1
|
Li KL, Lewis D, Zhu X, Coope DJ, Djoukhadar I, King AT, Cootes T, Jackson A. A Novel Multi-Model High Spatial Resolution Method for Analysis of DCE MRI Data: Insights from Vestibular Schwannoma Responses to Antiangiogenic Therapy in Type II Neurofibromatosis. Pharmaceuticals (Basel) 2023; 16:1282. [PMID: 37765090 PMCID: PMC10534691 DOI: 10.3390/ph16091282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/01/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
This study aimed to develop and evaluate a new DCE-MRI processing technique that combines LEGATOS, a dual-temporal resolution DCE-MRI technique, with multi-kinetic models. This technique enables high spatial resolution interrogation of flow and permeability effects, which is currently challenging to achieve. Twelve patients with neurofibromatosis type II-related vestibular schwannoma (20 tumours) undergoing bevacizumab therapy were imaged at 1.5 T both before and at 90 days following treatment. Using the new technique, whole-brain, high spatial resolution images of the contrast transfer coefficient (Ktrans), vascular fraction (vp), extravascular extracellular fraction (ve), capillary plasma flow (Fp), and the capillary permeability-surface area product (PS) could be obtained, and their predictive value was examined. Of the five microvascular parameters derived using the new method, baseline PS exhibited the strongest correlation with the baseline tumour volume (p = 0.03). Baseline ve showed the strongest correlation with the change in tumour volume, particularly the percentage tumour volume change at 90 days after treatment (p < 0.001), and PS demonstrated a larger reduction at 90 days after treatment (p = 0.0001) when compared to Ktrans or Fp alone. Both the capillary permeability-surface area product (PS) and the extravascular extracellular fraction (ve) significantly differentiated the 'responder' and 'non-responder' tumour groups at 90 days (p < 0.05 and p < 0.001, respectively). These results highlight that this novel DCE-MRI analysis approach can be used to evaluate tumour microvascular changes during treatment and the need for future larger clinical studies investigating its role in predicting antiangiogenic therapy response.
Collapse
Affiliation(s)
- Ka-Loh Li
- Division of Informatics, Imaging and Data Sciences, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK; (K.-L.L.); (T.C.); (A.J.)
- Geoffrey Jefferson Brain Research Centre, University of Manchester, Manchester M13 9PL, UK; (D.L.); (D.J.C.); (A.T.K.)
| | - Daniel Lewis
- Geoffrey Jefferson Brain Research Centre, University of Manchester, Manchester M13 9PL, UK; (D.L.); (D.J.C.); (A.T.K.)
- Department of Neurosurgery, Manchester Centre for Clinical Neurosciences, Salford Royal NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M13 9NT, UK
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK
| | - Xiaoping Zhu
- Division of Informatics, Imaging and Data Sciences, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK; (K.-L.L.); (T.C.); (A.J.)
- Wolfson Molecular Imaging Centre, University of Manchester, 27 Palatine Road, Manchester M20 3LJ, UK
| | - David J. Coope
- Geoffrey Jefferson Brain Research Centre, University of Manchester, Manchester M13 9PL, UK; (D.L.); (D.J.C.); (A.T.K.)
- Department of Neurosurgery, Manchester Centre for Clinical Neurosciences, Salford Royal NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M13 9NT, UK
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK
| | - Ibrahim Djoukhadar
- Department of Neuroradiology, Manchester Centre for Clinical Neurosciences, Salford Royal NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M13 9NT, UK;
| | - Andrew T. King
- Geoffrey Jefferson Brain Research Centre, University of Manchester, Manchester M13 9PL, UK; (D.L.); (D.J.C.); (A.T.K.)
- Department of Neurosurgery, Manchester Centre for Clinical Neurosciences, Salford Royal NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M13 9NT, UK
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester M13 9PL, UK
| | - Timothy Cootes
- Division of Informatics, Imaging and Data Sciences, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK; (K.-L.L.); (T.C.); (A.J.)
| | - Alan Jackson
- Division of Informatics, Imaging and Data Sciences, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK; (K.-L.L.); (T.C.); (A.J.)
| |
Collapse
|
2
|
Hicks WH, Bird CE, Traylor JI, Shi DD, El Ahmadieh TY, Richardson TE, McBrayer SK, Abdullah KG. Contemporary Mouse Models in Glioma Research. Cells 2021; 10:cells10030712. [PMID: 33806933 PMCID: PMC8004772 DOI: 10.3390/cells10030712] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 03/20/2021] [Accepted: 03/20/2021] [Indexed: 02/07/2023] Open
Abstract
Despite advances in understanding of the molecular pathogenesis of glioma, outcomes remain dismal. Developing successful treatments for glioma requires faithful in vivo disease modeling and rigorous preclinical testing. Murine models, including xenograft, syngeneic, and genetically engineered models, are used to study glioma-genesis, identify methods of tumor progression, and test novel treatment strategies. Since the discovery of highly recurrent isocitrate dehydrogenase (IDH) mutations in lower-grade gliomas, there is increasing emphasis on effective modeling of IDH mutant brain tumors. Improvements in preclinical models that capture the phenotypic and molecular heterogeneity of gliomas are critical for the development of effective new therapies. Herein, we explore the current status, advancements, and challenges with contemporary murine glioma models.
Collapse
Affiliation(s)
- William H. Hicks
- Department of Neurological Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA; (W.H.H.); (C.E.B.); (J.I.T.); (T.Y.E.A.)
| | - Cylaina E. Bird
- Department of Neurological Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA; (W.H.H.); (C.E.B.); (J.I.T.); (T.Y.E.A.)
| | - Jeffrey I. Traylor
- Department of Neurological Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA; (W.H.H.); (C.E.B.); (J.I.T.); (T.Y.E.A.)
| | - Diana D. Shi
- Department of Radiation Oncology, Brigham and Women’s Hospital and Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA;
| | - Tarek Y. El Ahmadieh
- Department of Neurological Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA; (W.H.H.); (C.E.B.); (J.I.T.); (T.Y.E.A.)
| | - Timothy E. Richardson
- Department of Pathology, Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, University of Texas Health San Antonio, San Antonio, TX 75229, USA;
| | - Samuel K. McBrayer
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Harrold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
- Correspondence: (S.K.M.); (K.G.A.)
| | - Kalil G. Abdullah
- Department of Neurological Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA; (W.H.H.); (C.E.B.); (J.I.T.); (T.Y.E.A.)
