1
|
Santos L, Moreira JN, Abrunhosa A, Gomes C. Brain metastasis: An insight into novel molecular targets for theranostic approaches. Crit Rev Oncol Hematol 2024; 198:104377. [PMID: 38710296 DOI: 10.1016/j.critrevonc.2024.104377] [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: 12/05/2023] [Revised: 04/11/2024] [Accepted: 04/25/2024] [Indexed: 05/08/2024] Open
Abstract
Brain metastases (BrM) are common malignant lesions in the central nervous system, and pose a significant threat in advanced-stage malignancies due to delayed diagnosis and limited therapeutic options. Their distinct genomic profiles underscore the need for molecular profiling to tailor effective treatments. Recent advances in cancer biology have uncovered molecular drivers underlying tumor initiation, progression, and metastasis. This, coupled with the advances in molecular imaging technology and radiotracer synthesis, has paved the way for the development of innovative radiopharmaceuticals with enhanced specificity and affinity for BrM specific targets. Despite the challenges posed by the blood-brain barrier to effective drug delivery, several radiolabeled compounds have shown promise in detecting and targeting BrM. This manuscript provides an overview of the recent advances in molecular biomarkers used in nuclear imaging and targeted radionuclide therapy in both clinical and preclinical settings. Additionally, it explores potential theranostic applications addressing the unique challenges posed by BrM.
Collapse
Affiliation(s)
- Liliana Santos
- Institute for Nuclear Sciences Applied to Health (ICNAS) and Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), University of Coimbra, Coimbra 3000-548, Portugal; Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra 3000-548, Portugal
| | - João Nuno Moreira
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra 3004-504, Portugal; Center for Innovative Biomedicine and Biotechnology Consortium (CIBB), University of Coimbra, Coimbra 3000-548, Portugal
| | - Antero Abrunhosa
- Institute for Nuclear Sciences Applied to Health (ICNAS) and Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), University of Coimbra, Coimbra 3000-548, Portugal
| | - Célia Gomes
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra 3000-548, Portugal; Center for Innovative Biomedicine and Biotechnology Consortium (CIBB), University of Coimbra, Coimbra 3000-548, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra 3000-075, Portugal.
| |
Collapse
|
2
|
Wang H, Yang J, Li X, Zhao H. Current state of immune checkpoints therapy for glioblastoma. Heliyon 2024; 10:e24729. [PMID: 38298707 PMCID: PMC10828821 DOI: 10.1016/j.heliyon.2024.e24729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 01/12/2024] [Accepted: 01/12/2024] [Indexed: 02/02/2024] Open
Abstract
Glioblastoma (GBM), one of the most aggressive forms of brain cancer, has limited treatment options. Recent years have witnessed the remarkable success of checkpoint inhibitor immunotherapy across various cancer types. Against this backdrop, several clinical trials investigating checkpoint inhibitors for GBM are underway in multiple countries. Furthermore, the integration of immunotherapy with traditional treatment approaches is now emerging as a highly promising strategy. This review summarizes the latest advancements in checkpoint inhibitor immunotherapy for GBM treatment. We provide a concise yet comprehensive overview of current GBM immunotherapy options. Additionally, this review underscores combination strategies and potential biomarkers for predicting response and resistance in GBM immunotherapies.
Collapse
Affiliation(s)
- He Wang
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, Shandong, 266005, China
| | - Jing Yang
- Department of Emergency Surgery, the Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, Shandong, 266005, China
| | - Xiangjun Li
- School of medicine, Department of Breast surgery, the Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, Shandong, 266000, China
| | - Hai Zhao
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, Shandong, 266005, China
| |
Collapse
|
3
|
Bartusik-Aebisher D, Serafin I, Dynarowicz K, Aebisher D. Photodynamic therapy and associated targeting methods for treatment of brain cancer. Front Pharmacol 2023; 14:1250699. [PMID: 37841921 PMCID: PMC10568033 DOI: 10.3389/fphar.2023.1250699] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 09/11/2023] [Indexed: 10/17/2023] Open
Abstract
Brain tumors, including glioblastoma multiforme, are currently a cause of suffering and death of tens of thousands of people worldwide. Despite advances in clinical treatment, the average patient survival time from the moment of diagnosis of glioblastoma multiforme and application of standard treatment methods such as surgical resection, radio- and chemotherapy, is less than 4 years. The continuing development of new therapeutic methods for targeting and treating brain tumors may extend life and provide greater comfort to patients. One such developing therapeutic method is photodynamic therapy. Photodynamic therapy is a progressive method of therapy used in dermatology, dentistry, ophthalmology, and has found use as an antimicrobial agent. It has also found wide application in photodiagnosis. Photodynamic therapy requires the presence of three necessary components: a clinically approved photosensitizer, oxygen and light. This paper is a review of selected literature from Pubmed and Scopus scientific databases in the field of photodynamic therapy in brain tumors with an emphasis on glioblastoma treatment.
Collapse
Affiliation(s)
- Dorota Bartusik-Aebisher
- Department of Biochemistry and General Chemistry, Medical College of the University of Rzeszów, Rzeszów, Poland
| | - Iga Serafin
- Students English Division Science Club, Medical College of the University of Rzeszów, Rzeszów, Poland
| | - Klaudia Dynarowicz
- Center for Innovative Research in Medical and Natural Sciences, Medical College of the University of Rzeszów, Rzeszów, Poland
| | - David Aebisher
- Department of Photomedicine and Physical Chemistry, Medical College of the University of Rzeszów, Rzeszów, Poland
| |
Collapse
|
4
|
Xiao X, Li X, Wang Y, Pan C, Zhang P, Gu G, Li T, Jiang Z, Zhang Y, Zhang L. Classification of Brainstem Gliomas Based on Tumor Microenvironment Status. Cancers (Basel) 2023; 15:4224. [PMID: 37686499 PMCID: PMC10487167 DOI: 10.3390/cancers15174224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/15/2023] [Accepted: 08/21/2023] [Indexed: 09/10/2023] Open
Abstract
The inter-tumor heterogeneity of the tumor microenvironment (TME) and how it correlates with clinical profiles and biological characteristics in brainstem gliomas (BSGs) remain unknown, dampening the development of novel therapeutics against BSGs. The TME status was determined with a list of pan-cancer conserved gene expression signatures using a single-sample gene set enrichment analysis (ssGSEA) and was subsequently clustered via consensus clustering. BSGs exhibited a high inter-tumor TME heterogeneity and were classified into four clusters: "immune-enriched, fibrotic", "immune-enriched, non-fibrotic", "fibrotic", and "depleted". The "fibrotic" cluster had a higher proportion of diffuse intrinsic pontine gliomas (p = 0.041), and "PA-like" tumors were more likely to be "immune-enriched, fibrotic" (p = 0.044). The four TME clusters exhibited distinct overall survival (p < 0.001) and independently impacted BSG outcomes. A four-gene panel as well as a radiomics approach were constructed to identify the TME clusters and achieved high accuracy for determining the classification. Together, BSGs exhibited high inter-tumor heterogeneity in the TME and were classified into four clusters with distinct clinical outcomes and tumor biological properties. The TME classification was accurately identified using a four-gene panel that can potentially be examined with the immunohistochemical method and a non-invasive radiomics method, facilitating its clinical application.
Collapse
Affiliation(s)
- Xiong Xiao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (X.X.); (X.L.); (Y.W.); (C.P.); (P.Z.); (G.G.); (T.L.); (Z.J.)
| | - Xiaoou Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (X.X.); (X.L.); (Y.W.); (C.P.); (P.Z.); (G.G.); (T.L.); (Z.J.)
| | - Yi Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (X.X.); (X.L.); (Y.W.); (C.P.); (P.Z.); (G.G.); (T.L.); (Z.J.)
| | - Changcun Pan
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (X.X.); (X.L.); (Y.W.); (C.P.); (P.Z.); (G.G.); (T.L.); (Z.J.)
| | - Peng Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (X.X.); (X.L.); (Y.W.); (C.P.); (P.Z.); (G.G.); (T.L.); (Z.J.)
| | - Guocan Gu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (X.X.); (X.L.); (Y.W.); (C.P.); (P.Z.); (G.G.); (T.L.); (Z.J.)
| | - Tian Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (X.X.); (X.L.); (Y.W.); (C.P.); (P.Z.); (G.G.); (T.L.); (Z.J.)
| | - Zhuang Jiang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (X.X.); (X.L.); (Y.W.); (C.P.); (P.Z.); (G.G.); (T.L.); (Z.J.)
| | - Yang Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (X.X.); (X.L.); (Y.W.); (C.P.); (P.Z.); (G.G.); (T.L.); (Z.J.)
| | - Liwei Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (X.X.); (X.L.); (Y.W.); (C.P.); (P.Z.); (G.G.); (T.L.); (Z.J.)
- China National Clinical Research Center for Neurological Diseases, Beijing 100070, China
- Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| |
Collapse
|
5
|
Chen Y, Zhao C, Li S, Wang J, Zhang H. Immune Microenvironment and Immunotherapies for Diffuse Intrinsic Pontine Glioma. Cancers (Basel) 2023; 15:cancers15030602. [PMID: 36765560 PMCID: PMC9913210 DOI: 10.3390/cancers15030602] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 01/12/2023] [Indexed: 01/20/2023] Open
Abstract
Diffuse intrinsic pontine glioma (DIPG) is a primary glial glioma that occurs in all age groups but predominates in children and is the main cause of solid tumor-related childhood mortality. Due to its rapid progression, the inability to operate and insensitivity to most chemotherapies, there is a lack of effective treatment methods in clinical practice for DIPG patients. The prognosis of DIPG patients is extremely poor, with a median survival time of no more than 12 months. In recent years, there have been continuous breakthroughs for immunotherapies in various hematological tumors and malignant solid tumors with extremely poor prognoses, which provides new insights into tumors without effective treatment strategies. Meanwhile, with the gradual development of stereotactic biopsy techniques, it is gradually becoming easier and safer to obtain live DIPG tissue, and the understanding of the immune properties of DIPG has also increased. On this basis, a series of immunotherapy studies of DIPG are under way, some of which have shown encouraging results. Herein, we review the current understanding of the immune characteristics of DIPG and critically reveal the limitations of current immune research, as well as the opportunities and challenges for immunological therapies in DIPG, hoping to clarify the development of novel immunotherapies for DIPG treatment.
Collapse
|
6
|
Sun K, Fei X, Xu M, Xu R, Xu M. FCGR3A Is a Prognostic Biomarker and Correlated with Immune Infiltrates in Lower-Grade Glioma. JOURNAL OF ONCOLOGY 2022; 2022:9499317. [PMID: 39280892 PMCID: PMC11401682 DOI: 10.1155/2022/9499317] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 05/22/2022] [Accepted: 05/31/2022] [Indexed: 09/18/2024]
Abstract
Low-grade gliomas (LGGs) are primary invasive brain tumors that grow slowly but are incurable and eventually develop into high malignant glioma. Fc fragment of IgG receptor IIIa (FCGR3A) gene polymorphism may correlate with some cancers' treatment responses. However, the expression and prognosis value of FCGR3A and correlation with tumor-immune infiltrate in LGG remain unclear. FCGR3A mRNA expression in gastric cancer (GC) was examined using TIMER and GEPIA databases. Correlations between FCGR3A expression and clinicopathological parameters were analyzed using ULACAN and CGGA databases. GEPIA, OncoLnc, and ULACAN databases were used to examine the clinical prognostic significance of FCGR3A in LGG. TIMER was used to analyze the correlations among FCGR3A and tumor-infiltrating immune cells. Signaling pathways related to FCGR3A expression were identified by LinkedOmics. We found that FCGR3A expression was higher in LGG than in normal tissue and was correlated with various clinical parameters. In addition, high FCGR3A expression predicted poor overall survival in LGG. More importantly, FCGR3A expression positively correlated with immune checkpoint molecules, including PD1, PD-L1, PD-L2, CTLA4, LAG-3 and TIM-3, and tumor-associated macrophage (TAM) gene markers in LGG. GO and KEGG pathway analyses indicated that TUBA1C may potentially regulate the pathogenesis of LGG through immune-related pathways. These findings indicated that FCGR3A plays a vital role in the infiltration of immune cells and could constitute a promising prognostic biomarker in LGG patients.
Collapse
Affiliation(s)
- Kai Sun
- Department of Neurosurgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Xiaowei Fei
- Department of Neurosurgery, The First Affiliated Hospital of the Fourth Military Medical University, Xi'an 710032, China
| | - Mingwei Xu
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Ruxiang Xu
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Minhui Xu
- Department of Neurosurgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| |
Collapse
|
7
|
du Chatinier A, Meel MH, Das AI, Metselaar DS, Waranecki P, Bugiani M, Breur M, Simonds EF, Lu ED, Weiss WA, Garcia Vallejo JJ, Hoving EW, Phoenix TN, Hulleman E. Generation of Immunocompetent Syngeneic Allograft Mouse Models for Pediatric Diffuse Midline Glioma. Neurooncol Adv 2022; 4:vdac079. [PMID: 35733514 PMCID: PMC9210310 DOI: 10.1093/noajnl/vdac079] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Abstract
Background
Diffuse midline gliomas (DMG) are highly malignant incurable pediatric brain tumors. A lack of effective treatment options highlights the need to investigate novel therapeutic strategies. This includes the use of immunotherapy, which has shown promise in other hard-to-treat tumors. To facilitate preclinical immunotherapeutic research, immunocompetent mouse models that accurately reflect the unique genetic, anatomical, and histological features of DMG patients are warranted.
Methods
We established cell cultures from primary DMG mouse models (C57BL/6) that were generated by brainstem targeted intra-uterine electroporation (IUE). We subsequently created allograft DMG mouse models by orthotopically implanting these tumor cells into syngeneic mice. Immunohistochemistry and -fluorescence, mass cytometry, and cell-viability assays were then used to verify that these murine tumors recapitulated human DMG.
