1
|
Yang G, Zhang K, Xu W, Xu S. A review of clinical use of surface-enhanced Raman scattering-based biosensing for glioma. Front Neurol 2024; 15:1287213. [PMID: 38651101 PMCID: PMC11033440 DOI: 10.3389/fneur.2024.1287213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 02/27/2024] [Indexed: 04/25/2024] Open
Abstract
Glioma is the most common malignant tumor of the nervous system in recent centuries, and the incidence rate of glioma is increasing year by year. Its invasive growth and malignant biological behaviors make it one of the most challenging malignant tumors. Maximizing the resection range (EOR) while minimizing the impact on normal brain tissue is crucial for patient prognosis. Changes in metabolites produced by tumor cells and their microenvironments might be important indicators. As a powerful spectroscopic technique, surface-enhanced Raman scattering (SERS) has many advantages, including ultra-high sensitivity, high specificity, and non-invasive features, which allow SERS technology to be widely applied in biomedicine, especially in the differential diagnosis of malignant tumor tissues. This review first introduced the clinical use of responsive SERS probes. Next, the sensing mechanisms of microenvironment-responsive SERS probes were summarized. Finally, the biomedical applications of these responsive SERS probes were listed in four sections, detecting tumor boundaries due to the changes of pH-responsive SERS probes, SERS probes to guide tumor resection, SERS for liquid biopsy to achieve early diagnosis of tumors, and the application of free-label SERS technology to detect fresh glioma specimens. Finally, the challenges and prospects of responsive SERS detections were summarized for clinical use.
Collapse
Affiliation(s)
- Guohui Yang
- China-Japan Union Hospital of Jilin University, Changchun, China
| | - Kaizhi Zhang
- China-Japan Union Hospital of Jilin University, Changchun, China
| | - Weiqing Xu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, China
| | - Shuping Xu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, China
- Institute of Theoretical Chemistry, College of Chemistry, Jilin University, Changchun, China
- Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun, China
| |
Collapse
|
2
|
Liu Y, Zhu W, Zhu H, Zhang J, Zhang J, Shen N, Jiang J, Xue Y, Jiang R. Characterization of orthotopic xenograft tumor of glioma stem cells (GSCs) on MRI, PET and immunohistochemical staining. Front Oncol 2022; 12:1085015. [PMID: 36591483 PMCID: PMC9797975 DOI: 10.3389/fonc.2022.1085015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 12/02/2022] [Indexed: 12/23/2022] Open
Abstract
Introduction The orthotopic xenograft tumors of human glioma stem cells (GSCs) is a recent glioma model with genotype and phenotypic characteristics close to human gliomas. This study aimed to explore the imaging and immunohistochemical characteristics of GSCs gliomas. Methods The rats underwent MRI and 18F-FDG PET scan in 6th-8th weeks after GSCs implantation. The MRI morphologic, DWI and PET features of the tumor lesions were assessed. In addition, the immunohistochemical features of the tumor tissues were further analyzed. Results Twenty-five tumor lesions were identified in 20 tumor-bearing rats. On structural MRI, the average tumor size was 30.04±17.31mm2, and the intensity was inhomogeneous in 76.00% (19/25) of the lesions. The proportion of the lesions mainly presented as solid, cystic and patchy tumor were 60.00% (15/25), 16.00% (4/25) and 24.00% (6/25), respectively. The boundary was unclear in 88.00% (22/25), and peritumoral mass effect was observed in 92.00% (23/25) of the lesions. On DWI, 80.00% (20/25) of the lesions showed increased intensity. Of the 14 lesions in the 11 rats underwent PET scan, 57.14% (8/14) showed increased FDG uptake. On immunohistochemical staining, the expression of Ki-67 was strong in all the lesions (51.67%±11.82%). Positive EGFR and VEGF expression were observed in 64.71% (11/17) and 52.94% (9/17) of the rats, whereas MGMT and HIF-1α showed negative expression in all the lesions. Discussion GSC gliomas showed significant heterogeneity and invasiveness on imaging, and exhibited strong expression of Ki-67, partial expression of EGFR and VEGF, and weak expression of MGMT and HIF-1α on immunohistochemical staining.
