1
|
van Bree N, Oppelt AS, Lindström S, Zhou L, Boutin L, Coyle B, Swartling FJ, Johnsen JI, Bräutigam L, Wilhelm M. Development of an orthotopic medulloblastoma zebrafish model for rapid drug testing. Neuro Oncol 2025; 27:779-794. [PMID: 39383211 PMCID: PMC11889719 DOI: 10.1093/neuonc/noae210] [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: 03/22/2024] [Indexed: 10/11/2024] Open
Abstract
BACKGROUND Medulloblastoma (MB) is one of the most common malignant brain tumors in children. Current preclinical in vivo model systems for MB have increased our understanding of molecular mechanisms regulating MB development. However, they may not be suitable for large-scale studies. The aim of this study was to investigate if a zebrafish-based xenograft model can recapitulate MB growth and enable rapid drug testing. METHODS Nine different MB cell lines or patient-derived cells were transplanted into blastula-stage zebrafish embryos. Tumor development and migration were then monitored using live imaging. RNA sequencing was performed to investigate transcriptome changes after conditioning cells in a neural stem cell-like medium. Furthermore, drug treatments were tested in a 96-well format. RESULTS We demonstrate here that transplantation of MB cells into the blastula stage of zebrafish embryos leads to orthotopic tumor growth that can be observed within 24 h after transplantation. Importantly, the homing of transplanted cells to the hindbrain region and the aggressiveness of tumor growth are enhanced by pre-culturing cells in a neural stem cell-like medium. The change in culture conditions rewires the transcriptome toward a more migratory and neuronal phenotype, including the expression of guidance molecules SEMA3A and EFNB1, both of which correlate with lower overall survival in MB patients. Furthermore, we highlight that the orthotopic zebrafish MB model has the potential to be used for rapid drug testing. CONCLUSIONS Blastula-stage zebrafish MB xenografts present an alternative to current MB mouse xenograft models, enabling quick evaluation of tumor cell growth, neurotropism, and drug efficacy.
Collapse
Affiliation(s)
- Niek van Bree
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Ann-Sophie Oppelt
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | | | - Leilei Zhou
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Lola Boutin
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Beth Coyle
- Children’s Brain Tumour Research Centre, School of Medicine, Biodiscovery Institute, University Park, University of Nottingham, Nottingham, UK
| | - Fredrik J Swartling
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - John Inge Johnsen
- Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
| | - Lars Bräutigam
- Comparative Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Margareta Wilhelm
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
2
|
Sigaud R, Brummer T, Kocher D, Milde T, Selt F. MOST wanted: navigating the MAPK-OIS-SASP-tumor microenvironment axis in primary pediatric low-grade glioma and preclinical models. Childs Nerv Syst 2024; 40:3209-3221. [PMID: 38789691 PMCID: PMC11511703 DOI: 10.1007/s00381-024-06463-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024]
Abstract
Understanding the molecular and cellular mechanisms driving pediatric low-grade glioma (pLGG)-the most prevalent brain tumor in children-is essential for the identification and evaluation of novel effective treatments. This review explores the intricate relationship between the mitogen-activated protein kinase (MAPK) pathway, oncogene-induced senescence (OIS), the senescence-associated secretory phenotype (SASP), and the tumor microenvironment (TME), integrating these elements into a unified framework termed the MAPK/OIS/SASP/TME (MOST) axis. This integrated approach seeks to deepen our understanding of pLGG and improve therapeutic interventions by examining the MOST axis' critical influence on tumor biology and response to treatment. In this review, we assess the axis' capacity to integrate various biological processes, highlighting new targets for pLGG treatment, and the need for characterized in vitro and in vivo preclinical models recapitulating pLGG's complexity to test targets. The review underscores the need for a comprehensive strategy in pLGG research, positioning the MOST axis as a pivotal approach in understanding pLGG. This comprehensive framework will open promising avenues for patient care and guide future research towards inventive treatment options.
Collapse
Affiliation(s)
- Romain Sigaud
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany.
- National Center for Tumor Diseases (NCT), Heidelberg, Germany.
| | - Tilman Brummer
- Institute, of Molecular Medicine and Cell Research (IMMZ), Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Centre for Biological Signaling Studies BIOSS, University of Freiburg and German Consortium for Translational Cancer Research (DKTK), Freiburg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Daniela Kocher
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Till Milde
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
- Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Florian Selt
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany.
- National Center for Tumor Diseases (NCT), Heidelberg, Germany.
- Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany.
| |
Collapse
|
3
|
Lee SC, Peterson C, Wang K, Alaali L, Eshleman J, Mahoney NR, Li E, Eberhart CG, Campbell AA. Establishment and Characterization of Three Human Ocular Adnexal Sebaceous Carcinoma Cell Lines. Int J Mol Sci 2024; 25:10183. [PMID: 39337668 PMCID: PMC11432008 DOI: 10.3390/ijms251810183] [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: 08/16/2024] [Revised: 09/14/2024] [Accepted: 09/21/2024] [Indexed: 09/30/2024] Open
Abstract
Ocular adnexal sebaceous carcinoma (SebCA) represents one of the most clinically problematic periocular tumors, often requiring aggressive surgical resection. The pathobiology of this tumor remains poorly understood, and few models exist that are suitable for preclinical testing. The aim of this study was to establish new cell lines to serve as models for pathobiological and drug testing. With patient consent, freshly resected tumor tissue was cultured using conditional reprogramming cell conditions. Standard techniques were used to characterize the cell lines in terms of overall growth, clonogenicity, apoptosis, and differentiation in vitro. Additional analyses including Western blotting, short tandem repeat (STR) profiling, and next-generation sequencing (NGS) were performed. Drug screening using mitomycin-C (MMC), 5-fluorouricil (5-FU), and 6-Diazo-5-oxo-L-norleucine (DON) were performed. JHH-SebCA01, JHH-SebCA02, and JHH-SebCA03 cell lines were established from two women and one man undergoing surgical resection of eyelid tumors. At passage 15, they each showed a doubling time of two to three days, and all could form colonies in anchorage-dependent conditions, but not in soft agar. The cells contained cytoplasmic vacuoles consistent with sebaceous differentiation, and adipophilin protein was present in all three lines. STR profiling confirmed that all lines were derived from their respective patients. NGS of the primary tumors and their matched cell lines identified numerous shared mutations, including alterations similar to those previously described in SebCA. Treatment with MMC or 5-FU resulted in dose-dependent growth inhibition and the induction of both apoptosis and differentiation. MYC protein was abundant in all three lines, and the glutamine metabolism inhibitor DON, previously shown to target high MYC tumors, slowed the growth of all our SebCA models. Ocular adnexal SebCA cell lines can be established using conditional reprogramming cell conditions, and our three new models are useful for testing therapies and interrogating the functional role of MYC and other possible molecular drivers. Current topical chemotherapies promote both apoptosis and differentiation in SebCA cells, and these tumors appear sensitive to inhibition or MYC-associated metabolic changes.
