1
|
Lim S, Kwak M, Kang J, Cesaire M, Tang K, Robey RW, Frye WJE, Karim B, Butcher D, Lizak MJ, Dalmage M, Foster B, Nuechterlein N, Eberhart C, Cimino PJ, Gottesman MM, Jackson S. Ibrutinib disrupts blood-tumor barrier integrity and prolongs survival in rodent glioma model. Acta Neuropathol Commun 2024; 12:56. [PMID: 38589905 PMCID: PMC11003129 DOI: 10.1186/s40478-024-01763-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 03/24/2024] [Indexed: 04/10/2024] Open
Abstract
In malignant glioma, cytotoxic drugs are often inhibited from accessing the tumor site due to the blood-tumor barrier (BTB). Ibrutinib, FDA-approved lymphoma agent, inhibits Bruton tyrosine kinase (BTK) and has previously been shown to independently impair aortic endothelial adhesion and increase rodent glioma model survival in combination with cytotoxic therapy. Yet additional research is required to understand ibrutinib's effect on BTB function. In this study, we detail baseline BTK expression in glioma cells and its surrounding vasculature, then measure endothelial junctional expression/function changes with varied ibrutinib doses in vitro. Rat glioma cells and rodent glioma models were treated with ibrutinib alone (1-10 µM and 25 mg/kg) and in combination with doxil (10-100 µM and 3 mg/kg) to assess additive effects on viability, drug concentrations, tumor volume, endothelial junctional expression and survival. We found that ibrutinib, in a dose-dependent manner, decreased brain endothelial cell-cell adhesion over 24 h, without affecting endothelial cell viability (p < 0.005). Expression of tight junction gene and protein expression was decreased maximally 4 h after administration, along with inhibition of efflux transporter, ABCB1, activity. We demonstrated an additive effect of ibrutinib with doxil on rat glioma cells, as seen by a significant reduction in cell viability (p < 0.001) and increased CNS doxil concentration in the brain (56 ng/mL doxil alone vs. 74.6 ng/mL combination, p < 0.05). Finally, Ibrutinib, combined with doxil, prolonged median survival in rodent glioma models (27 vs. 16 days, p < 0.0001) with brain imaging showing a - 53% versus - 75% volume change with doxil alone versus combination therapy (p < 0.05). These findings indicate ibrutinib's ability to increase brain endothelial permeability via junctional disruption and efflux inhibition, to increase BTB drug entry and prolong rodent glioma model survival. Our results motivate the need to identify other BTB modifiers, all with the intent of improving survival and reducing systemic toxicities.
Collapse
Affiliation(s)
- Sanghee Lim
- Develomental Therapeutics and Pharmacology Unit, Surgical Neurology Branch, National Institute of Neurologic Disorders and Stroke (NINDS), NIH, Building 10, Room 7D45, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Minhye Kwak
- Develomental Therapeutics and Pharmacology Unit, Surgical Neurology Branch, National Institute of Neurologic Disorders and Stroke (NINDS), NIH, Building 10, Room 7D45, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Jeonghan Kang
- Develomental Therapeutics and Pharmacology Unit, Surgical Neurology Branch, National Institute of Neurologic Disorders and Stroke (NINDS), NIH, Building 10, Room 7D45, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Melissa Cesaire
- Develomental Therapeutics and Pharmacology Unit, Surgical Neurology Branch, National Institute of Neurologic Disorders and Stroke (NINDS), NIH, Building 10, Room 7D45, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Kayen Tang
- Develomental Therapeutics and Pharmacology Unit, Surgical Neurology Branch, National Institute of Neurologic Disorders and Stroke (NINDS), NIH, Building 10, Room 7D45, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Robert W Robey
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, MD, 20892, USA
| | - William J E Frye
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, MD, 20892, USA
| | - Baktiar Karim
- Molecular Histopathology Laboratory, Frederick National Laboratory, Leidos Biomedical Research, Frederick, MD, 21702, USA
| | - Donna Butcher
- Molecular Histopathology Laboratory, Frederick National Laboratory, Leidos Biomedical Research, Frederick, MD, 21702, USA
| | - Martin J Lizak
- NIH MRI Research Facility and Mouse Imaging Facility, National Institute of Neurologic Disorders and Stroke (NINDS), NIH, Bethesda, MD, 20814, USA
| | - Mahalia Dalmage
- Develomental Therapeutics and Pharmacology Unit, Surgical Neurology Branch, National Institute of Neurologic Disorders and Stroke (NINDS), NIH, Building 10, Room 7D45, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Brandon Foster
- Develomental Therapeutics and Pharmacology Unit, Surgical Neurology Branch, National Institute of Neurologic Disorders and Stroke (NINDS), NIH, Building 10, Room 7D45, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Nicholas Nuechterlein
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Charles Eberhart
- Neuropathology Unit, Surgical Neurology Branch, National Institute of Neurologic Disorders and Stroke (NINDS), NIH, Bethesda, MD, 20892, USA
| | - Patrick J Cimino
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael M Gottesman
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, MD, 20892, USA
| | - Sadhana Jackson
- Develomental Therapeutics and Pharmacology Unit, Surgical Neurology Branch, National Institute of Neurologic Disorders and Stroke (NINDS), NIH, Building 10, Room 7D45, 10 Center Drive, Bethesda, MD, 20892, USA.
| |
Collapse
|
2
|
Pournajaf S, Afsordeh N, Pourgholami MH. In vivo C6 glioma models: an update and a guide toward a more effective preclinical evaluation of potential anti-glioblastoma drugs. Rev Neurosci 2024; 35:183-195. [PMID: 37651618 DOI: 10.1515/revneuro-2023-0067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 08/11/2023] [Indexed: 09/02/2023]
Abstract
Glioblastoma multiform (GBM) is the most common primary brain tumor with a poor prognosis and few therapeutic choices. In vivo, tumor models are useful for enhancing knowledge of underlying GBM pathology and developing more effective therapies/agents at the preclinical level, as they recapitulate human brain tumors. The C6 glioma cell line has been one of the most widely used cell lines in neuro-oncology research as they produce tumors that share the most similarities with human GBM regarding genetic, invasion, and expansion profiles and characteristics. This review provides an overview of the distinctive features and the different animal models produced by the C6 cell line. We also highlight specific applications of various C6 in vivo models according to the purpose of the study and offer some technical notes for more convenient/repeatable modeling. This work also includes novel findings discovered in our laboratory, which would further enhance the feasibility of the model in preclinical GBM investigations.
Collapse
Affiliation(s)
- Safura Pournajaf
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran 1411713116, Iran
| | - Nastaran Afsordeh
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran 1411713116, Iran
| | | |
Collapse
|
3
|
Slika H, Karimov Z, Alimonti P, Abou-Mrad T, De Fazio E, Alomari S, Tyler B. Preclinical Models and Technologies in Glioblastoma Research: Evolution, Current State, and Future Avenues. Int J Mol Sci 2023; 24:16316. [PMID: 38003507 PMCID: PMC10671665 DOI: 10.3390/ijms242216316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/07/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
Glioblastoma is the most common malignant primary central nervous system tumor and one of the most debilitating cancers. The prognosis of patients with glioblastoma remains poor, and the management of this tumor, both in its primary and recurrent forms, remains suboptimal. Despite the tremendous efforts that are being put forward by the research community to discover novel efficacious therapeutic agents and modalities, no major paradigm shifts have been established in the field in the last decade. However, this does not mirror the abundance of relevant findings and discoveries made in preclinical glioblastoma research. Hence, developing and utilizing appropriate preclinical models that faithfully recapitulate the characteristics and behavior of human glioblastoma is of utmost importance. Herein, we offer a holistic picture of the evolution of preclinical models of glioblastoma. We further elaborate on the commonly used in vitro and vivo models, delving into their development, favorable characteristics, shortcomings, and areas of potential improvement, which aids researchers in designing future experiments and utilizing the most suitable models. Additionally, this review explores progress in the fields of humanized and immunotolerant mouse models, genetically engineered animal models, 3D in vitro models, and microfluidics and highlights promising avenues for the future of preclinical glioblastoma research.
Collapse
Affiliation(s)
- Hasan Slika
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (H.S.); (Z.K.); (S.A.)
| | - Ziya Karimov
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (H.S.); (Z.K.); (S.A.)
- Faculty of Medicine, Ege University, 35100 Izmir, Turkey
| | - Paolo Alimonti
- School of Medicine, Vita-Salute San Raffaele University, 20132 Milan, Italy; (P.A.); (E.D.F.)
| | - Tatiana Abou-Mrad
- Faculty of Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon;
- Department of Neurosurgery, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Emerson De Fazio
- School of Medicine, Vita-Salute San Raffaele University, 20132 Milan, Italy; (P.A.); (E.D.F.)
| | - Safwan Alomari
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (H.S.); (Z.K.); (S.A.)
| | - Betty Tyler
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (H.S.); (Z.K.); (S.A.)
| |
Collapse
|
4
|
Wang S, Yan W, Kong L, Zuo S, Wu J, Zhu C, Huang H, He B, Dong J, Wei J. Oncolytic viruses engineered to enforce cholesterol efflux restore tumor-associated macrophage phagocytosis and anti-tumor immunity in glioblastoma. Nat Commun 2023; 14:4367. [PMID: 37474548 PMCID: PMC10359270 DOI: 10.1038/s41467-023-39683-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 06/23/2023] [Indexed: 07/22/2023] Open
Abstract
The codependency of cholesterol metabolism sustains the malignant progression of glioblastoma (GBM) and effective therapeutics remain scarce. In orthotopic GBM models in male mice, we identify that codependent cholesterol metabolism in tumors induces phagocytic dysfunction in monocyte-derived tumor-associated macrophages (TAMs), resulting in disease progression. Manipulating cholesterol efflux with apolipoprotein A1 (ApoA1), a cholesterol reverse transporter, restores TAM phagocytosis and reactivates TAM-T cell antitumor immunity. Cholesterol metabolomics analysis of in vivo-sorted TAMs further reveals that ApoA1 mediates lipid-related metabolic remodeling and lowers 7-ketocholesterol levels, which directly inhibits tumor necrosis factor signaling in TAMs through mitochondrial translation inhibition. An ApoA1-armed oncolytic adenovirus is also developed, which restores antitumor immunity and elicits long-term tumor-specific immune surveillance. Our findings provide insight into the mechanisms by which cholesterol metabolism impairs antitumor immunity in GBM and offer an immunometabolic approach to target cholesterol disturbances in GBM.
Collapse
Affiliation(s)
- Shiqun Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, Nanjing, Jiangsu, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Wei Yan
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hang Zhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Lingkai Kong
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Shuguang Zuo
- Liuzhou Key Laboratory of Molecular Diagnosis, Guangxi Key Laboratory of Molecular Diagnosis and Application, Affiliated Liutie Central Hospital of Guangxi Medical University, Liuzhou, Guangxi, China
| | - Jingyi Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Chunxiao Zhu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, UCAS, Hangzhou, Zhejiang, China
| | - Huaping Huang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hang Zhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Bohao He
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Jie Dong
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, Nanjing, Jiangsu, China.
| | - Jiwu Wei
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, Nanjing, Jiangsu, China.
