1
|
Sun L, Jiang Y, Tan H, Liang R. Collagen and derivatives-based materials as substrates for the establishment of glioblastoma organoids. Int J Biol Macromol 2024; 254:128018. [PMID: 37967599 DOI: 10.1016/j.ijbiomac.2023.128018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 10/31/2023] [Accepted: 11/09/2023] [Indexed: 11/17/2023]
Abstract
Glioblastoma (GBM) is a common primary brain malignancy known for its ability to invade the brain, resistance to chemotherapy and radiotherapy, tendency to recur frequently, and unfavorable prognosis. Attempts have been undertaken to create 2D and 3D models, such as glioblastoma organoids (GBOs), to recapitulate the glioma microenvironment, explore tumor biology, and develop efficient therapies. However, these models have limitations and are unable to fully recapitulate the complex networks formed by the glioma microenvironment that promote tumor cell growth, invasion, treatment resistance, and immune escape. Therefore, it is necessary to develop advanced experimental models that could better simulate clinical physiology. Here, we review recent advances in natural biomaterials (mainly focus on collagen and its derivatives)-based GBO models, as in vitro experimental platforms to simulate GBM tumor biology and response to tested drugs. Special attention will be given to 3D models that use collagen, gelatin, further modified derivatives, and composite biomaterials (e.g., with other natural or synthetic polymers) as substrates. Application of these collagen/derivatives-constructed GBOs incorporate the physical as well as chemical characteristics of the GBM microenvironment. A perspective on future research is given in terms of current issues. Generally, natural materials based on collagen/derivatives (monomers or composites) are expected to enrich the toolbox of GBO modeling substrates and potentially help to overcome the limitations of existing models.
Collapse
Affiliation(s)
- Lu Sun
- Department of Targeting Therapy & Immunology; Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuelin Jiang
- West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Hong Tan
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China.
| | - Ruichao Liang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
2
|
Srivastava R, Dodda M, Zou H, Li X, Hu B. Tumor Niches: Perspectives for Targeted Therapies in Glioblastoma. Antioxid Redox Signal 2023; 39:904-922. [PMID: 37166370 PMCID: PMC10654996 DOI: 10.1089/ars.2022.0187] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 04/10/2023] [Accepted: 04/12/2023] [Indexed: 05/12/2023]
Abstract
Significance: Glioblastoma (GBM), the most common and lethal primary brain tumor with a median survival rate of only 15 months and a 5-year survival rate of only 6.8%, remains largely incurable despite the intensive multimodal treatment of surgical resection and radiochemotherapy. Developing effective new therapies is an unmet need for patients with GBM. Recent Advances: Targeted therapies, such as antiangiogenesis therapy and immunotherapy, show great promise in treating GBM based upon increasing knowledge about brain tumor biology. Single-cell transcriptomics reveals the plasticity, heterogeneity, and dynamics of tumor cells during GBM development and progression. Critical Issues: While antiangiogenesis therapy and immunotherapy have been highly effective in some types of cancer, the disappointing results from clinical trials represent continued challenges in applying these treatments to GBM. Molecular and cellular heterogeneity of GBM is developed temporally and spatially, which profoundly contributes to therapeutic resistance and tumor recurrence. Future Directions: Deciphering mechanisms of tumor heterogeneity and mapping tumor niche trajectories and functions will provide a foundation for the development of more effective therapies for GBM patients. In this review, we discuss five different tumor niches and the intercellular and intracellular communications among these niches, including the perivascular, hypoxic, invasive, immunosuppressive, and glioma-stem cell niches. We also highlight the cellular and molecular biology of these niches and discuss potential strategies to target these tumor niches for GBM therapy. Antioxid. Redox Signal. 39, 904-922.
Collapse
Affiliation(s)
- Rashmi Srivastava
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- John G. Rangos Sr. Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Meghana Dodda
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- John G. Rangos Sr. Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Han Zou
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- John G. Rangos Sr. Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Xuejun Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Changsha, China
| | - Baoli Hu
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- John G. Rangos Sr. Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Cancer Biology Program, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
3
|
Ah-Pine F, Khettab M, Bedoui Y, Slama Y, Daniel M, Doray B, Gasque P. On the origin and development of glioblastoma: multifaceted role of perivascular mesenchymal stromal cells. Acta Neuropathol Commun 2023; 11:104. [PMID: 37355636 PMCID: PMC10290416 DOI: 10.1186/s40478-023-01605-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 06/18/2023] [Indexed: 06/26/2023] Open
Abstract
Glioblastoma, IDH wild-type is the most common and aggressive form of glial tumors. The exact mechanisms of glioblastoma oncogenesis, including the identification of the glioma-initiating cell, are yet to be discovered. Recent studies have led to the hypothesis that glioblastoma arises from neural stem cells and glial precursor cells and that cell lineage constitutes a key determinant of the glioblastoma molecular subtype. These findings brought significant advancement to the comprehension of gliomagenesis. However, the cellular origin of glioblastoma with mesenchymal molecular features remains elusive. Mesenchymal stromal cells emerge as potential glioblastoma-initiating cells, especially with regard to the mesenchymal molecular subtype. These fibroblast-like cells, which derive from the neural crest and reside in the perivascular niche, may underlie gliomagenesis and exert pro-tumoral effects within the tumor microenvironment. This review synthesizes the potential roles of mesenchymal stromal cells in the context of glioblastoma and provides novel research avenues to better understand this lethal disease.
Collapse
Affiliation(s)
- F. Ah-Pine
- Unité de Recherche en Pharmaco-Immunologie (UR-EPI), Université et CHU de La Réunion, 97400 Saint-Denis, France
- Service d’Anatomie et Cytologie Pathologiques, CHU de La Réunion sites SUD – Saint-Pierre, BP 350, 97448 Saint-Pierre Cedex, France
| | - M. Khettab
- Unité de Recherche en Pharmaco-Immunologie (UR-EPI), Université et CHU de La Réunion, 97400 Saint-Denis, France
- Service d’Oncologie Médicale, CHU de La Réunion sites SUD – Saint-Pierre, BP 350, 97448 Saint-Pierre Cedex, France
| | - Y. Bedoui
- Unité de Recherche en Pharmaco-Immunologie (UR-EPI), Université et CHU de La Réunion, 97400 Saint-Denis, France
- Service d’Anatomie et Cytologie Pathologiques, CHU de La Réunion sites SUD – Saint-Pierre, BP 350, 97448 Saint-Pierre Cedex, France
| | - Y. Slama
- Unité de Recherche en Pharmaco-Immunologie (UR-EPI), Université et CHU de La Réunion, 97400 Saint-Denis, France
| | - M. Daniel
- Unité de Recherche en Pharmaco-Immunologie (UR-EPI), Université et CHU de La Réunion, 97400 Saint-Denis, France
- Service de Médecine d’Urgences-SAMU-SMUR, CHU de La Réunion - Site Félix Guyon, Allée Des Topazes CS 11 021, 97400 Saint-Denis, France
| | - B. Doray
- Unité de Recherche en Pharmaco-Immunologie (UR-EPI), Université et CHU de La Réunion, 97400 Saint-Denis, France
- Service de Génétique, CHU de La Réunion - Site Félix Guyon, Allée Des Topazes CS 11 021, 97400 Saint-Denis, France
| | - P. Gasque
- Unité de Recherche en Pharmaco-Immunologie (UR-EPI), Université et CHU de La Réunion, 97400 Saint-Denis, France
| |
Collapse
|
4
|
Yao X, Zeng Y. Tumour associated endothelial cells: origin, characteristics and role in metastasis and anti-angiogenic resistance. Front Physiol 2023; 14:1199225. [PMID: 37389120 PMCID: PMC10301839 DOI: 10.3389/fphys.2023.1199225] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 05/23/2023] [Indexed: 07/01/2023] Open
Abstract
Tumour progression and metastasis remain the leading causes of cancer-related death worldwide. Tumour angiogenesis is essential for tumour progression. The vasculature surrounding tumours is not only a transport channel for nutrients, oxygen, and metabolites, but also a pathway for metastasis. There is a close interaction between tumour cells and endothelial cells in the tumour microenvironment. Recent studies have shown that tumour-associated endothelial cells have different characteristics from normal vascular endothelial cells, play an important role in tumour progression and metastasis, and are expected to be a key target for cancer therapy. This article reviews the tissue and cellular origin of tumour-associated endothelial cells and analyses the characteristics of tumour-associated endothelial cells. Finally, it summarises the role of tumour-associated endothelial cells in tumour progression and metastasis and the prospects for their use in clinical anti-angiogenic therapy.
Collapse
Affiliation(s)
- Xinghong Yao
- Radiation Oncology Key Laboratory of Sichuan Province, Department of Radiotherapy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Ye Zeng
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| |
Collapse
|
5
|
The diagnostic and prognostic potential of the EGFR/MUC4/MMP9 axis in glioma patients. Sci Rep 2022; 12:19868. [PMID: 36400876 PMCID: PMC9674618 DOI: 10.1038/s41598-022-24099-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 11/10/2022] [Indexed: 11/19/2022] Open
Abstract
Glioblastoma is the most aggressive form of brain cancer, presenting poor prognosis despite current advances in treatment. There is therefore an urgent need for novel biomarkers and therapeutic targets. Interactions between mucin 4 (MUC4) and the epidermal growth factor receptor (EGFR) are involved in carcinogenesis, and may lead to matrix metalloproteinase-9 (MMP9) overexpression, exacerbating cancer cell invasiveness. In this study, the role of MUC4, MMP9, and EGFR in the progression and clinical outcome of glioma patients was investigated. Immunohistochemistry (IHC) and immunofluorescence (IF) in fixed tissue samples of glioma patients were used to evaluate the expression and localization of EGFR, MMP9, and MUC4. Kaplan-Meier survival analysis was also performed to test the prognostic utility of the proteins for glioma patients. The protein levels were assessed with enzyme-linked immunosorbent assay (ELISA) in serum of glioma patients, to further investigate their potential as non-invasive serum biomarkers. We demonstrated that MUC4 and MMP9 are both significantly upregulated during glioma progression. Moreover, MUC4 is co-expressed with MMP9 and EGFR in the proliferative microvasculature of glioblastoma, suggesting a potential role for MUC4 in microvascular proliferation and angiogenesis. The combined high expression of MUC4/MMP9, and MUC4/MMP9/EGFR was associated with poor overall survival (OS). Finally, MMP9 mean protein level was significantly higher in the serum of glioblastoma compared with grade III glioma patients, whereas MUC4 mean protein level was minimally elevated in higher glioma grades (III and IV) compared with control. Our results suggest that MUC4, along with MMP9, might account for glioblastoma progression, representing potential therapeutic targets, and suggesting the 'MUC4/MMP9/EGFR axis' may play a vital role in glioblastoma diagnostics.
Collapse
|
6
|
Pucko EB, Ostrowski RP. Inhibiting CK2 among Promising Therapeutic Strategies for Gliomas and Several Other Neoplasms. Pharmaceutics 2022; 14:331. [PMID: 35214064 PMCID: PMC8877581 DOI: 10.3390/pharmaceutics14020331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/24/2022] [Accepted: 01/28/2022] [Indexed: 11/17/2022] Open
Abstract
In gliomas, casein kinase 2 (CK2) plays a dominant role in cell survival and tumour invasiveness and is upregulated in many brain tumours. Among CK2 inhibitors, benzimidazole and isothiourea derivatives hold a dominant position. While targeting glioma tumour cells, they show limited toxicity towards normal cells. Research in recent years has shown that these compounds can be suitable as components of combined therapies with hyperbaric oxygenation. Such a combination increases the susceptibility of glioma tumour cells to cell death via apoptosis. Moreover, researchers planning on using any other antiglioma investigational pharmaceutics may want to consider using these agents in combination with CK2 inhibitors. However, different compounds are not equally effective when in such combination. More research is needed to elucidate the mechanism of treatment and optimize the treatment regimen. In addition, the role of CK2 in gliomagenesis and maintenance seems to have been challenged recently, as some compounds structurally similar to CK2 inhibitors do not inhibit CK2 while still being effective at reducing glioma viability and invasion. Furthermore, some newly developed inhibitors specific for CK2 do not appear to have strong anticancer properties. Further experimental and clinical studies of these inhibitors and combined therapies are warranted.
