1
|
Wu J, Liang F, Wei R, Lai S, Lv X, Luo S, Wu Z, Chen H, Zhang W, Zeng X, Ye X, Wu Y, Wei X, Jiang X, Zhen X, Yang R. A Multiparametric MR-Based RadioFusionOmics Model with Robust Capabilities of Differentiating Glioblastoma Multiforme from Solitary Brain Metastasis. Cancers (Basel) 2021; 13:cancers13225793. [PMID: 34830943 PMCID: PMC8616314 DOI: 10.3390/cancers13225793] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 11/13/2021] [Accepted: 11/13/2021] [Indexed: 11/24/2022] Open
Abstract
Simple Summary Glioblastoma multiforme (GBM) and solitary brain metastasis (SBM) are common brain tumors in adults. The two tumors often pose a diagnostic dilemma owing to their similar features on conventional magnetic resonance imaging (MRI). Ability to discriminate the two tumors is critical as it informs clinical treatment strategies. This pilot study attempts to employ the machine learning technique to identify GBM and SBM by fusing radiomics features of multiple MRI sequences and multiple models. A multiparametric MR-based RadioFusionOmics (RFO) model was developed and has demonstrated promising prediction accuracy for the identifications of GBM and SBM. Abstract This study aimed to evaluate the diagnostic potential of a novel RFO model in differentiating GBM and SBM with multiparametric MR sequences collected from 244 (131 GBM and 113 SBM) patients. Three basic volume of interests (VOIs) were delineated on the conventional axial MR images (T1WI, T2WI, T2_FLAIR, and CE_T1WI), including volumetric non-enhanced tumor (nET), enhanced tumor (ET), and peritumoral edema (pTE). Using the RFO model, radiomics features extracted from different multiparametric MRI sequence(s) and VOI(s) were fused and the best sequence and VOI, or possible combinations, were determined. A multi-disciplinary team (MDT)-like fusion was performed to integrate predictions from the high-performing models for the final discrimination of GBM vs. SBM. Image features extracted from the volumetric ET (VOIET) had dominant predictive performances over features from other VOI combinations. Fusion of VOIET features from the T1WI and T2_FLAIR sequences via the RFO model achieved a discrimination accuracy of AUC = 0.925, accuracy = 0.855, sensitivity = 0.856, and specificity = 0.853, on the independent testing cohort 1, and AUC = 0.859, accuracy = 0.836, sensitivity = 0.708, and specificity = 0.919 on the independent testing cohort 2, which significantly outperformed three experienced radiologists (p = 0.03, 0.01, 0.02, and 0.01, and p = 0.02, 0.01, 0.45, and 0.02, respectively) and the MDT-decision result of three experienced experts (p = 0.03, 0.02, 0.03, and 0.02, and p = 0.03, 0.02, 0.44, and 0.03, respectively).
Collapse
Affiliation(s)
- Jialiang Wu
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China; (J.W.); (R.W.); (S.L.); (Z.W.); (H.C.); (W.Z.); (X.W.); (X.J.)
- Department of Radiology, The University of Hong Kong Shenzhen Hospital, Shenzhen 518000, China
| | - Fangrong Liang
- School of Biomedical Engineering, Southern Medical University, Guangzhou 510515, China;
| | - Ruili Wei
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China; (J.W.); (R.W.); (S.L.); (Z.W.); (H.C.); (W.Z.); (X.W.); (X.J.)
| | - Shengsheng Lai
- School of Medical Equipment, Guangdong Food and Drug Vocational College, Guangzhou 510520, China;
| | - Xiaofei Lv
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China;
- Department of Medical Imaging, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Shiwei Luo
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China; (J.W.); (R.W.); (S.L.); (Z.W.); (H.C.); (W.Z.); (X.W.); (X.J.)
| | - Zhe Wu
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China; (J.W.); (R.W.); (S.L.); (Z.W.); (H.C.); (W.Z.); (X.W.); (X.J.)
| | - Huixian Chen
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China; (J.W.); (R.W.); (S.L.); (Z.W.); (H.C.); (W.Z.); (X.W.); (X.J.)
| | - Wanli Zhang
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China; (J.W.); (R.W.); (S.L.); (Z.W.); (H.C.); (W.Z.); (X.W.); (X.J.)
| | - Xiangling Zeng
- Department of Radiology, Huizhou Municipal Central Hospital, Huizhou 516001, China;
| | - Xianghua Ye
- Department of Radiation Oncology, 1st Affiliated Hospital, Zhejiang University, Hangzhou 310009, China;
| | - Yong Wu
- Department of Oncology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China;
| | - Xinhua Wei
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China; (J.W.); (R.W.); (S.L.); (Z.W.); (H.C.); (W.Z.); (X.W.); (X.J.)
| | - Xinqing Jiang
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China; (J.W.); (R.W.); (S.L.); (Z.W.); (H.C.); (W.Z.); (X.W.); (X.J.)
| | - Xin Zhen
- School of Biomedical Engineering, Southern Medical University, Guangzhou 510515, China;
- Correspondence: (X.Z.); (R.Y.); Tel.: +86-20-62789323 (X.Z.); +86-20-81048873 (R.Y.)
| | - Ruimeng Yang
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China; (J.W.); (R.W.); (S.L.); (Z.W.); (H.C.); (W.Z.); (X.W.); (X.J.)
- Correspondence: (X.Z.); (R.Y.); Tel.: +86-20-62789323 (X.Z.); +86-20-81048873 (R.Y.)
| |
Collapse
|
2
|
Evaluating survival in subjects with astrocytic brain tumors by dynamic susceptibility-weighted perfusion MR imaging. PLoS One 2021; 16:e0244275. [PMID: 33406116 PMCID: PMC7787526 DOI: 10.1371/journal.pone.0244275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 12/07/2020] [Indexed: 12/02/2022] Open
Abstract
Purpose Studies have evaluated the application of perfusion MR for predicting survival in patients with astrocytic brain tumors, but few of them statistically adjust their results to reflect the impact of the variability of treatment administered in the patients. Our aim was to analyze the association between the perfusion values and overall survival time, with adjustment for various clinical factors, including initial treatments and follow-up treatments. Materials and methods This study consisted of 51 patients with astrocytic brain tumors who underwent perfusion-weighted MRI with MultiHance® at a dose of 0.1 mmol/kg prior to initial surgery. We measured the mean rCBV, the 5% & 10% maximum rCBV, and the variation of rCBV in the tumors. Comparisons were made between patients with and without 2-year survival using two-sample t-test or Wilcoxon rank-sum test for the continuous data, or chi-square and Fisher exact tests for categorical data. The multivariate cox-proportional hazard regression was fit to evaluate the association between rCBV and overall survival time, with adjustment for clinical factors. Results Patients who survived less than 2 years after diagnosis had a higher mean and maximum rCBV and a larger variation of rCBV. After adjusting for clinical factors including therapeutic measures, we found no significant association of overall survival time within 2 years with any of these rCBV values. Conclusions Although patients who survived less than 2 years had a higher mean and maximum rCBV and a larger variation of rCBV, rCBV itself may not be used independently for predicting 2-year survival of patients with astrocytic brain tumors.