Collapse
Affiliation(s)
- Su-Chan Lee
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (S.-C.L.)
| | - Cornelia Peterson
- Department of Comparative Pathobiology, Tufts University Cummings School of Veterinary Medicine, North Grafton, MA 01536, USA;
| | - Kaixuan Wang
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Lujain Alaali
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (S.-C.L.)
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - James Eshleman
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (S.-C.L.)
| | - Nicholas R. Mahoney
- Department of Ophthalmology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Emily Li
- Department of Ophthalmology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Charles G. Eberhart
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (S.-C.L.)
- Department of Ophthalmology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ashley A. Campbell
- Department of Ophthalmology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
4
|
Miyagishima KJ, Qiao F, Stasheff SF, Nadal-Nicolás FM. Visual Deficits and Diagnostic and Therapeutic Strategies for Neurofibromatosis Type 1: Bridging Science and Patient-Centered Care. Vision (Basel) 2024; 8:31. [PMID: 38804352 PMCID: PMC11130890 DOI: 10.3390/vision8020031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 05/03/2024] [Accepted: 05/04/2024] [Indexed: 05/29/2024] Open
Abstract
Neurofibromatosis type 1 (NF1) is an inherited autosomal dominant disorder primarily affecting children and adolescents characterized by multisystemic clinical manifestations. Mutations in neurofibromin, the protein encoded by the Nf1 tumor suppressor gene, result in dysregulation of the RAS/MAPK pathway leading to uncontrolled cell growth and migration. Neurofibromin is highly expressed in several cell lineages including melanocytes, glial cells, neurons, and Schwann cells. Individuals with NF1 possess a genetic predisposition to central nervous system neoplasms, particularly gliomas affecting the visual pathway, known as optic pathway gliomas (OPGs). While OPGs are typically asymptomatic and benign, they can induce visual impairment in some patients. This review provides insight into the spectrum and visual outcomes of NF1, current diagnostic techniques and therapeutic interventions, and explores the influence of NF1-OPGS on visual abnormalities. We focus on recent advancements in preclinical animal models to elucidate the underlying mechanisms of NF1 pathology and therapies targeting NF1-OPGs. Overall, our review highlights the involvement of retinal ganglion cell dysfunction and degeneration in NF1 disease, and the need for further research to transform scientific laboratory discoveries to improved patient outcomes.
Collapse
Affiliation(s)
- Kiyoharu J. Miyagishima
- Retinal Neurophysiology Section, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA; (K.J.M.); (F.Q.); (S.F.S.)
| | - Fengyu Qiao
- Retinal Neurophysiology Section, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA; (K.J.M.); (F.Q.); (S.F.S.)
| | - Steven F. Stasheff
- Retinal Neurophysiology Section, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA; (K.J.M.); (F.Q.); (S.F.S.)
- Center for Neuroscience and Behavioral Medicine, Gilbert Neurofibromatosis Institute, Children’s National Health System, Washington, DC 20010, USA
- Neurology Department, George Washington University School of Medicine, Washington, DC 20037, USA
| | - Francisco M. Nadal-Nicolás
- Retinal Neurophysiology Section, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA; (K.J.M.); (F.Q.); (S.F.S.)
| |
Collapse
|
5
|
Roy D, Subramaniam B, Chong WC, Bornhorst M, Packer RJ, Nazarian J. Zebrafish-A Suitable Model for Rapid Translation of Effective Therapies for Pediatric Cancers. Cancers (Basel) 2024; 16:1361. [PMID: 38611039 PMCID: PMC11010887 DOI: 10.3390/cancers16071361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 03/27/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
Pediatric cancers are the leading cause of disease-related deaths in children and adolescents. Most of these tumors are difficult to treat and have poor overall survival. Concerns have also been raised about drug toxicity and long-term detrimental side effects of therapies. In this review, we discuss the advantages and unique attributes of zebrafish as pediatric cancer models and their importance in targeted drug discovery and toxicity assays. We have also placed a special focus on zebrafish models of pediatric brain cancers-the most common and difficult solid tumor to treat.
Collapse
Affiliation(s)
- Debasish Roy
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC 20012, USA; (D.R.)
| | - Bavani Subramaniam
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC 20012, USA; (D.R.)
| | - Wai Chin Chong
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC 20012, USA; (D.R.)
| | - Miriam Bornhorst
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC 20012, USA; (D.R.)
| | - Roger J. Packer
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC 20012, USA; (D.R.)
| | - Javad Nazarian
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC 20012, USA; (D.R.)
- DIPG/DMG Research Center Zurich, Children’s Research Center, Department of Pediatrics, University Children’s Hospital Zürich, 8032 Zurich, Switzerland
| |
Collapse
|
6
|
Furnari FB, Anastasaki C, Bian S, Fine HA, Koga T, Le LQ, Rodriguez FJ, Gutmann DH. Stem cell modeling of nervous system tumors. Dis Model Mech 2024; 17:dmm050533. [PMID: 38353122 PMCID: PMC10886724 DOI: 10.1242/dmm.050533] [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: 10/01/2023] [Accepted: 12/18/2023] [Indexed: 02/16/2024] Open
Abstract
Nervous system tumors, particularly brain tumors, represent the most common tumors in children and one of the most lethal tumors in adults. Despite decades of research, there are few effective therapies for these cancers. Although human nervous system tumor cells and genetically engineered mouse models have served as excellent platforms for drug discovery and preclinical testing, they have limitations with respect to accurately recapitulating important aspects of the pathobiology of spontaneously arising human tumors. For this reason, attention has turned to the deployment of human stem cell engineering involving human embryonic or induced pluripotent stem cells, in which genetic alterations associated with nervous system cancers can be introduced. These stem cells can be used to create self-assembling three-dimensional cerebral organoids that preserve key features of the developing human brain. Moreover, stem cell-engineered lines are amenable to xenotransplantation into mice as a platform to investigate the tumor cell of origin, discover cancer evolutionary trajectories and identify therapeutic vulnerabilities. In this article, we review the current state of human stem cell models of nervous system tumors, discuss their advantages and disadvantages, and provide consensus recommendations for future research.