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, China.
| |
Collapse
|
5
|
Lim J, Baig AA, Donnelly BM, Chaves LD, Pol SU, Koenigsknecht C, Pionessa D, Levy BR, Gutierrez L, Tutino VM, Levy EI, Siddiqui AH. The first endovascular rat glioma model for pre-clinical evaluation of intra-arterial therapeutics. Interv Neuroradiol 2023:15910199231169597. [PMID: 37157800 DOI: 10.1177/15910199231169597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2023] Open
Abstract
BACKGROUND Several translational animal models have been described assessing intra-arterial (IA) treatments for malignant gliomas. We describe the first endovascular animal model that allows testing of IA drug delivery as a first-line treatment, which is difficult to do in actual patients. We report a unique protocol for vascular access and IA delivery in the rat model that, unlike prior reports, does not require direct puncture and opening of proximal cerebrovasculature which carries risk of ischemia in the animal brain post-delivery. METHODS Wistar rats underwent left femoral artery catherization with a Balt Magic 1.2F catheter or Marathon Flow directed 1.5F Microcatheter with an Asahi Chikai 0.008 micro-guidewire which was navigated to the left internal carotid artery under x-ray. 25% mannitol was administered to test blood brain barrier breakdown (BBBB). Additional rats were implanted with C6 glioma cells in the left frontal lobe. C6 Glioma-Implanted Rats (C6GRs) were monitored for overall survival and tumor growth. Tumor volumes from MRI images were calculated utilizing 3D slicer. Additional rats underwent femoral artery catheterization with Bevacizumab, carboplatin, or irinotecan injected into the left internal carotid artery to test feasibility and safety. RESULTS A successful endovascular access and BBBB protocol was established. BBBB was confirmed with positive Evans blue staining. 10 rats were successfully implanted with C6 gliomas with confirmed growths on MRI. Overall survival was 19.75 ± 2.21 days. 5 rats were utilized for the development of our femoral catheterization protocol and BBBB testing. With regards to IA chemotherapy dosage testing, control rats tolerated targeted 10 mg/kg of bevascizumab, 2.4 mg/kg of carboplatin, and 15 mg/kg of irinotecan IA ICA injections without any complications. CONCLUSIONS We present the first endovascular IA rat glioma model that allows selective catheterization of the intracranial vasculature and assessment of IA therapies for gliomas without need for access and sacrifice of proximal cerebrovasculature.
Collapse
Affiliation(s)
- Jaims Lim
- Department of Neurosurgery, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
- Department of Neurosurgery, Gates Vascular Institute at Kaleida Health, Buffalo, NY, USA
| | - Ammad A Baig
- Department of Neurosurgery, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
- Department of Neurosurgery, Gates Vascular Institute at Kaleida Health, Buffalo, NY, USA
| | - Brianna M Donnelly
- Department of Neurosurgery, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
- Department of Neurosurgery, Gates Vascular Institute at Kaleida Health, Buffalo, NY, USA
| | - Lee D Chaves
- Department of Medicine, University of Kansas Medical Center, Kansas City, USA
| | - Suyog U Pol
- Canon Stroke and Vascular Research Center, University at Buffalo, Buffalo, NY, USA
| | - Carmon Koenigsknecht
- Canon Stroke and Vascular Research Center, University at Buffalo, Buffalo, NY, USA
| | - Donald Pionessa
- Canon Stroke and Vascular Research Center, University at Buffalo, Buffalo, NY, USA
| | - Bennett R Levy
- George Washington University School of Medicine, Washington, DC, USA
| | - Liza Gutierrez
- Canon Stroke and Vascular Research Center, University at Buffalo, Buffalo, NY, USA
| | - Vincent M Tutino
- Canon Stroke and Vascular Research Center, University at Buffalo, Buffalo, NY, USA
- Jacobs Institute, Buffalo, NY, USA
- Mechanical and Aerospace Engineering, University at Buffalo School of Engineering and Applied Sciences, Buffalo, NY, USA
| | - Elad I Levy
- Department of Neurosurgery, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
- Department of Neurosurgery, Gates Vascular Institute at Kaleida Health, Buffalo, NY, USA
- Canon Stroke and Vascular Research Center, University at Buffalo, Buffalo, NY, USA
- Jacobs Institute, Buffalo, NY, USA
| | - Adnan H Siddiqui
- Department of Neurosurgery, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
- Department of Neurosurgery, Gates Vascular Institute at Kaleida Health, Buffalo, NY, USA
- Canon Stroke and Vascular Research Center, University at Buffalo, Buffalo, NY, USA
- Jacobs Institute, Buffalo, NY, USA
| |
Collapse
|
6
|
Sahu U, Barth RF, Otani Y, McCormack R, Kaur B. Rat and Mouse Brain Tumor Models for Experimental Neuro-Oncology Research. J Neuropathol Exp Neurol 2022; 81:312-329. [PMID: 35446393 PMCID: PMC9113334 DOI: 10.1093/jnen/nlac021] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Rodent brain tumor models have been useful for developing effective therapies for glioblastomas (GBMs). In this review, we first discuss the 3 most commonly used rat brain tumor models, the C6, 9L, and F98 gliomas, which are all induced by repeated injections of nitrosourea to adult rats. The C6 glioma arose in an outbred Wistar rat and its potential to evoke an alloimmune response is a serious limitation. The 9L gliosarcoma arose in a Fischer rat and is strongly immunogenic, which must be taken into consideration when using it for therapy studies. The F98 glioma may be the best of the 3 but it does not fully recapitulate human GBMs because it is weakly immunogenic. Next, we discuss a number of mouse models. The first are human patient-derived xenograft gliomas in immunodeficient mice. These have failed to reproduce the tumor-host interactions and microenvironment of human GBMs. Genetically engineered mouse models recapitulate the molecular alterations of GBMs in an immunocompetent environment and “humanized” mouse models repopulate with human immune cells. While the latter are rarely isogenic, expensive to produce, and challenging to use, they represent an important advance. The advantages and limitations of each of these brain tumor models are discussed. This information will assist investigators in selecting the most appropriate model for the specific focus of their research.
Collapse
Affiliation(s)
- Upasana Sahu
- From the Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Rolf F Barth
- Department of Pathology, The Ohio State University, Columbus, Ohio, USA
| | - Yoshihiro Otani
- From the Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Ryan McCormack
- From the Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Balveen Kaur
- From the Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
7
|
Quader S, Kataoka K, Cabral H. Nanomedicine for brain cancer. Adv Drug Deliv Rev 2022; 182:114115. [PMID: 35077821 DOI: 10.1016/j.addr.2022.114115] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 12/18/2021] [Accepted: 01/12/2022] [Indexed: 02/06/2023]
Abstract
CNS tumors remain among the deadliest forms of cancer, resisting conventional and new treatment approaches, with mortality rates staying practically unchanged over the past 30 years. One of the primary hurdles for treating these cancers is delivering drugs to the brain tumor site in therapeutic concentration, evading the blood-brain (tumor) barrier (BBB/BBTB). Supramolecular nanomedicines (NMs) are increasingly demonstrating noteworthy prospects for addressing these challenges utilizing their unique characteristics, such as improving the bioavailability of the payloadsviacontrolled pharmacokinetics and pharmacodynamics, BBB/BBTB crossing functions, superior distribution in the brain tumor site, and tumor-specific drug activation profiles. Here, we review NM-based brain tumor targeting approaches to demonstrate their applicability and translation potential from different perspectives. To this end, we provide a general overview of brain tumor and their treatments, the incidence of the BBB and BBTB, and their role on NM targeting, as well as the potential of NMs for promoting superior therapeutic effects. Additionally, we discuss critical issues of NMs and their clinical trials, aiming to bolster the potential clinical applications of NMs in treating these life-threatening diseases.
Collapse
Affiliation(s)
- Sabina Quader
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 212-0821, Japan
| | - Kazunori Kataoka
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 212-0821, Japan.
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.
| |
Collapse
|
8
|
Preclinical models of glioblastoma: limitations of current models and the promise of new developments. Expert Rev Mol Med 2021; 23:e20. [PMID: 34852856 DOI: 10.1017/erm.2021.20] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Glioblastoma (GBM) is the most common and aggressive primary brain tumour, yet little progress has been made towards providing better treatment options for patients diagnosed with this devastating condition over the last few decades. The complex nature of the disease, heterogeneity, highly invasive potential of GBM tumours and until recently, reduced investment in research funding compared with other cancer types, are contributing factors to few advancements in disease management. Survival rates remain low with less than 5% of patients surviving 5 years. Another important contributing factor is the use of preclinical models that fail to fully recapitulate GBM pathophysiology, preventing efficient translation from the lab into successful therapies in the clinic. This review critically evaluates current preclinical GBM models, highlighting advantages and disadvantages of using such models, and outlines several emerging techniques in GBM modelling using animal-free approaches. These novel approaches to a highly complex disease such as GBM show evidence of a more truthful recapitulation of GBM pathobiology with high reproducibility. The resulting advancements in this field will offer new biological insights into GBM and its aetiology with potential to contribute towards the development of much needed improved treatments for GBM in future.
Collapse
|
9
|
Kútna V, O'Leary VB, Newman E, Hoschl C, Ovsepian SV. Revisiting Brain Tuberous Sclerosis Complex in Rat and Human: Shared Molecular and Cellular Pathology Leads to Distinct Neurophysiological and Behavioral Phenotypes. Neurotherapeutics 2021; 18:845-858. [PMID: 33398801 PMCID: PMC8423952 DOI: 10.1007/s13311-020-01000-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2020] [Indexed: 12/27/2022] Open
Abstract
Tuberous sclerosis complex (TSC) is a dominant autosomal genetic disorder caused by loss-of-function mutations in TSC1 and TSC2, which lead to constitutive activation of the mammalian target of rapamycin C1 (mTORC1) with its decoupling from regulatory inputs. Because mTORC1 integrates an array of molecular signals controlling protein synthesis and energy metabolism, its unrestrained activation inflates cell growth and division, resulting in the development of benign tumors in the brain and other organs. In humans, brain malformations typically manifest through a range of neuropsychiatric symptoms, among which mental retardation, intellectual disabilities with signs of autism, and refractory seizures, which are the most prominent. TSC in the rat brain presents the first-rate approximation of cellular and molecular pathology of the human brain, showing many instructive characteristics. Nevertheless, the developmental profile and distribution of lesions in the rat brain, with neurophysiological and behavioral manifestation, deviate considerably from humans, raising numerous research and translational questions. In this study, we revisit brain TSC in human and Eker rats to relate their histopathological, electrophysiological, and neurobehavioral characteristics. We discuss shared and distinct aspects of the pathology and consider factors contributing to phenotypic discrepancies. Given the shared genetic cause and molecular pathology, phenotypic deviations suggest an incomplete understanding of the disease. Narrowing the knowledge gap in the future should not only improve the characterization of the TSC rat model but also explain considerable variability in the clinical manifestation of the disease in humans.
Collapse
Affiliation(s)
- Viera Kútna
- Department of Experimental Neurobiology, National Institute of Mental Health, Topolová 748, 250 67, Klecany, Czech Republic.
| | - Valerie B O'Leary
- Department of Medical Genetics, Third Faculty of Medicine of Charles University, Ruská 87, 100 00, Prague, Czech Republic
| | - Ehren Newman
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Cyril Hoschl
- Department of Experimental Neurobiology, National Institute of Mental Health, Topolová 748, 250 67, Klecany, Czech Republic
- Department of Psychiatry and Medical Psychology, Third Faculty of Medicine of Charles University, Ruská 87, 100 00, Prague, Czech Republic
| | - Saak V Ovsepian
- Department of Experimental Neurobiology, National Institute of Mental Health, Topolová 748, 250 67, Klecany, Czech Republic.
- Department of Psychiatry and Medical Psychology, Third Faculty of Medicine of Charles University, Ruská 87, 100 00, Prague, Czech Republic.
| |
Collapse
|
10
|
Sanai N. Phase 0 Clinical Trial Strategies for the Neurosurgical Oncologist. Neurosurgery 2020; 85:E967-E974. [PMID: 31245813 PMCID: PMC6855937 DOI: 10.1093/neuros/nyz218] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 03/08/2019] [Indexed: 12/04/2022] Open
Abstract
In an era of escalating drug discovery costs, shifting priorities within the pharmaceutical industry, and longstanding challenges in central nervous system drug delivery, surgical trials offer an avenue to identify promising agents with demonstrable tumor penetration and molecular effects. The rise of pharmacodynamic- and pharmacokinetic-driven clinical trials, including phase 0 study designs, creates an opportunity for the neurosurgical oncologist to engage drug development for brain tumor patients directly. Here, we review the phase 0 clinical trial mechanism as well as its current and future applications within neurosurgical oncology.