Collapse
|
3
|
Zhao X, Eyo UB, Murguan M, Wu LJ. Microglial interactions with the neurovascular system in physiology and pathology. Dev Neurobiol 2018; 78:604-617. [PMID: 29318762 PMCID: PMC5980686 DOI: 10.1002/dneu.22576] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 01/01/2018] [Accepted: 01/06/2018] [Indexed: 01/11/2023]
Abstract
Microglia as immune cells of the central nervous system (CNS) play significant roles not only in pathology but also in physiology, such as shaping of the CNS during development and its proper maintenance in maturity. Emerging research is showing a close association between microglia and the neurovasculature that is critical for brain energy supply. In this review, we summarize the current literature on microglial interaction with the vascular system in the normal and diseased brain. First, we highlight data that indicate interesting potential involvement of microglia in developmental angiogenesis. Then we discuss the evidence for microglial participation with the vasculature in neuropathologies from brain tumors to acute injuries such as ischemic stroke to chronic neurodegenerative conditions. We conclude by suggesting future areas of research to advance the field in light of current technical progress and outstanding questions. © 2018 Wiley Periodicals, Inc. Develop Neurobiol 78: 604-617, 2018.
Collapse
Affiliation(s)
- Xiaoliang Zhao
- Department of Neurology, Mayo Clinic, Rochester, MN 55905
| | - Ukpong B. Eyo
- Department of Neurology, Mayo Clinic, Rochester, MN 55905
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854
| | - Madhuvika Murguan
- Department of Neurology, Mayo Clinic, Rochester, MN 55905
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN 55905
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854
| |
Collapse
|
4
|
Dudvarski Stankovic N, Teodorczyk M, Ploen R, Zipp F, Schmidt MHH. Microglia-blood vessel interactions: a double-edged sword in brain pathologies. Acta Neuropathol 2016; 131:347-63. [PMID: 26711460 DOI: 10.1007/s00401-015-1524-y] [Citation(s) in RCA: 194] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 11/09/2015] [Accepted: 12/12/2015] [Indexed: 12/12/2022]
Abstract
Microglia are long-living resident immune cells of the brain, which secure a stable chemical and physical microenvironment necessary for the proper functioning of the central nervous system (CNS). These highly dynamic cells continuously scan their environment for pathogens and possess the ability to react to damage-induced signals in order to protect the brain. Microglia, together with endothelial cells (ECs), pericytes and astrocytes, form the functional blood-brain barrier (BBB), a specialized endothelial structure that selectively separates the sensitive brain parenchyma from blood circulation. Microglia are in bidirectional and permanent communication with ECs and their perivascular localization enables them to survey the influx of blood-borne components into the CNS. Furthermore, they may stimulate the opening of the BBB, extravasation of leukocytes and angiogenesis. However, microglia functioning requires tight control as their dysregulation is implicated in the initiation and progression of numerous neurological diseases. Disruption of the BBB, changes in blood flow, introduction of pathogens in the sensitive CNS niche, insufficient nutrient supply, and abnormal secretion of cytokines or expression of endothelial receptors are reported to prime and attract microglia. Such reactive microglia have been reported to even escalate the damage of the brain parenchyma as is the case in ischemic injuries, brain tumors, multiple sclerosis, Alzheimer's and Parkinson's disease. In this review, we present the current state of the art of the causes and mechanisms of pathological interactions between microglia and blood vessels and explore the possibilities of targeting those dysfunctional interactions for the development of future therapeutics.
Collapse
Affiliation(s)
- Nevenka Dudvarski Stankovic
- Molecular Signal Transduction Laboratories, Institute for Microscopic Anatomy and Neurobiology, Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn²), University Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131, Mainz, Germany.
- German Cancer Consortium (DKTK), Heidelberg, Germany.
- German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Marcin Teodorczyk
- Molecular Signal Transduction Laboratories, Institute for Microscopic Anatomy and Neurobiology, Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn²), University Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131, Mainz, Germany.
| | - Robert Ploen
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Research Center for Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn²), University Medical Center of the Johannes Gutenberg University, Mainz, Germany.
| | - Frauke Zipp
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Research Center for Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn²), University Medical Center of the Johannes Gutenberg University, Mainz, Germany.
| | - Mirko H H Schmidt
- Molecular Signal Transduction Laboratories, Institute for Microscopic Anatomy and Neurobiology, Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn²), University Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131, Mainz, Germany.
- German Cancer Consortium (DKTK), Heidelberg, Germany.
- German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
5
|
Sun XR, Patel NV, Danish SF. Tissue Ablation Dynamics During Magnetic Resonance-Guided, Laser-Induced Thermal Therapy. Neurosurgery 2016; 77:51-8; discussion 58. [PMID: 26086908 DOI: 10.1227/neu.0000000000000732] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Magnetic resonance-guided, laser-induced thermal therapy is a real-time magnetic resonance thermometry-guided, minimally invasive procedure used in the treatment of intracranial tumors, epilepsy, and pain. Little is known about its dynamics and the effects of various pathologies on overall ablation. OBJECTIVE To determine the relationship between thermal energy delivery and the time to maximal estimated thermal damage and whether differences exist between various intracranial pathologies. METHODS We used real-time ablation data from 28 patients across 5 unique intracranial pathologies. All ablations were performed using the Visualase Thermal Therapy System (Medtronic, Inc, Minneapolis, Minnesota), which uses a 980-nm diffusing tip diode laser. The thermal damage area was plotted against time for each ablation. We then estimated the duration of time required to reach 50% (t50) and 97% (t97) of maximal damage. Comparisons were then made between different intracranial pathologies. RESULTS The duration required to reach maximal thermal damage estimate (TDE) among all ablations was 159 ± 62 seconds, and the t50 and t97 were 43 ± 21 and 136 ± 57 seconds, respectively, where t97 was reached at an average of 23 seconds before the maximal TDE. The t97 was shorter in the recurrent metastasis/radiation necrosis and epilepsy groups compared with the previously untreated glioblastoma multiforme group. CONCLUSION The optimal duration can be estimated by the t97, which can be achieved in less than 3 minutes and differs across ablation targets. TDE expansion decelerates with prolonged ablation. Future studies are needed to examine the radiographic and clinical outcomes as well as the effects of ablation power, irrigation speed, and the effect of previous therapies on ablation dynamics.
Collapse
Affiliation(s)
- Xiaonan R Sun
- *Division of Neurosurgery, Rutgers University, Robert Wood Johnson Medical School, New Brunswick, New Jersey; ‡Department of Neurological Surgery, Rutgers University, New Jersey Medical School, Newark, New Jersey
| | | | | |
Collapse
|
6
|
Transmembrane-4-L-six-family-1, a potential predictor for poor prognosis, overexpressed in human glioma. Neuroreport 2016; 26:455-61. [PMID: 25855954 DOI: 10.1097/wnr.0000000000000370] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Transmembrane-4-L-six-family-1 (TM4SF1), a tumor-associated antigen, is expressed in various human epithelial malignancies including breast, ovarian, lung, and colon carcinomas. The aim of the present study was to measure TM4SF1 gene expression in human glioma tissues and to investigate its relationship with patient outcome. We measured TM4SF1 expression in tumor tissue from 72 patients with glioma and in eight control brain tissues by means of quantitative reverse transcription-PCR, western blotting, and immunohistochemistry. The survival data including age, sex, Karnofsky performance scores, epilepsy, size of tumor, extent of resection, pathological grade, and TM4SF1 expression were analyzed using Kaplan-Meier analysis and the multivariate test method (Cox's proportional hazards model). We observed a higher level of TM4SF1 expression in human glioma tissues than in control brain tissues. Furthermore, TM4SF1 expression increased with ascending tumor grade (rs=0.950, P<0.05). Kaplan-Meier analysis with the log-rank test indicated that high TM4SF1 expression had a significant negative impact on overall survival (P<0.001). Moreover, multivariate Cox regression analysis revealed that TM4SF1 was an independent prognostic marker in glioma patients. These findings indicate that (a) TM4SF1 is overexpressed in human gliomas in general and (b) the precise level of expression might predict outcome and could be of clinical value.
