1
|
Chen M, Zhao D. Invisible Bridges: Unveiling the Role and Prospects of Tunneling Nanotubes in Cancer Therapy. Mol Pharm 2024; 21:5413-5429. [PMID: 39373242 PMCID: PMC11539062 DOI: 10.1021/acs.molpharmaceut.4c00563] [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: 05/23/2024] [Revised: 09/20/2024] [Accepted: 09/23/2024] [Indexed: 10/08/2024]
Abstract
Tunneling nanotubes (TNTs) are essential intercellular communication channels that significantly impact cancer pathophysiology, affecting tumor progression and resistance. This review methodically examines the mechanisms of TNTs formation, their structural characteristics, and their functional roles in material and signal transmission between cells. Highlighting their regulatory functions within the tumor microenvironment, TNTs are crucial for modulating cell survival, proliferation, drug resistance, and immune evasion. The review critically evaluates the therapeutic potential of TNTs, focusing on their applications in targeted drug delivery and gene therapy. It also proposes future research directions to thoroughly understand TNTs biogenesis, identify cell-specific molecular targets, and develop advanced technologies for the real-time monitoring of TNTs. By integrating insights from molecular biology, nanotechnology, and immunology, this review highlights the transformative potential of TNTs in advancing cancer treatment strategies.
Collapse
Affiliation(s)
- Meiru Chen
- Department
of Gastroenterology, The Second Hospital of Hebei Medical University,
Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Shijiazhuang, Hebei 050000, China
- Department
of Gastroenterology, Hengshui People’s
Hospital, Hengshui, Hebei 053000, China
| | - Dongqiang Zhao
- Department
of Gastroenterology, The Second Hospital of Hebei Medical University,
Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Shijiazhuang, Hebei 050000, China
| |
Collapse
|
2
|
Wang L, Liang J, Ji S, Wang C, Huang Q. Potential Mechanism and Involvement of p120-Catenin in the Malignant Biology of Glioma. J Korean Neurosurg Soc 2024; 67:609-621. [PMID: 38956806 PMCID: PMC11540527 DOI: 10.3340/jkns.2024.0053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/19/2024] [Accepted: 07/02/2024] [Indexed: 07/04/2024] Open
Abstract
OBJECTIVE This study analyzed the influence of p120-catenin (catenin [cadherin-associated protein], delta 1 [CTNND1]) on the malignant characteristics of glioma and elucidated the potential underlying mechanism. METHODS The p120 expression level was assessed in the brain tissues of 42 glioma patients and 10 patients with epilepsy by using the immunohistochemical method. Meanwhile, quantitative polymerase chain reaction (QT-PCR) technology was employed to assess the expression of p120 in the brain tissues of 71 glioma patients and 13 epilepsy patients. LN229, U251, and U87 glioma cells were used for in vitro analysis and categorized into four treatment groups : siRNA-blank control (BC) group (no RNA sequence was transfected), siRNA-negative control (NC) group (transfected control RNA sequences with no effect), and siRNA-1 and siRNA-2 groups (two p120-specific interfering RNA transfection). p120 expression in these treatment groups was quantified by western blotting assay. The migratory and invasive capabilities of glioma cells were studied by wound healing assay and Transwell invasion assay, respectively, under different treatment conditions. MTT (3-(4,5)-dimethylthiahiazo (-z-y1)-3,5-di-phenytetrazoliumromide) assay and cell cycle and apoptosis assay were used to determine glioma cell proliferation and apoptosis, respectively. Enzymelabeled assay was performed to measure intracellular calcium ion concentration. Immunofluorescence assay was performed for determining microtubule formation and glioma cell distribution. RESULTS Brain tissues of the glioma group exhibited a remarkable increase in the p120 expression level as compared to brain tissues of the nontumor group (p<0.05). Furthermore, a strong positive correlation was noted between the malignancy degree in glioma brain tissues and p120 expression in Western blotting (r=0.906, p<0.0001) and QT-PCR (F=830.6, p<0.01). Compared to the BC and NC groups, the siRNA transfection groups showed a significant suppression in p120 expression in glioma cells (p<0.05), with a marked attenuation in the invasive, migratory, and proliferative capabilities of glioma cells as well as an increase in apoptotic potential (p<0.05). Enzyme-labeled assay showed a remarkable increase in calcium concentration in glioma cells after siRNA treatment. Immunofluorescence assay revealed that the microtubule formation ability of glioma cells reduced after siRNA treatment. CONCLUSION p120 has a pivotal involvement in facilitating glioma cell invasion and proliferation by potentially modulating these processes through its involvement in microtubule formation and regulation of intracellular calcium ion levels.
Collapse
Affiliation(s)
- Leilei Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Department of Neurosurgery, Cangzhou Central Hospital, Cangzhou, China
| | - Jianshen Liang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Suzhen Ji
- Department of Emergency, Cangzhou Central Hospital, Cangzhou, China
| | - Chunlou Wang
- Department of Pathology, Cangzhou Central Hospital, Cangzhou, China
| | - Qiang Huang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
3
|
Lv W, Wang Y. Neural Influences on Tumor Progression Within the Central Nervous System. CNS Neurosci Ther 2024; 30:e70097. [PMID: 39469896 PMCID: PMC11519750 DOI: 10.1111/cns.70097] [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: 07/08/2024] [Revised: 09/21/2024] [Accepted: 10/13/2024] [Indexed: 10/30/2024] Open
Abstract
For decades, researchers have studied how brain tumors, the immune system, and drugs interact. With the advances in cancer neuroscience, which centers on defining and therapeutically targeting nervous system-cancer interactions, both within the local tumor microenvironment (TME) and on a systemic level, the subtle relationship between neurons and tumors in the central nervous system (CNS) has been deeply studied. Neurons, as the executors of brain functional activities, have been shown to significantly influence the emergence and development of brain tumors, including both primary and metastatic tumors. They engage with tumor cells via chemical or electrical synapses, directly regulating tumors or via intricate coupling networks, and also contribute to the TME through paracrine signaling, secreting proteins that exert regulatory effects. For instance, in a study involving a mouse model of glioblastoma, the authors observed a 42% increase in tumor volume when neuronal activity was stimulated, compared to controls (p < 0.01), indicating a direct correlation between neural activity and tumor growth. These thought-provoking results offer promising new strategies for brain tumor therapies, highlighting the potential of neuronal modulation to curb tumor progression. Future strategies may focus on developing drugs to inhibit or neutralize proteins and other bioactive substances secreted by neurons, break synaptic connections and interactions between infiltrating cells and tumor cells, as well as disrupt electrical coupling within glioma cell networks. By harnessing the insights gained from this research, we aspire to usher in a new era of brain tumor therapies that are both more potent and precise.
Collapse
Affiliation(s)
- Wenhao Lv
- Affiliated Hospital of Hangzhou Normal UniversityHangzhou Normal UniversityHangzhouZhejiangChina
- School of PharmacyHangzhou Normal UniversityHangzhouZhejiangChina
| | - Yongjie Wang
- School of PharmacyHangzhou Normal UniversityHangzhouZhejiangChina
| |
Collapse
|
4
|
Sloan AR, Silver DJ, Kint S, Gallo M, Lathia JD. Cancer stem cell hypothesis 2.0 in glioblastoma: Where are we now and where are we going? Neuro Oncol 2024; 26:785-795. [PMID: 38394444 PMCID: PMC11066900 DOI: 10.1093/neuonc/noae011] [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] [Indexed: 02/25/2024] Open
Abstract
Over the past 2 decades, the cancer stem cell (CSC) hypothesis has provided insight into many malignant tumors, including glioblastoma (GBM). Cancer stem cells have been identified in patient-derived tumors and in some mouse models, allowing for a deeper understanding of cellular and molecular mechanisms underlying GBM growth and therapeutic resistance. The CSC hypothesis has been the cornerstone of cellular heterogeneity, providing a conceptual and technical framework to explain this longstanding phenotype in GBM. This hypothesis has evolved to fit recent insights into how cellular plasticity drives tumor growth to suggest that CSCs do not represent a distinct population but rather a cellular state with substantial plasticity that can be achieved by non-CSCs under specific conditions. This has further been reinforced by advances in genomics, including single-cell approaches, that have used the CSC hypothesis to identify multiple putative CSC states with unique properties, including specific developmental and metabolic programs. In this review, we provide a historical perspective on the CSC hypothesis and its recent evolution, with a focus on key functional phenotypes, and provide an update on the definition for its use in future genomic studies.