Collapse
Affiliation(s)
- Nader Sanai
- Ivy Brain Tumor Center, Barrow Neurological Institute, Phoenix, Arizona
| |
Collapse
|
11
|
Beccaria K, Sabbagh A, de Groot J, Canney M, Carpentier A, Heimberger AB. Blood-brain barrier opening with low intensity pulsed ultrasound for immune modulation and immune therapeutic delivery to CNS tumors. J Neurooncol 2020; 151:65-73. [PMID: 32112296 DOI: 10.1007/s11060-020-03425-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 02/05/2020] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Opening of the blood-brain barrier (BBB) by pulsed low intensity ultrasound has been developed during the last decade and is now recognized as a safe technique to transiently and repeatedly open the BBB. This non- or minimally invasive technique allows for a targeted and uniform dispersal of a wide range of therapeutic substances throughout the brain, including immune cells and antibodies. METHODS In this review article, we summarize pre-clinical studies that have used BBB-opening by pulsed low intensity ultrasound to enhance the delivery of immune therapeutics and effector cell populations, as well as several recent clinical studies that have been initiated. Based on this analysis, we propose immune therapeutic strategies that are most likely to benefit from this strategy. The literature review and trial data research were performed using Medline/Pubmed databases and clinical trial registry www.clinicaltrials.gov . The reference lists of all included articles were searched for additional studies. RESULTS A wide range of immune therapeutic agents, including small molecular weight drugs, antibodies or NK cells, have been safely and efficiently delivered to the brain with pulsed low intensity ultrasound in preclinical models, and both tumor control and increased survival have been demonstrated in different types of brain tumor models in rodents. Ultrasound-induced BBB disruption may also stimulate innate and cellular immune responses. CONCLUSIONS Ultrasound BBB opening has just recently entered clinical trials with encouraging results, and the association of this strategy with immune therapeutics creates a new field of brain tumor treatment.
Collapse
Affiliation(s)
- Kevin Beccaria
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Aria Sabbagh
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - John de Groot
- Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Michael Canney
- CarThera, Institut du Cerveau Et de La Moelle épinière (ICM), 75013, Paris, France
| | - Alexandre Carpentier
- Department of Neurosurgery, Sorbonne Université, UPMC Univ Paris 06, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpitaux Universitaires La Pitié-Salpêtrière, Paris, France
| | - Amy B Heimberger
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Unit 422, P.O. Box 301402, Houston, TX, 77230-1402, USA.
| |
Collapse
|
12
|
Linsenmann T, Jawork A, Westermaier T, Homola G, Monoranu CM, Vince GH, Kessler AF, Ernestus RI, Löhr M. Tumor growth under rhGM-CSF application in an orthotopic rodent glioma model. Oncol Lett 2019; 17:4843-4850. [PMID: 31186691 PMCID: PMC6507467 DOI: 10.3892/ol.2019.10179] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 02/14/2019] [Indexed: 12/14/2022] Open
Abstract
Regulation of the host immune response serves a pivotal role in the persistence and progression of malignant glioma. To date, cytotoxic cluster of differentiation (CD)-8+ T and natural killer cells are considered the main cellular components of host tumor control. The influence of macrophages in an orthotropic C6 tumor implantation model was investigated and the aim of the present study was to characterize the effects of systemic macrophage-activation on glioma growth by using the granulocyte macrophage colony stimulating factor (rhGM-CSF). A total of 20 male Sprague-Dawley rats were orthotopically implanted with C6 glioma spheroids and treated subcutaneously with 10 µg/kg rhGM-CSF every other day; 9 animals served as controls. Serial magnetic resonance imaging was performed on days 7, 14, 21, 28, 32 and 42 post-implantation to monitor tumor volume. Histological work-up included hematoxylin and eosin, CD68/ED-1 macrophage, CD8 T-cell and Ki-67 MIB1 proliferation staining in gliomas and spleen. Experimental C6-gliomas developed in 15/20 (75%) animals. In rhGM-CSF treated rats, tumors developed significantly later and reached a smaller size (median, 134 mm3) compared with the controls (median, 262 mm3). On day 14, solid tumors presented in 11/17 (65%) rhGM-CSF-treated animals; in control animals tumor growth was detected in 3/9 animals on day 7 and in all animals on day 14. The mean survival time was 35 days in the rhGM-CSF group and significantly longer when compared with the control group (24 days). Immunohistochemistry exhibited significantly more macrophages in tumors, particularly in the perivascular zone of the rhGM-CSF group when compared with untreated animals; intratumoral CD8+ counts were equal in both groups. A systemic stimulation of macrophages by rhGM-CSF resulted in significantly reduced and delayed tumor growth in the rodent C6 glioma model. The present data suggested a significant role of macrophages in host control of experimental gliomas on the innate immune response. Until now, the role of macrophages may have been underestimated in host glioma control.
Collapse
Affiliation(s)
- Thomas Linsenmann
- Department of Neurosurgery, Julius Maximilians University, Wuerzburg, D-97080 Wuerzburg, Germany
| | - Anna Jawork
- Department of Neurosurgery, Julius Maximilians University, Wuerzburg, D-97080 Wuerzburg, Germany
| | - Thomas Westermaier
- Department of Neurosurgery, Julius Maximilians University, Wuerzburg, D-97080 Wuerzburg, Germany
| | - György Homola
- Department of Neuroradiology, Julius Maximilians University, Wuerzburg, D-97080 Wuerzburg, Germany
| | - Camelia Maria Monoranu
- Department of Neuropathology, Julius Maximilians University, Wuerzburg, D-97080 Wuerzburg, Germany
| | - Giles Hamilton Vince
- Department of Neurosurgery, Clinical Centre of Aschaffenburg-Alzenau, D-63739 Aschaffenburg, Germany
| | | | - Ralf-Ingo Ernestus
- Department of Neurosurgery, Julius Maximilians University, Wuerzburg, D-97080 Wuerzburg, Germany
| | - Mario Löhr
- Department of Neurosurgery, Julius Maximilians University, Wuerzburg, D-97080 Wuerzburg, Germany
| |
Collapse
|
13
|
Löhr M, Linsenmann T, Jawork A, Kessler AF, Timmermann N, Homola GA, Ernestus RI, Hagemann C. Implanting Glioblastoma Spheroids into Rat Brains and Monitoring Tumor Growth by MRI Volumetry. Methods Mol Biol 2017; 1622:149-159. [PMID: 28674808 DOI: 10.1007/978-1-4939-7108-4_12] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The outcome of patients suffering from glioblastoma multiforme (GBM) remains poor with a median survival of less than 15 months. To establish innovative therapeutical approaches or to analyze the effect of protein overexpression or protein knockdown by RNA interference in vivo, animal models are mandatory. Here, we describe the implantation of C6 glioma spheroids into the rats' brain and how to follow tumor growth by MRI scans. We show that C6 cells grown in Sprague-Dawley rats share several morphologic features of human glioblastoma like pleomorphic cells, areas of necrosis, vascular proliferation, and tumor cell invasion into the surrounding brain tissue. In addition, we describe a method for tumor volumetry utilizing the CISS 3D- or contrast-enhanced T1-weighted 3D sequence and freely available post-processing software.
Collapse
Affiliation(s)
- Mario Löhr
- Tumorbiology Laboratory, Department of Neurosurgery, University of Würzburg, Josef-Schneider-Str. 11, D-97080, Würzburg, Germany
| | - Thomas Linsenmann
- Tumorbiology Laboratory, Department of Neurosurgery, University of Würzburg, Josef-Schneider-Str. 11, D-97080, Würzburg, Germany
| | - Anna Jawork
- Tumorbiology Laboratory, Department of Neurosurgery, University of Würzburg, Josef-Schneider-Str. 11, D-97080, Würzburg, Germany
| | - Almuth F Kessler
- Tumorbiology Laboratory, Department of Neurosurgery, University of Würzburg, Josef-Schneider-Str. 11, D-97080, Würzburg, Germany
| | - Nils Timmermann
- Tumorbiology Laboratory, Department of Neurosurgery, University of Würzburg, Josef-Schneider-Str. 11, D-97080, Würzburg, Germany
| | - György A Homola
- Department of Neuroradiology, University of Würzburg, Josef-Schneider-Str. 11, D-97080, Würzburg, Germany
| | - Ralf-Ingo Ernestus
- Tumorbiology Laboratory, Department of Neurosurgery, University of Würzburg, Josef-Schneider-Str. 11, D-97080, Würzburg, Germany
| | - Carsten Hagemann
- Tumorbiology Laboratory, Department of Neurosurgery, University of Würzburg, Josef-Schneider-Str. 11, D-97080, Würzburg, Germany.
| |
Collapse
|
14
|
Spontaneous complete regression of hypothalamic pilocytic astrocytoma after partial resection in a child, complicated with Stevens-Johnson syndrome: a case report and literature review. Neurosurg Rev 2015; 39:335-40; discussion 340. [PMID: 26662551 DOI: 10.1007/s10143-015-0672-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 06/28/2015] [Accepted: 08/09/2015] [Indexed: 10/22/2022]
Abstract
Pilocytic astrocytoma (PA) is the most common pediatric central nervous system glial neoplasm and the most common pediatric cerebellar tumor. The spontaneous regression that occurs after partial/subtotal resection is multifactorial, depending on multiple factors, as for the case of humoral and cell-mediated immune responses of the host to the implanted tumor. A 7-year-old boy was referred to a neurosurgery clinic with headache. Further imaging workup revealed hypothalamic PA. Partial resection of the lesions was performed with right-side pterional approach. The patient developed a severe panmucositis [Stevens-Johnson syndrome (SJS)] and respiratory failure plus conjunctivitis, due to phenytoin allergy. During the patient's 6-month follow-up, postoperative magnetic resonance imaging (MRI) revealed a residual tumor, and about 9 months later (at 15 months postoperatively), the MRI showed total regression of the tumor. Clinically, symptomatic PA may undergo spontaneous regression after partial resection. We report a well-documented case of spontaneous regression hypothalamic PA after partial resection that complicated with SJS. Immune system reaction in SJS may have a role in tumor behavior and spontaneous regression. Multiple studies confirmed spontaneous regression in PA after partial/subtotal resection. This phenomenon occurs due to humoral and cell-mediated host immune responses to the implanted tumor. The immune system reaction in SJS may have a role in tumor behavior and spontaneous regression.
Collapse
|
15
|
Kegelman TP, Hu B, Emdad L, Das SK, Sarkar D, Fisher PB. In vivo modeling of malignant glioma: the road to effective therapy. Adv Cancer Res 2015; 121:261-330. [PMID: 24889534 DOI: 10.1016/b978-0-12-800249-0.00007-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Despite an increased emphasis on developing new therapies for malignant gliomas, they remain among the most intractable tumors faced today as they demonstrate a remarkable ability to evade current treatment strategies. Numerous candidate treatments fail at late stages, often after showing promising preclinical results. This disconnect highlights the continued need for improved animal models of glioma, which can be used to both screen potential targets and authentically recapitulate the human condition. This review examines recent developments in the animal modeling of glioma, from more established rat models to intriguing new systems using Drosophila and zebrafish that set the stage for higher throughput studies of potentially useful targets. It also addresses the versatility of mouse modeling using newly developed techniques recreating human protocols and sophisticated genetically engineered approaches that aim to characterize the biology of gliomagenesis. The use of these and future models will elucidate both new targets and effective combination therapies that will impact on disease management.
