1
|
Kanvinde PP, Malla AP, Connolly NP, Szulzewsky F, Anastasiadis P, Ames HM, Kim AJ, Winkles JA, Holland EC, Woodworth GF. Leveraging the replication-competent avian-like sarcoma virus/tumor virus receptor-A system for modeling human gliomas. Glia 2021; 69:2059-2076. [PMID: 33638562 PMCID: PMC8591561 DOI: 10.1002/glia.23984] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/15/2021] [Accepted: 02/16/2021] [Indexed: 12/20/2022]
Abstract
Gliomas are the most common primary intrinsic brain tumors occurring in adults. Of all malignant gliomas, glioblastoma (GBM) is considered the deadliest tumor type due to diffuse brain invasion, immune evasion, cellular, and molecular heterogeneity, and resistance to treatments resulting in high rates of recurrence. An extensive understanding of the genomic and microenvironmental landscape of gliomas gathered over the past decade has renewed interest in pursuing novel therapeutics, including immune checkpoint inhibitors, glioma-associated macrophage/microglia (GAMs) modulators, and others. In light of this, predictive animal models that closely recreate the conditions and findings found in human gliomas will serve an increasingly important role in identifying new, effective therapeutic strategies. Although numerous syngeneic, xenograft, and transgenic rodent models have been developed, few include the full complement of pathobiological features found in human tumors, and therefore few accurately predict bench-to-bedside success. This review provides an update on how genetically engineered rodent models based on the replication-competent avian-like sarcoma (RCAS) virus/tumor virus receptor-A (tv-a) system have been used to recapitulate key elements of human gliomas in an immunologically intact host microenvironment and highlights new approaches using this model system as a predictive tool for advancing translational glioma research.
Collapse
Affiliation(s)
- Pranjali P Kanvinde
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, USA.,Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Adarsha P Malla
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, USA.,Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Nina P Connolly
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, USA.,Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Frank Szulzewsky
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Pavlos Anastasiadis
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, USA.,Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Heather M Ames
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, USA.,Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Anthony J Kim
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, USA.,Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Jeffrey A Winkles
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, USA.,Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, USA.,Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA.,Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Eric C Holland
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Seattle Tumor Translational Research Center, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Graeme F Woodworth
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, USA.,Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
2
|
Pigeon H, Pérès EA, Truillet C, Jego B, Boumezbeur F, Caillé F, Zinnhardt B, Jacobs AH, Le Bihan D, Winkeler A. TSPO-PET and diffusion-weighted MRI for imaging a mouse model of infiltrative human glioma. Neuro Oncol 2020; 21:755-764. [PMID: 30721979 DOI: 10.1093/neuonc/noz029] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Glioblastoma (GBM) is the most devastating brain tumor. Despite the use of multimodal treatments, most patients relapse, often due to the highly invasive nature of gliomas. However, the detection of glioma infiltration remains challenging. The aim of this study was to assess advanced PET and MRI techniques for visualizing biological activity and infiltration of the tumor. METHODS Using multimodality imaging, we investigated [18F]DPA-714, a radiotracer targeting the 18 kDa translocator protein (TSPO), [18F]FET PET, non-Gaussian diffusion MRI (apparent diffusion coefficient, kurtosis), and the S-index, a composite diffusion metric, to detect tumor infiltration in a human invasive glioma model. In vivo imaging findings were confirmed by autoradiography and immunofluorescence. RESULTS Increased tumor-to-contralateral [18F]DPA-714 uptake ratios (1.49 ± 0.11) were found starting 7 weeks after glioma cell implantation. TSPO-PET allowed visualization of glioma infiltration into the contralateral hemisphere 2 weeks earlier compared with the clinically relevant biomarker for biological glioma activity [18F]FET. Diffusion-weighted imaging (DWI), in particular kurtosis, was more sensitive than standard T2-weighted MRI to detect differences between the glioma-bearing and the contralateral hemisphere at 5 weeks. Immunofluorescence data reflect in vivo findings. Interestingly, labeling for tumoral and stromal TSPO indicates a predominant expression of TSPO by tumor cells. CONCLUSION These results suggest that advanced PET and MRI methods, such as [18F]DPA-714 and DWI, may be superior to standard imaging methods to visualize glioma growth and infiltration at an early stage.