Collapse
|
7
|
Andre JB, Nagpal S, Hippe DS, Ravanpay AC, Schmiedeskamp H, Bammer R, Palagallo GJ, Recht L, Zaharchuk G. Cerebral Blood Flow Changes in Glioblastoma Patients Undergoing Bevacizumab Treatment Are Seen in Both Tumor and Normal Brain. Neuroradiol J 2015; 28:112-9. [PMID: 25923677 DOI: 10.1177/1971400915576641] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
UNLABELLED Bevacizumab (BEV) is increasingly used to treat recurrent glioblastoma (GBM) with some reported improvement in neurocognitive function despite potential neurotoxicities. We examined the effects of BEV on cerebral blood flow (CBF) within recurrent GBM tumor and in the contralateral middle cerebral artery (MCA) territory.Post-chemoradiation patients with histologically confirmed GBM were treated with BEV and underwent routine, serial tumor imaging with additional pseudocontinuous arterial spin labeling (pcASL) following informed consent. Circular regions-of-interest were placed on pcASL images directly over the recurrent tumor and in the contralateral MCA territory. CBF changes before and during BEV treatment were evaluated in tumor and normal tissue. Linear mixed models were used to assess statistical significance.Fifty-three pcASL studies in 18 patients were acquired. Evaluation yielded lower mean tumoral CBF during BEV treatment compared with pre-treatment (45 ± 27 vs. 65 ± 27 ml/100 g/min, p = 0.002), and in the contralateral MCA territory during, compared with pre-BEV treatment (35 ± 8.4 vs. 41 ± 8.4 ml/100 g/min, p = 0.03). The decrease in mean CBF tended to be greater in the tumoral region than in the contralateral MCA, though the difference did not reach statistical significance (31% vs. 13%; p = 0.082). CONCLUSIONS BEV administration results in statistically significant global CBF decrease with a potentially preferential decrease in tumor perfusion compared with normal brain tissue.
Collapse
Affiliation(s)
- Jalal B Andre
- Department of Radiology, University of Washington; Seattle, WA, USA Department of Radiology, Stanford University, Stanford, CA, USA
| | - Seema Nagpal
- Department of Neurology and Neurological Sciences, Stanford University; Stanford, CA, USA
| | - Daniel S Hippe
- Department of Radiology, University of Washington; Seattle, WA, USA
| | - Ali C Ravanpay
- Department of Neurological Surgery, University of Washington; Seattle, WA, USA
| | | | - Roland Bammer
- Department of Radiology, Stanford University, Stanford, CA, USA
| | | | - Lawrence Recht
- Department of Neurological Surgery, University of Washington; Seattle, WA, USA
| | - Greg Zaharchuk
- Department of Radiology, Stanford University, Stanford, CA, USA
| |
Collapse
|
8
|
Wang J, Lei Y, Xie C, Lu W, Yan Z, Gao J, Xie Z, Zhang X, Liu M. Targeted gene delivery to glioblastoma using a C-end rule RGERPPR peptide-functionalised polyethylenimine complex. Int J Pharm 2013; 458:48-56. [DOI: 10.1016/j.ijpharm.2013.10.017] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Accepted: 10/03/2013] [Indexed: 01/13/2023]
|
9
|
Virág J, Kenessey I, Haberler C, Piurkó V, Bálint K, Döme B, Tímár J, Garami M, Hegedűs B. Angiogenesis and angiogenic tyrosine kinase receptor expression in pediatric brain tumors. Pathol Oncol Res 2013; 20:417-26. [PMID: 24190638 DOI: 10.1007/s12253-013-9711-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 10/10/2013] [Indexed: 01/07/2023]
Abstract
Tumor angiogenesis and receptor tyrosine kinases (RTK) are major novel targets in anticancer molecular therapy. Accordingly, we characterized the vascular network and the expression pattern of angiogenic RTK in the most frequent pediatric brain tumors. In a retrospective collection of 44 cases (14 astrocytoma, 16 ependymoma and 14 medulloblastoma), immunohistochemistry for VEGFR1, VEGFR2, PDGFRα, PDGFRβ, and c-Kit as well as microvessel labeling with CD34 and SMA were conducted on surgical specimens. We found a significantly higher vascular density in ependymoma. Glomeruloid formations were abundant in medulloblastoma but rare or almost absent in astrocytoma and ependymoma, respectively. C-Kit and VEGFR2 labeled blood vessels were more abundant in ependymoma than in the other two types of tumors. In contrast, medulloblastoma contained higher number of PDGFRα expressing vessels. In tumor cells, we found no VEGFR2 but VEGFR1 expression in all three tumor types. PDGFRα was strongly expressed on the tumor cells in all three malignancies, while PDGFRβ tumor cell expression was present in the majority of medulloblastoma cases. Interestingly, small populations of c-Kit expressing cancer cells were found in a number of medulloblastoma and ependymoma cases. Our study suggests that different angiogenic mechanisms are present in ependymoma and medulloblastoma. Furthermore ependymoma patients may benefit from anti-angiogenic therapies based on the high vascularization as well as the endothelial expression of c-kit and VEGFR2. The expression pattern of the receptors on tumor cells also suggests the targeting of specific angiogenic tyrosine kinase receptors may have direct antitumor activity. Further preclinical and biomarker driven clinical investigations are needed to establish the application of tyrosine kinase inhibitors in the treatment of pediatric brain tumors.
Collapse
Affiliation(s)
- József Virág
- 2nd Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Onishi M, Kurozumi K, Ichikawa T, Date I. Mechanisms of tumor development and anti-angiogenic therapy in glioblastoma multiforme. Neurol Med Chir (Tokyo) 2013; 53:755-63. [PMID: 24162241 PMCID: PMC4508716 DOI: 10.2176/nmc.ra2013-0200] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Despite advances in surgical and medical therapy, glioblastoma multiforme (GBM) remains a fatal disease. There has been no significant increase in survival for patients with this disease over the last 20 years. Tumor vasculature formation and glioma cell invasion along the white matter tracts both play a pivotal role in glioma development. Angiogenesis and invasion are the major factors believed to be responsible for treatment resistance in tumors, and a better understanding of the glioma invasion and angiogenesis mechanisms will lead to the development of potential new treatments. In this review, we focus on the molecular characteristics of angiogenesis and invasion in human malignant glioma. We discuss bevacizumab and cilengitide, which are used to inhibit angiogenesis in GBM.
Collapse
Affiliation(s)
- Manabu Onishi
- Department of Neurological Surgery, Okayama, University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Okayama
- Address reprint requests to: Manabu Onishi, MD, PhD, Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Okayama, Okayama 700-8558, Japan. e-mail:
| | - Kazuhiko Kurozumi
- Department of Neurological Surgery, Okayama, University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Okayama
| | - Tomotsugu Ichikawa
- Department of Neurological Surgery, Okayama, University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Okayama
| | - Isao Date
- Department of Neurological Surgery, Okayama, University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Okayama
| |
Collapse
|
11
|
Veeravagu A, Hou LC, Hsu AR, Cai W, Greve JM, Chen X, Tse V. The temporal correlation of dynamic contrast-enhanced magnetic resonance imaging with tumor angiogenesis in a murine glioblastoma model. Neurol Res 2013; 30:952-9. [DOI: 10.1179/174313208x322761] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
12
|
Abstract
Angiogenesis, recruitment of new blood vessels, is an essential component of the metastatic pathway. These vessels provide the principal route by which tumor cells exit the primary tumor site and enter the circulation. For many tumors, the vascular density can provide a prognostic indicator of metastatic potential, with the highly vascular primary tumors having a higher incidence of metastasis than poorly vascular tumors. The discovery and characterization of tumor-derived angiogenesis modulators greatly contributed to our understanding of how tumors regulate angiogenesis. However, although angiogenesis appears to be a rate-limiting event in tumor growth and metastatic dissemination, a direct connection between the induction of angiogenesis and the progression to tumor malignancy is less well understood. In this review, we discuss the observations concerning the modulation of angiogenesis and their implications in various neurological disorders, as well as their potential impact on cancer therapy.