Collapse
Affiliation(s)
- Timothy P Kegelman
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Bin Hu
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Luni Emdad
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Swadesh K Das
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Devanand Sarkar
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Paul B Fisher
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA.
| |
Collapse
|
16
|
Khurram MA, Stamp S, Sheerin NS, Rix D, Cunningham AC, Carter N, Talbot D. Behaviour of transplanted tumours and role of matching in rejection. Transpl Immunol 2015; 32:121-5. [DOI: 10.1016/j.trim.2015.02.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Revised: 02/19/2015] [Accepted: 02/20/2015] [Indexed: 01/14/2023]
|
17
|
Zanotto-Filho A, Braganhol E, Klafke K, Figueiró F, Terra SR, Paludo FJ, Morrone M, Bristot IJ, Battastini AM, Forcelini CM, Bishop AJR, Gelain DP, Moreira JCF. Autophagy inhibition improves the efficacy of curcumin/temozolomide combination therapy in glioblastomas. Cancer Lett 2015; 358:220-231. [DOI: 10.1016/j.canlet.2014.12.044] [Citation(s) in RCA: 132] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 12/18/2014] [Accepted: 12/19/2014] [Indexed: 12/14/2022]
|
18
|
Yang L, Zhao J, Zhou G, Wang Y, Li L, Yuan H, Nan X, Guan L, Pei X. The 9L(LUC)/Wistar rat glioma model is not suitable for immunotherapy. Neural Regen Res 2015; 7:1406-11. [PMID: 25657674 PMCID: PMC4308791 DOI: 10.3969/j.issn.1673-5374.2012.18.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Accepted: 03/22/2012] [Indexed: 01/08/2023] Open
Abstract
The availability of a well-characterized animal brain tumor model will play an important role in identifying treatments for human brain tumors. Wistar rats bearing 9L glioma cells can develop solid, well-circumcised tumors, and may be a useful animal model for the evaluation of various therapeutic approaches for gliosarcomas. In this study, the 9L/Wistar rat glioma model was produced by intracerebral implantation of 9LLUC glioma cells syngenic to Fischer 344 (F344) rats. Bioluminescence imaging showed that tumors progressively grew from day 7 to day 21 in 9LLUC/F344 rats, and tumor regression was found in some 9LLUC/Wistar rats. Hematoxylin-eosin staining verified that intracranial tumors were gliomas. Immunohistochemistry results demonstrated that no CD4- and CD8-positive cells were found in the syngeneic 9LLUC/F344 model. However, many infiltrating CD4- and CD8-positive cells were observed within the tumors of the 9LLUC/Wistar model. Our data suggests that compared with 9L/F344 rats, 9L glioma Wistar rats may not be suitable for evaluating brain glioma immunotherapies, even though the model induced an immune response and exhibited tumor regression.
Collapse
Affiliation(s)
- Liping Yang
- Department of Medicine, the 263 Hospital, Beijing 101149, China ; Department of Neurology, South West Hospital, the Third Military Medical University of Chinese PLA, Chongqing 400038, China
| | - Jingxiang Zhao
- Department of Stem Cell and Regenerative Medicine, Beijing Institute of Transfusion Medicine, Beijing 100850, China
| | - Guihong Zhou
- Department of Neurosurgery, the 263 Hospital, Beijing 101149, China
| | - Yunfang Wang
- Department of Stem Cell and Regenerative Medicine, Beijing Institute of Transfusion Medicine, Beijing 100850, China
| | - Lusi Li
- Department of Neurology, South West Hospital, the Third Military Medical University of Chinese PLA, Chongqing 400038, China
| | - Hongfeng Yuan
- Department of Stem Cell and Regenerative Medicine, Beijing Institute of Transfusion Medicine, Beijing 100850, China
| | - Xue Nan
- Department of Stem Cell and Regenerative Medicine, Beijing Institute of Transfusion Medicine, Beijing 100850, China
| | - Lidong Guan
- Department of Stem Cell and Regenerative Medicine, Beijing Institute of Transfusion Medicine, Beijing 100850, China
| | - Xuetao Pei
- Department of Stem Cell and Regenerative Medicine, Beijing Institute of Transfusion Medicine, Beijing 100850, China
| |
Collapse
|
19
|
Chae M, Peterson TE, Balgeman A, Chen S, Zhang L, Renner DN, Johnson AJ, Parney IF. Increasing glioma-associated monocytes leads to increased intratumoral and systemic myeloid-derived suppressor cells in a murine model. Neuro Oncol 2014; 17:978-91. [PMID: 25537019 DOI: 10.1093/neuonc/nou343] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Accepted: 10/31/2014] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Patients with glioblastoma multiforme (GBM) exhibit marked intratumoral and systemic immunosuppression. GBM is heavily infiltrated with monocytic cells. Monocytes contacting GBM cells develop features of immunosuppressive myeloid-derived suppressor cells (MDSCs), which are elevated in GBM patients. Therefore, we hypothesized that circulating MDSC levels could be raised in vivo by increasing glioma-associated macrophages. METHODS GL261-luciferase glioma was implanted intracranially in C57BL/6 mice with or without additional normal syngeneic CD11b+ monocytes. Tumor growth and intratumoral and systemic MDSC (CD11b+/Gr-1+) levels were determined. Green fluorescent protein (GFP)-transgenic monocytes were coinjected intracranially with GL261-luciferase cells. GFP+ cell frequency among splenic and bone marrow MDSCs was determined. Impact of increased MDSC's on spontaneous immune responses to tumor cells expressing a model antigen (ovalbumin [OVA]) was determined. RESULTS Tumors grew faster and MDSC's were increased in tumor, spleen, and bone marrow in mice receiving GL261-Luc plus monocytes. Many (30%-50%) systemic MDSC's were GFP+ in mice receiving intracranial tumor plus GFP-transgenic monocytes, suggesting that they originated from glioma-associated monocytes. Tumor-infiltrating OVA-specific CD8+ T cells were markedly reduced in mice receiving GL261-OVA and monocytes compared with mice receiving GL261-OVA alone. CONCLUSIONS Increasing glioma-associated macrophages in intracranial GL261 glioma decreases survival and markedly increases intratumoral and systemic MDSC's, many of which originate directly from glioma-associated macrophages. This is associated with decreased spontaneous immune responses to a model antigen. To our knowledge, this is the first evidence in cancer that systemic MDSC's can arise directly from normal monocytes that have undergone intratumoral immunosuppressive education.
Collapse
Affiliation(s)
- Michael Chae
- Department of Neurological Surgery (M.C., T.E.P., A.B., S.C., L.Z., I.F.P.) and Department of Immunology, Mayo Clinic, Rochester, Minnesota (D.N.R., A.J.J.)
| | - Timothy E Peterson
- Department of Neurological Surgery (M.C., T.E.P., A.B., S.C., L.Z., I.F.P.) and Department of Immunology, Mayo Clinic, Rochester, Minnesota (D.N.R., A.J.J.)
| | - Alexis Balgeman
- Department of Neurological Surgery (M.C., T.E.P., A.B., S.C., L.Z., I.F.P.) and Department of Immunology, Mayo Clinic, Rochester, Minnesota (D.N.R., A.J.J.)
| | - Selby Chen
- Department of Neurological Surgery (M.C., T.E.P., A.B., S.C., L.Z., I.F.P.) and Department of Immunology, Mayo Clinic, Rochester, Minnesota (D.N.R., A.J.J.)
| | - Lei Zhang
- Department of Neurological Surgery (M.C., T.E.P., A.B., S.C., L.Z., I.F.P.) and Department of Immunology, Mayo Clinic, Rochester, Minnesota (D.N.R., A.J.J.)
| | - Danielle N Renner
- Department of Neurological Surgery (M.C., T.E.P., A.B., S.C., L.Z., I.F.P.) and Department of Immunology, Mayo Clinic, Rochester, Minnesota (D.N.R., A.J.J.)
| | - Aaron J Johnson
- Department of Neurological Surgery (M.C., T.E.P., A.B., S.C., L.Z., I.F.P.) and Department of Immunology, Mayo Clinic, Rochester, Minnesota (D.N.R., A.J.J.)
| | - Ian F Parney
- Department of Neurological Surgery (M.C., T.E.P., A.B., S.C., L.Z., I.F.P.) and Department of Immunology, Mayo Clinic, Rochester, Minnesota (D.N.R., A.J.J.)
| |
Collapse
|
20
|
Mathieu D, Lecomte R, Tsanaclis AM, Larouche A, Fortin D. Standardization and Detailed Characterization of the Syngeneic Fischer/F98 Glioma Model. Can J Neurol Sci 2014; 34:296-306. [PMID: 17803026 DOI: 10.1017/s0317167100006715] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Introduction:Adequate animal glioma models are mandatory for the pursuit of preclinical research in neuro-oncology. Many implantation models have been described, but none perfectly emulate human malignant gliomas. This work reports our experience in standardizing, optimizing and characterizing the Fischer/F98 glioma model on the clinical, pathological, radiological and metabolic aspects.Materials and methods:F98 cells were implanted in 70 Fischer rats, varying the quantity of cells and volume of implantation solution, and using a micro-infusion pump to minimize implantation trauma, after adequate coordinates were established. Pathological analysis consisted in hematoxylin and eosin (H&E) staining and immunohistochemistry for GFAP, vimentin, albumin, TGF-b1, TGF-b2, CD3 and CD45. Twelve animals were used for MR imaging at 5, 10, 15 and 20 days. Corresponding MR images were compared with pathological slides. Two animals underwent 18F-FDG and 11C-acetate PET studies for metabolic characterization of the tumors.Results:Implantation with 1x104 cells produced a median survival of 26 days and a tumor take of 100%. Large infiltrative neoplasms with a necrotic core were seen on H&E. Numerous mitosis, peritumoral infiltrative behavior, and neovascular proliferation were also obvious. GFAP and vimentin staining was positive inside the tumor cells. Albumin staining was observed in the extracellular space around the tumors. CD3 staining was negligible. The MR images correlated the pathologic findings. 18F-FDG uptake was strong in the tumors.Conclusion:The standardized model described in this study behaves in a predictable and reproducible fashion, and could be considered for future pre-clinical studies. It adequately mimics the behavior of human malignant astrocytomas.
Collapse
Affiliation(s)
- David Mathieu
- Department of Surgery, Division of Neurosurgery and Neuro-oncology, Centre Hospitalier Universitaire de Sherbrooke. Sherbrooke University, Sherbrooke, Quebec, Canada
| | | | | | | | | |
Collapse
|
21
|
De K, Behera A, Banerjee I, Sarkar B, Ganguly S, Misra M. Radiolabeled novel peptide for imaging somatostatin-receptor expressing tumor: synthesis and radiobiological evaluation. J Radioanal Nucl Chem 2014. [DOI: 10.1007/s10967-014-3199-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
22
|
Ebrahimi Shahmabadi H, Movahedi F, Koohi Moftakhari Esfahani M, Alavi SE, Eslamifar A, Mohammadi Anaraki G, Akbarzadeh A. Efficacy of Cisplatin-loaded polybutyl cyanoacrylate nanoparticles on the glioblastoma. Tumour Biol 2014; 35:4799-806. [PMID: 24443270 DOI: 10.1007/s13277-014-1630-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Accepted: 01/06/2014] [Indexed: 12/24/2022] Open
Abstract
Glioblastoma is known as one of the most aggressive human cancers. To gain access of the brain, therapeutic agents must overcome blood-brain barrier (BBB). In this study, Cisplatin (Cispt)-loaded polybutylcyanoacrylate (PBCA) nanoparticles (NPs) were prepared through miniemulsion polymerization technique. They were coated with polysorbate 80 to cross the BBB of glioblastoma-bearing rats. Prepared NPs were characterized with respect to their size, size distribution, zeta potential, drug loading and encapsulation efficiency, cytotoxicity effects, drug release, and stability pattern. Size and zeta potential of nanodrug were found to be 489 nm and -20 mV, while drug loading and encapsulation efficiency were determined to be 5% and 25%, respectively. Release studies demonstrated high retention capability of nanodrug in that 3.18% of Cispt was released from NPs in a period of 51 h. NPs presented acceptable stability after 2 months and lyophilization. Mean survival time in nanodrug receivers was 19.6 days, while it was 17.5 days for free drug receivers. Histological studies demonstrated efficacy of PBCA NPs in reducing side effects. Finally, such preparation can be considered as a promising nanocarrier for other types of tumor.