- Harrold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
- Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
- Correspondence: (S.K.M.); (K.G.A.)
| |
Collapse
|
3
|
Lorusso G, Rüegg C, Kuonen F. Targeting the Extra-Cellular Matrix-Tumor Cell Crosstalk for Anti-Cancer Therapy: Emerging Alternatives to Integrin Inhibitors. Front Oncol 2020; 10:1231. [PMID: 32793493 PMCID: PMC7387567 DOI: 10.3389/fonc.2020.01231] [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: 01/10/2020] [Accepted: 06/16/2020] [Indexed: 12/11/2022] Open
Abstract
The extracellular matrix (ECM) is a complex network composed of a multitude of different macromolecules. ECM components typically provide a supportive structure to the tissue and engender positional information and crosstalk with neighboring cells in a dynamic reciprocal manner, thereby regulating tissue development and homeostasis. During tumor progression, tumor cells commonly modify and hijack the surrounding ECM to sustain anchorage-dependent growth and survival, guide migration, store pro-tumorigenic cell-derived molecules and present them to enhance receptor activation. Thereby, ECM potentially supports tumor progression at various steps from initiation, to local growth, invasion, and systemic dissemination and ECM-tumor cells interactions have long been considered promising targets for cancer therapy. Integrins represent key surface receptors for the tumor cell to sense and interact with the ECM. Yet, attempts to therapeutically impinge on these interactions using integrin inhibitors have failed to deliver anticipated results, and integrin inhibitors are still missing in the emerging arsenal of drugs for targeted therapies. This paradox situation should urge the field to reconsider the role of integrins in cancer and their targeting, but also to envisage alternative strategies. Here, we review the therapeutic targets implicated in tumor cell adhesion to the ECM, whose inhibitors are currently in clinical trials and may offer alternatives to integrin inhibition.
Collapse
Affiliation(s)
- Girieca Lorusso
- Experimental and Translational Oncology, Department of Oncology Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Curzio Rüegg
- Experimental and Translational Oncology, Department of Oncology Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - François Kuonen
- Department of Dermatology and Venereology, Hôpital de Beaumont, Lausanne University Hospital Center, Lausanne, Switzerland
| |
Collapse
|
4
|
van Lith SAM, Roodink I, Verhoeff JJC, Mäkinen PI, Lappalainen JP, Ylä-Herttuala S, Raats J, van Wijk E, Roepman R, Letteboer SJ, Verrijp K, Leenders WPJ. In vivo phage display screening for tumor vascular targets in glioblastoma identifies a llama nanobody against dynactin-1-p150Glued. Oncotarget 2018; 7:71594-71607. [PMID: 27689404 PMCID: PMC5342104 DOI: 10.18632/oncotarget.12261] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 09/19/2016] [Indexed: 12/23/2022] Open
Abstract
Diffuse gliomas are primary brain cancers that are characterised by infiltrative growth. Whereas high-grade glioma characteristically presents with perinecrotic neovascularisation, large tumor areas thrive on pre-existent vasculature as well. Clinical studies have revealed that pharmacological inhibition of the angiogenic process does not improve survival of glioblastoma patients. Direct targeting of tumor vessels may however still be an interesting therapeutic approach as it allows pinching off the blood supply to tumor cells. Such tumor vessel targeting requires the identification of tumor-specific vascular targeting agents (TVTAs). Here we describe a novel TVTA, C-C7, which we identified via in vivo biopanning of a llama nanobody phage display library in an orthotopic mouse model of diffuse glioma. We show that C-C7 recognizes a subpopulation of tumor blood vessels in glioma xenografts and clinical glioma samples. Additionally, C-C7 recognizes macrophages and activated endothelial cells in atherosclerotic lesions. By using C-C7 as bait in yeast-2-hybrid (Y2H) screens we identified dynactin-1-p150Glued as its binding partner. The interaction was confirmed by co-immunostainings with C-C7 and a commercial anti-dynactin-1-p150Glued antibody, and via co-immunoprecipitation/western blot studies. Normal brain vessels do not express dynactin-1-p150Glued and its expression is reduced under anti-VEGF therapy, suggesting that dynactin-1-p150Glued is a marker for activated endothelial cells. In conclusion, we show that in vivo phage display combined with Y2H screenings provides a powerful approach to identify tumor-targeting nanobodies and their binding partners. Using this combination of methods we identify dynactin-1-p150Glued as a novel targetable protein on activated endothelial cells and macrophages.
Collapse
Affiliation(s)
| | - Ilse Roodink
- Department of Pathology, RadboudUMC, 6500 HB, Nijmegen, The Netherlands.,Modiquest BV, LSP, Molenstraat 110, 5342 CC, Oss, The Netherlands
| | | | - Petri I Mäkinen
- Department of Biotechnology and Molecular Medicine, University of Eastern Finland, FI-70211, Kuopio, Finland
| | - Jari P Lappalainen
- Department of Biotechnology and Molecular Medicine, University of Eastern Finland, FI-70211, Kuopio, Finland
| | - Seppo Ylä-Herttuala
- Department of Biotechnology and Molecular Medicine, University of Eastern Finland, FI-70211, Kuopio, Finland.,Science Service Center and Gene Therapy Unit, Kuopio University Hospital, 70210 Kuopio, Finland
| | - Jos Raats
- Modiquest BV, LSP, Molenstraat 110, 5342 CC, Oss, The Netherlands
| | - Erwin van Wijk
- Department of Otorhinolaryngology, RadboudUMC, 6500 HB, Nijmegen, The Netherlands
| | - Ronald Roepman
- Department of Genetics, RadboudUMC, 6500 HB, Nijmegen,The Netherlands
| | - Stef J Letteboer
- Department of Genetics, RadboudUMC, 6500 HB, Nijmegen,The Netherlands
| | - Kiek Verrijp
- Department of Pathology, RadboudUMC, 6500 HB, Nijmegen, The Netherlands
| | | |
Collapse
|
5
|
Cenciarelli C, Marei HE, Felsani A, Casalbore P, Sica G, Puglisi MA, Cameron AJM, Olivi A, Mangiola A. PDGFRα depletion attenuates glioblastoma stem cells features by modulation of STAT3, RB1 and multiple oncogenic signals. Oncotarget 2018; 7:53047-53063. [PMID: 27344175 PMCID: PMC5288168 DOI: 10.18632/oncotarget.10132] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 06/09/2016] [Indexed: 12/15/2022] Open
Abstract
Platelet derived growth factor receptors (PDGFRs) play an important role in tumor pathogenesis, and they are frequently overexpressed in glioblastoma (GBM). Earlier we have shown a higher protein expression of PDGFR isoforms (α and β) in peritumoral-tissue derived cancer stem cells (p-CSC) than in tumor core (c-CSC) of several GBM affected patients. In the current study, in order to assess the activity of PDGFRα/PDGF-AA signaling axis, we performed time course experiments to monitor the effects of exogenous PDGF-AA on the expression of downstream target genes in c-CSC vs p-CSC. Interestingly, in p-CSC we detected the upregulation of Y705-phosphorylated Stat3, concurrent with a decrement of Rb1 protein in its active state, within minutes of PDGF-AA addition. This finding prompted us to elucidate the role of PDGFRα in self-renewal, invasion and differentiation in p-CSC by using short hairpin RNA depletion of PDGFRα expression. Notably, in PDGFRα-depleted cells, protein analysis revealed attenuation of stemness-related and glial markers expression, alongside early activation of the neuronal marker MAP2a/b that correlated with the induction of tumor suppressor Rb1. The in vitro reduction of the invasive capacity of PDGFRα-depleted CSC as compared to parental cells correlated with the downmodulation of markers of epithelial-mesenchymal transition phenotype and angiogenesis. Surprisingly, we observed the induction of anti-apoptotic proteins and compensatory oncogenic signals such as EDN1, EDNRB, PRKCB1, PDGF-C and PDGF-D. To conclude, we hypothesize that the newly discovered PDGFRα/Stat3/Rb1 regulatory axis might represent a potential therapeutic target for GBM treatment.