Collapse
Affiliation(s)
- Hayet Pigeon
- UMR 1023, IMIV, Service Hospitalier Frédéric Joliot, CEA, Inserm, Université Paris Sud, CNRS, Université Paris-Saclay, Orsay, France
| | - Elodie A Pérès
- UMR 1023, IMIV, Service Hospitalier Frédéric Joliot, CEA, Inserm, Université Paris Sud, CNRS, Université Paris-Saclay, Orsay, France.,NeuroSpin, CEA/Université Paris-Saclay, Gif sur Yvette, France.,Normandie Université, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, Caen, France
| | - Charles Truillet
- UMR 1023, IMIV, Service Hospitalier Frédéric Joliot, CEA, Inserm, Université Paris Sud, CNRS, Université Paris-Saclay, Orsay, France
| | - Benoit Jego
- UMR 1023, IMIV, Service Hospitalier Frédéric Joliot, CEA, Inserm, Université Paris Sud, CNRS, Université Paris-Saclay, Orsay, France
| | | | - Fabien Caillé
- UMR 1023, IMIV, Service Hospitalier Frédéric Joliot, CEA, Inserm, Université Paris Sud, CNRS, Université Paris-Saclay, Orsay, France
| | - Bastian Zinnhardt
- EIMI and Department of Nuclear Medicine, University Hospital Münster, Westfälische Wilhelms University Münster, Münster, Germany
| | - Andreas H Jacobs
- EIMI and Department of Nuclear Medicine, University Hospital Münster, Westfälische Wilhelms University Münster, Münster, Germany.,Department of Geriatrics, Johanniter Hospital, Evangelische Kliniken, Bonn, Germany
| | - Denis Le Bihan
- NeuroSpin, CEA/Université Paris-Saclay, Gif sur Yvette, France
| | - Alexandra Winkeler
- UMR 1023, IMIV, Service Hospitalier Frédéric Joliot, CEA, Inserm, Université Paris Sud, CNRS, Université Paris-Saclay, Orsay, France
| |
Collapse
|
3
|
Bongers A, Hau E, Shen H. Short Diffusion Time Diffusion-Weighted Imaging With Oscillating Gradient Preparation as an Early Magnetic Resonance Imaging Biomarker for Radiation Therapy Response Monitoring in Glioblastoma: A Preclinical Feasibility Study. Int J Radiat Oncol Biol Phys 2018; 102:1014-1023. [DOI: 10.1016/j.ijrobp.2017.12.280] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 11/21/2017] [Accepted: 12/19/2017] [Indexed: 12/24/2022]
|
4
|
Bastiancich C, Bastiat G, Lagarce F. Gemcitabine and glioblastoma: challenges and current perspectives. Drug Discov Today 2017; 23:416-423. [PMID: 29074439 DOI: 10.1016/j.drudis.2017.10.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 09/22/2017] [Accepted: 10/12/2017] [Indexed: 12/15/2022]
Abstract
Gemcitabine is a nucleoside analog currently used for the treatment of various solid tumors as a single agent or in combination with other chemotherapeutic drugs. Its use against highly aggressive brain tumors (glioblastoma) has been evaluated in preclinical and clinical trials leading to controversial results. Gemcitabine can inhibit DNA chain elongation, is a potent radiosensitizer and it can enhance antitumor immune activity, but it also presents some drawbacks (e.g., short half-life, side effects, chemoresistance). The aim of this review is to discuss the challenges related to the use of gemcitabine for glioblastoma and to report recent studies that suggest overcoming these obstacles opening new perspectives for its use in the field (e.g., gemcitabine derivatives and/or nanomedicines).
Collapse
Affiliation(s)
- Chiara Bastiancich
- MINT, UNIV Angers, INSERM 1066, CNRS 6021, Université Bretagne Loire, Angers, France; Université Catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Brussels, Belgium
| | - Guillaume Bastiat
- MINT, UNIV Angers, INSERM 1066, CNRS 6021, Université Bretagne Loire, Angers, France
| | - Frederic Lagarce
- MINT, UNIV Angers, INSERM 1066, CNRS 6021, Université Bretagne Loire, Angers, France; Pharmacy Department, CHU Angers, Angers University Hospital, France.
| |
Collapse
|
5
|
Castle KD, Chen M, Wisdom AJ, Kirsch DG. Genetically engineered mouse models for studying radiation biology. Transl Cancer Res 2017; 6:S900-S913. [PMID: 30733931 PMCID: PMC6363345 DOI: 10.21037/tcr.2017.06.19] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Genetically engineered mouse models (GEMMs) are valuable research tools that have transformed our understanding of cancer. The first GEMMs generated in the 1980s and 1990s were knock-in and knock-out models of single oncogenes or tumor suppressors. The advances that made these models possible catalyzed both technological and conceptual shifts in the way cancer research was conducted. As a result, dozens of mouse models of cancer exist today, covering nearly every tissue type. The advantages inherent to GEMMs compared to in vitro and in vivo transplant models are compounded in preclinical radiobiology research for several reasons. First, they accurately and robustly recapitulate primary cancers anatomically, histopathologically, and genetically. Reliable models are a prerequisite for predictive preclinical studies. Second, they preserve the tumor microenvironment, including the immune, vascular, and stromal compartments, which enables the study of radiobiology at a systems biology level. Third, they provide exquisite control over the genetics and kinetics of tumor initiation, which enables the study of specific gene mutations on radiation response and functional genomics in vivo. Taken together, these facets allow researchers to utilize GEMMs for rigorous and reproducible preclinical research. In the three decades since the generation of the first GEMMs of cancer, advancements in modeling approaches have rapidly progressed and expanded the mouse modeling toolbox with techniques such as in vivo short hairpin RNA (shRNA) knockdown, inducible gene expression, site-specific recombinases, and dual recombinase systems. Our lab and many others have utilized these tools to study cancer and radiobiology. Recent advances in genome engineering with CRISPR/Cas9 technology have made GEMMs even more accessible to researchers. Here, we review current and future approaches to mouse modeling with a focus on applications in preclinical radiobiology research.