Collapse
|
23
|
Marabelle A, Kohrt H, Sagiv-Barfi I, Ajami B, Axtell RC, Zhou G, Rajapaksa R, Green MR, Torchia J, Brody J, Luong R, Rosenblum MD, Steinman L, Levitsky HI, Tse V, Levy R. Depleting tumor-specific Tregs at a single site eradicates disseminated tumors. J Clin Invest 2013; 123:2447-63. [PMID: 23728179 DOI: 10.1172/jci64859] [Citation(s) in RCA: 295] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Accepted: 03/14/2013] [Indexed: 01/22/2023] Open
Abstract
Activation of TLR9 by direct injection of unmethylated CpG nucleotides into a tumor can induce a therapeutic immune response; however, Tregs eventually inhibit the antitumor immune response and thereby limit the power of cancer immunotherapies. In tumor-bearing mice, we found that Tregs within the tumor preferentially express the cell surface markers CTLA-4 and OX40. We show that intratumoral coinjection of anti-CTLA-4 and anti-OX40 together with CpG depleted tumor-infiltrating Tregs. This in situ immunomodulation, which was performed with low doses of antibodies in a single tumor, generated a systemic antitumor immune response that eradicated disseminated disease in mice. Further, this treatment modality was effective against established CNS lymphoma with leptomeningeal metastases, sites that are usually considered to be tumor cell sanctuaries in the context of conventional systemic therapy. These results demonstrate that antitumor immune effectors elicited by local immunomodulation can eradicate tumor cells at distant sites. We propose that, rather than using mAbs to target cancer cells systemically, mAbs could be used to target the tumor infiltrative immune cells locally, thereby eliciting a systemic immune response.
Collapse
Affiliation(s)
- Aurélien Marabelle
- Department of Medicine, Division of Oncology, Stanford University, Stanford, California 94305, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Sho A, Kondo S, Kamitani H, Otake M, Watanabe T. Establishment of experimental glioma models at the intrinsic brainstem region of the rats. Neurol Res 2013; 29:36-42. [PMID: 17427273 DOI: 10.1179/016164106x115080] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
OBJECTIVES As the treatment of human intrinsic brainstem gliomas remains challenging, experimental glioma models are needed. METHODS We developed a rat model of intrinsic brain stem glioma that uses a stereotactic frame to fix the head for the delivery of C6 glioma cells to target sites via a permanently implanted cannula. We inoculated the rat midbrain, pons or cerebral cortex with 5 x 10(4) cells suspended in 1 microl culture medium over the course of 2 minutes. RESULTS Three days post-implantation, tumor formation was visible in the periaqueductal gray matter in the midbrain and the tegmentum of the pons. On the tenth day, the tumor diameter exceeded over 2 mm; there was no tumor cell seeding into the cerebrospinal fluid space. The tumor manifested the histological features typical of glioblastoma; Ki-67 labeling index was 32%. DISCUSSION Because in our model the cannula is permanently implanted, additional inocula can be delivered. Here we detail our rat brainstem glioma model and discuss its usefulness for the investigation of these tumor in humans.
Collapse
Affiliation(s)
- Atsuko Sho
- Department of Neurosurgery, Institute of Neurological Sciences, Faculty of Medicine, Tottori University, 36-1 Nishi-cho, Yonago 683-8504, Japan.
| | | | | | | | | |
Collapse
|
25
|
Illán-Cabeza NA, García-García AR, Martínez-Martos JM, Ramírez-Expósito MJ, Peña-Ruiz T, Moreno-Carretero MN. A potential antitumor agent, (6-amino-1-methyl-5-nitrosouracilato-N3)-triphenylphosphine-gold(I): structural studies and in vivo biological effects against experimental glioma. Eur J Med Chem 2013; 64:260-72. [PMID: 23644209 DOI: 10.1016/j.ejmech.2013.03.067] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Revised: 03/29/2013] [Accepted: 03/30/2013] [Indexed: 02/07/2023]
Abstract
The synthesis and molecular and supramolecular structures of the compound (6-amino-1-methyl-5-nitrosouracilato-N3)-triphenylphosphine-gold(I) with interesting abilities to inhibit tumor growth in an animal model of experimental glioma are reported. Thus, its antitumor properties, effects on both enzyme and non-enzyme antioxidant defense systems and the response of several biochemical biomarkers have been analyzed. After seven days of treatment, the gold compound decreased the tumor growth to ca. one-tenth and reduced oxidative stress biomarkers (thiobarbituric acid-reactive substances (TBARS) and protein oxidation levels) compared to animals treated with the vehicle. Also, gold compound maintained non-enzyme antioxidant defense systems as in non-tumor animals and increased enzyme antioxidant defenses, such as superoxide dismutase and glutathione peroxidase activities, and decreased catalase activity. Analysis of serum levels of electrolytes, nitrogenous compounds, glucose, lipids, total protein, albumin, transaminases and alkaline phosphatase indicated that gold compound treatment showed few adverse effects, while effectively inhibiting tumor growth through mechanisms that involved endogenous antioxidant defenses.
Collapse
|
26
|
Huszthy PC, Daphu I, Niclou SP, Stieber D, Nigro JM, Sakariassen PØ, Miletic H, Thorsen F, Bjerkvig R. In vivo models of primary brain tumors: pitfalls and perspectives. Neuro Oncol 2012; 14:979-93. [PMID: 22679124 PMCID: PMC3408261 DOI: 10.1093/neuonc/nos135] [Citation(s) in RCA: 175] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Animal modeling for primary brain tumors has undergone constant development over the last 60 years, and significant improvements have been made recently with the establishment of highly invasive glioblastoma models. In this review we discuss the advantages and pitfalls of model development, focusing on chemically induced models, various xenogeneic grafts of human cell lines, including stem cell–like cell lines and biopsy spheroids. We then discuss the development of numerous genetically engineered models available to study mechanisms of tumor initiation and progression. At present it is clear that none of the current animal models fully reflects human gliomas. Yet, the various model systems have provided important insight into specific mechanisms of tumor development. In particular, it is anticipated that a combined comprehensive knowledge of the various models currently available will provide important new knowledge on target identification and the validation and development of new therapeutic strategies.
Collapse
Affiliation(s)
- Peter C Huszthy
- NorLux, Neuro-Oncology Laboratory, Department of Biomedicine, University of Bergen, Bergen, Norway
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
[Animal models of human glioma: the progress of application and investigation]. DONG WU XUE YAN JIU = ZOOLOGICAL RESEARCH 2012; 33:337-42. [PMID: 22653863 DOI: 10.3724/sp.j.1141.2012.03337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The glioma accounts for half of the central nervous tumors, among which the glioblastoma multiforme (GBM) is one of the most aggressive and lethal brain tumors. The difficulties in glioma therapy indicate the need of appropriate animal models for preclinical studies. Benefiting from the development of molecular biology, genetics, and transgenic technology, variable animal models of glioma have been established. These animal models of human glioma are reviewed in this paper.
Collapse
|
28
|
Chung DS, Kim CH, Hong YK. Animal models for vaccine therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 746:143-50. [PMID: 22639165 DOI: 10.1007/978-1-4614-3146-6_11] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Animal models are important for defining paradigms of tumor immunology and for evaluating therapeutic efficacy of immunotherapy. Many animal models have been used for evaluating in vivo characteristics of malignant gliomas and their responses to therapy. No animal model, however, is perfect because malignant glioma has a very heterogeneous biological behavior. There are so many parallels between mouse and human immunology, but there are significant discrepancies in immune system. Animal models for vaccine therapy can be classified as transplantable tumor models and models of spontaneous tumor in genetically engineered animals. Although transplantable tumor models have been used to test immunotherapeutic efficacy and remain a mainstay in study of brain tumor immunology, a lot of tumor vaccines that look promising in experimental animals have turned out to be ineffective clinically. Recent advances of laboratory techniques and understanding of genetic and molecular characteristics of gliomas allows for animal models of gliomas with similar biologic characteristics. Well-designed glioma models that accurately reflect the biology, pathology and clinical behaviors of human gliomas can provide more useful preclinical informations to predict clinical efficacy of novel immunotherapies and cancer vaccines.
Collapse
Affiliation(s)
- Dong-Sup Chung
- Department of Neurosurgery, The Catholic University of Korea, Seoul, Republic of Korea
| | | | | |
Collapse
|
29
|
Anatomical differences determine distribution of adenovirus after convection-enhanced delivery to the rat brain. PLoS One 2011; 6:e24396. [PMID: 22022354 PMCID: PMC3192704 DOI: 10.1371/journal.pone.0024396] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Accepted: 08/09/2011] [Indexed: 11/19/2022] Open
Abstract
Background Convection-enhanced delivery (CED) of adenoviruses offers the potential of widespread virus distribution in the brain. In CED, the volume of distribution (Vd) should be related to the volume of infusion (Vi) and not to dose, but when using adenoviruses contrasting results have been reported. As the characteristics of the infused tissue can affect convective delivery, this study was performed to determine the effects of the gray and white matter on CED of adenoviruses and similar sized super paramagnetic iron oxide nanoparticles (SPIO). Methodology/Principal Findings We convected AdGFP, an adenovirus vector expressing Green Fluorescent Protein, a virus sized SPIO or trypan blue in the gray and white matter of the striatum and external capsule of Wistar rats and towards orthotopic infiltrative brain tumors. The resulting Vds were compared to Vi and transgene expression to SPIO distribution. Results show that in the striatum Vd is not determined by the Vi but by the infused virus dose, suggesting diffusion, active transport or receptor saturation rather than convection. Distribution of virus and SPIO in the white matter is partly volume dependent, which is probably caused by preferential fluid pathways from the external capsule to the surrounding gray matter, as demonstrated by co-infusing trypan blue. Distant tumors were reached using the white matter tracts but tumor penetration was limited. Conclusions/Significance CED of adenoviruses in the rat brain and towards infiltrative tumors is feasible when regional anatomical differences are taken into account while SPIO infusion could be considered to validate proper catheter positioning and predict adenoviral distribution.
Collapse
|
30
|
Abstract
GBM (glioblastoma multiforme) is a highly aggressive brain tumour with very poor prognosis despite multi-modalities of treatment. Furthermore, recent failure of targeted therapy for these tumours highlights the need of appropriate rodent models for preclinical studies. In this review, we highlight the most commonly used rodent models (U251, U86, GL261, C6, 9L and CNS-1) with a focus on the pathological and genetic similarities to the human disease. We end with a comprehensive review of the CNS-1 rodent model.
