally mediated dependencies of GBM on key pathways, such as the PI3K/AKT axis, for survival across recurrences.
Collapse
Affiliation(s)
- Mylan R Blomquist
- Department of Cancer Biology, Mayo Clinic Arizona, Scottsdale, Arizona, USA.,Department of Neurosurgery, Mayo Clinic Arizona, Scottsdale, Arizona, USA
| | | | - Fulvio D'Angelo
- Institute for Cancer Genetics, Columbia University Medical Center, New York, New York, USA
| | - Joanna J Phillips
- Department of Pathology, University of California, San Francisco, San Francisco, California, USA
| | | | - Sen Peng
- Cancer and Cell Biology Division, Translational Genomics Research Institute, Phoenix, Arizona, USA
| | - Rebecca F Halperin
- Integrated Cancer Genomics Division, The Translational Genomics Research Institute, Phoenix, Arizona, USA
| | - Francesca P Caruso
- Department of Science and Technology, Università degli Studi del Sannio, Benevento, Italy
| | - Luciano Garofano
- Institute for Cancer Genetics, Columbia University Medical Center, New York, New York, USA.,Department of Science and Technology, Università degli Studi del Sannio, Benevento, Italy
| | - Sara A Byron
- Integrated Cancer Genomics Division, The Translational Genomics Research Institute, Phoenix, Arizona, USA
| | - Winnie S Liang
- Integrated Cancer Genomics Division, The Translational Genomics Research Institute, Phoenix, Arizona, USA
| | - David W Craig
- Department of Translational Genomics, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - John D Carpten
- Department of Translational Genomics, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Michael D Prados
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Jeffrey M Trent
- Integrated Cancer Genomics Division, The Translational Genomics Research Institute, Phoenix, Arizona, USA
| | - Michael E Berens
- Cancer and Cell Biology Division, Translational Genomics Research Institute, Phoenix, Arizona, USA
| | - Antonio Iavarone
- Institute for Cancer Genetics, Columbia University Medical Center, New York, New York, USA
| | - Harshil Dhruv
- Cancer and Cell Biology Division, Translational Genomics Research Institute, Phoenix, Arizona, USA
| | - Nhan L Tran
- Department of Cancer Biology, Mayo Clinic Arizona, Scottsdale, Arizona, USA.,Department of Neurosurgery, Mayo Clinic Arizona, Scottsdale, Arizona, USA
| |
Collapse
|
344
|
Vatter T, Klumpp L, Ganser K, Stransky N, Zips D, Eckert F, Huber SM. Against Repurposing Methadone for Glioblastoma Therapy. Biomolecules 2020; 10:biom10060917. [PMID: 32560384 PMCID: PMC7356722 DOI: 10.3390/biom10060917] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/08/2020] [Accepted: 06/11/2020] [Indexed: 02/06/2023] Open
Abstract
Methadone, which is used as maintenance medication for outpatient treatment of opioid dependence or as an analgesic drug, has been suggested by preclinical in vitro and mouse studies to induce cell death and sensitivity to chemo- or radiotherapy in leukemia, glioblastoma, and carcinoma cells. These data together with episodical public reports on long-term surviving cancer patients who use methadone led to a hype of methadone as an anti-cancer drug in social and public media. However, clinical evidence for a tumoricidal effect of methadone is missing and prospective clinical trials, except in colorectal cancer, are not envisaged because of the limited preclinical data available. The present article reviews the pharmacokinetics, potential molecular targets, as well as the evidence for a tumoricidal effect of methadone in view of the therapeutically achievable doses in the brain. Moreover, it provides original in vitro data showing that methadone at clinically relevant concentrations fails to impair clonogenicity or radioresistance of glioblastoma cells.
Collapse
Affiliation(s)
- Tatjana Vatter
- Department of Radiation Oncology, University of Tübingen, 72076 Tübingen, Germany; (T.V.); (L.K.); (K.G.); (N.S.); (D.Z.); (F.E.)
| | - Lukas Klumpp
- Department of Radiation Oncology, University of Tübingen, 72076 Tübingen, Germany; (T.V.); (L.K.); (K.G.); (N.S.); (D.Z.); (F.E.)
| | - Katrin Ganser
- Department of Radiation Oncology, University of Tübingen, 72076 Tübingen, Germany; (T.V.); (L.K.); (K.G.); (N.S.); (D.Z.); (F.E.)
| | - Nicolai Stransky
- Department of Radiation Oncology, University of Tübingen, 72076 Tübingen, Germany; (T.V.); (L.K.); (K.G.); (N.S.); (D.Z.); (F.E.)
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, 72076 Tübingen, Germany
| | - Daniel Zips
- Department of Radiation Oncology, University of Tübingen, 72076 Tübingen, Germany; (T.V.); (L.K.); (K.G.); (N.S.); (D.Z.); (F.E.)
