1
|
De Vleeschauwer SI, van de Ven M, Oudin A, Debusschere K, Connor K, Byrne AT, Ram D, Rhebergen AM, Raeves YD, Dahlhoff M, Dangles-Marie V, Hermans ER. OBSERVE: guidelines for the refinement of rodent cancer models. Nat Protoc 2024; 19:2571-2596. [PMID: 38992214 DOI: 10.1038/s41596-024-00998-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 02/23/2024] [Indexed: 07/13/2024]
Abstract
Existing guidelines on the preparation (Planning Research and Experimental Procedures on Animals: Recommendations for Excellence (PREPARE)) and reporting (Animal Research: Reporting of In Vivo Experiments (ARRIVE)) of animal experiments do not provide a clear and standardized approach for refinement during in vivo cancer studies, resulting in the publication of generic methodological sections that poorly reflect the attempts made at accurately monitoring different pathologies. Compliance with the 3Rs guidelines has mainly focused on reduction and replacement; however, refinement has been harder to implement. The Oncology Best-practices: Signs, Endpoints and Refinements for in Vivo Experiments (OBSERVE) guidelines are the result of a European initiative supported by EurOPDX and INFRAFRONTIER, and aim to facilitate the refinement of studies using in vivo cancer models by offering robust and practical recommendations on approaches to research scientists and animal care staff. We listed cancer-specific clinical signs as a reference point and from there developed sets of guidelines for a wide variety of rodent models, including genetically engineered models and patient derived xenografts. In this Consensus Statement, we systematically and comprehensively address refinement and monitoring approaches during the design and execution of murine cancer studies. We elaborate on the appropriate preparation of tumor-initiating biologicals and the refinement of tumor-implantation methods. We describe the clinical signs to monitor associated with tumor growth, the appropriate follow-up of animals tailored to varying clinical signs and humane endpoints, and an overview of severity assessment in relation to clinical signs, implantation method and tumor characteristics. The guidelines provide oncology researchers clear and robust guidance for the refinement of in vivo cancer models.
Collapse
Affiliation(s)
| | - Marieke van de Ven
- Laboratory Animal Facility, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Anaïs Oudin
- NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Karlijn Debusschere
- Animal Core Facility VUB, Brussels, Belgium
- Core ARTH Animal Facilities, Medicine and Health Sciences Ghent University, Ghent, Belgium
| | - Kate Connor
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Annette T Byrne
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Doreen Ram
- Laboratory Animal Facility, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | | | | | - Maik Dahlhoff
- Institute of in vivo and in vitro Models, University of Veterinary Medicine Vienna, Vienna, Austria
| | | | - Els R Hermans
- Laboratory Animal Facility, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| |
Collapse
|
2
|
Xu P, Wang H, Pan H, Chen J, Deng C. Anlotinib combined with temozolomide suppresses glioblastoma growth via mediation of JAK2/STAT3 signaling pathway. Cancer Chemother Pharmacol 2022; 89:183-196. [PMID: 34997858 PMCID: PMC8807469 DOI: 10.1007/s00280-021-04380-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 11/22/2021] [Indexed: 12/12/2022]
Abstract
Purpose Anlotinib protects against carcinogenesis through the induction of autophagy and apoptosis. The current study evaluated the role and molecular mechanisms of anlotinib in glioblastoma, and the effects of anlotinib in combination with temozolomide (TMZ). Methods Cell Counting Kit-8 and colony-forming assays were used to evaluate cell viability. Cell migration and invasion were assessed by wound-healing, Transwell migration, and Matrigel invasion assays. Cellular apoptosis and cell cycle analysis were determined by flow cytometry. Angiogenesis was assessed using human umbilical vein endothelial cells (HUVECs). Vascular endothelial growth factor A (VEGFA) was measured by enzyme-linked immunosorbent assay. Protein expression was determined by western blotting or immunofluorescence staining. The in vivo anti-glioblastoma effect was assessed with live imaging of tumor xenografts in nude mice. Results Anlotinib restricted the proliferation, migration, and invasion of glioblastoma cells in a dose-dependent manner. Tumor supernatant from glioblastoma cells treated with anlotinib inhibited angiogenesis in HUVECs. Anlotinib induced autophagy in glioblastoma cells by increasing Beclin-1 and microtubule-associated protein 1 light chain 3B (LC3B) levels. Mechanistically, anlotinib inhibited the Janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3)/VEGFA signaling pathway. STAT3 inhibition by S3I-201 decreased VEGFA and suppressed cellular proliferation and movement. TMZ enhanced the anti-glioblastoma ability of anlotinib. Finally, anlotinib inhibited tumor growth and JAK2/STAT3/VEGFA signaling in xenografts. Conclusion Anlotinib exerts anti-glioblastoma activity possibly through the JAK2/STAT3/VEGFA signaling pathway. TMZ potentiated the anti-glioblastoma effect of anlotinib via the same signaling pathway, indicating the potential application of anlotinib as a treatment option for glioblastoma.