Results
We generated three genetically distinct allograft models representing histone 3 wildtype (H3 WT) and K27M-mutant DMG (H3.3 K27M and H3.1 K27M). These allograft models recapitulated the histopathologic phenotype of their human counterparts, including their diffuse infiltrative growth and expression of DMG-associated antigens. These murine pontine tumors also exhibited an immune microenvironment similar to human DMG, characterized by considerable myeloid cell infiltration and a paucity of T-lymphocytes and NK cells. Finally, we show that these murine DMG cells display similar sensitivity to histone deacetylase (HDAC) inhibition as patient-derived DMG cells.
Conclusions
We created and validated an accessible method to generate immunocompetent allograft models reflecting different subtypes of DMG. These models adequately recapitulated the histopathology, immune microenvironment, and therapeutic response of human DMG, providing useful tools for future preclinical studies.
Collapse
Affiliation(s)
| | - Michaël H Meel
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Arvid I Das
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | | | - Piotr Waranecki
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Marianna Bugiani
- Department of Pathology, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Marjolein Breur
- Department of Pathology, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Erin F Simonds
- Departments of Neurology, Neurological Surgery, and Pediatrics, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California, USA
| | - Edbert D Lu
- Departments of Neurology, Neurological Surgery, and Pediatrics, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California, USA
| | - William A Weiss
- Departments of Neurology, Neurological Surgery, and Pediatrics, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California, USA
| | - Juan J Garcia Vallejo
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Eelco W Hoving
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Timothy N Phoenix
- Division of Pharmaceutical Sciences, College of Pharmacy, University of Cincinnati/ Research in Patient Services, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Esther Hulleman
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| |
Collapse
|
8
|
Pinho MP, Lepski GA, Rehder R, Chauca-Torres NE, Evangelista GCM, Teixeira SF, Flatow EA, de Oliveira JV, Fogolin CS, Peres N, Arévalo A, Alves V, Barbuto JAM, Bergami-Santos PC. Near-Complete Remission of Glioblastoma in a Patient Treated with an Allogenic Dendritic Cell-Based Vaccine: The Role of Tumor-Specific CD4+T-Cell Cytokine Secretion Pattern in Predicting Response and Recurrence. Int J Mol Sci 2022; 23:5396. [PMID: 35628206 PMCID: PMC9141410 DOI: 10.3390/ijms23105396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/19/2022] [Accepted: 05/03/2022] [Indexed: 01/27/2023] Open
Abstract
Immunotherapy has brought hope to the fight against glioblastoma, but its efficacy remains unclear. We present the case of CST, a 25-year-old female patient with a large right-hemisphere glioblastoma treated with a dendritic-tumor cell fusion vaccine. CST showed a near-complete tumor response, with a marked improvement in her functional status and simultaneous increases in tumor-specific CD8+ and CD4+ T cells. Two months before recurrence, the frequency of tumor-specific T cells decreased, while that of IL-17 and CD4+ T cells increased. CST passed away 15 months after enrollment. In this illustrative case, the tumor-specific CD4+ T-cell numbers and phenotype behaved as treatment efficacy biomarkers, highlighting the key role of the latter in glioblastoma immunotherapy.
Collapse
Affiliation(s)
- Mariana P. Pinho
- Department of Immunology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-000, Brazil; (M.P.P.); (N.E.C.-T.); (G.C.M.E.); (S.F.T.); (E.A.F.); (J.V.d.O.); (C.S.F.); (N.P.)
| | - Guilherme A. Lepski
- Hospital das Clínicas HCFMUSP, LIM26, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-000, Brazil; (G.A.L.); (A.A.)
- Department of Neurosurgery, Eberhard-Karls University, 72074 Tuebingen, Germany
| | | | - Nadia E. Chauca-Torres
- Department of Immunology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-000, Brazil; (M.P.P.); (N.E.C.-T.); (G.C.M.E.); (S.F.T.); (E.A.F.); (J.V.d.O.); (C.S.F.); (N.P.)
| | - Gabriela C. M. Evangelista
- Department of Immunology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-000, Brazil; (M.P.P.); (N.E.C.-T.); (G.C.M.E.); (S.F.T.); (E.A.F.); (J.V.d.O.); (C.S.F.); (N.P.)
| | - Sarah F. Teixeira
- Department of Immunology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-000, Brazil; (M.P.P.); (N.E.C.-T.); (G.C.M.E.); (S.F.T.); (E.A.F.); (J.V.d.O.); (C.S.F.); (N.P.)
| | - Elizabeth A. Flatow
- Department of Immunology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-000, Brazil; (M.P.P.); (N.E.C.-T.); (G.C.M.E.); (S.F.T.); (E.A.F.); (J.V.d.O.); (C.S.F.); (N.P.)
| | - Jaqueline V. de Oliveira
- Department of Immunology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-000, Brazil; (M.P.P.); (N.E.C.-T.); (G.C.M.E.); (S.F.T.); (E.A.F.); (J.V.d.O.); (C.S.F.); (N.P.)
| | - Carla S. Fogolin
- Department of Immunology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-000, Brazil; (M.P.P.); (N.E.C.-T.); (G.C.M.E.); (S.F.T.); (E.A.F.); (J.V.d.O.); (C.S.F.); (N.P.)
| | - Nataly Peres
- Department of Immunology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-000, Brazil; (M.P.P.); (N.E.C.-T.); (G.C.M.E.); (S.F.T.); (E.A.F.); (J.V.d.O.); (C.S.F.); (N.P.)
- Laboratory of Experimental Surgery (LIM-26), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-000, Brazil
| | - Analía Arévalo
- Hospital das Clínicas HCFMUSP, LIM26, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-000, Brazil; (G.A.L.); (A.A.)
| | - Venâncio Alves
- Department of Pathology, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 01246-903, Brazil;
| | - José A. M. Barbuto
- Department of Immunology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-000, Brazil; (M.P.P.); (N.E.C.-T.); (G.C.M.E.); (S.F.T.); (E.A.F.); (J.V.d.O.); (C.S.F.); (N.P.)
- Laboratory of Medical Investigation in Pathogenesis and Targeted Therapy in Onco-Immuno-Hematology (LIM-31), Department of Hematology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-000, Brazil
| | - Patricia C. Bergami-Santos
- Department of Immunology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-000, Brazil; (M.P.P.); (N.E.C.-T.); (G.C.M.E.); (S.F.T.); (E.A.F.); (J.V.d.O.); (C.S.F.); (N.P.)
| |
Collapse
|
9
|
Liu L, Shah K. The Potential of the Gut Microbiome to Reshape the Cancer Therapy Paradigm: A Review. JAMA Oncol 2022; 8:1059-1067. [PMID: 35482355 DOI: 10.1001/jamaoncol.2022.0494] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Importance The gut microbiome, home to the vast kingdom of diverse commensal bacteria and other microorganisms residing within the gut, was once thought to only have roles primarily centered on digestive functions. However, recent advances in sequencing technology have elucidated intricate roles of the gut microbiome in cancer development and efficacy of therapeutic response that need to be comprehensively addressed from a clinically translational angle. Observations This review aims to highlight the current understanding of the association of the gut microbiome with the therapeutic response to immunotherapy, chemotherapy, radiotherapy, cancer surgery, and more, while also contextualizing possible synergistic strategies with the microbiome for tackling some of the most challenging tumors. It also provides insights on contemporary methods that target the microbiota and the current progression of findings being translated from bench to bedside. Conclusions and Relevance Ultimately, the importance of gut bacteria in cancer therapy cannot be overstated in its potential for ushering in a new era of cancer treatments. With the understanding that the microbiome may play critical roles in the tumor microenvironment, holistic approaches that integrate microbiome-modulating treatments with biological, immune, cell-based, and surgical cancer therapies should be explored.
Collapse
Affiliation(s)
- Longsha Liu
- Center for Stem Cell and Translational Immunotherapy (CSTI), Harvard Medical School, Boston, Massachusetts.,Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Khalid Shah
- Center for Stem Cell and Translational Immunotherapy (CSTI), Harvard Medical School, Boston, Massachusetts.,Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.,Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts
| |
Collapse
|
10
|
Jiang C, Zhang H, Wu W, Wang Z, Dai Z, Zhang L, Liu Z, Cheng Q. Immune Characteristics of LYN in Tumor Microenvironment of Gliomas. Front Cell Dev Biol 2022; 9:760929. [PMID: 35186945 PMCID: PMC8847791 DOI: 10.3389/fcell.2021.760929] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 12/20/2021] [Indexed: 01/22/2023] Open
Abstract
The prognosis of gliomas is poor and there are limited therapeutic approaches. Immunotherapy has become a promising treatment for gliomas. Here, we explored the expression pattern of Lck/yes-related protein tyrosine kinase (LYN) in gliomas and assessed its value as an immunotherapy biomarker. Transcriptional data was mined from two publicly available datasets, TCGA and CGGA, and used to investigate the correlation between LYN and clinical characteristics including patient prognosis, somatic mutation, and immune infiltrating features in gliomas. Besides, the correlation between LYN and classical immune checkpoint molecules was explored. Glioma samples obtained from the Xiangya Hospital cohort were used for immunohistochemistry staining. High expression level of LYN was observed in advanced gliomas and other cancer types, which predicted a worse prognosis. LYN stratified patients’ survival in the Xiangya cohort and was also significantly associated with infiltrating immune cell types and inflammatory activities in the tumor microenvironment. LYN was involved in tumor mutation, correlated with the regulation of oncogenic genes, and also showed a significant positive correlation with PD-L1. LYN can be a potential diagnostic marker and immunotherapy marker in gliomas.
Collapse
Affiliation(s)
- Chonghua Jiang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Hao Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Wantao Wu
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Zeyu Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Ziyu Dai
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Liyang Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Clinical Diagnosis and Therapeutic Center of Glioma, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Quan Cheng, ; Zhixiong Liu, ; Liyang Zhang,
| | - Zhixiong Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Quan Cheng, ; Zhixiong Liu, ; Liyang Zhang,
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Quan Cheng, ; Zhixiong Liu, ; Liyang Zhang,
| |
Collapse
|
11
|
Abstract
RNA-based therapeutics have entered the mainstream with seemingly limitless possibilities to treat all categories of neurological disease. Here, common RNA-based drug modalities such as antisense oligonucleotides, small interfering RNAs, RNA aptamers, RNA-based vaccines and mRNA drugs are reviewed highlighting their current and potential applications. Rapid progress has been made across rare genetic diseases and neurodegenerative disorders, but safe and effective delivery to the brain remains a significant challenge for many applications. The advent of individualized RNA-based therapies for ultra-rare diseases is discussed against the backdrop of the emergence of this field into more common conditions such as Alzheimer's disease and ischaemic stroke. There remains significant untapped potential in the use of RNA-based therapeutics for behavioural disorders and tumours of the central nervous system; coupled with the accelerated development expected over the next decade, the true potential of RNA-based therapeutics to transform the therapeutic landscape in neurology remains to be uncovered.
Collapse
Affiliation(s)
- Karen Anthony
- Centre for Physical Activity and Life Sciences, University of Northampton, Northampton, UK
| |
Collapse
|
12
|
Bausart M, Préat V, Malfanti A. Immunotherapy for glioblastoma: the promise of combination strategies. J Exp Clin Cancer Res 2022; 41:35. [PMID: 35078492 PMCID: PMC8787896 DOI: 10.1186/s13046-022-02251-2] [Citation(s) in RCA: 106] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 01/11/2022] [Indexed: 12/11/2022] Open
Abstract
Glioblastoma (GBM) treatment has remained almost unchanged for more than 20 years. The current standard of care involves surgical resection (if possible) followed by concomitant radiotherapy and chemotherapy. In recent years, immunotherapy strategies have revolutionized the treatment of many cancers, increasing the hope for GBM therapy. However, mostly due to the high, multifactorial immunosuppression occurring in the microenvironment, the poor knowledge of the neuroimmune system and the presence of the blood-brain barrier, the efficacy of immunotherapy in GBM is still low. Recently, new strategies for GBM treatments have employed immunotherapy combinations and have provided encouraging results in both preclinical and clinical studies. The lessons learned from clinical trials highlight the importance of tackling different arms of immunity. In this review, we aim to summarize the preclinical evidence regarding combination immunotherapy in terms of immune and survival benefits for GBM management. The outcomes of recent studies assessing the combination of different classes of immunotherapeutic agents (e.g., immune checkpoint blockade and vaccines) will be discussed. Finally, future strategies to ameliorate the efficacy of immunotherapy and facilitate clinical translation will be provided to address the unmet medical needs of GBM.
Collapse
Affiliation(s)
- Mathilde Bausart
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
| | - Véronique Préat
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium.
| | - Alessio Malfanti
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
| |
Collapse
|
13
|
Lu J, Ding J, Liu Z, Chen T. Retrospective analysis of the preparation and application of immunotherapy in cancer treatment (Review). Int J Oncol 2022; 60:12. [PMID: 34981814 PMCID: PMC8759346 DOI: 10.3892/ijo.2022.5302] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 12/20/2021] [Indexed: 12/11/2022] Open
Abstract
Monoclonal antibody technology plays a vital role in biomedical and immunotherapy, which greatly promotes the study of the structure and function of genes and proteins. To date, monoclonal antibodies have gone through four stages: murine monoclonal antibody, chimeric monoclonal antibody, humanised monoclonal antibody and fully human monoclonal antibody; thousands of monoclonal antibodies have been used in the fields of biology and medicine, playing a special role in the pathogenesis, diagnosis and treatment of disease. In this review, we compare the advantages and disadvantages of hybridoma technology, phage display technology, ribosome display technology, transgenic mouse technology, single B cell monoclonal antibody generation technologies, and forecast the promising applications of these technologies in clinical medicine, disease diagnosis and tumour treatment.