Collapse
Affiliation(s)
- Frank B Furnari
- Department of Medicine, University of California, San Diego, San Diego, CA 92037, USA
| | - Corina Anastasaki
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Shan Bian
- Institute for Regenerative Medicine, School of Life Sciences and Technology, Tongji University, 200070 Shanghai, China
| | - Howard A Fine
- Department of Neurology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Tomoyuki Koga
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN 55455, USA
| | - Lu Q Le
- Department of Dermatology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Fausto J Rodriguez
- Division of Neuropathology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
7
|
Yvone GM, Breunig JJ. Pediatric low-grade glioma models: advances and ongoing challenges. Front Oncol 2024; 13:1346949. [PMID: 38318325 PMCID: PMC10839015 DOI: 10.3389/fonc.2023.1346949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 12/29/2023] [Indexed: 02/07/2024] Open
Abstract
Pediatric low-grade gliomas represent the most common childhood brain tumor class. While often curable, some tumors fail to respond and even successful treatments can have life-long side effects. Many clinical trials are underway for pediatric low-grade gliomas. However, these trials are expensive and challenging to organize due to the heterogeneity of patients and subtypes. Advances in sequencing technologies are helping to mitigate this by revealing the molecular landscapes of mutations in pediatric low-grade glioma. Functionalizing these mutations in the form of preclinical models is the next step in both understanding the disease mechanisms as well as for testing therapeutics. However, such models are often more difficult to generate due to their less proliferative nature, and the heterogeneity of tumor microenvironments, cell(s)-of-origin, and genetic alterations. In this review, we discuss the molecular and genetic alterations and the various preclinical models generated for the different types of pediatric low-grade gliomas. We examined the different preclinical models for pediatric low-grade gliomas, summarizing the scientific advances made to the field and therapeutic implications. We also discuss the advantages and limitations of the various models. This review highlights the importance of preclinical models for pediatric low-grade gliomas while noting the challenges and future directions of these models to improve therapeutic outcomes of pediatric low-grade gliomas.
Collapse
Affiliation(s)
- Griselda Metta Yvone
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Joshua J. Breunig
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Center for Neural Sciences in Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
8
|
Selt F, El Damaty A, Schuhmann MU, Sigaud R, Ecker J, Sievers P, Kocher D, Herold-Mende C, Oehme I, von Deimling A, Pfister SM, Sahm F, Jones DTW, Witt O, Milde T. Generation of patient-derived pediatric pilocytic astrocytoma in-vitro models using SV40 large T: evaluation of a modeling workflow. J Neurooncol 2023; 165:467-478. [PMID: 37999877 PMCID: PMC10752915 DOI: 10.1007/s11060-023-04500-6] [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: 09/27/2023] [Accepted: 11/02/2023] [Indexed: 11/25/2023]
Abstract
PURPOSE Although pediatric low-grade gliomas (pLGG) are the most common pediatric brain tumors, patient-derived cell lines reflecting pLGG biology in culture are scarce. This also applies to the most common pLGG subtype pilocytic astrocytoma (PA). Conventional cell culture approaches adapted from higher-grade tumors fail in PA due to oncogene-induced senescence (OIS) driving tumor cells into arrest. Here, we describe a PA modeling workflow using the Simian Virus large T antigen (SV40-TAg) to circumvent OIS. METHODS 18 pLGG tissue samples (17 (94%) histological and/or molecular diagnosis PA) were mechanically dissociated. Tumor cell positive-selection using A2B5 was perfomed in 8/18 (44%) cases. All primary cell suspensions were seeded in Neural Stem Cell Medium (NSM) and Astrocyte Basal Medium (ABM). Resulting short-term cultures were infected with SV40-TAg lentivirus. Detection of tumor specific alterations (BRAF-duplication and BRAF V600E-mutation) by digital droplet PCR (ddPCR) at defined time points allowed for determination of tumor cell fraction (TCF) and evaluation of the workflow. DNA-methylation profiling and gene-panel sequencing were used for molecular profiling of primary samples. RESULTS Primary cell suspensions had a mean TCF of 55% (+/- 23% (SD)). No sample in NSM (0/18) and ten samples in ABM (10/18) were successfully transduced. Three of these ten (30%) converted into long-term pLGG cell lines (TCF 100%), while TCF declined to 0% (outgrowth of microenvironmental cells) in 7/10 (70%) cultures. Young patient age was associated with successful model establishment. CONCLUSION A subset of primary PA cultures can be converted into long-term cell lines using SV40-TAg depending on sample intrinsic (patient age) and extrinsic workflow-related (e.g. type of medium, successful transduction) parameters. Careful monitoring of sample-intrinsic and extrinsic factors optimizes the process.
Collapse
Affiliation(s)
- Florian Selt
- Hopp Children's Cancer Center Heidelberg (KiTZ), Im Neuenheimer Feld 430, 69120, Heidelberg, Germany.
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany.
- KiTZ Clinical Trial Unit (ZIPO), Department of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany.
- National Center for Tumor Diseases (NCT), Heidelberg, Germany.
| | - Ahmed El Damaty
- Hopp Children's Cancer Center Heidelberg (KiTZ), Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
- Pediatric Neurosurgery Division, Department of Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Martin U Schuhmann
- Section of Pediatric Neurosurgery, Department of Neurosurgery, University Hospital Tübingen, Tübingen, Germany
| | - Romain Sigaud
- Hopp Children's Cancer Center Heidelberg (KiTZ), Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Jonas Ecker
- Hopp Children's Cancer Center Heidelberg (KiTZ), Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- KiTZ Clinical Trial Unit (ZIPO), Department of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Philipp Sievers
- Hopp Children's Cancer Center Heidelberg (KiTZ), Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
- Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Daniela Kocher
- Hopp Children's Cancer Center Heidelberg (KiTZ), Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | | | - Ina Oehme
- Hopp Children's Cancer Center Heidelberg (KiTZ), Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Andreas von Deimling
- Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefan M Pfister
- Hopp Children's Cancer Center Heidelberg (KiTZ), Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
- KiTZ Clinical Trial Unit (ZIPO), Department of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
| | - Felix Sahm
- Hopp Children's Cancer Center Heidelberg (KiTZ), Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
- Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - David T W Jones
- Hopp Children's Cancer Center Heidelberg (KiTZ), Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
- Division of Pediatric Glioma Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Olaf Witt
- Hopp Children's Cancer Center Heidelberg (KiTZ), Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- KiTZ Clinical Trial Unit (ZIPO), Department of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Till Milde
- Hopp Children's Cancer Center Heidelberg (KiTZ), Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- KiTZ Clinical Trial Unit (ZIPO), Department of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
| |
Collapse
|
9
|
Milde T, Fangusaro J, Fisher MJ, Hawkins C, Rodriguez FJ, Tabori U, Witt O, Zhu Y, Gutmann DH. Optimizing preclinical pediatric low-grade glioma models for meaningful clinical translation. Neuro Oncol 2023; 25:1920-1931. [PMID: 37738646 PMCID: PMC10628935 DOI: 10.1093/neuonc/noad125] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/24/2023] Open
Abstract
Pediatric low-grade gliomas (pLGGs) are the most common brain tumor in young children. While they are typically associated with good overall survival, children with these central nervous system tumors often experience chronic tumor- and therapy-related morbidities. Moreover, individuals with unresectable tumors frequently have multiple recurrences and persistent neurological symptoms. Deep molecular analyses of pLGGs reveal that they are caused by genetic alterations that converge on a single mitogenic pathway (MEK/ERK), but their growth is heavily influenced by nonneoplastic cells (neurons, T cells, microglia) in their local microenvironment. The interplay between neoplastic cell MEK/ERK pathway activation and stromal cell support necessitates the use of predictive preclinical models to identify the most promising drug candidates for clinical evaluation. As part of a series of white papers focused on pLGGs, we discuss the current status of preclinical pLGG modeling, with the goal of improving clinical translation for children with these common brain tumors.