Collapse
Affiliation(s)
- Anthony R Sloan
- Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Case Comprehensive Cancer Center, Cleveland, Ohio, USA
| | - Daniel J Silver
- Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Case Comprehensive Cancer Center, Cleveland, Ohio, USA
| | - Sam Kint
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Marco Gallo
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Pediatrics, Section of Hematology and Oncology, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Cancer Center, Texas Children’s Hospital, Houston, Texas, USA
| | - Justin D Lathia
- Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Case Comprehensive Cancer Center, Cleveland, Ohio, USA
- Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
5
|
Virtuoso A, D’Amico G, Scalia F, De Luca C, Papa M, Maugeri G, D’Agata V, Caruso Bavisotto C, D’Amico AG. The Interplay between Glioblastoma Cells and Tumor Microenvironment: New Perspectives for Early Diagnosis and Targeted Cancer Therapy. Brain Sci 2024; 14:331. [PMID: 38671983 PMCID: PMC11048111 DOI: 10.3390/brainsci14040331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 03/26/2024] [Accepted: 03/28/2024] [Indexed: 04/28/2024] Open
Abstract
Glioblastoma multiforme (GBM) stands out as the most tremendous brain tumor, constituting 60% of primary brain cancers, accompanied by dismal survival rates. Despite advancements in research, therapeutic options remain limited to chemotherapy and surgery. GBM molecular heterogeneity, the intricate interaction with the tumor microenvironment (TME), and non-selective treatments contribute to the neoplastic relapse. Diagnostic challenges arise from GBM advanced-stage detection, necessitating the exploration of novel biomarkers for early diagnosis. Using data from the literature and a bioinformatic tool, the current manuscript delineates the molecular interplay between human GBM, astrocytes, and myeloid cells, underscoring selected protein pathways belonging to astroglia and myeloid lineage, which can be considered for targeted therapies. Moreover, the pivotal role of extracellular vesicles (EVs) in orchestrating a favorable microenvironment for cancer progression is highlighted, suggesting their utility in identifying biomarkers for GBM early diagnosis.
Collapse
Affiliation(s)
- Assunta Virtuoso
- Laboratory of Neuronal Networks Morphology and System Biology, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.V.); (C.D.L.); (M.P.)
| | - Giuseppa D’Amico
- Department of Biomedicine, Neurosciences and Advanced Diagnostic (BiND), Human Anatomy Section, University of Palermo, 90127 Palermo, Italy; (G.D.); (F.S.)
| | - Federica Scalia
- Department of Biomedicine, Neurosciences and Advanced Diagnostic (BiND), Human Anatomy Section, University of Palermo, 90127 Palermo, Italy; (G.D.); (F.S.)
| | - Ciro De Luca
- Laboratory of Neuronal Networks Morphology and System Biology, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.V.); (C.D.L.); (M.P.)
| | - Michele Papa
- Laboratory of Neuronal Networks Morphology and System Biology, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.V.); (C.D.L.); (M.P.)
| | - Grazia Maugeri
- Department of Biomedical and Biotechnological Sciences, Section of Anatomy, Histology and Movement Sciences, University of Catania, 95100 Catania, Italy; (G.M.); (V.D.)
| | - Velia D’Agata
- Department of Biomedical and Biotechnological Sciences, Section of Anatomy, Histology and Movement Sciences, University of Catania, 95100 Catania, Italy; (G.M.); (V.D.)
| | - Celeste Caruso Bavisotto
- Department of Biomedicine, Neurosciences and Advanced Diagnostic (BiND), Human Anatomy Section, University of Palermo, 90127 Palermo, Italy; (G.D.); (F.S.)
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy
| | - Agata Grazia D’Amico
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy;
| |
Collapse
|
6
|
Radin DP, Shifman S, Outhwaite IR, Sharma A, Bases R, Seeliger MA, Tsirka SE. Lucanthone, a Potential PPT1 Inhibitor, Perturbs Stemness, Reduces Tumor Microtube Formation, and Slows the Growth of Temozolomide-Resistant Gliomas In Vivo. J Pharmacol Exp Ther 2024; 389:51-60. [PMID: 38296645 PMCID: PMC10949164 DOI: 10.1124/jpet.123.002021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 12/28/2023] [Accepted: 01/12/2024] [Indexed: 02/02/2024] Open
Abstract
Glioblastoma (GBM) is the most frequently diagnosed primary central nervous system tumor in adults. Despite the standard of care therapy, which includes surgical resection, temozolomide chemotherapy, radiation and the newly added tumor-treating fields, median survival remains only ∼20 months. Unfortunately, GBM has a ∼100% recurrence rate, but after recurrence there are no Food and Drug Administration-approved therapies to limit tumor growth and enhance patient survival, as these tumors are resistant to temozolomide (TMZ). Recently, our laboratory reported that lucanthone slows GBM by inhibiting autophagic flux through lysosome targeting and decreases the number of Olig2+ glioma stem-like cells (GSC) in vitro and in vivo. We now additionally report that lucanthone efficiently abates stemness in patient-derived GSC and reduces tumor microtube formation in GSC, an emerging hallmark of treatment resistance in GBM. In glioma tumors derived from cells with acquired resistance to TMZ, lucanthone retains the ability to perturb tumor growth, inhibits autophagy by targeting lysosomes, and reduces Olig2 positivity. We also find that lucanthone may act as an inhibitor of palmitoyl protein thioesterase 1. Our results suggest that lucanthone may function as a potential treatment option for GBM tumors that are not amenable to TMZ treatment. SIGNIFICANCE STATEMENT: We report that the antischistosome agent lucanthone impedes tumor growth in a preclinical model of temozolomide-resistant glioblastoma and reduces the numbers of stem-like glioma cells. In addition, it acts as an autophagy inhibitor, and its mechanism of action may be via inhibition of palmitoyl protein thioesterase 1. As there are no defined therapies approved for recurrent, TMZ-resistant tumor, lucanthone could emerge as a treatment for glioblastoma tumors that may not be amenable to TMZ both in the newly diagnosed and recurrent settings.