Collapse
|
31
|
Jantaratnotai N, McLarnon JG. Calcium dependence of purinergic subtype P2Y₁ receptor modulation of C6 glioma cell migration. Neurosci Lett 2011; 497:80-4. [PMID: 21540076 DOI: 10.1016/j.neulet.2011.04.034] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2011] [Revised: 04/12/2011] [Accepted: 04/12/2011] [Indexed: 10/18/2022]
Abstract
We have examined activation of purinergic P2Y₁ receptor-dependent Ca²⁺-signaling pathways in mediating C6 glioma cell migration. The administration of 2-methylthioadenosine 5'-diphosphate (2MeSADP), a selective agonist for P2Y₁R, induced marked increases in patterns of glioma migration in both scratch wound and Boyden chamber assays. Antagonism of P2Y₁R with either the broad spectrum purinergic blocker, pyridoxal-phosphate-6-azophenyl-2',4'-disulfonate (PPADS) or the specific P2Y₁R antagonist, 2'-deoxy-N⁶-methyladenosine-3',5'-bisphosphate (MRS2179), significantly inhibited C6 cell migration. Calcium-sensitive spectrofluorometry showed 2MeSADP stimulation of glioma cells caused a biphasic change in intracellular Ca²⁺ ([Ca²⁺]i). The rapid transient phase was unchanged in Ca²⁺-free solution reflecting a [Ca²⁺]i component due to intracellular stores release subsequent to activation of a metabotropic P2Y subtype receptor. The secondary prolonged phase of [Ca²⁺]i was abolished in Ca²⁺-free solution or in glioma cells treated with the store-operated channel (SOC) blocker, SKF96365. Treatment of glioma with either MRS2179 or PPADS significantly attenuated both the rapid and prolonged phases of [Ca²⁺]i. These results suggest critical roles for activation of P2Y₁R in mediating glioma cell mobility and migration with changes in [Ca²⁺]i contributing as a mechanistic link between activated receptor and functional response. Our findings suggest that pharmacological modulation of metabotropic P2Y₁R-dependent signaling pathways may serve as a novel therapeutic procedure to slow glioma progression.
Collapse
Affiliation(s)
- Nattinee Jantaratnotai
- Department of Anesthesiology, Pharmacology and Therapeutics, Faculty of Medicine, University of British Columbia, 2176 Health Sciences Mall, Vancouver, BC V6T1Z3 Canada
| | | |
Collapse
|
32
|
Bai RY, Staedtke V, Riggins GJ. Molecular targeting of glioblastoma: Drug discovery and therapies. Trends Mol Med 2011; 17:301-312. [PMID: 21411370 DOI: 10.1016/j.molmed.2011.01.011] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2010] [Revised: 01/10/2011] [Accepted: 01/21/2011] [Indexed: 12/19/2022]
Abstract
Despite advances in treatment for glioblastoma multiforme (GBM), patient prognosis remains poor. Although there is growing evidence that molecular targeting could translate into better survival for GBM, current clinical data show limited impact on survival. Recent progress in GBM genomics implicate several activated pathways and numerous mutated genes. This molecular diversity can partially explain therapeutic resistance and several approaches have been postulated to target molecular changes. Furthermore, most drugs are unable to reach effective concentrations within the tumor owing to elevated intratumoral pressure, restrictive vasculature and other limiting factors. Here, we describe the preclinical and clinical developments in treatment strategies of GBM. We review the current clinical trials for GBM and discuss the challenges and future directions of targeted therapies.
Collapse
Affiliation(s)
- Ren-Yuan Bai
- Departments of Neurosurgery, Johns Hopkins University School of Medicine, CRB II Rm. 257, 1550 Orleans Street, Baltimore, MD 21231, USA
| | - Verena Staedtke
- Departments of Neurosurgery, Johns Hopkins University School of Medicine, CRB II Rm. 257, 1550 Orleans Street, Baltimore, MD 21231, USA
| | - Gregory J Riggins
- Departments of Neurosurgery, Johns Hopkins University School of Medicine, CRB II Rm. 257, 1550 Orleans Street, Baltimore, MD 21231, USA
| |
Collapse
|
33
|
Miura FK, Alves MJF, Rocha MC, da Silva R, Oba-Shinjo SM, Marie SKN. Xenograft transplantation of human malignant astrocytoma cells into immunodeficient rats: an experimental model of glioblastoma. Clinics (Sao Paulo) 2010; 65:305-9. [PMID: 20360922 PMCID: PMC2845772 DOI: 10.1590/s1807-59322010000300011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2009] [Accepted: 12/14/2009] [Indexed: 11/22/2022] Open
Abstract
INTRODUCTION Astrocytic gliomas are the most common intracranial central nervous system neoplasias, accounting for about 60% of all primary central nervous system tumors. Despite advances in the treatment of gliomas, no effective therapeutic approach is yet available; hence, the search for a more realistic model to generate more effective therapies is essential. OBJECTIVE To develop an experimental malignant astrocytoma model with the characteristics of the human tumor. METHOD Primary cells from subcutaneous xenograft tumors produced with malignant astrocytoma U87MG cells were inoculated intracerebrally by stereotaxis into immunosuppressed (athymic) Rowett rats. RESULTS All four injected animals developed non-infiltrative tumors, although other glioblastoma characteristics, such as necrosis, pseudopalisading cells and intense mitotic activity, were observed. CONCLUSION A malignant astrocytoma intracerebral xenograft model with poorly invasive behavior was achieved in athymic Rowett rats. Tumor invasiveness in an experimental animal model may depend on a combination of several factors, including the cell line used to induce tumor formation, the rat strains and the status of the animal's immune system.
Collapse
Affiliation(s)
- Flávio Key Miura
- Laboratory of Molecular and Cellular Biology, Faculdade de Medicina da Universidade de São Paulo - São Paulo/SP, Brazil.
| | | | | | | | | | | |
Collapse
|
34
|
Abstract
Abnormalities of cellular immunity are commonly seen in patients with glioblastoma (GBM), and the subsequent relative immunosuppression likely contributes to poor tumor-specific responses in affected individuals. Endogenous immune regulation is likely to limit the efficacy of a wide array of immunotherapeutic strategies, therefore mandating consideration in the continued development of novel treatments for GBM. Various tumor-associated factors have been implicated as potential generators of the immunosuppressive effect. This article outlines relevant experimentation exploring the nature of immune defects in patients with GBM, including a critical discussion of tumor-secreted factors, cell-surface proteins, and more recently described populations of immunoregulatory leukocytes that have potential roles in the subversion of cellular immunity.
Collapse
Affiliation(s)
- Allen Waziri
- Department of Neurosurgery, University of Colorado Health Sciences Center, 12631 E. 17th Avenue, Aurora, CO 80045, USA.
| |
Collapse
|
35
|
Sughrue ME, Yang I, Kane AJ, Rutkowski MJ, Fang S, James CD, Parsa AT. Immunological considerations of modern animal models of malignant primary brain tumors. J Transl Med 2009; 7:84. [PMID: 19814820 PMCID: PMC2768693 DOI: 10.1186/1479-5876-7-84] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2009] [Accepted: 10/08/2009] [Indexed: 12/26/2022] Open
Abstract
Recent advances in animal models of glioma have facilitated a better understanding of biological mechanisms underlying gliomagenesis and glioma progression. The limitations of existing therapy, including surgery, chemotherapy, and radiotherapy, have prompted numerous investigators to search for new therapeutic approaches to improve quantity and quality of survival from these aggressive lesions. One of these approaches involves triggering a tumor specific immune response. However, a difficulty in this approach is the the scarcity of animal models of primary CNS neoplasms which faithfully recapitulate these tumors and their interaction with the host's immune system. In this article, we review the existing methods utilized to date for modeling gliomas in rodents, with a focus on the known as well as potential immunological aspects of these models. As this review demonstrates, many of these models have inherent immune system limitations, and the impact of these limitations on studies on the influence of pre-clinical therapeutics testing warrants further attention.
Collapse
Affiliation(s)
- Michael E Sughrue
- Department of Neurological Surgery, University of California at San Francisco, San Francisco, California, USA.
| | | | | | | | | | | | | |
Collapse
|
36
|
Ghulam Muhammad AKM, Candolfi M, King GD, Yagiz K, Foulad D, Mineharu Y, Kroeger KM, Treuer KA, Nichols WS, Sanderson NS, Yang J, Khayznikov M, Van Rooijen N, Lowenstein PR, Castro MG. Antiglioma immunological memory in response to conditional cytotoxic/immune-stimulatory gene therapy: humoral and cellular immunity lead to tumor regression. Clin Cancer Res 2009; 15:6113-27. [PMID: 19789315 DOI: 10.1158/1078-0432.ccr-09-1087] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Glioblastoma multiforme is a deadly primary brain cancer. Because the tumor kills due to recurrences, we tested the hypothesis that a new treatment would lead to immunological memory in a rat model of recurrent glioblastoma multiforme. EXPERIMENTAL DESIGN We developed a combined treatment using an adenovirus (Ad) expressing fms-like tyrosine kinase-3 ligand (Flt3L), which induces the infiltration of immune cells into the tumor microenvironment, and an Ad expressing herpes simplex virus-1-thymidine kinase (TK), which kills proliferating tumor cells in the presence of ganciclovir. RESULTS This treatment induced immunological memory that led to rejection of a second glioblastoma multiforme implanted in the contralateral hemisphere and of an extracranial glioblastoma multiforme implanted intradermally. Rechallenged long-term survivors exhibited anti-glioblastoma multiforme-specific T cells and displayed specific delayed-type hypersensitivity. Using depleting antibodies, we showed that rejection of the second tumor was dependent on CD8(+) T cells. Circulating anti-glioma antibodies were observed when glioblastoma multiforme cells were implanted intradermally in naïve rats or in long-term survivors. However, rats bearing intracranial glioblastoma multiforme only exhibited circulating antitumoral antibodies upon treatment with Ad-Flt3L + Ad-TK. This combined treatment induced tumor regression and release of the chromatin-binding protein high mobility group box 1 in two further intracranial glioblastoma multiforme models, that is, Fisher rats bearing intracranial 9L and F98 glioblastoma multiforme cells. CONCLUSIONS Treatment with Ad-Flt3L + Ad-TK triggered systemic anti-glioblastoma multiforme cellular and humoral immune responses, and anti-glioblastoma multiforme immunological memory. Release of the chromatin-binding protein high mobility group box 1 could be used as a noninvasive biomarker of therapeutic efficacy for glioblastoma multiforme. The robust treatment efficacy lends further support to its implementation in a phase I clinical trial.
Collapse
Affiliation(s)
- A K M Ghulam Muhammad
- Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California 90048, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Oh S, Ohlfest JR, Todhunter DA, Vallera VD, Hall WA, Chen H, Vallera DA. Intracranial elimination of human glioblastoma brain tumors in nude rats using the bispecific ligand-directed toxin, DTEGF13 and convection enhanced delivery. J Neurooncol 2009; 95:331-342. [PMID: 19517064 DOI: 10.1007/s11060-009-9932-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2008] [Accepted: 05/25/2009] [Indexed: 10/20/2022]
Abstract
A bispecific ligand-directed toxin (BLT) consisting of human interleukin-13, epithelial growth factor, and the first 389 amino acids of diphtheria toxin was assembled in order to target human glioblastoma. In vitro, DTEGF13 selectively killed the human glioblastoma cell line U87-luc as well as other human glioblastomas. DTEGF13 fulfilled the requirement of a successful BLT by having greater activity than either of its monospecific counterparts or their mixture proving it necessary to have both ligands on the same single chain molecule. Aggressive brain tumors established intracranially (IC) in nude rats with U87 glioma genetically marked with a firefly luciferase reporter gene were treated with two injections of DTEGF13 using convection enhanced delivery resulting in tumor eradication in 50% of the rats which survived with tumor free status at least 110 days post tumor inoculation. An irrelevant BLT control did not protect establishing specificity. The bispecific DTEGF13 MTD dose was measured at 2 microg/injection or 0.5 microg/kg and toxicity studies indicated safety in this dose. Combination of monospecific DTEGF and DTIL13 did not inhibit tumor growth. ELISA assay indicated that anti-DT antibodies were not generated in normal immunocompetent rats given identical intracranial DTEGF13 therapy. Thus, DTEGF13 is safe and efficacious as an alternative drug for glioblastoma therapy and warrants further study.