Collapse
Affiliation(s)
- Till Milde
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- KiTZ Clinical Trial Unit (ZIPO), Department of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Jason Fangusaro
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Michael J Fisher
- Division of Oncology, Children’s Hospital of Philadelphia, Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Cynthia Hawkins
- Department of Laboratory Medicine and Pathobiology, Hospital for Sick Children, Toronto, Canada
| | - Fausto J Rodriguez
- Department of Pathology, University of California Los Angeles, Los Angeles, California, USA
| | - Uri Tabori
- Department of Medical Biophysics, Institute of Medical Science and Paediatrics, University of Toronto, Toronto, Canada
| | - Olaf Witt
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- KiTZ Clinical Trial Unit (ZIPO), Department of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Yuan Zhu
- Gilbert Family Neurofibromatosis Institute Center for Cancer and Immunology Research, Children’s National Hospital, Washington, DC, USA
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
10
|
Tang-Schomer MD, Bookland MJ, Sargent JE, N Jackvony T. Human Patient-Derived Brain Tumor Models to Recapitulate Ependymoma Tumor Vasculature. Bioengineering (Basel) 2023; 10:840. [PMID: 37508868 PMCID: PMC10376907 DOI: 10.3390/bioengineering10070840] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/25/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
Despite in vivo malignancy, ependymoma lacks cell culture models, thus limiting therapy development. Here, we used a tunable three-dimensional (3D) culture system to approximate the ependymoma microenvironment for recapitulating a patient's tumor in vitro. Our data showed that the inclusion of VEGF in serum-free, mixed neural and endothelial cell culture media supported the in vitro growth of all four ependymoma patient samples. The growth was driven by Nestin and Ki67 double-positive cells in a putative cancer stem cell niche, which was manifested as rosette-looking clusters in 2D and spheroids in 3D. The effects of extracellular matrix (ECM) such as collagen or Matrigel superseded that of the media conditions, with Matrigel resulting in the greater enrichment of Nestin-positive cells. When mixed with endothelial cells, the 3D co-culture models developed capillary networks resembling the in vivo ependymoma vasculature. The transcriptomic analysis of two patient cases demonstrated the separation of in vitro cultures by individual patients, with one patient's culture samples closely clustered with the primary tumor tissue. While VEGF was found to be necessary for preserving the transcriptomic features of in vitro cultures, the presence of endothelial cells shifted the gene's expression patterns, especially genes associated with ECM remodeling. The homeobox genes were mostly affected in the 3D in vitro models compared to the primary tumor tissue and between different 3D formats. These findings provide a basis for understanding the ependymoma microenvironment and enabling the further development of patient-derived in vitro ependymoma models for personalized medicine.
Collapse
Affiliation(s)
- Min D Tang-Schomer
- UConn Health, Department of Pediatrics, 263 Farmington Avenue, Farmington, CT 06030, USA
| | - Markus J Bookland
- Connecticut Children's Medical Center, 282 Washington St., Hartford, CT 06106, USA
| | - Jack E Sargent
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06030, USA
| | - Taylor N Jackvony
- UConn Health, Department of Pediatrics, 263 Farmington Avenue, Farmington, CT 06030, USA
| |
Collapse
|
11
|
Eberhart C. Killing zombies: Senolytic therapy in pilocytic astrocytoma. Neuro Oncol 2023; 25:748-749. [PMID: 36702510 PMCID: PMC10076928 DOI: 10.1093/neuonc/noad016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Indexed: 01/28/2023] Open
Affiliation(s)
- Charles G Eberhart
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| |
Collapse
|
12
|
Selt F, Sigaud R, Valinciute G, Sievers P, Zaman J, Alcon C, Schmid S, Peterziel H, Tsai JW, Guiho R, Martínez-Barbera JP, Pusch S, Deng J, Zhai Y, van Tilburg CM, Schuhman MU, El Damaty A, Bandopadhayay P, Herold-Mende C, von Deimling A, Pfister SM, Montero J, Capper D, Oehme I, Sahm F, Jones DTW, Witt O, Milde T. BH3 mimetics targeting BCL-XL impact the senescent compartment of pilocytic astrocytoma. Neuro Oncol 2023; 25:735-747. [PMID: 35977048 PMCID: PMC10076946 DOI: 10.1093/neuonc/noac199] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Pilocytic astrocytoma (PA) is the most common pediatric brain tumor and a mitogen-activated protein kinase (MAPK)-driven disease. Oncogenic MAPK-signaling drives the majority of cells into oncogene-induced senescence (OIS). While OIS induces resistance to antiproliferative therapies, it represents a potential vulnerability exploitable by senolytic agents. METHODS We established new patient-derived PA cell lines that preserve molecular features of the primary tumors and can be studied in OIS and proliferation depending on expression or repression of the SV40 large T antigen. We determined expression of anti-apoptotic BCL-2 members in these models and primary PA. Dependence of senescent PA cells on anti-apoptotic BCL-2 members was investigated using a comprehensive set of BH3 mimetics. RESULTS Senescent PA cells upregulate BCL-XL upon senescence induction and show dependency on BCL-XL for survival. BH3 mimetics with high affinity for BCL-XL (BCL-XLi) reduce metabolic activity and induce mitochondrial apoptosis in senescent PA cells at nano-molar concentrations. In contrast, BH3 mimetics without BCL-XLi activity, conventional chemotherapy, and MEK inhibitors show no effect. CONCLUSIONS Our data demonstrate that BCL-XL is critical for survival of senescent PA tumor cells and provides proof-of-principle for the use of clinically available BCL-XL-dependent senolytics.