Collapse
Affiliation(s)
- Katherine D. Castle
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA
| | - Mark Chen
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA
- Medical Scientist Training Program, Duke University Medical Center, Durham, North Carolina, USA
| | - Amy J. Wisdom
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA
- Medical Scientist Training Program, Duke University Medical Center, Durham, North Carolina, USA
| | - David G. Kirsch
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
6
|
Lai YL, Wu CY, Chao KSC. Biological imaging in clinical oncology: radiation therapy based on functional imaging. Int J Clin Oncol 2016; 21:626-632. [PMID: 27384183 DOI: 10.1007/s10147-016-1000-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 05/29/2016] [Indexed: 12/25/2022]
Abstract
Radiation therapy is one of the most effective tools for cancer treatment. In recent years, intensity-modulated radiation therapy has become increasingly popular in that target dose-escalation can be done while sparing adjacent normal tissues. For this reason, the development of measures to pave the way for accurate target delineation is of great interest. With the integration of functional information obtained by biological imaging with radiotherapy, strategies using advanced biological imaging to visualize metabolic pathways and to improve therapeutic index and predict treatment response are discussed in this article.
Collapse
Affiliation(s)
- Yo-Liang Lai
- Department of Radiation Oncology, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Chun-Yi Wu
- Department of Biomedical Imaging and Radiological Science, China Medical University, Taichung, Taiwan
| | - K S Clifford Chao
- China Medical University, 91 Hsueh-Shih Road, Taichung, 40402, Taiwan.
| |
Collapse
|
7
|
Lauroyl-gemcitabine-loaded lipid nanocapsule hydrogel for the treatment of glioblastoma. J Control Release 2016; 225:283-93. [DOI: 10.1016/j.jconrel.2016.01.054] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 01/25/2016] [Accepted: 01/27/2016] [Indexed: 12/19/2022]
|
8
|
Kim MM, Camelo-Piragua S, Schipper M, Tao Y, Normolle D, Junck L, Mammoser A, Betz BL, Cao Y, Kim CJ, Heth J, Sagher O, Lawrence TS, Tsien CI. Gemcitabine Plus Radiation Therapy for High-Grade Glioma: Long-Term Results of a Phase 1 Dose-Escalation Study. Int J Radiat Oncol Biol Phys 2015; 94:305-11. [PMID: 26853339 DOI: 10.1016/j.ijrobp.2015.10.032] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 09/15/2015] [Accepted: 10/20/2015] [Indexed: 01/25/2023]
Abstract
PURPOSE To evaluate the tolerability and efficacy of gemcitabine plus radiation therapy (RT) in this phase 1 study of patients with newly diagnosed malignant glioma (HGG). PATIENTS AND METHODS Between 2004 and 2012, 29 adults with HGG were enrolled. After any extent of resection, RT (60 Gy over 6 weeks) was given concurrent with escalating doses of weekly gemcitabine. Using a time-to-event continual reassessment method, 5 dose levels were evaluated starting at 500 mg/m(2) during the last 2 weeks of RT and advanced stepwise into earlier weeks. The primary objective was to determine the recommended phase 2 dose of gemcitabine plus RT. Secondary objectives included progression-free survival, overall survival (OS), and long-term toxicity. RESULTS Median follow-up was 38.1 months (range, 8.9-117.5 months); 24 patients were evaluable for toxicity. After 2005 when standard practice changed, patients with World Health Organization grade 4 tumors were no longer enrolled. Median progression-free survival for 22 patients with grade 3 tumors was 26.0 months (95% confidence interval [CI] 15.6-inestimable), and OS was 48.5 months (95% CI 26.8-inestimable). In 4 IDH mutated, 1p/19q codeleted patients, no failures occurred, with all but 1 alive at time of last follow-up. Seven with IDH mutated, non-codeleted tumors with ATRX loss had intermediate OS of 73.5 months (95% CI 32.8-inestimable). Six nonmutated, non-codeleted patients had a median OS of 26.5 months (95% CI 25.4-inestimable). The recommended phase 2 dose of gemcitabine plus RT was 750 mg/m(2)/wk given the last 4 weeks of RT. Dose reductions were most commonly due to grade 3 neutropenia; no grade 4 or 5 toxicities were seen. CONCLUSIONS Gemcitabine concurrent with RT is well-tolerated and yields promising outcomes, including in patients with adverse molecular features. It is a candidate for further study, particularly for poor-prognosis patient subgroups with HGG.
Collapse
Affiliation(s)
- Michelle M Kim
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan.
| | | | - Matthew Schipper
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan; Department of Biostatistics, University of Michigan, Ann Arbor, Michigan
| | - Yebin Tao
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan; Department of Biostatistics, University of Michigan, Ann Arbor, Michigan
| | - Daniel Normolle
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Larry Junck
- Department of Neurology, University of Michigan, Ann Arbor, Michigan
| | - Aaron Mammoser
- Department of Neurology, University of Michigan, Ann Arbor, Michigan
| | - Bryan L Betz
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Yue Cao
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan; Department of Radiology, University of Michigan, Ann Arbor, Michigan; Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Christopher J Kim
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Jason Heth
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan
| | - Oren Sagher
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan
| | - Theodore S Lawrence
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Christina I Tsien
- Department of Radiation Oncology, Washington University, St. Louis, Missouri
| |
Collapse
|
9
|
|
10
|
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: 1.9] [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
|
11
|
Thorwarth D. Functional imaging for radiotherapy treatment planning: current status and future directions-a review. Br J Radiol 2015; 88:20150056. [PMID: 25827209 PMCID: PMC4628531 DOI: 10.1259/bjr.20150056] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
In recent years, radiotherapy (RT) has been subject to a number of technological innovations. Today, RT is extremely flexible, allowing irradiation of tumours with high doses, whilst also sparing normal tissues from doses. To make use of these additional degrees of freedom, integration of functional image information may play a key role (i) for better staging and tumour detection, (ii) for more accurate RT target volume delineation, (iii) to assess functional information about biological characteristics and individual radiation resistance and (iv) to apply personalized dose prescriptions. In this article, we discuss the current status and future directions of different clinically available functional imaging modalities; CT, MRI, positron emission tomography (PET) as well as the hybrid imaging techniques PET/CT and PET/MRI and their potential for individualized RT.