Collapse
Affiliation(s)
- Yufei Liu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wenzhen Zhu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hongquan Zhu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jiaxuan Zhang
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ju Zhang
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Nanxi Shen
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jingjing Jiang
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yunjing Xue
- Department of Radiology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Rifeng Jiang
- Department of Radiology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China,*Correspondence: Rifeng Jiang,
| |
Collapse
|
3
|
Ratiometric pH-responsive SERS strategy for glioma boundary determination. Talanta 2022; 250:123750. [DOI: 10.1016/j.talanta.2022.123750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/24/2022] [Accepted: 07/15/2022] [Indexed: 11/19/2022]
|
4
|
Study on the Function and Mechanism of miR-585-3p Inhibiting the Progression of Ovarian Cancer Cells by Targeting FSCN1 to Block the MAPK Signaling Pathway. Anal Cell Pathol (Amst) 2022; 2022:1732365. [PMID: 35602576 PMCID: PMC9122712 DOI: 10.1155/2022/1732365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 03/22/2022] [Accepted: 04/09/2022] [Indexed: 11/17/2022] Open
Abstract
Ovarian cancer (OC) is the leading cause of death for women diagnosed with gynecological cancer. Studies have shown that dysregulated miRNA expression is related to various cancers, including OC. Here, we aimed to explore the biological function and mechanism of miR-585-3p in the occurrence and development of OC. The expression level of miR-585-3p was found to be low in OC tissues and cells. We analyzed the biological function of miR-585-3p in OC through in vitro cell experiments. The results indicated that overexpression of miR-585-3p inhibited the proliferation, invasion, and migration of SW626 cells, while low expression of miR-585-3p had the opposite effect in SKOV3 cells. We then screened the target genes of miR-585-3p through miRDB database and detected the expression of target genes in OC cells. FSCN1 was found to be most significantly upregulated in OC cells. Dual-luciferase reporter assays revealed FSCN1 as a potential target of miR-585-3p. Western blot analysis showed that miR-585-3p targeted FSCN1 to inhibit protein phosphorylation of ERK. In vivo animal experiments also confirmed that miR-585-3p targets FSCN1 to inhibit tumor growth and block the MAPK signaling pathway. In summary, miR-585-3p inhibits the proliferation, migration, and invasion of OC cells by targeting FSCN1, and its mechanism of action may be achieved by inhibiting the activation of the MAPK signaling pathway. miR-585-3p may serve as a potential biomarker and therapeutic target for OC.
Collapse
|
5
|
Molecular imaging of a fluorescent antibody against epidermal growth factor receptor detects high-grade glioma. Sci Rep 2021; 11:5710. [PMID: 33707521 PMCID: PMC7952570 DOI: 10.1038/s41598-021-84831-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 02/16/2021] [Indexed: 01/31/2023] Open
Abstract
The prognosis for high-grade glioma (HGG) remains dismal and the extent of resection correlates with overall survival and progression free disease. Epidermal growth factor receptor (EGFR) is a biomarker heterogeneously expressed in HGG. We assessed the feasibility of detecting HGG using near-infrared fluorescent antibody targeting EGFR. Mice bearing orthotopic HGG xenografts with modest EGFR expression were imaged in vivo after systemic panitumumab-IRDye800 injection to assess its tumor-specific uptake macroscopically over 14 days, and microscopically ex vivo. EGFR immunohistochemical staining of 59 tumor specimens from 35 HGG patients was scored by pathologists and expression levels were compared to that of mouse xenografts. Intratumoral distribution of panitumumab-IRDye800 correlated with near-infrared fluorescence and EGFR expression. Fluorescence distinguished tumor cells with 90% specificity and 82.5% sensitivity. Target-to-background ratios peaked at 14 h post panitumumab-IRDye800 infusion, reaching 19.5 in vivo and 7.6 ex vivo, respectively. Equivalent or higher EGFR protein expression compared to the mouse xenografts was present in 77.1% HGG patients. Age, combined with IDH-wildtype cerebral tumor, was predictive of greater EGFR protein expression in human tumors. Tumor specific uptake of panitumumab-IRDye800 provided remarkable contrast and a flexible imaging window for fluorescence-guided identification of HGGs despite modest EGFR expression.
