Article Text
Abstract
Background Chimeric antigen receptor (CAR)-T cells have revolutionized treatment of multiple types of hematological malignancies, but have shown limited efficacy in patients with glioblastoma (GBM) or other solid tumors. This may be largely due to the immunosuppressive tumor microenvironment (TME) that compromises the delivery and anti-tumor activity of CAR-T cells. We previously showed that blocking VEGF (vascular endothelial growth factor) signaling can normalize tumor vessels in murine and human tumors, including GBM, breast, liver, and rectal carcinomas. Moreover, we demonstrated that vascular normalization can improve the delivery of CD8+ T cells and efficacy of immunotherapy in breast cancer models in mice. In fact, the US FDA has approved 7 different combinations of anti-VEGF drugs and immune-checkpoint blockers for liver, kidney, lung and endometrial cancers in the past 3 years.1-25 Here we tested the hypothesis that anti-VEGF therapy can improve the delivery and efficacy of CAR-T cells in immunocompetent mice bearing orthotopic GBM tumors
Methods We engineered two syngeneic mouse GBM cell lines (CT2A and GSC005) to express EGFRvIII – one of the most common neoantigens in human GBM – and CAR T cells to recognize EGFRvIII. We tested our CAR T cells in orthotopic GBMs for their efficacy in recognizing, and killing tumor cells, and the survival advantage when tumor vessels are normalized.
Results We found that treatment with the anti-mouse VEGF antibody (B20) improved CAR-T cell infiltration and distribution throughout the GBM TME, delayed tumor growth, and prolonged survival of GBM-bearing mice compared to EGFRvIII-CAR-T cell therapy alone (figure 1).
Conclusions Our study provides a strategy to overcome major challenges in CAR-T cell therapy in GBM by: (i) increasing the CAR-T cell infiltration, intratumoral distribution, and activation in murine GBM models, and (ii) reprograming TME by increasing the number and activation of endogenous effector T cells, resulting in improved anti-tumor efficacy of CAR-T therapy in two GBM mouse models. Given that anti-VEGF therapies have been approved for a number of solid tumors, including GBM, our study provides mechanistic insights and compelling preclinical data in support of testing the combination of vascular normalizing agents and CAR-T therapies in GBM patients. Furthermore, this approach may also improve CAR-T therapy of other solid tumors that share similar TME features as well as for other cell-based therapies using autologous or allogenic immune cells (e.g., NK cells, macrophages).
References
Young RM, Engel NW, Uslu U, Wellhausen N & June CH. Next-Generation CAR T-cell Therapies. Cancer Discov 2022;12:1625–1633, doi:10.1158/2159-8290.Cd-21-1683.
Rafiq S, Hackett CS & Brentjens RJ. Engineering strategies to overcome the current roadblocks in CAR T cell therapy. Nat Rev Clin Oncol 2020;17, 147–167, doi:10.1038/s41571-019-0297-y.
Brown CE. et al. Regression of glioblastoma after chimeric antigen receptor T-Cell Therapy. N Engl J Med 375, 2561–2569, doi:10.1056/NEJMoa1610497 (2016).
O'Rourke DM, et al. A single dose of peripherally infused EGFRvIII-directed CAR T cells mediates antigen loss and induces adaptive resistance in patients with recurrent glioblastoma. Sci Transl Med 2017;9, doi:10.1126/scitranslmed.aaa0984.
Good CR, et al. An NK-like CAR T cell transition in CAR T cell dysfunction. Cell 2021;184, 6081–6100.e6026, doi:10.1016/j.cell.2021.11.016.
Johnson L A, et al. Rational development and characterization of humanized anti-EGFR variant III chimeric antigen receptor T cells for glioblastoma. Sci Transl Med 2015;7, 275ra222, doi:10.1126/scitranslmed.aaa4963.
Huang Y, et al. Vascular normalizing doses of antiangiogenic treatment reprogram the immunosuppressive tumor microenvironment and enhance immunotherapy. Proc Natl Acad Sci U S A 2012;109:17561–17566, doi:10.1073/pnas.1215397109.
Fukumura D, Kloepper J, Amoozgar Z, Duda DG & Jain RK. Enhancing cancer immunotherapy using antiangiogenics: opportunities and challenges. Nat Rev Clin Oncol 2018;15:325–340, doi:10.1038/nrclinonc.2018.29.