Collapse
Affiliation(s)
- D Thorwarth
- Section for Biomedical Physics, Department of Radiation Oncology, Eberhard Karls University Tübingen, Tübingen, Germany
| |
Collapse
|
12
|
Schmainda KM. Diffusion-weighted MRI as a biomarker for treatment response in glioma. CNS Oncol 2015; 1:169-80. [PMID: 23936625 DOI: 10.2217/cns.12.25] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Diffusion-weighted imaging (DWI) is a powerful MRI method, which probes abnormalities of tissue structure by detecting microscopic changes in water mobility at a cellular level beyond what is available with other imaging techniques. Accordingly, DWI has the potential to identify pathology before gross anatomic changes are evident on standard anatomical brain images. These features of tissue characterization and earlier detection are what make DWI particularly appealing for the evaluation of gliomas and the newer therapies where standard anatomical imaging is proving insufficient. This article focuses on the basic principles and applications of DWI, and its derived parameter, the apparent diffusion coefficient, for the purposes of diagnosis and evaluation of glioma, especially in the context of monitoring response to therapy.
Collapse
|
13
|
Pre-treatment apparent diffusion coefficient mapping: differentiation of benign from malignant laryngeal lesions. J Laryngol Otol 2015; 129:57-62. [DOI: 10.1017/s0022215114003338] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
AbstractObjective:To determine whether a threshold apparent diffusion coefficient value may help to differentiate laryngeal carcinomas from benign lesions.Methods:Fifty-three patients with laryngeal masses were recruited; four of them were excluded because of susceptibility artefacts. In the remaining 49 patients, the pathological results showed 32 laryngeal carcinomas and 17 benign lesions. The diagnostic value of diffusion-weighted magnetic resonance imaging for the identification of malignant lesions was determined. In addition, the agreement between diffusion-weighted magnetic resonance imaging and histopathology was assessed. Moreover, the sensitivity, specificity, and negative and positive predictive values of the apparent diffusion coefficient in detecting benign and malignant lesions were analysed. An apparent diffusion coefficient histogram was also produced.Results:An apparent diffusion coefficient value of 1.1 × 10−3 mm2/second produced the best result when used as the cut-off point to differentiate malignant from benign masses.Conclusion:An apparent diffusion coefficient threshold of 1.1 × 10−3 mm2/second is optimal for distinguishing laryngeal carcinomas from benign lesions. Apparent diffusion coefficient values were lower for patients with laryngeal carcinomas than for those with benign lesions.
Collapse
|
14
|
Atkins RJ, Ng W, Stylli SS, Hovens CM, Kaye AH. Repair mechanisms help glioblastoma resist treatment. J Clin Neurosci 2014; 22:14-20. [PMID: 25444993 DOI: 10.1016/j.jocn.2014.09.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 09/03/2014] [Accepted: 09/03/2014] [Indexed: 12/28/2022]
Abstract
Glioblastoma multiforme (GBM) is a malignant and incurable glial brain tumour. The current best treatment for GBM includes maximal safe surgical resection followed by concomitant radiotherapy and adjuvant temozolomide. Despite this, median survival is still only 14-16 months. Mechanisms that lead to chemo- and radio-resistance underpin treatment failure. Insights into the DNA repair mechanisms that permit resistance to chemoradiotherapy in GBM may help improve patient responses to currently available therapies.
Collapse
Affiliation(s)
- Ryan J Atkins
- Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Grattan Street, Parkville, VIC 3050, Australia.
| | - Wayne Ng
- Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Grattan Street, Parkville, VIC 3050, Australia; Department of Neurosurgery, The Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Stanley S Stylli
- Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Grattan Street, Parkville, VIC 3050, Australia; Department of Neurosurgery, The Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Christopher M Hovens
- Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Grattan Street, Parkville, VIC 3050, Australia; Australian Prostate Cancer Research Centre at Epworth, Richmond, VIC, Australia
| | - Andrew H Kaye
- Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Grattan Street, Parkville, VIC 3050, Australia; Department of Neurosurgery, The Royal Melbourne Hospital, Parkville, VIC, Australia
| |
Collapse
|
15
|
Hassan O, Taha MS, Farag W. Diffusion-weighted MRI versus PET/CT in evaluation of clinically N0 neck in patients with HNSCC. Systematic review and meta-analysis study. ACTA ACUST UNITED AC 2014. [DOI: 10.1016/j.ejenta.2014.04.003] [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]
|
16
|
Fan Y, Potdar AA, Gong Y, Eswarappa SM, Donnola S, Lathia JD, Hambardzumyan D, Rich JN, Fox PL. Profilin-1 phosphorylation directs angiocrine expression and glioblastoma progression through HIF-1α accumulation. Nat Cell Biol 2014; 16:445-56. [PMID: 24747440 PMCID: PMC4036069 DOI: 10.1038/ncb2954] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Accepted: 03/20/2014] [Indexed: 12/18/2022]
Abstract
The tumour vascular microenvironment supports tumorigenesis not only by supplying oxygen and diffusible nutrients but also by secreting soluble factors that promote tumorigenesis. Here we identify a feedforward mechanism in which endothelial cells (ECs), in response to tumour-derived mediators, release angiocrines driving aberrant vascularization and glioblastoma multiforme (GBM) progression through a hypoxia-independent induction of hypoxia-inducible factor (HIF)-1α. Phosphorylation of profilin-1 (Pfn-1) at Tyr 129 in ECs induces binding to the tumour suppressor protein von Hippel-Lindau (VHL), and prevents VHL-mediated degradation of prolyl-hydroxylated HIF-1α, culminating in HIF-1α accumulation even in normoxia. Elevated HIF-1α induces expression of multiple angiogenic factors, leading to vascular abnormality and tumour progression. In a genetic model of GBM, mice with an EC-specific defect in Pfn-1 phosphorylation exhibit reduced tumour angiogenesis, normalized vasculature and improved survival. Moreover, EC-specific Pfn-1 phosphorylation is associated with tumour aggressiveness in human glioma. These findings suggest that targeting Pfn-1 phosphorylation may offer a selective strategy for therapeutic intervention of malignant solid tumours.