Collapse
Affiliation(s)
- Carlo Cenciarelli
- Institute of Translational Pharmacology, Department of Biomedical Sciences-National Research Council (IFT-CNR), Rome, Italy
| | - Hany E Marei
- Biomedical Research Center, Qatar University, Doha, Qatar
| | - Armando Felsani
- Institute of Cell Biology and Neurobiology, Dept. of Biomedical Sciences-National Research Council (IBCN-CNR), Rome, Italy
| | - Patrizia Casalbore
- Institute of Cell Biology and Neurobiology, Dept. of Biomedical Sciences-National Research Council (IBCN-CNR), Rome, Italy
| | - Gigliola Sica
- Institute of Histology and Embryology, Catholic University-School of Medicine, Rome, Italy
| | | | - Angus J M Cameron
- Barts Cancer Institute, John Vane Science Centre, Queen Mary University of London, London, United Kingdom
| | - Alessandro Olivi
- Institute of Neurosurgery, Department of Head and Neck, Catholic University-School of Medicine, Rome, Italy
| | - Annunziato Mangiola
- Institute of Neurosurgery, Department of Head and Neck, Catholic University-School of Medicine, Rome, Italy
| |
Collapse
|
6
|
Veldhuijzen van Zanten SEM, Sewing ACP, van Lingen A, Hoekstra OS, Wesseling P, Meel MH, van Vuurden DG, Kaspers GJL, Hulleman E, Bugiani M. Multiregional Tumor Drug-Uptake Imaging by PET and Microvascular Morphology in End-Stage Diffuse Intrinsic Pontine Glioma. J Nucl Med 2017; 59:612-615. [PMID: 28818988 DOI: 10.2967/jnumed.117.197897] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 08/07/2017] [Indexed: 11/16/2022] Open
Abstract
Inadequate tumor uptake of the vascular endothelial growth factor antibody bevacizumab could explain lack of effect in diffuse intrinsic pontine glioma. Methods: By combining data from a PET imaging study using 89Zr-labeled bevacizumab and an autopsy study, a 1-on-1 analysis of multiregional in vivo and ex vivo 89Zr-bevacizumab uptake, tumor histology, and vascular morphology in a diffuse intrinsic pontine glioma patient was performed. Results: In vivo 89Zr-bevacizumab measurements showed heterogeneity between lesions. Additional ex vivo measurements and immunohistochemistry of cervicomedullary metastasis samples showed uptake to be highest in the area with marked microvascular proliferation. In the primary pontine tumor, all samples showed similar vascular morphology. Other histologic features were similar between the samples studied. Conclusion: In vivo 89Zr-bevacizumab PET serves to identify heterogeneous uptake between tumor lesions, whereas subcentimeter intralesional heterogeneity could be identified only by ex vivo measurements. 89Zr-bevacizumab uptake is enhanced by vascular proliferation, although our results suggest it is not the only determinant of intralesional uptake heterogeneity.
Collapse
Affiliation(s)
- Sophie E M Veldhuijzen van Zanten
- Division of Oncology/Haematology, Department of Pediatrics, VUmc, Amsterdam, The Netherlands .,Neuro-Oncology Research Group, Cancer Center Amsterdam, VUmc, Amsterdam, The Netherlands
| | - A Charlotte P Sewing
- Division of Oncology/Haematology, Department of Pediatrics, VUmc, Amsterdam, The Netherlands.,Neuro-Oncology Research Group, Cancer Center Amsterdam, VUmc, Amsterdam, The Netherlands
| | - Arthur van Lingen
- Department of Radiology and Nuclear Medicine, VUmc, Amsterdam, The Netherlands
| | - Otto S Hoekstra
- Department of Radiology and Nuclear Medicine, VUmc, Amsterdam, The Netherlands
| | - Pieter Wesseling
- Neuro-Oncology Research Group, Cancer Center Amsterdam, VUmc, Amsterdam, The Netherlands.,Department of Pathology, VUmc, Amsterdam, The Netherlands.,Department of Pathology, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands; and
| | - Michaël H Meel
- Division of Oncology/Haematology, Department of Pediatrics, VUmc, Amsterdam, The Netherlands.,Neuro-Oncology Research Group, Cancer Center Amsterdam, VUmc, Amsterdam, The Netherlands
| | - Dannis G van Vuurden
- Division of Oncology/Haematology, Department of Pediatrics, VUmc, Amsterdam, The Netherlands.,Neuro-Oncology Research Group, Cancer Center Amsterdam, VUmc, Amsterdam, The Netherlands
| | - Gertjan J L Kaspers
- Division of Oncology/Haematology, Department of Pediatrics, VUmc, Amsterdam, The Netherlands.,Department of Pediatrics, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Esther Hulleman
- Division of Oncology/Haematology, Department of Pediatrics, VUmc, Amsterdam, The Netherlands.,Neuro-Oncology Research Group, Cancer Center Amsterdam, VUmc, Amsterdam, The Netherlands
| | | |
Collapse
|
7
|
van den Heuvel CNAM, Navis AC, de Bitter T, Amiri H, Verrijp K, Heerschap A, Rex K, Dussault I, Caenepeel S, Coxon A, Span PN, Wesseling P, Hendriks W, Leenders WPJ. Selective MET Kinase Inhibition in MET-Dependent Glioma Models Alters Gene Expression and Induces Tumor Plasticity. Mol Cancer Res 2017; 15:1587-1597. [PMID: 28751462 DOI: 10.1158/1541-7786.mcr-17-0177] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 06/15/2017] [Accepted: 07/24/2017] [Indexed: 11/16/2022]
Abstract
The receptor tyrosine kinase (RTK) MET represents a promising tumor target in a subset of glioblastomas. Most RTK inhibitors available in the clinic today, including those inhibiting MET, affect multiple targets simultaneously. Previously, it was demonstrated that treatment with cabozantinib (MET/VEGFR2/RET inhibitor) prolonged survival of mice carrying orthotopic patient-derived xenografts (PDX) of the MET-addicted glioblastoma model E98, yet did not prevent development of recurrent and cabozantinib-resistant tumors. To exclude VEGFR2 inhibition-inflicted blood-brain barrier normalization and diminished tumor distribution of the drug, we have now investigated the effects of the novel MET-selective inhibitor Compound A in the orthotopic E98 xenograft model. In vitro, Compound A proved a highly potent inhibitor of proliferation of MET-addicted cell lines. In line with its target selectivity, Compound A did not restore the leaky blood-brain barrier and was more effective than cabozantinib in inhibiting MET phosphorylation in vivo Compound A treatment significantly prolonged survival of mice carrying E98 tumor xenografts, but did not prevent eventual progression. Contrasting in vitro results, the Compound A-treated xenografts displayed high levels of AKT phosphorylation despite the absence of phosphorylated MET. Profiling by RNA sequencing showed that in vivo transcriptomes differed significantly from those in control xenografts.Implications: Collectively, these findings demonstrate the plasticity of paracrine growth factor receptor signaling in vivo and urge for prudency with in vitro drug-testing strategies to validate monotherapies. Mol Cancer Res; 15(11); 1587-97. ©2017 AACR.