Collapse
Affiliation(s)
- Daniel P Radin
- Department of Pharmacological Sciences (D.P.R., S.S., I.R.O., A.S., M.A.S., S.E.T.) and Stony Brook Medical Scientist Training Program (D.P.R., S.S., I.R.O.), Renaissance School of Medicine at Stony Brook University, Stony Brook, New York
| | - Sophie Shifman
- Department of Pharmacological Sciences (D.P.R., S.S., I.R.O., A.S., M.A.S., S.E.T.) and Stony Brook Medical Scientist Training Program (D.P.R., S.S., I.R.O.), Renaissance School of Medicine at Stony Brook University, Stony Brook, New York
| | - Ian R Outhwaite
- Department of Pharmacological Sciences (D.P.R., S.S., I.R.O., A.S., M.A.S., S.E.T.) and Stony Brook Medical Scientist Training Program (D.P.R., S.S., I.R.O.), Renaissance School of Medicine at Stony Brook University, Stony Brook, New York
| | - Aryan Sharma
- Department of Pharmacological Sciences (D.P.R., S.S., I.R.O., A.S., M.A.S., S.E.T.) and Stony Brook Medical Scientist Training Program (D.P.R., S.S., I.R.O.), Renaissance School of Medicine at Stony Brook University, Stony Brook, New York
| | - Robert Bases
- Department of Pharmacological Sciences (D.P.R., S.S., I.R.O., A.S., M.A.S., S.E.T.) and Stony Brook Medical Scientist Training Program (D.P.R., S.S., I.R.O.), Renaissance School of Medicine at Stony Brook University, Stony Brook, New York
| | - Markus A Seeliger
- Department of Pharmacological Sciences (D.P.R., S.S., I.R.O., A.S., M.A.S., S.E.T.) and Stony Brook Medical Scientist Training Program (D.P.R., S.S., I.R.O.), Renaissance School of Medicine at Stony Brook University, Stony Brook, New York
| | - Stella E Tsirka
- Department of Pharmacological Sciences (D.P.R., S.S., I.R.O., A.S., M.A.S., S.E.T.) and Stony Brook Medical Scientist Training Program (D.P.R., S.S., I.R.O.), Renaissance School of Medicine at Stony Brook University, Stony Brook, New York
| |
Collapse
|
7
|
De Fazio E, Pittarello M, Gans A, Ghosh B, Slika H, Alimonti P, Tyler B. Intrinsic and Microenvironmental Drivers of Glioblastoma Invasion. Int J Mol Sci 2024; 25:2563. [PMID: 38473812 PMCID: PMC10932253 DOI: 10.3390/ijms25052563] [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: 01/02/2024] [Revised: 02/07/2024] [Accepted: 02/16/2024] [Indexed: 03/14/2024] Open
Abstract
Gliomas are diffusely infiltrating brain tumors whose prognosis is strongly influenced by their extent of invasion into the surrounding brain tissue. While lower-grade gliomas present more circumscribed borders, high-grade gliomas are aggressive tumors with widespread brain infiltration and dissemination. Glioblastoma (GBM) is known for its high invasiveness and association with poor prognosis. Its low survival rate is due to the certainty of its recurrence, caused by microscopic brain infiltration which makes surgical eradication unattainable. New insights into GBM biology at the single-cell level have enabled the identification of mechanisms exploited by glioma cells for brain invasion. In this review, we explore the current understanding of several molecular pathways and mechanisms used by tumor cells to invade normal brain tissue. We address the intrinsic biological drivers of tumor cell invasion, by tackling how tumor cells interact with each other and with the tumor microenvironment (TME). We focus on the recently discovered neuronal niche in the TME, including local as well as distant neurons, contributing to glioma growth and invasion. We then address the mechanisms of invasion promoted by astrocytes and immune cells. Finally, we review the current literature on the therapeutic targeting of the molecular mechanisms of invasion.
Collapse
Affiliation(s)
- Emerson De Fazio
- Department of Medicine, Vita-Salute San Raffaele University School of Medicine, 20132 Milan, Italy; (E.D.F.); (P.A.)
| | - Matilde Pittarello
- Department of Medicine, Humanitas University School of Medicine, 20089 Rozzano, Italy;
| | - Alessandro Gans
- Department of Neurology, University of Milan, 20122 Milan, Italy;
| | - Bikona Ghosh
- School of Medicine and Surgery, Dhaka Medical College, Dhaka 1000, Bangladesh;
| | - Hasan Slika
- Hunterian Neurosurgical Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA;
| | - Paolo Alimonti
- Department of Medicine, Vita-Salute San Raffaele University School of Medicine, 20132 Milan, Italy; (E.D.F.); (P.A.)
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Betty Tyler
- Hunterian Neurosurgical Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA;
| |
Collapse
|
8
|
Hai L, Hoffmann DC, Wagener RJ, Azorin DD, Hausmann D, Xie R, Huppertz MC, Hiblot J, Sievers P, Heuer S, Ito J, Cebulla G, Kourtesakis A, Kaulen LD, Ratliff M, Mandelbaum H, Jung E, Jabali A, Horschitz S, Ernst KJ, Reibold D, Warnken U, Venkataramani V, Will R, Suvà ML, Herold-Mende C, Sahm F, Winkler F, Schlesner M, Wick W, Kessler T. A clinically applicable connectivity signature for glioblastoma includes the tumor network driver CHI3L1. Nat Commun 2024; 15:968. [PMID: 38320988 PMCID: PMC10847113 DOI: 10.1038/s41467-024-45067-8] [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: 01/25/2023] [Accepted: 01/12/2024] [Indexed: 02/08/2024] Open
Abstract
Tumor microtubes (TMs) connect glioma cells to a network with considerable relevance for tumor progression and therapy resistance. However, the determination of TM-interconnectivity in individual tumors is challenging and the impact on patient survival unresolved. Here, we establish a connectivity signature from single-cell RNA-sequenced (scRNA-Seq) xenografted primary glioblastoma (GB) cells using a dye uptake methodology, and validate it with recording of cellular calcium epochs and clinical correlations. Astrocyte-like and mesenchymal-like GB cells have the highest connectivity signature scores in scRNA-sequenced patient-derived xenografts and patient samples. In large GB cohorts, TM-network connectivity correlates with the mesenchymal subtype and dismal patient survival. CHI3L1 gene expression serves as a robust molecular marker of connectivity and functionally influences TM networks. The connectivity signature allows insights into brain tumor biology, provides a proof-of-principle that tumor cell TM-connectivity is relevant for patients' prognosis, and serves as a robust prognostic biomarker.
Collapse
Affiliation(s)
- Ling Hai
- Bioinformatics and Omics Data Analytics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neurology and Neurooncology Program, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Dirk C Hoffmann
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neurology and Neurooncology Program, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Robin J Wagener
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neurology and Neurooncology Program, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Daniel D Azorin
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - David Hausmann
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neurology and Neurooncology Program, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Ruifan Xie
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Magnus-Carsten Huppertz
- Department of Chemical Biology, Max Planck Institute for Medical Research, Heidelberg, Germany
| | - Julien Hiblot
- Department of Chemical Biology, Max Planck Institute for Medical Research, Heidelberg, Germany
| | - Philipp Sievers
- Department of Neuropathology, Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, DKTK, DKFZ, Heidelberg, Germany
| | - Sophie Heuer
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neurology and Neurooncology Program, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Jakob Ito
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Gina Cebulla
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Alexandros Kourtesakis
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neurology and Neurooncology Program, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Leon D Kaulen
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neurology and Neurooncology Program, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Miriam Ratliff
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Neurosurgery Clinic, University Hospital Mannheim, Mannheim, Germany
| | - Henriette Mandelbaum
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Erik Jung
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neurology and Neurooncology Program, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Ammar Jabali
- Medical Faculty Mannheim, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
- Hector Institute for Translational Brain Research, Mannheim, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute of Reconstructive Neurobiology, School of Medicine & University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Sandra Horschitz
- Medical Faculty Mannheim, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
- Hector Institute for Translational Brain Research, Mannheim, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kati J Ernst
- Pediatric Glioma Research Group, DKTK, DKFZ, Heidelberg, Germany
- Hopp Children's Cancer Center at the NCT Heidelberg (KiTZ), Heidelberg, Germany
| | - Denise Reibold
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Uwe Warnken
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Varun Venkataramani
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neurology and Neurooncology Program, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
- Department of Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Rainer Will
- Genomics and Proteomics Core Facility, DKTK, DKFZ, Heidelberg, Germany
| | - Mario L Suvà
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Felix Sahm
- Department of Neuropathology, Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, DKTK, DKFZ, Heidelberg, Germany
| | - Frank Winkler
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neurology and Neurooncology Program, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Matthias Schlesner
- Bioinformatics and Omics Data Analytics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Biomedical Informatics, Data Mining and Data Analytics, Faculty of Applied Computer Science and Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Wolfgang Wick
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neurology and Neurooncology Program, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Tobias Kessler
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Department of Neurology and Neurooncology Program, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany.