Collapse
Affiliation(s)
- Göksemin Acar
- Department of Neurology, Faculty of Medicine, Pamukkale University, Denizli, Turkey.
| | | | | |
Collapse
|
13
|
El Sayed SM, El-Magd RMA, Shishido Y, Yorita K, Chung SP, Tran DH, Sakai T, Watanabe H, Kagami S, Fukui K. D-Amino acid oxidase-induced oxidative stress, 3-bromopyruvate and citrate inhibit angiogenesis, exhibiting potent anticancer effects. J Bioenerg Biomembr 2012; 44:513-23. [PMID: 22802136 DOI: 10.1007/s10863-012-9455-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Accepted: 05/29/2012] [Indexed: 11/30/2022]
Abstract
Angiogenesis is critical for cancer growth and metastasis. Steps of angiogenesis are energy consuming, while vascular endothelial cells are highly glycolytic. Glioblastoma multiforme (GBM) is a highly vascular tumor and this enhances its aggressiveness. D-amino acid oxidase (DAO) is a promising therapeutic protein that induces oxidative stress upon acting on its substrates. Oxidative stress-energy depletion (OSED) therapy was recently reported (El Sayed et al., Cancer Gene Ther, 19, 1-18, 2012). OSED combines DAO-induced oxidative stress with energy depletion caused by glycolytic inhibitors such as 3-bromopyruvate (3BP), a hexokinase II inhibitor that depleted ATP in cancer cells and induced production of hydrogen peroxide. 3BP disturbs the Warburg effect and antagonizes effects of lactate and pyruvate (El Sayed et al., J Bioenerg Biomembr, 44, 61-79, 2012). Citrate is a natural organic acid capable of inhibiting glycolysis by targeting phosphofructokinase. Here, we report that DAO, 3BP and citrate significantly inhibited angiogenesis, decreased the number of vascular branching points and shortened the length of vascular tubules. OSED delayed the growth of C6/DAO glioma cells. 3BP combined with citrate delayed the growth of C6 glioma cells and decreased significantly the number and size of C6 glioma colonies in soft agar. Human GBM cells (U373MG) were resistant to chemotherapy e.g. cisplatin and cytosine arabinoside, while 3BP was effective in decreasing the viability and disturbing the morphology of U373MG cells.
Collapse
Affiliation(s)
- S M El Sayed
- Division of Enzyme Pathophysiology, The Institute for Enzyme Research (KOSOKEN), The University of Tokushima, Kuramoto-cho, Tokushima, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Jiang G, Zhang L, Xin Y, Pei DS, Wei ZP, Liu YQ, Zheng JN. Conditionally replicating adenoviruses carrying mda-7/IL-24 for cancer therapy. Acta Oncol 2012; 51:285-92. [PMID: 21995527 DOI: 10.3109/0284186x.2011.621447] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
BACKGROUND Melanoma differentiation associated gene-7/interleukin-24 (mda-7/IL-24) suppresses growth and induces apoptosis in a broad range of human cancers without significant cytotoxicity to normal cells. Conditionally replicating adenoviruses (CRAds) not only have the ability to destroy cancer cells but may also be potential vectors for the expression of therapeutic genes. METHODS This review provides an overview of specifications for a novel anti-tumor approach CRAds carrying IL-24, and discusses recent progress in this field. RESULTS Studies in multiple laboratories report that CRAds carrying IL-24 selectively induced apoptosis in some cancer cells, and enhanced selective toxicity to cancer cells when combined with chemotherapeutic agents. CONCLUSION CRAds carrying IL-24 may prove a novel and effective approach for the treatment of cancers.
Collapse
Affiliation(s)
- Guan Jiang
- Laboratory of Biological Cancer Therapy, Xuzhou Medical College, Xuzhou, China
| | | | | | | | | | | | | |
Collapse
|
15
|
Ullrich NJ, Zimmerman M, Smith E, Irons M, Marcus K, Kieran MW. Association of rapidly progressive moyamoya syndrome with bevacizumab treatment for glioblastoma in a child with neurofibromatosis type 1. J Child Neurol 2011; 26:228-30. [PMID: 20823033 DOI: 10.1177/0883073810379639] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Neurofibromatosis type 1 is a common multisystemic disorder that can result in tumors of the central and peripheral nervous system. Individuals with neurofibromatosis type 1 are also at increased risk to develop moyamoya syndrome, which is a cerebrovascular condition that predisposes affected individuals to develop strokes as a result of progressive narrowing of the intracranial internal carotid arteries and failure of adequate blood supply through collateral vessels. We report a case of a young boy with neurofibromatosis type 1 with glioblastoma who developed rapidly progressive moyamoya vasculopathy after treatment with the angiogenesis inhibitor bevacizumab.
Collapse
Affiliation(s)
- Nicole J Ullrich
- Department of Neurology, Children's Hospital Boston, Boston, MA 02446, USA.
| | | | | | | | | | | |
Collapse
|
16
|
Onishi M, Ichikawa T, Kurozumi K, Date I. Angiogenesis and invasion in glioma. Brain Tumor Pathol 2011; 28:13-24. [PMID: 21221826 DOI: 10.1007/s10014-010-0007-z] [Citation(s) in RCA: 189] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2010] [Accepted: 09/22/2010] [Indexed: 10/18/2022]
Abstract
Despite advances in surgical and medical therapy, glioblastoma consistently remains a fatal disease. Over the last 20 years, no significant increase in survival has been achieved for patients with this disease. The formation of abnormal tumor vasculature and glioma cell invasion along white matter tracts are believed to be the major factors responsible for the resistance of these tumors to treatment. Therefore, investigation of angiogenesis and invasion in glioblastoma is essential for the development of a curative therapy. In our report, we first reviewed certain histopathological studies that focus on angiogenesis and invasion of human malignant gliomas. Second, we considered several animal models of glioma available for studying angiogenesis and invasion, including our novel animal models. Third, we focused on the molecular aspects of glioma angiogenesis and invasion, and the key mediators of these processes. Finally, we discussed the recent and ongoing clinical trials targeting tumor angiogenesis and invasion in glioma patients. A better understanding of the mechanism of glioma angiogenesis and invasion will lead to the development of new treatment methods.
Collapse
Affiliation(s)
- Manabu Onishi
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1, Shikata-cho, Kita-ku, Okayama 700-8558, Japan.
| | | | | | | |
Collapse
|
17
|
Toktas ZO, Akgun E, Ozkan A, Bozkurt SU, Bekiroglu N, Seker A, Konya D, Kilic T. Relationship of Angiogenic Potential With Clinical Features in Cranial Meningiomas: A Corneal Angiogenesis Study. Neurosurgery 2010; 67:1724-32; discussion 1732. [DOI: 10.1227/neu.0b013e3181f9f310] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
18
|
Kawai H, Yamashita T, Ohta Y, Deguchi K, Nagotani S, Zhang X, Ikeda Y, Matsuura T, Abe K. Tridermal tumorigenesis of induced pluripotent stem cells transplanted in ischemic brain. J Cereb Blood Flow Metab 2010; 30:1487-93. [PMID: 20216552 PMCID: PMC2949240 DOI: 10.1038/jcbfm.2010.32] [Citation(s) in RCA: 145] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Stroke is a major neurologic disorder. Induced pluripotent stem (iPS) cells can be produced from basically any part of patients, with high reproduction ability and pluripotency to differentiate into various types of cells, suggesting that iPS cells can provide a hopeful therapy for cell transplantation. However, transplantation of iPS cells into ischemic brain has not been reported. In this study, we showed that the iPS cells fate in a mouse model of transient middle cerebral artery occlusion (MCAO). Undifferentiated iPS cells (5 x 10(5)) were transplanted into ipsilateral striatum and cortex at 24 h after 30 mins of transient MCAO. Behavioral and histologic analyses were performed at 28 day after the cell transplantation. To our surprise, the transplanted iPS cells expanded and formed much larger tumors in mice postischemic brain than in sham-operated brain. The clinical recovery of the MCAO+iPS group was delayed as compared with the MCAO+PBS (phosphate-buffered saline) group. iPS cells formed tridermal teratoma, but could supply a great number of Dcx-positive neuroblasts and a few mature neurons in the ischemic lesion. iPS cells have a promising potential to provide neural cells after ischemic brain injury, if tumorigenesis is properly controlled.