Collapse
Affiliation(s)
- Jiachen Lu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Jianing Ding
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zhaoxia Liu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Tingtao Chen
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
14
|
Burster T, Traut R, Yermekkyzy Z, Mayer K, Westhoff MA, Bischof J, Knippschild U. Critical View of Novel Treatment Strategies for Glioblastoma: Failure and Success of Resistance Mechanisms by Glioblastoma Cells. Front Cell Dev Biol 2021; 9:695325. [PMID: 34485282 PMCID: PMC8415230 DOI: 10.3389/fcell.2021.695325] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 07/29/2021] [Indexed: 12/28/2022] Open
Abstract
According to the invasive nature of glioblastoma, which is the most common form of malignant brain tumor, the standard care by surgery, chemo- and radiotherapy is particularly challenging. The presence of glioblastoma stem cells (GSCs) and the surrounding tumor microenvironment protects glioblastoma from recognition by the immune system. Conventional therapy concepts have failed to completely remove glioblastoma cells, which is one major drawback in clinical management of the disease. The use of small molecule inhibitors, immunomodulators, immunotherapy, including peptide and mRNA vaccines, and virotherapy came into focus for the treatment of glioblastoma. Although novel strategies underline the benefit for anti-tumor effectiveness, serious challenges need to be overcome to successfully manage tumorigenesis, indicating the significance of developing new strategies. Therefore, we provide insights into the application of different medications in combination to boost the host immune system to interfere with immune evasion of glioblastoma cells which are promising prerequisites for therapeutic approaches to treat glioblastoma patients.
Collapse
Affiliation(s)
- Timo Burster
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Rebecca Traut
- Department of General and Visceral Surgery, Surgery Center, Ulm University Hospital, Ulm, Germany
| | - Zhanerke Yermekkyzy
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Katja Mayer
- Department of General and Visceral Surgery, Surgery Center, Ulm University Hospital, Ulm, Germany
| | - Mike-Andrew Westhoff
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Joachim Bischof
- Department of General and Visceral Surgery, Surgery Center, Ulm University Hospital, Ulm, Germany
| | - Uwe Knippschild
- Department of General and Visceral Surgery, Surgery Center, Ulm University Hospital, Ulm, Germany
| |
Collapse
|
15
|
Taxifolin Targets PI3K and mTOR and Inhibits Glioblastoma Multiforme. JOURNAL OF ONCOLOGY 2021; 2021:5560915. [PMID: 34462635 PMCID: PMC8403040 DOI: 10.1155/2021/5560915] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 07/31/2021] [Indexed: 01/12/2023]
Abstract
Glioblastoma multiforme (GBM), the most common malignant primary brain tumor, has a very poor prognosis. With increasing knowledge of tumor molecular biology, targeted therapies are becoming increasingly integral to comprehensive GBM treatment strategies. mTOR is a key downstream molecule of the PI3K/Akt signaling pathway, integrating input signals from growth factors, nutrients, and energy sources to regulate cell growth and cell proliferation through multiple cellular responses. mTOR/PI3K dual-targeted therapy has shown promise in managing various cancers. Here, we report that taxifolin, a flavanone commonly found in milk thistle, inhibited mTOR/PI3K, promoted autophagy, and suppressed lipid synthesis in GBM. In silico analysis showed that taxifolin can bind to the rapamycin binding site of mTOR and the catalytic site of PI3K (p110α). In in vitro experiments, taxifolin inhibited mTOR and PI3K activity in five different glioma cell lines. Lastly, we showed that taxifolin suppressed tumors in mice; stimulated expression of autophagy-related genes LC3B-II, Atg7, atg12, and Beclin-1; and inhibited expression of fatty acid synthesis-related genes C/EBPα, PPARγ, FABP4, and FAS. Our observations suggest that taxifolin is potentially a valuable drug for treating GBM.
Collapse
|
16
|
Chen D, Li G, Ji C, Lu Q, Qi Y, Tang C, Xiong J, Hu J, Yasar FBA, Zhang Y, Hoon DSB, Yao Y, Zhou L. Enhanced B7-H4 expression in gliomas with low PD-L1 expression identifies super-cold tumors. J Immunother Cancer 2021; 8:jitc-2019-000154. [PMID: 32457124 PMCID: PMC7253052 DOI: 10.1136/jitc-2019-000154] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2020] [Indexed: 12/29/2022] Open
Abstract
Background Characterizing expression profiles of different immune checkpoint molecules are promising for personalized checkpoint inhibitory immunotherapy. Gliomas have been shown as potential targets for immune checkpoint inhibitors recently. Our study was performed to determine coexpression levels of two major B7 immune regulatory molecules programmed death ligand 1 (PD-L1) and B7-H4, both of which have been demonstrated to inhibit antitumor host immunity in gliomas. Methods We assessed tumor tissues from stage II–IV primary gliomas (n=505) by immunohistochemistry (IHC) for protein levels of both PD-L1 and B7-H4. Gene coexpression analysis assessing clusters based on extent of PD-L1/B7-H4 classifier genes expression were investigated in two transcriptome datasets (The Cancer Genome Atlas and Chinese Glioma Genome Atlas). In addition, levels of immune cell infiltrates were estimated with IHC and RNA-seq data for assessing the tumor immune microenvironment of PD-L1/B7-H4 subgroups. Results High expression of PD-L1 and B7-H4 in gliomas was 23% and 20%, respectively, whereas coexpression of two proteins at high levels was limited to 2% of the cases. Comparable results were seen in RNA-seq datasets where PD-L1 mRNA expression levels negatively correlated with that of B7-H4. Gene coexpression modules clustered within each grade of gliomas demonstrated lack of double-high modules (cluster with high expression of both PD-L1 and B7-H4 classifier genes). B7-H4 mRNA expression levels showed negative correlation with extent of immune cell infiltration and High-B7-H4 module gliomas (high B7-H4 but low PD-L1 classifier genes expression) had less tumor-infiltrating lymphocytes (TILs) and tumor-associated macrophages (TAMs). IHC assessment also showed few TILs and TAMs in High-B7-H4 subgroup gliomas. Conclusions The majority of gliomas express PD-L1 or B7-H4, however, coexpression of both at high levels is minimal. The high-B7-H4 patients could be considered as ‘super-cold’ gliomas with significantly deficient in TILs, suggesting that B7-H4 might inhibit T-cell trafficking into the central nervous system. This study demonstrated that PD-L1 and B7-H4 may serve as mutually compensatory immune checkpoint molecules in gliomas for immune targeted or active-specific immunotherapy. The distinct B7-H4 pathways modulating T-cell function and immune evasion in glioma patients deserved to be further explored in the future during immunotherapy.
Collapse
Affiliation(s)
- Di Chen
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Gaopeng Li
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, Brain Disease and Big Data Research Institute, College of Life Sciences & and Oceanography, Shenzhen University, Shenzhen, Guangdong, China
| | - Chunxia Ji
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China.,Immunology Laboratory, Neurosurgical Institute of Fudan University, Shanghai, China
| | - Qiqi Lu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China.,Immunology Laboratory, Neurosurgical Institute of Fudan University, Shanghai, China
| | - Ying Qi
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Chao Tang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China.,Immunology Laboratory, Neurosurgical Institute of Fudan University, Shanghai, China
| | - Ji Xiong
- Department of Pathology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jian Hu
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Fatma Betul Aksoy Yasar
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yan Zhang
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, Brain Disease and Big Data Research Institute, College of Life Sciences & and Oceanography, Shenzhen University, Shenzhen, Guangdong, China
| | - Dave S B Hoon
- Department of Translational Molecular Medicine, John Wayne Cancer Institute, Providence Health Systems, Santa Monica, California, USA
| | - Yu Yao
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Liangfu Zhou
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China.,Immunology Laboratory, Neurosurgical Institute of Fudan University, Shanghai, China
| |
Collapse
|
17
|
Liu F, Xu XH, Li CY, Zhang TT, Yin SL, Liu GQ, Hu F, Yu SB, Chen XQ. Rapid tumor recurrence in a novel murine GBM surgical model is associated with Akt/PD-L1/vimentin signaling. Biochem Biophys Res Commun 2021; 569:1-9. [PMID: 34216991 DOI: 10.1016/j.bbrc.2021.06.072] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/15/2021] [Accepted: 06/21/2021] [Indexed: 12/20/2022]
Abstract
Glioblastoma multiforme (GBM) is the most common primary malignant brain tumor without curable therapy. Surgical resection remains the first choice of patients with GBM but tumors relapse rapidly even combined with conventional chemoradiotherapy. The mechanism of GBM rapid recurrence is poorly understood, which is largely due to the lack of an appropriate animal model, thus heavily impedes the improvement of postoperative therapy. Here we established a highly reproducible mouse GBM surgical model by using the syngeneic G422TN-GBM cells, which faithfully recapitulates the features of rapid recurrence of human GBM after surgery. Implanting 2 × 103-5 × 104 of G422TN-GBM cells in mouse cerebral cortex caused death in all animal within 23 days, while surgery was an effective therapy but not curable. After complete removal of visible tumors on day 5-9 of tumor growth, the tumors recurred macroscopically within 5 days accompanied by increasing infiltrative cancer foci. Mechanistically, the rapid recurrence of resected tumors was positively correlated to early Akt activation, which subsequently upregulated PD-L1/Vimentin and promoted proliferation/migration of cancer cells. In addition, environmental astrocytic activation with strong PD-L1 signal was prominent. Taken together, we provided a novel GBM surgical recurrence model for preclinical studies and suggested complicated recurring mechanisms involving in strong oncogenic signaling as well as immune inhibitory signals from both GBM cells and their neighboring astrocytes.
Collapse
Affiliation(s)
- Feng Liu
- Department of Pharmacy, The First Affiliated Hospital of Yangtze University, Jingzhou, 434000, Hubei, China
| | - Xiao Hong Xu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Ministry of Education for Neurological Disorders, Wuhan, 430030, Hubei, China
| | - Chun Yang Li
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Ministry of Education for Neurological Disorders, Wuhan, 430030, Hubei, China
| | - Ting Ting Zhang
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Ministry of Education for Neurological Disorders, Wuhan, 430030, Hubei, China
| | - Song Lin Yin
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Ministry of Education for Neurological Disorders, Wuhan, 430030, Hubei, China
| | - Guo Qiang Liu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Ministry of Education for Neurological Disorders, Wuhan, 430030, Hubei, China
| | - Feng Hu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Shang Bin Yu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Ministry of Education for Neurological Disorders, Wuhan, 430030, Hubei, China
| | - Xiao Qian Chen
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Ministry of Education for Neurological Disorders, Wuhan, 430030, Hubei, China.
| |
Collapse
|
18
|
Patel V, Shah J. The current and future aspects of glioblastoma: Immunotherapy a new hope? Eur J Neurosci 2021; 54:5120-5142. [PMID: 34107127 DOI: 10.1111/ejn.15343] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/28/2021] [Accepted: 06/03/2021] [Indexed: 12/13/2022]
Abstract
Glioblastoma (GBM) is the most perilous and highly malignant in all the types of brain tumor. Regardless of the treatment, the diagnosis of the patients in GBM is very poor. The average survival rate is only 21 months after multimodal combinational therapies, which include chemotherapy, radiation, and surgery. Due to the intrusive and infiltrative nature of GBM, it requires elective therapy for specific targeting of tumor cells. Tumor vaccine in a form of immunotherapy has potential to address this need. Nanomedicine-based immunotherapies have clutch the trigger of systemic and specific immune response against tumor cells, which might be the approach to eliminating the unrelieved cancer. In this mechanism, combination of immunomodulators with specific target and appropriate strategic vaccines can stifle tumor anti-immune defense system and/or increase the capabilities of the body to move up immunity against the tumor. Here, we explore the different types of immunotherapies and vaccines for brain tumor treatment and their clinical trials, which bring the feasibility of the future of personalized vaccine of nanomedicine-based immunotherapies for the brain tumor. We believe that immunotherapy could result in a significantly more stable reaction in GBM patients.
Collapse
Affiliation(s)
- Vimal Patel
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, India
| | - Jigar Shah
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, India
| |
Collapse
|
19
|
Yokota C, Nakata J, Takano K, Nakajima H, Hayashibara H, Minagawa H, Chiba Y, Hirayama R, Kijima N, Kinoshita M, Hashii Y, Tsuboi A, Oka Y, Oji Y, Kumanogoh A, Sugiyama H, Kagawa N, Kishima H. Distinct difference in tumor-infiltrating immune cells between Wilms' tumor gene 1 peptide vaccine and anti-programmed cell death-1 antibody therapies. Neurooncol Adv 2021; 3:vdab091. [PMID: 34355173 PMCID: PMC8331049 DOI: 10.1093/noajnl/vdab091] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Background Wilms’ tumor gene 1 (WT1) peptide vaccine and anti-programmed cell death-1 (anti-PD-1) antibody are expected as immunotherapies to improve the clinical outcome of glioblastoma. The aims of this study were to clarify how each immunotherapy affects tumor-infiltrating immune cells (TIIs) and to determine whether the combination of these two therapies could synergistically work. Methods Mice were transplanted with WT1 and programmed cell death-ligand 1 doubly expressing glioblastoma cells into brain followed by treatment with WT1 peptide vaccine, anti-PD-1 antibody, or the combination of the two, and survival of each therapy was compared. CD45+ cells were positively selected as TIIs from the brains with tumors, and TIIs were compared between WT1 peptide vaccine and anti-PD-1 antibody therapies. Results Most mice seemed to be cured by the combination therapy with WT1 peptide vaccine and anti-PD-1 antibody, which was much better survival than each monotherapy. A large number of CD4+ T cells, CD8+ T cells, and NK cells including WT1-specific CD8+ and CD4+ T cells infiltrated into the glioblastoma in WT1 peptide vaccine-treated mice. On the other hand, the number of TIIs did not increase, but instead PD-1 molecule expression was decreased on the majority of the tumor-infiltrating CD8+ T cells in the anti-PD-1 antibody-treated mice. Conclusion Our results clearly demonstrated that WT1 peptide vaccine and anti-PD-1 antibody therapies worked in the different steps of cancer-immunity cycle and that the combination of the two therapies could work synergistically against glioblastoma.
