Molecular profiling of an osseous metastasis in glioblastoma during checkpoint inhibition: potential mechanisms of immune escape

Acta Neuropathol Commun. 2020 Mar 9;8(1):28. doi: 10.1186/s40478-020-00906-9.

Abstract

Peripheral metastases of glioblastoma (GBM) are very rare despite the ability of GBM cells to pass through the blood-brain barrier and be disseminated through the peripheral blood. Here, we describe a detailed genetic and immunological characterization of a GBM metastasis in the skeleton, which occurred during anti-PD-1 immune checkpoint therapy. We performed whole genome sequencing (WGS) and 850 K methylation profiling of the primary and recurrent intracranial GBM as well as one of the bone metastases. Copy number alterations (CNA) and mutational profiles were compared to known genomic alterations in the TCGA data base. In addition, immunophenotyping of the peripheral blood was performed. The patient who was primarily diagnosed with IDH-wildtype GBM. After the resection of the first recurrence, progressive intracranial re-growth was again detected, and chemotherapy was replaced by PD-1 checkpoint inhibition, which led to a complete intracranial remission. Two months later MR-imaging revealed multiple osseous lesions. Biopsy confirmed the GBM origin of the skeleton metastases. Immunophenotyping reflected the effective activation of a peripheral T-cell response, with, however, increase of regulatory T cells during disease progression. WGS sequencing demonstrated distinct genomic alterations of the GBM metastasis, with gains along chromosomes 3 and 9 and losses along chromosome 4, 10, and 11. Mutational analysis showed mutations in potentially immunologically relevant regions. Additionally, we correlated tumour-infiltrating lymphocyte and microglia presence to the occurrence of circulating tumour cells (CTCs) in a larger cohort and found a decreased infiltration of cytotoxic T cells in patients positive for CTCs. This study exemplifies that the tumour microenvironment may dictate the response to immune checkpoint therapy. In addition, our study highlights the fact that despite an effective control of intracranial GBM, certain tumour clones have the ability to evade the tumour-specific T-cell response and cause progression even outside of the CNS.

Keywords: Circulating tumour cells; Glioblastoma; Immune escape; Metastasis; T-cells; Tumour-infiltrating lymphocytes; Whole genome sequencing.

Publication types

  • Case Reports
  • Research Support, Non-U.S. Gov't

MeSH terms

  • Aged
  • Angiogenesis Inhibitors / therapeutic use
  • Antigens, CD / metabolism
  • Antigens, Differentiation, Myelomonocytic / metabolism
  • B7-H1 Antigen / metabolism
  • Bevacizumab / therapeutic use
  • Brain Neoplasms / metabolism*
  • Brain Neoplasms / pathology
  • Brain Neoplasms / therapy
  • CD3 Complex / metabolism
  • Chemoradiotherapy
  • Glial Fibrillary Acidic Protein / metabolism
  • Glioblastoma / diagnostic imaging
  • Glioblastoma / metabolism*
  • Glioblastoma / secondary
  • Glioblastoma / therapy
  • Humans
  • Immune Checkpoint Inhibitors / therapeutic use*
  • Magnetic Resonance Imaging
  • Male
  • Neoplasm Recurrence, Local
  • Nivolumab / therapeutic use*
  • Spinal Neoplasms / diagnostic imaging
  • Spinal Neoplasms / metabolism*
  • Spinal Neoplasms / secondary
  • Spinal Neoplasms / therapy
  • Tumor Suppressor Protein p53 / metabolism

Substances

  • Angiogenesis Inhibitors
  • Antigens, CD
  • Antigens, Differentiation, Myelomonocytic
  • B7-H1 Antigen
  • CD274 protein, human
  • CD3 Complex
  • CD68 antigen, human
  • GFAP protein, human
  • Glial Fibrillary Acidic Protein
  • Immune Checkpoint Inhibitors
  • TP53 protein, human
  • Tumor Suppressor Protein p53
  • Bevacizumab
  • Nivolumab