Collapse
Affiliation(s)
- Hiromi Kawai
- Department of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Mustafa D, van der Weiden M, Zheng P, Nigg A, Luider TM, Kros JM. Expression sites of colligin 2 in glioma blood vessels. Brain Pathol 2009; 20:50-65. [PMID: 19067716 PMCID: PMC2805918 DOI: 10.1111/j.1750-3639.2008.00248.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
In a previous study using state-of-the-art proteomic techniques, we identified colligin 2 (HSP47) as a glioma blood vessel-specific protein. In the present study we precisely localized the expression of colligin 2 in the blood vessels of diffusely infiltrating gliomas and relate the expression to the distinct cellular components of the vessels by using multiple immunolabeling and confocal microscopy. We grouped the glioma blood vessels into morphological categories ranging from normal looking capillaries to vessels with hypertrophic and sclerotic changes. The expression patterns of various markers of endothelial and pericytic differentiation were correlated with the position of the cells in the vessels and the expression of colligin 2. We found that colligin 2 is expressed in all categories of glioma blood vessels in cells with endothelial and pericytic lineage. Expression of colligin 2 was also found in cells scattered around blood vessels and in few glial fibrillary acidic protein-positive cells within the blood vessels. There is overlap in the expression of colligin 2 and the collagens type I and IV for which colligin 2 is a chaperon. We conclude that colligin 2 is expressed in all cellular components of glioma blood vessels and may serve as a general marker for active angiogenesis.
Collapse
Affiliation(s)
- Dana Mustafa
- Department of Pathology and Laboratory of Neuro-oncology and Clinical Proteomics, Erasmus Medical Center, Dr. Molewaterplein 50, 3015 GD Rotterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
20
|
Zhang X, Zheng X, Jiang F, Zhang ZG, Katakowski M, Chopp M. Dual-color fluorescence imaging in a nude mouse orthotopic glioma model. J Neurosci Methods 2009; 181:178-85. [PMID: 19447136 DOI: 10.1016/j.jneumeth.2009.05.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2009] [Revised: 04/30/2009] [Accepted: 05/03/2009] [Indexed: 11/30/2022]
Abstract
We sought to establish a new orthotopic glioma model of nude mice by transfer of DsRed2, a red fluorescent protein gene, to malignant glioma cells and to perfuse the tissue with fluorescein isothiocyanate (FITC) dextran in vivo, which would permit the concurrent detection of brain tumor invasion and angiogenesis in vivo by florescence microscopy. 9L or U87 malignant glioma cells with DsRed2 expression were intracerebrally injected into the nude mice. FITC-dextran was administered intravenously to the mice bearing DsRed2-9L or DsRed2-U87 cells immediately before they were sacrificed at 10 days or 15 days after the implantation, respectively. Coronal vibratome sections were examined using 2D and 3D fluorescence microscopy and the results were compared with those examined by routine hematoxylin and eosin (H & E) staining. Angiogenesis induced by glioma was confirmed by two-dimensional and three-dimensional imaging analysis. DsRed2 fluorescence clearly demarcated the primary tumor margins and readily allowed for the visualization of local invasion at the single-cell level in the brain adjacent to tumor. We found that a few tumor cells migrated from the tumor mass along the aberrant microvasculature, but did not extend out of the angiogenic areas. However, locally invasive foci were very difficult to detect by H & E staining. We demonstrated, for the first time, that abnormal vascular structure and glioma cells can be visualized concurrently by fluorescence microscopy. This method is superior to H & E staining for the detection and study of physiologically relevant patterns of brain tumor invasion and angiogenesis in vivo.
Collapse
Affiliation(s)
- Xuepeng Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, MI 48202, USA
| | | | | | | | | | | |
Collapse
|
21
|
CRAdRGDflt-IL24 virotherapy in combination with chemotherapy of experimental glioma. Cancer Gene Ther 2009; 16:794-805. [PMID: 19363468 DOI: 10.1038/cgt.2009.23] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Malignant forms of glioma, the most common primary brain tumors, remain poorly responsive to multimodality therapeutic interventions, including chemotherapy. Suppressed apoptosis and extraordinary invasiveness are important distinctive features that contribute to the malignant phenotype of glioma. We have developed the vascular endothelial growth factor receptor 1 (VEGFR-1/flt-1) conditional replicating adenoviral vector (CRAdRGDflt-IL24) encoding the interleukin-24 (IL-24) gene. We investigated whether a combination of CRAdRGDflt-IL24-mediated oncolytic virotherapy and chemotherapy using temozolomide (TMZ) produces increased cytotoxicity against human glioma cells in comparison with these agents alone. Combination of CRAdRGDflt-IL24 and TMZ significantly enhanced cytotoxicity in vitro, inhibited D54MG tumor growth and prolonged survival of mice harboring intracranial human glioma xenografts in comparison with CRAdRGDflt-IL24 or TMZ alone. These data indicate that combined treatment with CRAdRGDflt-IL24-mediated oncolytic virotherapy and TMZ chemotherapy provides a promising approach for glioma therapy.
Collapse
|
22
|
Brown MC, Staniszewska I, Lazarovici P, Tuszynski GP, Del Valle L, Marcinkiewicz C. Regulatory effect of nerve growth factor in alpha9beta1 integrin-dependent progression of glioblastoma. Neuro Oncol 2009; 10:968-80. [PMID: 19074980 DOI: 10.1215/15228517-2008-047] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
In the present study we described the role of alpha9beta1 integrin in glioblastoma progression following its interaction with nerve growth factor (NGF). The level of expression of alpha9beta1 on astrocytomas is correlated with increased grade of this brain tumor and is highest on glioblastoma, whereas normal astrocytes do not express this integrin. Two glioblastoma cell lines, LN229 and LN18, that are alpha9beta1 integrin positive and negative, respectively, were used for alpha9beta1 integrin-dependent NGF-induced tumor progression. NGF was a significant promoter of promigratory and pro-proliferative activities of glioblastoma cells through direct interaction with alpha9beta1 integrin and activation of MAPK Erk1/2 pathway. The level of NGF increases approximately threefold in the most malignant glioma tissue when compared with normal brain. This increase is related to secretion of NGF by tumor cells. Specific inhibitors of alpha9beta1 integrin or gene silencing inhibited NGF-induced proliferation of LN229 cell line to the level shown by LN18 cells. VLO5 promoted alpha9beta1-dependent programmed cell death by induction of intrinsic apoptosis pathway in cancer cells. LN229 cells were rescued from proapoptotic effect of VLO5 by the presence of NGF. This disintegrin significantly inhibited tumor growth induced by implantation of LN229 cells to the chorioallantoic membrane (CAM) of quail embryonic model, and this inhibitory effect was significantly abolished by the presence of NGF. alpha9beta1 integrin appears to be an interesting target for blocking the progression of malignant gliomas, especially in light of the stimulatory effect of NGF on the development of these tumors and its ability to transfer proapoptotic signals in cancer cells.