Collapse
Affiliation(s)
- Chisato Yokota
- Department of Neurosurgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Jun Nakata
- Department of Clinical Laboratory and Biomedical Sciences, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Koji Takano
- Department of Neurosurgery, Osaka International Cancer Institute, Osaka, Osaka, Japan
| | - Hiroko Nakajima
- Department of Cancer Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hiromu Hayashibara
- Department of Clinical Laboratory and Biomedical Sciences, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hikaru Minagawa
- Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yasuyoshi Chiba
- Department of Neurosurgery, Osaka Women's and Children's Hospital, Osaka, Izumi, Japan
| | - Ryuichi Hirayama
- Department of Neurosurgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Noriyuki Kijima
- Department of Neurosurgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Manabu Kinoshita
- Department of Neurosurgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yoshiko Hashii
- Department of Cancer Immunotherapy, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Akihiro Tsuboi
- Department of Cancer Immunotherapy, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yoshihiro Oka
- Department of Cancer Stem Cell Biology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.,Department of Immunopathology, WP1 Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Yusuke Oji
- Department of Clinical Laboratory and Biomedical Sciences, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.,Department of Immunopathology, WP1 Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Haruo Sugiyama
- Department of Cancer Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Naoki Kagawa
- Department of Neurosurgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Haruhiko Kishima
- Department of Neurosurgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| |
Collapse
|
20
|
Alghamri MS, McClellan BL, Hartlage MS, Haase S, Faisal SM, Thalla R, Dabaja A, Banerjee K, Carney SV, Mujeeb AA, Olin MR, Moon JJ, Schwendeman A, Lowenstein PR, Castro MG. Targeting Neuroinflammation in Brain Cancer: Uncovering Mechanisms, Pharmacological Targets, and Neuropharmaceutical Developments. Front Pharmacol 2021; 12:680021. [PMID: 34084145 PMCID: PMC8167057 DOI: 10.3389/fphar.2021.680021] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 05/04/2021] [Indexed: 12/11/2022] Open
Abstract
Gliomas are one of the most lethal types of cancers accounting for ∼80% of all central nervous system (CNS) primary malignancies. Among gliomas, glioblastomas (GBM) are the most aggressive, characterized by a median patient survival of fewer than 15 months. Recent molecular characterization studies uncovered the genetic signatures and methylation status of gliomas and correlate these with clinical prognosis. The most relevant molecular characteristics for the new glioma classification are IDH mutation, chromosome 1p/19q deletion, histone mutations, and other genetic parameters such as ATRX loss, TP53, and TERT mutations, as well as DNA methylation levels. Similar to other solid tumors, glioma progression is impacted by the complex interactions between the tumor cells and immune cells within the tumor microenvironment. The immune system’s response to cancer can impact the glioma’s survival, proliferation, and invasiveness. Salient characteristics of gliomas include enhanced vascularization, stimulation of a hypoxic tumor microenvironment, increased oxidative stress, and an immune suppressive milieu. These processes promote the neuro-inflammatory tumor microenvironment which can lead to the loss of blood-brain barrier (BBB) integrity. The consequences of a compromised BBB are deleteriously exposing the brain to potentially harmful concentrations of substances from the peripheral circulation, adversely affecting neuronal signaling, and abnormal immune cell infiltration; all of which can lead to disruption of brain homeostasis. In this review, we first describe the unique features of inflammation in CNS tumors. We then discuss the mechanisms of tumor-initiating neuro-inflammatory microenvironment and its impact on tumor invasion and progression. Finally, we also discuss potential pharmacological interventions that can be used to target neuro-inflammation in gliomas.
Collapse
Affiliation(s)
- Mahmoud S Alghamri
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Brandon L McClellan
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Margaret S Hartlage
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Santiago Haase
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Syed Mohd Faisal
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Rohit Thalla
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Ali Dabaja
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Kaushik Banerjee
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Stephen V Carney
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Anzar A Mujeeb
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Michael R Olin
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, United States.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| | - James J Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, United States.,Biointerfaces Institute, University of Michigan, Ann Arbor, MI, United States.,Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Anna Schwendeman
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, United States.,Biointerfaces Institute, University of Michigan, Ann Arbor, MI, United States
| | - Pedro R Lowenstein
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States.,Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States.,Biosciences Initiative in Brain Cancer, University of Michigan, Ann Arbor, MI, United States
| | - Maria G Castro
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States.,Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States.,Biosciences Initiative in Brain Cancer, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
21
|
Riva M, Wouters R, Sterpin E, Giovannoni R, Boon L, Himmelreich U, Gsell W, Van Ranst M, Coosemans A. Radiotherapy, Temozolomide, and Antiprogrammed Cell Death Protein 1 Treatments Modulate the Immune Microenvironment in Experimental High-Grade Glioma. Neurosurgery 2021; 88:E205-E215. [PMID: 33289503 DOI: 10.1093/neuros/nyaa421] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 07/02/2020] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND The lack of immune synergy with conventional chemoradiation could explain the failure of checkpoint inhibitors in current clinical trials for high-grade gliomas (HGGs). OBJECTIVE To analyze the impact of radiotherapy (RT), Temozolomide (TMZ) and antiprogrammed cell death protein 1 (αPD1) (as single or combined treatments) on the immune microenvironment of experimental HGGs. METHODS Mice harboring neurosphere /CT-2A HGGs received RT (4 Gy, single dose), TMZ (50 mg/kg, 4 doses) and αPD1 (100 μg, 3 doses) as monotherapies or combinations. The influence on survival, tumor volume, and tumor-infiltrating immune cells was analyzed. RESULTS RT increased total T cells (P = .0159) and cluster of differentiation (CD)8+ T cells (P = .0078) compared to TMZ. Lymphocyte subpopulations resulting from TMZ or αPD1 treatment were comparable with those of controls. RT reduced M2 tumor-associated macrophages/microglia (P = .0019) and monocytic myeloid derived suppressor cells (mMDSCs, P = .0003) compared to controls. The effect on mMDSC was also seen following TMZ and αPD1 treatment, although less pronounced (P = .0439 and P = .0538, respectively). Combining RT with TMZ reduced CD8+ T cells (P = .0145) compared to RT alone. Adding αPD1 partially mitigated this effect as shown by the increased CD8+ T cells/Tregs ratio, even if this result failed to reach statistical significance (P = .0973). Changing the combination sequence of RT, TMZ, and αPD1 did not alter survival nor the immune effects. CONCLUSION RT, TMZ, and αPD1 modify the immune microenvironment of HGG. The combination of RT with TMZ induces a strong immune suppression which cannot be effectively counteracted by αPD1.
Collapse
Affiliation(s)
- Matteo Riva
- Department of Oncology, Laboratory of Tumor Immunology and Immunotherapy, KU Leuven, Leuven, Belgium.,Department of Neurosurgery, University Hospital of Godinne, UCL Namur, Yvoir, Belgium
| | - Roxanne Wouters
- Department of Oncology, Laboratory of Tumor Immunology and Immunotherapy, KU Leuven, Leuven, Belgium
| | - Edmond Sterpin
- Department of Oncology, Laboratory of Experimental Radiotherapy, KU Leuven, Leuven, Belgium
| | - Roberto Giovannoni
- School of Medicine and Surgery, University of Milano Bicocca, Monza, Italy
| | - Louis Boon
- Polpharma Biologics, Utrecht, the Netherlands
| | - Uwe Himmelreich
- Department of Imaging and Pathology and Molecular Small Animal Imaging Center (MoSAIC), Biomedical MRI, KU Leuven, Leuven, Belgium
| | - Willy Gsell
- Department of Imaging and Pathology and Molecular Small Animal Imaging Center (MoSAIC), Biomedical MRI, KU Leuven, Leuven, Belgium
| | - Marc Van Ranst
- Rega Institute for Medical Research, Laboratory for Clinical and Epidemiological Virology, KU Leuven, Leuven, Belgium
| | - An Coosemans
- Department of Oncology, Laboratory of Tumor Immunology and Immunotherapy, KU Leuven, Leuven, Belgium.,Department of Gynaecology and Obstetrics, Leuven Cancer Institute, UZ Leuven, Leuven, Belgium
| |
Collapse
|
22
|
Wu J, Xie S, Li H, Zhang Y, Yue J, Yan C, Liu K, Liu Y, Xu R, Zheng G. Antitumor effect of IL-12 gene-modified bone marrow mesenchymal stem cells combined with Fuzheng Yiliu decoction in an in vivo glioma nude mouse model. J Transl Med 2021; 19:143. [PMID: 33827606 PMCID: PMC8028710 DOI: 10.1186/s12967-021-02809-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 03/26/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Glioma is a complex cancer with a high morbidity and high mortality. Bone marrow mesenchymal stem cells (BMSCs) have shown promise as an excellent cell/drug delivery vehicle for gene-targeted therapy; however, maintaining genetic stability and biological activity remains difficult. Furthermore, whether BMSCs support or inhibit tumor growth remains debated. This study investigated whether a traditional Chinese medicine fomular, Fuzheng Yiliu decoction (FYD) had a synergistic antitumor effect with IL-12 gene-modified BMSCs in glioma-bearing nude mice METHODS: The lentivirus-mediated IL-12 gene was transfected into primarily cultured BMSCs. A total of 72 BALB/c nude mice were used to establish xenograft models with glioma U251 cells and were divided into groups (n = 12) including blank control group, nude mouse model group (model group), lentiviral transfection of BMSC group with no gene loading (BMSC group), IL-12 lentivirus-transfected BMSC group (IL-12 + BMSC group), FYD treatment group (FYD group), and FYD treatment in IL-12 lentivirus-transfected BMSC group (FYD + IL-12 + BMSC group).. After treatment for 14 days, all mice were sacrificed to collect tumor tissue and serum for more detection, such as distribution of BMSCs, cell apoptosis in xenograft tumors, serum IL-12 and INF-γ levels, mouse weight and tumor volume were measured RESULTS: There were significantly more apoptotic cells in tumor tissue in IL-12 gene transfected group, FYD treatment group and FYD combining with IL-12 gene transfected group than that in the model group (P < 0.05). The FYD + IL-12 + BMSC group showed significantly higher Bax and lower Bcl-2 expression (P < 0.05), and serum IL-12 and INF-γ levels (P < 0.05) were higher than that in all other groups. After the intervention, this group also showed a strong inhibitory effect against tumor growth (P < 0.05) CONCLUSIONS: This study suggested FYD treatment combined with IL-12 gene-modified BMSCs shows synergistic antitumor effect in glioma-bearing nude mice.
Collapse
Affiliation(s)
- Jianjun Wu
- School of Public Health, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
- Key Laboratory of Dunhuang Medicine and Transformation at Provincial and Ministerial Level, Lanzhou, 730000, Gansu, China
- Provincial Key Laboratory of Chinese Medicine Prevention and Control of Chronic Diseases, Lanzhou, 730000, Gansu, China
| | - Shoupin Xie
- Department of Neurology, The First People's Hospital of Lanzhou City, Lanzhou, 730050, China
| | - Hailong Li
- School of Public Health, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
- Provincial Key Laboratory of Chinese Medicine Prevention and Control of Chronic Diseases, Lanzhou, 730000, Gansu, China
| | - Yanxia Zhang
- Key Laboratory of Dunhuang Medicine and Transformation at Provincial and Ministerial Level, Lanzhou, 730000, Gansu, China
- Provincial Key Laboratory of Chinese Medicine Prevention and Control of Chronic Diseases, Lanzhou, 730000, Gansu, China
| | - Jia Yue
- School of Public Health, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
- Provincial Key Laboratory of Chinese Medicine Prevention and Control of Chronic Diseases, Lanzhou, 730000, Gansu, China
| | - Chunlu Yan
- Key Laboratory of Dunhuang Medicine and Transformation at Provincial and Ministerial Level, Lanzhou, 730000, Gansu, China
- School of Integrated Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Kai Liu
- Key Laboratory of Dunhuang Medicine and Transformation at Provincial and Ministerial Level, Lanzhou, 730000, Gansu, China
- School of Integrated Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Yongqi Liu
- Key Laboratory of Dunhuang Medicine and Transformation at Provincial and Ministerial Level, Lanzhou, 730000, Gansu, China
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Rui Xu
- School of Public Health, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
- Key Laboratory of Dunhuang Medicine and Transformation at Provincial and Ministerial Level, Lanzhou, 730000, Gansu, China
| | - Guisen Zheng
- School of Public Health, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China.
- Provincial Key Laboratory of Chinese Medicine Prevention and Control of Chronic Diseases, Lanzhou, 730000, Gansu, China.
| |
Collapse
|
23
|
Checkpoint Inhibitors as High-Grade Gliomas Treatment: State of the Art and Future Perspectives. J Clin Med 2021; 10:jcm10071367. [PMID: 33810532 PMCID: PMC8036455 DOI: 10.3390/jcm10071367] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/15/2021] [Accepted: 03/17/2021] [Indexed: 12/18/2022] Open
Abstract
Glioblastoma (GBM) is the most common and aggressive malignant brain tumor in adults. Despite significant efforts, no therapies have demonstrated valuable survival benefit beyond the current standard of care. Immune checkpoint inhibitors (ICI) have revolutionized the treatment landscape and improved patient survival in many advanced malignancies. Unfortunately, these clinical successes have not been replicated in the neuro-oncology field so far. This review summarizes the status of ICI investigation in high-grade gliomas, critically presenting the available data from preclinical models and clinical trials. Moreover, we explore new approaches to increase ICI efficacy, with a particular focus on combinatorial strategies, and the potential biomarkers to identify patients most likely to benefit from immune checkpoint blockade.
Collapse
|
24
|
Wu S, Calero-Pérez P, Arús C, Candiota AP. Anti-PD-1 Immunotherapy in Preclinical GL261 Glioblastoma: Influence of Therapeutic Parameters and Non-Invasive Response Biomarker Assessment with MRSI-Based Approaches. Int J Mol Sci 2020; 21:ijms21228775. [PMID: 33233585 PMCID: PMC7699815 DOI: 10.3390/ijms21228775] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/16/2020] [Accepted: 11/18/2020] [Indexed: 12/21/2022] Open
Abstract
Glioblastomas (GBs) are malignant brain tumours with poor prognosis even after aggressive therapy. Programmed cell death-1 (PD-1) immune checkpoint blockade is a promising strategy in many types of cancer, but its therapeutic effects in GB remain low and associated with immune infiltration. Previous work suggests that oscillations of magnetic resonance spectroscopic imaging (MRSI)-based response pattern with chemotherapy could act as a biomarker of efficient immune system attack onto GBs. The presence of such oscillations with other monotherapies such as anti-PD-1 would reinforce its monitoring potential. Here, we confirm that the oscillatory behaviour of the response biomarker is also detected in mice treated with anti PD-1 immunotherapy both in combination with temozolomide and as monotherapy. This indicates that the spectral pattern changes observed during therapy response are shared by different therapeutic strategies, provided the host immune system is elicited and able to productively attack tumour cells. Moreover, the participation of the immune system in response is also supported by the rate of cured animals observed with different therapeutic strategies (in the range of 50–100% depending on the treatment), which also held long-term immune memory against tumour cells re-challenge. Taken together, our findings open the way for a translational use of the MRSI-based biomarker in patient-tailored GB therapy, including immunotherapy, for which reliable non-invasive biomarkers are still missing.