| |
Collapse
|
9
|
Yadav N, Purow BW. Understanding current experimental models of glioblastoma-brain microenvironment interactions. J Neurooncol 2024; 166:213-229. [PMID: 38180686 PMCID: PMC11056965 DOI: 10.1007/s11060-023-04536-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 12/07/2023] [Indexed: 01/06/2024]
Abstract
Glioblastoma (GBM) is a common and devastating primary brain tumor, with median survival of 16-18 months after diagnosis in the setting of substantial resistance to standard-of-care and inevitable tumor recurrence. Recent work has implicated the brain microenvironment as being critical for GBM proliferation, invasion, and resistance to treatment. GBM does not operate in isolation, with neurons, astrocytes, and multiple immune populations being implicated in GBM tumor progression and invasiveness. The goal of this review article is to provide an overview of the available in vitro, ex vivo, and in vivo experimental models for assessing GBM-brain interactions, as well as discuss each model's relative strengths and limitations. Current in vitro models discussed will include 2D and 3D co-culture platforms with various cells of the brain microenvironment, as well as spheroids, whole organoids, and models of fluid dynamics, such as interstitial flow. An overview of in vitro and ex vivo organotypic GBM brain slices is also provided. Finally, we conclude with a discussion of the various in vivo rodent models of GBM, including xenografts, syngeneic grafts, and genetically-engineered models of GBM.
Collapse
Affiliation(s)
- Niket Yadav
- Department of Neurology, University of Virginia Comprehensive Cancer Center, University of Virginia Health System, Charlottesville, VA, 22903, USA
- Medical Scientist Training Program, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Benjamin W Purow
- Department of Neurology, University of Virginia Comprehensive Cancer Center, University of Virginia Health System, Charlottesville, VA, 22903, USA.
| |
Collapse
|
10
|
Ratliff M, Karimian-Jazi K, Hoffmann DC, Rauschenbach L, Simon M, Hai L, Mandelbaum H, Schubert MC, Kessler T, Uhlig S, Dominguez Azorin D, Jung E, Osswald M, Solecki G, Maros ME, Venkataramani V, Glas M, Etminan N, Scheffler B, Wick W, Winkler F. Individual glioblastoma cells harbor both proliferative and invasive capabilities during tumor progression. Neuro Oncol 2023; 25:2150-2162. [PMID: 37335907 PMCID: PMC10708941 DOI: 10.1093/neuonc/noad109] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Indexed: 06/21/2023] Open
Abstract
BACKGROUND Glioblastomas are characterized by aggressive and infiltrative growth, and by striking heterogeneity. The aim of this study was to investigate whether tumor cell proliferation and invasion are interrelated, or rather distinct features of different cell populations. METHODS Tumor cell invasion and proliferation were longitudinally determined in real-time using 3D in vivo 2-photon laser scanning microscopy over weeks. Glioblastoma cells expressed fluorescent markers that permitted the identification of their mitotic history or their cycling versus non-cycling cell state. RESULTS Live reporter systems were established that allowed us to dynamically determine the invasive behavior, and previous or actual proliferation of distinct glioblastoma cells, in different tumor regions and disease stages over time. Particularly invasive tumor cells that migrated far away from the main tumor mass, when followed over weeks, had a history of marked proliferation and maintained their proliferative capacity during brain colonization. Infiltrating cells showed fewer connections to the multicellular tumor cell network, a typical feature of gliomas. Once tumor cells colonized a new brain region, their phenotype progressively transitioned into tumor microtube-rich, interconnected, slower-cycling glioblastoma cells. Analysis of resected human glioblastomas confirmed a higher proliferative potential of tumor cells from the invasion zone. CONCLUSIONS The detection of glioblastoma cells that harbor both particularly high proliferative and invasive capabilities during brain tumor progression provides valuable insights into the interrelatedness of proliferation and migration-2 central traits of malignancy in glioma. This contributes to our understanding of how the brain is efficiently colonized in this disease.
Collapse
Affiliation(s)
- Miriam Ratliff
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neurosurgery, University Hospital Mannheim, University of Heidelberg, Mannheim, Germany
| | - Kianush Karimian-Jazi
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dirk C Hoffmann
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Neurology Clinic and Neurooncology Program and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Laurèl Rauschenbach
- DKFZ-Division Translational Neurooncology at the WTZ, DKTK Partner Site, University Hospital Essen, Essen, Germany
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty and University Hospital Bonn, Bonn, Germany
- Department of Neurosurgery and Spine Surgery, University Hospital Essen, Essen, Germany
| | - Matthias Simon
- Department of Neurosurgery, University of Bonn Medical Center, Bonn, Germany
- Department of Neurosurgery, Bethel Clinic, University of Bielefeld Medical Center, OWL, Bielefeld, Germany
| | - Ling Hai
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Henriette Mandelbaum
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Marc C Schubert
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Neurology Clinic and Neurooncology Program and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Tobias Kessler
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Neurology Clinic and Neurooncology Program and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Stefanie Uhlig
- FlowCore Mannheim and Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Daniel Dominguez Azorin
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Neurology Clinic and Neurooncology Program and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Erik Jung
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Neurology Clinic and Neurooncology Program and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Matthias Osswald
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Neurology Clinic and Neurooncology Program and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Gergely Solecki
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Máté E Maros
- Department of Biomedical Informatics at the Center for Preventive Medicine and Digital Health (CPD-BW), University Hospital Mannheim, University of Heidelberg, Mannheim, Germany
| | - Varun Venkataramani
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Neurology Clinic and Neurooncology Program and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Martin Glas
- DKFZ-Division Translational Neurooncology at the WTZ, DKTK Partner Site, University Hospital Essen, Essen, Germany
- Division of Clinical Neurooncology, Department of Neurology, University Hospital Essen, Essen, Germany
| | - Nima Etminan
- Department of Neurosurgery, University Hospital Mannheim, University of Heidelberg, Mannheim, Germany
| | - Björn Scheffler
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- DKFZ-Division Translational Neurooncology at the WTZ, DKTK Partner Site, University Hospital Essen, Essen, Germany
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty and University Hospital Bonn, Bonn, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Wolfgang Wick
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Neurology Clinic and Neurooncology Program and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Frank Winkler
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Neurology Clinic and Neurooncology Program and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
11
|
Heuer S, Winkler F. Glioblastoma revisited: from neuronal-like invasion to pacemaking. Trends Cancer 2023; 9:887-896. [PMID: 37586918 DOI: 10.1016/j.trecan.2023.07.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/13/2023] [Accepted: 07/17/2023] [Indexed: 08/18/2023]
Abstract
In recent years, two developments have helped us to better understand the fundamental biology of glioblastoma: the description of a striking intratumoral heterogeneity including gene expression-based cell states, and the discovery that neuro-cancer interactions and cancer-intrinsic neurodevelopmental mechanisms are fundamental features of glioblastoma. In this opinion article, we aim to integrate both developments. We explain how two key disease features are characterized by different neural mechanisms related to distinct but plastic cancer cell states: first, the single cell-dominated invasive parts and second, the more solid parts which are dominated by communicating cell networks constantly activated by pacemaker-like glioblastoma cells. The resulting integrative roadmap of molecular and functional heterogeneity contributes to the Cancer Neuroscience of glioblastoma and suggests novel therapeutic strategies.