Collapse
Affiliation(s)
- Meghan C Brown
- Department of Neuroscience, Center for Neurovirology and Cancer Biology, School of Medicine, Temple University, Philadelphia, PA, USA
| | | | | | | | | | | |
Collapse
|
23
|
Matiasek L, Platt S, Adams V, Scase T, Keys D, Miller J, Adamo F, Long S, Matiasek K. Ki-67 and Vascular Endothelial Growth Factor Expression in Intracranial Meningiomas in Dogs. J Vet Intern Med 2009; 23:146-51. [DOI: 10.1111/j.1939-1676.2008.0235.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
24
|
Brown MC, Staniszewska I, Lazarovici P, Tuszynski GP, Del Valle L, Marcinkiewicz C. Regulatory effect of nerve growth factor in α9β1 integrin–dependent progression of glioblastoma. Neuro Oncol 2008. [DOI: 10.1215/15228517-2008-0047] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
|
25
|
Yunker CK, Golembieski W, Lemke N, Schultz CR, Cazacu S, Brodie C, Rempel SA. SPARC-induced increase in glioma matrix and decrease in vascularity are associated with reduced VEGF expression and secretion. Int J Cancer 2008; 122:2735-43. [PMID: 18350569 PMCID: PMC3644882 DOI: 10.1002/ijc.23450] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2007] [Accepted: 12/27/2007] [Indexed: 11/23/2022]
Abstract
Glioblastomas are heterogeneous tumors displaying regions of necrosis, proliferation, angiogenesis, apoptosis and invasion. SPARC, a matricellular protein that negatively regulates angiogenesis and cell proliferation, but enhances cell deadhesion from matrix, is upregulated in gliomas (Grades II-IV). We previously demonstrated that SPARC promotes invasion while concomitantly decreasing tumor growth, in part by decreasing proliferation of the tumor cells. In other cancer types, SPARC has been shown to influence tumor growth by altering matrix production, and by decreasing angiogenesis via interfering with the VEGF-VEGFR1 signaling pathway. We therefore examined whether the SPARC-induced decrease in glioma tumor growth was also, in part, due to alterations in matrix and/or decreased vascularity, and assessed SPARC-VEGF interactions. The data demonstrate that SPARC upregulates glioma matrix, collagen I is a constituent of the matrix and SPARC promotes collagen fibrillogenesis. Furthermore, SPARC suppressed glioma vascularity, and this was accompanied by decreased VEGF expression and secretion, which was, in part, due to reduced VEGF165 transcript abundance. These data indicate that SPARC modulates glioma growth by altering the tumor microenvironment and by suppressing tumor vascularity through suppression of VEGF expression and secretion. These experiments implicate a novel mechanism, whereby SPARC regulates VEGF function by limiting the available growth factor. Because SPARC is considered to be a therapeutic target for gliomas, a further understanding of its complex signaling mechanisms is important, as targeting SPARC to decrease invasion could undesirably lead to the growth of more vascular and proliferative tumors.
Collapse
Affiliation(s)
- Christopher K Yunker
- Barbara Jane Levy Laboratory of Molecular Neuro-Oncology, Hermelin Brain Tumor Center, Department of Neurosurgery, Henry Ford HospitalDetroit, MI
| | - William Golembieski
- Barbara Jane Levy Laboratory of Molecular Neuro-Oncology, Hermelin Brain Tumor Center, Department of Neurosurgery, Henry Ford HospitalDetroit, MI
| | - Nancy Lemke
- Barbara Jane Levy Laboratory of Molecular Neuro-Oncology, Hermelin Brain Tumor Center, Department of Neurosurgery, Henry Ford HospitalDetroit, MI
| | - Chad R Schultz
- Barbara Jane Levy Laboratory of Molecular Neuro-Oncology, Hermelin Brain Tumor Center, Department of Neurosurgery, Henry Ford HospitalDetroit, MI
| | - Simona Cazacu
- William and Karen Davidson Laboratory of Cell Signaling and Tumorigenesis, Hermelin Brain Tumor Center, Department of Neurosurgery, Henry Ford HospitalDetroit, MI
| | - Chaya Brodie
- William and Karen Davidson Laboratory of Cell Signaling and Tumorigenesis, Hermelin Brain Tumor Center, Department of Neurosurgery, Henry Ford HospitalDetroit, MI
| | - Sandra A Rempel
- Barbara Jane Levy Laboratory of Molecular Neuro-Oncology, Hermelin Brain Tumor Center, Department of Neurosurgery, Henry Ford HospitalDetroit, MI
| |
Collapse
|
26
|
|
27
|
Fukushima Y, Tamura M, Nakagawa H, Itoh K. Induction of glioma cell migration by vitronectin in human serum and cerebrospinal fluid. J Neurosurg 2007; 107:578-85. [PMID: 17886558 DOI: 10.3171/jns-07/09/0578] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT Malignant gliomas are often highly invasive and can migrate along blood vessels. The purpose of the current study was to identify the substance in human serum and/or cerebrospinal fluid (CSF) that promotes glioma cell migration. METHODS The authors used a Boyden chamber cell migration assay to study the effect of serum from patients with glioma and healthy volunteers on chemotaxis of A172 human glioma cells. Heat inactivation, trypsinization, and ultrafiltration of serum were used to establish the nature of the active factor. Vitronectin and fibronectin were chosen for further investigations; chemotactic effects were studied in both serum and CSF. RESULTS Serum from both patients with glioma and healthy volunteers was found to promote chemotaxis of human glioma cells. This activity was greatly reduced by heat inactivation or trypsinization. Fractionation of the serum by ultrafiltration through membranes with various pore sizes showed that the active molecule was larger than 50 kD. Antibodies against integrin alphav or alphavbeta5 or arginine-glycine-aspartic acid-containing peptides, both of which block the vitronectin-glioma cell interactions, significantly reduced serum-induced cell migration, whereas blocking the interaction of glioma cells with fibronectin had no effect. Furthermore, the ability of serum to promote the migration of A 172 or T98G glioma cells was suppressed by immunodepletion of vitronectin and restored by the addition of exogenous vitronectin. The migration of glioma cells induced by CSF collected from the postoperative cavity of a malignant glioma patient was also reduced by blocking the interaction of glioma cells with vitronectin. CONCLUSIONS These results suggest that vitronectin is one of the major factors in serum- and CSF-induced glioma cell migration.
Collapse
Affiliation(s)
- Yuji Fukushima
- Department of Biology, Osaka Medical Center for Cancer and Cardiovascular Diseases, Osaka, Japan
| | | | | | | |
Collapse
|
28
|
Lightfoot NJ, Lucas PG, Finnis NDM. Disseminated haemangioblastoma without evidence of the von Hippel-Lindau syndrome or haemangioblastomatosis--A case report and clinico-pathological correlation. Clin Neurol Neurosurg 2007; 109:305-10. [PMID: 17250956 DOI: 10.1016/j.clineuro.2006.12.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2006] [Revised: 12/16/2006] [Accepted: 12/18/2006] [Indexed: 10/23/2022]
Abstract
A 71-year-old Caucasian female presented to our service with disseminated cranial and spinal haemangioblastomata but no other features to suggest the von Hippel-Lindau syndrome. We feel that this represents cellular dissemination through the cerebro-spinal fluid and may be an intermediate step to the development of frank haemangioblastomatosis. By comparing this presentation to that of other tumours we have suggested a potential pathological mechanism and have discussed its management.