- German Cancer Consortium (DKTK), Partner Site Tübingen, Tübingen, Germany, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Franziska Eckert
- Department of Radiation Oncology, University of Tübingen, 72076 Tübingen, Germany; (T.V.); (L.K.); (K.G.); (N.S.); (D.Z.); (F.E.)
- German Cancer Consortium (DKTK), Partner Site Tübingen, Tübingen, Germany, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Stephan M. Huber
- Department of Radiation Oncology, University of Tübingen, 72076 Tübingen, Germany; (T.V.); (L.K.); (K.G.); (N.S.); (D.Z.); (F.E.)
- Correspondence: ; Tel.: +49-(0)7071-29-82183
| |
Collapse
|
345
|
Cirotti C, Contadini C, Barilà D. SRC Kinase in Glioblastoma News from an Old Acquaintance. Cancers (Basel) 2020; 12:cancers12061558. [PMID: 32545574 PMCID: PMC7352599 DOI: 10.3390/cancers12061558] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/05/2020] [Accepted: 06/10/2020] [Indexed: 12/20/2022] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most recalcitrant brain tumors characterized by a tumor microenvironment (TME) that strongly supports GBM growth, aggressiveness, invasiveness, and resistance to therapy. Importantly, a common feature of GBM is the aberrant activation of receptor tyrosine kinases (RTKs) and of their downstream signaling cascade, including the non-receptor tyrosine kinase SRC. SRC is a central downstream intermediate of many RTKs, which triggers the phosphorylation of many substrates, therefore, promoting the regulation of a wide range of different pathways involved in cell survival, adhesion, proliferation, motility, and angiogenesis. In addition to the aforementioned pathways, SRC constitutive activity promotes and sustains inflammation and metabolic reprogramming concurring with TME development, therefore, actively sustaining tumor growth. Here, we aim to provide an updated picture of the molecular pathways that link SRC to these events in GBM. In addition, SRC targeting strategies are discussed in order to highlight strengths and weaknesses of SRC inhibitors in GBM management, focusing our attention on their potentialities in combination with conventional therapeutic approaches (i.e., temozolomide) to ameliorate therapy effectiveness.
Collapse
Affiliation(s)
- Claudia Cirotti
- Department of Biology, University of Rome “Tor Vergata”, 00133 Rome, Italy; (C.C.); (C.C.)
- Laboratory of Signal Transduction, IRCCS-Fondazione Santa Lucia, 00179 Rome, Italy
| | - Claudia Contadini
- Department of Biology, University of Rome “Tor Vergata”, 00133 Rome, Italy; (C.C.); (C.C.)
- Laboratory of Signal Transduction, IRCCS-Fondazione Santa Lucia, 00179 Rome, Italy
| | - Daniela Barilà
- Department of Biology, University of Rome “Tor Vergata”, 00133 Rome, Italy; (C.C.); (C.C.)
- Laboratory of Signal Transduction, IRCCS-Fondazione Santa Lucia, 00179 Rome, Italy
- Correspondence: ; Tel.: +39-065-0170-3168
| |
Collapse
|
346
|
Choi PJ, Park TI, Cooper E, Dragunow M, Denny WA, Jose J. Heptamethine Cyanine Dye Mediated Drug Delivery: Hype or Hope. Bioconjug Chem 2020; 31:1724-1739. [DOI: 10.1021/acs.bioconjchem.0c00302] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Peter J. Choi
- Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Thomas I−H. Park
- Department of Pharmacology & The Centre for Brain Research, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
- Neurosurgical Research Unit, The Centre for Brain Research, University of Auckland, Private Bag
92019, Auckland 1142, New Zealand
| | - Elizabeth Cooper
- Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
- Department of Pharmacology & The Centre for Brain Research, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
- Neurosurgical Research Unit, The Centre for Brain Research, University of Auckland, Private Bag
92019, Auckland 1142, New Zealand
| | - Mike Dragunow
- Department of Pharmacology & The Centre for Brain Research, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
- Neurosurgical Research Unit, The Centre for Brain Research, University of Auckland, Private Bag
92019, Auckland 1142, New Zealand
| | - William A. Denny
- Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Jiney Jose
- Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| |
Collapse
|
347
|
Ho KH, Kuo TC, Lee YT, Chen PH, Shih CM, Cheng CH, Liu AJ, Lee CC, Chen KC. Xanthohumol regulates miR-4749-5p-inhibited RFC2 signaling in enhancing temozolomide cytotoxicity to glioblastoma. Life Sci 2020; 254:117807. [PMID: 32422304 DOI: 10.1016/j.lfs.2020.117807] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 05/07/2020] [Accepted: 05/13/2020] [Indexed: 02/01/2023]
Abstract
