1
|
Wang Y, Yu Y, Yu J, Wang C, Wang Y, Fu R, Zhang C. The intersections between neuroscience and medulloblastoma. Cancer Lett 2025; 620:217660. [PMID: 40154912 DOI: 10.1016/j.canlet.2025.217660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/17/2025] [Accepted: 03/19/2025] [Indexed: 04/01/2025]
Abstract
Medulloblastoma (MB) represents the most common malignant central nervous system tumor in childhood. The nervous system plays a critical role in the progression of MB, with interactions between the nervous system and cancer significantly influencing oncogenesis, tumor growth, invasion, stemness, and metabolism. These interactions also regulate angiogenesis, metastatic dissemination, the tumor immune microenvironment, and drug resistance. Investigating the nervous system-MB axis holds promise for identifying diagnostic markers, prognostic biomarkers, and therapeutic targets. It also provides insights into the molecular mechanisms underlying MB and informs the development of novel therapeutic strategies. This review summarizes the latest advancements in understanding the interplay between the nervous system and MB, including the role of glial cells in MB and the potential of drug repurposing targeting nervous system components for MB treatment. These findings underscore promising diagnostic and therapeutic opportunities for MB management. Additionally, we outline future research directions in neurosciences that may pave the way for innovative therapeutic approaches and deepen our understanding of this complex disease.
Collapse
Affiliation(s)
- Yafei Wang
- Department of Pediatric Neurosurgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Ying Yu
- Department of Pediatric Neurosurgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Jiahua Yu
- Department of Pediatric Neurosurgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Cheng Wang
- Department of Pediatric Neurosurgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China; Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yunkun Wang
- Department of Pediatric Neurosurgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Runxi Fu
- Department of Pediatric Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China; Shanghai Institute for Pediatric Research, Shanghai, China
| | - Chenran Zhang
- Department of Pediatric Neurosurgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
2
|
Stockwell CA, Thang M, Kram DE, Satterlee AB, Hingtgen S. Therapeutic approaches for targeting the pediatric brain tumor microenvironment. Drug Deliv Transl Res 2025:10.1007/s13346-025-01839-3. [PMID: 40257744 DOI: 10.1007/s13346-025-01839-3] [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] [Accepted: 03/14/2025] [Indexed: 04/22/2025]
Abstract
Central nervous system (CNS) tumors are the most frequent solid malignant tumors in pediatric patients and are the leading cause of tumor-related death in children. Treatment for this heterogeneous group of tumors consists of various combinations of safe maximal surgical resection, chemotherapy, and radiation therapy which offer a cure for some children but often cause debilitating adverse late effects in others. While therapies targeting the tumor microenvironment (TME) like immune checkpoint inhibition (ICI) have been successful in treating some cancers, these therapies failed to exhibit treatment efficacy in the majority of pediatric brain tumors in the clinic. Importantly, the pediatric TME is unique and distinct from adult brain tumors and designing therapies to effectively target these tumors requires understanding the unique biology of pediatric brain tumors and the use of translational models that recapitulate the TME. Here we describe the TME of medulloblastoma (MB) and diffuse midline glioma (DMG), specifically diffuse intrinsic pontine glioma (DIPG), and further present the current drug delivery approaches and clinical administration routes targeting the TME in these tumors, including preclinical and clinical studies.
Collapse
Affiliation(s)
- Caroline A Stockwell
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Morrent Thang
- Neuroscience Program, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - David E Kram
- Division of Pediatric Hematology-Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Andrew B Satterlee
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Eshelman Innovation, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Shawn Hingtgen
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
3
|
Zhang J, Li S, Wang Y, Liu J, Liu Y, Gong X, Sun Y, Sun L, Li Z, Wang T, Du S, Wu W. Tumor-associated macrophages correlate with better outcome in SHH medulloblastoma. Front Oncol 2025; 15:1557313. [PMID: 40303994 PMCID: PMC12037381 DOI: 10.3389/fonc.2025.1557313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 03/20/2025] [Indexed: 05/02/2025] Open
Abstract
Objective Tumor-associated macrophages (TAMs) constitute a significant proportion of the immune cell population within brain tumors. The polarization of macrophages exerts an important influence on the tumor microenvironment (TME). Nevertheless, the specific role of TAMs in sonic hedgehog (SHH) medulloblastoma remains unclear. To investigate the polarization characteristics and effects of TAMs in SHH medulloblastoma, we evaluated the infiltration of M1 and M2 macrophages in SHH medulloblastoma tissues and analyzed the correlation between TAMs recruitment and the clinical outcome of SHH medulloblastoma patients. Methods We enrolled a total of 42 patients diagnosed with SHH medulloblastoma. Using multiple immunofluorescence staining on paraffin-embedded sections, we detected the activated phenotype (M1/M2) by monoclonal antibodies for CD68, HLA-DR and CD163. Subsequently, we analyzed the correlation between TAMs and clinical characteristics as well as prognostic factors. Results The median age of 42 patients (31 boys, 11 girls) was 5.3 years (range: 0.8-15.1 years). All patients had confirmed pathological types, including 4 cases of classic medulloblastoma (CMB), 33 cases of desmoplastic/nodular medulloblastoma (DNMB), 3 cases of medulloblastoma with extensive nodularity (MBEN), and 2 cases of large-cell/anaplastic medulloblastoma (LCA). Thirteen cases presented with metastasis at diagnosis, while twenty-nine cases were without metastasis. Four cases had high-risk genetic abnormalities. Different proportions of macrophages were found in the collected medulloblastoma tissues, and large amounts of CD68+HLA-DR+CD163+ cells were found. The study revealed that Mtotal (total macrophages) and Mmix (CD68+HLA-DR+CD163+ cells) were significantly higher in group of patients <5 years old (P < 0.05), and Mtotal in non-metastatic group were significantly higher than that in metastatic group (P = 0.043). M2 macrophages in CMB group were significantly higher than that in DNMB/MBEN group (P = 0.036), M1 macrophages were significantly higher in children without high-risk genetic abnormalities (P = 0.007). Five-year PFS was significantly poorer in patients ≥5 years old and metastatic group (P < 0.05). High Mtotal group had a better 5-year PFS (P = 0.000), whereas high M2 group had both better 5-year PFS and OS (P = 0.001, P = 0.001). Multivariate analysis showed that Mtotal and M2 macrophages were independent prognostic factors for 5-year PFS, and M2 macrophages were an independent prognostic factor for 5-year OS. Conclusion The increase in total macrophages and M2 macrophages predicts a better outcome of SHH medulloblastoma. TAMs especially M2 macrophages might be a therapeutic target for SHH medulloblastoma.
Collapse
Affiliation(s)
- Jin Zhang
- Department of Pediatrics, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Shuting Li
- Department of Pediatrics, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Yuan Wang
- Department of Pediatrics, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Jingjing Liu
- Department of Pediatrics, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Yan Liu
- Department of Pediatrics, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xiaojun Gong
- Department of Pediatrics, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Yanling Sun
- Department of Pediatrics, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Liming Sun
- Department of Pediatrics, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Zhigang Li
- Hematologic Disease Laboratory, Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology, National Key Discipline of Pediatrics (Capital Medical University), Key Laboratory of Major Disease in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Tianyou Wang
- Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology, National Key Discipline of Pediatrics (Capital Medical University), Key Laboratory of Major Disease in Children, Ministry of Education, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Shuxu Du
- Department of Pediatrics, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Wanshui Wu
- Department of Pediatrics, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
4
|
de Santis JO, de Sousa GR, Queiroz RGDP, Cândido MF, Almeida F, de Rezende CP, de Ruy PC, Arini GS, Coyle B, Wade P, Brassesco MS, Scrideli CA, Tone LG, Valera ET. Immunomodulatory role of exosome-derived content in pediatric medulloblastoma: a molecular subgroup perspective. Hum Cell 2025; 38:55. [PMID: 39960575 DOI: 10.1007/s13577-025-01181-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 01/07/2025] [Indexed: 02/20/2025]
Abstract
Medulloblastoma (MB) is the most common malignant brain tumor in children, comprising four distinct subgroups: wingless (WNT), sonic hedgehog (SHH), Group 3, and Group 4. MYC amplification and metastatic dissemination are challenges in clinical management, and tumor-associated macrophages (TAMs) play an essential role in these intricate molecular processes. However, the influence of immune cells in MB metastasis and MYC-amp is unclear. Secretion of extracellular vesicles (EVs) has emerged as a pivotal mediator facilitating communication within the tumor microenvironment, orchestrating coordinated responses among immune cells during tumor initiation, progression, and tumor dissemination. Here, we sought to elucidate the role of exosome-derived MBs in promoting specific patterns of TAM polarization across different molecular subgroups of MB cell lines. CIBERSORTx analysis using a single-cell RNA dataset revealed an increase in M0 macrophages and a decreased proportion of M2 macrophages in MB patients with tumor dissemination in the central nervous system (CNS). Cell-derived exosomes were found to secrete high levels of IL-4, IL-10, and TGF-β, indicative of a protumor M2-profile pattern. Moreover, EVs from SHH TP53-mutated, Group 3/4, and MYC-amplified MBs induced dissimilar patterns of TNF-α and/or IL-1β overexpression. This study demonstrates that exosomes from pediatric MBs promote a predominant M2-macrophage phenotype and Group 3, Group 4, SHH TP53-mutated, and MYC-amplified MBs induced a mixed M1/M2 response pattern. These findings shed light on the pivotal role of exosomes in modulating the immune response, potentially contributing to immune escape in this malignant neoplasm.