Collapse
Affiliation(s)
| | - Robert P. Ostrowski
- Department of Experimental and Clinical Neuropathology, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 Pawińskiego St., 02-106 Warsaw, Poland;
| |
Collapse
|
7
|
Staedtke V, Gray-Bethke T, Liu G, Liapi E, Riggins GJ, Bai RY. Neutrophil depletion enhanced the Clostridium novyi-NT therapy in mouse and rabbit tumor models. Neurooncol Adv 2022; 4:vdab184. [PMID: 35118381 PMCID: PMC8807082 DOI: 10.1093/noajnl/vdab184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Hypoxia is a prominent feature of solid tumors and can function as fertile environment for oncolytic anaerobic bacteria such as Clostridium novyi-NT (C. novyi-NT) where it can induce tumor destruction in mice and patients. However, two major obstacles have limited its use, namely the host inflammatory response and the incomplete clearance of normoxic tumor areas. METHODS In this study, we first used a subcutaneous tumor model of a glioblastoma (GBM) cell line in immunocompetent mice to investigate the local distribution of tumor hypoxia, kinetics of C. novyi-NT germination and spread, and the local host immune response. We subsequently applied the acquired knowledge to develop a C. novyi-NT therapy in an orthotopic rabbit brain tumor model. RESULTS We found that local accumulation of granular leukocytes, mainly neutrophils, could impede the spread of bacteria through the tumor and prevent complete oncolysis. Depletion of neutrophils via anti-Ly6G antibody or bone marrow suppression using hydroxyurea significantly improved tumor clearance. We then applied this approach to rabbits implanted with an aggressive intracranial brain tumor and achieved long-term survival in majority of the animals without apparent toxicity. CONCLUSION These results indicated that depleting neutrophils can greatly enhance the safety and efficacy of C. novyi-NT cancer therapy for brain tumors.
Collapse
Affiliation(s)
- Verena Staedtke
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Tyler Gray-Bethke
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Guanshu Liu
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, USA
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Eleni Liapi
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Gregory J Riggins
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ren-Yuan Bai
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
8
|
Recent Advances in Glioma Therapy: Combining Vascular Normalization and Immune Checkpoint Blockade. Cancers (Basel) 2021; 13:cancers13153686. [PMID: 34359588 PMCID: PMC8345045 DOI: 10.3390/cancers13153686] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/09/2021] [Accepted: 07/19/2021] [Indexed: 12/28/2022] Open
Abstract
Glioblastoma (GBM) accounts for more than 50% of all primary malignancies of the brain. Current standard treatment regimen for GBM includes maximal surgical resection followed by radiation and adjuvant chemotherapy. However, due to the heterogeneity of the tumor cells, tumor recurrence is often inevitable. The prognosis of patients with glioma is, thus, dismal. Glioma is a highly angiogenic tumor yet immunologically cold. As such, evolving studies have focused on designing strategies that specifically target the tyrosine kinase receptors of angiokines and encourage immune infiltration. Recent promising results from immunotherapies on other cancer types have prompted further investigations of this therapy in GBM. In this article, we reviewed the pathological angiogenesis and immune reactivity in glioma, as well as its target for drug development, and we discussed future directions in glioma therapy.
Collapse
|
9
|
Carlson JC, Cantu Gutierrez M, Lozzi B, Huang-Hobbs E, Turner WD, Tepe B, Zhang Y, Herman AM, Rao G, Creighton CJ, Wythe JD, Deneen B. Identification of diverse tumor endothelial cell populations in malignant glioma. Neuro Oncol 2021; 23:932-944. [PMID: 33367832 DOI: 10.1093/neuonc/noaa297] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Glioblastoma is the most common and aggressive type of primary brain tumor, as most patients succumb to the disease less than two years after diagnosis. Critically, studies demonstrate that glioma recruits surrounding blood vessels, while some work suggests that tumor stem cells themselves directly differentiate into endothelial cells, yet the molecular and cellular dynamics of the endothelium in glioma are poorly characterized. The goal of this study was to establish molecular and morphological benchmarks for tumor associated vessels (TAVs) and tumor derived endothelial cells (TDECs) during glioblastoma progression. METHODS Using In-Utero Electroporation and CRISPR/Cas9 genome engineering to generate a native, immunocompetent mouse model of glioma, we characterized vascular-tumor dynamics in three dimensions during tumor progression. We employed bulk and single-cell RNA-Sequencing to elucidate the relationship between TAVs and TDECs. We confirmed our findings in a patient derived orthotopic xenograft (PDOX) model. RESULTS Using a mouse model of glioma, we identified progressive alteration of vessel function and morphogenesis over time. We also showed in our mouse model that TDECs are a rare subpopulation that contributes to vessels within the tumor, albeit to a limited degree. Furthermore, transcriptional profiling demonstrates that both TAVs and TDECs are molecularly distinct, and both populations feature extensive molecular heterogeneity. Finally, the distinct molecular signatures of these heterogeneous populations are also present in human glioma. CONCLUSIONS Our findings show extensive endothelial heterogeneity within the tumor and tumor microenvironment and provide insights into the diverse cellular and molecular mechanisms that drive glioma vascularization and angiogenesis during tumorigenesis.
Collapse
Affiliation(s)
- Jeff C Carlson
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas.,Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
| | - Manuel Cantu Gutierrez
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas.,Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas.,Cardiovascular Research Institute, Baylor College of Medicine, Houston, Texas
| | - Brittney Lozzi
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
| | - Emmet Huang-Hobbs
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas.,The Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, Texas
| | - Williamson D Turner
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas.,Cardiovascular Research Institute, Baylor College of Medicine, Houston, Texas.,Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas
| | - Burak Tepe
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Yiqun Zhang
- Dan L Duncan Cancer Center, Division of Biostatistics, Baylor College of Medicine, Houston, Texas
| | - Alexander M Herman
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas.,Cardiovascular Research Institute, Baylor College of Medicine, Houston, Texas
| | - Ganesh Rao
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas
| | - Chad J Creighton
- Dan L Duncan Cancer Center, Division of Biostatistics, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Joshua D Wythe
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas.,Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas.,Cardiovascular Research Institute, Baylor College of Medicine, Houston, Texas.,Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas
| | - Benjamin Deneen
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas.,Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas.,Department of Neurosurgery, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
10
|
Winkler F. In glioma, all endothelial cells are not created the same. Neuro Oncol 2021; 23:863-864. [PMID: 33705550 DOI: 10.1093/neuonc/noab065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Frank Winkler
- Neurology Clinic, University Hospital Heidelberg, Neurooncology Program at the National Center for Tumor Disease, German Cancer Consortium (DKTK), Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
11
|
Li F, Xu J, Liu S. Cancer Stem Cells and Neovascularization. Cells 2021; 10:cells10051070. [PMID: 33946480 PMCID: PMC8147173 DOI: 10.3390/cells10051070] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/19/2021] [Accepted: 04/27/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer stem cells (CSCs) refer to a subpopulation of cancer cells responsible for tumorigenesis, metastasis, and drug resistance. Increasing evidence suggests that CSC-associated tumor neovascularization partially contributes to the failure of cancer treatment. In this review, we discuss the roles of CSCs on tumor-associated angiogenesis via trans-differentiation or forming the capillary-like vasculogenic mimicry, as well as the roles of CSCs on facilitating endothelial cell-involved angiogenesis to support tumor progression and metastasis. Furthermore, we discuss the underlying regulation mechanisms, including the intrinsic signals of CSCs and the extrinsic signals such as cytokines from the tumor microenvironment. Further research is required to identify and verify some novel targets to develop efficient therapeutic approaches for more efficient cancer treatment through interfering CSC-mediated neovascularization.
Collapse
Affiliation(s)
- Fengkai Li
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences, Cancer Institutes, Fudan University, Shanghai 200032, China; (F.L.); (J.X.)
- Key Laboratory of Breast Cancer in Shanghai, The Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai 200032, China
- The International Co-Laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jiahui Xu
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences, Cancer Institutes, Fudan University, Shanghai 200032, China; (F.L.); (J.X.)
- Key Laboratory of Breast Cancer in Shanghai, The Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai 200032, China
- The International Co-Laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Suling Liu
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences, Cancer Institutes, Fudan University, Shanghai 200032, China; (F.L.); (J.X.)
- Key Laboratory of Breast Cancer in Shanghai, The Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai 200032, China
- The International Co-Laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Correspondence: ; Tel.: +86-21-34771023
| |
Collapse
|
12
|
Piantino M, Figarol A, Matsusaki M. Three-Dimensional in vitro Models of Healthy and Tumor Brain Microvasculature for Drug and Toxicity Screening. FRONTIERS IN TOXICOLOGY 2021; 3:656254. [PMID: 35295158 PMCID: PMC8915870 DOI: 10.3389/ftox.2021.656254] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 03/31/2021] [Indexed: 12/16/2022] Open
Abstract
Tissue vascularization is essential for its oxygenation and the homogenous diffusion of nutrients. Cutting-edge studies are focusing on the vascularization of three-dimensional (3D) in vitro models of human tissues. The reproduction of the brain vasculature is particularly challenging as numerous cell types are involved. Moreover, the blood-brain barrier, which acts as a selective filter between the vascular system and the brain, is a complex structure to replicate. Nevertheless, tremendous advances have been made in recent years, and several works have proposed promising 3D in vitro models of the brain microvasculature. They incorporate cell co-cultures organized in 3D scaffolds, often consisting of components of the native extracellular matrix (ECM), to obtain a micro-environment similar to the in vivo physiological state. These models are particularly useful for studying adverse effects on the healthy brain vasculature. They provide insights into the molecular and cellular events involved in the pathological evolutions of this vasculature, such as those supporting the appearance of brain cancers. Glioblastoma multiform (GBM) is the most common form of brain cancer and one of the most vascularized solid tumors. It is characterized by a high aggressiveness and therapy resistance. Current conventional therapies are unable to prevent the high risk of recurrence of the disease. Most of the new drug candidates fail to pass clinical trials, despite the promising results shown in vitro. The conventional in vitro models are unable to efficiently reproduce the specific features of GBM tumors. Recent studies have indeed suggested a high heterogeneity of the tumor brain vasculature, with the coexistence of intact and leaky regions resulting from the constant remodeling of the ECM by glioma cells. In this review paper, after summarizing the advances in 3D in vitro brain vasculature models, we focus on the latest achievements in vascularized GBM modeling, and the potential applications for both healthy and pathological models as platforms for drug screening and toxicological assays. Particular attention will be paid to discuss the relevance of these models in terms of cell-cell, cell-ECM interactions, vascularization and permeability properties, which are crucial parameters for improving in vitro testing accuracy.
Collapse
Affiliation(s)
- Marie Piantino
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Osaka, Japan
| | - Agathe Figarol
- Institut Jean Lamour, UMR 7198 CNRS, Université de Lorraine, Nancy, France
| | - Michiya Matsusaki
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Osaka, Japan
- *Correspondence: Michiya Matsusaki
| |
Collapse
|
13
|
Tumor Endothelial Cell-A Biological Tool for Translational Cancer Research. Int J Mol Sci 2020; 21:ijms21093238. [PMID: 32375250 PMCID: PMC7247330 DOI: 10.3390/ijms21093238] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 04/29/2020] [Accepted: 04/30/2020] [Indexed: 12/14/2022] Open
Abstract
Going from bench to bedside is a simplified description of translational research, with the ultimate goal being to improve the health status of mankind. Tumor endothelial cells (TECs) perform angiogenesis to support the growth, establishment, and dissemination of tumors to distant organs. TECs have various features that distinguish them from normal endothelial cells, which include alterations in gene expression patterns, higher angiogenic and metabolic activities, and drug resistance tendencies. The special characteristics of TECs enhance the vulnerability of tumor blood vessels toward antiangiogenic therapeutic strategies. Therefore, apart from being a viable therapeutic target, TECs would act as a better mediator between the bench (i.e., angiogenesis research) and the bedside (i.e., clinical application of drugs discovered through research). Exploitation of TEC characteristics could reveal unidentified strategies of enhancing and monitoring antiangiogenic therapy in the treatment of cancer, which are discussed in this review.