Collapse
Affiliation(s)
- Sophie Heuer
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, INF 400, 69120 Heidelberg, Germany; Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Frank Winkler
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, INF 400, 69120 Heidelberg, Germany; Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| |
Collapse
|
12
|
Lin X, Huang Z, Wang Y. Neuronal Mechanisms Govern Glioblastoma Cell Invasion. Neurosci Bull 2023; 39:1027-1030. [PMID: 36723779 PMCID: PMC10264303 DOI: 10.1007/s12264-023-01028-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 12/18/2022] [Indexed: 02/02/2023] Open
Affiliation(s)
- Xiaowu Lin
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou Normal University, Hangzhou, 311121, China
- Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Hangzhou Normal University, Hangzhou, 311121, China
- Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, China
| | - Zhihui Huang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China.
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou Normal University, Hangzhou, 311121, China.
- Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Hangzhou Normal University, Hangzhou, 311121, China.
- Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, China.
| | - Yongjie Wang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China.
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou Normal University, Hangzhou, 311121, China.
- Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Hangzhou Normal University, Hangzhou, 311121, China.
- Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, China.
| |
Collapse
|
13
|
Kardani K, Sanchez Gil J, Rabkin SD. Oncolytic herpes simplex viruses for the treatment of glioma and targeting glioblastoma stem-like cells. Front Cell Infect Microbiol 2023; 13:1206111. [PMID: 37325516 PMCID: PMC10264819 DOI: 10.3389/fcimb.2023.1206111] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 05/15/2023] [Indexed: 06/17/2023] Open
Abstract
Glioblastoma (GBM) is one of the most lethal cancers, having a poor prognosis and a median survival of only about 15 months with standard treatment (surgery, radiation, and chemotherapy), which has not been significantly extended in decades. GBM demonstrates remarkable cellular heterogeneity, with glioblastoma stem-like cells (GSCs) at the apex. GSCs are a subpopulation of GBM cells that possess the ability to self-renew, differentiate, initiate tumor formation, and manipulate the tumor microenvironment (TME). GSCs are no longer considered a static population of cells with specific markers but are quite flexible phenotypically and in driving tumor heterogeneity and therapeutic resistance. In light of these features, they are a critical target for successful GBM therapy. Oncolytic viruses, in particular oncolytic herpes simplex viruses (oHSVs), have many attributes for therapy and are promising agents to target GSCs. oHSVs are genetically-engineered to selectively replicate in and kill cancer cells, including GSCs, but not normal cells. Moreover, oHSV can induce anti-tumor immune responses and synergize with other therapies, such as chemotherapy, DNA repair inhibitors, and immune checkpoint inhibitors, to potentiate treatment effects and reduce GSC populations that are partly responsible for chemo- and radio-resistance. Herein, we present an overview of GSCs, activity of different oHSVs, clinical trial results, and combination strategies to enhance efficacy, including therapeutic arming of oHSV. Throughout, the therapeutic focus will be on GSCs and studies specifically targeting these cells. Recent clinical trials and approval of oHSV G47Δ in Japan for patients with recurrent glioma demonstrate the efficacy and promise of oHSV therapy.
Collapse
Affiliation(s)
| | | | - Samuel D. Rabkin
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
14
|
Tu SM, Aydin AM, Maraboyina S, Chen Z, Singh S, Gokden N, Langford T. Stem Cell Origin of Cancer: Implications of Oncogenesis Recapitulating Embryogenesis in Cancer Care. Cancers (Basel) 2023; 15:cancers15092516. [PMID: 37173982 PMCID: PMC10177345 DOI: 10.3390/cancers15092516] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 04/18/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
From this perspective, we wonder about the clinical implications of oncology recapturing ontogeny in the contexts of neoantigens, tumor biomarkers, and cancer targets. We ponder about the biological ramifications of finding remnants of mini-organs and residuals of tiny embryos in some tumors. We reminisce about classical experiments showing that the embryonic microenvironment possesses antitumorigenic properties. Ironically, a stem-ness niche-in the wrong place at the wrong time-is also an onco-niche. We marvel at the paradox of TGF-beta both as a tumor suppressor and a tumor promoter. We query about the dualism of EMT as a stem-ness trait engaged in both normal development and abnormal disease states, including various cancers. It is uncanny that during fetal development, proto-oncogenes wax, while tumor-suppressor genes wane. Similarly, during cancer development, proto-oncogenes awaken, while tumor-suppressor genes slumber. Importantly, targeting stem-like pathways has therapeutic implications because stem-ness may be the true driver, if not engine, of the malignant process. Furthermore, anti-stem-like activity elicits anti-cancer effects for a variety of cancers because stem-ness features may be a universal property of cancer. When a fetus survives and thrives despite immune surveillance and all the restraints of nature and the constraints of its niche, it is a perfect baby. Similarly, when a neoplasm survives and thrives in an otherwise healthy and immune-competent host, is it a perfect tumor? Therefore, a pertinent narrative of cancer depends on a proper perspective of cancer. If malignant cells are derived from stem cells, and both cells are intrinsically RB1 negative and TP53 null, do the absence of RB1 and loss of TP53 really matter in this whole narrative and an entirely different perspective of cancer?
Collapse
Affiliation(s)
- Shi-Ming Tu
- Division of Hematology and Oncology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Ahmet Murat Aydin
- Department of Urology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Sanjay Maraboyina
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Zhongning Chen
- Division of Hematology and Oncology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Sunny Singh
- Division of Hematology and Oncology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Neriman Gokden
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Timothy Langford
- Department of Urology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
15
|
Yang Y, Schubert MC, Kuner T, Wick W, Winkler F, Venkataramani V. Brain Tumor Networks in Diffuse Glioma. Neurotherapeutics 2022; 19:1832-1843. [PMID: 36357661 PMCID: PMC9723066 DOI: 10.1007/s13311-022-01320-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/10/2022] [Indexed: 11/12/2022] Open
Abstract
Diffuse gliomas are primary brain tumors associated with a poor prognosis. Cellular and molecular mechanisms driving the invasive growth patterns and therapeutic resistance are incompletely understood. The emerging field of cancer neuroscience offers a novel approach to study these brain tumors in the context of their intricate interactions with the nervous system employing and combining methodological toolsets from neuroscience and oncology. Increasing evidence has shown how neurodevelopmental and neuronal-like mechanisms are hijacked leading to the discovery of multicellular brain tumor networks. Here, we review how gap junction-coupled tumor-tumor-astrocyte networks, as well as synaptic and paracrine neuron-tumor networks drive glioma progression. Molecular mechanisms of these malignant, homo- and heterotypic networks, and their complex interplay are reviewed. Lastly, potential clinical-translational implications and resulting therapeutic strategies are discussed.
Collapse
Affiliation(s)
- Yvonne Yang
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, INF 400, 69120, Heidelberg, Germany
- Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), INF 280, 69120, Heidelberg, Germany
| | - Marc C Schubert
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, INF 400, 69120, Heidelberg, Germany
- Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), INF 280, 69120, Heidelberg, Germany
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, INF 307, 69120, Heidelberg, Germany
| | - Thomas Kuner
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, INF 307, 69120, Heidelberg, Germany
| | - Wolfgang Wick
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, INF 400, 69120, Heidelberg, Germany
- Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), INF 280, 69120, Heidelberg, Germany
| | - Frank Winkler
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, INF 400, 69120, Heidelberg, Germany
- Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), INF 280, 69120, Heidelberg, Germany
| | - Varun Venkataramani
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, INF 400, 69120, Heidelberg, Germany.
- Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), INF 280, 69120, Heidelberg, Germany.