Shrimali RK, et al. Antiangiogenic agents can increase lymphocyte infiltration into tumor and enhance the effectiveness of adoptive immunotherapy of cancer. Cancer Res 2010;70, 6171–6180, doi:10.1158/0008-5472.Can-10-0153.
10. Amoozgar Z, et al. Targeting Treg cells with GITR activation alleviates resistance to immunotherapy in murine glioblastomas. Nat Commun 2021;12, 2582, doi:10.1038/s41467-021-22885-8.
11. Ma L, et al. Enhanced CAR-T cell activity against solid tumors by vaccine boosting through the chimeric receptor. Science 2019;365:162-168, doi:10.1126/science.aav8692.
12. Marsh J, Mukherjee P & Seyfried TN. Akt-dependent proapoptotic effects of dietary restriction on late-stage management of a phosphatase and tensin homologue/tuberous sclerosis complex 2-deficient mouse astrocytoma. Clin Cancer Res 2008;14, 7751–7762, doi:10.1158/1078-0432.ccr-08-0213.
13. Saha D, Martuza RL & Rabkin SD. Oncolytic herpes simplex virus immunovirotherapy in combination with immune checkpoint blockade to treat glioblastoma. Immunotherapy 2018;10, 779–786, doi:10.2217/imt-2018-0009.
14. Kloepper, J. et al. Ang-2/VEGF bispecific antibody reprograms macrophages and resident microglia to anti-tumor phenotype and prolongs glioblastoma survival. Proc Natl Acad Sci U S A 2016;113:4476–4481, doi:10.1073/pnas.1525360113.
15. Seano G, et al. Solid stress in brain tumours causes neuronal loss and neurological dysfunction and can be reversed by lithium. Nat Biomed Eng 2019;3, 230–245, doi:10.1038/s41551-018-0334-7.
16. Gibson VB, et al. A novel method to allow noninvasive, longitudinal imaging of the murine immune system in vivo. Blood 2012;119:2545–2551, doi:10.1182/blood-2011-09-378356.
17. Aldape K, et al. Challenges to curing primary brain tumours. Nat Rev Clin Oncol 2019:16:509–520, doi:10.1038/s41571-019-0177-5.
18. Jain RK. Antiangiogenesis strategies revisited: from starving tumors to alleviating hypoxia. Cancer Cell 2014;26:605–622, doi:10.1016/j.ccell.2014.10.006.
19. Martin JD, Seano G & Jain RK. Normalizing Function of Tumor Vessels: Progress, Opportunities, and Challenges. Annu Rev Physiol 2019;81:505–534, doi:10.1146/annurev-physiol-020518-114700.
20. Liu CJ, et al. Treatment of an aggressive orthotopic murine glioblastoma model with combination checkpoint blockade and a multivalent neoantigen vaccine. Neuro Oncol 2020;22:1276–1288, doi:10.1093/neuonc/noaa050.
21. Saha D, Martuza RL & Rabkin SD. Macrophage polarization contributes to glioblastoma eradication by combination immunovirotherapy and immune checkpoint blockade. Cancer Cell 2017;32:253–267.e255, doi:10.1016/j.ccell.2017.07.006.
22. Akbari P, et al. Directing CAR T cells towards the tumor vasculature for the treatment of solid tumors. Biochim Biophys Acta Rev Cancer 2022;1877:188701, doi:10.1016/j.bbcan.2022.188701.
23. Lanitis E, et al. VEGFR-2 redirected CAR-T cells are functionally impaired by soluble VEGF-A competition for receptor binding. J Immunother Cancer 2021;9: doi:10.1136/jitc-2020-002151.
24. Bocca P, et al. Bevacizumab-mediated tumor vasculature remodelling improves tumor infiltration and antitumor efficacy of GD2-CAR T cells in a human neuroblastoma preclinical model. Oncoimmunology 2021;7, e1378843, doi:10.1080/2162402X.2017.1378843.
25. Martinez Bedoya D, Dutoit V & Migliorini D. Allogeneic CAR T Cells: An Alternative to Overcome Challenges of CAR T Cell Therapy in Glioblastoma. Front Immunol 2021;12:640082, doi:10.3389/fimmu.2021.640082.