Collapse
Affiliation(s)
| | - Anna C Navis
- Department of Pathology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Tessa de Bitter
- Department of Pathology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Houshang Amiri
- Department of Radiology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Kiek Verrijp
- Department of Pathology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Arend Heerschap
- Department of Radiology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Karen Rex
- Department of Oncology Research, Amgen Inc., Thousand Oaks, California
| | - Isabelle Dussault
- Department of Oncology Research, Amgen Inc., Thousand Oaks, California
| | - Sean Caenepeel
- Department of Oncology Research, Amgen Inc., Thousand Oaks, California
| | - Angela Coxon
- Department of Oncology Research, Amgen Inc., Thousand Oaks, California
| | - Paul N Span
- Department of Radiation Oncology, Radboud University Medical Centre, Radiotherapy and Oncoimmunology Laboratory, Nijmegen, the Netherlands
| | - Pieter Wesseling
- Department of Pathology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Wiljan Hendriks
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Nijmegen, the Netherlands
| | - William P J Leenders
- Department of Pathology, Radboud University Medical Centre, Nijmegen, the Netherlands.
| |
Collapse
|
8
|
Simon T, Gagliano T, Giamas G. Direct Effects of Anti-Angiogenic Therapies on Tumor Cells: VEGF Signaling. Trends Mol Med 2017; 23:282-292. [DOI: 10.1016/j.molmed.2017.01.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 01/04/2017] [Accepted: 01/09/2017] [Indexed: 12/18/2022]
|
9
|
Lenting K, Verhaak R, Ter Laan M, Wesseling P, Leenders W. Glioma: experimental models and reality. Acta Neuropathol 2017; 133:263-282. [PMID: 28074274 PMCID: PMC5250671 DOI: 10.1007/s00401-017-1671-4] [Citation(s) in RCA: 191] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 01/06/2017] [Accepted: 01/06/2017] [Indexed: 12/12/2022]
Abstract
In theory, in vitro and in vivo models for human gliomas have great potential to not only enhance our understanding of glioma biology, but also to facilitate the development of novel treatment strategies for these tumors. For reliable prediction and validation of the effects of different therapeutic modalities, however, glioma models need to comply with specific and more strict demands than other models of cancer, and these demands are directly related to the combination of genetic aberrations and the specific brain micro-environment gliomas grow in. This review starts with a brief introduction on the pathological and molecular characteristics of gliomas, followed by an overview of the models that have been used in the last decades in glioma research. Next, we will discuss how these models may play a role in better understanding glioma development and especially in how they can aid in the design and optimization of novel therapies. The strengths and weaknesses of the different models will be discussed in light of genotypic, phenotypic and metabolic characteristics of human gliomas. The last part of this review provides some examples of how therapy experiments using glioma models can lead to deceptive results when such characteristics are not properly taken into account.
Collapse
Affiliation(s)
- Krissie Lenting
- Department of Pathology, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Roel Verhaak
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Mark Ter Laan
- Department of Neurosurgery, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Pieter Wesseling
- Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands
- Department of Pathology, Princess Máxima Center for Pediatric Oncology and University Medical Center Utrecht, Utrecht, The Netherlands
| | - William Leenders
- Department of Pathology, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands.
| |
Collapse
|
10
|
Abstract
Pericytes are a heterogeneous population of cells located in the blood vessel wall. They were first identified in the 19th century by Rouget, however their biological role and potential for drug targeting have taken time to be recognised. Isolation of pericytes from several different tissues has allowed a better phenotypic and functional characterization. These findings revealed a tissue-specific, multi-functional group of cells with multilineage potential. Given this emerging evidence, pericytes have acquired specific roles in pathobiological events in vascular diseases. In this review article, we will provide a compelling overview of the main diseases in which pericytes are involved, from well-established mechanisms to the latest findings. Pericyte involvement in diabetes and cancer will be discussed extensively. In the last part of the article we will review therapeutic approaches for these diseases in light of the recently acquired knowledge. To unravel pericyte-related vascular pathobiological events is pivotal not only for more tailored treatments of disease but also to establish pericytes as a therapeutic tool.