Collapse
Affiliation(s)
- Florian Selt
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- KiTZ Clinical Trial Unit (ZIPO), Department of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany
| | - Romain Sigaud
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
| | - Gintvile Valinciute
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
| | - Philipp Sievers
- Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Julia Zaman
- Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Clara Alcon
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), 08028, Barcelona, Spain
| | - Simone Schmid
- Charité-Universitaetsmedizin Berlin, Corporate Member of Freie Universitaet Berlin, Humboldt-Universitaet zu Berlin, and Berlin Institute of Health, Department of Neuropathology, Berlin, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Berlin, Berlin, Germany
| | - Heike Peterziel
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
| | - Jessica W Tsai
- Dana-Farber/Boston Children’s Cancer and Blood Disorder Center, Boston, Massachusetts, USA
| | - Romain Guiho
- Developmental Biology and Cancer Research & Teaching Department, Birth Defects Research Centre, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Juan Pedro Martínez-Barbera
- Developmental Biology and Cancer Research & Teaching Department, Birth Defects Research Centre, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Stefan Pusch
- Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jing Deng
- Ascentage Pharma (Suzhou) Co, Ltd, Suzhou, Jiangsu Province, China
| | - Yifan Zhai
- Ascentage Pharma (Suzhou) Co, Ltd, Suzhou, Jiangsu Province, China
| | - Cornelis M van Tilburg
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- KiTZ Clinical Trial Unit (ZIPO), Department of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany
| | - Martin U Schuhman
- Department of Neurosurgery, University Hospital Tübingen, Tübingen, Germany
| | - Ahmed El Damaty
- Pediatric Neurosurgery Division, Department of Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Pratiti Bandopadhayay
- Dana-Farber/Boston Children’s Cancer and Blood Disorder Center, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Andreas von Deimling
- Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefan M Pfister
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- KiTZ Clinical Trial Unit (ZIPO), Department of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
| | - Joan Montero
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), 08028, Barcelona, Spain
| | - David Capper
- Charité-Universitaetsmedizin Berlin, Corporate Member of Freie Universitaet Berlin, Humboldt-Universitaet zu Berlin, and Berlin Institute of Health, Department of Neuropathology, Berlin, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Berlin, Berlin, Germany
| | - Ina Oehme
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
| | - Felix Sahm
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - David T W Jones
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Division of Pediatric Glioma Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Olaf Witt
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- KiTZ Clinical Trial Unit (ZIPO), Department of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany
| | - Till Milde
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- KiTZ Clinical Trial Unit (ZIPO), Department of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
13
|
Tang LW, Mallela AN, Deng H, Richardson TE, Hervey-Jumper SL, McBrayer SK, Abdullah KG. Preclinical modeling of lower-grade gliomas. Front Oncol 2023; 13:1139383. [PMID: 37051530 PMCID: PMC10083350 DOI: 10.3389/fonc.2023.1139383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 03/16/2023] [Indexed: 03/28/2023] Open
Abstract
Models for human gliomas prove critical not only to advancing our understanding of glioma biology but also to facilitate the development of therapeutic modalities. Specifically, creating lower-grade glioma (LGG) models has been challenging, contributing to few investigations and the minimal progress in standard treatment over the past decade. In order to reliably predict and validate the efficacies of novel treatments, however, LGG models need to adhere to specific standards that recapitulate tumor genetic aberrations and micro-environment. This underscores the need to revisit existing models of LGG and explore prospective models that may bridge the gap between preclinical insights and clinical translation. This review first outlines a set of criteria aimed to address the current challenges hindering model development. We then evaluate the strengths and weaknesses of existing preclinical models of LGG with respect to these established standards. To conclude, the review discusses potential future directions for integrating existing models to maximize the exploration of disease mechanisms and therapeutics development.
Collapse
Affiliation(s)
- Lilly W. Tang
- Physician Scientist Training Program, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Arka N. Mallela
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Hansen Deng
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Timothy E. Richardson
- Department of Pathology, Cell and Molecular Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Shawn L. Hervey-Jumper
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, United States
| | - Samuel K. McBrayer
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Kalil G. Abdullah
- Physician Scientist Training Program, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| |
Collapse
|
14
|
Kelly NA, Shontz KM, Bergman M, Manning AM, Reynolds SD, Chiang T. Biobanked tracheal basal cells retain the capacity to differentiate. Laryngoscope Investig Otolaryngol 2022; 7:2119-2125. [PMID: 36544928 PMCID: PMC9764751 DOI: 10.1002/lio2.925] [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: 05/26/2022] [Revised: 08/19/2022] [Accepted: 09/07/2022] [Indexed: 12/24/2022] Open
Abstract
Objective While airway epithelial biorepositories have established roles in the study of bronchial progenitor stem (basal) cells, the utility of a bank of tracheal basal cells from pediatric patients, who have or are suspected of having an airway disease, has not been established. In vitro study of these cells can enhance options for tracheal restoration, graft design, and disease modeling. Development of a functional epithelium in these settings is a key measure. The aim of this study was the creation a tracheal basal cell biorepository and assessment of recovered cells. Methods Pediatric patients undergoing bronchoscopy were identified and endotracheal brush (N = 29) biopsies were collected. Cells were cultured using the modified conditional reprogramming culture (mCRC) method. Samples producing colonies by day 14 were passaged and cryopreserved. To explore differentiation potential, cells were thawed and differentiated using the air-liquid interface (ALI) method. Results No adverse events were associated with biopsy collection. Of 29 brush biopsies, 16 (55%) were successfully cultured to passage 1/cryopreserved. Samples with higher initial cell yields were more likely to achieve this benchmark. Ten unique donors were then thawed for analysis of differentiation. The average age was 2.2 ± 2.2 years with five donors (50%) having laryngotracheal pathology. Nine donors (90%) demonstrated differentiation capacity at 21 days of culture, as indicated by detection of ciliated cells (ACT+) and mucous cells (MUC5B+). Conclusion Pediatric tracheal basal cells can be successfully collected and cryopreserved. Recovered cells retain the ability to differentiate into epithelial cell types in vitro. Level of Evidence Level 3.