Collapse
Affiliation(s)
- Jessica Oliveira de Santis
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
| | - Graziella Ribeiro de Sousa
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, Universidade of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | | | - Marina Ferreira Cândido
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Fausto Almeida
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Caroline Patini de Rezende
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Patricia Cassia de Ruy
- Center for Genomic Medicine, Clinical Hospital of Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Gabriel Santos Arini
- Department of Cellular and Molecular Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Beth Coyle
- Children's Brain Tumour Research Centre, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - Philippa Wade
- Children's Brain Tumour Research Centre, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - María Sol Brassesco
- Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Carlos Alberto Scrideli
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Luiz Gonzaga Tone
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Elvis Terci Valera
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
5
|
Liu M, Ren Y, Zhou Z, Yang J, Shi X, Cai Y, Arreola AX, Luo W, Fung KM, Xu C, Nipp RD, Bronze MS, Zheng L, Li YP, Houchen CW, Zhang Y, Li M. The crosstalk between macrophages and cancer cells potentiates pancreatic cancer cachexia. Cancer Cell 2024; 42:885-903.e4. [PMID: 38608702 PMCID: PMC11162958 DOI: 10.1016/j.ccell.2024.03.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 12/18/2023] [Accepted: 03/15/2024] [Indexed: 04/14/2024]
Abstract
With limited treatment options, cachexia remains a major challenge for patients with cancer. Characterizing the interplay between tumor cells and the immune microenvironment may help identify potential therapeutic targets for cancer cachexia. Herein, we investigate the critical role of macrophages in potentiating pancreatic cancer induced muscle wasting via promoting TWEAK (TNF-like weak inducer of apoptosis) secretion from the tumor. Specifically, depletion of macrophages reverses muscle degradation induced by tumor cells. Macrophages induce non-autonomous secretion of TWEAK through CCL5/TRAF6/NF-κB pathway. TWEAK promotes muscle atrophy by activating MuRF1 initiated muscle remodeling. Notably, tumor cells recruit and reprogram macrophages via the CCL2/CCR2 axis and disrupting the interplay between macrophages and tumor cells attenuates muscle wasting. Collectively, this study identifies a feedforward loop between pancreatic cancer cells and macrophages, underlying the non-autonomous activation of TWEAK secretion from tumor cells thereby providing promising therapeutic targets for pancreatic cancer cachexia.
Collapse
Affiliation(s)
- Mingyang Liu
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Yu Ren
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Zhijun Zhou
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Jingxuan Yang
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Xiuhui Shi
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Yang Cai
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Alex X Arreola
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Wenyi Luo
- Department of Pathology, Yale School of Medicine, New Haven, CT 06519, USA
| | - Kar-Ming Fung
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Chao Xu
- Department of Biostatistics and Epidemiology, Hudson College of Public Health, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Ryan D Nipp
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Michael S Bronze
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Lei Zheng
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Yi-Ping Li
- Department of Integrative Biology & Pharmacology, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Courtney W Houchen
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Yuqing Zhang
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| | - Min Li
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| |
Collapse
|
6
|
Nellan A, Jackson S. Targeted therapy done right: Direct sonic hedgehog inhibition for sonic hedgehog medulloblastoma. Neuro Oncol 2024; 26:623-624. [PMID: 38290484 PMCID: PMC10995513 DOI: 10.1093/neuonc/noae018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Indexed: 02/01/2024] Open
Affiliation(s)
- Anandani Nellan
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
- Surgical Neurology Branch, National Institutes of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Sadhana Jackson
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
7
|
Yang H, Li M, Deng Y, Wen H, Luo M, Zhang W. Roles and interactions of tumor microenvironment components in medulloblastoma with implications for novel therapeutics. Genes Chromosomes Cancer 2024; 63:e23233. [PMID: 38607297 DOI: 10.1002/gcc.23233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 03/16/2024] [Indexed: 04/13/2024] Open
Abstract
Medulloblastomas, the most common malignant pediatric brain tumors, can be classified into the wingless, sonic hedgehog (SHH), group 3, and group 4 subgroups. Among them, the SHH subgroup with the TP53 mutation and group 3 generally present with the worst patient outcomes due to their high rates of recurrence and metastasis. A novel and effective treatment for refractory medulloblastomas is urgently needed. To date, the tumor microenvironment (TME) has been shown to influence tumor growth, recurrence, and metastasis through immunosuppression, angiogenesis, and chronic inflammation. Treatments targeting TME components have emerged as promising approaches to the treatment of solid tumors. In this review, we summarize progress in research on medulloblastoma microenvironment components and their interactions. We also discuss challenges and future research directions for TME-targeting medulloblastoma therapy.
Collapse
Affiliation(s)
- Hanjie Yang
- Department of Pediatric Neurosurgery, Neurosurgery Center, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Min Li
- Department of Pediatric Neurosurgery, Neurosurgery Center, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yuhao Deng
- Department of Pediatric Neurosurgery, Neurosurgery Center, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Huantao Wen
- Department of Pediatric Neurosurgery, Neurosurgery Center, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Minjie Luo
- Department of Pediatric Neurosurgery, Neurosurgery Center, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Wangming Zhang
- Department of Pediatric Neurosurgery, Neurosurgery Center, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
8
|
Yang S, Wang M, Hua Y, Li J, Zheng H, Cui M, Huang N, Liu Q, Liao Q. Advanced insights on tumor-associated macrophages revealed by single-cell RNA sequencing: The intratumor heterogeneity, functional phenotypes, and cellular interactions. Cancer Lett 2024; 584:216610. [PMID: 38244910 DOI: 10.1016/j.canlet.2024.216610] [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: 11/23/2022] [Revised: 11/28/2023] [Accepted: 12/18/2023] [Indexed: 01/22/2024]
Abstract
Single-cell RNA sequencing (scRNA-seq) is an emerging technology used for cellular transcriptome analysis. The application of scRNA-seq has led to profoundly advanced oncology research, continuously optimizing novel therapeutic strategies. Intratumor heterogeneity extensively consists of all tumor components, contributing to different tumor behaviors and treatment responses. Tumor-associated macrophages (TAMs), the core immune cells linking innate and adaptive immunity, play significant roles in tumor progression and resistance to therapies. Moreover, dynamic changes occur in TAM phenotypes and functions subject to the regulation of the tumor microenvironment. The heterogeneity of TAMs corresponding to the state of the tumor microenvironment has been comprehensively recognized using scRNA-seq. Herein, we reviewed recent research and summarized variations in TAM phenotypes and functions from a developmental perspective to better understand the significance of TAMs in the tumor microenvironment.
Collapse
Affiliation(s)
- Sen Yang
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science, and Peking Union Medical College, Beijing, 100730, China
| | - Mengyi Wang
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science, and Peking Union Medical College, Beijing, 100730, China
| | - Yuze Hua
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science, and Peking Union Medical College, Beijing, 100730, China
| | - Jiayi Li
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science, and Peking Union Medical College, Beijing, 100730, China
| | - Huaijin Zheng
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science, and Peking Union Medical College, Beijing, 100730, China
| | - Ming Cui
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science, and Peking Union Medical College, Beijing, 100730, China
| | - Nan Huang
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science, and Peking Union Medical College, Beijing, 100730, China
| | - Qiaofei Liu
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science, and Peking Union Medical College, Beijing, 100730, China.
| | - Quan Liao
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science, and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
9
|
Sheng H, Li H, Zeng H, Zhang B, Lu Y, Liu X, Xu Z, Zhang J, Zhang L. Heterogeneity and tumoral origin of medulloblastoma in the single-cell era. Oncogene 2024; 43:839-850. [PMID: 38355808 PMCID: PMC10942862 DOI: 10.1038/s41388-024-02967-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 02/05/2024] [Indexed: 02/16/2024]
Abstract
Medulloblastoma is one of the most common malignant pediatric brain tumors derived from posterior fossa. The current treatment includes maximal safe surgical resection, radiotherapy, whole cranio-spinal radiation and adjuvant with chemotherapy. However, it can only limitedly prolong the survival time with severe side effects and relapse. Defining the intratumoral heterogeneity, cellular origin and identifying the interaction network within tumor microenvironment are helpful for understanding the mechanisms of medulloblastoma tumorigenesis and relapse. Due to technological limitations, the mechanisms of cellular heterogeneity and tumor origin have not been fully understood. Recently, the emergence of single-cell technology has provided a powerful tool for achieving the goal of understanding the mechanisms of tumorigenesis. Several studies have demonstrated the intratumoral heterogeneity and tumor origin for each subtype of medulloblastoma utilizing the single-cell RNA-seq, which has not been uncovered before using conventional technologies. In this review, we present an overview of the current progress in understanding of cellular heterogeneity and tumor origin of medulloblastoma and discuss novel findings in the age of single-cell technologies.