Collapse
|
6
|
Yan Y, Zeng S, Gong Z, Xu Z. Clinical implication of cellular vaccine in glioma: current advances and future prospects. J Exp Clin Cancer Res 2020; 39:257. [PMID: 33228738 PMCID: PMC7685666 DOI: 10.1186/s13046-020-01778-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 11/12/2020] [Indexed: 02/08/2023] Open
Abstract
Gliomas, especially glioblastomas, represent one of the most aggressive and difficult-to-treat human brain tumors. In the last few decades, clinical immunotherapy has been developed and has provided exceptional achievements in checkpoint inhibitors and vaccines for cancer treatment. Immunization with cellular vaccines has the advantage of containing specific antigens and acceptable safety to potentially improve cancer therapy. Based on T cells, dendritic cells (DC), tumor cells and natural killer cells, the safety and feasibility of cellular vaccines have been validated in clinical trials for glioma treatment. For TAA engineered T cells, therapy mainly uses chimeric antigen receptors (IL13Rα2, EGFRvIII and HER2) and DNA methylation-induced technology (CT antigen) to activate the immune response. Autologous dendritic cells/tumor antigen vaccine (ADCTA) pulsed with tumor lysate and peptides elicit antigen-specific and cytotoxic T cell responses in patients with malignant gliomas, while its pro-survival effect is biased. Vaccinations using autologous tumor cells modified with TAAs or fusion with fibroblast cells are characterized by both effective humoral and cell-mediated immunity. Even though few therapeutic effects have been observed, most of this therapy showed safety and feasibility, asking for larger cohort studies and better guidelines to optimize cellular vaccine efficiency in anti-glioma therapy.
Collapse
Affiliation(s)
- Yuanliang Yan
- Department of Pharmacy, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
| | - Shuangshuang Zeng
- Department of Pharmacy, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
| | - Zhicheng Gong
- Department of Pharmacy, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
| | - Zhijie Xu
- Department of Pathology, Xiangya Hospital, Central South University, 87 Xiangya Road, Hunan, 410008, Changsha, China.
| |
Collapse
|
7
|
Chen W, Hong L, Hou C, Wang Y, Wang F, Zhang J. MicroRNA-585 inhibits human glioma cell proliferation by directly targeting MDM2. Cancer Cell Int 2020; 20:469. [PMID: 33005102 PMCID: PMC7523344 DOI: 10.1186/s12935-020-01528-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 09/01/2020] [Indexed: 02/07/2023] Open
Abstract
Background MicroRNAs (miRNAs) are important regulators for cancer cell proliferation. miR-585 has been shown to inhibit the proliferation of several types of cancer, however, little is known about its role in human glioma cells. Methods miR-585 levels in human glioma clinical samples and cell lines were examined by quantitative real-time PCR (qRT-PCR) analysis. Cell proliferation was measured by Cell Counting Kit-8 (CCK-8) and EdU incorporation assays in vitro. For in vivo investigations, U251 cells were intracranially inoculated in BALB/c nude mice and xenografted tumors were visualized by magnetic resonance imaging (MRI). Results miR-585 expression is downregulated in human glioma tissues and cell lines compared with non-cancerous counterparts. Additionally, miR-585 overexpression inhibits and its knockdown promotes human glioma cell proliferation in vitro. Moreover, miR-585 overexpression also inhibits the growth of glioma xenografts in vivo, suggesting that miR-585 may act as a tumor suppressor to inhibit the proliferation of human glioma. Furthermore, miR-585 directly targets and decreases the expression of oncoprotein murine double minute 2 (MDM2). More importantly, the restoration of MDM2 via enforced overexpression markedly rescues miR-585 inhibitory effect on human glioma cell proliferation, thus demonstrating that targeting MDM2 is a critical mechanism by which miR-585 inhibits human glioma cell proliferation. Conclusions Our study unveils the anti-proliferative role of miR-585 in human glioma cells, and also implicates its potential application in clinical therapy.