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, INF 307, 69120, Heidelberg, Germany.
| |
Collapse
|
16
|
Wang L, Wang Z, Liang Z, Li G, Duan S, Xi Y. Short- and long-term efficacy of sustained-release chemotherapy in tumor bed interstitium combined with surgical resection for recurrent malignant glioma. Am J Transl Res 2022; 14:5669-5676. [PMID: 36105035 PMCID: PMC9452319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 07/01/2022] [Indexed: 06/15/2023]
Abstract
OBJECTIVE To discuss and analyze the short- and long-term curative effect of sustained-release chemotherapy combined with surgical resection on recurrent malignant glioma. METHODS The clinical data of 137 patients with recurrent glioma admitted to our hospital from March 2016 to July 2018 were retrospectively analyzed. Among them, 67 patients who received local chemotherapy with cisplatin slow-release polymer after surgical resection were included in the observation group, and the other 70 patients who did not receive chemotherapy after surgical resection were regarded as the control group. The short-term therapeutic efficacy, quality of life score before and after surgery, incidence of toxic and side effects, long-term recurrence rate and survival were compared between the two groups. RESULTS The total remission rate of clinical treatment in observation group was remarkably higher than that in control group (P<0.05). There was no statistically significant difference in quality of life score between the two groups before and after surgery (P>0.05). There was no significant difference in the incidence of postoperative side effects between the two groups (P>0.05). While the recurrence rates of the observation group at 12 and 24 months after surgery were significantly lower than those in the control group (P<0.05). The overall postoperative survival of observation group was obviously superior to that of control group (P<0.05). In patients who received sustained-release chemotherapy in tumor bed interstitium combined with surgical resection, older ones and the ones with partial surgical resection or pathological grade IV had worse long-term survival (P<0.05). CONCLUSION The combined treatment of sustained release chemotherapy in tumor bed interstitium and surgical resection for recurrent malignant glioma can effectively improve the clinical efficacy, reduce postoperative recurrence rate and prolong the survival time of the patients.
Collapse
Affiliation(s)
- Longlong Wang
- Department of Neurosurgery, Cangzhou Central Hospital Cangzhou 061000, Hebei, China
| | - Zhifeng Wang
- Department of Neurosurgery, Cangzhou Central Hospital Cangzhou 061000, Hebei, China
| | - Zan Liang
- Department of Neurosurgery, Cangzhou Central Hospital Cangzhou 061000, Hebei, China
| | - Guojing Li
- Department of Neurosurgery, Cangzhou Central Hospital Cangzhou 061000, Hebei, China
| | - Shibo Duan
- Department of Neurosurgery, Cangzhou Central Hospital Cangzhou 061000, Hebei, China
| | - Yanguo Xi
- Department of Neurosurgery, Cangzhou Central Hospital Cangzhou 061000, Hebei, China
| |
Collapse
|
17
|
Venkataramani V, Yang Y, Schubert MC, Reyhan E, Tetzlaff SK, Wißmann N, Botz M, Soyka SJ, Beretta CA, Pramatarov RL, Fankhauser L, Garofano L, Freudenberg A, Wagner J, Tanev DI, Ratliff M, Xie R, Kessler T, Hoffmann DC, Hai L, Dörflinger Y, Hoppe S, Yabo YA, Golebiewska A, Niclou SP, Sahm F, Lasorella A, Slowik M, Döring L, Iavarone A, Wick W, Kuner T, Winkler F. Glioblastoma hijacks neuronal mechanisms for brain invasion. Cell 2022; 185:2899-2917.e31. [PMID: 35914528 DOI: 10.1016/j.cell.2022.06.054] [Citation(s) in RCA: 217] [Impact Index Per Article: 72.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 04/10/2022] [Accepted: 06/28/2022] [Indexed: 12/29/2022]
Abstract
Glioblastomas are incurable tumors infiltrating the brain. A subpopulation of glioblastoma cells forms a functional and therapy-resistant tumor cell network interconnected by tumor microtubes (TMs). Other subpopulations appear unconnected, and their biological role remains unclear. Here, we demonstrate that whole-brain colonization is fueled by glioblastoma cells that lack connections with other tumor cells and astrocytes yet receive synaptic input from neurons. This subpopulation corresponds to neuronal and neural-progenitor-like tumor cell states, as defined by single-cell transcriptomics, both in mouse models and in the human disease. Tumor cell invasion resembled neuronal migration mechanisms and adopted a Lévy-like movement pattern of probing the environment. Neuronal activity induced complex calcium signals in glioblastoma cells followed by the de novo formation of TMs and increased invasion speed. Collectively, superimposing molecular and functional single-cell data revealed that neuronal mechanisms govern glioblastoma cell invasion on multiple levels. This explains how glioblastoma's dissemination and cellular heterogeneity are closely interlinked.
Collapse
Affiliation(s)
- Varun Venkataramani
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, 69120 Heidelberg, Germany; Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, 69120 Heidelberg, Germany; Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| | - Yvonne Yang
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Marc Cicero Schubert
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, 69120 Heidelberg, Germany
| | - Ekin Reyhan
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Svenja Kristin Tetzlaff
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, 69120 Heidelberg, Germany
| | - Niklas Wißmann
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, 69120 Heidelberg, Germany
| | - Michael Botz
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, 69120 Heidelberg, Germany
| | - Stella Judith Soyka
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, 69120 Heidelberg, Germany
| | - Carlo Antonio Beretta
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, 69120 Heidelberg, Germany
| | - Rangel Lyubomirov Pramatarov
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, 69120 Heidelberg, Germany
| | - Laura Fankhauser
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Luciano Garofano
- Institute for Cancer Genetics, Columbia University Medical Center, New York, NY 10027, USA
| | | | - Julia Wagner
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, 69120 Heidelberg, Germany
| | - Dimitar Ivanov Tanev
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, 69120 Heidelberg, Germany; Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, 69120 Heidelberg, Germany
| | - Miriam Ratliff
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Neurosurgery Clinic, University Hospital Mannheim, 68167 Mannheim, Germany
| | - Ruifan Xie
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, 69120 Heidelberg, Germany; Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Tobias Kessler
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, 69120 Heidelberg, Germany; Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Dirk C Hoffmann
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, 69120 Heidelberg, Germany; Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Ling Hai
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, 69120 Heidelberg, Germany; Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Yvette Dörflinger
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, 69120 Heidelberg, Germany
| | - Simone Hoppe
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, 69120 Heidelberg, Germany
| | - Yahaya A Yabo
- NORLUX Neuro-Oncology Laboratory, Department of Cancer Research, Luxembourg Institute of Health, 1526 Luxembourg, Luxembourg
| | - Anna Golebiewska
- NORLUX Neuro-Oncology Laboratory, Department of Cancer Research, Luxembourg Institute of Health, 1526 Luxembourg, Luxembourg
| | - Simone P Niclou
- NORLUX Neuro-Oncology Laboratory, Department of Cancer Research, Luxembourg Institute of Health, 1526 Luxembourg, Luxembourg
| | - Felix Sahm
- Department of Neuropathology, Institute of Pathology, University Hospital Heidelberg, 69120 Heidelberg, Germany; Clinical Cooperation Unit Neuropathology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Anna Lasorella
- Institute for Cancer Genetics, Columbia University Medical Center, New York, NY 10027, USA
| | - Martin Slowik
- Institute of Mathematics, University of Mannheim, 68131 Mannheim, Germany
| | - Leif Döring
- Institute of Mathematics, University of Mannheim, 68131 Mannheim, Germany
| | - Antonio Iavarone
- Institute for Cancer Genetics, Columbia University Medical Center, New York, NY 10027, USA
| | - Wolfgang Wick
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, 69120 Heidelberg, Germany; Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Thomas Kuner
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, 69120 Heidelberg, Germany.
| | - Frank Winkler
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, 69120 Heidelberg, Germany; Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| |
Collapse
|
18
|
Venkataramani V, Schneider M, Giordano FA, Kuner T, Wick W, Herrlinger U, Winkler F. Disconnecting multicellular networks in brain tumours. Nat Rev Cancer 2022; 22:481-491. [PMID: 35488036 DOI: 10.1038/s41568-022-00475-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/24/2022] [Indexed: 12/13/2022]
Abstract
Cancer cells can organize and communicate in functional networks. Similarly to other networks in biology and sociology, these can be highly relevant for growth and resilience. In this Perspective, we demonstrate by the example of glioblastomas and other incurable brain tumours how versatile multicellular tumour networks are formed by two classes of long intercellular membrane protrusions: tumour microtubes and tunnelling nanotubes. The resulting networks drive tumour growth and resistance to standard therapies. This raises the question of how to disconnect brain tumour networks to halt tumour growth and whether this can make established therapies more effective. Emerging principles of tumour networks, their potential relevance for tumour types outside the brain and translational implications, including clinical trials that are already based on these discoveries, are discussed.