Collapse
Affiliation(s)
- Nicholas J Lightfoot
- Department of Neurosurgery, Christchurch Hospital, Private Bag 4710, Christchurch, New Zealand.
| | | | | |
Collapse
|
29
|
Invernici G, Ponti D, Corsini E, Cristini S, Frigerio S, Colombo A, Parati E, Alessandri G. Human microvascular endothelial cells from different fetal organs demonstrate organ-specific CAM expression. Exp Cell Res 2005; 308:273-82. [PMID: 15936757 DOI: 10.1016/j.yexcr.2005.04.033] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2005] [Revised: 04/27/2005] [Accepted: 04/28/2005] [Indexed: 10/25/2022]
Abstract
In this work, we isolated and produced long-term cultures of human fetal endothelial cells (fECs) deriving from different organs of the same 12-week-old embryos. Highly pure endothelium cultures were obtained from specimens of brain, heart, lung, liver, aorta and kidney by using magnetic microspheres coated with CD31 or CD34 specific endothelial antibodies. The endothelial nature of these cells was confirmed by the presence of von Willebrand Factor (vWf), Flk-1/VEGFR2 and CD31. The fECs cultures showed organ-specific differences as regards to the morphological appearance, the growth rate and the expression of cellular adhesion molecules (CAMs) before or after stimulation by the inflammatory cytokines IL-1beta and TNF-alpha. For instance, TNF-alpha showed a specific effect on fetal heart ECs by stimulating E-selectin expression. Our findings indicate that fECs may represent an innovative tool to study differences among ECs of different vascular districts of the same individual, thus increasing the possibility to compare many pathological aspects of human adult and fetal microvasculature.
Collapse
Affiliation(s)
- G Invernici
- SSD Neurobiology and Neuroregenerative Therapies, Carlo Besta Neurological Institute, Milan, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Jiang F, Zhang Z, Kalkanis S, Katakowksi M, Robin AM, Zhang X, Gotlib A, Chelst I, Mikkelsen T, Chopp M. A Quantitative Model of Tumor-induced Angiogenesis in the Nude Mouse. Neurosurgery 2005; 57:320-4. [PMID: 16094162 DOI: 10.1227/01.neu.0000166665.43838.0d] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE Novel animal models allowing for the quantification of tumor-induced angiogenesis and cell migration may offer significant insight into the characterization and multidisciplinary treatment of brain tumors. In this study, we seek to establish such a model in tumor-bearing brain, allowing for a clear demarcation of primary and satellite tumor tissue in conjunction with precise quantification of cerebral microvasculature. METHODS We used green fluorescent protein-transfected 9L-gliosarcoma cells stereotactically injected into the brain parenchyma of nude mice perfused with tetramethylrhodamine-dextran immediately before they were killed. New three-dimensional analytical software developed in our laboratory provided a quantitative analysis of laser-scanning confocal microscopy images of dextran-labeled cerebral microvessels. RESULTS Our data confirm significant angiogenesis in tumor and brain adjacent to tumor. CONCLUSION Because these highly infiltrative malignant brain tumors interdigitate with normal brain parenchyma through finger-like projections at the periphery of the solid tumor boundary, therapeutic options targeting tumor blood flow--combined with novel three-dimensional imaging to localize and track such interventions--may offer new hope for glioma management. To our knowledge, this system represents the first animal brain tumor model allowing for the precise colocalization and quantification of angiogenesis and tumor cell invasion, which may play an important role in the development of future therapy for brain tumors.
Collapse
Affiliation(s)
- Feng Jiang
- Department of Neurology, Henry Ford Health Sciences Center, Detroit, Michigan, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Charalambous C, Hofman FM, Chen TC. Functional and phenotypic differences between glioblastoma multiforme-derived and normal human brain endothelial cells. J Neurosurg 2005; 102:699-705. [PMID: 15871513 DOI: 10.3171/jns.2005.102.4.0699] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
OBJECT Glioblastomas multiforme (GBMs) are hypervascular tumors characterized by endothelial cell (EC) proliferation. There is increasing evidence that ECs that infiltrate systemic tumors are different from normal blood vessel cells; whether this difference is seen in the central nervous system between GBM and normal brain tissue is not known. The goal of this investigation was to characterize and compare the functional and phenotypic properties of GBM-associated ECs and normal brain ECs. METHODS Human ECs were isolated from fresh tissue specimens, purified using flow cytometry, and characterized by immunostaining. Proliferation was measured by determining bromodeoxyuridine incorporation and Ki-67 staining, and by performing the monotetrazolium assay. The migration rate of the cells was determined using the modified Boyden chamber technique. Apoptosis was evaluated by performing the TUNEL assay, cell death enzyme-linked immunosorbent assay (ELISA), and annexin V staining. Growth factor production was analyzed using the ELISA technique. The brain tumor ECs differed from normal brain ECs morphologically and by their expression and distribution of specific markers (that is, vascular endothelial cadherin [VE-cadherin] and CD31). Functional differences between the two cell populations were also evident. The brain tumor ECs proliferated more slowly and underwent less apoptosis than normal brain ECs; however, the tumor ECs migrated faster than the normal ECs. The normal ECs were sensitive to growth factors such as vascular endothelial growth factor (VEGF) and endothelin-1 (ET-1), whereas the tumor ECs were not. In addition, the brain tumor ECs constitutively produced higher levels of ET-1 and VEGF, compared with the normal ECs. CONCLUSIONS The data demonstrated that ECs derived from normal brain and from GBMs have significant phenotypic and functional distinctions. Further characterization of brain tumor ECs is essential for efficient antiangiogenic treatment of gliomas.
Collapse
Affiliation(s)
- Christiana Charalambous
- Department of Molecular Microbiology, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | | | | |
Collapse
|
32
|
Harrer JU, Parker GJM, Haroon HA, Buckley DL, Embelton K, Roberts C, Balériaux D, Jackson A. Comparative study of methods for determining vascular permeability and blood volume in human gliomas. J Magn Reson Imaging 2004; 20:748-57. [PMID: 15503330 DOI: 10.1002/jmri.20182] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
PURPOSE To characterize human gliomas using T1-weighted dynamic contrast-enhanced MRI (DCE-MRI), and directly compare three pharmacokinetic analysis techniques: a conventional established technique and two novel techniques that aim to reduce erroneous overestimation of the volume transfer constant between plasma and the extravascular extracellular space (EES) (Ktrans) in areas of high blood volume. MATERIALS AND METHODS Eighteen patients with high-grade gliomas underwent DCE-MRI. Three kinetic models were applied to estimate Ktrans and fractional blood plasma volume (vp). We applied the Tofts and Kermode (TK) model without arterial input function (AIF) estimation, the TK model modified to include vp and AIF estimation (mTK), and a "first pass" variant of the TK model (FP). RESULTS KTK values were considerably higher than KmTK and KFP values (P <0.001). KmTK and KFP were more comparable and closely correlated (rho=0.744), with KmTK generally higher than KFP (P <0.001). Estimates of vp(mTK) and vp(FP) also showed a significant difference (P <0.001); however, these values were very closely correlated (rho=0.901). KTK parameter maps showed "pseudopermeability" effects displaying numerous vessels. These were not visualized on KmTK and KFP maps but appeared on the corresponding vp maps, indicating a failure of the TK model in commonly occurring vascular regions. CONCLUSION Both of the methods that incorporate a measured AIF and an estimate of vp provide similar pathophysiological information and avoid erroneous overestimation of Ktrans in areas of significant vessel density, and thus allow a more accurate estimation of endothelial permeability.