Collapse
Affiliation(s)
- Wangsheng Chen
- Department of Radiology, Hainan General Hospital/Hainan Hospital of Hainan Medical University, Haikou, 570311 China
| | - Lan Hong
- Department of Gynecology, Hainan General Hospital/Hainan Hospital of Hainan Medical University, Haikou, 570311 China
| | - Changlong Hou
- Department of Radiology, Shanghai East Hospital, Tongji University School of Medicine, No 150, JiMo Road, Pudong New Area, Shanghai, 200120 China
| | - Yibin Wang
- Department of Radiology, Shanghai East Hospital, Tongji University School of Medicine, No 150, JiMo Road, Pudong New Area, Shanghai, 200120 China
| | - Fei Wang
- Department of Radiology, Hainan General Hospital/Hainan Hospital of Hainan Medical University, Haikou, 570311 China
| | - Jianhua Zhang
- Department of Radiology, Shanghai East Hospital, Tongji University School of Medicine, No 150, JiMo Road, Pudong New Area, Shanghai, 200120 China
| |
Collapse
|
8
|
Xue W, Ton H, Zhang J, Xie T, Chen X, Zhou B, Guo Y, Fang J, Wang S, Zhang W. Patient‑derived orthotopic xenograft glioma models fail to replicate the magnetic resonance imaging features of the original patient tumor. Oncol Rep 2020; 43:1619-1629. [PMID: 32323818 PMCID: PMC7107810 DOI: 10.3892/or.2020.7538] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 02/12/2020] [Indexed: 12/14/2022] Open
Abstract
Patient-derived orthotopic glioma xenograft models are important platforms used for pre-clinical research of glioma. In the present study, the diagnostic ability of magnetic resonance imaging (MRI) was examined with regard to the identification of biomarkers obtained from patient-derived glioma xenografts and human tumors. Conventional MRI, diffusion weighted imaging and dynamic contrast-enhanced (DCE)-MRI were used to analyze seven pairs of high grade gliomas with their corresponding xenografts obtained from non-obese diabetic-severe-combined immunodeficiency nude mice. Tumor samples were collected for transcriptome sequencing and histopathological staining, and differentially expressed genes were screened between the original tumors and the corresponding xenografts. Gene Ontology (GO) analysis was performed to predict the functions of these genes. In 6 cases of xenografts with diffuse growth, the degree of enhancement was significantly lower compared with the original tumors. Histopathological staining indicated that the microvascular area and microvascular diameter of the xenografts were significantly lower compared with the original tumors (P=0.009 and P=0.007, respectively). In one case, there was evidence of nodular tumor growth in the mouse. Both MRI and histopathological staining showed a clear demarcation between the transplanted tumors and the normal brain tissues. The relative apparent diffusion coefficient values of the 7 cases examined were significantly higher compared with the corresponding original tumors (P=0.001) and transfer coefficient values derived from DCE-MRI of the tumor area was significantly lower compared with the original tumors (P=0.016). GO analysis indicated that the expression levels of extracellular matrix-associated genes, angiogenesis-associated genes and immune function-associated genes in the original tumors were higher compared with the corresponding xenografts. In conclusion, the data demonstrated that the MRI features of patient-derived xenograft glioma models in mice were different compared with those of the original patient tumors. Differential gene expression may underlie the differences noted in the MRI features between original tumors and corresponding xenografts. The results of the present study highlight the precautions that should be taken when extrapolating data from patient-derived xenograft studies, and their applicability to humans.
Collapse
Affiliation(s)
- Wei Xue
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing 400042, P.R. China
| | - Haipeng Ton
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing 400042, P.R. China
| | - Junfeng Zhang
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing 400042, P.R. China
| | - Tian Xie
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing 400042, P.R. China
| | - Xiao Chen
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing 400042, P.R. China
| | - Bo Zhou
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing 400042, P.R. China
| | - Yu Guo
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing 400042, P.R. China
| | - Jingqin Fang
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing 400042, P.R. China
| | - Shunan Wang
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing 400042, P.R. China
| | - Weiguo Zhang
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing 400042, P.R. China
| |
Collapse
|
9
|
Hope TR, White NS, Kuperman J, Chao Y, Yamin G, Bartch H, Schenker-Ahmed NM, Rakow-Penner R, Bussell R, Nomura N, Kesari S, Bjørnerud A, Dale AM. Demonstration of Non-Gaussian Restricted Diffusion in Tumor Cells Using Diffusion Time-Dependent Diffusion-Weighted Magnetic Resonance Imaging Contrast. Front Oncol 2016; 6:179. [PMID: 27532028 PMCID: PMC4970563 DOI: 10.3389/fonc.2016.00179] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 07/19/2016] [Indexed: 12/31/2022] Open