Collapse
Affiliation(s)
- Varun Venkataramani
- Neurology Clinic, University Hospital Heidelberg, Heidelberg, Germany.
- National Center for Tumour Diseases, University Hospital Heidelberg, Heidelberg, Germany.
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany.
| | | | - Frank Anton Giordano
- Department of Radiation Oncology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Thomas Kuner
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Wolfgang Wick
- Neurology Clinic, University Hospital Heidelberg, Heidelberg, Germany
- National Center for Tumour Diseases, University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ulrich Herrlinger
- Division of Clinical Neurooncology, Department of Neurology, University Hospital Bonn, University of Bonn, Bonn, Germany.
| | - Frank Winkler
- Neurology Clinic, University Hospital Heidelberg, Heidelberg, Germany.
- National Center for Tumour Diseases, University Hospital Heidelberg, Heidelberg, Germany.
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
19
|
Wang X, Liang J, Sun H. The Network of Tumor Microtubes: An Improperly Reactivated Neural Cell Network With Stemness Feature for Resistance and Recurrence in Gliomas. Front Oncol 2022; 12:921975. [PMID: 35847909 PMCID: PMC9277150 DOI: 10.3389/fonc.2022.921975] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 05/18/2022] [Indexed: 11/13/2022] Open
Abstract
Gliomas are known as an incurable brain tumor for the poor prognosis and robust recurrence. In recent years, a cellular subpopulation with tumor microtubes (TMs) was identified in brain tumors, which may provide a new angle to explain the invasion, resistance, recurrence, and heterogeneity of gliomas. Recently, it was demonstrated that the cell subpopulation also expresses neural stem cell markers and shares a lot of features with both immature neurons and cancer stem cells and may be seen as an improperly reactivated neural cell network with a stemness feature at later time points of life. TMs may also provide a new angle to understand the resistance and recurrence mechanisms of glioma stem cells. In this review, we innovatively focus on the common features between TMs and sprouting axons in morphology, formation, and function. Additionally, we summarized the recent progress in the resistance and recurrence mechanisms of gliomas with TMs and explained the incurability and heterogeneity in gliomas with TMs. Moreover, we discussed the recently discovered overlap between cancer stem cells and TM-positive glioma cells, which may contribute to the understanding of resistant glioma cell subpopulation and the exploration of the new potential therapeutic target for gliomas.
Collapse
Affiliation(s)
- Xinyue Wang
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jianhao Liang
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Haitao Sun
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou, China
| |
Collapse
|
20
|
Fu Z, Yuan Y. The role of tumor neogenesis pipelines in tumor progression and their therapeutic potential. Cancer Med 2022; 12:1558-1571. [PMID: 35832030 PMCID: PMC9883577 DOI: 10.1002/cam4.4979] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 05/27/2022] [Accepted: 06/11/2022] [Indexed: 02/02/2023] Open
Abstract
Pipeline formation between tumor cells and the tumor microenvironment (TME) is a key event leading to tumor progression. These pipelines include blood vessels, lymphatics, and membranous vessels (the former two can be collectively referred to as vasculature). Pipeline regeneration is a feature of all solid tumors; it delivers nutrients to tumors and promotes tumor invasion and metastasis such that cancer cells grow rapidly, escape unfavorable TME, spread to secondary sites, generate tumor drug resistance, and promote postoperative tumor recurrence. Novel tumor therapy strategies must exploit the molecular mechanisms underpinning these pipelines to facilitate more targeted drug therapies. In this review, pipeline generation, influencing factors, pipeline functions during tumor progression, and pipeline potential as drug targets are systematically summarized.
Collapse
Affiliation(s)
- Zhanqi Fu
- Tumor Etiology and Screening Department of Cancer Institute and General SurgeryThe First Hospital of China Medical UniversityShenyangChina,Key Laboratory of Cancer Etiology and Prevention in Liaoning Education DepartmentThe First Hospital of China Medical UniversityShenyangChina,Key Laboratory of GI Cancer Etiology and Prevention in Liaoning ProvinceThe First Hospital of China Medical UniversityShenyangChina
| | - Yuan Yuan
- Tumor Etiology and Screening Department of Cancer Institute and General SurgeryThe First Hospital of China Medical UniversityShenyangChina,Key Laboratory of Cancer Etiology and Prevention in Liaoning Education DepartmentThe First Hospital of China Medical UniversityShenyangChina,Key Laboratory of GI Cancer Etiology and Prevention in Liaoning ProvinceThe First Hospital of China Medical UniversityShenyangChina
| |
Collapse
|
21
|
van Ineveld RL, van Vliet EJ, Wehrens EJ, Alieva M, Rios AC. 3D imaging for driving cancer discovery. EMBO J 2022; 41:e109675. [PMID: 35403737 PMCID: PMC9108604 DOI: 10.15252/embj.2021109675] [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: 09/09/2021] [Revised: 03/09/2022] [Accepted: 03/09/2022] [Indexed: 11/09/2022] Open
Abstract
Our understanding of the cellular composition and architecture of cancer has primarily advanced using 2D models and thin slice samples. This has granted spatial information on fundamental cancer biology and treatment response. However, tissues contain a variety of interconnected cells with different functional states and shapes, and this complex organization is impossible to capture in a single plane. Furthermore, tumours have been shown to be highly heterogenous, requiring large-scale spatial analysis to reliably profile their cellular and structural composition. Volumetric imaging permits the visualization of intact biological samples, thereby revealing the spatio-phenotypic and dynamic traits of cancer. This review focuses on new insights into cancer biology uniquely brought to light by 3D imaging and concomitant progress in cancer modelling and quantitative analysis. 3D imaging has the potential to generate broad knowledge advance from major mechanisms of tumour progression to new strategies for cancer treatment and patient diagnosis. We discuss the expected future contributions of the newest imaging trends towards these goals and the challenges faced for reaching their full application in cancer research.
