Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Targeting PAK4 to reprogram the vascular microenvironment and improve CAR-T immunotherapy for glioblastoma

Abstract

Malignant solid tumors are characterized by aberrant vascularity that fuels the formation of an immune-hostile microenvironment and induces resistance to immunotherapy. Vascular abnormalities may be driven by pro-angiogenic pathway activation and genetic reprogramming in tumor endothelial cells (ECs). Here, our kinome-wide screening of mesenchymal-like transcriptional activation in human glioblastoma (GBM)-derived ECs identifies p21-activated kinase 4 (PAK4) as a selective regulator of genetic reprogramming and aberrant vascularization. PAK4 knockout induces adhesion protein re-expression in ECs, reduces vascular abnormalities, improves T cell infiltration and inhibits GBM growth in mice. Moreover, PAK4 inhibition normalizes the tumor vascular microenvironment and sensitizes GBM to chimeric antigen receptor–T cell immunotherapy. Finally, we reveal a MEF2D/ZEB1- and SLUG-mediated mechanism by which PAK4 reprograms the EC transcriptome and downregulates claudin-14 and VCAM-1 expression, enhancing vessel permeability and reducing T cell adhesion to the endothelium. Thus, targeting PAK4-mediated EC plasticity may offer a unique opportunity to recondition the vascular microenvironment and strengthen cancer immunotherapy.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Identification of PAK4 as a critical regulator of mesenchymal-like transcriptional activation and functional abnormalities in GBM ECs.
Fig. 2: Endothelial-specific deletion of PAK4 inhibits vascular abnormalities and enhances T cell infiltration, leading to reduced tumor growth and increased mouse survival.
Fig. 3: Pharmacological PAK4 inhibition reduces proliferation selectively in GBM ECs and normalizes the tumor vasculature.
Fig. 4: PAK4 is critical for mesenchymal-like transcriptional reprogramming in GBM ECs.
Fig. 5: PAK4 suppresses adhesion protein expression via ZEB1 and SLUG, enhancing vessel permeability and reducing T cell adhesion to GBM ECs.
Fig. 6: PAK4 induces ZEB1 expression via MEF2D in GBM ECs.
Fig. 7: PAK4 inhibition sensitizes GBM to CAR-T immunotherapy.

Similar content being viewed by others

Data availability

RNA-seq data have been deposited in the National Center for Biotechnology Information’s Gene Expression Omnibus under accession code GSE154133. The MSigDB database used in the study is available at https://www.gsea-msigdb.org/gsea/msigdb. All of the other data supporting the findings of this study are available from the corresponding authors upon reasonable request. Source data are provided with this paper.

References

  1. Carmeliet, P. & Jain, R. K. Principles and mechanisms of vessel normalization for cancer and other angiogenic diseases. Nat. Rev. Drug Discov. 10, 417–427 (2011).

    Article  CAS  PubMed  Google Scholar 

  2. Weis, S. M. & Cheresh, D. A. Tumor angiogenesis: molecular pathways and therapeutic targets. Nat. Med. 17, 1359–1370 (2011).

    Article  CAS  PubMed  Google Scholar 

  3. Hanahan, D. & Weinberg, R. A. Hallmarks of cancer: the next generation. Cell 144, 646–674 (2011).

    Article  CAS  PubMed  Google Scholar 

  4. Lanitis, E., Irving, M. & Coukos, G. Targeting the tumor vasculature to enhance T cell activity. Curr. Opin. Immunol. 33, 55–63 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Wang, Q. et al. Vascular niche IL-6 induces alternative macrophage activation in glioblastoma through HIF-2α. Nat. Commun. 9, 559 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  6. Huang, Y. et al. Improving immune–vascular crosstalk for cancer immunotherapy. Nat. Rev. Immunol. 18, 195–203 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Klein, D. The tumor vascular endothelium as decision maker in cancer therapy. Front. Oncol. 8, 367 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Zheng, X. et al. Increased vessel perfusion predicts the efficacy of immune checkpoint blockade. J. Clin. Invest. 128, 2104–2115 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Schaaf, M. B., Garg, A. D. & Agostinis, P. Defining the role of the tumor vasculature in antitumor immunity and immunotherapy. Cell Death Dis. 9, 115 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Batchelor, T. T. et al. AZD2171, a pan-VEGF receptor tyrosine kinase inhibitor, normalizes tumor vasculature and alleviates edema in glioblastoma patients. Cancer Cell 11, 83–95 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Jain, R. K. Antiangiogenesis strategies revisited: from starving tumors to alleviating hypoxia. Cancer Cell 26, 605–622 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Gilbert, M. R. et al. A randomized trial of bevacizumab for newly diagnosed glioblastoma. N. Engl. J. Med. 370, 699–708 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Huang, M. et al. c-Met-mediated endothelial plasticity drives aberrant vascularization and chemoresistance in glioblastoma. J. Clin. Invest. 126, 1801–1814 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Liu, T. et al. PDGF-mediated mesenchymal transformation renders endothelial resistance to anti-VEGF treatment in glioblastoma. Nat. Commun. 9, 3439 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  15. Fan, Y. Vascular detransformation for cancer therapy. Trends Cancer 5, 460–463 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Stupp, R. et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N. Engl. J. Med. 352, 987–996 (2005).