Abstract
The diffusion-weighted magnetic resonance imaging (DWI) technique enables quantification of water mobility for probing microstructural properties of biological tissue and has become an effective tool for collecting information about the underlying pathology of cancerous tissue. Measurements using multiple b-values have indicated biexponential signal attenuation, ascribed to “fast” (high ADC) and “slow” (low ADC) diffusion components. In this empirical study, we investigate the properties of the diffusion time (Δ)-dependent components of the diffusion-weighted (DW) signal in a constant b-value experiment. A xenograft gliobastoma mouse was imaged using Δ = 11 ms, 20 ms, 40 ms, 60 ms, and b = 500–4000 s/mm2 in intervals of 500 s/mm2. Data were corrected for EPI distortions, and the Δ-dependence on the DW-signal was measured within three regions of interest [intermediate- and high-density tumor regions and normal-appearing brain (NAB) tissue regions]. In this study, we verify the assumption that the slow decaying component of the DW-signal is non-Gaussian and dependent on Δ, consistent with restricted diffusion of the intracellular space. As the DW-signal is a function of Δ and is specific to restricted diffusion, manipulating Δ at constant b-value (cb) provides a complementary and direct approach for separating the restricted from the hindered diffusion component. We found that Δ-dependence is specific to the tumor tissue signal. Based on an extended biexponential model, we verified the interpretation of the diffusion time-dependent contrast and successfully estimated the intracellular restricted ADC, signal volume fraction, and cell size within each ROI.
Collapse
Affiliation(s)
- Tuva R Hope
- The Interventional Centre, Oslo University Hospital, Oslo, Norway; Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | - Nathan S White
- Department of Radiology, University of California San Diego , La Jolla, CA , USA
| | - Joshua Kuperman
- Department of Radiology, University of California San Diego , La Jolla, CA , USA
| | - Ying Chao
- Department of Neurosciences, University of California San Diego , La Jolla, CA , USA
| | - Ghiam Yamin
- Department of Radiology, University of California San Diego , La Jolla, CA , USA
| | - Hauke Bartch
- Department of Radiology, University of California San Diego , La Jolla, CA , USA
| | | | - Rebecca Rakow-Penner
- Department of Radiology, University of California San Diego , La Jolla, CA , USA
| | - Robert Bussell
- Department of Radiology, University of California San Diego , La Jolla, CA , USA
| | - Natsuko Nomura
- Department of Neurosciences, University of California San Diego , La Jolla, CA , USA
| | - Santosh Kesari
- Department of Neurosciences, University of California San Diego , La Jolla, CA , USA
| | - Atle Bjørnerud
- The Interventional Centre, Oslo University Hospital, Oslo, Norway; Department of Physics, University of Oslo, Oslo, Norway
| | - Anders M Dale
- Department of Radiology, University of California San Diego, La Jolla, CA, USA; Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
10
|
Jensen SS, Meyer M, Petterson SA, Halle B, Rosager AM, Aaberg-Jessen C, Thomassen M, Burton M, Kruse TA, Kristensen BW. Establishment and Characterization of a Tumor Stem Cell-Based Glioblastoma Invasion Model. PLoS One 2016; 11:e0159746. [PMID: 27454178 PMCID: PMC4959755 DOI: 10.1371/journal.pone.0159746] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 07/07/2016] [Indexed: 11/18/2022] Open
Abstract
Aims Glioblastoma is the most frequent and malignant brain tumor. Recurrence is inevitable and most likely connected to tumor invasion and presence of therapy resistant stem-like tumor cells. The aim was therefore to establish and characterize a three-dimensional in vivo-like in vitro model taking invasion and tumor stemness into account. Methods Glioblastoma stem cell-like containing spheroid (GSS) cultures derived from three different patients were established and characterized. The spheroids were implanted in vitro into rat brain slice cultures grown in stem cell medium and in vivo into brains of immuno-compromised mice. Invasion was followed in the slice cultures by confocal time-lapse microscopy. Using immunohistochemistry, we compared tumor cell invasion as well as expression of proliferation and stem cell markers between the models. Results We observed a pronounced invasion into brain slice cultures both by confocal time-lapse microscopy and immunohistochemistry. This invasion closely resembled the invasion in vivo. The Ki-67 proliferation indexes in spheroids implanted into brain slices were lower than in free-floating spheroids. The expression of stem cell markers varied between free-floating spheroids, spheroids implanted into brain slices and tumors in vivo. Conclusion The established invasion model kept in stem cell medium closely mimics tumor cell invasion into the brain in vivo preserving also to some extent the expression of stem cell markers. The model is feasible and robust and we suggest the model as an in vivo-like model with a great potential in glioma studies and drug discovery.