Collapse
Affiliation(s)
- Shuang Wu
- Departament de Bioquímica i Biologia Molecular, Unitat de Bioquímica de Biociències, Edifici Cs, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain; (S.W.); (P.C.-P.); (C.A.)
| | - Pilar Calero-Pérez
- Departament de Bioquímica i Biologia Molecular, Unitat de Bioquímica de Biociències, Edifici Cs, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain; (S.W.); (P.C.-P.); (C.A.)
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 09183 Cerdanyola del Vallès, Spain
| | - Carles Arús
- Departament de Bioquímica i Biologia Molecular, Unitat de Bioquímica de Biociències, Edifici Cs, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain; (S.W.); (P.C.-P.); (C.A.)
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 09183 Cerdanyola del Vallès, Spain
- Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
| | - Ana Paula Candiota
- Departament de Bioquímica i Biologia Molecular, Unitat de Bioquímica de Biociències, Edifici Cs, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain; (S.W.); (P.C.-P.); (C.A.)
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 09183 Cerdanyola del Vallès, Spain
- Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
- Correspondence:
| |
Collapse
|
25
|
Lin X, Wang Z, Yang G, Wen G, Zhang H. YTHDF2 correlates with tumor immune infiltrates in lower-grade glioma. Aging (Albany NY) 2020; 12:18476-18500. [PMID: 32986017 PMCID: PMC7585119 DOI: 10.18632/aging.103812] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 07/20/2020] [Indexed: 01/24/2023]
Abstract
Immunotherapy is an effective treatment for many cancer types. However, YTHDF2 effects on the prognosis of different tumors and correlation with tumor immune infiltration are unclear. Here, we analyzed The Cancer Genome Atlas and Gene Expression Omnibus data obtained through various web-based platforms. The analyses showed that YTHDF2 expression and associated prognoses may depend on cancer type. High YTHDF2 expression was associated with poor overall survival in lower-grade glioma (LGG). In addition, YTHDF2 expression positively correlated with expression of several immune cell markers, including PD-1, TIM-3, and CTLA-4, as well as tumor-associated macrophage gene markers, and isocitrate dehydrogenase 1 in LGG. These findings suggest that YTHDF2 is a potential prognostic biomarker that correlates with LGG tumor-infiltrating immune cells.
Collapse
Affiliation(s)
- Xiangan Lin
- Department of Cancer Chemotherapy, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangzhou 510000, China
| | - Zhichao Wang
- Department of Cancer Chemotherapy, Zengcheng District People’s Hospital of Guangzhou, Guangzhou 511300, China
| | - Guangda Yang
- Department of Cancer Chemotherapy, Zengcheng District People’s Hospital of Guangzhou, Guangzhou 511300, China
| | - Guohua Wen
- Department of Cancer Chemotherapy, Zengcheng District People’s Hospital of Guangzhou, Guangzhou 511300, China
| | - Hailiang Zhang
- Department of Cancer Chemotherapy, Zengcheng District People’s Hospital of Guangzhou, Guangzhou 511300, China
| |
Collapse
|
26
|
Zhang H, Fan F, Yu Y, Wang Z, Liu F, Dai Z, Zhang L, Liu Z, Cheng Q. Clinical characterization, genetic profiling, and immune infiltration of TOX in diffuse gliomas. J Transl Med 2020; 18:305. [PMID: 32762688 PMCID: PMC7409670 DOI: 10.1186/s12967-020-02460-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 07/26/2020] [Indexed: 01/19/2023] Open
Abstract
Background Immunotherapies targeting glioblastoma (GBM) have led to significant improvements in patient outcomes. TOX is closely associated with the immune environment surrounding tumors, but its role in gliomas is not fully understood. Methods Using data from The Cancer Genome Atlas (TCGA) and the Chinese Glioma Genome Atlas (CGGA), we analyzed the transcriptomes of 1691 WHO grade I-IV human glioma samples. The R language was used to perform most of the statistical analyses. Somatic mutations and somatic copy number variation (CNV) were analyzed using GISTIC 2.0. Results TOX was down-regulated in malignant gliomas compared to low grade gliomas, and upregulated in the proneural and IDH mutant subtypes of GBM. TOXlow tumours are associated with the loss of PTEN and amplification of EGFR, while TOXhigh tumours harbor frequent mutations in IDH1 (91%). TOX was highly expressed in leading edge regions of tumours. Gene ontology and pathway analyses demonstrated that TOX was enriched in multiple immune related processes including lymphocyte migration in GBM. Finally, TOX had a negative association with the infiltration of several immune cell types in the tumour microenvironment. Conclusion TOX has the potential to be a new prognostic marker for GBM.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fan Fan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China.,Center for Medical Genetics and Hunan Provincial Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuanqiang Yu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zeyu Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fangkun Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ziyu Dai
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Liyang Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China. .,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China. .,Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA. .,Clinical Diagnosis and Therapeutic Center of Glioma, Xiangya Hospital, Central South University, Changsha, 410078, Hunan, People's Republic of China.
| | - Zhixiong Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China. .,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China. .,Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China. .,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
27
|
He M, Chen X, Luo M, Ouyang L, Xie L, Huang Z, Liu A. Suppressor of cytokine signaling 1 inhibits the maturation of dendritic cells involving the nuclear factor kappa B signaling pathway in the glioma microenvironment. Clin Exp Immunol 2020; 202:47-59. [PMID: 32516488 DOI: 10.1111/cei.13476] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 04/22/2020] [Accepted: 05/24/2020] [Indexed: 12/30/2022] Open
Abstract
Recurrence and diffuse infiltration challenge traditional therapeutic strategies for malignant glioma. Immunotherapy appears to be a promising approach to obtain long-term survival. Dendritic cells (DCs), the most specialized and potent antigen-presenting cells (APCs), play an important part in initiating and amplifying both the innate and adaptive immune responses against cancer cells. However, cancer cells can escape from immune surveillance by inhibiting maturation of DCs. Until the present, molecular mechanisms of maturation inhibition of DCs in the tumor microenvironment (TME) have not been fully revealed. Our study showed that pretreatment with tumor-conditioned medium (TCM) collected from supernatant of primary glioma cells significantly suppressed the maturation of DCs. TCM pretreatment significantly changed the morphology of DCs, TCM decreased the expression levels of CD80, CD83, CD86 and interleukin (IL)-12p70, while it increased the expression levels of IL-10, transforming growth factor (TGF)-β and IL-6. RNA-Seq showed that TCM pretreatment significantly increased the gene expression level of suppressor of cytokine signaling 1 (SOCS1) in DCs. suppressor of cytokine signaling 1 (SOCS1) knock-down significantly antagonized the maturation inhibition of DCs by TCM, which was demonstrated by the restoration of maturation markers. TCM pretreatment also significantly suppressed T cell viability and T helper type 1 (Th1) response, and SOCS1 knock-down significantly antagonized this suppressive effect. Further, TCM pretreatment significantly suppressed p65 nuclear translocation and transcriptional activity in DCs, and SOCS1 knock-down significantly attenuated this suppressive effect. In conclusion, our research demonstrates that TCM up-regulate SOCS1 to suppress the maturation of DCs via the nuclear factor-kappa signaling pathway.
Collapse
Affiliation(s)
- M He
- Department of Neurosurgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - X Chen
- Department of Neurosurgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - M Luo
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - L Ouyang
- Department of Neurosurgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - L Xie
- Department of Neurosurgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Z Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Department of Neurosurgery, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - A Liu
- Department of Neurosurgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
28
|
Immunotherapy Approaches for Pediatric CNS Tumors and Associated Neurotoxicity. Pediatr Neurol 2020; 107:7-15. [PMID: 32113728 DOI: 10.1016/j.pediatrneurol.2020.01.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 12/07/2019] [Accepted: 01/15/2020] [Indexed: 12/24/2022]
Abstract
Treatment for brain tumors has recently shifted to using the power of the immune system to destroy cancer cells with promising results. Many immunotherapeutic approaches that have been used in adults, including checkpoint inhibitors, vaccine therapy, adoptive immunotherapy, such as chimeric antigen receptor T cell therapy, and viral therapy, are now being evaluated in children. Although these treatments work through different mechanisms, they all activate the immune system and can result in inflammation at the site of disease. This can be especially problematic in the confined area of the brain causing potentially severe neurological side effects, which are of special concern in children with central nervous system malignancies. Steroids can be helpful in the management of neurological complications but carry the risk of making immunotherapeutic approaches less effective. Alternative therapeutic interventions to mitigate side effects are being evaluated. This review describes the most common immunotherapeutic modalities that are now under study for the treatment of pediatric brain tumors, their rationale, associated neurotoxicities, and current management.
Collapse
|
29
|
Kuo YC, Rajesh R. Biomaterial-based drug delivery systems used to improve chemotherapeutic activity of pharmaceuticals and to target inhibitors of apoptosis proteins. J Taiwan Inst Chem Eng 2020. [DOI: 10.1016/j.jtice.2020.05.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
30
|
Abstract
INTRODUCTION Glioblastoma multiforme (GBM) is the most prevalent primary brain tumor. In spite of the rigorous multimodal treatment involving surgery and radiochemotherapy, GBM has a dismal prognosis and rapid relapsing potential. Hence, search for novel therapeutic agents still continues. Neoantigens are the tumor-specific antigens which arise due to somatic mutations in the tumor genome. In recent years, personalized vaccine approach targeting neoantigens has been explored widely in cancer immunotherapy and several efforts have also been made to revolutionize the immunotherapy of cold tumors such as GBM using neoantigen targeted vaccines. AREAS COVERED In this review, we discuss the clinical application of personalized neoantigen targeted vaccine strategy in GBM immunotherapy. While discussing this strategy, we brief about the current challenges faced in GBM treatment by the novel immunotherapeutics. EXPERT OPINION To date, very few vaccines developed for GBM have reached till phase III clinical development. Early-phase clinical trials of GBM neoantigen vaccines have shown promising clinical outcomes and therefore, its rapid clinical development is warranted. Advent of newer and faster techniques such as next-generation sequencing will drive the faster clinical development of multiplex neoantigen vaccines and hence, increase in the clinical trials is expected.
Collapse
Affiliation(s)
- Vaishali Y Londhe
- Shobhaben Pratapbhai Patel School of Pharmacy &, Technology Management, SVKM's NMIMS University , Mumbai, India
| | - Varada Date
- Shobhaben Pratapbhai Patel School of Pharmacy &, Technology Management, SVKM's NMIMS University , Mumbai, India
| |
Collapse
|
31
|
Gong H, Gao S, Yu C, Li M, Liu P, Zhang G, Song J, Zheng J. Effect and mechanism of YB-1 knockdown on glioma cell growth, migration, and apoptosis. Acta Biochim Biophys Sin (Shanghai) 2020; 52:168-179. [PMID: 32047913 DOI: 10.1093/abbs/gmz161] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Indexed: 12/13/2022] Open
Abstract
Y-box binding protein 1 (YB-1) is manifested as its involvement in cell proliferation and differentiation and malignant cell transformation. Overexpression of YB-1 is associated with glioma progression and patient survival. The aim of this study is to investigate the influence of YB-1 knockdown on glioma cell progression and reveal the mechanisms of YB-1 knockdown on glioma cell growth, migration, and apoptosis. It was found that the knockdown of YB-1 decreased the mRNA and protein levels of YB-1 in U251 glioma cells. The knockdown of YB-1 significantly inhibited cell proliferation, colony formation, and migration in vitro and tumor growth in vivo. Proteome and phosphoproteome data revealed that YB-1 is involved in glioma progression through regulating the expression and phosphorylation of major proteins involved in cell cycle, adhesion, and apoptosis. The main regulated proteins included CCNB1, CCNDBP1, CDK2, CDK3, ADGRG1, CDH-2, MMP14, AIFM1, HO-1, and BAX. Furthermore, it was also found that YB-1 knockdown is associated with the hypo-phosphorylation of ErbB, mTOR, HIF-1, cGMP-PKG, and insulin signaling pathways, and proteoglycans in cancer. Our findings indicated that YB-1 plays a key role in glioma progression in multiple ways, including regulating the expression and phosphorylation of major proteins associated with cell cycle, adhesion, and apoptosis.