    Article  CAS  PubMed  Google Scholar 

  17. Huse, J. T. & Holland, E. C. Targeting brain cancer: advances in the molecular pathology of malignant glioma and medulloblastoma. Nat. Rev. Cancer 10, 319–331 (2010).

    Article  CAS  PubMed  Google Scholar 

  18. Kim, K. J. et al. Inhibition of vascular endothelial growth factor-induced angiogenesis suppresses tumour growth in vivo. Nature 362, 841–844 (1993).

    Article  CAS  PubMed  Google Scholar 

  19. Friedman, H. S. et al. Bevacizumab alone and in combination with irinotecan in recurrent glioblastoma. J. Clin. Oncol. 27, 4733–4740 (2009).

    Article  CAS  PubMed  Google Scholar 

  20. Chinot, O. L. et al. Bevacizumab plus radiotherapy–temozolomide for newly diagnosed glioblastoma. N. Engl. J. Med. 370, 709–722 (2014).

    Article  CAS  PubMed  Google Scholar 

  21. Zhao, J. et al. Immune and genomic correlates of response to anti-PD-1 immunotherapy in glioblastoma. Nat. Med. 25, 462–469 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  22. O’Rourke, D. M. 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. 9, eaaa0984 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Xie, Y. J. et al. Nanobody-based CAR T cells that target the tumor microenvironment inhibit the growth of solid tumors in immunocompetent mice. Proc. Natl Acad. Sci. USA 116, 7624–7631 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Datta, M., Coussens, L. M., Nishikawa, H., Hodi, F. S. & Jain, R. K. Reprogramming the tumor microenvironment to improve immunotherapy: emerging strategies and combination therapies. Am. Soc. Clin. Oncol. Educ. Book 39, 165–174 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Zeng, Q. et al. Endothelial cell retraction is induced by PAK2 monophosphorylation of myosin II. J. Cell Sci. 113, 471–482 (2000).

    Article  CAS  PubMed  Google Scholar 

  26. Koh, W. et al. Formation of endothelial lumens requires a coordinated PKCϵ-, Src-, Pak- and Raf-kinase-dependent signaling cascade downstream of Cdc42 activation. J. Cell Sci. 122, 1812–1822 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Radu, M. et al. p21-activated kinase 2 regulates endothelial development and function through the Bmk1/Erk5 pathway. Mol. Cell. Biol. 35, 3990–4005 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Acloque, H., Adams, M. S., Fishwick, K., Bronner-Fraser, M. & Nieto, M. A. Epithelial–mesenchymal transitions: the importance of changing cell state in development and disease. J. Clin. Invest. 119, 1438–1449 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Kalluri, R. & Weinberg, R. A. The basics of epithelial–mesenchymal transition. J. Clin. Invest. 119, 1420–1428 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Lamouille, S., Xu, J. & Derynck, R. Molecular mechanisms of epithelial–mesenchymal transition. Nat. Rev. Mol. Cell Biol. 15, 178–196 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Melder, R. J. et al. During angiogenesis, vascular endothelial growth factor and basic fibroblast growth factor regulate natural killer cell adhesion to tumor endothelium. Nat. Med. 2, 992–997 (1996).

    Article  CAS  PubMed  Google Scholar 

  32. Griffioen, A. W., Damen, C. A., Martinotti, S., Blijham, G. H. & Groenewegen, G. Endothelial intercellular adhesion molecule-1 expression is suppressed in human malignancies: the role of angiogenic factors. Cancer Res. 56, 1111–1117 (1996).

    CAS  PubMed  Google Scholar 

  33. Hellebrekers, D. M. et al. Epigenetic regulation of tumor endothelial cell anergy: silencing of intercellular adhesion molecule-1 by histone modifications. Cancer Res. 66, 10770–10777 (2006).

    Article  CAS  PubMed  Google Scholar 

  34. Su, L. et al. MEF2D transduces microenvironment stimuli to ZEB1 to promote epithelial–mesenchymal transition and metastasis in colorectal cancer. Cancer Res. 76, 5054–5067 (2016).

    Article  CAS  PubMed  Google Scholar 

  35. Sampson, J. H. et al. EGFRvIII mCAR-modified T-cell therapy cures mice with established intracerebral glioma and generates host immunity against tumor-antigen loss. Clin. Cancer Res. 20, 972–984 (2014).