Collapse
Affiliation(s)
- Stine Skov Jensen
- Department of Pathology, Odense University Hospital, Denmark, Odense C, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense C, Denmark
| | - Morten Meyer
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense C, Denmark
| | - Stine Asferg Petterson
- Department of Pathology, Odense University Hospital, Denmark, Odense C, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense C, Denmark
- * E-mail:
| | - Bo Halle
- Department of Pathology, Odense University Hospital, Denmark, Odense C, Denmark
- Department of Neurosurgery, Odense University Hospital, Odense C, Denmark
| | - Ann Mari Rosager
- Department of Pathology, Odense University Hospital, Denmark, Odense C, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense C, Denmark
| | - Charlotte Aaberg-Jessen
- Department of Pathology, Odense University Hospital, Denmark, Odense C, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense C, Denmark
| | - Mads Thomassen
- Department of Clinical Research, University of Southern Denmark, Odense C, Denmark
- Department of Clinical Genetics, Odense University Hospital, Odense C, Denmark
| | - Mark Burton
- Department of Clinical Research, University of Southern Denmark, Odense C, Denmark
- Department of Clinical Genetics, Odense University Hospital, Odense C, Denmark
| | - Torben A. Kruse
- Department of Clinical Research, University of Southern Denmark, Odense C, Denmark
- Department of Clinical Genetics, Odense University Hospital, Odense C, Denmark
| | - Bjarne Winther Kristensen
- Department of Pathology, Odense University Hospital, Denmark, Odense C, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense C, Denmark
| |
Collapse
|
11
|
Mukthavaram R, Ouyang X, Saklecha R, Jiang P, Nomura N, Pingle SC, Guo F, Makale M, Kesari S. Effect of the JAK2/STAT3 inhibitor SAR317461 on human glioblastoma tumorspheres. J Transl Med 2015; 13:269. [PMID: 26283544 PMCID: PMC4539675 DOI: 10.1186/s12967-015-0627-5] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 08/03/2015] [Indexed: 01/19/2023] Open
Abstract
Background The STAT3 transcription factor is a major intracellular signaling protein and is frequently dysregulated in the most
common and lethal brain malignancy in adults, glioblastoma multiforme (GBM). Activation of STAT3 in GBM correlates with malignancy and poor prognosis. The phosphorylating signal transducer JAK2 activates STAT3 in response to cytokines and growth factors. Currently there are no JAK-STAT pathway inhibitors in clinical trials for GBM, so we sought to examine the anti-GBM activity of SAR317461 (Sanofi-Aventis), a newer generation, highly potent JAK2 inhibitor that exhibits low toxicity and good pharmacokinetics. SAR317461 was initially approved for patient testing in the treatment of primary myelofibrosis (PMF), and has shown activity in preclinical models of melanoma and pulmonary cancer, but has not been tested in GBM. Methods We hypothesized that a potent small molecule JAK2 inhibitor could overcome the heterogeneous nature of GBM, and suppress a range of patient derived GBM tumorsphere lines and immortalized GBM cell lines. We treated with SAR317461 to determine IC50 values, and using Western blot analysis we asked whether the response was linked to STAT3 expression. Western blot analysis, FACS, and cell viability studies were used to identify the mechanism of SAR317461 induced cell death. Results We report for the first time that the JAK2 inhibitor SAR317461 clearly inhibited STAT3 phosphorylation and had substantial activity against cells (IC50 1–10 µM) from 6 of 7 different patient GSC derived GBM tumorsphere lines and three immortalized GBM lines. One patient GSC derived line did not constitutively express STAT3 and was more resistant to SAR317461 (IC50 ≈25 µM). In terms of mechanism we found cleaved PARP and clear apoptosis following SAR317461. SAR317461 also induced autophagy and the addition of an autophagy inhibitor markedly enhanced cell killing by SAR317461. Conclusions We conclude that SAR317461 potently inhibits STAT3 phosphorylation and that it has significant activity against those GBM cells which express activated STAT3. Further studies are warranted in terms of the potential of SAR317461 as single and combined therapy for selectively treating human patients afflicted with GBMs expressing activation of the JAK2-STAT3 signaling axis.