Collapse
Affiliation(s)
- Peng Xu
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Southeast University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, China
| | - Handong Wang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Southeast University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, China.
| | - Hao Pan
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Southeast University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, China.
| | - Jiakai Chen
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Chulei Deng
- Department of Neurosurgery, Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing, 210002, China
| |
Collapse
|
3
|
Sousa JFD, Serafim RB, Freitas LMD, Fontana CR, Valente V. DNA repair genes in astrocytoma tumorigenesis, progression and therapy resistance. Genet Mol Biol 2019; 43:e20190066. [PMID: 31930277 PMCID: PMC7198033 DOI: 10.1590/1678-4685-gmb-2019-0066] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 07/21/2019] [Indexed: 12/26/2022] Open
Abstract
Glioblastoma (GBM) is the most common and malignant type of primary brain tumor,
showing rapid development and resistance to therapies. On average, patients
survive 14.6 months after diagnosis and less than 5% survive five years or more.
Several pieces of evidence have suggested that the DNA damage signaling and
repair activities are directly correlated with GBM phenotype and exhibit
opposite functions in cancer establishment and progression. The functions of
these pathways appear to present a dual role in tumorigenesis and cancer
progression. Activation and/or overexpression of ATRX, ATM and RAD51 genes were
extensively characterized as barriers for GBM initiation, but paradoxically the
exacerbated activity of these genes was further associated with cancer
progression to more aggressive stages. Excessive amounts of other DNA repair
proteins, namely HJURP, EXO1, NEIL3, BRCA2, and BRIP, have also been connected
to proliferative competence, resistance and poor prognosis. This scenario
suggests that these networks help tumor cells to manage replicative stress and
treatment-induced damage, diminishing genome instability and conferring therapy
resistance. Finally, in this review we address promising new drugs and
therapeutic approaches with potential to improve patient survival. However,
despite all technological advances, the prognosis is still dismal and further
research is needed to dissect such complex mechanisms.
Collapse
Affiliation(s)
- Juliana Ferreira de Sousa
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, U.S.A
| | - Rodolfo Bortolozo Serafim
- Universidade de São Paulo, Faculdade de Medicina de Ribeirão Preto, Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Universidade de São Paulo, Ribeirão Preto, SP, Brazil
| | - Laura Marise de Freitas
- Universidade de São Paulo, Instituto de Química, Departamento de Bioquímica, São Paulo, SP, Brazil
| | - Carla Raquel Fontana
- Universidade Estadual Paulista "Júlio de Mesquita Filho" (UNESP), Faculdade de Ciências Farmacêuticas, Departamento de Análises Clínicas, Araraquara, SP, Brazil
| | - Valeria Valente
- Universidade de São Paulo, Faculdade de Medicina de Ribeirão Preto, Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Universidade de São Paulo, Ribeirão Preto, SP, Brazil.,Universidade Estadual Paulista "Júlio de Mesquita Filho" (UNESP), Faculdade de Ciências Farmacêuticas, Departamento de Análises Clínicas, Araraquara, SP, Brazil.,Centro de Terapia Celular (CEPID-FAPESP), Ribeirão Preto, SP, Brazil
| |
Collapse
|
4
|
[ 18F]FET PET is a useful tool for treatment evaluation and prognosis prediction of anti-angiogenic drug in an orthotopic glioblastoma mouse model. Lab Anim Res 2019; 34:248-256. [PMID: 30671112 PMCID: PMC6333614 DOI: 10.5625/lar.2018.34.4.248] [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: 10/22/2018] [Revised: 12/04/2018] [Accepted: 12/07/2018] [Indexed: 11/21/2022] Open