Collapse
|
14
|
Lugano R, Ramachandran M, Dimberg A. Tumor angiogenesis: causes, consequences, challenges and opportunities. Cell Mol Life Sci 2019; 77:1745-1770. [PMID: 31690961 PMCID: PMC7190605 DOI: 10.1007/s00018-019-03351-7] [Citation(s) in RCA: 954] [Impact Index Per Article: 190.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 10/10/2019] [Accepted: 10/21/2019] [Indexed: 02/07/2023]
Abstract
Tumor vascularization occurs through several distinct biological processes, which not only vary between tumor type and anatomic location, but also occur simultaneously within the same cancer tissue. These processes are orchestrated by a range of secreted factors and signaling pathways and can involve participation of non-endothelial cells, such as progenitors or cancer stem cells. Anti-angiogenic therapies using either antibodies or tyrosine kinase inhibitors have been approved to treat several types of cancer. However, the benefit of treatment has so far been modest, some patients not responding at all and others acquiring resistance. It is becoming increasingly clear that blocking tumors from accessing the circulation is not an easy task to accomplish. Tumor vessel functionality and gene expression often differ vastly when comparing different cancer subtypes, and vessel phenotype can be markedly heterogeneous within a single tumor. Here, we summarize the current understanding of cellular and molecular mechanisms involved in tumor angiogenesis and discuss challenges and opportunities associated with vascular targeting.
Collapse
Affiliation(s)
- Roberta Lugano
- The Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, 75185, Uppsala, Sweden
| | - Mohanraj Ramachandran
- The Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, 75185, Uppsala, Sweden
| | - Anna Dimberg
- The Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, 75185, Uppsala, Sweden.
| |
Collapse
|
15
|
Civita P, Franceschi S, Aretini P, Ortenzi V, Menicagli M, Lessi F, Pasqualetti F, Naccarato AG, Mazzanti CM. Laser Capture Microdissection and RNA-Seq Analysis: High Sensitivity Approaches to Explain Histopathological Heterogeneity in Human Glioblastoma FFPE Archived Tissues. Front Oncol 2019; 9:482. [PMID: 31231613 PMCID: PMC6568189 DOI: 10.3389/fonc.2019.00482] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 05/21/2019] [Indexed: 12/21/2022] Open
Abstract
Laser capture microdissection (LCM) coupled with RNA-seq is a powerful tool to identify genes that are differentially expressed in specific histological tumor subtypes. To better understand the role of single tumor cell populations in the complex heterogeneity of glioblastoma, we paired microdissection and NGS technology to study intra-tumoral differences into specific histological regions and cells of human GBM FFPE tumors. We here isolated astrocytes, neurons and endothelial cells in 6 different histological contexts: tumor core astrocytes, pseudopalisading astrocytes, perineuronal astrocytes in satellitosis, neurons with satellitosis, tumor blood vessels, and normal blood vessels. A customized protocol was developed for RNA amplification, library construction, and whole transcriptome analysis of each single portion. We first validated our protocol comparing the obtained RNA expression pattern with the gene expression levels of RNA-seq raw data experiments from the BioProject NCBI database, using Spearman's correlation coefficients calculation. We found a good concordance for pseudopalisading and tumor core astrocytes compartments (0.5 Spearman correlation) and a high concordance for perineuronal astrocytes, neurons, normal, and tumor endothelial cells compartments (0.7 Spearman correlation). Then, Principal Component Analysis and differential expression analysis were employed to find differences between tumor compartments and control tissue and between same cell types into distinct tumor contexts. Data consistent with the literature emerged, in which multiple therapeutic targets significant for glioblastoma (such as Integrins, Extracellular Matrix, transmembrane transport, and metabolic processes) play a fundamental role in the disease progression. Moreover, specific cellular processes have been associated with certain cellular subtypes within the tumor. Our results are promising and suggest a compelling method for studying glioblastoma heterogeneity in FFPE samples and its application in both prospective and retrospective studies.
Collapse
Affiliation(s)
| | | | | | - Valerio Ortenzi
- Department of Translational Research and New Technologies in Medicine and Surgery, Pisa University Hospital, Pisa, Italy
| | | | | | | | - Antonio Giuseppe Naccarato
- Department of Translational Research and New Technologies in Medicine and Surgery, Pisa University Hospital, Pisa, Italy
| | | |
Collapse
|
16
|
Vokuda RS, B H S, Madhugiri VS, Velusamy SK, Verma SK. The Expression of Leptin and Its Receptor During Tumorigenesis of Diffuse Gliomas such as Astrocytoma and Oligodendroglioma- Grade II, III and IV (NOS). Asian Pac J Cancer Prev 2019; 20:479-485. [PMID: 30803210 PMCID: PMC6897042 DOI: 10.31557/apjcp.2019.20.2.479] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Background: Leptin, an adipocytokine functions via the leptin receptor, OB-Rb that contains an intact intracellular
domain and activates the JAK/STAT signalling cascade. It stimulates growth, migration and invasion of cancer cells in
vitro potentiating angiogenesis. Recently, the involvement of leptin in tumor progression is being explored. Gliomas
exhibit poor prognosis, low survival rates demanding for novel therapeutic regimens resulting in discovery of many
potential biomarkers and pharmaceutical targets. We analysed the potential role of leptin and OB-Rb in carcinogenesis
of malignant gliomas. Methods: Sixty fresh tissue samples of diffuse gliomas were collected after tumor excision. Real
time PCR, immunohistochemical (IHC) analysis and western blot analysis were carried out to assess the expression of
leptin and its receptor. Results: The present study demonstrates the expression of leptin and LepR and their involvement
in tumor progression. Of the 60 cases, 57 cases (95%) and 53 cases (88.3%) showed amplification for leptin and
OB-Rb respectively. The expression of these proteins were measured semi-quantitatively and correlated with degree of
malignancy (p<0.05). The bands were visualised on western blot. Conclusion: Leptin may be valued as a pharmaceutical
target and anti-leptin compounds could be developed as drugs in mono- or combined therapies for these tumors.
Collapse
Affiliation(s)
- Ramya S Vokuda
- Department of Pathology, Jawaharlal Institute of Post Graduate Medical Education and Research (JIPMER), Puducherry, India.
| | | | | | | | | |
Collapse
|
17
|
De Pascalis I, Morgante L, Pacioni S, D'Alessandris QG, Giannetti S, Martini M, Ricci-Vitiani L, Malinverno M, Dejana E, Larocca LM, Pallini R. Endothelial trans-differentiation in glioblastoma recurring after radiotherapy. Mod Pathol 2018; 31:1361-1366. [PMID: 29713042 DOI: 10.1038/s41379-018-0046-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 02/03/2018] [Accepted: 02/04/2018] [Indexed: 01/09/2023]
Abstract
We hypothesized that in glioblastoma recurring after radiotherapy, a condition whereby the brain endothelium undergoes radiation-induced senescence, tumor cells with endothelial phenotype may be relevant for tumor neovascularization. Matched glioblastoma samples obtained at primary surgery and at surgery for tumor recurrence after radiotherapy, all expressing epidermal growth factor receptor variant III (EGFRvIII), were assessed by a technique that combines fluorescent in situ hybridization (FISH) for the EGFR/CEP7 chromosomal probe with immunostaining for endothelial cells (CD31) and activated pericytes (α Smooth Muscle Actin). Five EGFRvIII-expressing paired primary/recurrent glioblastoma samples, in which the tumor cells showed EGFR/CEP7 amplification, were then assessed by CD31 and α Smooth Muscle Actin immunofluorescence. In glomeruloid bodies, the ratio between CD31+ cells with amplified EGFR/CEP7 signal and the total CD31+ cells was 0.23 ± 0.09 (mean ± sem) and 0.63 ± 0.07 in primary tumors and in recurrent ones, respectively (p < 0.002, Student-t test). In capillaries, the ratio of CD31+ cells with amplified EGFR/CEP7 over the total CD31+ cells lining the capillary lumen was 0.21 ± 0.06 (mean ± sem) and 0.42 ± 0.07 at primary surgery and at recurrence, respectively (p < 0.005, Student-t test). Expression of α Smooth Muscle Actin by cells with EGFR/CEP7 amplification was not observed. Then, in glioblastoma recurring after radiotherapy, where the brain endothelium suffers from radiation-induced cell senescence, tumor-derived endothelium plays a role in neo-vascularization.
Collapse
Affiliation(s)
- Ivana De Pascalis
- Institute of Neurosurgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Liliana Morgante
- Institute of Human Anatomy, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Simone Pacioni
- Institute of Neurosurgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | | | - Stefano Giannetti
- Institute of Human Anatomy, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Maurizio Martini
- Institute of Pathology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Lucia Ricci-Vitiani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Matteo Malinverno
- New Strategies to Inhibit Tumor Angiogenesis Program Fondazione Italiana per la Ricerca sul Cancro, Institute of Molecular Oncology Fondazione, Milan, Italy
| | - Elisabetta Dejana
- New Strategies to Inhibit Tumor Angiogenesis Program Fondazione Italiana per la Ricerca sul Cancro, Institute of Molecular Oncology Fondazione, Milan, Italy
| | - Luigi M Larocca
- Institute of Pathology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Roberto Pallini
- Institute of Neurosurgery, Università Cattolica del Sacro Cuore, Rome, Italy.
| |
Collapse
|
18
|
Hira VV, Aderetti DA, van Noorden CJ. Glioma Stem Cell Niches in Human Glioblastoma Are Periarteriolar. J Histochem Cytochem 2018; 66:349-358. [PMID: 29328867 PMCID: PMC5958355 DOI: 10.1369/0022155417752676] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 12/11/2017] [Indexed: 12/22/2022] Open
Abstract
Survival of primary brain tumor (glioblastoma) patients is seriously hampered by glioma stem cells (GSCs) that are distinct therapy-resistant self-replicating pluripotent cancer cells. GSCs reside in GSC niches, which are specific protective microenvironments in glioblastoma tumors. We have recently found that GSC niches are hypoxic periarteriolar, whereas in most studies, GSC niches are identified as hypoxic perivascular. The aim of this review is to critically evaluate the literature on perivascular GSC niches to establish whether these are periarteriolar, pericapillary, perivenular, and/or perilymphatic. We found six publications showing images of human glioblastoma tissue containing perivascular GSC niches without any specification of the vessel type. However, it is frequently assumed that these vessels are capillaries which are exchange vessels, whereas arterioles and venules are transport vessels. Closer inspection of the figures of these publications showed vessels that were not capillaries. Whether these vessels were arterioles or venules was difficult to determine in one case, but in the other cases, these were clearly arterioles and their perivascular niches were similar to the periarteriolar niches we have found. Therefore, we conclude that in human glioblastoma tumors, GSC niches are hypoxic periarteriolar and are structurally and functionally look-alikes of hematopoietic stem cell niches in the bone marrow.
Collapse
Affiliation(s)
- Vashendriya V.V. Hira
- Cancer Center Amsterdam, Department of Medical Biology at the Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Diana A. Aderetti
- Cancer Center Amsterdam, Department of Medical Biology at the Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Cornelis J.F. van Noorden
- Cancer Center Amsterdam, Department of Medical Biology at the Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
19
|
Aderetti DA, Hira VVV, Molenaar RJ, van Noorden CJF. The hypoxic peri-arteriolar glioma stem cell niche, an integrated concept of five types of niches in human glioblastoma. Biochim Biophys Acta Rev Cancer 2018; 1869:346-354. [PMID: 29684521 DOI: 10.1016/j.bbcan.2018.04.008] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Revised: 04/17/2018] [Accepted: 04/18/2018] [Indexed: 12/22/2022]
Abstract
Glioblastoma is the most lethal primary brain tumor and poor survival of glioblastoma patients is attributed to the presence of glioma stem cells (GSCs). These therapy-resistant, quiescent and pluripotent cells reside in GSC niches, which are specific microenvironments that protect GSCs against radiotherapy and chemotherapy. We previously showed the existence of hypoxic peri-arteriolar GSC niches in glioblastoma tumor samples. However, other studies have described peri-vascular niches, peri-hypoxic niches, peri-immune niches and extracellular matrix niches of GSCs. The aim of this review was to critically evaluate the literature on these five different types of GSC niches. In the present review, we describe that the five niche types are not distinct from one another, but should be considered to be parts of one integral GSC niche model, the hypoxic peri-arteriolar GSC niche. Moreover, hypoxic peri-arteriolar GSC niches are structural and functional look-alikes of hematopoietic stem cell (HSC) niches in the bone marrow. GSCs are maintained in peri-arteriolar niches by the same receptor-ligand interactions as HSCs in bone marrow. Our concept should be rigidly tested in the near future and applied to develop therapies to expel and keep GSCs out of their protective niches to render them more vulnerable to standard therapies.