Collapse
Affiliation(s)
- Yi Fan
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio, USA 44195
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, Pennsylvania, USA 19104
| | - Alka A. Potdar
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio, USA 44195
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio, USA 44106
| | - Yanqing Gong
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio, USA 44195
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, Pennsylvania, USA 19104
| | - Sandeepa M. Eswarappa
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio, USA 44195
| | - Shannon Donnola
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio, USA 44195
| | - Justin D. Lathia
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio, USA 44195
| | - Dolores Hambardzumyan
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio, USA 44195
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio, USA 44195
| | - Jeremy N. Rich
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio, USA 44195
| | - Paul L. Fox
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio, USA 44195
| |
Collapse
|
17
|
Mathematical modeling of PDGF-driven glioblastoma reveals optimized radiation dosing schedules. Cell 2014; 156:603-616. [PMID: 24485463 DOI: 10.1016/j.cell.2013.12.029] [Citation(s) in RCA: 184] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Revised: 09/18/2013] [Accepted: 12/24/2013] [Indexed: 12/19/2022]
Abstract
Glioblastomas (GBMs) are the most common and malignant primary brain tumors and are aggressively treated with surgery, chemotherapy, and radiotherapy. Despite this treatment, recurrence is inevitable and survival has improved minimally over the last 50 years. Recent studies have suggested that GBMs exhibit both heterogeneity and instability of differentiation states and varying sensitivities of these states to radiation. Here, we employed an iterative combined theoretical and experimental strategy that takes into account tumor cellular heterogeneity and dynamically acquired radioresistance to predict the effectiveness of different radiation schedules. Using this model, we identified two delivery schedules predicted to significantly improve efficacy by taking advantage of the dynamic instability of radioresistance. These schedules led to superior survival in mice. Our interdisciplinary approach may also be applicable to other human cancer types treated with radiotherapy and, hence, may lay the foundation for significantly increasing the effectiveness of a mainstay of oncologic therapy. PAPERCLIP:
Collapse
|
18
|
BUB1 and BUBR1 inhibition decreases proliferation and colony formation, and enhances radiation sensitivity in pediatric glioblastoma cells. Childs Nerv Syst 2013; 29:2241-8. [PMID: 23728478 DOI: 10.1007/s00381-013-2175-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2013] [Accepted: 05/16/2013] [Indexed: 01/13/2023]
Abstract
PURPOSE Glioblastoma (GBM) is a very aggressive and lethal brain tumor with poor prognosis. Despite new treatment strategies, patients' median survival is still lower than 1 year in most cases. The expression of the BUB gene family has demonstrated to be altered in a variety of solid tumors, pointing to a role as putative therapeutic target. The purpose of this study was to determine BUB1, BUB3, and BUBR1 gene expression profiles in glioblastoma and to analyze the effects of BUB1 and BUBR1 inhibition combined or not with Temozolomide and radiation in the pediatric SF188 GBM cell line. METHODS For gene expression analysis, 8 cell lines and 18 tumor samples were used. The effect of BUB1 and BUBR1 inhibition was evaluated using siRNA. Apoptosis, cell proliferation, cell cycle kinetics, micronuclei formation, and clonogenic capacity were analyzed after BUB1 and BUBR1 inhibition. Additionally, combinatorial effects of gene inhibition and radiation or Temozolomide (TMZ) treatment were evaluated through proliferation and clonogenic capacity assays. RESULTS We report the upregulation of BUB1 and BUBR1 expression and the downregulation of BUB3 in GBM samples and cell lines when compared to white matter samples (p < 0.05). Decreased cell proliferation and colony formation after BUB1 and BUBR1 inhibition were observed, along with increased micronuclei formation. Combinations with TMZ also caused cell cycle arrest and increased apoptosis. Moreover, our results demonstrate that BUB1 and BUBR1 inhibition sensitized SF188 cells to γ-irradiation as shown by decreased growth and abrogation of colony formation capacity. CONCLUSION BUB1 and BUBR1 inhibition decreases proliferation and shows radiosensitizing effects on pediatric GBM cells, which could improve treatment strategies for this devastating tumor. Collectively, these findings highlight the potentials of BUB1 and BUBR1 as putative therapeutic targets for glioblastoma treatment.