Collapse
Affiliation(s)
- Rajesh Mukthavaram
- Translational Neuro-Oncology Laboratories, Moores Cancer Center, UC San Diego, 3855 Health Sciences Drive, MC#0819, La Jolla, CA, 92093-0819, USA. .,Department of Neurosciences, UC San Diego, La Jolla, CA, USA.
| | - Xiao Ouyang
- Department of Orthopedic Surgery, Xuzhou 3rd Hospital, Affiliated Hospital of Jiangsu University, No. 131 Huancheng Road, 221005, Xuzhou, China.
| | - Rohit Saklecha
- Translational Neuro-Oncology Laboratories, Moores Cancer Center, UC San Diego, 3855 Health Sciences Drive, MC#0819, La Jolla, CA, 92093-0819, USA.
| | - Pengfei Jiang
- Translational Neuro-Oncology Laboratories, Moores Cancer Center, UC San Diego, 3855 Health Sciences Drive, MC#0819, La Jolla, CA, 92093-0819, USA.
| | - Natsuko Nomura
- Translational Neuro-Oncology Laboratories, Moores Cancer Center, UC San Diego, 3855 Health Sciences Drive, MC#0819, La Jolla, CA, 92093-0819, USA.
| | - Sandeep C Pingle
- Translational Neuro-Oncology Laboratories, Moores Cancer Center, UC San Diego, 3855 Health Sciences Drive, MC#0819, La Jolla, CA, 92093-0819, USA.
| | - Fang Guo
- Laboratory of Tumor Targeted Therapy, Shanghai Advanced Research Institute, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 99 Haike Road, 201210, Shanghai, China.
| | - Milan Makale
- Translational Neuro-Oncology Laboratories, Moores Cancer Center, UC San Diego, 3855 Health Sciences Drive, MC#0819, La Jolla, CA, 92093-0819, USA.
| | - Santosh Kesari
- Translational Neuro-Oncology Laboratories, Moores Cancer Center, UC San Diego, 3855 Health Sciences Drive, MC#0819, La Jolla, CA, 92093-0819, USA. .,Department of Neurosciences, UC San Diego, La Jolla, CA, USA. .,Department of Radiation Medicine and Applied Sciences, UC San Diego, La Jolla, CA, USA.
| |
Collapse
|
12
|
Highly penetrative, drug-loaded nanocarriers improve treatment of glioblastoma. Proc Natl Acad Sci U S A 2013; 110:11751-6. [PMID: 23818631 DOI: 10.1073/pnas.1304504110] [Citation(s) in RCA: 196] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Current therapy for glioblastoma multiforme is insufficient, with nearly universal recurrence. Available drug therapies are unsuccessful because they fail to penetrate through the region of the brain containing tumor cells and they fail to kill the cells most responsible for tumor development and therapy resistance, brain cancer stem cells (BCSCs). To address these challenges, we combined two major advances in technology: (i) brain-penetrating polymeric nanoparticles that can be loaded with drugs and are optimized for intracranial convection-enhanced delivery and (ii) repurposed compounds, previously used in Food and Drug Administration-approved products, which were identified through library screening to target BCSCs. Using fluorescence imaging and positron emission tomography, we demonstrate that brain-penetrating nanoparticles can be delivered to large intracranial volumes in both rats and pigs. We identified several agents (from Food and Drug Administration-approved products) that potently inhibit proliferation and self-renewal of BCSCs. When loaded into brain-penetrating nanoparticles and administered by convection-enhanced delivery, one of these agents, dithiazanine iodide, significantly increased survival in rats bearing BCSC-derived xenografts. This unique approach to controlled delivery in the brain should have a significant impact on treatment of glioblastoma multiforme and suggests previously undescribed routes for drug and gene delivery to treat other diseases of the central nervous system.