Collapse
Affiliation(s)
- Huilin Gong
- Department of Pathology, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Shan Gao
- Department of Kidney Transplant, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Chenghuan Yu
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou 310013, China
| | - Meihe Li
- Department of Kidney Transplant, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Ping Liu
- Department of Dermatology, the Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Guanjun Zhang
- Department of Pathology, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Jinning Song
- Department of Neurosurgery, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Jin Zheng
- Department of Kidney Transplant, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| |
Collapse
|
32
|
Sun J, Zhang Y, Zhen Y, Cui J, Hu G, Lin Y. Antitumor activity of tetrandrine citrate in human glioma U87 cells in vitro and in vivo. Oncol Rep 2019; 42:2345-2354. [PMID: 31638254 PMCID: PMC6826321 DOI: 10.3892/or.2019.7372] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 09/11/2019] [Indexed: 12/18/2022] Open
Abstract
Since the current methods of treatment for malignant glioma, radiotherapy and chemotherapy, are unsatisfactory, the development of novel therapeutic compounds is required. In the present study, the inhibitory effect of tetrandrine citrate (TetC) on the proliferation of human glioma U87 cells, as well as its mechanism of action, were investigated. An MTT assay was used to assess cell viability in vitro, and the production of intracellular reactive oxygen species (ROS) was determined by assessing the fluorescence intensity of 2,7-dichlorofluorescein (DCF). Flow cytometry was used to determine the level of apoptosis and cell cycle status, and the protein expression levels of apoptosis-associated proteins were determined using western blotting. Additionally, the antitumor activity of TetC was assessed in vivo using a nude mouse xenograft model. The results revealed that in vitro, the proliferative rate of U87, U251 and human umbilical vein endothelial cells (HUVECs) was significantly reduced in a dose-dependent manner following treatment with TetC, although TetC had the greatest inhibitory effect on U87 cells. The vacuolization and apoptosis of U87 cells was induced using 10 and 20 µmol/l TetC, respectively. The overall proliferative inhibition was associated with an increase in the levels of ROS and apoptosis. In TetC-treated cells, the expression levels of apoptosis-related proteins, including cleaved (CL) caspase-3, Fas, phosphorylated (p)-p38 and p-JNK, were increased, whereas those of caspase-3 and Bcl-2 were decreased. In vivo, TetC was highly effective at inhibiting the growth of human glioma U87 ×enografts in BALB/c nude mice, with a percentage growth inhibition of ≥68.7%. These findings indicated that the potent antitumor activity of TetC may be mediated through an increase in ROS levels, the downregulation of Bcl-2, and the upregulation of CL caspase-3, Fas, p-p38 and p-JNK expression levels.
Collapse
Affiliation(s)
- Jingyu Sun
- Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing 100730, P.R. China
| | - Yang Zhang
- Peking University Fifth School of Clinical Medicine, Beijing 100730, P.R. China
| | - Yongzhan Zhen
- Tangshan Key Laboratory for Preclinical and Basic Research on Chronic Diseases, School of Basic Medical Sciences, North China University of Science and Technology, Tangshan 063000, P.R. China
| | - Ju Cui
- Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing 100730, P.R. China
| | - Gang Hu
- Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing 100730, P.R. China
| | - Yajun Lin
- Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing 100730, P.R. China
| |
Collapse
|
33
|
Cao G, Cui R, Liu C, Zhang G, Zhang Z. MTBHsp70-exFPR1-pulsed Dendritic Cells Enhance the Immune Response against Cervical Cancer. J Cancer 2019; 10:6364-6373. [PMID: 31772669 PMCID: PMC6856742 DOI: 10.7150/jca.29779] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 09/19/2019] [Indexed: 12/18/2022] Open
Abstract
Cervical cancer is the most common malignancy of the female reproductive system. Dendritic cell (DC)-based immunological therapy is a novel treatment for this cancer. DCs are specialized antigen-presenting cells (APCs) in the human immune system, and they can activate the T cells used in tumor immunological therapy. In this study, we developed a novel immunotherapeutic peptide by linking the Mycobacterium tuberculosis (MTB) heat shock protein 70 (Hsp70) functional peptide to the extracellular domain of FPR1, a protein overexpressed in cervical cancer, to obtain an MTBHsp70-exFPR1 fusion protein. Our experiments confirmed that the MTBHsp70-exFPR1 protein could promote DC maturation and induce the secretion of IL-12p70, IL-1β, and TNF-α. The antitumor effect of human cytotoxic T lymphocytes (CTLs) activated by autologous DCs was assessed in NOG mice. These results indicate that DCs pulsed with MTBHsp70-exFPR1 can enhance antitumor immunity against cervical cancer, providing a novel immune therapeutic strategy.
Collapse
Affiliation(s)
- Guangming Cao
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University
| | - Ran Cui
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University
| | - Chongdong Liu
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University
| | - Guyu Zhang
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University
| | - Zhenyu Zhang
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University
| |
Collapse
|
34
|
Drug-Loaded Microbubbles Combined with Ultrasound for Thrombolysis and Malignant Tumor Therapy. BIOMED RESEARCH INTERNATIONAL 2019; 2019:6792465. [PMID: 31662987 PMCID: PMC6791276 DOI: 10.1155/2019/6792465] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 07/22/2019] [Accepted: 09/14/2019] [Indexed: 12/14/2022]
Abstract
Cardiac-cerebral thrombosis and malignant tumor endanger the safety of human life seriously. Traditional chemotherapy drugs have side effects which restrict their applications. Drug-loaded microbubbles can be destroyed by ultrasound irradiation at the focus position and be used for thrombolysis and tumor therapy. Compared with traditional drug treatment, the drug-loaded microbubbles can be excited by ultrasound and release drugs to lesion sites, increasing the local drug concentration and the exposure dose to nonfocal regions, thus reducing the cytotoxicity and side effects of drugs. This article reviews the applications of drug-loaded microbubbles combined with ultrasound for thrombolysis and tumor therapy. We focus on highlighting the advantages of using this new technique for disease treatment and concluding with recommendations for future efforts on the applications of this technology.
Collapse
|
35
|
Riva M, Wouters R, Weerasekera A, Belderbos S, Nittner D, Thal DR, Baert T, Giovannoni R, Gsell W, Himmelreich U, Van Ranst M, Coosemans A. CT-2A neurospheres-derived high-grade glioma in mice: a new model to address tumor stem cells and immunosuppression. Biol Open 2019; 8:bio.044552. [PMID: 31511246 PMCID: PMC6777368 DOI: 10.1242/bio.044552] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Recently, several promising treatments for high-grade gliomas (HGGs) failed to provide significant benefit when translated from the preclinical setting to patients. Improving the animal models is fundamental to overcoming this translational gap. To address this need, we developed and comprehensively characterized a new in vivo model based on the orthotopic implantation of CT-2A cells cultured in neurospheres (NS/CT-2A). Murine CT-2A methylcholanthrene-induced HGG cells (C57BL/6 background) were cultured in monolayers (ML) or NS and orthotopically inoculated in syngeneic animals. ML/CT-2A and NS/CT-2A tumors' characterization included the analysis of tumor growth, immune microenvironment, glioma stem cells (GSCs), vascularization and metabolites. The immuno-modulating properties of NS/CT-2A and ML/CT-2A cells on splenocytes were tested in vitro. Mice harboring NS/CT-2A tumors had a shorter survival than those harboring ML/CT-2A tumors (P=0.0033). Compared to standard ML/CT-2A tumors, NS/CT-2A tumors showed more abundant GSCs (P=0.0002 and 0.0770 for Nestin and CD133, respectively) and regulatory T cells (Tregs, P=0.0074), and a strong tendency towards an increased vascularization (P=0.0503). There were no significant differences in metabolites' composition between NS/ and ML/CT-2A tumors. In vitro, NS were able to drive splenocytes towards a more immunosuppressive status by reducing CD8+ T cells (P=0.0354) and by promoting Tregs (P=0.0082), macrophages (MF, P=0.0019) and their M2 subset (P=0.0536). Compared to standard ML/CT-2A tumors, NS/CT-2A tumors show a more aggressive phenotype with increased immunosuppression and GSCs proliferation. Because of these specific features, the NS/CT-2A model represents a clinically relevant platform in the search for new HGG treatments aimed at reducing immunosuppression and eliminating GSCs. Summary: The NS/CT-2A tumor model represents a valuable research platform for the study of innovative treatments aimed at eliminating GSCs and reversing the tumor-induced immunosuppression in HGGs.
Collapse
Affiliation(s)
- Matteo Riva
- Department of Oncology, Laboratory of Tumor Immunology and Immunotherapy, KU Leuven, Leuven 3000, Belgium .,Department of Neurosurgery, Erasme Hospital, Bruxelles 1070, Belgium
| | - Roxanne Wouters
- Department of Oncology, Laboratory of Tumor Immunology and Immunotherapy, KU Leuven, Leuven 3000, Belgium
| | - Akila Weerasekera
- Biomedical MRI, Department of Imaging and Pathology and Molecular Small Animal Imaging Center (MoSAIC), KU Leuven, Leuven 3000, Belgium
| | - Sarah Belderbos
- Biomedical MRI, Department of Imaging and Pathology and Molecular Small Animal Imaging Center (MoSAIC), KU Leuven, Leuven 3000, Belgium
| | - David Nittner
- Center for the Biology of Disease, KU Leuven Center for Human Genetics - InfraMouse, VIB, University of Leuven, Leuven 3000, Belgium
| | - Dietmar R Thal
- Laboratory of Neuropathology, Department of Imaging and Pathology, Leuven Brain Institute, KU Leuven, Leuven 3000, Belgium.,Department of Pathology, UZ-Leuven, Leuven 3000, Belgium
| | - Thaïs Baert
- Department of Oncology, Laboratory of Tumor Immunology and Immunotherapy, KU Leuven, Leuven 3000, Belgium.,Department of Gynecology and Gynecologic Oncology, Kliniken Essen Mitte (KEM), Essen 2910, Germany
| | - Roberto Giovannoni
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza 20900, Italy
| | - Willy Gsell
- Biomedical MRI, Department of Imaging and Pathology and Molecular Small Animal Imaging Center (MoSAIC), KU Leuven, Leuven 3000, Belgium
| | - Uwe Himmelreich
- Biomedical MRI, Department of Imaging and Pathology and Molecular Small Animal Imaging Center (MoSAIC), KU Leuven, Leuven 3000, Belgium
| | - Marc Van Ranst
- Laboratory for Clinical and Epidemiological Virology, Rega Institute for Medical Research, KU Leuven, Leuven 3000, Belgium
| | - An Coosemans
- Department of Oncology, Laboratory of Tumor Immunology and Immunotherapy, KU Leuven, Leuven 3000, Belgium.,Department of Gynaecology and Obstetrics, Leuven Cancer Institute, UZ Leuven, Leuven 3000, Belgium
| |
Collapse
|
36
|
Puttemans J, Lahoutte T, D'Huyvetter M, Devoogdt N. Beyond the Barrier: Targeted Radionuclide Therapy in Brain Tumors and Metastases. Pharmaceutics 2019; 11:pharmaceutics11080376. [PMID: 31374991 PMCID: PMC6723032 DOI: 10.3390/pharmaceutics11080376] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 07/12/2019] [Accepted: 07/17/2019] [Indexed: 01/10/2023] Open
Abstract
Brain tumors are notoriously difficult to treat. The blood-brain barrier provides a sanctuary site where residual and metastatic cancer cells can evade most therapeutic modalities. The delicate nature of the brain further complicates the decision of eliminating as much tumorous tissue as possible while protecting healthy tissue. Despite recent advances in immunotherapy, radiotherapy and systemic treatments, prognosis of newly diagnosed patients remains dismal, and recurrence is still a universal problem. Several strategies are now under preclinical and clinical investigation to optimize delivery and maximize the cytotoxic potential of pharmaceuticals with regards to brain tumors. This review provides an overview of targeted radionuclide therapy approaches for the treatment of primary brain tumors and brain metastases, with an emphasis on biological targeting moieties that specifically target key biomarkers involved in cancer development.
Collapse
Affiliation(s)
- Janik Puttemans
- In Vivo Cellular and Molecular Imaging Lab, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium.
| | - Tony Lahoutte
- In Vivo Cellular and Molecular Imaging Lab, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
- Nuclear Medicine Department, UZ Brussel, Laarbeeklaan 101, 1090 Brussels, Belgium
| | - Matthias D'Huyvetter
- In Vivo Cellular and Molecular Imaging Lab, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Nick Devoogdt
- In Vivo Cellular and Molecular Imaging Lab, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
| |
Collapse
|
37
|
Lee TH, Sung WW, Chan L, Lee HL, Chen SL, Huang YH, Kwan AL. The Association between Mortality-to-Incidence Ratios and Health Expenditures in Brain and Nervous System Cancers. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:ijerph16152739. [PMID: 31370357 PMCID: PMC6696604 DOI: 10.3390/ijerph16152739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 07/18/2019] [Accepted: 07/27/2019] [Indexed: 12/24/2022]
Abstract
Mortality-to-incidence ratios (MIRs) are alternative parameters used to evaluate the prognosis of a disease. In addition, MIRs are associated with the ranking of health care systems and expenditures for certain types of cancer. However, a lack of association between MIRs and pancreatic cancer has been noted. Given the poor prognosis of brain and nervous system cancers, similar to pancreatic cancer, the relation of MIRs and health care disparities is worth investigating. We used the Spearman's rank correlation coefficient (CC) to analyze the correlation between the MIRs in brain and nervous system cancers and inter-country disparities, including expenditures on health and human development index. Interestingly, the MIRs in brain and nervous system cancers are associated with the human development index score (N = 157, CC = -0.394, p < 0.001), current health expenditure (CHE) per capita (N = 157, CC = -0.438, p < 0.001), and CHE as percentage of gross domestic product (N = 157, CC = -0.245, p = 0.002). In conclusion, the MIRs in the brain and nervous system cancer are significantly associated with health expenditures and human development index. However, their role as an indicator of health disparity warrants further investigation.
Collapse
Affiliation(s)
- Tsung-Han Lee
- Division of Neurosurgery, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Wen-Wei Sung
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
- Department of Urology, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| | - Lung Chan
- School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
| | - Hsiang-Lin Lee
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
- Department of Surgery, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| | - Sung-Lang Chen
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
- Department of Urology, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| | - Yu-Hui Huang
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan.
- School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan.