Collapse
Affiliation(s)
- Hui Sheng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Haotai Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Han Zeng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Bin Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yu Lu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xixi Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhongwen Xu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jing Zhang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Liguo Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
10
|
du Chatinier A, Velilla IQ, Meel MH, Hoving EW, Hulleman E, Metselaar DS. Microglia in pediatric brain tumors: The missing link to successful immunotherapy. Cell Rep Med 2023; 4:101246. [PMID: 37924816 PMCID: PMC10694606 DOI: 10.1016/j.xcrm.2023.101246] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 08/10/2023] [Accepted: 09/26/2023] [Indexed: 11/06/2023]
Abstract
Brain tumors are the leading cause of cancer-related mortality in children. Despite the development of immunotherapeutic strategies for adult brain tumors, progress in pediatric neuro-oncology has been hindered by the complex and poorly understood nature of the brain's immune system during early development, a phase that is critical for the onset of many pediatric brain tumors. A defining characteristic of these tumors is the abundance of microglia, the resident immune cells of the central nervous system. In this review, we explore the concept of microglial diversity across brain regions and throughout development and discuss how their maturation stage may contribute to tumor growth in children. We also summarize the current knowledge on the roles of microglia in common pediatric brain tumor entities and provide examples of myeloid-based immunotherapeutic strategies. Our review underscores the importance of microglial plasticity in pediatric brain tumors and its significance for developing effective immunotherapeutic strategies.
Collapse
Affiliation(s)
- Aimée du Chatinier
- Department of Neuro-oncology, Princess Máxima Center for Paediatric Oncology, Heidelberglaan 25, 3584CS Utrecht, the Netherlands
| | - Irene Querol Velilla
- Department of Neuro-oncology, Princess Máxima Center for Paediatric Oncology, Heidelberglaan 25, 3584CS Utrecht, the Netherlands
| | - Michaël Hananja Meel
- Department of Neuro-oncology, Princess Máxima Center for Paediatric Oncology, Heidelberglaan 25, 3584CS Utrecht, the Netherlands
| | - Eelco Wieger Hoving
- Department of Neuro-oncology, Princess Máxima Center for Paediatric Oncology, Heidelberglaan 25, 3584CS Utrecht, the Netherlands
| | - Esther Hulleman
- Department of Neuro-oncology, Princess Máxima Center for Paediatric Oncology, Heidelberglaan 25, 3584CS Utrecht, the Netherlands
| | - Dennis Serge Metselaar
- Department of Neuro-oncology, Princess Máxima Center for Paediatric Oncology, Heidelberglaan 25, 3584CS Utrecht, the Netherlands.
| |
Collapse
|
11
|
Ji ZZ, Chan MKK, Chan ASW, Leung KT, Jiang X, To KF, Wu Y, Tang PMK. Tumour-associated macrophages: versatile players in the tumour microenvironment. Front Cell Dev Biol 2023; 11:1261749. [PMID: 37965573 PMCID: PMC10641386 DOI: 10.3389/fcell.2023.1261749] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 10/12/2023] [Indexed: 11/16/2023] Open
Abstract
Tumour-Associated Macrophages (TAMs) are one of the pivotal components of the tumour microenvironment. Their roles in the cancer immunity are complicated, both pro-tumour and anti-cancer activities are reported, including not only angiogenesis, extracellular matrix remodeling, immunosuppression, drug resistance but also phagocytosis and tumour regression. Interestingly, TAMs are highly dynamic and versatile in solid tumours. They show anti-cancer or pro-tumour activities, and interplay between the tumour microenvironment and cancer stem cells and under specific conditions. In addition to the classic M1/M2 phenotypes, a number of novel dedifferentiation phenomena of TAMs are discovered due to the advanced single-cell technology, e.g., macrophage-myofibroblast transition (MMT) and macrophage-neuron transition (MNT). More importantly, emerging information demonstrated the potential of TAMs on cancer immunotherapy, suggesting by the therapeutic efficiency of the checkpoint inhibitors and chimeric antigen receptor engineered cells based on macrophages. Here, we summarized the latest discoveries of TAMs from basic and translational research and discussed their clinical relevance and therapeutic potential for solid cancers.
Collapse
Affiliation(s)
- Zoey Zeyuan Ji
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Max Kam-Kwan Chan
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Alex Siu-Wing Chan
- Department of Applied Social Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Kam-Tong Leung
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Xiaohua Jiang
- Key Laboratory for Regenerative Medicine of the Ministry of Education of China, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Ka-Fai To
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Yi Wu
- MOE Key Laboratory of Environment and Genes Related to Diseases, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, China
| | - Patrick Ming-Kuen Tang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| |
Collapse
|
12
|
Pokrajac NT, Tokarew NJA, Gurdita A, Ortin-Martinez A, Wallace VA. Meningeal macrophages inhibit chemokine signaling in pre-tumor cells to suppress mouse medulloblastoma initiation. Dev Cell 2023; 58:2015-2031.e8. [PMID: 37774709 DOI: 10.1016/j.devcel.2023.08.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 07/10/2023] [Accepted: 08/30/2023] [Indexed: 10/01/2023]
Abstract
The microenvironment profoundly influences tumor initiation across numerous tissues but remains understudied in brain tumors. In the cerebellum, canonical Wnt signaling controlled by Norrin/Frizzled4 (Fzd4) activation in meningeal endothelial cells is a potent inhibitor of preneoplasia and tumor progression in mouse models of Sonic hedgehog medulloblastoma (Shh-MB). Single-cell transcriptome profiling and phenotyping of the meninges indicate that Norrin/Frizzled4 sustains the activation of meningeal macrophages (mMΦs), characterized by Lyve1 and CXCL4 expression, during the critical preneoplastic period. Depleting mMΦs during this period enhances preneoplasia and tumorigenesis, phenocopying the effects of Norrin loss. The anti-tumorigenic function of mMΦs is derived from the expression of CXCL4, which counters CXCL12/CXCR4 signaling in pre-tumor cells, thereby inhibiting cell-cycle progression and promoting migration away from the pre-tumor niche. These findings identify a pivotal role for mMΦs as key mediators in chemokine-regulated anti-cancer crosstalk between the stroma and pre-tumor cells in the control of MB initiation.
Collapse
Affiliation(s)
- Nenad T Pokrajac
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Toronto, ON M5T 2S8, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Nicholas J A Tokarew
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Toronto, ON M5T 2S8, Canada
| | - Akshay Gurdita
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Toronto, ON M5T 2S8, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Arturo Ortin-Martinez
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Toronto, ON M5T 2S8, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Valerie A Wallace
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Toronto, ON M5T 2S8, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Ophthalmology and Vision Sciences, University of Toronto, Toronto, ON M5T 3A9, Canada.
| |
Collapse
|
13
|
Hedberg J, Studebaker A, Smith L, Chen CY, Westfall JJ, Cam M, Gross A, Hernandez-Aguirre I, Martin A, Kim D, Dhital R, Kim Y, Roberts RD, Cripe TP, Mardis ER, Cassady KA, Leonard J, Miller KE. Oncolytic virus-driven immune remodeling revealed in mouse medulloblastomas at single cell resolution. Mol Ther Oncolytics 2023; 30:39-55. [PMID: 37583388 PMCID: PMC10424001 DOI: 10.1016/j.omto.2023.07.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 07/17/2023] [Indexed: 08/17/2023] Open
Abstract
Oncolytic viruses, modified for tumor-restricted infection, are a promising cancer immunotherapeutic, yet much remains to be understood about factors driving their activity and outcome in the tumor microenvironment. Here, we report that oncolytic herpes simplex virus C134, previously found to exert T cell-dependent efficacy in mouse models of glioblastoma, exerts T cell-independent efficacy in mouse models of medulloblastoma, indicating this oncolytic virus uses different mechanisms in different tumors. We investigated C134's behavior in mouse medulloblastomas, using single cell RNA sequencing to map C134-induced gene expression changes across cell types, timepoints, and medulloblastoma subgroup models at whole-transcriptome resolution. Our work details substantial oncolytic virus-induced transcriptional remodeling of medulloblastoma-infiltrating immune cells, 10 subpopulations of monocytes and macrophages collectively demonstrating M1-like responses to C134, and suggests C134 be investigated as a potential new therapy for medulloblastoma.