AIMS Xanthohumol (XN), a natural prenylated flavonoid isolated from Humulus lupulus L. (hops), possess the therapeutic effects in glioblastoma multiforme (GBM), which is a grade IV aggressive glioma in adults. However, low bioavailability and extractive yield limit the clinical applications of XN. To comprehensively investigate XN-mediated gene networks in inducing cell death is helpful for drug development and cancer research. Therefore, we aim to identify the detailed molecular mechanisms of XN's effects on exhibiting cytotoxicity for GBM therapy. METHODS AND KEY FINDINGS XN significantly induced GBM cell death and enhanced temozolomide (TMZ) cytotoxicity, a first-line therapeutic drug of GBM. XN-mediated transcriptome profiles and canonical pathways were identified. DNA repair signaling, a well-established mechanism against TMZ cytotoxicity, was significantly correlated with XN-downregulated genes. Replication factor C subunit 2 (RFC2), a DNA repair-related gene, was obviously downregulated in XN-treated cells. Higher RFC2 levels which occupied poor patient survival were also observed in high grade GBM patients and tumors. Inhibition of RFC2 reduced cell viability, induced cell apoptosis, and enhanced both XN and TMZ cytotoxicity. By intersecting array data, bioinformatic prediction, and in vitro experiments, microRNA (miR)-4749-5p, a XN-upregulated microRNA, was identified to target to RFC2 3'UTR and inhibited RFC2 expression. A negative correlation existed between miR-4749-5p and RFC2 in GBM patients. Overexpression of miR-4749-5p significantly promoted XN- and TMZ-mediated cytotoxicity, and reduced RFC2 levels. SIGNIFICANCE Consequently, we suggest that miR-4749-5p targeting RFC2 signaling participates in XN-enhanced TMZ cytotoxicity of GBM. Our findings provide new potential therapeutic directions for future GBM therapy.
Collapse
Affiliation(s)
- Kuo-Hao Ho
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taiwan; Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taiwan
| | - Tai-Chih Kuo
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taiwan; Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taiwan
| | - Yi-Ting Lee
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taiwan; Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taiwan
| | - Peng-Hsu Chen
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taiwan; Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taiwan
| | - Chwen-Ming Shih
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taiwan; Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taiwan
| | - Chia-Hsiung Cheng
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taiwan; Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taiwan
| | - Ann-Jeng Liu
- Department of Neurosurgery, Taipei City Hospital Ren-Ai Branch, Taipei, Taiwan
| | - Chin-Cheng Lee
- Department of Pathology and Laboratory Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan.
| | - Ku-Chung Chen
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taiwan; Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taiwan.
| |
Collapse
|
348
|
Brighi C, Reid L, Genovesi LA, Kojic M, Millar A, Bruce Z, White AL, Day BW, Rose S, Whittaker AK, Puttick S. Comparative study of preclinical mouse models of high-grade glioma for nanomedicine research: the importance of reproducing blood-brain barrier heterogeneity. Theranostics 2020; 10:6361-6371. [PMID: 32483457 PMCID: PMC7255036 DOI: 10.7150/thno.46468] [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: 03/29/2020] [Accepted: 04/30/2020] [Indexed: 12/22/2022] Open
Abstract
The clinical translation of new nanoparticle-based therapies for high-grade glioma (HGG) remains extremely poor. This has partly been due to the lack of suitable preclinical mouse models capable of replicating the complex characteristics of recurrent HGG (rHGG), namely the heterogeneous structural and functional characteristics of the blood-brain barrier (BBB). The goal of this study is to compare the characteristics of the tumor BBB of rHGG with two different mouse models of HGG, the ubiquitously used U87 cell line xenograft model and a patient-derived cell line WK1 xenograft model, in order to assess their suitability for nanomedicine research. Method: Structural MRI was used to assess the extent of BBB opening in mouse models with a fully developed tumor, and dynamic contrast enhanced MRI was used to obtain values of BBB permeability in contrast enhancing tumor. H&E and immunofluorescence staining were used to validate results obtained from the in vivo imaging studies. Results: The extent of BBB disruption and permeability in the contrast enhancing tumor was significantly higher in the U87 model than in rHGG. These values in the WK1 model are similar to those of rHGG. The U87 model is not infiltrative, has an entirely abnormal and leaky vasculature and it is not of glial origin. The WK1 model infiltrates into the non-neoplastic brain parenchyma, it has both regions with intact BBB and regions with leaky BBB and remains of glial origin. Conclusion: The WK1 mouse model more accurately reproduces the extent of BBB disruption, the level of BBB permeability and the histopathological characteristics found in rHGG patients than the U87 mouse model, and is therefore a more clinically relevant model for preclinical evaluations of emerging nanoparticle-based therapies for HGG.