- Department of Physical Medicine & Rehabilitation, Chung Shan Medical University Hospital, Taichung 40201, Taiwan.
| | - Aij-Lie Kwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
- Department of Neurosurgery, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan.
| |
Collapse
|
38
|
Alghamri MS, Núñez FJ, Kamran N, Carney S, Altshuler D, Lowenstein PR, Castro MG. Functional characterization of tumor antigen-specific T-cells isolated from the tumor microenvironment of sleeping beauty induced murine glioma models. Methods Enzymol 2019; 631:91-106. [PMID: 31948569 PMCID: PMC7021207 DOI: 10.1016/bs.mie.2019.05.032] [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: 04/24/2024]
Abstract
Diffuse Gliomas represent 80% of brain tumors with an average survival of the most aggressive form glioblastoma (GBM) 15-22 months from the time of diagnosis. The current standard of care includes tumor resection, chemotherapy and radiation, nevertheless, the incidence of recurrence remains high and there is a critical need for developing new therapeutic strategies. T-cell mediated immunotherapy that triggers an anti-tumor T cell-mediated memory response is a promising approach since it will not only attack the primary tumor but also prevent recurrence. Multiple immunotherapeutic strategies against glioma are currently being tested in clinical trials. We have developed an immune-mediated gene therapy (Thymidine kinase plus Fms-like tyrosine kinase 3 ligand: TK/Flt3L) which induces a robust anti-tumor T cell response leading to tumor regression, long-term survival and immunological memory in GBM models. Efficacy of the anti-glioma T cell therapy is determined by anti-tumor specific effector T cells. Therefore, assessing effector T cell activation status and function are critical readouts for determining the effectiveness of the therapy. Here, we detail methodologies to evaluate tumor specific T-cell responses using a genetically engineered Sleeping Beauty transposase-mediated glioma model. We first describe the glioma model and the generation of neurospheres (NS) that express the surrogate antigen cOVA. Then, we describe functional assays to determine anti-tumor T-cell response.
Collapse
Affiliation(s)
- Mahmoud S Alghamri
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States; Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Felipe J Núñez
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States; Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Neha Kamran
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States; Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Stephen Carney
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States; Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - David Altshuler
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States; Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Pedro R Lowenstein
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States; Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Maria G Castro
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States; Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States.
| |
Collapse
|
39
|
Meel MH, Kaspers GJL, Hulleman E. Preclinical therapeutic targets in diffuse midline glioma. Drug Resist Updat 2019; 44:15-25. [PMID: 31202081 DOI: 10.1016/j.drup.2019.06.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 05/29/2019] [Accepted: 06/04/2019] [Indexed: 12/29/2022]
Abstract
Diffuse midline gliomas (DMG) are rapidly fatal tumors of the midbrain in children, characterized by a diffuse growing pattern and high levels of intrinsic resistance to therapy. The location of these tumors, residing behind the blood-brain barrier (BBB), and the limited knowledge about the biology of these tumors, has hindered the development of effective treatment strategies. However, the introduction of diagnostic biopsies and the implementation of autopsy protocols in several large centers world-wide has allowed for a detailed characterization of these rare tumors. This has resulted in the identification of novel therapeutic targets, as well as major advances in understanding the biology of DMG in relation to therapy resistance. We here provide an overview of the cellular pathways and tumor-specific aberrations that have been targeted in preclinical DMG research, and discuss the advantages and limitations of these therapeutic strategies in relation to therapy resistance and BBB-penetration. Therewith, we aim to provide researchers with a framework for successful preclinical therapy development.
Collapse
Affiliation(s)
- Michaël Hananja Meel
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Pediatric Oncology, Cancer Center Amsterdam, the Netherlands
| | - Gertjan J L Kaspers
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Pediatric Oncology, Cancer Center Amsterdam, the Netherlands
| | - Esther Hulleman
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Pediatric Oncology, Cancer Center Amsterdam, the Netherlands.
| |
Collapse
|
40
|
Mehrian-Shai R, Reichardt JKV, Harris CC, Toren A. The Gut-Brain Axis, Paving the Way to Brain Cancer. Trends Cancer 2019; 5:200-207. [PMID: 30961828 DOI: 10.1016/j.trecan.2019.02.008] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 02/12/2019] [Accepted: 02/15/2019] [Indexed: 12/15/2022]
Abstract
The gut-brain axis formed by blood and lymphatic vessels paves the way for microbiota to impact the brain. Bacterial populations in the gut are a good candidate for a nongenetic factor contributing substantively to brain tumor development and to the success of therapy. Specifically, suppression of the immune system and induction of inflammation by microbiota sustain proliferative signaling, limit cell death, and induce angiogenesis as well as invasiveness. In addition, altered microbial metabolites and their levels could stimulate cell proliferation. We propose here a novel gear model connecting these complex interdisciplinary fields. Our model may impact mechanistic studies of brain cancer and better treatment outcomes through precision oncology.
Collapse
Affiliation(s)
| | - Juergen K V Reichardt
- Australian Institute for Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Curtis C Harris
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Amos Toren
- Pediatric Hemato-Oncology, Sheba Medical Center, Ramat Gan, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
41
|
Dong B, Wang L, Nie S, Li X, Xiao Y, Yang L, Meng X, Zhao P, Cui C, Tu L, Lu W, Sun W, Yu Y. Anti-glioma effect of intracranial vaccination with tumor cell lysate plus flagellin in mice. Vaccine 2018; 36:8148-8157. [PMID: 30449633 DOI: 10.1016/j.vaccine.2018.04.053] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 02/19/2018] [Accepted: 04/19/2018] [Indexed: 02/06/2023]
Abstract
The adjuvant effects of flagellin on regulation of immune response have been proved; whether flagellin could assist tumor cell lysate (TCL) to enhance anti-glioma immunity remains to be investigated. This study tests a hypothesis that therapeuticly intracranial administration with flagellin plus TCL enhances the effects of specific immunotherapy on glioma in mice. In this study, GL261 cells were transferred into C57BL/6 mice and the GL261-bearing mice were subcutaneously or intracranially inoculated with flagellin plus TCL, flagellin, TCL or saline. Our results showed that prophylacticly subcutaneous administration with TCL and flagellin could induce potent cytotoxic T lymphocyte (CTL) and prolong the survival of GL261-bearing mice significantly, but therapeuticly subcutaneous administration failed to. However, therapeuticly intracranial administration of TCL plus flagellin could prolong the survival. Moreover, intracranial administration of flagellin could recruit CD4+ T cells and CD8+ T cells to brain tissues, induce proliferation of natural killer (NK) cells, CD4+ T cells and CD8+ T cells in peripheral blood mononuclear cells and induce to splenomegaly. The results suggested that flagellin could be acted as an efficient adjuvant for TCL based vaccine.
Collapse
Affiliation(s)
- Boqi Dong
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Liying Wang
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Shu Nie
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Xin Li
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Yue Xiao
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Lei Yang
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Xiuping Meng
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Peiyan Zhao
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Cuiyun Cui
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Liqun Tu
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Wenting Lu
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Wei Sun
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China.
| | - Yongli Yu
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China.
| |
Collapse
|
42
|
Abstract
OBJECTIVES To describe the immunotherapy approaches currently under investigation for the treatment of gliomas. To discuss the management of immune-related adverse effects. DATA SOURCES Published literature, clinical trials, and oncology association guidance documents. CONCLUSION There are numerous modalities of immune treatment currently being evaluated in patients with glioma, including peptide vaccines, dendritic cell vaccines, oncolytic viruses, CAR-T cells, and checkpoint inhibitor therapy. Immunotherapy utilizes new mechanisms of treatment that may lead us to the eradication of gliomas. IMPLICATIONS FOR NURSING PRACTICE Immunotherapy is a rapidly growing field in the treatment of gliomas. Oncology nurses are often involved in the safe administration of these therapies, as well as the identification and management of immune-related toxicities.
Collapse
|
43
|
Guan X, Hasan MN, Begum G, Kohanbash G, Carney KE, Pigott VM, Persson AI, Castro MG, Jia W, Sun D. Blockade of Na/H exchanger stimulates glioma tumor immunogenicity and enhances combinatorial TMZ and anti-PD-1 therapy. Cell Death Dis 2018; 9:1010. [PMID: 30262908 PMCID: PMC6160445 DOI: 10.1038/s41419-018-1062-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 08/21/2018] [Accepted: 09/10/2018] [Indexed: 12/28/2022]
Abstract
The weak immunogenicity of gliomas presents a barrier for effective immunotherapy. Na/H exchanger isoform 1 (NHE1) maintains alkaline intracellular pH (pHi) of glioma cells and acidic microenvironment. In addition, NHE1 is expressed in tumor-associated microglia and tumor-associated macrophages (TAMs) and involved in protumoral communications between glioma and TAMs. Therefore, we hypothesize that NHE1 plays a role in developing tumor resistance and immunosuppressive tumor microenvironment. In this study, we investigated the efficacy of pharmacological inhibition of NHE1 on combinatorial therapies. Here we show that temozolomide (TMZ) treatment stimulates NHE1 protein expression in two intracranial syngeneic mouse glioma models (SB28, GL26). Pharmacological inhibition of NHE1 potentiated the cytotoxic effects of TMZ, leading to reduced tumor growth and increased median survival of mice. Blockade of NHE1 stimulated proinflammatory activation of TAM and increased cytotoxic T cell infiltration into tumors. Combining TMZ, anti-PD-1 antibody treatment with NHE1 blockade significantly prolonged the median survival in the mouse glioma model. These results demonstrate that pharmacological inhibition of NHE1 protein presents a new strategy for potentiating TMZ-induced cytotoxicity and increasing tumor immunogenicity for immunotherapy to improve glioma therapy.
Collapse
Affiliation(s)
- Xiudong Guan
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
- Chinese National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Md Nabiul Hasan
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Gulnaz Begum
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Gary Kohanbash
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Karen E Carney
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Victoria M Pigott
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Anders I Persson
- Department of Neurology, University of California, San Francisco, CA, USA
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - Maria G Castro
- Department of Neurological Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Wang Jia
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
- Chinese National Clinical Research Center for Neurological Diseases, Beijing, China.
- Beijing Neurosurgical Institute, Beijing, China.
| | - Dandan Sun
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
44
|
Alphandéry E. Glioblastoma Treatments: An Account of Recent Industrial Developments. Front Pharmacol 2018; 9:879. [PMID: 30271342 PMCID: PMC6147115 DOI: 10.3389/fphar.2018.00879] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 07/20/2018] [Indexed: 12/28/2022] Open
Abstract
The different drugs and medical devices, which are commercialized or under industrial development for glioblastoma treatment, are reviewed. Their different modes of action are analyzed with a distinction being made between the effects of radiation, the targeting of specific parts of glioma cells, and immunotherapy. Most of them are still at a too early stage of development to firmly conclude about their efficacy. Optune, which triggers antitumor activity by blocking the mitosis of glioma cells under the application of an alternating electric field, seems to be the only recently developed therapy with some efficacy reported on a large number of GBM patients. The need for early GBM diagnosis is emphasized since it could enable the treatment of GBM tumors of small sizes, possibly easier to eradicate than larger tumors. Ways to improve clinical protocols by strengthening preclinical studies using of a broader range of different animal and tumor models are also underlined. Issues related with efficient drug delivery and crossing of blood brain barrier are discussed. Finally societal and economic aspects are described with a presentation of the orphan drug status that can accelerate the development of GBM therapies, patents protecting various GBM treatments, the different actors tackling GBM disease, the cost of GBM treatments, GBM market figures, and a financial analysis of the different companies involved in the development of GBM therapies.
Collapse
Affiliation(s)
- Edouard Alphandéry
- Institut de Minéralogie, de Physique des Matériaux et de Cosmochimie, UMR 7590 CNRS, Sorbonne Universités, UPMC, University Paris 06, Paris, France.,Nanobacterie SARL, Paris, France
| |
Collapse
|
45
|
Qian J, Luo F, Yang J, Liu J, Liu R, Wang L, Wang C, Deng Y, Lu Z, Wang Y, Lu M, Wang JY, Chu Y. TLR2 Promotes Glioma Immune Evasion by Downregulating MHC Class II Molecules in Microglia. Cancer Immunol Res 2018; 6:1220-1233. [PMID: 30131377 DOI: 10.1158/2326-6066.cir-18-0020] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 06/15/2018] [Accepted: 08/15/2018] [Indexed: 11/16/2022]
Abstract
Gliomas, the most common primary neoplasms in the brain, are notorious for their ability to evade the immune response. Despite microglial infiltration in gliomas, expression of MHC class II molecules in those microglia is compromised. Here, we report that Toll-like receptor 2 (TLR2) activation downregulated expression of MHC class II molecules in microglia in an orthotopic murine glioma model. TLR2-induced microglial impairment hindered the proliferation and activation of CD4+ T cells, which facilitated glioma immune evasion. TLR2-induced downregulation of MHC class II molecules was caused by suppression of the master regulator of MHC class II molecule transcription, Ciita TLR2 activation triggered downstream MAPK/ERK1/2 signaling and loss of histone H3 acetylation at Ciita promoters, which in turn inhibited Ciita expression. In glioblastoma tissues, various endogenous TLR2 ligands, including the heat shock proteins that are endogenous TLR2 ligands, were upregulated, a response that correlated with CIITA inhibition. Thus, TLR2 promotes glioma immune-system evasion. These results advance our understanding of microglia as antigen-presenting cells in the context of glioma. In the glioma tumor microenvironment, TLR2 activation of microglia induces downregulation of microglial MHC class II expression. Impaired MHC class II expression limits T-cell-dependent antitumor immunity. Cancer Immunol Res; 6(10); 1220-33. ©2018 AACR.