Collapse
|
19
|
Diffusion-Weighted MRI as a Biomarker of Tumor Radiation Treatment Response Heterogeneity: A Comparative Study of Whole-Volume Histogram Analysis versus Voxel-Based Functional Diffusion Map Analysis. Transl Oncol 2013; 6:554-61. [PMID: 24151536 DOI: 10.1593/tlo.13532] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Revised: 07/29/2013] [Accepted: 09/05/2013] [Indexed: 12/21/2022] Open
Abstract
RATIONALE Treatment of glioblastoma (GBM) remains challenging due in part to its histologic intratumoral heterogeneity that contributes to its overall poor treatment response. Our goal was to evaluate a voxel-based biomarker, the functional diffusion map (fDM), as an imaging biomarker to detect heterogeneity of tumor response in a radiation dose escalation protocol using a genetically engineered murine GBM model. EXPERIMENTAL DESIGN Twenty-four genetically engineered murine GBM models [Ink4a-Arf(-/-)/Pten(loxp/loxp)/Ntv-a RCAS/PDGF(+)/Cre(+)] were randomized in four treatment groups (n = 6 per group) consisting of daily doses of 0, 1, 2, and 4 Gy delivered for 5 days. Contrast-enhanced T1-weighted and diffusion-weighted magnetic resonance imaging (MRI) scans were acquired for tumor delineation and quantification of apparent diffusion coefficient (ADC) maps, respectively. MRI experiments were performed daily for a week and every 2 days thereafter. For each animal, the area under the curve (AUC) of the percentage change of the ADC (AUCADC) and that of the increase in fDM values (AUCfDM+) were determined within the first 5 days following therapy initiation. RESULTS Animal survival increased with increasing radiation dose. Treatment induced a dose-dependent increase in tumor ADC values. The strongest correlation between survival and ADC measurements was observed using the AUCfDM+ metric (R (2) = 0.88). CONCLUSION This study showed that the efficacy of a voxel-based imaging biomarker (fDM) was able to detect spatially varying changes in tumors, which were determined to be a more sensitive predictor of overall response versus whole-volume tumor measurements (AUCADC). Finally, fDM provided for visualization of treatment-associated spatial heterogeneity within the tumor.
Collapse
|
20
|
Differentiating laryngeal carcinomas from precursor lesions by diffusion-weighted magnetic resonance imaging at 3.0 T: a preliminary study. PLoS One 2013; 8:e68622. [PMID: 23874693 PMCID: PMC3706423 DOI: 10.1371/journal.pone.0068622] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Accepted: 05/31/2013] [Indexed: 11/19/2022] Open
Abstract
Background Diffusion-weighted magnetic resonance imaging (DWI) has been introduced in head and neck cancers. Due to limitations in the performance of laryngeal DWI, including the complex anatomical structure of the larynx leading to susceptibility effects, the value of DWI in differentiating benign from malignant laryngeal lesions has largely been ignored. We assessed whether a threshold for the apparent diffusion coefficient (ADC) was useful in differentiating preoperative laryngeal carcinomas from precursor lesions by turbo spin-echo (TSE) DWI and 3.0-T magnetic resonance. Methods We evaluated DWI and the ADC value in 33 pathologically proven laryngeal carcinomas and 17 precancerous lesions. Results The sensitivity, specificity, and accuracy were 81.8%, 64.7%, 76.0% by laryngostroboscopy, respectively. The sensitivity, specificity, and accuracy of conventional magnetic resonance imaging were 90.9%, 76.5%, 86.0%, respectively. Qualitative DWI analysis produced sensitivity, specificity, and accuracy values of 100.0, 88.2, and 96.0%, respectively. The ADC values were lower for patients with laryngeal carcinoma (mean 1.195±0.32×10−3 mm2/s) versus those with laryngeal precancerous lesions (mean 1.780±0.32×10−3 mm2/s; P<0.001). ROC analysis showed that the area under the curve was 0.956 and the optimum threshold for the ADC was 1.455×10−3 mm2/s, resulting in a sensitivity of 94.1%, a specificity of 90.9%, and an accuracy of 92.9%. Conclusions Despite some limitations, including the small number of laryngeal carcinomas included, DWI may detect changes in tumor size and shape before they are visible by laryngostroboscopy. The ADC values were lower for patients with laryngeal carcinoma than for those with laryngeal precancerous lesions. The proposed cutoff for the ADC may help distinguish laryngeal carcinomas from laryngeal precancerous lesions.