Collapse
Affiliation(s)
- Jack Hedberg
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43215, USA
- The Center for Childhood Cancer, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43215, USA
| | - Adam Studebaker
- The Center for Childhood Cancer, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43215, USA
| | - Luke Smith
- Department of Neurosurgery, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Chun-Yu Chen
- The Center for Childhood Cancer, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43215, USA
| | - Jesse J. Westfall
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43215, USA
| | - Maren Cam
- The Center for Childhood Cancer, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43215, USA
| | - Amy Gross
- The Center for Childhood Cancer, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43215, USA
| | - Ilse Hernandez-Aguirre
- The Center for Childhood Cancer, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43215, USA
| | - Alexia Martin
- The Center for Childhood Cancer, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43215, USA
| | - Doyeon Kim
- The Center for Childhood Cancer, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43215, USA
| | - Ravi Dhital
- The Center for Childhood Cancer, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43215, USA
| | - Yeaseul Kim
- The Center for Childhood Cancer, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43215, USA
| | - Ryan D. Roberts
- The Center for Childhood Cancer, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43215, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43210, USA
- Division of Hematology/Oncology/BMT, Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Timothy P. Cripe
- The Center for Childhood Cancer, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43215, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43210, USA
- Division of Hematology/Oncology/BMT, Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Elaine R. Mardis
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43215, USA
- Department of Neurosurgery, The Ohio State University College of Medicine, Columbus, OH 43210, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Kevin A. Cassady
- The Center for Childhood Cancer, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43215, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Jeffrey Leonard
- The Center for Childhood Cancer, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43215, USA
- Department of Neurosurgery, The Ohio State University College of Medicine, Columbus, OH 43210, USA
- Department of Neurosurgery, Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Katherine E. Miller
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43215, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| |
Collapse
|
14
|
Shapiro JA, Gaonkar KS, Spielman SJ, Savonen CL, Bethell CJ, Jin R, Rathi KS, Zhu Y, Egolf LE, Farrow BK, Miller DP, Yang Y, Koganti T, Noureen N, Koptyra MP, Duong N, Santi M, Kim J, Robins S, Storm PB, Mack SC, Lilly JV, Xie HM, Jain P, Raman P, Rood BR, Lulla RR, Nazarian J, Kraya AA, Vaksman Z, Heath AP, Kline C, Scolaro L, Viaene AN, Huang X, Way GP, Foltz SM, Zhang B, Poetsch AR, Mueller S, Ennis BM, Prados M, Diskin SJ, Zheng S, Guo Y, Kannan S, Waanders AJ, Margol AS, Kim MC, Hanson D, Van Kuren N, Wong J, Kaufman RS, Coleman N, Blackden C, Cole KA, Mason JL, Madsen PJ, Koschmann CJ, Stewart DR, Wafula E, Brown MA, Resnick AC, Greene CS, Rokita JL, Taroni JN, Children’s Brain Tumor Network, Pacific Pediatric Neuro-Oncology Consortium. OpenPBTA: The Open Pediatric Brain Tumor Atlas. CELL GENOMICS 2023; 3:100340. [PMID: 37492101 PMCID: PMC10363844 DOI: 10.1016/j.xgen.2023.100340] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 02/28/2023] [Accepted: 05/04/2023] [Indexed: 07/27/2023]
Abstract
Pediatric brain and spinal cancers are collectively the leading disease-related cause of death in children; thus, we urgently need curative therapeutic strategies for these tumors. To accelerate such discoveries, the Children's Brain Tumor Network (CBTN) and Pacific Pediatric Neuro-Oncology Consortium (PNOC) created a systematic process for tumor biobanking, model generation, and sequencing with immediate access to harmonized data. We leverage these data to establish OpenPBTA, an open collaborative project with over 40 scalable analysis modules that genomically characterize 1,074 pediatric brain tumors. Transcriptomic classification reveals universal TP53 dysregulation in mismatch repair-deficient hypermutant high-grade gliomas and TP53 loss as a significant marker for poor overall survival in ependymomas and H3 K28-mutant diffuse midline gliomas. Already being actively applied to other pediatric cancers and PNOC molecular tumor board decision-making, OpenPBTA is an invaluable resource to the pediatric oncology community.
Collapse
Affiliation(s)
- Joshua A. Shapiro
- Childhood Cancer Data Lab, Alex’s Lemonade Stand Foundation, Bala Cynwyd, PA 19004, USA
| | - Krutika S. Gaonkar
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Bioinformatics and Health Informatics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Stephanie J. Spielman
- Childhood Cancer Data Lab, Alex’s Lemonade Stand Foundation, Bala Cynwyd, PA 19004, USA
- Rowan University, Glassboro, NJ 08028, USA
| | - Candace L. Savonen
- Childhood Cancer Data Lab, Alex’s Lemonade Stand Foundation, Bala Cynwyd, PA 19004, USA
| | - Chante J. Bethell
- Childhood Cancer Data Lab, Alex’s Lemonade Stand Foundation, Bala Cynwyd, PA 19004, USA
| | - Run Jin
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Komal S. Rathi
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Bioinformatics and Health Informatics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Yuankun Zhu
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Laura E. Egolf
- Cell and Molecular Biology Graduate Group, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
- Division of Oncology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Bailey K. Farrow
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Daniel P. Miller
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Yang Yang
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL 60637, USA
| | - Tejaswi Koganti
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Nighat Noureen
- Greehey Children’s Cancer Research Institute, UT Health San Antonio, San Antonio, TX 78229, USA
| | - Mateusz P. Koptyra
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Nhat Duong
- Department of Bioinformatics and Health Informatics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Mariarita Santi
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Jung Kim
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD 20850, USA
| | - Shannon Robins
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Phillip B. Storm
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Stephen C. Mack
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Jena V. Lilly
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Hongbo M. Xie
- Department of Bioinformatics and Health Informatics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Payal Jain
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Pichai Raman
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Bioinformatics and Health Informatics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Brian R. Rood
- Children’s National Research Institute, Washington, DC 20012, USA
- George Washington University School of Medicine and Health Sciences, Washington, DC 20052, USA
| | - Rishi R. Lulla
- Division of Hematology/Oncology, Hasbro Children’s Hospital, Providence, RI 02903, USA
- Department of Pediatrics, The Warren Alpert School of Brown University, Providence, RI 02912, USA
| | - Javad Nazarian
- Children’s National Research Institute, Washington, DC 20012, USA
- George Washington University School of Medicine and Health Sciences, Washington, DC 20052, USA
- Department of Pediatrics, University of Zurich, Zurich, Switzerland
| | - Adam A. Kraya
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Zalman Vaksman
- Division of Oncology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Allison P. Heath
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Cassie Kline
- Division of Oncology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Laura Scolaro
- Division of Oncology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Angela N. Viaene
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Xiaoyan Huang
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Gregory P. Way
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Steven M. Foltz
- Childhood Cancer Data Lab, Alex’s Lemonade Stand Foundation, Bala Cynwyd, PA 19004, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bo Zhang
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Anna R. Poetsch
- Biotechnology Center, Technical University Dresden, Dresden, Germany
- National Center for Tumor Diseases, Dresden, Germany
| | - Sabine Mueller
- Department of Neurology, Neurosurgery and Pediatrics, University of California, San Francisco, San Francisco, CA 94115, USA
| | - Brian M. Ennis
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Michael Prados
- University of California, San Francisco, San Francisco, CA 94115, USA
| | - Sharon J. Diskin
- Division of Oncology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pediatrics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Siyuan Zheng
- Greehey Children’s Cancer Research Institute, UT Health San Antonio, San Antonio, TX 78229, USA
| | - Yiran Guo
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Shrivats Kannan
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Angela J. Waanders
- Division of Hematology, Oncology, Neuro-Oncology, and Stem Cell Transplant, Ann & Robert H Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Ashley S. Margol
- Division of Hematology and Oncology, Children’s Hospital of Los Angeles, Los Angeles, CA 90027, USA
- Department of Pediatrics, Keck School of Medicine of University of Southern California, Los Angeles, CA 90033, USA
| | - Meen Chul Kim
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Derek Hanson
- Hackensack Meridian School of Medicine, Nutley, NJ 07110, USA
- Hackensack University Medical Center, Hackensack, NJ 07601, USA
| | - Nicholas Van Kuren
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Jessica Wong
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Rebecca S. Kaufman
- Department of Bioinformatics and Health Informatics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of Oncology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Noel Coleman
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Christopher Blackden
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Kristina A. Cole
- Division of Oncology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pediatrics, University of Pennsylvania, Philadelphia, PA 19104, USA
- Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jennifer L. Mason
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Peter J. Madsen
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Carl J. Koschmann
- Department of Pediatrics, University of Michigan Health, Ann Arbor, MI 48105, USA
- Pediatric Hematology Oncology, Mott Children’s Hospital, Ann Arbor, MI 48109, USA
| | - Douglas R. Stewart
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD 20850, USA
| | - Eric Wafula
- Department of Bioinformatics and Health Informatics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Miguel A. Brown
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Adam C. Resnick
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Casey S. Greene
- Childhood Cancer Data Lab, Alex’s Lemonade Stand Foundation, Bala Cynwyd, PA 19004, USA
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO 80045, USA
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jo Lynne Rokita
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Bioinformatics and Health Informatics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Jaclyn N. Taroni