Collapse
|
11
|
van Lith SAM, Navis AC, Lenting K, Verrijp K, Schepens JTG, Hendriks WJAJ, Schubert NA, Venselaar H, Wevers RA, van Rooij A, Wesseling P, Molenaar RJ, van Noorden CJF, Pusch S, Tops B, Leenders WPJ. Identification of a novel inactivating mutation in Isocitrate Dehydrogenase 1 (IDH1-R314C) in a high grade astrocytoma. Sci Rep 2016; 6:30486. [PMID: 27460417 PMCID: PMC4962051 DOI: 10.1038/srep30486] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 07/06/2016] [Indexed: 12/16/2022] Open
Abstract
The majority of low-grade and secondary high-grade gliomas carry heterozygous hotspot mutations in cytosolic isocitrate dehydrogenase 1 (IDH1) or the mitochondrial variant IDH2. These mutations mostly involve Arg132 in IDH1, and Arg172 or Arg140 in IDH2. Whereas IDHs convert isocitrate to alpha-ketoglutarate (α-KG) with simultaneous reduction of NADP+ to NADPH, these IDH mutants reduce α-KG to D-2-hydroxyglutarate (D-2-HG) while oxidizing NADPH. D-2-HG is a proposed oncometabolite, acting via competitive inhibition of α-KG-dependent enzymes that are involved in metabolism and epigenetic regulation. However, much less is known about the implications of the metabolic stress, imposed by decreased α-KG and NADPH production, for tumor biology. We here present a novel heterozygous IDH1 mutation, IDH1R314C, which was identified by targeted next generation sequencing of a high grade glioma from which a mouse xenograft model and a cell line were generated. IDH1R314C lacks isocitrate-to-α-KG conversion activity due to reduced affinity for NADP+, and differs from the IDH1R132 mutants in that it does not produce D-2-HG. Because IDH1R314C is defective in producing α-KG and NADPH, without concomitant production of the D-2-HG, it represents a valuable tool to study the effects of IDH1-dysfunction on cellular metabolism in the absence of this oncometabolite.
Collapse
Affiliation(s)
| | - Anna C Navis
- Department of Pathology, Radboudumc, Nijmegen, The Netherlands
| | - Krissie Lenting
- Department of Pathology, Radboudumc, Nijmegen, The Netherlands
| | - Kiek Verrijp
- Department of Pathology, Radboudumc, Nijmegen, The Netherlands
| | - Jan T G Schepens
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, The Netherlands
| | - Wiljan J A J Hendriks
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, The Netherlands
| | - Nil A Schubert
- Department of Pathology, Radboudumc, Nijmegen, The Netherlands
| | - Hanka Venselaar
- Centre for Molecular and Biomolecular Informatics, Radboud Institute for Molecular Life Sciences, The Netherlands
| | - Ron A Wevers
- Translational Metabolic Laboratory, Department Laboratory Medicine, Radboudumc, Nijmegen, The Netherlands
| | - Arno van Rooij
- Translational Metabolic Laboratory, Department Laboratory Medicine, Radboudumc, Nijmegen, The Netherlands
| | - Pieter Wesseling
- Department of Pathology, Radboudumc, Nijmegen, The Netherlands.,Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands
| | - Remco J Molenaar
- Department of Cell Biology and Histology, Academic Medical Center, Amsterdam, The Netherlands
| | | | - Stefan Pusch
- Clinical Cooperation Unit Neuropathology, German Cancer Center (DKFZ), Heidelberg, Germany
| | - Bastiaan Tops
- Department of Pathology, Radboudumc, Nijmegen, The Netherlands
| | | |
Collapse
|
12
|
Jansen MHA, Lagerweij T, Sewing ACP, Vugts DJ, van Vuurden DG, Molthoff CFM, Caretti V, Veringa SJE, Petersen N, Carcaboso AM, Noske DP, Vandertop WP, Wesseling P, van Dongen GAMS, Kaspers GJL, Hulleman E. Bevacizumab Targeting Diffuse Intrinsic Pontine Glioma: Results of 89Zr-Bevacizumab PET Imaging in Brain Tumor Models. Mol Cancer Ther 2016; 15:2166-74. [PMID: 27325687 DOI: 10.1158/1535-7163.mct-15-0558] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 05/26/2016] [Indexed: 11/16/2022]
Abstract
The role of the VEGF inhibitor bevacizumab in the treatment of diffuse intrinsic pontine glioma (DIPG) is unclear. We aim to study the biodistribution and uptake of zirconium-89 ((89)Zr)-labeled bevacizumab in DIPG mouse models. Human E98-FM, U251-FM glioma cells, and HSJD-DIPG-007-FLUC primary DIPG cells were injected into the subcutis, pons, or striatum of nude mice. Tumor growth was monitored by bioluminescence imaging (BLI) and visualized by MRI. Seventy-two to 96 hours after (89)Zr-bevacizumab injections, mice were imaged by positron emission tomography (PET), and biodistribution was analyzed ex vivo High VEGF expression in human DIPG was confirmed in a publically available mRNA database, but no significant (89)Zr-bevacizumab uptake could be detected in xenografts located in the pons and striatum at an early or late stage of the disease. E98-FM, and to a lesser extent the U251-FM and HSJD-DIPG-007 subcutaneous tumors, showed high accumulation of (89)Zr-bevacizumab. VEGF expression could not be demonstrated in the intracranial tumors by in situ hybridization (ISH) but was clearly present in the perinecrotic regions of subcutaneous E98-FM tumors. The poor uptake of (89)Zr-bevacizumab in xenografts located in the brain suggests that VEGF targeting with bevacizumab has limited efficacy for diffuse infiltrative parts of glial brain tumors in mice. Translating these results to the clinic would imply that treatment with bevacizumab in patients with DIPG is only justified after targeting of VEGF has been demonstrated by (89)Zr-bevacizumab immuno-PET. We aim to confirm this observation in a clinical PET study with patients with DIPG. Mol Cancer Ther; 15(9); 2166-74. ©2016 AACR.
Collapse
Affiliation(s)
- Marc H A Jansen
- Department of Pediatrics, Pediatric Hematology and Oncology, Cancer Center, Amsterdam, the Netherlands
| | - Tonny Lagerweij
- Neuro-oncology Research Group Cancer Center, Amsterdam, the Netherlands. Department of Neurosurgery VU University Medical Center and Academic Medical Center, Amsterdam, the Netherlands
| | - A Charlotte P Sewing
- Department of Pediatrics, Pediatric Hematology and Oncology, Cancer Center, Amsterdam, the Netherlands. Neuro-oncology Research Group Cancer Center, Amsterdam, the Netherlands
| | - Danielle J Vugts
- Department of Radiology & Nuclear Medicine VU University Medical Center, Amsterdam, the Netherlands
| | - Dannis G van Vuurden
- Department of Pediatrics, Pediatric Hematology and Oncology, Cancer Center, Amsterdam, the Netherlands. Neuro-oncology Research Group Cancer Center, Amsterdam, the Netherlands
| | - Carla F M Molthoff
- Department of Radiology & Nuclear Medicine VU University Medical Center, Amsterdam, the Netherlands
| | - Viola Caretti
- Department of Pediatrics, Pediatric Hematology and Oncology, Cancer Center, Amsterdam, the Netherlands. Neuro-oncology Research Group Cancer Center, Amsterdam, the Netherlands. Departments of Neurology, Pediatrics and Neurosurgery, Stanford University School of Medicine, Stanford, California
| | - Susanna J E Veringa
- Department of Pediatrics, Pediatric Hematology and Oncology, Cancer Center, Amsterdam, the Netherlands. Neuro-oncology Research Group Cancer Center, Amsterdam, the Netherlands
| | - Naomi Petersen
- Neuro-oncology Research Group Cancer Center, Amsterdam, the Netherlands
| | - Angel M Carcaboso
- Preclinical Therapeutics and Drug Delivery Research Program, Department of Oncology, Hospital Sant Joan de Déu, Barcelona, Spain
| | - David P Noske
- Neuro-oncology Research Group Cancer Center, Amsterdam, the Netherlands. Department of Neurosurgery VU University Medical Center and Academic Medical Center, Amsterdam, the Netherlands
| | - W Peter Vandertop
- Department of Neurosurgery VU University Medical Center and Academic Medical Center, Amsterdam, the Netherlands
| | - Pieter Wesseling
- Neuro-oncology Research Group Cancer Center, Amsterdam, the Netherlands. Department of Pathology VU University Medical Center. Amsterdam, the Netherlands. Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Guus A M S van Dongen
- Department of Radiology & Nuclear Medicine VU University Medical Center, Amsterdam, the Netherlands
| | - Gertjan J L Kaspers
- Department of Pediatrics, Pediatric Hematology and Oncology, Cancer Center, Amsterdam, the Netherlands
| | - Esther Hulleman
- Department of Pediatrics, Pediatric Hematology and Oncology, Cancer Center, Amsterdam, the Netherlands. Neuro-oncology Research Group Cancer Center, Amsterdam, the Netherlands.