Collapse
Affiliation(s)
- Judith U Harrer
- Department of Neurology, Aachen University Hospital, Aachen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Yanamandra N, Gumidyala KV, Waldron KG, Gujrati M, Olivero WC, Dinh DH, Rao JS, Mohanam S. Blockade of cathepsin B expression in human glioblastoma cells is associated with suppression of angiogenesis. Oncogene 2004; 23:2224-30. [PMID: 14730346 DOI: 10.1038/sj.onc.1207338] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The cysteine proteinase cathepsin B has been implicated in tumor progression by virtue of its increased mRNA and protein levels, as well as its localization at the invading front of the tumor. In this study, we examined whether blocking cathepsin B expression in human glioblastoma SNB19 cells affects angiogenesis. Stable transfectants of human glioblastoma cells with a plasmid containing antisense cathepsin B cDNA showed decreased migration rates in wound- and spheroid-migration assays. Analysis showed a reduction in VEGF protein and MMP-9 activity in the cathepsin B antisense cDNA-transfected cells. Regarding angiogenesis in vitro, we found that the conditioned medium of glioblastoma cells with downregulated cathepsin B expression reduced cell-cell interaction of human microvascular endothelial cells, resulting in the disruption of capillary-like network formation. Furthermore, a marked reduction in microvasculature development was seen in an in vivo dorsal air sac assay of glioblastoma cells with downregulated cathepsin B expression. Taken together, these results provide evidence that inhibition of cathepsin B expression can suppress glioblastoma-induced neovascularization.
Collapse
Affiliation(s)
- Niranjan Yanamandra
- Program of Cancer Biology, University of Illinois College of Medicine at Peoria, Peoria, One Illini Drive, Box 1649, IL 61656, USA
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Régina A, Jodoin J, Khoueir P, Rolland Y, Berthelet F, Moumdjian R, Fenart L, Cecchelli R, Demeule M, Béliveau R. Down-regulation of caveolin-1 in glioma vasculature: modulation by radiotherapy. J Neurosci Res 2004; 75:291-299. [PMID: 14705150 DOI: 10.1002/jnr.10865] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Primary brain tumors, particularly glioblastomas (GB), remain a challenge for oncology. An element of the malignant brain tumors' aggressive behavior is the fact that GB are among the most densely vascularized tumors. To determine some of the molecular regulations occuring at the brain tumor endothelium level during tumoral progression would be an asset in understanding brain tumor biology. Caveolin-1 is an essential structural constituent of caveolae that has been implicated in mitogenic signaling, oncogenesis, and angiogenesis. In this work we investigated regulation of caveolin-1 expression in brain endothelial cells (ECs) under angiogenic conditions. In vitro, brain EC caveolin-1 is down-regulated by angiogenic factors treament and by hypoxia. Coculture of brain ECs with tumoral cells induced a similar down-regulation. In addition, activation of the p42/44 MAP kinase is demonstrated. By using an in vivo brain tumor model, we purified ECs from gliomas as well as from normal brain to investigate possible regulation of caveolin-1 expression in tumoral brain vasculature. We show that caveolin-1 expression is strikingly down-regulated in glioma ECs, whereas an increase of phosphorylated caveolin-1 is observed. Whole-brain radiation treatment, a classical way in which GB is currently being treated, resulted in increased caveolin-1 expression in tumor isolated ECs. The level of tumor cells spreading around newly formed blood vessels was also elevated. The regulation of caveolin-1 expression in tumoral ECs may reflect the tumoral vasculature state and correlates with angiogenesis kinetics.
Collapse
Affiliation(s)
- Anthony Régina
- Laboratoire de Médecine Moléculaire, Université du Québec à Montréal-Hôpital Ste.-Justine, Montréal, Québec, Canada
| | - Julie Jodoin
- Laboratoire de Médecine Moléculaire, Université du Québec à Montréal-Hôpital Ste.-Justine, Montréal, Québec, Canada
| | - Paul Khoueir
- Laboratoire de Médecine Moléculaire, Université du Québec à Montréal-Hôpital Ste.-Justine, Montréal, Québec, Canada
| | - Yannève Rolland
- Laboratoire de Médecine Moléculaire, Université du Québec à Montréal-Hôpital Ste.-Justine, Montréal, Québec, Canada
| | - France Berthelet
- Département de Pathologie, Hôpital Notre-Dame, Montréal, Québec, Canada
| | - Robert Moumdjian
- Département de Pathologie, Hôpital Notre-Dame, Montréal, Québec, Canada
| | - Laurence Fenart
- Laboratoire Mixte Institut Pasteur de Lille, Université d'Artois, Lens Cedex, France
| | - Romeo Cecchelli
- Laboratoire Mixte Institut Pasteur de Lille, Université d'Artois, Lens Cedex, France
| | - Michel Demeule
- Laboratoire de Médecine Moléculaire, Université du Québec à Montréal-Hôpital Ste.-Justine, Montréal, Québec, Canada
| | - Richard Béliveau
- Laboratoire de Médecine Moléculaire, Université du Québec à Montréal-Hôpital Ste.-Justine, Montréal, Québec, Canada
| |
Collapse
|
35
|
Kirk S, Frank JA, Karlik S. Angiogenesis in multiple sclerosis: is it good, bad or an epiphenomenon? J Neurol Sci 2004; 217:125-30. [PMID: 14706213 DOI: 10.1016/j.jns.2003.10.016] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Characteristic pathological features of multiple sclerosis (MS) include inflammation, demyelination and axonal and oligodendrocyte loss. In addition, lesions can also have a significant vascular component. In this review, morphological, biochemical and radiological evidence is presented suggesting angiogenesis as a potential focus for investigation in MS. We hypothesize that angiogenesis plays a significant role in the MS lesion, perpetuating disease progression. Thus, treatment strategies that inhibit angiogenesis may decrease clinical and pathological signs of disease. Several approaches for testing this hypothesis are outlined.
Collapse
Affiliation(s)
- Shauna Kirk
- Department of Pathology, University of Western Ontario, Ontario, London, Canada
| | | | | |
Collapse
|
36
|
Tsuneki H, Ito K, Sekizaki N, Ma EL, You Y, Kawakami J, Adachi I, Sasaoka T, Kimura I. Nicotinic Enhancement of Proliferation in Bovine and Porcine Cerebral Microvascular Endothelial Cells. Biol Pharm Bull 2004; 27:1951-6. [PMID: 15577211 DOI: 10.1248/bpb.27.1951] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Nicotinic acetylcholine receptors are found in microvascular endothelial cells. To reveal the functional role in cerebral angiogenic processes, we studied the nicotinic modulation of proliferation activity in cultured bovine and porcine cerebral microvascular endothelial cells. The proliferation activity was determined by an increase in the number of cells present in culture dishes. When the bovine cerebral endothelial cells at different passages were cultured in the presence of nicotine (10 nM), the proliferation activities were significantly increased in the cells at passage 1 and passage 3, but not at passage 4. Reverse transcriptase-polymerase chain reaction studies demonstrated that the expression of mRNAs coding for alpha3 nicotinic receptor subunit was significantly reduced in the bovine cerebral endothelial cells at passage 4, compared with that at passage 1. The proliferation of porcine cerebral endothelial cells (passage 1) was enhanced by acetylcholine (10 nM-100 microM) in the presence of atropine, a muscarinic antagonist, and this enhancing effect was inhibited by hexamethonium (100 microM, a nicotinic antagonist). The stimulation by acetylcholine (1 microM, with atropine) or nicotine (10 nM) induced the phosphorylation of a mitogen-activated protein (MAP) kinase (extracellular-signal regulated kinase: ERK) in the serum-starved endothelial cells. In the presence of PD98059 (2 microM, a MAP kinase kinase inhibitor) and atropine, acetylcholine (1 microM) failed to enhance the proliferation of porcine cerebral endothelial cells. These results demonstrate that nicotinic stimulation promotes the proliferation of bovine and porcine cerebral microvascular endothelial cells, at least in part, through the MAP kinase activation.
Collapse
Affiliation(s)
- Hiroshi Tsuneki
- Department of Clinical Pharmacology, Toyama Medical and Pharmaceutical University, Toyama, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|