Collapse
Affiliation(s)
- Jiawen Qian
- Department of Immunology, School of Basic Medical Sciences, and Institute of Biomedical Sciences, Fudan University, Shanghai, P.R. China.,Biotherapy Research Center, Fudan University, Shanghai, P.R. China
| | - Feifei Luo
- Biotherapy Research Center, Fudan University, Shanghai, P.R. China.,Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, P.R. China
| | - Jiao Yang
- Department of Immunology, School of Basic Medical Sciences, and Institute of Biomedical Sciences, Fudan University, Shanghai, P.R. China.,Biotherapy Research Center, Fudan University, Shanghai, P.R. China
| | - Jun Liu
- Department of Immunology, School of Basic Medical Sciences, and Institute of Biomedical Sciences, Fudan University, Shanghai, P.R. China
| | - Ronghua Liu
- Department of Immunology, School of Basic Medical Sciences, and Institute of Biomedical Sciences, Fudan University, Shanghai, P.R. China
| | - Luman Wang
- Department of Immunology, School of Basic Medical Sciences, and Institute of Biomedical Sciences, Fudan University, Shanghai, P.R. China
| | - Chen Wang
- Department of Immunology, School of Basic Medical Sciences, and Institute of Biomedical Sciences, Fudan University, Shanghai, P.R. China.,Biotherapy Research Center, Fudan University, Shanghai, P.R. China
| | - Yuting Deng
- Department of Immunology, School of Basic Medical Sciences, and Institute of Biomedical Sciences, Fudan University, Shanghai, P.R. China.,Biotherapy Research Center, Fudan University, Shanghai, P.R. China
| | - Zhou Lu
- Department of Immunology, School of Basic Medical Sciences, and Institute of Biomedical Sciences, Fudan University, Shanghai, P.R. China
| | - Yuedi Wang
- Department of Immunology, School of Basic Medical Sciences, and Institute of Biomedical Sciences, Fudan University, Shanghai, P.R. China.,Biotherapy Research Center, Fudan University, Shanghai, P.R. China
| | - Mingfang Lu
- Department of Immunology, School of Basic Medical Sciences, and Institute of Biomedical Sciences, Fudan University, Shanghai, P.R. China
| | - Ji-Yang Wang
- Department of Immunology, School of Basic Medical Sciences, and Institute of Biomedical Sciences, Fudan University, Shanghai, P.R. China
| | - Yiwei Chu
- Department of Immunology, School of Basic Medical Sciences, and Institute of Biomedical Sciences, Fudan University, Shanghai, P.R. China. .,Biotherapy Research Center, Fudan University, Shanghai, P.R. China
| |
Collapse
|
46
|
González-Morales A, Zabaleta A, Guruceaga E, Alonso MM, García-Moure M, Fernández-Irigoyen J, Santamaría E. Spatial and temporal proteome dynamics of glioma cells during oncolytic adenovirus Delta-24-RGD infection. Oncotarget 2018; 9:31045-31065. [PMID: 30123426 PMCID: PMC6089549 DOI: 10.18632/oncotarget.25774] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 06/22/2018] [Indexed: 11/25/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and aggressive type of malignant glioma. Oncolytic adenoviruses are being modified to exploit the aberrant expression of proteins in tumor cells to increase the antiglioma efficacy. E1A mutant adenovirus Delta-24-RGD (DNX-2401) has shown a favorable toxicity profile and remarkable efficacy in a first-in-human phase I clinical trial. However, the comprehensive modulation of glioma metabolism in response to Delta-24-RGD infection is poorly understood. Integrating mass spectrometry based-quantitative proteomics, physical and functional interaction data, and biochemical approaches, we conducted a cell-wide study of cytosolic, nuclear, and secreted glioma proteomes throughout the early time course of Delta-24-RGD infection. In addition to the severe proteostasis impairment detected during the first hours post-infection (hpi), Delta-24-RGD induces a transient inhibition of signal transducer and activator of transcription 3 (STAT3), and transcription factor AP-1 (c-JUN) between 3 and 10hpi, increasing the nuclear factor kappa B (NF-κB) activity at 6hpi. Furthermore, Delta-24-RGD specifically modulates the activation dynamics of protein kinase C (PKC), extracellular signal-regulated kinase 1/2 (ERK1/2), and p38 mitogen-activated protein kinase (p38 MAPK) pathways early in infection. At extracellular level, Delta-24-RGD triggers a time -dependent dynamic production of multitasking cytokines, and chemotactic factors, suggesting potential pleiotropic effects on the immune system reactivation. Taken together, these data help us to understand the mechanisms used by Delta-24-RGD to exploit glioma proteome organization. Further mining of this proteomic resource may enable design and engineering complementary adenoviral based-vectors to increase the specificity and potency against glioma.
Collapse
Affiliation(s)
- Andrea González-Morales
- Clinical Neuroproteomics Group, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Irunlarrea, Pamplona, Spain.,IDISNA, Navarra Institute for Health Research, Pamplona, Spain.,Proteored-ISCIII, Proteomics Unit, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Irunlarrea, Pamplona, Spain
| | - Aintzane Zabaleta
- IDISNA, Navarra Institute for Health Research, Pamplona, Spain.,Oncohematology Area, University Hospital of Navarra, Center for Applied Medical Research, CIBERONC, Pamplona, Spain
| | - Elizabeth Guruceaga
- IDISNA, Navarra Institute for Health Research, Pamplona, Spain.,Bioinformatics Unit, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - Marta M Alonso
- IDISNA, Navarra Institute for Health Research, Pamplona, Spain.,Program in Solid Tumors and Biomarkers, Foundation for the Applied Medical Research, Pamplona, Spain.,Department of Pediatrics, University Hospital of Navarra, Pamplona, Spain
| | - Marc García-Moure
- IDISNA, Navarra Institute for Health Research, Pamplona, Spain.,Program in Solid Tumors and Biomarkers, Foundation for the Applied Medical Research, Pamplona, Spain.,Department of Pediatrics, University Hospital of Navarra, Pamplona, Spain
| | - Joaquín Fernández-Irigoyen
- Clinical Neuroproteomics Group, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Irunlarrea, Pamplona, Spain.,IDISNA, Navarra Institute for Health Research, Pamplona, Spain.,Proteored-ISCIII, Proteomics Unit, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Irunlarrea, Pamplona, Spain
| | - Enrique Santamaría
- Clinical Neuroproteomics Group, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Irunlarrea, Pamplona, Spain.,IDISNA, Navarra Institute for Health Research, Pamplona, Spain.,Proteored-ISCIII, Proteomics Unit, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Irunlarrea, Pamplona, Spain
| |
Collapse
|
47
|
Broestl L, Rubin JB, Dahiya S. Fetal microchimerism in human brain tumors. Brain Pathol 2018; 28:484-494. [PMID: 28921714 PMCID: PMC5884742 DOI: 10.1111/bpa.12557] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Accepted: 09/13/2017] [Indexed: 11/26/2022] Open
Abstract
Sex differences in cancer incidence and survival, including central nervous system tumors, are well documented. Multiple mechanisms contribute to sex differences in health and disease. Recently, the presence of fetal-in-maternal microchimeric cells has been shown to have prognostic significance in breast and colorectal cancers. The frequency and potential role of these cells has not been investigated in brain tumors. We therefore selected two common primary adult brain tumors for this purpose: meningioma, which is sex hormone responsive and has a higher incidence in women, and glioblastoma, which is sex hormone independent and occurs more commonly in men. Quantitative PCR was used to detect the presence of male DNA in tumor samples from women with a positive history of male pregnancy and a diagnosis of either glioblastoma or meningioma. Fluorescence in situ hybridization for the X and Y chromosomes was used to verify the existence of intact male cells within tumor tissue. Fetal microchimerism was found in approximately 80% of glioblastoma cases and 50% of meningioma cases. No correlations were identified between the presence of microchimerism and commonly used clinical or molecular diagnostic features of disease. The impact of fetal microchimeric cells should be evaluated prospectively.
Collapse
Affiliation(s)
- Lauren Broestl
- Department of PediatricsWashington University School of MedicineSt. LouisMO
| | - Joshua B. Rubin
- Department of PediatricsWashington University School of MedicineSt. LouisMO
- Department of NeuroscienceWashington University School of MedicineSt. LouisMO
| | - Sonika Dahiya
- Department of Pathology and ImmunologyWashington University School of MedicineSt. LouisMO
| |
Collapse
|
48
|
Checkpoint inhibitors as treatment for malignant gliomas: "A long way to the top". Cancer Treat Rev 2018; 69:121-131. [PMID: 29966936 DOI: 10.1016/j.ctrv.2018.06.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 06/20/2018] [Indexed: 01/10/2023]
Abstract
Glioblastoma is the most common and lethal malignant brain tumor in adults, with a very poor prognosis of less than two years despite surgical resection followed by radiotherapy and chemotherapy. To date, targeted agents and antiangiogenic therapy have failed to show survival benefits and novel treatment approaches are urgently needed. Immune checkpoint inhibitors have recently revolutionized the landscape of cancer immunotherapy achieving regulatory approvals for a number of other 'historically' resistant cancers. These exciting successes have generated great interest in investigating if these agents could be such effective also in brain tumors field. Moreover, the traditional dogma that considers the central nervous system (CNS) as an immune-privileged site lacking the potential for immunosurveillance has been challenged as it has become clear that the CNS is immunoactive. Critical barriers to an effective antitumor immunity in brain tumor patients are still represented by the peculiar CNS immunological milieu and the numerous systemic and local immunosuppressive forces exhibited by malignant gliomas to avoid immune recognition and cellular death. This review describes the current status of checkpoint modulation as treatment for malignant gliomas. We start illustrating the compelling molecular and immunological rationale, than we show striking preclinical evidence of activity and discuss available data from prospective clinical trials. Furthermore, we explore the role of predictive biomarkers of responsiveness to checkpoint blockade in the context of gliomas, along with the development of combinatorial and potentially synergistic approaches with other established anti-cancer treatments or complementary immunotherapeutic modalities.
Collapse
|
49
|
Chen YH, Zeng WJ, Wen ZP, Cheng Q, Chen XP. Under explored epigenetic modulators: role in glioma chemotherapy. Eur J Pharmacol 2018; 833:201-209. [PMID: 29864410 DOI: 10.1016/j.ejphar.2018.05.047] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Revised: 05/31/2018] [Accepted: 05/31/2018] [Indexed: 12/15/2022]
Abstract
Patients with somatic mutations of epigenetic regulators are characterized by aberrant chromatin modification patterns. Recent mechanistic studies pairing chemical tool compounds and deep-sequencing technology have greatly broadened our understanding of epigenetic regulation in glioma progression and underpinned alternative treatment of epigenetic inhibitors. However, the effect of most inhibitors is condition-dependent, and the overall results of clinical trials still have not been applied to patients. There is an intense need to develop more potent and specific compounds as well as identify the population who may achieve clinical benefits. Besides, combination therapy with conventional therapeutics is another alternative strategy. In this review, we summarize well-characterized chemical probes in glioma research and clinical translation. We also discuss the target population and combination of therapy regimens of various agents. In a holistic sense, we try to provide guidance for selecting targeted chemical probes and pave the way for personalized rational therapy.
Collapse
Affiliation(s)
- Yan-Hong Chen
- Department of Clinical pharmacology, Xiangya Hospital, Central South University, Changsha 410078, China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, China
| | - Wen-Jing Zeng
- Department of Clinical pharmacology, Xiangya Hospital, Central South University, Changsha 410078, China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, China
| | - Zhi-Peng Wen
- Department of Clinical pharmacology, Xiangya Hospital, Central South University, Changsha 410078, China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, China
| | - Quan Cheng
- Department of Clinical pharmacology, Xiangya Hospital, Central South University, Changsha 410078, China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, China
| | - Xiao-Ping Chen
- Department of Clinical pharmacology, Xiangya Hospital, Central South University, Changsha 410078, China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, China.
| |
Collapse
|
50
|
Guo X, Qiu W, Liu Q, Qian M, Wang S, Zhang Z, Gao X, Chen Z, Xue H, Li G. Immunosuppressive effects of hypoxia-induced glioma exosomes through myeloid-derived suppressor cells via the miR-10a/Rora and miR-21/Pten Pathways. Oncogene 2018; 37:4239-4259. [PMID: 29713056 DOI: 10.1038/s41388-018-0261-9] [Citation(s) in RCA: 207] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Revised: 02/16/2018] [Accepted: 03/21/2018] [Indexed: 12/22/2022]
Abstract
While immunosuppressive environments mediated by myeloid-derived suppressor cells (MDSCs) have been well documented in glioma patients, the mechanisms of MDSC development and activation have not been clearly defined. Here, we elucidated a role for glioma-derived exosomes (GDEs) in potentiating an MDSC pathway. We isolated normoxia-stimulated and hypoxia-stimulated GDEs and studied their MDSC induction abilities in vivo and in vitro. Analyses of spleen and bone marrow MDSC proportions (flow cytometry) and reactive oxygen species (ROS), arginase activity, nitric oxide (NO), T-cell proliferation and immunosuppressive cytokine (IL-10 and TGF-β, ELISA) levels were used to assess MDSC expansion and functional capacity. We also performed microRNA (miRNA) sequencing analysis of two types of GDEs to find miRNAs that potentially mediate the development and activation of MDSCs. GDE miRNA intracellular signaling in MDSCs was also studied. Hypoxia promoted the secretion of GDEs, and mouse MDSCs could uptake GDEs. Hypoxia-stimulated GDEs had a stronger ability to induce MDSCs than N-GDEs. The hypoxia-inducible expression of miR-10a and miR-21 in GDEs mediated GDE-induced MDSC expansion and activation by targeting RAR-related orphan receptor alpha (RORA) and phosphatase and tensin homolog (PTEN). Mice inoculated with miR-10a or miR-21 knockout glioma cells generated fewer MDSCs than those inoculated with normal glioma cells. These data elucidated a mechanism by which glioma cells influence the differentiation and activation of MDSCs via exosomes and demonstrated how local glioma hypoxia affects the entirety of tumor immune environments.
Collapse
Affiliation(s)
- Xiaofan Guo
- Brain Science Research Institute, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong Province, 250012, P.R. China
| | - Wei Qiu
- Brain Science Research Institute, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong Province, 250012, P.R. China
| | - Qinglin Liu
- Department of Neurosurgery, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Jinan, Shandong Province, 250012, P.R. China
| | - Mingyu Qian
- Brain Science Research Institute, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong Province, 250012, P.R. China
| | - Shaobo Wang
- Brain Science Research Institute, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong Province, 250012, P.R. China
| | - Zongpu Zhang
- Brain Science Research Institute, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong Province, 250012, P.R. China
| | - Xiao Gao
- Brain Science Research Institute, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong Province, 250012, P.R. China
| | - Zihang Chen
- Brain Science Research Institute, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong Province, 250012, P.R. China
| | - Hao Xue
- Brain Science Research Institute, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong Province, 250012, P.R. China. .,Department of Neurosurgery, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Jinan, Shandong Province, 250012, P.R. China.
| | - Gang Li
- Brain Science Research Institute, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong Province, 250012, P.R. China. .,Department of Neurosurgery, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Jinan, Shandong Province, 250012, P.R. China.
| |
Collapse
|