| |
Collapse
|
13
|
Bourgonje AM, Navis AC, Schepens JTG, Verrijp K, Hovestad L, Hilhorst R, Harroch S, Wesseling P, Leenders WPJ, Hendriks WJAJ. Intracellular and extracellular domains of protein tyrosine phosphatase PTPRZ-B differentially regulate glioma cell growth and motility. Oncotarget 2015; 5:8690-702. [PMID: 25238264 PMCID: PMC4226714 DOI: 10.18632/oncotarget.2366] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Gliomas are primary brain tumors for which surgical resection and radiotherapy is difficult because of the diffuse infiltrative growth of the tumor into the brain parenchyma. For development of alternative, drug-based, therapies more insight in the molecular processes that steer this typical growth and morphodynamic behavior of glioma cells is needed. Protein tyrosine phosphatase PTPRZ-B is a transmembrane signaling molecule that is found to be strongly up-regulated in glioma specimens. We assessed the contribution of PTPRZ-B protein domains to tumor cell growth and migration, via lentiviral knock-down and over-expression using clinically relevant glioma xenografts and their derived cell models. PTPRZ-B knock-down resulted in reduced migration and proliferation of glioma cells in vitro and also inhibited tumor growth in vivo. Interestingly, expression of only the PTPRZ-B extracellular segment was sufficient to rescue the in vitro migratory phenotype that resulted from PTPRZ-B knock-down. In contrast, PTPRZ-B knock-down effects on proliferation could be reverted only after re-expression of PTPRZ-B variants that contained its C-terminal PDZ binding domain. Thus, distinct domains of PTPRZ-B are differentially required for migration and proliferation of glioma cells, respectively. PTPRZ-B signaling pathways therefore represent attractive therapeutic entry points to combat these tumors.
Collapse
Affiliation(s)
- Annika M Bourgonje
- Department of Cell Biology , Radboud University Medical Center, Nijmegen, The Netherlands
| | - Anna C Navis
- Department of Pathology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jan T G Schepens
- Department of Cell Biology , Radboud University Medical Center, Nijmegen, The Netherlands
| | - Kiek Verrijp
- Department of Pathology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Riet Hilhorst
- PamGene International BV, 's-Hertogenbosch, The Netherlands
| | - Sheila Harroch
- Department of Neuroscience, Institut Pasteur, Paris, France
| | - Pieter Wesseling
- Department of Pathology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands. Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands
| | - William P J Leenders
- Department of Pathology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Wiljan J A J Hendriks
- Department of Cell Biology , Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
14
|
AXL as a modulator of sunitinib response in glioblastoma cell lines. Exp Cell Res 2015; 332:1-10. [PMID: 25637219 DOI: 10.1016/j.yexcr.2015.01.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2014] [Revised: 01/15/2015] [Accepted: 01/19/2015] [Indexed: 02/01/2023]
Abstract
Receptor tyrosine kinase (RTK) targeted therapy has been explored for glioblastoma treatment. However, it is unclear which RTK inhibitors are the most effective and there are no predictive biomarkers available. We recently identified the RTK AXL as a putative target for the pan-RTK inhibitors cediranib and sunitinib, which are under clinical trials for glioblastoma patients. Here, we provide evidence that AXL activity can modulate sunitinib response in glioblastoma cell lines. We found that AXL knockdown conferred lower sensitivity to sunitinib by rescuing migratory defects and inhibiting apoptosis in cells expressing high AXL basal levels. Accordingly, overactivation of AXL by its ligand GAS6 rendered AXL positive glioblastoma cells more sensitive to sunitinib. AXL knockdown induced a cellular rewiring of several growth signaling pathways through activation of RTKs, such as EGFR, as well as intracellular pathways such as MAPK and AKT. The combination of sunitinib with a specific AKT inhibitor reverted the resistance of AXL-silenced cells to sunitinib. Together, our results suggest that sunitinib inhibits AXL and AXL activation status modulates therapy response of glioblastoma cells to sunitinib. Moreover, it indicates that combining sunitinib therapy with AKT pathway inhibitors could overcome sunitinib resistance.
Collapse
|
15
|
Glutamate as chemotactic fuel for diffuse glioma cells: are they glutamate suckers? Biochim Biophys Acta Rev Cancer 2014; 1846:66-74. [PMID: 24747768 DOI: 10.1016/j.bbcan.2014.04.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 04/09/2014] [Accepted: 04/11/2014] [Indexed: 11/21/2022]
Abstract
Diffuse gliomas comprise a group of primary brain tumors that originate from glial (precursor) cells and present as a variety of malignancy grades which have in common that they grow by diffuse infiltration. This phenotype complicates treatment enormously as it precludes curative surgery and radiotherapy. Furthermore, diffusely infiltrating glioma cells often hide behind a functional blood-brain barrier, hampering delivery of systemically administered therapeutic and diagnostic compounds to the tumor cells. The present review addresses the biological mechanisms that underlie the diffuse infiltrative phenotype, knowledge of which may improve treatment strategies for this disastrous tumor type. The invasive phenotype is specific for glioma: most other brain tumor types, both primary and metastatic, grow as delineated lesions. Differences between the genetic make-up of glioma and that of other tumor types may therefore help to unravel molecular pathways, involved in diffuse infiltrative growth. One such difference concerns mutations in the NADP(+)-dependent isocitrate dehydrogenase (IDH1 and IDH2) genes, which occur in >80% of cases of low grade glioma and secondary glioblastoma. In this review we present a novel hypothesis which links IDH1 and IDH2 mutations to glutamate metabolism, possibly explaining the specific biological behavior of diffuse glioma.