Collapse
Affiliation(s)
- Diana A Aderetti
- Department of Medical Biology, Cancer Center Amsterdam at the Academic Medical Center, University of Amsterdam, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands
| | - Vashendriya V V Hira
- Department of Medical Biology, Cancer Center Amsterdam at the Academic Medical Center, University of Amsterdam, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands
| | - Remco J Molenaar
- Department of Medical Biology, Cancer Center Amsterdam at the Academic Medical Center, University of Amsterdam, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands; Department of Medical Oncology, Cancer Center Amsterdam at the Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Cornelis J F van Noorden
- Department of Medical Biology, Cancer Center Amsterdam at the Academic Medical Center, University of Amsterdam, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands; Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna pot 111, 1000 Ljubljana, Slovenia.
| |
Collapse
|
20
|
IDH1 mutation is associated with lower expression of VEGF but not microvessel formation in glioblastoma multiforme. Oncotarget 2018; 9:16462-16476. [PMID: 29662659 PMCID: PMC5893254 DOI: 10.18632/oncotarget.24536] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 02/10/2018] [Indexed: 12/18/2022] Open
Abstract
Introduction Glioblastoma multiforme (GBM) represents the most malignant primary brain tumor characterized by pathological vascularization. Mutations in isocitrate dehydrogenases 1 and 2 (IDH1 and IDH2) were observed in GBM. We aimed to assess the intra-tumor hypoxia, angiogenesis and microvessel formation in GBM and to find their associations with IDH1 mutation status and patients prognosis. Methods 52 patients with a diagnosis of GBM were included into the study. IDH1 R132H mutation was assessed by RT-PCR from FFPE tumor samples obtained during surgery. The expression of markers of hypoxia (HIF2α), angiogenesis (VEGF), tumor microvascularity (CD31, CD34, vWF, CD105), and proliferation (Ki-67) were assessed immunohistochemically (IHC). IDH1 mutation and IHC markers were correlated with the patient survival. Results 20 from 52 GBM tumor samples comprised IDH1 R132H mutation (38.5%). The majority of mutated tumors were classified as secondary glioblastomas (89.9%). Patients with IDH1 mutated tumors experienced better progression-free survival (P = 0.037) as well as overall survival (P = 0.035) compared with wild type tumors. The significantly lower expression of VEGF was observed in GBM with IDH1 mutation than in wild type tumors (P = 0.01). No such association was found for microvascular markers. The increased expression of newly-formed microvessels (ratio CD105/CD31) in tumor samples was associated with worse patient’s progression-free survival (P = 0.026). Summary No increase in HIF/VEGF-mediated angiogenesis was observed in IDH1-mutated GBM compared with IDH1 wild type tumors. The histological assessment of the portion of newly-formed microvessels in tumor tissue can be used for the prediction of GBM patient’s prognosis.
Collapse
|
21
|
Yan H, Romero-López M, Benitez LI, Di K, Frieboes HB, Hughes CCW, Bota DA, Lowengrub JS. Multiscale modeling of glioblastoma. Transl Cancer Res 2018; 7:S96-S98. [PMID: 30211018 PMCID: PMC6130886 DOI: 10.21037/tcr.2017.12.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Huaming Yan
- Department of Mathematics, University of California, Irvine, USA
| | | | - Lesly I. Benitez
- Department of Molecular Biology and Biochemistry, University of California, Irvine, USA
| | - Kaijun Di
- Department of Neurology, University of California, Irvine, USA
| | - Hermann B. Frieboes
- James Graham Brown Cancer Center, University of Louisville, Louisville, USA
- Department of Bioengineering, University of Louisville, Louisville, USA
| | - Christopher C. W. Hughes
- Department of Biomedical Engineering, University of California, Irvine, USA
- Department of Molecular Biology and Biochemistry, University of California, Irvine, USA
- Chao Comprehensive Cancer Center, University of California, Irvine, USA
- Center for Complex Biological Systems, University of California, Irvine, USA
| | - Daniela A. Bota
- Department of Neurology, University of California, Irvine, USA
- Chao Comprehensive Cancer Center, University of California, Irvine, USA
- Department of Neurological Surgery, University of California, Irvine, USA
| | - John S. Lowengrub
- Department of Mathematics, University of California, Irvine, USA
- Department of Biomedical Engineering, University of California, Irvine, USA
- Chao Comprehensive Cancer Center, University of California, Irvine, USA
- Center for Complex Biological Systems, University of California, Irvine, USA
| |
Collapse
|
22
|
Mansouri S, Singh S, Alamsahebpour A, Burrell K, Li M, Karabork M, Ekinci C, Koch E, Solaroglu I, Chang JT, Wouters B, Aldape K, Zadeh G. DICER governs characteristics of glioma stem cells and the resulting tumors in xenograft mouse models of glioblastoma. Oncotarget 2018; 7:56431-56446. [PMID: 27421140 PMCID: PMC5302925 DOI: 10.18632/oncotarget.10570] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 05/19/2016] [Indexed: 12/21/2022] Open
Abstract
The RNAse III endonuclease DICER is a key regulator of microRNA (miRNA) biogenesis and is frequently decreased in a variety of malignancies. We characterized the role of DICER in glioblastoma (GB), specifically demonstrating its effects on the ability of glioma stem-like cells (GSCs) to form tumors in a mouse model of GB. DICER silencing in GSCs reduced their stem cell characteristics, while tumors arising from these cells were more aggressive, larger in volume, and displayed a higher proliferation index and lineage differentiation. The resulting tumors, however, were more sensitive to radiation treatment. Our results demonstrate that DICER silencing enhances the tumorigenic potential of GSCs, providing a platform for analysis of specific relevant miRNAs and development of potentially novel therapies against GB.
Collapse
Affiliation(s)
- Sheila Mansouri
- Princess Margaret Cancer Centre and MacFeeters-Hamilton Centre for Neuro-Oncology Research, Toronto, ON, Canada
| | - Sanjay Singh
- Princess Margaret Cancer Centre and MacFeeters-Hamilton Centre for Neuro-Oncology Research, Toronto, ON, Canada
| | - Amir Alamsahebpour
- Princess Margaret Cancer Centre and MacFeeters-Hamilton Centre for Neuro-Oncology Research, Toronto, ON, Canada
| | - Kelly Burrell
- Princess Margaret Cancer Centre and MacFeeters-Hamilton Centre for Neuro-Oncology Research, Toronto, ON, Canada
| | - Mira Li
- Princess Margaret Cancer Centre and MacFeeters-Hamilton Centre for Neuro-Oncology Research, Toronto, ON, Canada
| | - Merve Karabork
- School of Medicine, Koç University, Rumelifeneri Yolu, Sariyer, Istanbul, Turkey
| | - Can Ekinci
- School of Medicine, Koç University, Rumelifeneri Yolu, Sariyer, Istanbul, Turkey
| | - Elizabeth Koch
- Ontario Cancer Institute and Campbell Family Institute for Cancer Research, Princess Margaret Cancer Centre, Toronto, ON, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Ihsan Solaroglu
- School of Medicine, Koç University, Rumelifeneri Yolu, Sariyer, Istanbul, Turkey.,Loma Linda University, School of Medicine, Loma Linda, CA, USA
| | - Jeffery T Chang
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas, Houston, TX, USA
| | - Bradly Wouters
- Ontario Cancer Institute and Campbell Family Institute for Cancer Research, Princess Margaret Cancer Centre, Toronto, ON, Canada.,Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, ON, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Kenneth Aldape
- Princess Margaret Cancer Centre and MacFeeters-Hamilton Centre for Neuro-Oncology Research, Toronto, ON, Canada
| | - Gelareh Zadeh
- Princess Margaret Cancer Centre and MacFeeters-Hamilton Centre for Neuro-Oncology Research, Toronto, ON, Canada.,Department of Neurosurgery, Toronto Western Hospital, University Health Network, 4W-436, Toronto, ON, Canada
| |
Collapse
|
23
|
|
24
|
Molecular crosstalk between tumour and brain parenchyma instructs histopathological features in glioblastoma. Oncotarget 2017; 7:31955-71. [PMID: 27049916 PMCID: PMC5077988 DOI: 10.18632/oncotarget.7454] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 01/29/2016] [Indexed: 12/11/2022] Open
Abstract
The histopathological and molecular heterogeneity of glioblastomas represents a major obstacle for effective therapies. Glioblastomas do not develop autonomously, but evolve in a unique environment that adapts to the growing tumour mass and contributes to the malignancy of these neoplasms. Here, we show that patient-derived glioblastoma xenografts generated in the mouse brain from organotypic spheroids reproducibly give rise to three different histological phenotypes: (i) a highly invasive phenotype with an apparent normal brain vasculature, (ii) a highly angiogenic phenotype displaying microvascular proliferation and necrosis and (iii) an intermediate phenotype combining features of invasion and vessel abnormalities. These phenotypic differences were visible during early phases of tumour development suggesting an early instructive role of tumour cells on the brain parenchyma. Conversely, we found that tumour-instructed stromal cells differentially influenced tumour cell proliferation and migration in vitro, indicating a reciprocal crosstalk between neoplastic and non-neoplastic cells. We did not detect any transdifferentiation of tumour cells into endothelial cells. Cell type-specific transcriptomic analysis of tumour and endothelial cells revealed a strong phenotype-specific molecular conversion between the two cell types, suggesting co-evolution of tumour and endothelial cells. Integrative bioinformatic analysis confirmed the reciprocal crosstalk between tumour and microenvironment and suggested a key role for TGFβ1 and extracellular matrix proteins as major interaction modules that shape glioblastoma progression. These data provide novel insight into tumour-host interactions and identify novel stroma-specific targets that may play a role in combinatorial treatment strategies against glioblastoma.
Collapse
|
25
|
Zhang L, Wang Y, Rashid MH, Liu M, Angara K, Mivechi NF, Maihle NJ, Arbab AS, Ko L. Malignant pericytes expressing GT198 give rise to tumor cells through angiogenesis. Oncotarget 2017; 8:51591-51607. [PMID: 28881671 PMCID: PMC5584272 DOI: 10.18632/oncotarget.18196] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 05/01/2017] [Indexed: 12/21/2022] Open
Abstract
Angiogenesis promotes tumor development. Understanding the crucial factors regulating tumor angiogenesis may reveal new therapeutic targets. Human GT198 (PSMC3IP or Hop2) is an oncoprotein encoded by a DNA repair gene that is overexpressed in tumor stromal vasculature to stimulate the expression of angiogenic factors. Here we show that pericytes expressing GT198 give rise to tumor cells through angiogenesis. GT198+ pericytes and perivascular cells are commonly present in the stromal compartment of various human solid tumors and rodent xenograft tumor models. In human oral cancer, GT198+ pericytes proliferate into GT198+ tumor cells, which migrate into lymph nodes. Increased GT198 expression is associated with increased lymph node metastasis and decreased progression-free survival in oral cancer patients. In rat brain U-251 glioblastoma xenografts, GT198+ pericytes of human tumor origin encase endothelial cells of rat origin to form mosaic angiogenic blood vessels, and differentiate into pericyte-derived tumor cells. The net effect is continued production of glioblastoma tumor cells from malignant pericytes via angiogenesis. In addition, activation of GT198 induces the expression of VEGF and promotes tube formation in cultured U251 cells. Furthermore, vaccination using GT198 protein as an antigen in mouse xenograft of GL261 glioma delayed tumor growth and prolonged mouse survival. Together, these findings suggest that GT198-expressing malignant pericytes can give rise to tumor cells through angiogenesis, and serve as a potential source of cells for distant metastasis. Hence, the oncoprotein GT198 has the potential to be a new target in anti-angiogenic therapies in human cancer.