Collapse
Affiliation(s)
- Ravian L van Ineveld
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
- Oncode InstituteUtrechtThe Netherlands
| | - Esmée J van Vliet
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
- Oncode InstituteUtrechtThe Netherlands
| | - Ellen J Wehrens
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
- Oncode InstituteUtrechtThe Netherlands
| | - Maria Alieva
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
- Oncode InstituteUtrechtThe Netherlands
| | - Anne C Rios
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
- Oncode InstituteUtrechtThe Netherlands
| |
Collapse
|
22
|
Intercellular Communication in the Brain through Tunneling Nanotubes. Cancers (Basel) 2022; 14:cancers14051207. [PMID: 35267518 PMCID: PMC8909287 DOI: 10.3390/cancers14051207] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 02/21/2022] [Accepted: 02/23/2022] [Indexed: 12/19/2022] Open
Abstract
Simple Summary Tunneling nanotubes (TNTs) are a means of cell communication which have been recently discovered. They allow the intercellular trafficking of many types of cellular compounds ranging from ions, such as Ca2+, to whole organelles such as mitochondria. TNTs are found in many tissues, both in physiological and pathological conditions. They are also found in the brain where they contribute to brain development and function and also to degenerative diseases and glioma. Abstract Intercellular communication is essential for tissue homeostasis and function. Understanding how cells interact with each other is paramount, as crosstalk between cells is often dysregulated in diseases and can contribute to their progression. Cells communicate with each other through several modalities, including paracrine secretion and specialized structures ensuring physical contact between them. Among these intercellular specialized structures, tunneling nanotubes (TNTs) are now recognized as a means of cell-to-cell communication through the exchange of cellular cargo, controlled by a variety of biological triggers, as described here. Intercellular communication is fundamental to brain function. It allows the dialogue between the many cells, including neurons, astrocytes, oligodendrocytes, glial cells, microglia, necessary for the proper development and function of the brain. We highlight here the role of TNTs in connecting these cells, for the physiological functioning of the brain and in pathologies such as stroke, neurodegenerative diseases, and gliomas. Understanding these processes could pave the way for future therapies.
Collapse
|
23
|
Yabo YA, Niclou SP, Golebiewska A. Cancer cell heterogeneity and plasticity: A paradigm shift in glioblastoma. Neuro Oncol 2021; 24:669-682. [PMID: 34932099 PMCID: PMC9071273 DOI: 10.1093/neuonc/noab269] [Citation(s) in RCA: 115] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Phenotypic plasticity has emerged as a major contributor to intra-tumoral heterogeneity and treatment resistance in cancer. Increasing evidence shows that glioblastoma (GBM) cells display prominent intrinsic plasticity and reversibly adapt to dynamic microenvironmental conditions. Limited genetic evolution at recurrence further suggests that resistance mechanisms also largely operate at the phenotypic level. Here we review recent literature underpinning the role of GBM plasticity in creating gradients of heterogeneous cells including those that carry cancer stem cell (CSC) properties. A historical perspective from the hierarchical to the nonhierarchical concept of CSCs towards the recent appreciation of GBM plasticity is provided. Cellular states interact dynamically with each other and with the surrounding brain to shape a flexible tumor ecosystem, which enables swift adaptation to external pressure including treatment. We present the key components regulating intra-tumoral phenotypic heterogeneity and the equilibrium of phenotypic states, including genetic, epigenetic, and microenvironmental factors. We further discuss plasticity in the context of intrinsic tumor resistance, where a variable balance between preexisting resistant cells and adaptive persisters leads to reversible adaptation upon treatment. Innovative efforts targeting regulators of plasticity and mechanisms of state transitions towards treatment-resistant states are needed to restrict the adaptive capacities of GBM.
Collapse
Affiliation(s)
- Yahaya A Yabo
- NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg.,Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Simone P Niclou
- NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg.,Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Anna Golebiewska
- Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| |
Collapse
|
24
|
Valdebenito S, Malik S, Luu R, Loudig O, Mitchell M, Okafo G, Bhat K, Prideaux B, Eugenin EA. Tunneling nanotubes, TNT, communicate glioblastoma with surrounding non-tumor astrocytes to adapt them to hypoxic and metabolic tumor conditions. Sci Rep 2021; 11:14556. [PMID: 34267246 PMCID: PMC8282675 DOI: 10.1038/s41598-021-93775-8] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/30/2021] [Indexed: 02/07/2023] Open
Abstract
Cell-to-cell communication is essential for the development and proper function of multicellular systems. We and others demonstrated that tunneling nanotubes (TNT) proliferate in several pathological conditions such as HIV, cancer, and neurodegenerative diseases. However, the nature, function, and contribution of TNT to cancer pathogenesis are poorly understood. Our analyses demonstrate that TNT structures are induced between glioblastoma (GBM) cells and surrounding non-tumor astrocytes to transfer tumor-derived mitochondria. The mitochondrial transfer mediated by TNT resulted in the adaptation of non-tumor astrocytes to tumor-like metabolism and hypoxia conditions. In conclusion, TNT are an efficient cell-to-cell communication system used by cancer cells to adapt the microenvironment to the invasive nature of the tumor.
Collapse
Affiliation(s)
- Silvana Valdebenito
- Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch (UTMB), Research Building 17, Fifth Floor, 105 11th Street, Galveston, TX, 77555, USA
| | - Shaily Malik
- Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch (UTMB), Research Building 17, Fifth Floor, 105 11th Street, Galveston, TX, 77555, USA
| | - Ross Luu
- Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch (UTMB), Research Building 17, Fifth Floor, 105 11th Street, Galveston, TX, 77555, USA
| | - Olivier Loudig
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, USA
| | - Megan Mitchell
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, USA
| | | | - Krishna Bhat
- Department of Translational Molecular Pathology, Division of Pathology and Laboratory Medicine, M.D. Anderson, Houston, TX, USA
| | - Brendan Prideaux
- Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch (UTMB), Research Building 17, Fifth Floor, 105 11th Street, Galveston, TX, 77555, USA
| | - Eliseo A Eugenin
- Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch (UTMB), Research Building 17, Fifth Floor, 105 11th Street, Galveston, TX, 77555, USA.
| |
Collapse
|
25
|
Two routes of direct intercellular communication in brain cancer. Biochem J 2021; 478:1283-1286. [PMID: 33755120 DOI: 10.1042/bcj20200990] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/02/2021] [Accepted: 03/04/2021] [Indexed: 12/11/2022]
Abstract
Glioblastoma is a particularly challenging disease characterized by the connection of tumor cells to functional multicellular networks that effectively resist therapies. In this issue of Biochemical Journal, Pinto et al. report the discovery of two distinct classes of intercellular membrane tube connections, tunneling nanotubes and tumor microtubes, in the same state-of-the-art culture model of patient-derived glioblastoma material. These findings contribute to our understanding of the heterogeneity of intercellular membrane tubes in health and disease, and pave the way for future functional studies on their various roles for disease progression and tumor resistance.
Collapse
|
26
|
Tumor cell plasticity, heterogeneity, and resistance in crucial microenvironmental niches in glioma. Nat Commun 2021; 12:1014. [PMID: 33579922 PMCID: PMC7881116 DOI: 10.1038/s41467-021-21117-3] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 01/08/2021] [Indexed: 12/15/2022] Open
Abstract
Both the perivascular niche (PVN) and the integration into multicellular networks by tumor microtubes (TMs) have been associated with progression and resistance to therapies in glioblastoma, but their specific contribution remained unknown. By long-term tracking of tumor cell fate and dynamics in the live mouse brain, differential therapeutic responses in both niches are determined. Both the PVN, a preferential location of long-term quiescent glioma cells, and network integration facilitate resistance against cytotoxic effects of radiotherapy and chemotherapy—independently of each other, but with additive effects. Perivascular glioblastoma cells are particularly able to actively repair damage to tumor regions. Population of the PVN and resistance in it depend on proficient NOTCH1 expression. In turn, NOTCH1 downregulation induces resistant multicellular networks by TM extension. Our findings identify NOTCH1 as a central switch between the PVN and network niche in glioma, and demonstrate robust cross-compensation when only one niche is targeted. Whether the perivascular niche (PVN) and the integration into multicellular networks by tumor microtubes (TMs) have a different role in glioblastoma progression and resistance to therapies is currently unclear. Here, the authors, by long-term tracking of individual glioma, demonstrate that both niches can partially compensate for each other and that glioma cells localized in both niches are resistant to radio- and chemotherapy.
Collapse
|