Collapse
Affiliation(s)
- Seunguk Oh
- Section on Molecular Cancer Therapeutics, Department of Therapeutic Radiology-Radiation Oncology, University of Minnesota Masonic Cancer Center, MMC: 367, Minneapolis, MN, 55455, USA
| | - John R Ohlfest
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - Deborah A Todhunter
- Section on Molecular Cancer Therapeutics, Department of Therapeutic Radiology-Radiation Oncology, University of Minnesota Masonic Cancer Center, MMC: 367, Minneapolis, MN, 55455, USA
| | - Vincent D Vallera
- Section on Molecular Cancer Therapeutics, Department of Therapeutic Radiology-Radiation Oncology, University of Minnesota Masonic Cancer Center, MMC: 367, Minneapolis, MN, 55455, USA
| | - Walter A Hall
- Department of Neurosurgery, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Hua Chen
- Section on Molecular Cancer Therapeutics, Department of Therapeutic Radiology-Radiation Oncology, University of Minnesota Masonic Cancer Center, MMC: 367, Minneapolis, MN, 55455, USA
| | - Daniel A Vallera
- Section on Molecular Cancer Therapeutics, Department of Therapeutic Radiology-Radiation Oncology, University of Minnesota Masonic Cancer Center, MMC: 367, Minneapolis, MN, 55455, USA.
| |
Collapse
|
38
|
Barth RF, Kaur B. Rat brain tumor models in experimental neuro-oncology: the C6, 9L, T9, RG2, F98, BT4C, RT-2 and CNS-1 gliomas. J Neurooncol 2009; 94:299-312. [PMID: 19381449 DOI: 10.1007/s11060-009-9875-7] [Citation(s) in RCA: 300] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2009] [Accepted: 03/16/2009] [Indexed: 02/08/2023]
Abstract
In this review we will describe eight commonly used rat brain tumor models and their application for the development of novel therapeutic and diagnostic modalities. The C6, 9L and T9 gliomas were induced by repeated injections of methylnitrosourea (MNU) to adult rats. The C6 glioma has been used extensively for a variety of studies, but since it arose in an outbred Wistar rat, it is not syngeneic to any inbred strain, and its potential to evoke an alloimmune response is a serious limitation. The 9L gliosarcoma has been used widely and has provided important information relating to brain tumor biology and therapy. The T9 glioma, although not generally recognized, was and probably still is the same as the 9L. Both of these tumors arose in Fischer rats and can be immunogenic in syngeneic hosts, a fact that must be taken into consideration when used in therapy studies, especially if survival is the endpoint. The RG2 and F98 gliomas were both chemically induced by administering ethylnitrosourea (ENU) to pregnant rats, the progeny of which developed brain tumors that subsequently were propagated in vitro and cloned. They are either weakly or non-immunogenic and have an invasive pattern of growth and uniform lethality, which make them particularly attractive models to test new therapeutic modalities. The CNS-1 glioma was induced by administering MNU to a Lewis rat. It has an infiltrative pattern of growth and is weakly immunogenic, which should make it useful in experimental neuro-oncology. Finally, the BT4C glioma was induced by administering ENU to a BD IX rat, following which brain cells were propagated in vitro until a tumorigenic clone was isolated. This tumor has been used for a variety of studies to evaluate new therapeutic modalities. The Avian Sarcoma Virus (ASV) induced tumors, and a continuous cell line derived from one of them designated RT-2, have been useful for studies in which de novo tumor induction is an important requirement. These tumors also are immunogenic and this limits their usefulness for therapy studies. It is essential to recognize the limitations of each of the models that have been described, and depending upon the nature of the study to be conducted, it is important that the appropriate model be selected.
Collapse
Affiliation(s)
- Rolf F Barth
- Department of Pathology, The Ohio State University, 165 Hamilton Hall, Columbus, OH 43210, USA.
| | | |
Collapse
|
39
|
Parney IF, Waldron JS, Parsa AT. Flow cytometry and in vitro analysis of human glioma-associated macrophages. Laboratory investigation. J Neurosurg 2009; 110:572-82. [PMID: 19199469 DOI: 10.3171/2008.7.jns08475] [Citation(s) in RCA: 126] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECT To date, glioma immunotherapy has been focused mostly on stimulating antitumor peripheral lymphocyte responses; however, some data suggest that microglia and/or macrophages (not lymphocytes) are the predominant inflammatory cells infiltrating gliomas. To study this hypothesis further, the authors analyzed inflammatory cell infiltrates in fresh human malignant glioma specimens and primary cultures. METHODS Single-cell suspensions from fresh operative malignant glioma specimens, obtained by stereotactic localization, were analyzed for CD11b and CD45 by using flow cytometry. A comparison was made with peripheral blood mononuclear cells. In a subset of patients, a more detailed flow cytometry analysis of Class I and II major histocompatibility complex, B7-1, B7-2, CD11c, and CD14 expression was performed. Macrophage-like cells in primary glioma cultures were similarly assessed. RESULTS Operative samples were obtained from 9 newly diagnosed malignant gliomas. The mean percent of CD45(+)/CD11b(-) cells (lymphocytes) was 2.48% (range 0.65-5.50%); CD45(dim)/CD11b(+) cells (microglia), 1.65% (range 0.37-3.92%); and CD45(bright)/CD11b+ (monocytes/macrophages), 6.25% (range 1.56-15.3%). More detailed fluorescence-activated cell sorting suggested that macrophage-like cells expressed Class I and II major histocompatibility complex, B7-2, and CD11c but not CD14 or B7-1. Primary human glioma cultures contained significant numbers of macrophage-like (CD45(bright)/CD11b(+)) cells, but these cells were lost with successive passages. These cells maintained the immunomarker profiles of macrophage-like cells from fresh specimens only if they were cultured in serum-free media. CONCLUSIONS The CD45(+)/CD11b(+) cells are the predominant inflammatory cell infiltrating human gliomas. Of this type, the CD45(bright)/CD11b(+) cells, a phenotype compatible with circulating macrophages in rodent models, and not microglia, are the most common. Their immunomarker profile is compatible with an immature antigen-presenting cell. They are present in primary glioma cultures but are lost in successive passages. Their role is enigmatic, and they may prove an important target for future glioma immunotherapy studies.
Collapse
Affiliation(s)
- Ian F Parney
- Department of Clinical Neurosciences, Southern Alberta Cancer Research Institute, and Hotchkiss Brain Institute, University of Calgary, Alberta, Canada
| | | | | |
Collapse
|
40
|
Liu Q, Liu R, Kashyap MV, Agarwal R, Shi X, Wang CC, Yang SH. Brainstem glioma progression in juvenile and adult rats. J Neurosurg 2008; 109:849-55. [PMID: 18976074 DOI: 10.3171/jns/2008/109/11/0849] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT Brainstem gliomas are common in children and have the worst prognosis of any brain tumor in this age group. On the other hand, brainstem gliomas are rare in adults, and the authors of some clinical studies have suggested that this lesion behaves differently in adults than in children. In the present study, the authors test an orthotopic C6 brainstem glioma model in juvenile and adult rats, and investigate the biological behavior of this lesion in the 2 age groups. METHODS The C6 glioma cells were stereotactically implanted into the pons of juvenile or adult male rats. Neurological presentation and survival time were recorded. Tumor proliferation and the number of apoptotic cells in brainstem gliomas of young and adult rats were determined by immunohistochemical staining with Ki 67 and terminal deoxynucleotidyl transferase 2'-deoxyuridine 5'-triphosphate-mediated nick-end labeling assay. RESULTS Striking differences in the onset of neurological signs, duration of symptoms, survival time, tumor growth pattern, tumor proliferation, and number of apoptotic cells were found between the gliomas in the 2 groups of rats. The lesions were relatively focal in adult rats but more diffuse in young rats. Furthermore, brainstem gliomas in adult rats were less proliferative and had more apoptotic cells than those in young rats. CONCLUSIONS The authors found that the C6 brainstem glioma model in young and adult rats closely imitates the course of brainstem glioma in humans both in neurological findings and histopathological characteristics. Their findings also suggest that the different growth pattern and invasiveness of these lesions in children compared with that in adults could be due to different cellular environments in the 2 age groups, and warrants further investigation into the difference in the host response to brainstem gliomas in children and adults.
Collapse
Affiliation(s)
- Qing Liu
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas 76107, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
Miura FK, Alves MJF, Rocha MC, Silva RS, Oba-Shinjo SM, Uno M, Colin C, Sogayar MC, Marie SKN. Experimental nodel of C6 brain tumors in athymic rats. ARQUIVOS DE NEURO-PSIQUIATRIA 2008; 66:238-41. [PMID: 18545790 DOI: 10.1590/s0004-282x2008000200019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2007] [Accepted: 02/11/2008] [Indexed: 11/21/2022]
Abstract
Malignant brain tumor experimental models tend to employ cells that are immunologically compatible with the receptor animal. In this study, we have proposed an experimental model of encephalic tumor development by injecting C6 cells into athymic Rowett rats, aiming at reaching a model which more closely resembles to the human glioma tumor. In our model, we observed micro-infiltration of tumor cell clusters in the vicinity of the main tumor mass, and of more distal isolated tumor cells immersed in normal encephalic parenchyma. This degree of infiltration is superior to that usually observed in other C6 models.
Collapse
Affiliation(s)
- Flávio K Miura
- Department of Neurology, Medical School, University of São Paulo, São Paulo, Brazil.
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Regnard P, Duc GL, Bräuer-Krisch E, Troprès I, Siegbahn EA, Kusak A, Clair C, Bernard H, Dallery D, Laissue JA, Bravin A. Irradiation of intracerebral 9L gliosarcoma by a single array of microplanar x-ray beams from a synchrotron: balance between curing and sparing. Phys Med Biol 2008; 53:861-78. [DOI: 10.1088/0031-9155/53/4/003] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
43
|
Karmakar S, Olive MF, Banik NL, Ray SK. Intracranial stereotaxic cannulation for development of orthotopic glioblastoma allograft in Sprague-Dawley rats and histoimmunopathological characterization of the brain tumor. Neurochem Res 2007; 32:2235-42. [PMID: 17701349 DOI: 10.1007/s11064-007-9450-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2007] [Accepted: 07/17/2007] [Indexed: 10/23/2022]
Abstract
Glioblastoma is the most common brain tumor that causes significant mortality annually. Limitations of the current therapeutic regimens warrant development of new techniques and treatment strategies in orthotopic animal model for better management of this devastating brain cancer. There are only a few experimental orthotopic models of glioblastoma for pre-clinical testing. In the present investigation, we successfully implanted rat C6 cells via intracranial stereotaxic cannulation in adult Sprague-Dawley rats for development and histoimmunopathological characterization of an advanced orthotopic glioblastoma allograft model, which could be useful for investigating the course of glioblastoma development as well as for testing efficacy of new therapeutic agents. The orthotopic glioblastoma allograft was generated by intracerebral injection of rat C6 cells through a guide-cannula system and after 21 post-inoculation days the brain tumor was characterized by histoimmunopathological experiments. Histological staining and immunofluorescent labelings for TERT, VEGF, Bcl-2, survivin, XIAP, and GFAP revealed the distinct characteristics of glioblastoma in C6 allograft, which could be useful as a target for treatment with emerging new therapeutic agents. Our investigation indicated the successful development of intracranial cannulated orthotopic glioblastoma allograft in adult Sprague-Dawley rats, making it as a useful animal model of glioblastoma for pre-clinical evaluation of various therapeutic strategies for the management of glioblastoma.