Collapse
Affiliation(s)
- Liyong Zhang
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Yan Wang
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Mohammad H. Rashid
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Min Liu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Kartik Angara
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Nahid F. Mivechi
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Nita J. Maihle
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA, USA
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Ali S. Arbab
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Lan Ko
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA, USA
- Department of Pathology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| |
Collapse
|
26
|
Audia A, Conroy S, Glass R, Bhat KPL. The Impact of the Tumor Microenvironment on the Properties of Glioma Stem-Like Cells. Front Oncol 2017; 7:143. [PMID: 28740831 PMCID: PMC5502267 DOI: 10.3389/fonc.2017.00143] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 06/20/2017] [Indexed: 01/22/2023] Open
Abstract
Glioblastoma is the most common and highly malignant primary brain tumor, and patients affected with this disease exhibit a uniformly dismal prognosis. Glioma stem-like cells (GSCs) are a subset of cells within the bulk tumor that possess self-renewal and multi-lineage differentiation properties similar to somatic stem cells. These cells also are at the apex of the cellular hierarchy and cause tumor initiation and expansion after chemo-radiation. These traits make them an attractive target for therapeutic development. Because GSCs are dependent on the brain microenvironment for their growth, and because non-tumorigenic cell types in the microenvironment can influence GSC phenotypes and treatment response, a better understanding of these cell types is needed. In this review, we provide a focused overview of the contributions from the microenvironment to GSC homing, maintenance, phenotypic plasticity, and tumor initiation. The interaction of GSCs with the vascular compartment, mesenchymal stem cells, immune system, and normal brain cell types are discussed. Studies that provide mechanistic insight into each of these GSC–microenvironment interactions are warranted in the future.
Collapse
Affiliation(s)
- Alessandra Audia
- Department of Translational Molecular Pathology, University of Texas, M.D. Anderson Cancer Center, Houston, TX, United States
| | - Siobhan Conroy
- Department of Translational Molecular Pathology, University of Texas, M.D. Anderson Cancer Center, Houston, TX, United States.,Department of Pathology and Medical Biology, University Medical Center, Groningen, Netherlands
| | - Rainer Glass
- Neurosurgical Research, Department of Neurosurgery, University Hospital, LMU Munich, Munich, Germany.,German Cancer Consortium (DKTK) partner site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Krishna P L Bhat
- Department of Translational Molecular Pathology, University of Texas, M.D. Anderson Cancer Center, Houston, TX, United States.,Department of Neurosurgery, University of Texas, M.D. Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
27
|
Contribution of the Microenvironmental Niche to Glioblastoma Heterogeneity. BIOMED RESEARCH INTERNATIONAL 2017. [PMID: 28630875 PMCID: PMC5467280 DOI: 10.1155/2017/9634172] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Glioblastoma is the most aggressive cancer of the brain. The dismal prognosis is largely attributed to the heterogeneous nature of the tumor, which in addition to intrinsic molecular and genetic changes is also influenced by the microenvironmental niche in which the glioma cells reside. The cancer stem cells (CSCs) hypothesis suggests that all cancers arise from CSCs that possess the ability to self-renew and initiate tumor formation. CSCs reside in specialized niches where interaction with the microenvironment regulates their stem cell behavior. The reciprocal interaction between glioma stem cells (GSCs) and cells from the microenvironment, such as endothelial cells, immune cells, and other parenchymal cells, may also promote angiogenesis, invasion, proliferation, and stemness of the GSCs and be likely to have an underappreciated role in their responsiveness to therapy. This crosstalk may also promote molecular transition of GSCs. Hence the inherent plasticity of GSCs can be seen as an adaptive response, changing according to the signaling cue from the niche. Given the association of GSCs with tumor recurrence and treatment sensitivity, understanding this bidirectional crosstalk between GSCs and its niche may provide a framework to identify more effective therapeutic targets and improve treatment outcome.
Collapse
|
28
|
Yan H, Romero-López M, Benitez LI, Di K, Frieboes HB, Hughes CCW, Bota DA, Lowengrub JS. 3D Mathematical Modeling of Glioblastoma Suggests That Transdifferentiated Vascular Endothelial Cells Mediate Resistance to Current Standard-of-Care Therapy. Cancer Res 2017; 77:4171-4184. [PMID: 28536277 DOI: 10.1158/0008-5472.can-16-3094] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 02/24/2017] [Accepted: 05/16/2017] [Indexed: 01/17/2023]
Abstract
Glioblastoma (GBM), the most aggressive brain tumor in human patients, is decidedly heterogeneous and highly vascularized. Glioma stem/initiating cells (GSC) are found to play a crucial role by increasing cancer aggressiveness and promoting resistance to therapy. Recently, cross-talk between GSC and vascular endothelial cells has been shown to significantly promote GSC self-renewal and tumor progression. Furthermore, GSC also transdifferentiate into bona fide vascular endothelial cells (GEC), which inherit mutations present in GSC and are resistant to traditional antiangiogenic therapies. Here we use three-dimensional mathematical modeling to investigate GBM progression and response to therapy. The model predicted that GSCs drive invasive fingering and that GEC spontaneously form a network within the hypoxic core, consistent with published experimental findings. Standard-of-care treatments using DNA-targeted therapy (radiation/chemo) together with antiangiogenic therapies reduced GBM tumor size but increased invasiveness. Anti-GEC treatments blocked the GEC support of GSCs and reduced tumor size but led to increased invasiveness. Anti-GSC therapies that promote differentiation or disturb the stem cell niche effectively reduced tumor invasiveness and size, but were ultimately limited in reducing tumor size because GECs maintain GSCs. Our study suggests that a combinatorial regimen targeting the vasculature, GSCs, and GECs, using drugs already approved by the FDA, can reduce both tumor size and invasiveness and could lead to tumor eradication. Cancer Res; 77(15); 4171-84. ©2017 AACR.
Collapse
Affiliation(s)
- Huaming Yan
- Department of Mathematics, University of California, Irvine, California
| | - Mónica Romero-López
- Department of Biomedical Engineering, University of California, Irvine, California
| | - Lesly I Benitez
- Department of Molecular Biology and Biochemistry, University of California, Irvine, California
| | - Kaijun Di
- Chao Comprehensive Cancer Center, University of California, Irvine, California.,Department of Neurological Surgery, University of California, Irvine, California
| | - Hermann B Frieboes
- James Graham Brown Cancer Center, University of Louisville.,Department of Bioengineering, University of Louisville, Louisville, Kentucky
| | - Christopher C W Hughes
- Department of Biomedical Engineering, University of California, Irvine, California.,Department of Molecular Biology and Biochemistry, University of California, Irvine, California.,Chao Comprehensive Cancer Center, University of California, Irvine, California.,Center for Complex Biological Systems, University of California, Irvine, California
| | - Daniela A Bota
- Chao Comprehensive Cancer Center, University of California, Irvine, California.,Department of Neurological Surgery, University of California, Irvine, California.,Department of Neurology, University of California, Irvine, California
| | - John S Lowengrub
- Department of Mathematics, University of California, Irvine, California. .,Department of Biomedical Engineering, University of California, Irvine, California.,Chao Comprehensive Cancer Center, University of California, Irvine, California.,Center for Complex Biological Systems, University of California, Irvine, California
| |
Collapse
|
29
|
Mahase S, Rattenni RN, Wesseling P, Leenders W, Baldotto C, Jain R, Zagzag D. Hypoxia-Mediated Mechanisms Associated with Antiangiogenic Treatment Resistance in Glioblastomas. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:940-953. [PMID: 28284719 PMCID: PMC5417003 DOI: 10.1016/j.ajpath.2017.01.010] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 12/31/2016] [Accepted: 01/05/2017] [Indexed: 12/28/2022]
Abstract
Glioblastomas (GBMs) are malignant tumors characterized by their vascularity and invasive capabilities. Antiangiogenic therapy (AAT) is a treatment option that targets GBM-associated vasculature to mitigate the growth of GBMs. However, AAT demonstrates transient effects because many patients eventually develop resistance to this treatment. Several recent studies attempt to explain the molecular and biochemical basis of resistance to AAT in GBM patients. Experimental investigations suggest that the induction of extensive intratumoral hypoxia plays a key role in GBM escape from AAT. In this review, we examine AAT resistance in GBMs, with an emphasis on six potential hypoxia-mediated mechanisms: enhanced invasion and migration, including increased expression of matrix metalloproteinases and activation of the c-MET tyrosine kinase pathway; shifts in cellular metabolism, including up-regulation of hypoxia inducible factor-1α's downstream processes and the Warburg effect; induction of autophagy; augmentation of GBM stem cell self-renewal; possible implications of GBM-endothelial cell transdifferentiation; and vasoformative responses, including vasculogenesis, alternative angiogenic pathways, and vascular mimicry. Juxtaposing recent studies on well-established resistance pathways with that of emerging mechanisms highlights the overall complexity of GBM treatment resistance while also providing direction for further investigation.
Collapse
Affiliation(s)
- Sean Mahase
- Microvascular and Molecular Neuro-Oncology Laboratory, New York University School of Medicine, New York, New York
| | - Rachel N Rattenni
- Microvascular and Molecular Neuro-Oncology Laboratory, New York University School of Medicine, New York, New York
| | - Pieter Wesseling
- Department of Pathology, VU University Medical Center, Amsterdam, the Netherlands; Department of Pathology, Princess Máxima Center for Pediatric Oncology and University Medical Center, Utrecht, the Netherlands
| | - William Leenders
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Clarissa Baldotto
- Medical Oncology, Instituto Nacionale de Cancer, Rio de Janeiro, Brazil
| | - Rajan Jain
- Department of Radiology, New York University School of Medicine, New York, New York; Department of Neurosurgery, New York University School of Medicine, New York, New York
| | - David Zagzag
- Microvascular and Molecular Neuro-Oncology Laboratory, New York University School of Medicine, New York, New York; Department of Neurosurgery, New York University School of Medicine, New York, New York; Division of Neuropathology, Department of Pathology, New York University School of Medicine, New York, New York; Laura and Isaac Perlmutter Cancer Center, New York University School of Medicine, New York, New York.
| |
Collapse
|
30
|
Hu B, Wang Q, Wang YA, Hua S, Sauvé CEG, Ong D, Lan ZD, Chang Q, Ho YW, Monasterio MM, Lu X, Zhong Y, Zhang J, Deng P, Tan Z, Wang G, Liao WT, Corley LJ, Yan H, Zhang J, You Y, Liu N, Cai L, Finocchiaro G, Phillips JJ, Berger MS, Spring DJ, Hu J, Sulman EP, Fuller GN, Chin L, Verhaak RGW, DePinho RA. Epigenetic Activation of WNT5A Drives Glioblastoma Stem Cell Differentiation and Invasive Growth. Cell 2017; 167:1281-1295.e18. [PMID: 27863244 DOI: 10.1016/j.cell.2016.10.039] [Citation(s) in RCA: 189] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 08/11/2016] [Accepted: 10/20/2016] [Indexed: 01/08/2023]
Abstract
Glioblastoma stem cells (GSCs) are implicated in tumor neovascularization, invasiveness, and therapeutic resistance. To illuminate mechanisms governing these hallmark features, we developed a de novo glioblastoma multiforme (GBM) model derived from immortalized human neural stem/progenitor cells (hNSCs) to enable precise system-level comparisons of pre-malignant and oncogene-induced malignant states of NSCs. Integrated transcriptomic and epigenomic analyses uncovered a PAX6/DLX5 transcriptional program driving WNT5A-mediated GSC differentiation into endothelial-like cells (GdECs). GdECs recruit existing endothelial cells to promote peritumoral satellite lesions, which serve as a niche supporting the growth of invasive glioma cells away from the primary tumor. Clinical data reveal higher WNT5A and GdECs expression in peritumoral and recurrent GBMs relative to matched intratumoral and primary GBMs, respectively, supporting WNT5A-mediated GSC differentiation and invasive growth in disease recurrence. Thus, the PAX6/DLX5-WNT5A axis governs the diffuse spread of glioma cells throughout the brain parenchyma, contributing to the lethality of GBM.
