Document Type

Journal Article

Publication Title

Frontiers in Immunology

Volume

13

PubMed ID

35309309

Publisher

Frontiers

School

School of Medical and Health Sciences

RAS ID

52754

Funders

The Pirate Ship Foundation Australian Lions Children’s Cancer Research Fundation Stan Perron Charitable Foundation Cancer Council of Western Australia Research Fellowship Stan Perron Chair of Paediatric Haematology and Oncology Richard Walter Gibbon Medical Research Scholarship Australian Government Research Training Program Scholarship, University of Western Australia Australian Postgraduate Award from the Australian Government Channel 7 Telethon Trust National Cancer Institute (2R01 CA159859, P30 CA30199) National Institute of Neurological Disorders and Stroke (R35 NS122339, R01 NS096368) V Foundation for Cancer Research (D2018-021).

Comments

Abbas, Z., George, C., Ancliffe, M., Howlett, M., Jones, A. C., Kuchibhotla, M., ... & Endersby, R. (2022). Conventional Therapies Deplete Brain-Infiltrating Adaptive Immune Cells in a Mouse Model of Group 3 Medulloblastoma Implicating Myeloid Cells as Favorable Immunotherapy Targets. Frontiers in immunology, 13, p. 1-16. https://doi.org/10.3389/fimmu.2022.837013

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.

DOI

10.3389/fimmu.2022.837013

Creative Commons License

Creative Commons Attribution 4.0 License
This work is licensed under a Creative Commons Attribution 4.0 License.

Share

 
COinS