Collapse
|
16
|
Hamans B, Navis AC, Wright A, Wesseling P, Heerschap A, Leenders W. Multivoxel ¹H MR spectroscopy is superior to contrast-enhanced MRI for response assessment after anti-angiogenic treatment of orthotopic human glioma xenografts and provides handles for metabolic targeting. Neuro Oncol 2013; 15:1615-24. [PMID: 24158109 DOI: 10.1093/neuonc/not129] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Anti-angiogenic treatment of glioblastoma characteristically results in therapy resistance and tumor progression via diffuse infiltration. Monitoring tumor progression in these patients is thwarted because therapy results in tumor invisibility in contrast-enhanced (CE) MRI. To address this problem, we examined whether tumor progression could be monitored by metabolic mapping using (1)H MR spectroscopic imaging (MRSI). METHODS We treated groups of BALB/c nu/nu mice carrying different orthotopic diffuse-infiltrative glioblastoma xenografts with bevacizumab (anti-vascular endothelial growth factor [VEGF] antibody, n = 13), cabozantinib (combined VEGF receptor 2/c-Met tyrosine kinase inhibitor, n = 11), or placebo (n = 15) and compared CE-MRI with MRS-derived metabolic maps before, during, and after treatment. Metabolic maps and CE-MRIs were subsequently correlated to histology and immunohistochemistry. RESULTS In vivo imaging of choline/n-acetyl aspartate ratios via multivoxel MRS is better able to evaluate response to therapy than CE-MRI. Lactate imaging revealed that diffuse infiltrative areas in glioblastoma xenografts did not present with excessive glycolysis. In contrast, glycolysis was observed in hypoxic areas in angiogenesis-dependent compact regions of glioma only, especially after anti-angiogenic treatment. CONCLUSION Our data present MRSI as a powerful and feasible approach that is superior to CE-MRI and may provide handles for optimizing treatment of glioma. Furthermore, we show that glycolysis is more prominent in hypoxic areas than in areas of diffuse infiltrative growth. The Warburg hypothesis of persisting glycolysis in tumors under normoxic conditions may thus not be valid for diffuse glioma.
Collapse
Affiliation(s)
- Bob Hamans
- Corresponding Author: William Leenders, PhD, Dept of Pathology, Radboud University Nijmegen Medical Centre, PO Box 9101, 6500 HB Nijmegen, the Netherlands.
| | | | | | | | | | | |
Collapse
|
17
|
Goggi JL, Bejot R, Moonshi SS, Bhakoo KK. Stratification of 18F-Labeled PET Imaging Agents for the Assessment of Antiangiogenic Therapy Responses in Tumors. J Nucl Med 2013; 54:1630-6. [DOI: 10.2967/jnumed.112.115824] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
|
18
|
Shen J, Zheng H, Ruan J, Fang W, Li A, Tian G, Niu X, Luo S, Zhao P. Autophagy inhibition induces enhanced proapoptotic effects of ZD6474 in glioblastoma. Br J Cancer 2013; 109:164-71. [PMID: 23799852 PMCID: PMC3708568 DOI: 10.1038/bjc.2013.306] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Revised: 05/21/2013] [Accepted: 05/24/2013] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Autophagy is a lysosomal degradation pathway that can provide energy through its recycling mechanism to act as a cytoprotective adaptive response mediating treatment resistance in cancer cells. We investigated the autophagy-inducing effects of ZD6474, a small-molecule inhibitor that blocks activities of vascular endothelial growth factor receptor (VEGFR), epidermal growth factor receptor (EGFR), and RET tyrosine kinases. METHODS We investigated the effects of ZD6474 on autophagy in glioblastomas cells. The ZD6474 mechanism of action was determined by western blot. We then examined the impacts of the inhibition of autophagy in combination with ZD6474 on cell apoptosis in vitro. Furthermore, we evaluated the synergistic anticancer activity of combination treatment with an autophagy inhibitor (chloroquine) and ZD6474 in U251 glioblastoma cells xenograft model. RESULTS ZD6474-induced autophagy was dependent on signalling through the phosphoinositide 3-kinase/Akt/mammalian target of rapamycin (PI3K/Akt/mTOR) pathway. ZD6474-induced autophagy was inhibited by both knockdown of the ATG7 and Beclin 1 gene, essential autophagy genes, and pharmacologic agents (chloroquine and 3-methyalanine) treatment. Both treatments also dramatically sensitised glioblastoma cells to ZD6474-induced apoptosis, decreasing cell viability in vitro. Furthermore, in a xenograft mouse model, combined treatment with ZD6474 and chloroquine significantly inhibited U251 tumour growth, and increased the numbers of apoptotic cells compared with treatment with either agent alone. CONCLUSION Autophagy protects glioblastoma cells from the proapoptotic effects of ZD6474, which might contribute to tumour resistance against ZD6474 treatment.
Collapse
Affiliation(s)
- J Shen
- Department of Cell Biology, Southern Medical University, Guangzhou, Gungdong, People's Republic of China
- Cancer Center, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - H Zheng
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - J Ruan
- Cancer Center, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - W Fang
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - A Li
- Cancer Center, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - G Tian
- Department of Cell Biology, Southern Medical University, Guangzhou, Gungdong, People's Republic of China
| | - X Niu
- Department of Cell Biology, Southern Medical University, Guangzhou, Gungdong, People's Republic of China
| | - S Luo
- Department of Cell Biology, Southern Medical University, Guangzhou, Gungdong, People's Republic of China
| | - P Zhao
- Cancer Center, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| |
Collapse
|
19
|
Navis AC, Niclou SP, Fack F, Stieber D, van Lith S, Verrijp K, Wright A, Stauber J, Tops B, Otte-Holler I, Wevers RA, van Rooij A, Pusch S, von Deimling A, Tigchelaar W, van Noorden CJF, Wesseling P, Leenders WPJ. Increased mitochondrial activity in a novel IDH1-R132H mutant human oligodendroglioma xenograft model: in situ detection of 2-HG and α-KG. Acta Neuropathol Commun 2013; 1:18. [PMID: 24252742 PMCID: PMC3893588 DOI: 10.1186/2051-5960-1-18] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 05/09/2013] [Indexed: 01/04/2023] Open
Abstract
Background Point mutations in genes encoding NADP+-dependent isocitrate dehydrogenases (especially IDH1) are common in lower grade diffuse gliomas and secondary glioblastomas and occur early during tumor development. The contribution of these mutations to gliomagenesis is not completely understood and research is hampered by the lack of relevant tumor models. We previously described the development of the patient-derived high-grade oligodendroglioma xenograft model E478 that carries the commonly occurring IDH1-R132H mutation. We here report on the analyses of E478 xenografts at the genetic, histologic and metabolic level. Results LC-MS and in situ mass spectrometric imaging by LESA-nano ESI-FTICR revealed high levels of the proposed oncometabolite D-2-hydroxyglutarate (D-2HG), the product of enzymatic conversion of α-ketoglutarate (α-KG) by IDH1-R132H, in the tumor but not in surrounding brain parenchyma. α-KG levels and total NADP+-dependent IDH activity were similar in IDH1-mutant and -wildtype xenografts, demonstrating that IDH1-mutated cancer cells maintain α-KG levels. Interestingly, IDH1-mutant tumor cells in vivo present with high densities of mitochondria and increased levels of mitochondrial activity as compared to IDH1-wildtype xenografts. It is not yet clear whether this altered mitochondrial activity is a driver or a consequence of tumorigenesis. Conclusions The oligodendroglioma model presented here is a valuable model for further functional elucidation of the effects of IDH1 mutations on tumor metabolism and may aid in the rational development of novel therapeutic strategies for the large subgroup of gliomas carrying IDH1 mutations.