- Childhood Cancer Data Lab, Alex’s Lemonade Stand Foundation, Bala Cynwyd, PA 19004, USA
| | - Children’s Brain Tumor Network
- Childhood Cancer Data Lab, Alex’s Lemonade Stand Foundation, Bala Cynwyd, PA 19004, USA
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Bioinformatics and Health Informatics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Rowan University, Glassboro, NJ 08028, USA
- Cell and Molecular Biology Graduate Group, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
- Division of Oncology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL 60637, USA
- Greehey Children’s Cancer Research Institute, UT Health San Antonio, San Antonio, TX 78229, USA
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD 20850, USA
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Children’s National Research Institute, Washington, DC 20012, USA
- George Washington University School of Medicine and Health Sciences, Washington, DC 20052, USA
- Division of Hematology/Oncology, Hasbro Children’s Hospital, Providence, RI 02903, USA
- Department of Pediatrics, The Warren Alpert School of Brown University, Providence, RI 02912, USA
- Department of Pediatrics, University of Zurich, Zurich, Switzerland
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO 80045, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
- Biotechnology Center, Technical University Dresden, Dresden, Germany
- National Center for Tumor Diseases, Dresden, Germany
- Department of Neurology, Neurosurgery and Pediatrics, University of California, San Francisco, San Francisco, CA 94115, USA
- University of California, San Francisco, San Francisco, CA 94115, USA
- Department of Pediatrics, University of Pennsylvania, Philadelphia, PA 19104, USA
- Division of Hematology, Oncology, Neuro-Oncology, and Stem Cell Transplant, Ann & Robert H Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Division of Hematology and Oncology, Children’s Hospital of Los Angeles, Los Angeles, CA 90027, USA
- Department of Pediatrics, Keck School of Medicine of University of Southern California, Los Angeles, CA 90033, USA
- Hackensack Meridian School of Medicine, Nutley, NJ 07110, USA
- Hackensack University Medical Center, Hackensack, NJ 07601, USA
- Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pediatrics, University of Michigan Health, Ann Arbor, MI 48105, USA
- Pediatric Hematology Oncology, Mott Children’s Hospital, Ann Arbor, MI 48109, USA
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Pacific Pediatric Neuro-Oncology Consortium
- Childhood Cancer Data Lab, Alex’s Lemonade Stand Foundation, Bala Cynwyd, PA 19004, USA
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Bioinformatics and Health Informatics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Rowan University, Glassboro, NJ 08028, USA
- Cell and Molecular Biology Graduate Group, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
- Division of Oncology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL 60637, USA
- Greehey Children’s Cancer Research Institute, UT Health San Antonio, San Antonio, TX 78229, USA
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD 20850, USA
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Children’s National Research Institute, Washington, DC 20012, USA
- George Washington University School of Medicine and Health Sciences, Washington, DC 20052, USA
- Division of Hematology/Oncology, Hasbro Children’s Hospital, Providence, RI 02903, USA
- Department of Pediatrics, The Warren Alpert School of Brown University, Providence, RI 02912, USA
- Department of Pediatrics, University of Zurich, Zurich, Switzerland
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO 80045, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
- Biotechnology Center, Technical University Dresden, Dresden, Germany
- National Center for Tumor Diseases, Dresden, Germany
- Department of Neurology, Neurosurgery and Pediatrics, University of California, San Francisco, San Francisco, CA 94115, USA
- University of California, San Francisco, San Francisco, CA 94115, USA
- Department of Pediatrics, University of Pennsylvania, Philadelphia, PA 19104, USA
- Division of Hematology, Oncology, Neuro-Oncology, and Stem Cell Transplant, Ann & Robert H Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Division of Hematology and Oncology, Children’s Hospital of Los Angeles, Los Angeles, CA 90027, USA
- Department of Pediatrics, Keck School of Medicine of University of Southern California, Los Angeles, CA 90033, USA
- Hackensack Meridian School of Medicine, Nutley, NJ 07110, USA
- Hackensack University Medical Center, Hackensack, NJ 07601, USA
- Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pediatrics, University of Michigan Health, Ann Arbor, MI 48105, USA
- Pediatric Hematology Oncology, Mott Children’s Hospital, Ann Arbor, MI 48109, USA
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
15
|
Yao B, Delaidelli A, Vogel H, Sorensen PH. Pediatric Brain Tumours: Lessons from the Immune Microenvironment. Curr Oncol 2023; 30:5024-5046. [PMID: 37232837 PMCID: PMC10217418 DOI: 10.3390/curroncol30050379] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/01/2023] [Accepted: 05/10/2023] [Indexed: 05/27/2023] Open
Abstract
In spite of recent advances in tumour molecular subtyping, pediatric brain tumours (PBTs) remain the leading cause of cancer-related deaths in children. While some PBTs are treatable with favourable outcomes, recurrent and metastatic disease for certain types of PBTs remains challenging and is often fatal. Tumour immunotherapy has emerged as a hopeful avenue for the treatment of childhood tumours, and recent immunotherapy efforts have been directed towards PBTs. This strategy has the potential to combat otherwise incurable PBTs, while minimizing off-target effects and long-term sequelae. As the infiltration and activation states of immune cells, including tumour-infiltrating lymphocytes and tumour-associated macrophages, are key to shaping responses towards immunotherapy, this review explores the immune landscape of the developing brain and discusses the tumour immune microenvironments of common PBTs, with hopes of conferring insights that may inform future treatment design.
Collapse
Affiliation(s)
- Betty Yao
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada; (B.Y.)
| | - Alberto Delaidelli
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada; (B.Y.)
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Hannes Vogel
- Department of Pathology, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Poul H. Sorensen
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada; (B.Y.)
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| |
Collapse
|
16
|
Chen Z, Soni N, Pinero G, Giotti B, Eddins DJ, Lindblad KE, Ross JL, Puigdelloses Vallcorba M, Joshi T, Angione A, Thomason W, Keane A, Tsankova NM, Gutmann DH, Lira SA, Lujambio A, Ghosn EEB, Tsankov AM, Hambardzumyan D. Monocyte depletion enhances neutrophil influx and proneural to mesenchymal transition in glioblastoma. Nat Commun 2023; 14:1839. [PMID: 37012245 PMCID: PMC10070461 DOI: 10.1038/s41467-023-37361-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 03/13/2023] [Indexed: 04/05/2023] Open
Abstract
Myeloid cells comprise the majority of immune cells in tumors, contributing to tumor growth and therapeutic resistance. Incomplete understanding of myeloid cells response to tumor driver mutation and therapeutic intervention impedes effective therapeutic design. Here, by leveraging CRISPR/Cas9-based genome editing, we generate a mouse model that is deficient of all monocyte chemoattractant proteins. Using this strain, we effectively abolish monocyte infiltration in genetically engineered murine models of de novo glioblastoma (GBM) and hepatocellular carcinoma (HCC), which show differential enrichment patterns for monocytes and neutrophils. Eliminating monocyte chemoattraction in monocyte enriched PDGFB-driven GBM invokes a compensatory neutrophil influx, while having no effect on Nf1-silenced GBM model. Single-cell RNA sequencing reveals that intratumoral neutrophils promote proneural-to-mesenchymal transition and increase hypoxia in PDGFB-driven GBM. We further demonstrate neutrophil-derived TNF-a directly drives mesenchymal transition in PDGFB-driven primary GBM cells. Genetic or pharmacological inhibiting neutrophils in HCC or monocyte-deficient PDGFB-driven and Nf1-silenced GBM models extend the survival of tumor-bearing mice. Our findings demonstrate tumor-type and genotype dependent infiltration and function of monocytes and neutrophils and highlight the importance of targeting them simultaneously for cancer treatments.
Collapse
Affiliation(s)
- Zhihong Chen
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, 30322, USA.
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| | - Nishant Soni
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Gonzalo Pinero
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Bruno Giotti
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Devon J Eddins
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Department of Medicine, Lowance Center for Human Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Katherine E Lindblad
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Graduate School of Biomedical Sciences at Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - James L Ross
- Emory University Department of Microbiology and Immunology, Emory Vaccine Center, Atlanta, GA, 30322, USA
| | | | - Tanvi Joshi
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Angelo Angione
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Wes Thomason
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Aislinn Keane
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Nadejda M Tsankova
- Department of Pathology and Laboratory Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Sergio A Lira
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Amaia Lujambio
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Graduate School of Biomedical Sciences at Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Eliver E B Ghosn
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Department of Medicine, Lowance Center for Human Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Alexander M Tsankov
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Dolores Hambardzumyan
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, 30322, USA.
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, 30322, USA.
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
17
|
Tan S, Yang Y, Yang W, Han Y, Huang L, Yang R, Hu Z, Tao Y, Liu L, Li Y, Oyang L, Lin J, Peng Q, Jiang X, Xu X, Xia L, Peng M, Wu N, Tang Y, Cao D, Liao Q, Zhou Y. Exosomal cargos-mediated metabolic reprogramming in tumor microenvironment. J Exp Clin Cancer Res 2023; 42:59. [PMID: 36899389 PMCID: PMC9999652 DOI: 10.1186/s13046-023-02634-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 02/28/2023] [Indexed: 03/12/2023] Open
Abstract
Metabolic reprogramming is one of the hallmarks of cancer. As nutrients are scarce in the tumor microenvironment (TME), tumor cells adopt multiple metabolic adaptations to meet their growth requirements. Metabolic reprogramming is not only present in tumor cells, but exosomal cargos mediates intercellular communication between tumor cells and non-tumor cells in the TME, inducing metabolic remodeling to create an outpost of microvascular enrichment and immune escape. Here, we highlight the composition and characteristics of TME, meanwhile summarize the components of exosomal cargos and their corresponding sorting mode. Functionally, these exosomal cargos-mediated metabolic reprogramming improves the "soil" for tumor growth and metastasis. Moreover, we discuss the abnormal tumor metabolism targeted by exosomal cargos and its potential antitumor therapy. In conclusion, this review updates the current role of exosomal cargos in TME metabolic reprogramming and enriches the future application scenarios of exosomes.