Collapse
Affiliation(s)
- Natalie A. Kelly
- Department of OtolaryngologyNationwide Children's HospitalColumbusOhioUSA
| | - Kimberly M. Shontz
- Center for Regenerative MedicineAbigail Wexner Research Institute at Nationwide Children's HospitalColumbusOhioUSA
| | - Maxwell Bergman
- Department of Otolaryngology‐Head and Neck SurgeryThe Ohio State Wexner Medical CenterColumbusOhioUSA
| | - Amy M. Manning
- Department of OtolaryngologyNationwide Children's HospitalColumbusOhioUSA
- Department of Otolaryngology‐Head and Neck SurgeryThe Ohio State Wexner Medical CenterColumbusOhioUSA
| | - Susan D. Reynolds
- Center for Perinatal MedicineAbigail Wexner Research Institute at Nationwide Children's HospitalColumbusOhioUSA
| | - Tendy Chiang
- Department of OtolaryngologyNationwide Children's HospitalColumbusOhioUSA
- Center for Regenerative MedicineAbigail Wexner Research Institute at Nationwide Children's HospitalColumbusOhioUSA
- Department of Otolaryngology‐Head and Neck SurgeryThe Ohio State Wexner Medical CenterColumbusOhioUSA
| |
Collapse
|
15
|
Basheer F, Dhar P, Samarasinghe RM. Zebrafish Models of Paediatric Brain Tumours. Int J Mol Sci 2022; 23:9920. [PMID: 36077320 PMCID: PMC9456103 DOI: 10.3390/ijms23179920] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/25/2022] [Accepted: 08/29/2022] [Indexed: 11/30/2022] Open
Abstract
Paediatric brain cancer is the second most common childhood cancer and is the leading cause of cancer-related deaths in children. Despite significant advancements in the treatment modalities and improvements in the 5-year survival rate, it leaves long-term therapy-associated side effects in paediatric patients. Addressing these impairments demands further understanding of the molecularity and heterogeneity of these brain tumours, which can be demonstrated using different animal models of paediatric brain cancer. Here we review the use of zebrafish as potential in vivo models for paediatric brain tumour modelling, as well as catalogue the currently available zebrafish models used to study paediatric brain cancer pathophysiology, and discuss key findings, the unique attributes that these models add, current challenges and therapeutic significance.
Collapse
Affiliation(s)
- Faiza Basheer
- School of Medicine, Deakin University, Geelong, VIC 3220, Australia
- Instiute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, VIC 3220, Australia
| | - Poshmaal Dhar
- School of Medicine, Deakin University, Geelong, VIC 3220, Australia
- Instiute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, VIC 3220, Australia
| | - Rasika M. Samarasinghe
- School of Medicine, Deakin University, Geelong, VIC 3220, Australia
- Instiute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, VIC 3220, Australia
| |
Collapse
|
16
|
Anastasaki C, Chatterjee J, Cobb O, Sanapala S, Scheaffer SM, De Andrade Costa A, Wilson AF, Kernan CM, Zafar AH, Ge X, Garbow JR, Rodriguez FJ, Gutmann DH. Human induced pluripotent stem cell engineering establishes a humanized mouse platform for pediatric low-grade glioma modeling. Acta Neuropathol Commun 2022; 10:120. [PMID: 35986378 PMCID: PMC9392324 DOI: 10.1186/s40478-022-01428-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 08/11/2022] [Indexed: 11/25/2022] Open
Abstract
A major obstacle to identifying improved treatments for pediatric low-grade brain tumors (gliomas) is the inability to reproducibly generate human xenografts. To surmount this barrier, we leveraged human induced pluripotent stem cell (hiPSC) engineering to generate low-grade gliomas (LGGs) harboring the two most common pediatric pilocytic astrocytoma-associated molecular alterations, NF1 loss and KIAA1549:BRAF fusion. Herein, we identified that hiPSC-derived neuroglial progenitor populations (neural progenitors, glial restricted progenitors and oligodendrocyte progenitors), but not terminally differentiated astrocytes, give rise to tumors retaining LGG histologic features for at least 6 months in vivo. Additionally, we demonstrated that hiPSC-LGG xenograft formation requires the absence of CD4 T cell-mediated induction of astrocytic Cxcl10 expression. Genetic Cxcl10 ablation is both necessary and sufficient for human LGG xenograft development, which additionally enables the successful long-term growth of patient-derived pediatric LGGs in vivo. Lastly, MEK inhibitor (PD0325901) treatment increased hiPSC-LGG cell apoptosis and reduced proliferation both in vitro and in vivo. Collectively, this study establishes a tractable experimental humanized platform to elucidate the pathogenesis of and potential therapeutic opportunities for childhood brain tumors.
Collapse
Affiliation(s)
- Corina Anastasaki
- Department of Neurology, Washington University School of Medicine, 660 S. Euclid Avenue, Box 8111, St. Louis, MO, 63110, USA
| | - Jit Chatterjee
- Department of Neurology, Washington University School of Medicine, 660 S. Euclid Avenue, Box 8111, St. Louis, MO, 63110, USA
| | - Olivia Cobb
- Department of Neurology, Washington University School of Medicine, 660 S. Euclid Avenue, Box 8111, St. Louis, MO, 63110, USA
| | - Shilpa Sanapala
- Department of Neurology, Washington University School of Medicine, 660 S. Euclid Avenue, Box 8111, St. Louis, MO, 63110, USA
| | - Suzanne M Scheaffer
- Department of Neurology, Washington University School of Medicine, 660 S. Euclid Avenue, Box 8111, St. Louis, MO, 63110, USA
| | - Amanda De Andrade Costa
- Department of Neurology, Washington University School of Medicine, 660 S. Euclid Avenue, Box 8111, St. Louis, MO, 63110, USA
| | - Anna F Wilson
- Department of Neurology, Washington University School of Medicine, 660 S. Euclid Avenue, Box 8111, St. Louis, MO, 63110, USA
| | - Chloe M Kernan
- Department of Neurology, Washington University School of Medicine, 660 S. Euclid Avenue, Box 8111, St. Louis, MO, 63110, USA
| | - Ameera H Zafar
- Department of Neurology, Washington University School of Medicine, 660 S. Euclid Avenue, Box 8111, St. Louis, MO, 63110, USA
| | - Xia Ge
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Joel R Garbow
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Fausto J Rodriguez
- Department of Pathology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, 660 S. Euclid Avenue, Box 8111, St. Louis, MO, 63110, USA.