Collapse
|
349
|
Lyakhova I, Piatkova M, Gulaia V, Romanishin A, Shmelev M, Bryukhovetskiy A, Sharma A, Sharma HS, Khotimchenko R, Bryukhovetskiy I. Alkaloids of fascaplysin are promising chemotherapeutic agents for the treatment of glioblastoma: Review. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 151:299-324. [PMID: 32448613 DOI: 10.1016/bs.irn.2020.03.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Glioblastoma is one of the most aggressive human brain tumors. Even following all the modern protocols of complex treatment, the median patient survival typically does not exceed 15 months. This review analyzes the main reasons for glioblastoma resistance to therapy, as well as attempts at categorizing the main approaches to increasing chemotherapy efficiency. Special emphasis is placed on the specific group of compounds, known as marine alkaloids and their synthetic derivatives exerting a general antitumor effect on glioblastoma cells. The unique mechanisms of marine alkaloid influence on the tumor cells prompt considering them as a promising basis for creating new chemotherapeutic agents for glioblastoma treatment.
Collapse
Affiliation(s)
- Irina Lyakhova
- Department of Fundamental Medicine, School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
| | - Mariia Piatkova
- Department of Fundamental Medicine, School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
| | - Valeriia Gulaia
- Laboratory of Biomedical Cell Technologies, Department of Medical Biology and Biotechnology, School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
| | - Aleksandr Romanishin
- Laboratory of Biomedical Cell Technologies, Department of Medical Biology and Biotechnology, School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
| | - Mikhail Shmelev
- Laboratory of Biomedical Cell Technologies, Department of Medical Biology and Biotechnology, School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
| | - Andrey Bryukhovetskiy
- NeuroVita Clinic of Interventional and Restorative Neurology and Therapy, Moscow, Russia
| | - Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, University Hospital, Uppsala University, S-75185 Uppsala, Sweden
| | - Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, University Hospital, Uppsala University, S-75185 Uppsala, Sweden
| | - Rodion Khotimchenko
- Department of Fundamental Medicine, School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia; Laboratory of Pharmacology, National Scientific Center of Marine Biology, Far East Branch of the Russian Academy of Sciences, Vladivostok, Russia
| | - Igor Bryukhovetskiy
- Department of Fundamental Medicine, School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia; Laboratory of Pharmacology, National Scientific Center of Marine Biology, Far East Branch of the Russian Academy of Sciences, Vladivostok, Russia.
| |
Collapse
|
350
|
Bryukhovetskiy I, Pak O, Khotimchenko Y, Bryukhovetskiy A, Sharma A, Sharma HS. Personalized therapy and stem cell transplantation for pro-inflammatory modulation of cancer stem cells microenvironment in glioblastoma: Review. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 151:67-98. [PMID: 32448615 DOI: 10.1016/bs.irn.2020.03.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Glioblastoma multiforme (GBM) is one of the most aggressive types of brain tumor in humans. The prognosis for patients with GBM is unfavorable and treatment is largely ineffective, where modern treatment regimens typically increase survival by 15 months. GBM relapse and progression are associated with cancer stem cells (CSCs). The present review provides a critical analysis of the primary reasons underlying the lack of effectiveness of modern CSC management methods. An emphasis is placed on the role of the blood-brain barrier in the development of treatment resistance. The existing methods for increasing the efficiency of antitumor genotoxic therapy are also described, and a strategy for personalized regulation of CSC based on post-genome technologies is suggested. The hypothesis that GBM cells employ a special mechanism for DNA repair based on their interactions with normal stem cells, is presented and the function of the tumor microenvironment in fulfilling the antitumor potential of normal stem cells is explained. Additionally, the mechanisms by which cancer stem cells regulate glioblastoma progression and recurrence are described based on novel biomedical technologies.
Collapse
Affiliation(s)
- Igor Bryukhovetskiy
- Department of Fundamental Medicine, School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia; Laboratory of Pharmacology, National Scientific Center of Marine Biology, Far East Branch of the Russian Academy of Sciences, Vladivostok, Russia.
| | - Oleg Pak
- Medical Center, Far Eastern Federal University, Vladivostok, Russia
| | - Yuri Khotimchenko
- Department of Fundamental Medicine, School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia; Laboratory of Pharmacology, National Scientific Center of Marine Biology, Far East Branch of the Russian Academy of Sciences, Vladivostok, Russia
| | - Andrey Bryukhovetskiy
- NeuroVita Clinic of Interventional and Restorative Neurology and Therapy, Moscow, Russia
| | - Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, University Hospital, Uppsala University, S-75185 Uppsala, Sweden
| | - Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, University Hospital, Uppsala University, S-75185 Uppsala, Sweden
| |
Collapse
|