Collapse
Affiliation(s)
- Shiming Tan
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Yiqing Yang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Wenjuan Yang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Yaqian Han
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Lisheng Huang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China.,University of South China, Hengyang, 421001, Hunan, China
| | - Ruiqian Yang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China.,University of South China, Hengyang, 421001, Hunan, China
| | - Zifan Hu
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China.,University of South China, Hengyang, 421001, Hunan, China
| | - Yi Tao
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China.,University of South China, Hengyang, 421001, Hunan, China
| | - Lin Liu
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Yun Li
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Linda Oyang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Jinguan Lin
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Qiu Peng
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Xianjie Jiang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Xuemeng Xu
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Longzheng Xia
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Mingjing Peng
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Nayiyuan Wu
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Yanyan Tang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Deliang Cao
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China.
| | - Qianjin Liao
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China. .,Hunan Key Laboratory of Translational Radiation Oncology, 283 Tongzipo Road, Changsha, 410013, Hunan, China.
| | - Yujuan Zhou
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China. .,Hunan Key Laboratory of Translational Radiation Oncology, 283 Tongzipo Road, Changsha, 410013, Hunan, China.
| |
Collapse
|
18
|
Gorini F, Miceli M, de Antonellis P, Amente S, Zollo M, Ferrucci V. Epigenetics and immune cells in medulloblastoma. Front Genet 2023; 14:1135404. [PMID: 36968588 PMCID: PMC10036437 DOI: 10.3389/fgene.2023.1135404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 02/27/2023] [Indexed: 03/12/2023] Open
Abstract
Medulloblastoma (MB) is a highly malignant childhood tumor of the cerebellum. Transcriptional and epigenetic signatures have classified MB into four molecular subgroups, further stratified into biologically different subtypes with distinct somatic copy-number aberrations, driver genes, epigenetic alterations, activated pathways, and clinical outcomes. The brain tumor microenvironment (BTME) is of importance to regulate a complex network of cells, including immune cells, involved in cancer progression in brain malignancies. MB was considered with a “cold” immunophenotype due to the low influx of immune cells across the blood brain barrier (BBB). Recently, this assumption has been reconsidered because of the identification of infiltrating immune cells showing immunosuppressive phenotypes in the BTME of MB tumors. Here, we are providing a comprehensive overview of the current status of epigenetics alterations occurring during cancer progression with a description of the genomic landscape of MB by focusing on immune cells within the BTME. We further describe how new immunotherapeutic approaches could influence concurring epigenetic mechanisms of the immunosuppressive cells in BTME. In conclusion, the modulation of these molecular genetic complexes in BTME during cancer progression might enhance the therapeutic benefit, thus firing new weapons to fight MB.
Collapse
Affiliation(s)
- Francesca Gorini
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples, Naples, Italy
| | - Marco Miceli
- CEINGE Biotecnologie Avanzate “Franco Salvatore”, Naples, Italy
| | - Pasqualino de Antonellis
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples, Naples, Italy
| | - Stefano Amente
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples, Naples, Italy
| | - Massimo Zollo
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples, Naples, Italy
- CEINGE Biotecnologie Avanzate “Franco Salvatore”, Naples, Italy
- DAI Medicina di Laboratorio e Trasfusionale, ‘AOU Federico II Policlinico, Naples, Italy
| | - Veronica Ferrucci
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples, Naples, Italy
- CEINGE Biotecnologie Avanzate “Franco Salvatore”, Naples, Italy
- *Correspondence: Veronica Ferrucci,
| |
Collapse
|
19
|
Guzman G, Pellot K, Reed MR, Rodriguez A. CAR T-cells to treat brain tumors. Brain Res Bull 2023; 196:76-98. [PMID: 36841424 DOI: 10.1016/j.brainresbull.2023.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 01/18/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023]
Abstract
Tremendous success using CAR T therapy in hematological malignancies has garnered significant interest in developing such treatments for solid tumors, including brain tumors. This success, however, has yet to be mirrored in solid organ neoplasms. CAR T function has shown limited efficacy against brain tumors due to several factors including the immunosuppressive tumor microenvironment, blood-brain barrier, and tumor-antigen heterogeneity. Despite these considerations, CAR T-cell therapy has the potential to be implemented as a treatment modality for brain tumors. Here, we review adult and pediatric brain tumors, including glioblastoma, diffuse midline gliomas, and medulloblastomas that continue to portend a grim prognosis. We describe insights gained from different preclinical models using CAR T therapy against various brain tumors and results gathered from ongoing clinical trials. Furthermore, we outline the challenges limiting CAR T therapy success against brain tumors and summarize advancements made to overcome these obstacles.
Collapse
Affiliation(s)
- Grace Guzman
- Department of Neurosurgery, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | | | - Megan R Reed
- Department of Neurosurgery, University of Arkansas for Medical Sciences, Little Rock, AR, United States; Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Analiz Rodriguez
- Department of Neurosurgery, University of Arkansas for Medical Sciences, Little Rock, AR, United States.
| |
Collapse
|
20
|
Myeloid cell heterogeneity in the tumor microenvironment and therapeutic implications for childhood central nervous system (CNS) tumors. J Neuroimmunol 2023; 374:578009. [PMID: 36508930 DOI: 10.1016/j.jneuroim.2022.578009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 11/07/2022] [Accepted: 11/30/2022] [Indexed: 12/08/2022]
Abstract
Central nervous system (CNS) tumors are the most common type of solid tumors in children and the leading cause of cancer deaths in ages 0-14. Recent advances in the field of tumor biology and immunology have underscored the disparate nature of these distinct CNS tumor types. In this review, we briefly introduce pediatric CNS tumors and discuss various components of the TME, with a particular focus on myeloid cells. Although most studies regarding myeloid cells have been done on adult CNS tumors and animal models, we discuss the role of myeloid cell heterogeneity in pediatric CNS tumors and describe how these cells may contribute to tumorigenesis and treatment response. In addition, we present studies within the last 5 years that highlight human CNS tumors, the utility of various murine CNS tumor models, and the latest multi-dimensional tools that can be leveraged to investigate myeloid cell infiltration in young adults and children diagnosed with select CNS tumors.
Collapse
|
21
|
SHH/GLI2-TGF-β1 feedback loop between cancer cells and tumor-associated macrophages maintains epithelial-mesenchymal transition and endoplasmic reticulum homeostasis in cholangiocarcinoma. Pharmacol Res 2023; 187:106564. [PMID: 36423790 DOI: 10.1016/j.phrs.2022.106564] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/31/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022]
Abstract
BACKGROUND Tumor-associated macrophages (TAMs) play a dual role in tumors. However, the factors which drive the function of TAMs in cholangiocarcinoma remain largely undefined. METHODS SHH signaling pathway and endoplasmic reticulum stress (ERS) indicators were detected in clinical tissues and cholangiocarcinoma cell lines. TAMs were co-cultured with cholangiocarcinoma cells under conditions of hypoxia/normoxia. Polarized TAMs were counted by flow cytometry, and TGF-β1 levels in cell supernatants were detected by ELISA. The effects of glioma-associated oncogene GLI2 on TAMs themselves and cholangiocarcinoma cells were examined by conducting interference and overexpression assays. RESULTS The SHH signaling pathway and ERS were both activated in tumor tissues or tumor cell lines under conditions of hypoxia. In co-culture experiments, the presence of cholangiocarcinoma cells increased the proportion of M2-polarized TAMs and the secretion of TGF-β1 by TAMs, while knockdown of SHH expression reversed those increases. Overexpression of GLI2 in TAMS or stimulation of TAMS with Hh-Ag1.5 increased their levels of TGF-β1 expression. Furthermore, under co-culture conditions, interference with GLI2 expression in TAMs reduced the tumor cell migration, invasion, and ER homeostasis induced by Hh-Ag1.5-pretreated TAMs. Under conditions of hypoxia, the presence of cholangiocarcinoma cells promoted the expression of GLI2 and TGF-β1 in Tams, and in turn, TAMs inhibited the apoptosis and promoted the migration and invasion of cholangiocarcinoma cells. In vivo, an injection of cholangiocarcinoma cells plus TAMs contributed to the growth, EMT, and ER homeostasis of tumor tissue, while an injection of TAMs with GLI2 knockdown had the opposite effects. CONCLUSION Cholangiocarcinoma cells regulated TAM polarization and TGF-β1 secretion via a paracrine SHH signaling pathway, and in turn, TAMs promoted the growth, EMT, and ER homeostasis of cholangiocarcinoma cells via TGF-β1.