Collapse
Affiliation(s)
- Surajit Karmakar
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | | | | | | |
Collapse
|
44
|
Nakagawa N, Akai F, Fukawa N, Fujita Y, Suzuki M, Ono K, Taneda M. Early effects of boron neutron capture therapy on rat glioma models. Brain Tumor Pathol 2007; 24:7-13. [PMID: 18095138 DOI: 10.1007/s10014-007-0214-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2007] [Accepted: 02/13/2007] [Indexed: 10/23/2022]
Abstract
Early effects of boron neutron capture therapy (BNCT) on malignant glioma are characterized by reduction of the enhancement area and regression of the peritumoral edema radiologically. The aim of this study was to investigate the early histological changes of tumors and inflammatory cells after BNCT in the rat brain. Rats were treated with BNCT using boronophenylalanine (BPA) 7 days after implantation of C6 glioma cells. The tumors were assessed with magnetic resonance imaging and histopathological examination at 4 days after BNCT. The mean tumor volumes were 39 +/- 2 mm3 in the BNCT group and 134 +/- 18 mm3 in the control group. In the BNCT group, tumor cells showed a less pleomorphic appearance with atypical nuclei and mitotic figures. The Ki-67 labeling index was 6.5% +/- 4.7% in the BNCT and 35% +/- 3.8% in the control group. The reactions of the inflammatory cells were examined with ED-1 as macrophage marker and OX42 as microglia marker. ED-1- and OX-42-positive cells were reduced both in the core and the marginal area of the tumor in the BNCT group. It is suggested that BNCT reduced tumor progression by suppression of proliferation. Inhibition of the activated macrophages may relate to reduced peritumoral edema in the early phase.
Collapse
Affiliation(s)
- Nobuhiro Nakagawa
- Department of Neurosurgery, Kinki University School of Medicine, 377-2 Ohnohigashi, Osakasayama, Osaka, 589-8511, Japan.
| | | | | | | | | | | | | |
Collapse
|
45
|
Dehnhardt M, Palm C, Vieten A, Bauer A, Pietrzyk U. Quantifying the A1AR distribution in peritumoural zones around experimental F98 and C6 rat brain tumours. J Neurooncol 2007; 85:49-63. [PMID: 17497078 DOI: 10.1007/s11060-007-9391-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2007] [Accepted: 04/06/2007] [Indexed: 01/24/2023]
Abstract
Quantification of growth in experimental F98 and C6 rat brain tumours was performed on 51 rat brains, 17 of which have been further assessed by 3D tumour reconstruction. Brains were cryosliced and radio-labelled with a ligand of the peripheral type benzodiazepine-receptor (pBR), (3)H-Pk11195 [(1-(2-chlorophenyl)-N-methyl-N-(1-methyl-propylene)-3-isoquinoline-carboxamide)] by receptor autoradiography. Manually segmented and automatically registered tumours have been 3D-reconstructed for volumetric comparison on the basis of (3)H-Pk11195-based tumour recognition. Furthermore automatically computed areas of -300 microm inner (marginal) zone as well as 300 microm and 600 microm outer tumour space were quantified. These three different regions were transferred onto other adjacent slices that had been labelled by receptor autoradiography with the A(1) Adenosine receptor (A(1)AR)-ligand (3)H-CPFPX ((3)H-8-cyclopentyl-3-(3-fluorpropyl)-1-propylxanthine) for quantitative assessment of A(1)AR in the three different tumour zones. Hence, a method is described for quantifying various receptor protein systems in the tumour as well as in the marginal invasive zones around experimentally implanted rat brain tumours and their representation in the tumour microenvironment as well as in 3D space. Furthermore, a tool for automatically reading out radio-labelled rat brain slices from auto radiographic films was developed, reconstructed into a consistent 3D-tumour model and the zones around the tumour were visualized. A(1)AR expression was found to depend upon the tumour volume in C6 animals, but is independent on the time of tumour development. In F98 animals, a significant increase in A(1)AR receptor protein was found in the Peritumoural zone as a function of time of tumour development and tumour volume.
Collapse
Affiliation(s)
- Markus Dehnhardt
- Institute of Neuroscience and Biophysics 3-Medicine, Research Centre Juelich, 52425 Juelich, Germany.
| | | | | | | | | |
Collapse
|
46
|
Rustamzadeh E, Hall WA, Todhunter DA, Vallera VD, Low WC, Liu H, Panoskaltsis-Mortari A, Vallera DA. Intracranial therapy of glioblastoma with the fusion protein DTAT in immunodeficient mice. Int J Cancer 2007; 120:411-9. [PMID: 17075792 DOI: 10.1002/ijc.22278] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
A gene splicing technique was used to create a hybrid fusion protein DTAT encoding the 390 amino acid portion of diphtheria toxin (DT(390)), a linker, and the downstream 135-amino terminal fragment portion of human urokinase plasminogen activator. DTAT was assembled to target human glioblastoma cell lines in a murine intracranial model. Previously published in vitro studies demonstrated that DTAT was highly selective and toxic to human glioblastoma cell lines in a flank tumor model. The purpose of this study was to determine the toxicity, specificity and possible therapeutic efficacy of DTAT in an intracranial model. Convection enhanced delivery of DTAT resulted in about a 16-fold increase in maximum tolerated dose. Intracranial administration of DTAT on an every-other-day basis in nude mice with established U87 MG brain tumors resulted in significant reductions in tumor volume and significantly prolonged survival (p < 0.0001). Magnetic resonance imaging proved to be a powerful tool in mice and rats for demonstrating tumor growth in a xenograft intracranial model, assessing the efficacy of DTAT in tumor volume reduction and detecting DTAT-associated intracranial toxicity and vascular damage. These results suggest that the DTAT recombinant fusion protein is highly effective in an intracranial model and DTAT might be an effective treatment for glioblastoma.
Collapse
Affiliation(s)
- Edward Rustamzadeh
- Department of Neurosurgery, University of Minnesota Cancer Center, Minneapolis, MN 55455, USA
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Lobão-Soares B, Alvarez-Silva M, Mendes de Aguiar CBN, Nicolau M, Trentin AG. Undersulfation of glycosaminoglycans induced by sodium chlorate treatment affects the progression of C6 rat glioma, in-vivo. Brain Res 2007; 1131:29-36. [PMID: 17174944 DOI: 10.1016/j.brainres.2006.11.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2006] [Revised: 09/22/2006] [Accepted: 11/04/2006] [Indexed: 11/23/2022]
Abstract
The stimulatory input of extracellular matrix (ECM) components has been implicated in the invasive properties of glioma cells. It has been demonstrated that undersulfation of glycosaminoglycans (GAGs) promoted by sodium chlorate (SC) treatment reduces C6 glioma cell proliferation and adhesion to ECM molecules, in-vitro. In the present study, the authors investigated the involvement of GAG undersulfation in glioma cell growth in the brain parenchyma. The in-vitro treatment of C6 cells with SC and subsequent intracerebral inoculation in vehicle containing SC resulted in a reduced proportion of animals bearing glioma and a reduced tumor mass diameter. It also promoted longer animal survival. Intracerebral inoculation of SC-treated C6 cells in vehicle without SC or the SC treatment after intracerebral implantation of untreated C6 cells did not result in any reduction of tumor growth. Alterations in clinical, hematological and behavioral parameters in the open field were observed near the point of death when tumors presented a greater size. The results suggest an important role of GAGs in glioma growth which possibly affects cell proliferation and/or interactions with the normal tissue environment.
Collapse
Affiliation(s)
- Bruno Lobão-Soares
- Departamento de Biologia Celular, Embriologia e Genética, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Campus Universitário, 88040-900, Trindade, Florianópolis, S.C., Brazil
| | | | | | | | | |
Collapse
|
48
|
Yimam MA, Bui T, Ho RJY. Effects of lipid association on lomustine (CCNU) administered intracerebrally to syngeneic 36B-10 rat brain tumors. Cancer Lett 2006; 244:211-9. [PMID: 16455196 DOI: 10.1016/j.canlet.2005.12.024] [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] [Received: 01/17/2005] [Revised: 08/02/2005] [Accepted: 12/10/2005] [Indexed: 11/29/2022]
Abstract
A syngeneic, intracerebral rat brain tumor model was developed and characterized and then used to evaluate the therapeutic enhancement of lipid-associated 1-(2-chloroethyl)-3-cyclohexyl-1-nitrosourea (CCNU). The Fisher rat glioma cell 36B-10 (100,000-500,000 cells) was implanted intracranially to Fisher F-344 rats into the caudate nucleus. Animals treated with lipid-associated CCNU showed a 2- to 10-fold decrease in tumor size compared with animals treated with free CCNU, indicating that lipid association increases the therapeutic index of intracerebral CCNU treatment. Moreover, the syngeneic rat brain tumor model may be useful for evaluation of other therapeutic modalities.
Collapse
Affiliation(s)
- Mesfin A Yimam
- Department of Pharmaceutics, University of Washington, P.O. Box 357610 H272 Health Science Building, Seattle, WA 98195, USA
| | | | | |
Collapse
|
49
|
Spaeth N, Wyss MT, Pahnke J, Biollaz G, Trachsel E, Drandarov K, Treyer V, Weber B, Neri D, Buck A. Radioimmunotherapy targeting the extra domain B of fibronectin in C6 rat gliomas: a preliminary study about the therapeutic efficacy of iodine-131-labeled SIP(L19). Nucl Med Biol 2006; 33:661-6. [PMID: 16843841 DOI: 10.1016/j.nucmedbio.2006.05.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2006] [Revised: 05/03/2006] [Accepted: 05/08/2006] [Indexed: 10/22/2022]
Abstract
UNLABELLED Despite aggressive treatment protocols, patients suffering from glioblastoma multiforme still experience poor outcome. Therefore, new adjuvant therapeutic options such as radioimmunotherapy (RIT) have been studied and have resulted in significant survival benefit. In this study, we assessed the efficacy of a novel radioimmunotherapeutic approach targeting the extra domain B (EDB) of fibronectin, a marker of angiogenesis, in glioma-bearing rats. METHODS C6 gliomas were induced intracerebrally in Wistar rats. Ten to 11 days later, 220-360 MBq of iodine-131-labeled anti-EDB SIP(L19) ("small immunoprotein") was administered intravenously into nine animals, yielding a radiation dose of 13-21 Gy. Another nine rats served as controls. Then the following parameters were compared: median survival time, tumor size and histology. RESULTS Histological examination of the tumors revealed typical glioblastoma characteristics. Eleven of 18 rats developed a tumor size bigger than 150 mm(3). When these animals were used for survival analysis, median survival did significantly differ between groups [22 days (therapy; n=7) vs. 16 days (control; n=4); P<.0176]. CONCLUSIONS In this preliminary trial, (131)I-SIP(L19)-RIT showed promising potential in treating C6 gliomas, warranting further studies. However, larger trials with preferentially higher doses are needed to confirm this finding and, potentially, to further increase the efficacy of this treatment.
Collapse
Affiliation(s)
- Nicolas Spaeth
- PET Center, Division of Nuclear Medicine, University Hospital Zurich, 8091 Zurich, Switzerland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
This study emphasizes the importance of Rho/ROCK pathway in lovastatin-induced apoptosis as replenishment with exogenous isoprenoid, geranylgeranylpyrophosphate (GGPP), resulted in inhibition of apoptosis in cultured tumor cells. Treatment of C6 glioma cells with Toxin B and exoenzyme C3 resulted in cell death suggesting the role of geranylgeranylated protein(s) in the survival of glioma cells. Relative apoptotic death observed in cells transfected with dominant negative constructs of RhoA, Rac, and cdc42 imply Rho A as playing the major role in cell survival. Furthermore, the inhibition of Rho A kinase (ROCK), a direct downstream effector of Rho A, by Y-27632 or dominant negative of ROCK, induced apoptosis in glioma cells. These findings indicate that RhoA/ROCK pathway is involved negatively in the regulation of glioma cell death pathway. Moreover, in vivo studies of lovastatin treatment in animals implanted with C6 glioma cell tumors also resulted in smaller tumor size and induced apoptosis in the tumor tissue. The implantation of stably transfected C6 glioma cells with expression vector of C3 exoenzyme, dominant negative of RhoA and ROCK, resulted in significant smaller tumor mass, further establishing the importance of geranylgeranylated proteins, specifically RhoA and its downstream effecter ROCK, in cell survival and tumor genesis.
Collapse
Affiliation(s)
- R Rattan
- Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | | | | | | |
Collapse
|