Abstract
O-2-18F-fluoroethyl-l-tyrosine ([18F]FET) has been widely used for glioblastomas (GBM) in clinical practice, although evaluation of its applicability in non-clinical research is still lacking. The objective of this study was to examine the value of [18F]FET for treatment evaluation and prognosis prediction of anti-angiogenic drug in an orthotopic mouse model of GBM. Human U87MG cells were implanted into nude mice and then bevacizumab, a representative anti-angiogenic drug, was administered. We monitored the effect of anti-angiogenic agents using multiple imaging modalities, including bioluminescence imaging (BLI), magnetic resonance imaging (MRI), and positron emission tomography-computed tomography (PET/CT). Among these imaging methods analyzed, only [18F]FET uptake showed a statistically significant decrease in the treatment group compared to the control group (P=0.02 and P=0.03 at 5 and 20 mg/kg, respectively). This indicates that [18F]FET PET is a sensitive method to monitor the response of GBM bearing mice to anti-angiogenic drug. Moreover, [18F]FET uptake was confirmed to be a significant parameter for predicting the prognosis of anti-angiogenic drug (P=0.041 and P=0.007, on Days 7 and 12, respectively, on Pearson's correlation; P=0.048 and P=0.030, on Days 7 and 12, respectively, on Cox regression analysis). However, results of BLI or MRI were not significantly associated with survival time. In conclusion, this study suggests that [18F]FET PET imaging is a pertinent imaging modality for sensitive monitoring and accurate prediction of treatment response to anti-angiogenic agents in an orthotopic model of GBM.
Collapse
|
5
|
Li C, Tan J, Chang J, Li W, Liu Z, Li N, Ji Y. Radioiodine-labeled anti-epidermal growth factor receptor binding bovine serum albumin-polycaprolactone for targeting imaging of glioblastoma. Oncol Rep 2017; 38:2919-2926. [DOI: 10.3892/or.2017.5937] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 08/02/2017] [Indexed: 11/06/2022] Open
|
6
|
Barboza T, Gomes T, Mizurini DM, Monteiro RQ, König S, Francischetti IMB, Signoretti PVP, Ramos IP, Gutfilen B, Souza SAL. (99m)Tc-ixolaris targets glioblastoma-associated tissue factor: in vitro and pre-clinical applications. Thromb Res 2015; 136:432-9. [PMID: 26070446 DOI: 10.1016/j.thromres.2015.05.032] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Revised: 04/18/2015] [Accepted: 05/30/2015] [Indexed: 11/18/2022]
Abstract
BACKGROUND The clotting initiator protein tissue factor (TF) has recently been described as a potential target that can be exploited to image aggressive tumors. Ixolaris is a specific TF inhibitor that blocks tumor cell procoagulant activity and tumor growth. OBJECTIVE Herein we evaluated the ability of (99m)Tc-ixolaris to target tumor-derived TF using an orthotopic glioblastoma (GBM) model in mice. METHODS The right forebrains of Swiss mice were stereotactically inoculated with U87-MG human GBM cells. Histological and immunohistochemical analyses were performed on the resulting tumors after 35-45 days. The biodistribution of (99m)Tc-ixolaris was evaluated by semi-quantitative whole-body scintigraphy and a quantitative analysis of radioactivity in isolated organs. RESULTS No (99m)Tc-ixolaris uptake was observed in brain of tumor-free mice, independently of the integrity of brain-blood barrier. In contrast, the presence of TF-expressing brain tumor masses determined a significant (99m)Tc-ixolaris uptake. CONCLUSION (99m)Tc-ixolaris recognized TF-expressing GBM cells in vivo. Given the proposed role of TF in tumor progression, (99m)Tc-ixolaris is a promising radiopharmaceutical agent for quantifying cancer-associated TF in aggressive tumors, including GBM.