Collapse
Affiliation(s)
- Baoli Hu
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Qianghu Wang
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Y Alan Wang
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Sujun Hua
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | - Derrick Ong
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Zheng D Lan
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Qing Chang
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Institute for Applied Cancer Science, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yan Wing Ho
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Marta Moreno Monasterio
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xin Lu
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yi Zhong
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jianhua Zhang
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Institute for Applied Cancer Science, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Pingna Deng
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Zhi Tan
- Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Guocan Wang
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Wen-Ting Liao
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Lynda J Corley
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Haiyan Yan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Junxia Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Yongping You
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Ning Liu
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Linbo Cai
- Department of Oncology, Guangdong 999 Brain Hospital, Guangzhou 510510, China
| | - Gaetano Finocchiaro
- Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico C. Besta, 20133 Milano, Italy
| | - Joanna J Phillips
- Departments of Neurological Surgery and Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Mitchel S Berger
- Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Denise J Spring
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jian Hu
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Erik P Sulman
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Gregory N Fuller
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Lynda Chin
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Roeland G W Verhaak
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ronald A DePinho
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
31
|
Huse JT, Snuderl M, Jones DTW, Brathwaite CD, Altman N, Lavi E, Saffery R, Sexton-Oates A, Blumcke I, Capper D, Karajannis MA, Benayed R, Chavez L, Thomas C, Serrano J, Borsu L, Ladanyi M, Rosenblum MK. Polymorphous low-grade neuroepithelial tumor of the young (PLNTY): an epileptogenic neoplasm with oligodendroglioma-like components, aberrant CD34 expression, and genetic alterations involving the MAP kinase pathway. Acta Neuropathol 2017; 133:417-429. [PMID: 27812792 PMCID: PMC5325850 DOI: 10.1007/s00401-016-1639-9] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 10/25/2016] [Accepted: 10/25/2016] [Indexed: 02/07/2023]
Abstract
Epileptogenic tumors affecting children and young adults are a morphologically diverse collection of neuroepithelial neoplasms that, as a group, exhibit varying levels of glial and/or neuronal differentiation. Recent advances in molecular profiling technology, including comprehensive DNA sequencing and methylation analysis, have enabled the application of more precise and biologically relevant classification schemes to these tumors. In this report, we describe a morphologically and molecularly distinct epileptogenic neoplasm, the polymorphous low-grade neuroepithelial tumor of the young (PLNTY), which likely accounts for a sizable portion of oligodendroglioma-like tumors affecting the pediatric population. Characteristic microscopic findings most notably include infiltrative growth, the invariable presence of oligodendroglioma-like cellular components, and intense immunolabeling for cluster of differentiation 34 (CD34). Moreover, integrative molecular profiling reveals a distinct DNA methylation signature for PLNTYs, along with frequent genetic abnormalities involving either B-Raf proto-oncogene (BRAF) or fibroblast growth factor receptors 2 and 3 (FGFR2, FGFR3). These findings suggest that PLNTY represents a distinct biological entity within the larger spectrum of pediatric, low-grade neuroepithelial tumors.
Collapse
Affiliation(s)
- Jason T Huse
- Departments of Pathology and Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, 2130 W Holcombe Blvd, LSP9.4009, Houston, TX, 77030, USA.
| | - Matija Snuderl
- Department of Pathology, New York University Langone Medical Center, New York, NY, 10016, USA
| | - David T W Jones
- Division of Pediatric Neuro-oncology, German Cancer Research Center (DFKZ), 69120, Heidelberg, Germany
| | - Carole D Brathwaite
- Department of Pathology, Nicklaus Children's Hospital, Miami, FL, 33155, USA
| | - Nolan Altman
- Department of Radiology, Nicklaus Children's Hospital, Miami, FL, 33155, USA
| | - Ehud Lavi
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Richard Saffery
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, VIC, 3052, Australia
| | - Alexandra Sexton-Oates
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, VIC, 3052, Australia
| | - Ingmar Blumcke
- Institute of Neuropathology, University of Erlangen, 91054, Erlangen, Germany
| | - David Capper
- Department of Pathology, University of Heidelberg, 69120, Heidelberg, Germany
| | - Matthias A Karajannis
- Department of Pediatrics, New York University Langone Medical Center, New York, NY, 10016, USA
- Department of Ototlaryngology, New York University Langone Medical Center, New York, NY, 10016, USA
| | - Ryma Benayed
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, 408 E 69th St. (Z564), New York, NY, 10065, USA
| | - Lukas Chavez
- Division of Pediatric Neuro-oncology, German Cancer Research Center (DFKZ), 69120, Heidelberg, Germany
| | - Cheddhi Thomas
- Department of Pathology, New York University Langone Medical Center, New York, NY, 10016, USA
| | - Jonathan Serrano
- Department of Pathology, New York University Langone Medical Center, New York, NY, 10016, USA
| | - Laetitia Borsu
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, 408 E 69th St. (Z564), New York, NY, 10065, USA
| | - Marc Ladanyi
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, 408 E 69th St. (Z564), New York, NY, 10065, USA
| | - Marc K Rosenblum
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, 408 E 69th St. (Z564), New York, NY, 10065, USA
| |
Collapse
|
32
|
Yi Y, Hsieh IY, Huang X, Li J, Zhao W. Glioblastoma Stem-Like Cells: Characteristics, Microenvironment, and Therapy. Front Pharmacol 2016; 7:477. [PMID: 28003805 PMCID: PMC5141588 DOI: 10.3389/fphar.2016.00477] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 11/23/2016] [Indexed: 01/01/2023] Open
Abstract
Glioblastoma multiforme (GBM), grade IV astrocytoma, is the most fatal malignant primary brain tumor. GBM contains functional subsets of cells called glioblastoma stem-like cells (GSCs), which are radioresistant and chemoresistant and eventually lead to tumor recurrence. Recent studies showed that GSCs reside in particular tumor niches that are necessary to support their behavior. To successfully eradicate GBM growth and recurrence, new strategies selectively targeting GSCs and/or their microenvironmental niche should be designed. In this regard, here we focus on elucidating the molecular mechanisms that govern these GSC properties and on understanding the mechanism of the microenvironmental signals within the tumor mass. Moreover, to overcome the blood–brain barrier, which represents a critical limitation of GBM treatments, a new drug delivery system should be developed. Nanoparticles can be easily modified by different methods to facilitate delivery efficiency of chemotherapeutics, to enhance the accumulation within the tumors, and to promote the capacity for targeting the GSCs. Therefore, nanotechnology has become the most promising approach to GSC-targeting therapy. Additionally, we discussed the future of nanotechnology-based targeted therapy and point out the disadvantages that should be overcome.
Collapse
Affiliation(s)
- Yang Yi
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen UniversityGuangzhou, China; Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen UniversityGuangzhou, China
| | - I-Yun Hsieh
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Sun Yat-sen University Guangzhou, China
| | - Xiaojia Huang
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen UniversityGuangzhou, China; Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen UniversityGuangzhou, China
| | - Jie Li
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Sun Yat-sen University Guangzhou, China
| | - Wei Zhao
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen UniversityGuangzhou, China; Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen UniversityGuangzhou, China
| |
Collapse
|
33
|
Cerqueira-Coutinho C, Missailidis S, Alessandra-Perini J, Machado DE, Perini JA, Santos-Oliveira R. Comparison of biodistribution profile of monoclonal antibodies nanoparticles and aptamers in rats with breast cancer. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2016; 45:598-601. [DOI: 10.3109/21691401.2016.1163717] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
| | - Sotiris Missailidis
- Laboratory of Research in Pharmaceutical Sciences, Zona Oeste State University, Rio de Janeiro, Brazil
| | - Jéssica Alessandra-Perini
- Laboratory of Research in Pharmaceutical Sciences, Zona Oeste State University, Rio de Janeiro, Brazil
- Graduate Program in Public Health and Environment National School of Public Health, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Daniel Escorsim Machado
- Laboratory of Research in Pharmaceutical Sciences, Zona Oeste State University, Rio de Janeiro, Brazil
| | - Jamila Alessandra Perini
- Laboratory of Research in Pharmaceutical Sciences, Zona Oeste State University, Rio de Janeiro, Brazil
- Graduate Program in Public Health and Environment National School of Public Health, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- National Institute of Traumatology and Orthopedics, Rio de Janeiro, Brazil
| | - Ralph Santos-Oliveira
- Laboratory of Radiopharmacy and Nanoradiopharmaceuticals, Zona Oeste State University, Rio de Janeiro, Brazil
| |
Collapse
|
34
|
Glioma Stem Cells and Their Microenvironments: Providers of Challenging Therapeutic Targets. Stem Cells Int 2016; 2016:5728438. [PMID: 26977157 PMCID: PMC4764748 DOI: 10.1155/2016/5728438] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 11/12/2015] [Accepted: 01/06/2016] [Indexed: 12/26/2022] Open
Abstract
Malignant gliomas are aggressive brain tumors with limited therapeutic options, possibly because of highly tumorigenic subpopulations of glioma stem cells. These cells require specific microenvironments to maintain their “stemness,” described as perivascular and hypoxic niches. Each of those niches induces particular signatures in glioma stem cells (e.g., activation of Notch signaling, secretion of VEGF, bFGF, SDF1 for the vascular niche, activation of HIF2α, and metabolic reprogramming for hypoxic niche). Recently, accumulated knowledge on tumor-associated macrophages, possibly delineating a third niche, has underlined the role of immune cells in glioma progression, via specific chemoattractant factors and cytokines, such as macrophage-colony stimulation factor (M-CSF). The local or myeloid origin of this new component of glioma stem cells niche is yet to be determined. Such niches are being increasingly recognized as key regulators involved in multiple stages of disease progression, therapy resistance, immune-escaping, and distant metastasis, thereby substantially impacting the future development of frontline interventions in clinical oncology. This review focuses on the microenvironment impact on the glioma stem cell biology, emphasizing GSCs cross talk with hypoxic, perivascular, and immune niches and their potential use as targeted therapy.
Collapse
|
35
|
Glioma Stem Cells: Signaling, Microenvironment, and Therapy. Stem Cells Int 2016; 2016:7849890. [PMID: 26880988 PMCID: PMC4736567 DOI: 10.1155/2016/7849890] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 10/25/2015] [Indexed: 12/15/2022] Open
Abstract
Glioblastoma remains the most common and devastating primary brain tumor despite maximal therapy with surgery, chemotherapy, and radiation. The glioma stem cell (GSC) subpopulation has been identified in glioblastoma and likely plays a key role in resistance of these tumors to conventional therapies as well as recurrent disease. GSCs are capable of self-renewal and differentiation; glioblastoma-derived GSCs are capable of de novo tumor formation when implanted in xenograft models. Further, GSCs possess unique surface markers, modulate characteristic signaling pathways to promote tumorigenesis, and play key roles in glioma vascular formation. These features, in addition to microenvironmental factors, present possible targets for specifically directing therapy against the GSC population within glioblastoma. In this review, the authors summarize the current knowledge of GSC biology and function and the role of GSCs in new vascular formation within glioblastoma and discuss potential therapeutic approaches to target GSCs.
Collapse
|
36
|
Popescu AM, Purcaru SO, Alexandru O, Dricu A. New perspectives in glioblastoma antiangiogenic therapy. Contemp Oncol (Pozn) 2015; 20:109-18. [PMID: 27358588 PMCID: PMC4925727 DOI: 10.5114/wo.2015.56122] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 10/15/2015] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma (GB) is highly vascularised tumour, known to exhibit enhanced infiltrative potential. One of the characteristics of glioblastoma is microvascular proliferation surrounding necrotic areas, as a response to a hypoxic environment, which in turn increases the expression of angiogenic factors and their signalling pathways (RAS/RAF/ERK/MAPK pathway, PI3K/Akt signalling pathway and WTN signalling cascade). Currently, a small number of anti-angiogenic drugs, extending glioblastoma patients survival, are available for clinical use. Most medications are ineffective in clinical therapy of glioblastoma due to acquired malignant cells or intrinsic resistance, angiogenic receptors cross-activation and redundant intracellular signalling, or the inability of the drug to cross the blood-brain barrier and to reach its target in vivo. Researchers have also observed that GB tumours are different in many aspects, even when they derive from the same tissue, which is the reason for personalised therapy. An understanding of the molecular mechanisms regulating glioblastoma angiogenesis and invasion may be important in the future development of curative therapeutic approaches for the treatment of this devastating disease.