| |
Collapse
|
17
|
Yuan M, Eberhart CG, Pratilas CA, Blakeley JO, Davis C, Stojanova M, Reilly K, Meeker AK, Heaphy CM, Rodriguez FJ. Therapeutic Vulnerability to ATR Inhibition in Concurrent NF1 and ATRX-Deficient/ALT-Positive High-Grade Solid Tumors. Cancers (Basel) 2022; 14:cancers14123015. [PMID: 35740680 PMCID: PMC9221513 DOI: 10.3390/cancers14123015] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 06/17/2022] [Indexed: 01/02/2023] Open
Abstract
Simple Summary Tumors of the brain and nerves develop frequently in patients with neurofibromatosis type 1. Many are benign growths, such as pilocytic astrocytomas in the brain and neurofibromas in the nerves. However, in some patients, the tumors become malignant and may cause local damage, disseminate to distant sites and result in death. We studied changes in the levels of chromatin proteins and changes in telomeres, in cells obtained from mouse gliomas that are deficient in neurofibromin as well as excess brain and nerve tumor tissue from patients with neurofibromatosis type 1 or sporadic tumors lacking neurofibromin expression. A decrease in the levels of these proteins in experimental cell lines resulted in susceptibility to a class of specific drugs knowns as ATR inhibitors, which may represent a specific vulnerability of these tumor subgroups. We expect our data to provide the required rationale for the development of more accurate animal models to study neurofibromatosis, as well as specific molecularly based drugs for treatment as alternatives to the current, often devastating approaches of surgery, radiation, and chemotherapy. Abstract Subsets of Neurofibromatosis Type 1 (NF1)-associated solid tumors have been shown to display high frequencies of ATRX mutations and the presence of alternative lengthening of telomeres (ALT). We studied the phenotype of combined NF1 and ATRX deficiency in malignant solid tumors. Cell lines derived from NF1-deficient sporadic glioblastomas (U251, SF188), an NF1-associated ATRX mutant glioblastoma cell line (JHH-NF1-GBM1), an NF1-derived sarcoma cell line (JHH-CRC65), and two NF1-deficient MPNST cell lines (ST88-14, NF90.8) were utilized. Cancer cells were treated with ATR inhibitors, with or without a MEK inhibitor or temozolomide. In contrast to the glioma cell line SF188, combined ATRX knockout (KO) and TERC KO led to ALT-like properties and sensitized U251 glioma cells to ATR inhibition in vitro and in vivo. In addition, ATR inhibitors sensitized U251 cells to temozolomide, but not MEK inhibition, irrespective of ATRX level manipulation; whereas, the JHH-NF1-GBM1 cell line demonstrated sensitivity to ATR inhibition, but not temozolomide. Similar effects were noted using the MPNST cell line NF90.8 after combined ATRX knockdown and TERC KO; however, not in ST88-14. Taken together, our study supports the feasibility of targeting the ATR pathway in subsets of NF1-deficient and associated tumors.
Collapse
Affiliation(s)
- Ming Yuan
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA; (M.Y.); (C.G.E.); (C.D.); (A.K.M.)
| | - Charles G. Eberhart
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA; (M.Y.); (C.G.E.); (C.D.); (A.K.M.)
| | - Christine A. Pratilas
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA; (C.A.P.); (J.O.B.)
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
| | - Jaishri O. Blakeley
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA; (C.A.P.); (J.O.B.)
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
| | - Christine Davis
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA; (M.Y.); (C.G.E.); (C.D.); (A.K.M.)
| | - Marija Stojanova
- Department of Medicine, Boston University School of Medicine and Boston Medical Center, Boston, MA 02118, USA;
| | | | - Alan K. Meeker
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA; (M.Y.); (C.G.E.); (C.D.); (A.K.M.)
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA; (C.A.P.); (J.O.B.)
| | - Christopher M. Heaphy
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA; (M.Y.); (C.G.E.); (C.D.); (A.K.M.)
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA; (C.A.P.); (J.O.B.)
- Department of Medicine, Boston University School of Medicine and Boston Medical Center, Boston, MA 02118, USA;
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine and Boston Medical Center, Boston, MA 02118, USA
- Correspondence: (C.M.H.); (F.J.R.)
| | - Fausto J. Rodriguez
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA; (M.Y.); (C.G.E.); (C.D.); (A.K.M.)
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA; (C.A.P.); (J.O.B.)
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles (UCLA), 10833 Le Conte Avenue, CHS Bldg., Suite 18-170B, Los Angeles, CA 90095, USA
- Correspondence: (C.M.H.); (F.J.R.)
| |
Collapse
|
18
|
Abdullah KG, Bird CE, Buehler JD, Gattie LC, Savani MR, Sternisha AC, Xiao Y, Levitt MM, Hicks WH, Li W, Ramirez DMO, Patel T, Garzon-Muvdi T, Barnett S, Zhang G, Ashley DM, Hatanpaa KJ, Richardson TE, McBrayer SK. Establishment of patient-derived organoid models of lower-grade glioma. Neuro Oncol 2022; 24:612-623. [PMID: 34850183 PMCID: PMC8972292 DOI: 10.1093/neuonc/noab273] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Historically, creating patient-derived models of lower-grade glioma (LGG) has been challenging, contributing to few experimental platforms that support laboratory-based investigations of this disease. Although organoid modeling approaches have recently been employed to create in vitro models of high-grade glioma (HGG), it is unknown whether this approach can be successfully applied to LGG. METHODS In this study, we developed an optimized protocol for the establishment of organoids from LGG primary tissue samples by utilizing physiologic (5%) oxygenation conditions and employed it to produce the first known suite of these models. To assess their fidelity, we surveyed key biological features of patient-derived organoids using metabolic, genomic, histologic, and lineage marker gene expression assays. RESULTS Organoid models were created with a success rate of 91% (n = 20/22) from primary tumor samples across glioma histological subtypes and tumor grades (WHO Grades 1-4), and a success rate of 87% (13/15) for WHO Grade 1-3 tumors. Patient-derived organoids recapitulated stemness, proliferative, and tumor-stromal composition profiles of their respective parental tumor specimens. Cytoarchitectural, mutational, and metabolic traits of parental tumors were also conserved. Importantly, LGG organoids were maintained in vitro for weeks to months and reanimated after biobanking without loss of integrity. CONCLUSIONS We report an efficient method for producing faithful in vitro models of LGG. New experimental platforms generated through this approach are well positioned to support preclinical studies of this disease, particularly those related to tumor immunology, tumor-stroma interactions, identification of novel drug targets, and personalized assessments of treatment response profiles.