Collapse
|
21
|
Ulasov I, Thaci B, Sarvaiya P, Yi R, Guo D, Auffinger B, Pytel P, Zhang L, Kim CK, Borovjagin A, Dey M, Han Y, Baryshnikov AY, Lesniak MS. Inhibition of MMP14 potentiates the therapeutic effect of temozolomide and radiation in gliomas. Cancer Med 2013; 2:457-67. [PMID: 24156018 PMCID: PMC3799280 DOI: 10.1002/cam4.104] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Revised: 05/21/2013] [Accepted: 05/23/2013] [Indexed: 12/12/2022] Open
Abstract
Metalloproteinases are membrane-bound proteins that play a role in the cellular responses to antiglioma therapy. Previously, it has been shown that treatment of glioma cells with temozolomide (TMZ) and radiation (XRT) induces the expression of metalloproteinase 14 (MMP14). To investigate the role of MMP14 in gliomagenesis, we used several chemical inhibitors which affect MMP14 expression. Of all the inhibitors tested, we found that Marimastat not only inhibits the expression of MMP14 in U87 and U251 glioma cells, but also induces cell cycle arrest. To determine the relationship between MMP14 inhibition and alteration of the cell cycle, we used an RNAi technique. Genetic knockdown of MMP14 in U87 and U251 glioma cells induced G2/M arrest and decreased proliferation. Mechanistically, we show that TMZ and XRT regulated expression of MMP14 in clinical samples and in vitro models through downregulation of microRNA374. In vivo genetic knockdown of MMP14 significantly decreased tumor growth of glioma xenografts and improved survival of glioma-bearing mice. Moreover, the combination of MMP14 silencing with TMZ and XRT significantly improved the survival of glioma-bearing mice compared to a single modality treatment group. Therefore, we show that the inhibition of MMP14 sensitizes tumor cells to TMZ and XRT and could be used as a future strategy for antiglioma therapy. Glioblastoma remains an incurable form of brain cancer. In this manuscript, we show that inhibition of MMP14 can potentiate the efficacy of current standard of care which includes chemo- and radiotherapy.
Collapse
Affiliation(s)
- Ilya Ulasov
- The Brain Tumor Center, The University of Chicago Chicago, Illinois, 60637
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Cheng JCH, Yuan A, Chen JH, Lu YC, Cho KH, Wu JK, Wu CJ, Chang YC, Yang PC. Early detection of Lewis lung carcinoma tumor control by irradiation using diffusion-weighted and dynamic contrast-enhanced MRI. PLoS One 2013; 8:e62762. [PMID: 23658769 PMCID: PMC3642149 DOI: 10.1371/journal.pone.0062762] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Accepted: 03/25/2013] [Indexed: 01/08/2023] Open
Abstract
PURPOSE To investigate the correlation between diffusion-weighted (DW) and dynamic contrast-enhanced (DCE) magnetic resonance imaging (MRI) derived parameters and radioresponsiveness of Lewis lung carcinoma (LLC) tumor. MATERIALS AND METHODS LLC tumor growth in C57BL/6 mouse limb was used for the experiment. The tumors were irradiated with 10 Gy×5, or 30 Gy×2 vs. sham irradiation. Fourteen tumors were subjected to DW-MRI and DCE-MRI pre-radiotherapy and weekly imaging after radiotherapy. The temporal changes in apparent diffusion coefficient (ADC) and DCE-MRI derived parameters (K(trans), k(ep), v(e), and v(p)) were correlated with tumor size, and were histologically compared with CD31 staining of resected tumors. RESULTS The 10 Gy×5 dose inhibited tumor growth for a week, while 30 Gy×2 controlled tumor growth for a 3-week observation period. One week after radiotherapy (week 2), irradiated tumors showed significantly higher values of ADC than untreated ones (10 Gy×5, p = 0.004; 30 Gy×2, p = 0.01). Significantly higher values of v(e) were shown earlier by 30 Gy×2 vs. sham (p = 0.01) and 10 Gy×5 vs. sham irradiation (p = 0.05). Sustained higher v(e) from 10 Gy×5 compared to sham irradiated tumors was evident at week 3 (p = 0.016) and week 4 (p = 0.046). A 13.8% early increase in ADC for 30 Gy×2 tumor group (p = 0.002) and a 16.5% increase for 10 Gy×5 group were noted (p = 0.01) vs. sham irradiation (which showed a 2.2% decrease). No differences were found for K(trans), k(ep), or v(p). Both radiotherapy groups demonstrated significant reduction in microvessel counts. CONCLUSION Early increase in ADC and v(e) correlated with tumor control by irradiation.
Collapse
Affiliation(s)
- Jason Chia-Hsien Cheng
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University College of Electrical Engineering and Computer Science, Taipei, Taiwan
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Ang Yuan
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Jyh-Horng Chen
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University College of Electrical Engineering and Computer Science, Taipei, Taiwan
| | - Yi-Chien Lu
- Department of Radiology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Kuan-Hung Cho
- Instrumentation Resource Center, National Yang-Ming University, Taipei, Taiwan
| | - Jian-Kuen Wu
- Institute of Electro-Optical Science and Technology, National Taiwan Normal University, Taipei, Taiwan
| | - Chien-Jang Wu
- Institute of Electro-Optical Science and Technology, National Taiwan Normal University, Taipei, Taiwan
| | - Yeun-Chung Chang
- Department of Radiology, National Taiwan University College of Medicine, Taipei, Taiwan
- Department of Medical Imaging, National Taiwan University Hospital, Taipei, Taiwan
- * E-mail:
| | - Pan-Chyr Yang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
23
|
DW-MRI as a Predictive Biomarker of Radiosensitization of GBM through Targeted Inhibition of Checkpoint Kinases. Transl Oncol 2013; 6:133-42. [PMID: 23544166 DOI: 10.1593/tlo.13214] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Revised: 02/13/2013] [Accepted: 02/28/2013] [Indexed: 01/22/2023] Open
Abstract
PURPOSE The inherent treatment resistance of glioblastoma (GBM) can involve multiple mechanisms including checkpoint kinase (Chk1/2)-mediated increased DNA repair capability, which can attenuate the effects of genotoxic chemotherapies and radiation. The goal of this study was to evaluate diffusion-weighted magnetic resonance imaging (DW-MRI) as a biomarker for Chk1/2 inhibitors in combination with radiation for enhancement of treatment efficacy in GBM. EXPERIMENTAL DESIGN We evaluated a specific small molecule inhibitor of Chk1/2, AZD7762, in combination with radiation using in vitro human cell lines and in vivo using a genetically engineered GBM mouse model. DW-MRI and T1-contrast MRI were used to follow treatment effects on intracranial tumor cellularity and growth rates, respectively. RESULTS AZD7762 inhibited clonal proliferation in a panel of GBM cell lines and increased radiosensitivity in p53-mutated GBM cell lines to a greater extent compared to p53 wild-type cells. In vivo efficacy of AZD7762 demonstrated a dose-dependent inhibitory effect on GBM tumor growth rate and a reduction in tumor cellularity based on DW-MRI scans along with enhancement of radiation efficacy. CONCLUSION DW-MRI was found to be a useful imaging biomarker for the detection of radiosensitization through inhibition of checkpoint kinases. Chk1/2 inhibition resulted in antiproliferative activity, prevention of DNA damage-induced repair, and radiosensitization in preclinical GBM tumor models, both in vitro and in vivo. The effects were found to be maximal in p53-mutated GBM cells. These results provide the rationale for integration of DW-MRI in clinical translation of Chk1/2 inhibition with radiation for the treatment of GBM.