Collapse
Affiliation(s)
- Thiago Barboza
- Laboratório de Marcação de Células e Moléculas, Departamento de Radiologia, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Tainá Gomes
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil; Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Daniella M Mizurini
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Robson Q Monteiro
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Sandra König
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Ivo M B Francischetti
- Vector Biology Section, Laboratory of Malaria and Vector Research, National Institutes of Health, Bethesda, MD, USA
| | - Paula V P Signoretti
- Laboratório de Físico-Química Biológica Aída Hassón Voloch e Laboratório Intermediário de Biomembranas, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Isalira P Ramos
- Laboratório de Cardiologia Celular e Molecular, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Bianca Gutfilen
- Laboratório de Marcação de Células e Moléculas, Departamento de Radiologia, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Sergio A L Souza
- Laboratório de Marcação de Células e Moléculas, Departamento de Radiologia, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil.
| |
Collapse
|
7
|
Lu-Emerson C, Duda DG, Emblem KE, Taylor JW, Gerstner ER, Loeffler JS, Batchelor TT, Jain RK. Lessons from anti-vascular endothelial growth factor and anti-vascular endothelial growth factor receptor trials in patients with glioblastoma. J Clin Oncol 2015; 33:1197-213. [PMID: 25713439 PMCID: PMC4517055 DOI: 10.1200/jco.2014.55.9575] [Citation(s) in RCA: 140] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Treatment of glioblastoma (GBM), the most common primary malignant brain tumor in adults, remains a significant unmet need in oncology. Historically, cytotoxic treatments provided little durable benefit, and tumors recurred within several months. This has spurred a substantial research effort to establish more effective therapies for both newly diagnosed and recurrent GBM. In this context, antiangiogenic therapy emerged as a promising treatment strategy because GBMs are highly vascular tumors. In particular, GBMs overexpress vascular endothelial growth factor (VEGF), a proangiogenic cytokine. Indeed, many studies have demonstrated promising radiographic response rates, delayed tumor progression, and a relatively safe profile for anti-VEGF agents. However, randomized phase III trials conducted to date have failed to show an overall survival benefit for antiangiogenic agents alone or in combination with chemoradiotherapy. These results indicate that antiangiogenic agents may not be beneficial in unselected populations of patients with GBM. Unfortunately, biomarker development has lagged behind in the process of drug development, and no validated biomarker exists for patient stratification. However, hypothesis-generating data from phase II trials that reveal an association between increased perfusion and/or oxygenation (ie, consequences of vascular normalization) and survival suggest that early imaging biomarkers could help identify the subset of patients who most likely will benefit from anti-VEGF agents. In this article, we discuss the lessons learned from the trials conducted to date and how we could potentially use recent advances in GBM biology and imaging to improve outcomes of patients with GBM who receive antiangiogenic therapy.
Collapse
Affiliation(s)
- Christine Lu-Emerson
- All authors, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA
| | - Dan G Duda
- All authors, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA
| | - Kyrre E Emblem
- All authors, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA
| | - Jennie W Taylor
- All authors, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA
| | - Elizabeth R Gerstner
- All authors, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA
| | - Jay S Loeffler
- All authors, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA
| | - Tracy T Batchelor
- All authors, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA
| | - Rakesh K Jain
- All authors, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA.
| |
Collapse
|
8
|
Liu SY, Chiang MF, Chen YJ. Role of WW domain proteins WWOX in development, prognosis, and treatment response of glioma. Exp Biol Med (Maywood) 2014; 240:315-23. [PMID: 25432984 DOI: 10.1177/1535370214561588] [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] [Indexed: 12/26/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most aggressive and malignant brain tumor. Delicate microenvironment and lineage heterogeneity of GBM cells including infiltration, hypoxia, angiogenesis, and stemness make them highly resistant to current conventional therapies, with an average life expectancy for GBM patients of less than 15 months. Poor response to cytotoxic agents of GBM cells remains the major challenge of GBM treatment. Resistance of GBM to clinical treatment is a result of genomic alternation and deregulated signaling pathways, such as p53 mutation and apoptosis signaling blockage, providing cancer cells more opportunities for survival rather than cell death. WW domain-containing oxidoreductase (WWOX) is a tumor suppressor gene, commonly downregulated in various types of tumors, including GBM. It has been found that the reintroduction of WWOX induced p53-mutant GBM cells to undergo apoptosis, but not in p53 wild-type GBM cells, indicating WWOX is likely to reopen apoptosis pathways in a p53-independent manner in GBM. Identifying the crucial target modulated by WWOX deficiency provides a potential therapeutic target for GBM treatment. Here, we have reviewed the literatures about the role of WWOX in development, signaling pathway, prognosis, and treatment response in malignant glioma.