Collapse
|
22
|
Borgenvik A, Holmberg KO, Bolin S, Zhao M, Savov V, Rosén G, Hutter S, Garancher A, Rahmanto AS, Bergström T, Olsen TK, Mainwaring OJ, Sattanino D, Verbaan AD, Rusert JM, Sundström A, Bravo MB, Dang Y, Wenz AS, Richardson S, Fotaki G, Hill RM, Dubuc AM, Kalushkova A, Remke M, Čančer M, Jernberg-Wiklund H, Giraud G, Chen X, Taylor MD, Sangfelt O, Clifford SC, Schüller U, Wechsler-Reya RJ, Weishaupt H, Swartling FJ. Dormant SOX9-Positive Cells Facilitate MYC-Driven Recurrence of Medulloblastoma. Cancer Res 2022; 82:4586-4603. [PMID: 36219398 PMCID: PMC9755969 DOI: 10.1158/0008-5472.can-22-2108] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 09/01/2022] [Accepted: 10/07/2022] [Indexed: 01/24/2023]
Abstract
Relapse is the leading cause of death in patients with medulloblastoma, the most common malignant pediatric brain tumor. A better understanding of the mechanisms underlying recurrence could lead to more effective therapies for targeting tumor relapses. Here, we observed that SOX9, a transcription factor and stem cell/glial fate marker, is limited to rare, quiescent cells in high-risk medulloblastoma with MYC amplification. In paired primary-recurrent patient samples, SOX9-positive cells accumulated in medulloblastoma relapses. SOX9 expression anti-correlated with MYC expression in murine and human medulloblastoma cells. However, SOX9-positive cells were plastic and could give rise to a MYC high state. To follow relapse at the single-cell level, an inducible dual Tet model of medulloblastoma was developed, in which MYC expression was redirected in vivo from treatment-sensitive bulk cells to dormant SOX9-positive cells using doxycycline treatment. SOX9 was essential for relapse initiation and depended on suppression of MYC activity to promote therapy resistance, epithelial-mesenchymal transition, and immune escape. p53 and DNA repair pathways were downregulated in recurrent tumors, whereas MGMT was upregulated. Recurrent tumor cells were found to be sensitive to treatment with an MGMT inhibitor and doxorubicin. These findings suggest that recurrence-specific targeting coupled with DNA repair inhibition comprises a potential therapeutic strategy in patients affected by medulloblastoma relapse. SIGNIFICANCE SOX9 facilitates therapy escape and recurrence in medulloblastoma via temporal inhibition of MYC/MYCN genes, revealing a strategy to specifically target SOX9-positive cells to prevent tumor relapse.
Collapse
Affiliation(s)
- Anna Borgenvik
- Department of Immunology, Genetics, and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Karl O. Holmberg
- Department of Immunology, Genetics, and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Sara Bolin
- Department of Immunology, Genetics, and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Miao Zhao
- Department of Immunology, Genetics, and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Vasil Savov
- Department of Immunology, Genetics, and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Gabriela Rosén
- Department of Immunology, Genetics, and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Sonja Hutter
- Department of Immunology, Genetics, and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Alexandra Garancher
- Tumor Initiation & Maintenance Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, San Diego, California
| | | | - Tobias Bergström
- Department of Immunology, Genetics, and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Thale Kristin Olsen
- Department of Immunology, Genetics, and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Oliver J. Mainwaring
- Department of Immunology, Genetics, and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Damiana Sattanino
- Department of Immunology, Genetics, and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Annemieke D. Verbaan
- Department of Immunology, Genetics, and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Jessica M. Rusert
- Tumor Initiation & Maintenance Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, San Diego, California
| | - Anders Sundström
- Department of Immunology, Genetics, and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Mar Ballester Bravo
- Department of Immunology, Genetics, and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Yonglong Dang
- Department of Immunology, Genetics, and Pathology, Science for Life Laboratory, Biomedical Centre, Uppsala University, Uppsala, Sweden
| | - Amelie S. Wenz
- Department of Immunology, Genetics, and Pathology, Science for Life Laboratory, Biomedical Centre, Uppsala University, Uppsala, Sweden
| | - Stacey Richardson
- Wolfson Childhood Cancer Research Centre, Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Newcastle upon Tyne, United Kingdom
| | - Grammatiki Fotaki
- Department of Immunology, Genetics, and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Rebecca M. Hill
- Wolfson Childhood Cancer Research Centre, Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Newcastle upon Tyne, United Kingdom
| | - Adrian M. Dubuc
- The Arthur and Sonia Labatt Brain Tumor Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Antonia Kalushkova
- Department of Immunology, Genetics, and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Marc Remke
- The Arthur and Sonia Labatt Brain Tumor Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Matko Čančer
- Department of Immunology, Genetics, and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Helena Jernberg-Wiklund
- Department of Immunology, Genetics, and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Géraldine Giraud
- Department of Immunology, Genetics, and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Xingqi Chen
- Department of Immunology, Genetics, and Pathology, Science for Life Laboratory, Biomedical Centre, Uppsala University, Uppsala, Sweden
| | - Michael D. Taylor
- The Arthur and Sonia Labatt Brain Tumor Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Olle Sangfelt
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Steven C. Clifford
- Wolfson Childhood Cancer Research Centre, Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Newcastle upon Tyne, United Kingdom
| | - Ulrich Schüller
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Paediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Research Institute Children's Cancer Center Hamburg, Hamburg, Germany
| | - Robert J. Wechsler-Reya
- Tumor Initiation & Maintenance Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, San Diego, California
| | - Holger Weishaupt
- Department of Immunology, Genetics, and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Fredrik J. Swartling
- Department of Immunology, Genetics, and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden.,Corresponding Author: Fredrik J. Swartling, Department of Immunology, Genetics and Pathology, Uppsala University, Rudbeck Laboratory, Uppsala 751 85, Sweden. E-mail:
| |
Collapse
|
23
|
van Bree NFHN, Wilhelm M. The Tumor Microenvironment of Medulloblastoma: An Intricate Multicellular Network with Therapeutic Potential. Cancers (Basel) 2022; 14:5009. [PMID: 36291792 PMCID: PMC9599673 DOI: 10.3390/cancers14205009] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/08/2022] [Accepted: 10/10/2022] [Indexed: 11/25/2022] Open
Abstract
Medulloblastoma (MB) is a heterogeneous disease in which survival is highly affected by the underlying subgroup-specific characteristics. Although the current treatment modalities have increased the overall survival rates of MB up to 70-80%, MB remains a major cause of cancer-related mortality among children. This indicates that novel therapeutic approaches against MB are needed. New promising treatment options comprise the targeting of cells and components of the tumor microenvironment (TME). The TME of MB consists of an intricate multicellular network of tumor cells, progenitor cells, astrocytes, neurons, supporting stromal cells, microglia, immune cells, extracellular matrix components, and vasculature systems. In this review, we will discuss all the different components of the MB TME and their role in MB initiation, progression, metastasis, and relapse. Additionally, we briefly introduce the effect that age plays on the TME of brain malignancies and discuss the MB subgroup-specific differences in TME components and how all of these variations could affect the progression of MB. Finally, we highlight the TME-directed treatments, in which we will focus on therapies that are being evaluated in clinical trials.
Collapse
Affiliation(s)
| | - Margareta Wilhelm
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institute, 17165 Stockholm, Sweden
| |
Collapse
|
24
|
Zhang J, Yuan X, Wang Y, Liu J, Li Z, Li S, Liu Y, Gong X, Sun Y, Wu W, Sun L, Du S, Wang T. Tumor-Associated Macrophages Correlate With Prognosis in Medulloblastoma. Front Oncol 2022; 12:893132. [PMID: 35860588 PMCID: PMC9289152 DOI: 10.3389/fonc.2022.893132] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 05/31/2022] [Indexed: 12/22/2022] Open
Abstract
Purpose Macrophage polarization plays an essential role in the tumor microenvironment of brain tumors. However, the role of tumor-associated macrophages (TAMs) in medulloblastoma still remains controversial. Thus, we investigated the distribution of macrophages in medulloblastoma tissues and analyzed the association of TAM recruitment and medulloblastoma patients’ outcomes. Methods We obtained a total of 71 paraffin sections from patients with medulloblastoma, and detected the activated phenotype (M1/M2) by monoclonal antibodies for CD68, HLA-DR and CD163 with multiple fluorescence immunohistochemistry method. The number of polarized macrophages was quantified using the InForm software. Outcomes were analyzed according to clinical data and quantified macrophage data. Results The study revealed that TAMs were significantly higher in sonic hedgehog (SHH) medulloblastoma than in other subgroups, and M1 macrophages in metastatic group were significantly higher than those in non-metastatic group. A Kaplan-Meier survival analysis and multivariate Cox regression model showed the correlation of high percentage of total macrophages (P = 0.038, HR = 0.241) and M1 macrophages (P = 0.034, HR = 0.333) with good 5-year progression-free survival (PFS); however, M2 macrophages had no correlation with survival of medulloblastoma patients (P> 0.05). Conclusion High percentage of total macrophages and M1 macrophages are correlated with good outcome of medulloblastoma patients. TAMs might be a target of therapy.
Collapse
Affiliation(s)
- Jin Zhang
- Department of Pediatrics, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology, National Key Discipline of Pediatrics (Capital Medical University), Key Laboratory of Major Disease in Children, Ministry of Education, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Xia Yuan
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Yuan Wang
- Department of Pediatrics, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Jingjing Liu
- Department of Pediatrics, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Zhigang Li
- Hematologic Disease Laboratory, Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology, National Key Discipline of Pediatrics (Capital Medical University), Key Laboratory of Major Disease in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Shuting Li
- Department of Pediatrics, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Yan Liu
- Department of Pediatrics, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xiaojun Gong
- Department of Pediatrics, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Yanling Sun
- Department of Pediatrics, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Wanshui Wu
- Department of Pediatrics, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Liming Sun
- Department of Pediatrics, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Shuxu Du
- Department of Pediatrics, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- *Correspondence: Tianyou Wang, ; Shuxu Du,
| | - Tianyou Wang
- Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology, National Key Discipline of Pediatrics (Capital Medical University), Key Laboratory of Major Disease in Children, Ministry of Education, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
- *Correspondence: Tianyou Wang, ; Shuxu Du,
| |
Collapse
|
25
|
Pediatric CNS cancer genomics and immunogenomics. Curr Opin Genet Dev 2022; 75:101918. [PMID: 35617766 DOI: 10.1016/j.gde.2022.101918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 04/18/2022] [Accepted: 04/21/2022] [Indexed: 11/20/2022]
Abstract
Large-scale genomic discovery in pediatric cancers established the importance of multiple platform-based characterizations of DNA and RNA to obtain a complete molecular landscape of these cancers, including actionable variants, diagnostic or prognostic evidence, and germline susceptibility. While these discoveries set the stage for pediatric cancer precision medicine, broad-based implementation has been quite slow compared with the adult-cancer precision medicine setting, due largely to the rarity of pediatric cancer. Here, we survey several large-cohort studies that utilize multiplex clinical characterization, including pediatric patients diagnosed with central nervous system (CNS) malignancies. The reported results demonstrate that molecularly guided precision therapeutics yield clinical benefit for these patients, establishing one important component needed for precision therapeutics to enter the pediatric CNS setting.