Collapse
|
24
|
|
25
|
Wang H, Galbán S, Wu R, Bowman BM, Witte A, Vetter K, Galbán CJ, Ross BD, Cho KR, Rehemtulla A. Molecular imaging reveals a role for AKT in resistance to cisplatin for ovarian endometrioid adenocarcinoma. Clin Cancer Res 2012; 19:158-69. [PMID: 23095324 DOI: 10.1158/1078-0432.ccr-12-2380] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE Ovarian cancer is the fifth leading cause of cancer-related deaths among American women. Platinum-based chemotherapy, such as cisplatin, represents the standard-of-care for ovarian cancer. However, toxicity and acquired resistance to cisplatin have proven challenging in the treatment of patients with ovarian cancer. EXPERIMENTAL DESIGN Using a genetically engineered mouse model of ovarian endometrioid adenocarcinoma (OEA) in combination with molecular-imaging technologies, we studied the activation of the AKT serine/threonine kinase in response to long-term cisplatin therapy. RESULTS Treatment of cells in culture and tumor-bearing animals with cisplatin resulted in activation of AKT, a key mediator of cell survival. On the basis of these results, we investigated the therapeutic use of AKT inhibition in combination with cisplatin, which resulted in enhanced and prolonged induction of apoptosis and in significantly improved tumor control as compared with either agent alone. CONCLUSION These results provide an impetus for clinical trials using combination therapy. To facilitate these trials, we also show the use of diffusion-weighted MRI as an imaging biomarker for evaluation of therapeutic efficacy in OEA.
Collapse
Affiliation(s)
- Hanxiao Wang
- Center for Molecular Imaging, Departments of Radiation Oncology, The University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Atkins RJ, Dimou J, Paradiso L, Morokoff AP, Kaye AH, Drummond KJ, Hovens CM. Regulation of glycogen synthase kinase-3 beta (GSK-3β) by the Akt pathway in gliomas. J Clin Neurosci 2012; 19:1558-63. [PMID: 22999562 DOI: 10.1016/j.jocn.2012.07.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Accepted: 07/04/2012] [Indexed: 12/12/2022]
Abstract
Gliomas are aggressive brain tumours that, despite advances in multimodal therapies, continue to portend a dismal prognosis. Glioblastoma multiforme (GBM) represents the most aggressive glioma and patients have a median survival of 14 months, even with the best available treatments. The phosphoinositide 3-kinase/Akt/glycogen synthase kinase-3 beta (GSK-3β) and Wnt/β-catenin pathways are dysregulated in a number of cancers, and these two pathways share a common node protein, GSK-3β. This protein is responsible for the regulation/degradation of β-catenin, which reduces β-catenin's translocation to the nucleus and influences the subsequent transcription of oncogenes. The non-specific small-molecule GSK-3β inhibitor, lithium chloride (LiCl), and the specific Akt inhibitor, AktX, were used to treat U87MG and U87MG.Δ2-7 human glioma cell lines. LiCl treatment significantly affected cell morphology of U87MG and U87MG.Δ2-7 cells, while also increasing levels of phospho-GSK-3β in a dose-dependent manner. Increased cell proliferation was observed at low-to-mid LiCl concentrations as determined by MTT cell growth assays. Treatment of U87MG and U87MG.Δ2-7 cells with AktX resulted in reduced levels of phospho-GSK-3β through its inhibition of Akt, in addition to decreased levels of phosphorylated (active) Akt in a dose-dependent fashion. We have shown in this study that GSK-3β regulation by phosphorylation is important for cell morphology and growth, and that LiCl enhances growth of U87MG and U87MG.Δ2-7 cells by inhibiting GSK-3β through its phosphorylation, whereas AktX reduces growth via activation of GSK-3β by inhibiting Akt's kinase activity.
Collapse
Affiliation(s)
- Ryan J Atkins
- Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Royal Parade, Parkville 3050, Victoria, Australia.
| | | | | | | | | | | | | |
Collapse
|