Collapse
Affiliation(s)
- Shin-Yi Liu
- Department of Medical Research, Mackay Memorial Hospital, Taipei 104, Taiwan
| | - Ming-Fu Chiang
- Department of Neurosurgery, Mackay Memorial Hospital, Taipei 104, Taiwan Graduate Institute of Injury Prevention and Control, Taipei Medical University, Taipei 110, Taiwan
| | - Yu-Jen Chen
- Department of Radiation Oncology, Mackay Memorial Hospital, Taipei 104, Taiwan Graduate Institute of Pharmacology, Taipei Medical University, Taipei 110, Taiwan
| |
Collapse
|
9
|
Interrogation of gossypol therapy in glioblastoma implementing cell line and patient-derived tumour models. Br J Cancer 2014; 111:2275-86. [PMID: 25375271 PMCID: PMC4264441 DOI: 10.1038/bjc.2014.529] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Revised: 09/07/2014] [Accepted: 09/08/2014] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Glioblastoma (GBM), being a highly vascularised and locally invasive tumour, is an attractive target for anti-angiogenic and anti-invasive therapies. The GBM/endothelial cell response to gossypol/temozolomide (TMZ) treatment was investigated with a particular aim to assess treatment effects on cancer hallmarks. METHODS Cell viability, endothelial tube formation and GBM tumour cell invasion were variously assessed following combined treatment in vitro. The U87MG-luc2 subcutaneous xenograft model was used to investigate therapeutic response in vivo. Viable tumour response to treatment was interrogated using immunohistochemistry. Combined treatment protocols were also tested in primary GBM patient-derived cultures. RESULTS An endothelial/GBM cell viability inhibitory effect, as well as an anti-angiogenic and anti-invasive response, to combined treatment have been demonstrated in vitro. A significantly greater anti-proliferative (P=0.020, P=0.030), anti-angiogenic (P=0.040, P<0.0001) and pro-apoptotic (P=0.0083, P=0.0149) response was observed when combined treatment was compared with single gossypol/TMZ treatment response, respectively. GBM cell line and patient-specific response to gossypol/TMZ treatment was observed. CONCLUSIONS Our results indicate that response to a combined gossypol/TMZ treatment is related to inhibition of tumour-associated angiogenesis, invasion and proliferation and warrants further investigation as a novel targeted GBM treatment strategy.
Collapse
|
10
|
Nakano I. Therapeutic potential of targeting glucose metabolism in glioma stem cells. Expert Opin Ther Targets 2014; 18:1233-6. [PMID: 25077882 DOI: 10.1517/14728222.2014.944899] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Glioblastoma is a highly lethal cancer. Glioma stem cells (GSCs) are potentially an attractive therapeutic target and eradication of GSCs may impact tumor growth and sensitize tumors to conventional therapies. The brain is one of the most metabolically active organs with glucose representing the most important, but not the only, source of energy and carbon. Like all other cancers, glioblastoma requires a continuous source of energy and molecular resources for new cell production with a preferential use of aerobic glycolysis, recognized as the Warburg effect. As selected metabolic nodes are amenable to therapeutic targeting, we observed that the Warburg effect may causally contribute to glioma heterogeneity. This Editorial summarizes recent studies that examine the relationship between GSCs and metabolism and briefly provides our views for the future directions. The ultimate goal is to establish a new concept by incorporating both the cellular hierarchical theory and the cellular evolution theory to explain tumor heterogeneity. Such concept may better elucidate the mechanisms of how tumors gain cellular and molecular complexity and guide us develop novel and effective targeted therapies.