Collapse
|
26
|
Eisemann T, Wechsler-Reya RJ. Coming in from the cold: overcoming the hostile immune microenvironment of medulloblastoma. Genes Dev 2022; 36:514-532. [PMID: 35680424 PMCID: PMC9186392 DOI: 10.1101/gad.349538.122] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Medulloblastoma is an aggressive brain tumor that occurs predominantly in children. Despite intensive therapy, many patients die of the disease, and novel therapies are desperately needed. Although immunotherapy has shown promise in many cancers, the low mutational burden, limited infiltration of immune effector cells, and immune-suppressive microenvironment of medulloblastoma have led to the assumption that it is unlikely to respond to immunotherapy. However, emerging evidence is challenging this view. Here we review recent preclinical and clinical studies that have identified mechanisms of immune evasion in medulloblastoma, and highlight possible therapeutic interventions that may give new hope to medulloblastoma patients and their families.
Collapse
Affiliation(s)
- Tanja Eisemann
- Tumor Initiation and Maintenance Program, National Cancer Institute-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, USA
| | - Robert J Wechsler-Reya
- Tumor Initiation and Maintenance Program, National Cancer Institute-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, USA.,Department of Pediatrics, University of California at San Diego, La Jolla, California 92161, USA
| |
Collapse
|
27
|
Medulloblastoma: Immune microenvironment and targeted nano-therapy. OPENNANO 2022. [DOI: 10.1016/j.onano.2022.100035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
28
|
Abbas Z, George C, Ancliffe M, Howlett M, Jones AC, Kuchibhotla M, Wechsler-Reya RJ, Gottardo NG, Endersby R. Conventional Therapies Deplete Brain-Infiltrating Adaptive Immune Cells in a Mouse Model of Group 3 Medulloblastoma Implicating Myeloid Cells as Favorable Immunotherapy Targets. Front Immunol 2022; 13:837013. [PMID: 35309309 PMCID: PMC8928748 DOI: 10.3389/fimmu.2022.837013] [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: 12/16/2021] [Accepted: 02/08/2022] [Indexed: 11/13/2022] Open
Abstract
Medulloblastoma is the most common childhood brain cancer. Mainstay treatments of radiation and chemotherapy have not changed in decades and new treatment approaches are crucial for the improvement of clinical outcomes. To date, immunotherapies for medulloblastoma have been unsuccessful, and studies investigating the immune microenvironment of the disease and the impact of current therapies are limited. Preclinical models that recapitulate both the disease and immune environment are essential for understanding immune-tumor interactions and to aid the identification of new and effective immunotherapies. Using an immune-competent mouse model of aggressive Myc-driven medulloblastoma, we characterized the brain immune microenvironment and changes induced in response to craniospinal irradiation, or the medulloblastoma chemotherapies cyclophosphamide or gemcitabine. The role of adaptive immunity in disease progression and treatment response was delineated by comparing survival outcomes in wildtype C57Bl/6J and in mice deficient in Rag1 that lack mature T and B cells. We found medulloblastomas in wildtype and Rag1-deficient mice grew equally fast, and that craniospinal irradiation and chemotherapies extended survival equally in wildtype and Rag1-deficient mice, suggesting that tumor growth and treatment response is independent of T and B cells. Medulloblastomas were myeloid dominant, and in wildtype mice, craniospinal irradiation and cyclophosphamide depleted T and B cells in the brain. Gemcitabine treatment was found to minimally alter the immune populations in the brain, resulting only in a depletion of neutrophils. Intratumorally, we observed an abundance of Iba1+ macrophages, and we show that CD45high cells comprise the majority of immune cells within these medulloblastomas but found that existing markers are insufficient to clearly delineate resident microglia from infiltrating macrophages. Ultimately, brain resident and peripheral macrophages dominate the brain and tumor microenvironment and are not depleted by standard-of-care medulloblastoma therapies. These populations therefore present a favorable target for immunotherapy in combination with front-line treatments.
Collapse
Affiliation(s)
- Zahra Abbas
- Centre for Child Health Research, University of Western Australia, Perth, WA, Australia.,Brain Tumour Research Program, Telethon Kids Cancer Centre, Telethon Kids Institute, Perth, WA, Australia
| | - Courtney George
- Brain Tumour Research Program, Telethon Kids Cancer Centre, Telethon Kids Institute, Perth, WA, Australia.,School of Medical and Health Sciences, Edith Cowan University, Perth, WA, Australia
| | - Mathew Ancliffe
- Brain Tumour Research Program, Telethon Kids Cancer Centre, Telethon Kids Institute, Perth, WA, Australia.,School of Veterinary and Life Sciences, Murdoch University, Perth, WA, Australia
| | - Meegan Howlett
- Centre for Child Health Research, University of Western Australia, Perth, WA, Australia.,Brain Tumour Research Program, Telethon Kids Cancer Centre, Telethon Kids Institute, Perth, WA, Australia
| | - Anya C Jones
- Centre for Child Health Research, University of Western Australia, Perth, WA, Australia.,Cancer Centre Core Research, Telethon Kids Cancer Centre, Telethon Kids Institute, Perth, WA, Australia
| | - Mani Kuchibhotla
- Brain Tumour Research Program, Telethon Kids Cancer Centre, Telethon Kids Institute, Perth, WA, Australia
| | - Robert J Wechsler-Reya
- NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Nicholas G Gottardo
- Centre for Child Health Research, University of Western Australia, Perth, WA, Australia.,Brain Tumour Research Program, Telethon Kids Cancer Centre, Telethon Kids Institute, Perth, WA, Australia.,Department of Paediatric and Adolescent Oncology and Haematology, Perth Children's Hospital, Perth, WA, Australia
| | - Raelene Endersby
- Centre for Child Health Research, University of Western Australia, Perth, WA, Australia.,Brain Tumour Research Program, Telethon Kids Cancer Centre, Telethon Kids Institute, Perth, WA, Australia
| |
Collapse
|
29
|
Dang MT, Mafra F, Haldar M. Isolation of myeloid cells from mouse brain tumors for single-cell RNA-seq analysis. STAR Protoc 2021; 2:100957. [PMID: 34825218 PMCID: PMC8605103 DOI: 10.1016/j.xpro.2021.100957] [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] [Indexed: 11/17/2022] Open
Abstract
Current single-cell RNA sequencing (scRNA-seq) protocols are limited by the number of cells that can be simultaneously sequenced, restricting the ability to resolve heterogeneity of rare cell types. We describe here a protocol for rapid isolation of myeloid cells from tumor-harboring mouse cerebellum without cell sorting to minimize cell damage for scRNA-seq. This protocol includes the procedures for further enrichment of myeloid cells using CD11b+ magnetic beads, followed by the generation of scRNA library and sequencing analysis. For complete details on the use and execution of this protocol, please refer to Dang et al. (2021).
Collapse
Affiliation(s)
- Mai T. Dang
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of Pediatric and Developmental Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Fernanda Mafra
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Malay Haldar
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
30
|
Melcher V, Kerl K. The Growing Relevance of Immunoregulation in Pediatric Brain Tumors. Cancers (Basel) 2021; 13:5601. [PMID: 34830753 PMCID: PMC8615622 DOI: 10.3390/cancers13225601] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 11/05/2021] [Indexed: 12/19/2022] Open
Abstract
Pediatric brain tumors are genetically heterogeneous solid neoplasms. With a prevailing poor prognosis and widespread resistance to conventional multimodal therapy, these aggressive tumors are the leading cause of childhood cancer-related deaths worldwide. Advancement in molecular research revealed their unique genetic and epigenetic characteristics and paved the way for more defined prognostication and targeted therapeutic approaches. Furthermore, uncovering the intratumoral metrics on a single-cell level placed non-malignant cell populations such as innate immune cells into the context of tumor manifestation and progression. Targeting immune cells in pediatric brain tumors entails unique challenges but promising opportunities to improve outcome. Herein, we outline the current understanding of the role of the immune regulation in pediatric brain tumors.
Collapse
Affiliation(s)
- Viktoria Melcher
- Department of Pediatric Hematology and Oncology, University Children’s Hospital Münster, 48149 Münster, Germany
| | - Kornelius Kerl
- Department of Pediatric Hematology and Oncology, University Children’s Hospital Münster, 48149 Münster, Germany
| |
Collapse
|