Collapse
|
13
|
Exploring the cancer stem cell phenotype with high-throughput screening applications. Future Med Chem 2012; 4:1229-41. [PMID: 22800368 DOI: 10.4155/fmc.12.73] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Several human cancer types consist of diverse cell populations that can differ in their tumor-driving potential. One breakthrough has been the identification of poorly differentiated tumor cells, herein termed cancer stem cells (CSCs). CSCs have been shown to initiate tumors in different model systems and have been implicated in cancer resistance to conventional therapies. The clinical relevance of CSCs has been increasingly recognized, and recent progress in their enrichment and characterization has paved the way for exploring CSC biology with high-throughput screening technologies. This article focuses on functional chemical and RNAi screens that have led to the identification of factors that control the CSC phenotype. Different experimental strategies, current challenges and perspectives in CSC drug discovery are discussed.
Collapse
|
14
|
Dunn GP, Rinne ML, Wykosky J, Genovese G, Quayle SN, Dunn IF, Agarwalla PK, Chheda MG, Campos B, Wang A, Brennan C, Ligon KL, Furnari F, Cavenee WK, Depinho RA, Chin L, Hahn WC. Emerging insights into the molecular and cellular basis of glioblastoma. Genes Dev 2012. [PMID: 22508724 DOI: 10.1101/gad.187922.112.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Glioblastoma is both the most common and lethal primary malignant brain tumor. Extensive multiplatform genomic characterization has provided a higher-resolution picture of the molecular alterations underlying this disease. These studies provide the emerging view that "glioblastoma" represents several histologically similar yet molecularly heterogeneous diseases, which influences taxonomic classification systems, prognosis, and therapeutic decisions.
Collapse
Affiliation(s)
- Gavin P Dunn
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Dunn GP, Rinne ML, Wykosky J, Genovese G, Quayle SN, Dunn IF, Agarwalla PK, Chheda MG, Campos B, Wang A, Brennan C, Ligon KL, Furnari F, Cavenee WK, Depinho RA, Chin L, Hahn WC. Emerging insights into the molecular and cellular basis of glioblastoma. Genes Dev 2012; 26:756-84. [PMID: 22508724 DOI: 10.1101/gad.187922.112] [Citation(s) in RCA: 415] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Glioblastoma is both the most common and lethal primary malignant brain tumor. Extensive multiplatform genomic characterization has provided a higher-resolution picture of the molecular alterations underlying this disease. These studies provide the emerging view that "glioblastoma" represents several histologically similar yet molecularly heterogeneous diseases, which influences taxonomic classification systems, prognosis, and therapeutic decisions.
Collapse
Affiliation(s)
- Gavin P Dunn
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Mazar AP, Ahn RW, O'Halloran TV. Development of novel therapeutics targeting the urokinase plasminogen activator receptor (uPAR) and their translation toward the clinic. Curr Pharm Des 2011; 17:1970-8. [PMID: 21711234 DOI: 10.2174/138161211796718152] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Accepted: 05/31/2011] [Indexed: 11/22/2022]
Abstract
The urokinase plasminogen activator receptor (uPAR) mediates cell motility and tissue remodeling. Although uPAR may be expressed transiently in many tissues during development and wound healing, its constitutive expression appears to be associated with several pathological conditions, including cancer. uPAR expression has been demonstrated in most solid tumors and several hematologic malignancies including multiple myeloma and acute leukemias.Unlike many tumor antigens, uPAR is present not only in tumor cells but also in a number of tumor-associated cells including angiogenic endothelial cells and macrophages. The expression of uPAR has been shown to be fairly high in tumor compared to normal, quiescent tissues, which has led to uPAR being proposed as a therapeutic target, as well as a targeting agent, for the treatment of cancer. The majority of therapeutic approaches that have been investigated to date have focused on inhibiting the urokinase plasminogen activator (uPA)-uPAR interaction but these have not led to the development of a viable uPAR targeted clinical candidate. Genetic knockdown approaches e.g. siRNA, shRNA focused on decreasing uPAR expression have demonstrated robust antitumor activity in pre-clinical studies but have been hampered by the obstacles of stability and drug delivery that have limited the field of RNA nucleic acid based therapeutics. More recently, novel approaches that target interactions of uPAR that are downstream of uPA binding e.g. with integrins or that exploit observations describing the biology of uPAR such as mediating uPA internalization and signaling have generated novel uPAR targeted candidates that are now advancing towards clinic evaluation. This review will discuss some of the pitfalls that have delayed progress on uPAR-targeted interventions and will summarize recent progress in the development of uPAR-targeted therapeutics.
Collapse
Affiliation(s)
- Andrew P Mazar
- Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA.
| | | | | |
Collapse
|