Collapse
Affiliation(s)
| | - Stefana Oana Purcaru
- Unit of Biochemistry, University of Medicine and Pharmacy of Craiova, Craiova, Romania
| | - Oana Alexandru
- Department of Neurology, University of Medicine and Pharmacy of Craiova and Clinical Hospital of Neuropsychiatry Craiova, Craiova, Romania
| | - Anica Dricu
- Unit of Biochemistry, University of Medicine and Pharmacy of Craiova, Craiova, Romania
| |
Collapse
|
37
|
Abstract
Glioblastomas (GBM) are one of the most recalcitrant brain tumors because of their aggressive invasive growth and resistance to therapy. They are highly heterogeneous malignancies at both the molecular and histological levels. Specific histological hallmarks including pseudopalisading necrosis and microvascular proliferation distinguish GBM from lower-grade gliomas, and make GBM one of the most hypoxic as well as angiogenic tumors. These microanatomical compartments present specific niches within the tumor microenvironment that regulate metabolic needs, immune surveillance, survival, invasion as well as cancer stem cell maintenance. Here we review features and functions of the distinct GBM niches, detail the different cell constituents and the functional status of the vasculature, and discuss prospects of therapeutically targeting GBM niche constituents.
Collapse
Affiliation(s)
- Dolores Hambardzumyan
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
| | - Gabriele Bergers
- Department of Neurological Surgery, University of California San Francisco, Helen Diller Family Cancer Research Center, 1450 3 Street, San Francisco, California 94158, USA; Brain Tumor Center, University of California San Francisco, Helen Diller Family Cancer Research Center, 1450 3 Street, San Francisco, California 94158, USA; UCSF Comprehensive Cancer Center, University of California San Francisco, Helen Diller Family Cancer Research Center, 1450 3 Street, San Francisco, California 94158, USA
| |
Collapse
|
38
|
Segarra M, Kirchmaier BC, Acker-Palmer A. A vascular perspective on neuronal migration. Mech Dev 2015; 138 Pt 1:17-25. [PMID: 26192337 DOI: 10.1016/j.mod.2015.07.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 07/09/2015] [Accepted: 07/13/2015] [Indexed: 12/21/2022]
Abstract
During CNS development and adult neurogenesis, immature neurons travel from the germinal zones towards their final destination using cellular substrates for their migration. Classically, radial glia and neuronal axons have been shown to act as physical scaffolds to support neuroblast locomotion in processes known as gliophilic and neurophilic migration, respectively (Hatten, 1999; Marin and Rubenstein, 2003; Rakic, 2003). In adulthood, long distance neuronal migration occurs in a glial-independent manner since radial glia cells differentiate into astrocytes after birth. A series of studies highlight a novel mode of neuronal migration that uses blood vessels as scaffolds, the so-called vasophilic migration. This migration mode allows neuroblast navigation in physiological and also pathological conditions, such as neuronal precursor migration after ischemic stroke or cerebral invasion of glioma tumor cells. Here we review the current knowledge about how vessels pave the path for migrating neurons and how trophic factors derived by glio-vascular structures guide neuronal migration both during physiological as well as pathological processes.
Collapse
Affiliation(s)
- Marta Segarra
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, Germany; Focus Program Translational Neurosciences (FTN), University of Mainz, Germany; Max Planck Institute for Brain Research, Frankfurt, Germany.
| | - Bettina C Kirchmaier
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, Germany; Focus Program Translational Neurosciences (FTN), University of Mainz, Germany; Max Planck Institute for Brain Research, Frankfurt, Germany
| | - Amparo Acker-Palmer
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, Germany; Focus Program Translational Neurosciences (FTN), University of Mainz, Germany; Max Planck Institute for Brain Research, Frankfurt, Germany.
| |
Collapse
|
39
|
Abstract
Glioblastoma is characterized by microvascular proliferation and a highly abnormal dysfunctional vasculature. The glioblastoma vessels differ significantly from normal brain vessels morphologically, functionally and molecularly. The present review provides a brief overview of the current understanding of the formation, functional abnormalities and specific gene expression of glioblastoma vessels and the consequences of vascular abnormalization for the tumour microenvironment.
Collapse
|
40
|
Mousavi HS, Monga V, Rao G, Rao AUK. Automated discrimination of lower and higher grade gliomas based on histopathological image analysis. J Pathol Inform 2015; 6:15. [PMID: 25838967 PMCID: PMC4382761 DOI: 10.4103/2153-3539.153914] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Accepted: 01/05/2015] [Indexed: 11/04/2022] Open
Abstract
INTRODUCTION Histopathological images have rich structural information, are multi-channel in nature and contain meaningful pathological information at various scales. Sophisticated image analysis tools that can automatically extract discriminative information from the histopathology image slides for diagnosis remain an area of significant research activity. In this work, we focus on automated brain cancer grading, specifically glioma grading. Grading of a glioma is a highly important problem in pathology and is largely done manually by medical experts based on an examination of pathology slides (images). To complement the efforts of clinicians engaged in brain cancer diagnosis, we develop novel image processing algorithms and systems to automatically grade glioma tumor into two categories: Low-grade glioma (LGG) and high-grade glioma (HGG) which represent a more advanced stage of the disease. RESULTS We propose novel image processing algorithms based on spatial domain analysis for glioma tumor grading that will complement the clinical interpretation of the tissue. The image processing techniques are developed in close collaboration with medical experts to mimic the visual cues that a clinician looks for in judging of the grade of the disease. Specifically, two algorithmic techniques are developed: (1) A cell segmentation and cell-count profile creation for identification of Pseudopalisading Necrosis, and (2) a customized operation of spatial and morphological filters to accurately identify microvascular proliferation (MVP). In both techniques, a hierarchical decision is made via a decision tree mechanism. If either Pseudopalisading Necrosis or MVP is found present in any part of the histopathology slide, the whole slide is identified as HGG, which is consistent with World Health Organization guidelines. Experimental results on the Cancer Genome Atlas database are presented in the form of: (1) Successful detection rates of pseudopalisading necrosis and MVP regions, (2) overall classification accuracy into LGG and HGG categories, and (3) receiver operating characteristic curves which can facilitate a desirable trade-off between HGG detection and false-alarm rates. CONCLUSION The proposed method demonstrates fairly high accuracy and compares favorably against best-known alternatives such as the state-of-the-art WND-CHARM feature set provided by NIH combined with powerful support vector machine classifier. Our results reveal that the proposed method can be beneficial to a clinician in effectively separating histopathology slides into LGG and HGG categories, particularly where the analysis of a large number of slides is needed. Our work also reveals that MVP regions are much harder to detect than Pseudopalisading Necrosis and increasing accuracy of automated image processing for MVP detection emerges as a significant future research direction.
Collapse
Affiliation(s)
- Hojjat Seyed Mousavi
- Department of Electrical Engineering, The Pennsylvania State University, University Park, PA, USA
| | - Vishal Monga
- Department of Electrical Engineering, The Pennsylvania State University, University Park, PA, USA
| | - Ganesh Rao
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Arvind U K Rao
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
41
|
Lu KV, Bergers G. Mechanisms of evasive resistance to anti-VEGF therapy in glioblastoma. CNS Oncol 2015; 2:49-65. [PMID: 23750318 DOI: 10.2217/cns.12.36] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Angiogenesis inhibitors targeting the VEGF signaling pathway have been US FDA approved for various cancers including glioblastoma (GBM), one of the most lethal and angiogenic tumors. This has led to the routine use of the anti-VEGF antibody bevacizumab in recurrent GBM, conveying substantial improvements in radiographic response, progression-free survival and quality of life. Despite these encouraging beneficial effects, patients inevitably develop resistance and frequently fail to demonstrate significantly better overall survival. Unlike chemotherapies, to which tumors exhibit resistance due to genetic mutation of drug targets, emerging evidence suggests that tumors bypass antiangiogenic therapy while VEGF signaling remains inhibited through a variety of mechanisms that are just beginning to be recognized. Because of the indirect nature of resistance to VEGF inhibitors there is promise that strategies combining angiogenesis inhibitors with drugs targeting such evasive resistance pathways will lead to more durable antiangiogenic efficacy and improved patient outcomes. Further identifying and understanding of evasive resistance mechanisms and their clinical importance in GBM relapse is therefore a timely and critical issue.
Collapse
|
42
|
Chroscinski D, Sampey D, Maherali N. Registered report: tumour vascularization via endothelial differentiation of glioblastoma stem-like cells. eLife 2015; 4. [PMID: 25714925 PMCID: PMC4383241 DOI: 10.7554/elife.04363] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 01/27/2015] [Indexed: 12/22/2022] Open
Abstract
The Reproducibility Project: Cancer Biology seeks to address growing
concerns about reproducibility in scientific research by conducting replications of
50 papers in the field of cancer biology published between 2010 and 2012. This
Registered report describes the proposed replication plan of key experiments from
‘Tumour vascularization via endothelial differentiation of glioblastoma
stem-like cells’ by Ricci-Vitiani and colleagues, published in
Nature in 2010 (Ricci-Vitiani
et al., 2010). The experiments that will be replicated are those reported
in Figure 4B and Supplementary Figure 10B (Ricci-Vitiani et al., 2010), which demonstrate that glioblastoma stem-like
cells can derive into endothelial cells, and can be selectively ablated to reduce
tumor progression in vivo, and Supplementary Figures S10C and S10D (Ricci-Vitiani et al., 2010), which demonstrate
that fully differentiated glioblastoma cells cannot form functionally relevant
endothelium. The Reproducibility Project: Cancer Biology is a collaboration between
the Center for Open Science and Science Exchange, and the
results of the replications will be published by eLife. DOI:http://dx.doi.org/10.7554/eLife.04363.001
Collapse
Affiliation(s)
| | | | - Nimet Maherali
- Harvard Stem Cell Institute, Cambridge, Massachusetts, United States
| | | | | |
Collapse
|
43
|
Orr BA, Eberhart CG. Molecular pathways: not a simple tube--the many functions of blood vessels. Clin Cancer Res 2014; 21:18-23. [PMID: 25074609 DOI: 10.1158/1078-0432.ccr-13-1641] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Although the ability of blood vessels to carry fluid and cells through neoplastic tissue is clearly important, other functions of vascular elements that drive tumor growth and progression are increasingly being recognized. Vessels can provide physical support and help regulate the stromal microenvironment within tumors, form niches for tumor-associated stem cells, serve as avenues for local tumor spread, and promote relative immune privilege. Understanding the molecular drivers of these phenotypes will be critical if we are to therapeutically target their protumorigenic effects. The potential for neoplastic cells to transdifferentiate into vascular and perivascular elements also needs to be better understood, as it has the potential to complicate such therapies. In this review, we provide a brief overview of these less conventional vascular functions in tumors.
Collapse
Affiliation(s)
- Brent A Orr
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Charles G Eberhart
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland. Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, Maryland. Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
44
|
Abstract
Glioblastoma (GBM) is the most malignant brain tumor where patients' survival is only 14.6 months, despite multimodal therapy with debulking surgery, concurrent chemotherapy and radiotherapy. There is an urgent, unmet need for novel, effective therapeutic strategies for this devastating disease. Although several immunotherapies are under development for the treatment of GBM patients, the use of natural killer (NK) cells is still marginal despite this being a promising approach to treat cancer. In regard of our knowledge on the role of NG2/CSPG4 in promoting GBM aggressiveness we investigated the potential of an innovative immunotherapeutic strategy combining mAb9.2.27 against NG2/CSPG4 and NK cells in preclinical animal models of GBM. Multiple immune escape mechanisms maintain the tumor microenvironment in an anti-inflammatory state to promote tumor growth, however, the distinct roles of resident microglia versus recruited macrophages is not elucidated. We hypothesized that exploiting the cytokine release capabilities of activated (NK) cells to reverse the anti-inflammatory axis combined with mAb9.2.27 targeting the NG2/CSPG4 may favor tumor destruction by editing pro-GBM immune responses. Combination treatment with NK+mAb9.2.27 diminished tumor growth that was associated with reduced tumor proliferation, increased cellular apoptosis and prolonged survival compared to vehicle and monotherapy controls. The therapeutic efficacy was mediated by recruitment of CCR2low macrophages into the tumor microenvironment, increased ED1 and MHC class II expression on microglia that might render them competent for GBM antigen presentation, as well as elevated IFN-γ and TNF-α levels in the cerebrospinal fluid compared to controls. Depletion of systemic macrophages by liposome-encapsulated clodronate decreased the CCR2low macrophages recruited to the brain and abolished the beneficial outcomes. Moreover, mAb9.2.27 reversed tumor-promoting effects of patient-derived tumor-associated macrophage/microglia(TAM) ex vivo.Taken together, these findings indicate thatNK+mAb9.2.27 treatment may be an amenable therapeutic strategy to treat NG2/CSPG4 expressing GBMs. We provide a novel conceptual approach of combination immunotherapy for glioblastoma. The results traverse beyond the elucidation of NG2/CSPG4 as a therapeutic target, but demonstrate a proof of concept that this antibody may hold potential for the treatment of GBM by activation of tumor infiltrated microglia/macrophages.