Collapse
Affiliation(s)
- Kalil G Abdullah
- Department of Neurological Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- O’Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas,USA
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas,USA
| | - Cylaina E Bird
- Department of Neurological Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Joseph D Buehler
- Department of Neurological Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Lauren C Gattie
- Department of Neurological Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Milan R Savani
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas,USA
| | - Alex C Sternisha
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas,USA
| | - Yi Xiao
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas,USA
| | - Michael M Levitt
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas,USA
| | - William H Hicks
- Department of Neurological Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Wenhao Li
- O’Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas,USA
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas,USA
| | - Denise M O Ramirez
- O’Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas,USA
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas,USA
| | - Toral Patel
- Department of Neurological Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- O’Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas,USA
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas,USA
| | - Tomas Garzon-Muvdi
- Department of Neurological Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- O’Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas,USA
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas,USA
| | - Samuel Barnett
- Department of Neurological Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- O’Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas,USA
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas,USA
| | - Gao Zhang
- Duke University School of Medicine, Duke University, Durham, North Carolina,USA
| | - David M Ashley
- Duke University School of Medicine, Duke University, Durham, North Carolina,USA
| | - Kimmo J Hatanpaa
- Department of Pathology, Division of Neuropathology, University of Texas Southwestern Medical Center, Dallas, Texas,USA
| | - Timothy E Richardson
- Department of Pathology and Laboratory Medicine and The Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, Texas,USA
| | - Samuel K McBrayer
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas,USA
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas,USA
| |
Collapse
|
19
|
Rota CM, Brown AT, Addleson E, Ives C, Trumper E, Pelton K, Teh WP, Schniederjan MJ, Castellino RC, Buhrlage S, Lauffenburger DA, Ligon KL, Griffith LG, Segal RA. Synthetic extracellular matrices and astrocytes provide a supportive microenvironment for the cultivation and investigation of primary pediatric gliomas. Neurooncol Adv 2022; 4:vdac049. [PMID: 35669012 PMCID: PMC9159660 DOI: 10.1093/noajnl/vdac049] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Background Pediatric gliomas comprise a diverse set of brain tumor entities that have substantial long-term ramifications for patient survival and quality of life. However, the study of these tumors is currently limited due to a lack of authentic models. Additionally, many aspects of pediatric brain tumor biology, such as tumor cell invasiveness, have been difficult to study with currently available tools. To address these issues, we developed a synthetic extracellular matrix (sECM)-based culture system to grow and study primary pediatric brain tumor cells. Methods We developed a brain-like sECM material as a supportive scaffold for the culture of primary, patient-derived pediatric glioma cells and established patient-derived cell lines. Primary juvenile brainstem-derived murine astrocytes were used as a feeder layer to support the growth of primary human tumor cells. Results We found that our culture system facilitated the proliferation of various primary pediatric brain tumors, including low-grade gliomas, and enabled ex vivo testing of investigational therapeutics. Additionally, we found that tuning this sECM material allowed us to assess high-grade pediatric glioma cell invasion and evaluate therapeutic interventions targeting invasive behavior. Conclusion Our sECM culture platform provides a multipurpose tool for pediatric brain tumor researchers that enables both a wide breadth of biological assays and the cultivation of diverse tumor types.
Collapse
Affiliation(s)
- Christopher M Rota
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Alexander T Brown
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Emily Addleson
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Clara Ives
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Ella Trumper
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Kristine Pelton
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Wei Pin Teh
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | | | | | - Sara Buhrlage
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Douglas A Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Keith L Ligon
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Linda G Griffith
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Rosalind A Segal
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| |
Collapse
|
20
|
de Andrade Costa A, Chatterjee J, Cobb O, Cordell E, Chao A, Schaeffer S, Goldstein A, Dahiya S, Gutmann DH. Immune deconvolution and temporal mapping identifies stromal targets and developmental intervals for abrogating murine low-grade optic glioma formation. Neurooncol Adv 2022; 4:vdab194. [PMID: 35187488 PMCID: PMC8852255 DOI: 10.1093/noajnl/vdab194] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Brain tumor formation and progression are dictated by cooperative interactions between neoplastic and non-neoplastic cells. This stromal dependence is nicely illustrated by tumors arising in the Neurofibromatosis type 1 (NF1) cancer predisposition syndrome, where children develop low-grade optic pathway gliomas (OPGs). Using several authenticated Nf1-OPG murine models, we previously demonstrated that murine Nf1-OPG growth is regulated by T cell function and microglia Ccl5 production, such that their inhibition reduces tumor proliferation in vivo. While these interactions are critical for established Nf1-OPG tumor growth, their importance in tumor formation has not been explored. METHODS A combination of bulk and single-cell RNA mouse optic nerve sequencing, immunohistochemistry, T cell assays, and pharmacologic and antibody-mediated inhibition methods were used in these experiments. RESULTS We show that T cells and microglia are the main non-neoplastic immune cell populations in both murine and human LGGs. Moreover, we demonstrate that CD8+ T cells, the predominant LGG-infiltrating lymphocyte population, are selectively recruited through increased Ccl2 receptor (Ccr4) expression in CD8+, but not CD4+, T cells, in a NF1/RAS-dependent manner. Finally, we identify the times during gliomagenesis when microglia Ccl5 production (3-6 weeks of age) and Ccl2-mediated T cell infiltration (7-10 weeks of age) occur, such that temporally-restricted Ccl2 or Ccl5 inhibition abrogates tumor formation >3.5 months following the cessation of treatment. CONCLUSIONS Collectively, these findings provide proof-of-concept demonstrations that targeting stromal support during early gliomagenesis durably blocks murine LGG formation.
Collapse
Affiliation(s)
| | - Jit Chatterjee
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Olivia Cobb
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Elizabeth Cordell
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Astoria Chao
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Suzanne Schaeffer
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Andrea Goldstein
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Sonika Dahiya
- Department of Pathology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
21
|
Milde T, Rodriguez FJ, Barnholtz-Sloan JS, Patil N, Eberhart CG, Gutmann DH. Reimagining Pilocytic Astrocytomas in the Context of Pediatric Low-Grade Gliomas. Neuro Oncol 2021; 23:1634-1646. [PMID: 34131743 DOI: 10.1093/neuonc/noab138] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Pediatric low-grade gliomas (pLGGs) are the most common brain tumor in children, and are associated with life-long clinical morbidity. Relative to their high-grade adult counterparts or other malignant childhood brain tumors, there is a paucity of authenticated preclinical models for these pediatric low-grade gliomas and an incomplete understanding of their molecular and cellular pathogenesis. While large scale genomic profiling efforts have identified the majority of pathogenic driver mutations, which converge on the MAPK/ERK signaling pathway, it is now appreciated that these events may not be sufficient by themselves for gliomagenesis and clinical progression. In light of the recent World Health Organization reclassification of pLGGs, and pilocytic astrocytoma (PA) in particular, we review our current understanding of these pediatric brain tumors, provide a conceptual framework for future mechanistic studies, and outline the challenges and pressing needs for the pLGG clinical and research communities.
Collapse
Affiliation(s)
- Till Milde
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany.,Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany.,Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Fausto J Rodriguez
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore MD, USA
| | - Jill S Barnholtz-Sloan
- Department of Population and Quantitative Health Sciences, Case Western Reserve School of Medicine, Cleveland OH, USA.,University Hospitals, Cleveland OH, USA.,Central Brain Tumor Registry of the United States (CBTRUS), Hinsdale, IL, USA
| | - Nirav Patil
- University Hospitals, Cleveland OH, USA.,Central Brain Tumor Registry of the United States (CBTRUS), Hinsdale, IL, USA
| | - Charles G Eberhart
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore MD, USA
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis MO, USA
| |
Collapse
|