Collapse
|
20
|
In Vitro and In Vivo Analysis of RTK Inhibitor Efficacy and Identification of Its Novel Targets in Glioblastomas. Transl Oncol 2013; 6:187-96. [PMID: 23544171 DOI: 10.1593/tlo.12400] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Revised: 01/07/2013] [Accepted: 01/08/2013] [Indexed: 01/31/2023] Open
Abstract
Treatment for glioblastoma consists of radiotherapy and temozolomide-based chemotherapy. However, virtually all patients recur, leading to a fatal outcome. Receptor tyrosine kinase (RTK)-targeted therapy has been the focus of attention in novel treatment options for these patients. Here, we compared the efficacy of imatinib, sunitinib, and cediranib in glioblastoma models. In the present work, the biologic effect of the drugs was screened by viability, cell cycle, apoptosis, migration, and invasion in vitro assays or in vivo by chick chorioallantoic membrane assay. Intracellular signaling was assessed by Western blot and the RTK targets were identified using phospho-RTK arrays. The amplified status of KIT, PDGFRA, and VEGFR2 genes was assessed by quantitative polymerase chain reaction. In a panel of 10 glioblastoma cell lines, we showed that cediranib was the most potent. In addition, cediranib and sunitinib synergistically sensitize the cells to temozolomide. Cediranib efficacy was shown to associate with higher cytostatic and unique cytotoxic effects in vitro and both antitumoral and antiangiogenic activity in vivo, which could associate with its great capacity to inhibit mitogen-activated protein kinase (MAPK) and AKT pathways. The molecular status of KIT, PDGFRA, and VEGFR2 did not predict glioblastoma cell responsiveness to any of the RTK inhibitors. Importantly, phospho-RTK arrays revealed novel targets for cediranib and sunitinib therapy. In conclusion, the novel targets found may be of value as future biomarkers for therapy response in glioblastoma and lead to the rational selection of patients for effective molecular targeted treatment.
Collapse
|
21
|
Effects of dual targeting of tumor cells and stroma in human glioblastoma xenografts with a tyrosine kinase inhibitor against c-MET and VEGFR2. PLoS One 2013; 8:e58262. [PMID: 23484006 PMCID: PMC3587584 DOI: 10.1371/journal.pone.0058262] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Accepted: 01/31/2013] [Indexed: 12/20/2022] Open
Abstract
Anti-angiogenic treatment of glioblastoma with Vascular Endothelial Growth Factor (VEGF)- or VEGF Receptor 2 (VEGFR2) inhibitors normalizes tumor vessels, resulting in a profound radiologic response and improved quality of life. This approach however does not halt tumor progression by diffuse infiltration, as this phenotype is less angiogenesis dependent. Combined inhibition of angiogenesis and diffuse infiltrative growth would therefore be a more effective treatment approach in these tumors. The HGF/c-MET axis is important in both angiogenesis and cell migration in several tumor types including glioma. We therefore analyzed the effects of the c-MET- and VEGFR2 tyrosine kinase inhibitor cabozantinib (XL184, Exelixis) on c-MET positive orthotopic E98 glioblastoma xenografts, which routinely present with angiogenesis-dependent areas of tumor growth, as well as diffuse infiltrative growth. In in vitro cultures of E98 cells, cabozantinib effectively inhibited c-MET phosphorylation, concomitant with inhibitory effects on AKT and ERK1/2 phosphorylation, and cell proliferation and migration. VEGFR2 activation in endothelial cells was also effectively inhibited in vitro. Treatment of BALB/c nu/nu mice carrying orthotopic E98 xenografts resulted in a significant increase in overall survival. Cabozantinib effectively inhibited angiogenesis, resulting in increased hypoxia in angiogenesis-dependent tumor areas, and induced vessel normalization. Yet, tumors ultimately escaped cabozantinib therapy by diffuse infiltrative outgrowth via vessel co-option. Of importance, in contrast to the results from in vitro experiments, in vivo blockade of c-MET activation was incomplete, possibly due to multiple factors including restoration of the blood-brain barrier resulting from cabozantinib-induced VEGFR2 inhibition. In conclusion, cabozantinib is a promising therapy for c-MET positive glioma, but improving delivery of the drug to the tumor and/or the surrounding tissue may be needed for full activity.
Collapse
|
22
|
Antiangiogenic therapy in the management of brain tumors: a clinical overview. Cancer Chemother Pharmacol 2012; 70:353-63. [DOI: 10.1007/s00280-012-1926-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2012] [Accepted: 07/05/2012] [Indexed: 12/15/2022]
|
23
|
González-Arenas A, Hansberg-Pastor V, Hernández-Hernández OT, González-García TK, Henderson-Villalpando J, Lemus-Hernández D, Cruz-Barrios A, Rivas-Suárez M, Camacho-Arroyo I. Estradiol increases cell growth in human astrocytoma cell lines through ERα activation and its interaction with SRC-1 and SRC-3 coactivators. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2012; 1823:379-86. [DOI: 10.1016/j.bbamcr.2011.11.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2011] [Revised: 11/10/2011] [Accepted: 11/11/2011] [Indexed: 02/07/2023]
|
24
|
Current World Literature. Curr Opin Nephrol Hypertens 2012; 21:106-18. [DOI: 10.1097/mnh.0b013e32834ee42b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|