Collapse
|
45
|
Growth arrest and forced differentiation of human primary glioblastoma multiforme by a novel small molecule. Sci Rep 2014; 4:5546. [PMID: 24989033 PMCID: PMC4080225 DOI: 10.1038/srep05546] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 05/30/2014] [Indexed: 12/18/2022] Open
Abstract
Glioblastoma multiforme is the most common malignant brain tumor in adults, with an average survival of less than one year due to its resistance to therapy. Recent studies reported that GBM initiates from CD133-expressing cancer stem cells (CSC). However, the efficacy of CSC targeting is limited. A newly developed approach in cancer treatment is the forced differentiation of cancer cells. Here, we show that the treatment of the novel small molecule, CG500354, into CD133-expressing human primary GBM cells induces growth arrest by cell cycle regulators, p53, p21, p27 and phase-specific cyclins, and neural differentiation, as confirmed by neural progenitor/precursor markers, nestin, GFAP and Tuj1. When GBM-derived cells caused the tumors in NOD/SCID mice, CG500354 induced GBM-derived cells differentiation into Tuj1 and GFAP expressing cells. We next demonstrated that CG500354 plays a tumor-suppressive role via cAMP/CREB signaling pathway. CG500354 increases not only the extracellular cAMP level but also the protein level of PKA and CREB. Additionally, both mimetic substances, Forskolin and Rolipram, revealed comparable results with CG500354. Our findings indicate that induction of growth arrest and neural differentiation via cAMP/CREB signaling pathway by CG500354 treatment suggests the novel targeting of PDE4D in the development of new drugs for brain tumor therapy.
Collapse
|
46
|
Würth R, Bajetto A, Harrison JK, Barbieri F, Florio T. CXCL12 modulation of CXCR4 and CXCR7 activity in human glioblastoma stem-like cells and regulation of the tumor microenvironment. Front Cell Neurosci 2014; 8:144. [PMID: 24904289 PMCID: PMC4036438 DOI: 10.3389/fncel.2014.00144] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 05/06/2014] [Indexed: 12/16/2022] Open
Abstract
Chemokines are crucial autocrine and paracrine players in tumor development. In particular, CXCL12, through its receptors CXCR4 and CXCR7, affects tumor progression by controlling cancer cell survival, proliferation and migration, and, indirectly, via angiogenesis or recruiting immune cells. Glioblastoma (GBM) is the most prevalent primary malignant brain tumor in adults and despite current multimodal therapies it remains almost incurable. The aggressive and recurrent phenotype of GBM is ascribed to high growth rate, invasiveness to normal brain, marked angiogenesis, ability to escape the immune system and resistance to standard of care therapies. Tumor molecular and cellular heterogeneity severely hinders GBM therapeutic improvement. In particular, a subpopulation of chemo- and radio-therapy resistant tumorigenic cancer stem-like cells (CSCs) is believed to be the main responsible for tumor cell dissemination to the brain. GBM cells display heterogeneous expression levels of CXCR4 and CXCR7 that are overexpressed in CSCs, representing a molecular correlate for the invasive potential of GBM. The microenvironment contribution in GBM development is increasingly emphasized. An interplay exists between CSCs, differentiated GBM cells, and the microenvironment, mainly through secreted chemokines (e.g., CXCL12) causing recruitment of fibroblasts, endothelial, mesenchymal and inflammatory cells to the tumor, via specific receptors such as CXCR4. This review covers recent developments on the role of CXCL12/CXCR4-CXCR7 networks in GBM progression and the potential translational impact of their targeting. The biological and molecular understanding of the heterogeneous GBM cell behavior, phenotype and signaling is still limited. Progress in the identification of chemokine-dependent mechanisms that affect GBM cell survival, trafficking and chemo-attractive functions, opens new perspectives for development of more specific therapeutic approaches that include chemokine-based drugs.
Collapse
Affiliation(s)
- Roberto Würth
- Sezione di Farmacologia, Dipartimento di Medicina Interna, University of Genova Genova, Italy ; Centro di Eccellenza per la Ricerca Biomedica, University of Genova Genova, Italy
| | - Adriana Bajetto
- Sezione di Farmacologia, Dipartimento di Medicina Interna, University of Genova Genova, Italy ; Centro di Eccellenza per la Ricerca Biomedica, University of Genova Genova, Italy
| | - Jeffrey K Harrison
- Department of Pharmacology and Therapeutics, College of Medicine, University of Florida Gainesville, FL, USA
| | - Federica Barbieri
- Sezione di Farmacologia, Dipartimento di Medicina Interna, University of Genova Genova, Italy ; Centro di Eccellenza per la Ricerca Biomedica, University of Genova Genova, Italy
| | - Tullio Florio
- Sezione di Farmacologia, Dipartimento di Medicina Interna, University of Genova Genova, Italy ; Centro di Eccellenza per la Ricerca Biomedica, University of Genova Genova, Italy
| |
Collapse
|
47
|
Stem cell niches in glioblastoma: a neuropathological view. BIOMED RESEARCH INTERNATIONAL 2014; 2014:725921. [PMID: 24834433 PMCID: PMC4009309 DOI: 10.1155/2014/725921] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 03/22/2014] [Accepted: 03/24/2014] [Indexed: 11/27/2022]
Abstract
Glioblastoma (GBM) stem cells (GSCs), responsible for tumor growth, recurrence, and resistance to therapies, are considered the real therapeutic target, if they had no molecular mechanisms of resistance, in comparison with the mass of more differentiated cells which are insensitive to therapies just because of being differentiated and nonproliferating. GSCs occur in tumor niches where both stemness status and angiogenesis are conditioned by the microenvironment. In both perivascular and perinecrotic niches, hypoxia plays a fundamental role. Fifteen glioblastomas have been studied by immunohistochemistry and immunofluorescence for stemness and differentiation antigens. It has been found that circumscribed necroses develop inside hyperproliferating areas that are characterized by high expression of stemness antigens. Necrosis developed inside them because of the imbalance between the proliferation of tumor cells and endothelial cells; it reduces the number of GSCs to a thin ring around the former hyperproliferating area. The perinecrotic GSCs are nothing else that the survivors remnants of those populating hyperproliferating areas. In the tumor, GSCs coincide with malignant areas so that the need to detect where they are located is not so urgent.
Collapse
|
48
|
Xu K, Wang L, Shu HKG. COX-2 overexpression increases malignant potential of human glioma cells through Id1. Oncotarget 2014; 5:1241-52. [PMID: 24659686 PMCID: PMC4012736 DOI: 10.18632/oncotarget.1370] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 10/29/2013] [Indexed: 01/19/2023] Open
Abstract
Increased COX-2 expression directly correlates with glioma grade and is associated with shorter survival in glioblastoma (GBM) patients. COX-2 is also regulated by epidermal growth factor receptor signaling which is important in the pathogenesis of GBMs. However, COX-2 expression has not been previously shown to directly alter malignancy of GBMs. Id1 is a member of the helix-loop-helix (HLH) family of transcriptional repressors that act as dominant-negative inhibitors of basic-HLH factors. This factor has been shown to be regulated by COX-2 in breast carcinoma cells and recent studies suggest that Id1 may also be involved in the genesis/progression of gliomas. We now show that COX-2 increases the aggressiveness of GBM cells. GBM cells with COX-2 overexpression show increased growth of colonies in soft agar. Tumorigenesis in vivo is also increased in both subcutaneous flank and orthotopic intracranial tumor models. COX-2 overexpression induces Id1 expression in two GBM cell lines suggesting a role for Id1 in glioma transformation/tumorigenesis. Furthermore, we find direct evidence of a role for Id1 with significant suppression of in vitro transformation and in vivo tumorigenesis in COX-2-overexpressing GBM cells where Id1 has been knocked down. In fact, Id1 is even more efficient at enhancing transformation/tumorigenesis of GBM cells than COX-2. Finally, GBM cells with COX-2 or Id1 overexpression show greater migration/invasive potential and tumors that arise from these cells also display increased microvessel density, results in line with the increased malignant potential seen in these cells. Thus, COX-2 enhances the malignancy of GBM cells through induction of Id1.
Collapse
Affiliation(s)
- Kaiming Xu
- Department of Radiation Oncology and the Winship Cancer Institute, Emory University, Atlanta, GA
| | - Lanfang Wang
- Department of Radiation Oncology and the Winship Cancer Institute, Emory University, Atlanta, GA
| | - Hui-Kuo G. Shu
- Department of Radiation Oncology and the Winship Cancer Institute, Emory University, Atlanta, GA
| |
Collapse
|
49
|
Glioblastoma multiforme therapy and mechanisms of resistance. Pharmaceuticals (Basel) 2013; 6:1475-506. [PMID: 24287492 PMCID: PMC3873674 DOI: 10.3390/ph6121475] [Citation(s) in RCA: 164] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Revised: 11/04/2013] [Accepted: 11/12/2013] [Indexed: 12/26/2022] Open
Abstract
Glioblastoma multiforme (GBM) is a grade IV brain tumor characterized by a heterogeneous population of cells that are highly infiltrative, angiogenic and resistant to chemotherapy. The current standard of care, comprised of surgical resection followed by radiation and the chemotherapeutic agent temozolomide, only provides patients with a 12–14 month survival period post-diagnosis. Long-term survival for GBM patients remains uncommon as cells with intrinsic or acquired resistance to treatment repopulate the tumor. In this review we will describe the mechanisms of resistance, and how they may be overcome to improve the survival of GBM patients by implementing novel chemotherapy drugs, new drug combinations and new approaches relating to DNA damage, angiogenesis and autophagy.
Collapse
|
50
|
Shin JH, Lee YS, Hong YK, Kang CS. Correlation between the prognostic value and the expression of the stem cell marker CD133 and isocitrate dehydrogenase1 in glioblastomas. J Neurooncol 2013; 115:333-41. [PMID: 24129546 DOI: 10.1007/s11060-013-1234-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Accepted: 08/21/2013] [Indexed: 12/25/2022]
Abstract
Cancer stem cells are thought to be responsible for tumor recurrence and resistance in glioblastomas. An isocitrate dehydrogenase1 (IDH1) mutation, affecting codon132 of the isocitrate dehydrogenase1 gene, has prognostic significance in glioblastomas. We investigated whether stem cell marker expression [CD133, CD34, and vascular endothelial growth factor (VEGF)] and IDH1 mutation correlate with clinical factors and prognosis in glioblastoma. CD133, CD34, and VEGF expression was evaluated by immunohistochemistry in 67 cases of glioblastoma identified between 2005 and 2012. IDH1 mutation was assessed by immunohistochemistry, peptide-nucleic-acid mediated PCR clamping, and direct gene sequencing. Diffuse CD133 expression was detected in 12 (17.9 %) cases and was associated with poor overall survival (OS) (P = 0.010) and progression-free survival (P = 0.017). CD34 and VEGF expression were not associated with prognosis in these samples. IDH1 mutation was detected in ten (14.9 %) cases. Eight were clinically secondary tumors and two were primary tumors (P < 0.001); the mean age of the secondary tumor patients was significantly younger (P = 0.001, 41.20 vs. 59.14). IDH1-positive patients had longer OS than IDH1-negative patients (25.78 vs. 22.95 months), but this difference was not significant. In addition, IDH1 and CD34 expression showed a negative correlation (P = 0.024). Multivariate analysis showed that age, extent of surgery, and diffuse CD133 expression correlated with OS. CD133 may be a survival marker for glioblastoma. Further characterization of CD133, IDH1, and vascular markers in glioblastoma may help identify new therapeutic targets.
Collapse
Affiliation(s)
- Jung Ha Shin
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul, Korea,
| | | | | | | |
Collapse
|