Collapse
Affiliation(s)
- Ichiro Nakano
- Associate Professor, Director of Neural Cancer Stem Cell Program,The Ohio State University, James Comprehensive Cancer Center, Department of Neurological Surgery , 385 Wiseman Hall, 400 W 12th St., Columbus, OH 43210 , USA +1 614 292 0358 ; +1 614 688 4882 ;
| |
Collapse
|
11
|
Heiss WD. Clinical Impact of Amino Acid PET in Gliomas. J Nucl Med 2014; 55:1219-20. [DOI: 10.2967/jnumed.114.142661] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 06/23/2014] [Indexed: 01/23/2023] Open
|
12
|
PARPi-FL--a fluorescent PARP1 inhibitor for glioblastoma imaging. Neoplasia 2014; 16:432-40. [PMID: 24970386 PMCID: PMC4198695 DOI: 10.1016/j.neo.2014.05.005] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Accepted: 05/13/2014] [Indexed: 11/23/2022] Open
Abstract
New intravital optical imaging technologies have revolutionized our understanding of mammalian biology and continue to evolve rapidly. However, there are only a limited number of imaging probes available to date. In this study, we investigated in mouse models of glioblastoma whether a fluorescent small molecule inhibitor of the DNA repair enzyme PARP1, PARPi-FL, can be used as an imaging agent to detect glioblastomas in vivo. We demonstrated that PARPi-FL has appropriate biophysical properties, low toxicity at concentrations used for imaging, high stability in vivo, and accumulates selectively in glioblastomas due to high PARP1 expression. Importantly, subcutaneous and orthotopic glioblastoma xenografts were imaged with high contrast clearly defining tumor tissue from normal surrounding tissue. This research represents a step toward exploring and developing PARPi-FL as an optical intraoperative imaging agent for PARP1 in the clinic.
Collapse
|
13
|
Heiss WD. [PET in gliomas. Overview of current studies]. Nuklearmedizin 2014; 53:163-71; quiz N32. [PMID: 24853278 DOI: 10.3413/nukmed-0662-14-04] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 05/20/2014] [Indexed: 11/20/2022]
Abstract
Gliomas which represent 30% of intracranial tumours are morphologic lesions and therefore CT and MRI are the first line diagnostic procedures with MRI giving better soft tissue resolution and permitting additional functional information. These mainly morphologic imaging modalities yield only restricted information on grade of malignancy, on infiltration into and effects on surrounding brain tissue, on differentiation between necrotic and recurrent tumour, on prognosis and on efficacy of treatment. Information on these important issues for patient management can be obtained by PET-studies of glucose metabolism with FDG, of aminoacid-uptake and protein synthesis with 11C-methionin, 18F-fluorethyltyrosin and 18F-fluor-deoxyphenylalanin and of proliferation by 18F-deoxythymidin. With the increasing availability of 18F-tracers PET has obtained wider spread clinical application. In all these applications a coregistration with morphologic imaging should be obtained, and for that purpose hybrid installations (PET-MR) are already being used.
Collapse
Affiliation(s)
- W-D Heiss
- Prof. Dr. W.-D. Heiss, Max-Planck-Institut für neurologische Forschung, Gleueler Str. 50, 50931 Köln, Tel. 02 21/472 62 20, Fax 02 21/472 63 49, E-Mail:
| |
Collapse
|
14
|
Early monitoring antiangiogenesis treatment response of Sunitinib in U87MG Tumor Xenograft by (18)F-FLT MicroPET/CT imaging. BIOMED RESEARCH INTERNATIONAL 2014; 2014:218578. [PMID: 24860813 PMCID: PMC4000939 DOI: 10.1155/2014/218578] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 03/19/2014] [Indexed: 11/17/2022]
Abstract
AIM It was aimed to monitor early treatment response of Sunitinib in U87MG models mimicking glioblastoma multiforme by longitudinal (18)F-FLT microPET/CT imaging in this study. METHODS U87MG tumor mice were intragastrically injected with Sunitinib at a dose of 80 mg/kg for consecutive 7 days. (18)F-FLT microPET/CT scans were acquired on days 0, 1, 3, 7, and 13 after therapy. Tumor sizes and body weight were measured. Tumor samples were collected for immunohistochemical analysis of proliferation and microvessel density (MVD) with anti-Ki67 and anti-CD31, respectively. RESULTS The uptake ratios of tumor to the contralateral muscle (T/M) of (18)F-FLT in the Sunitinib group decreased from baseline to day 3 (T/M0 = 2.98 ± 0.33; T/M3 = 2.23 ± 0.36; P < 0.001), reached the bottom on day 7 (T/M7 = 1.96 ± 0.35; P < 0.001), and then recovered on day 13. The T/M of (18)F-FLT uptake in the control group remained around 3.0. There was no difference for the tumor size between both groups until day 11. (18)F-FLT uptakes of tumor were correlated with Ki67 staining index and MVD. CONCLUSION Early therapy response to Sunitinib could be predicted via (18)F-FLT PET, which will contribute to monitoring antiangiogenesis treatment.
Collapse
|