Abstract

Clinical trials revealed limited response duration of glioblastomas to the anti-angiogenic therapy bevacizumab, a VEGF-neutralizing antibody. Bevacizumab-resistant glioblastomas exhibit increased invasiveness and 50% more tumor-associated macrophages (TAMs), which can be of classical M1 or pro-tumoral M2 subtypes. As macrophage migration inhibitory factor (MIF) regulates tumor invasion and macrophage recruitment, we investigated MIF's role in bevacizumab resistance in glioblastoma. Patient bevacizumab-resistant glioblastomas exhibited 2-fold less MIF protein than bevacizumab-naive glioblastomas (n = 10; p < 0.05). Bevacizumab-resistant xenografts exhibited 6-fold less MIF protein and increased invasiveness than isogeneic bevacizumab-responsive xenografts (p < 0.05). Immunoprecipitation revealed that bevacizumab bound MIF, likely reflecting its 30% homology to the VEGF protein structure. Furthermore, VEGF stimulation of U87 cells in vitro increased MIF transcription and secretion 5-fold (p < 0.05), suggesting VEGF depletion as another mechanism of MIF downregulation. Site-directed biopsies of bevacizumab-naive glioblastomas revealed enriched MIF expression at the enhancing tumor margin, which was lost in bevacizumab-resistant glioblastomas (p < 0.05). A similar regional pattern of expression was seen with VEGF, supporting its role in stimulating tumor cell MIF secretion. Continued bevacizumab treatment of bevacizumab-resistant xenografts increased M2-like CD11b+ TAMs, 3-fold, with preferential localization at the tumor margin (p < 0.05). The same pattern of recruitment and polarization was seen in U87/MIF-shRNA relative to U87/control-shRNA intracranial xenografts (p < 0.05). When co-cultured with cells from bevacizumab-resistant xenografts or U87/MIF-shRNA cells, bone marrow matured to an M2-like phenotype (p < 0.05), while an M1-like phenotype was generated with addition of recombinant MIF in a dose-dependent manner (p < 0.05). In conclusion, MIF expression is enriched at the glioblastoma margin, a pattern that may create a perimeter of inflammatory M1-like TAMs. This pattern is lost with bevacizumab resistance, likely due to bevacizumab-induced MIF depletion and diminished VEGF-induced MIF upregulation. MIF depletion leads to M2-like TAM polarization and recruitment to the tumor margin, thus softening the tumor border and promoting invasive bevacizumab resistance.

Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.