    Article  CAS  PubMed  Google Scholar 

  36. Sampson, J. H., Maus, M. V. & June, C. H. Immunotherapy for brain tumors. J. Clin. Oncol. 35, 2450–2456 (2017).

    Article  CAS  PubMed  Google Scholar 

  37. Bovenberg, M. S., Degeling, M. H. & Tannous, B. A. Cell-based immunotherapy against gliomas: from bench to bedside. Mol. Ther. 21, 1297–1305 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. June, C. H. Principles of adoptive T cell cancer therapy. J. Clin. Invest. 117, 1204–1212 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Cho, J. G., Lee, A., Chang, W., Lee, M. S. & Kim, J. Endothelial to mesenchymal transition represents a key link in the interaction between inflammation and endothelial dysfunction. Front. Immunol. 9, 294 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Kovacic, J. C., Mercader, N., Torres, M., Boehm, M. & Fuster, V. Epithelial-to-mesenchymal and endothelial-to-mesenchymal transition: from cardiovascular development to disease. Circulation 125, 1795–1808 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Zeisberg, E. M. et al. Endothelial-to-mesenchymal transition contributes to cardiac fibrosis. Nat. Med. 13, 952–961 (2007).

    Article  CAS  PubMed  Google Scholar 

  42. Li, J., Qu, X. & Bertram, J. F. Endothelial–myofibroblast transition contributes to the early development of diabetic renal interstitial fibrosis in streptozotocin-induced diabetic mice. Am. J. Pathol. 175, 1380–1388 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Zeisberg, E. M., Potenta, S. E., Sugimoto, H., Zeisberg, M. & Kalluri, R. Fibroblasts in kidney fibrosis emerge via endothelial-to-mesenchymal transition. J. Am. Soc. Nephrol. 19, 2282–2287 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Chen, P. Y. et al. FGF regulates TGF-β signaling and endothelial-to-mesenchymal transition via control of let-7 miRNA expression. Cell Rep. 2, 1684–1696 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Cooley, B. C. et al. TGF-β signaling mediates endothelial-to-mesenchymal transition (EndMT) during vein graft remodeling. Sci. Transl. Med. 6, 227ra234 (2014).

    Article  Google Scholar 

  46. Maddaluno, L. et al. EndMT contributes to the onset and progression of cerebral cavernous malformations. Nature 498, 492–496 (2013).

    Article  CAS  PubMed  Google Scholar 

  47. Eilken, H. M., Nishikawa, S. & Schroeder, T. Continuous single-cell imaging of blood generation from haemogenic endothelium. Nature 457, 896–900 (2009).

    Article  CAS  PubMed  Google Scholar 

  48. Boisset, J. C. et al. In vivo imaging of haematopoietic cells emerging from the mouse aortic endothelium. Nature 464, 116–120 (2010).

    Article  CAS  PubMed  Google Scholar 

  49. Kissa, K. & Herbomel, P. Blood stem cells emerge from aortic endothelium by a novel type of cell transition. Nature 464, 112–115 (2010).

    Article  CAS  PubMed  Google Scholar 

  50. Bertrand, J. Y. et al. Haematopoietic stem cells derive directly from aortic endothelium during development. Nature 464, 108–111 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Lancrin, C. et al. The haemangioblast generates haematopoietic cells through a haemogenic endothelium stage. Nature 457, 892–895 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Krebs, A. M. et al. The EMT-activator Zeb1 is a key factor for cell plasticity and promotes metastasis in pancreatic cancer. Nat. Cell Biol. 19, 518–529 (2017).

    Article  CAS  PubMed  Google Scholar 

  53. Aghdassi, A. et al. Recruitment of histone deacetylases HDAC1 and HDAC2 by the transcriptional repressor ZEB1 downregulates E-cadherin expression in pancreatic cancer. Gut 61, 439–448 (2012).

    Article  CAS  PubMed  Google Scholar 

  54. Bolos, V. et al. The transcription factor Slug represses E-cadherin expression and induces epithelial to mesenchymal transitions: a comparison with Snail and E47 repressors. J. Cell Sci. 116, 499–511 (2003).

    Article  CAS  PubMed  Google Scholar 

  55. Radu, M., Semenova, G., Kosoff, R. & Chernoff, J. PAK signalling during the development and progression of cancer. Nat. Rev. Cancer 14, 13–25 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Kesanakurti, D. et al. A novel interaction of PAK4 with PPARγ to regulate Nox1 and radiation-induced epithelial-to-mesenchymal transition in glioma. Oncogene 36, 5309–5320 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Pitts, T. M. et al. Association of the epithelial-to-mesenchymal transition phenotype with responsiveness to the p21-activated kinase inhibitor, PF-3758309, in colon cancer models. Front. Pharmacol. 4, 35 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  58. Kumar, R., Gururaj, A. E. & Barnes, C. J. p21-activated kinases in cancer. Nat. Rev. Cancer 6, 459–471 (2006).

    Article  CAS  PubMed  Google Scholar 

  59. Wang, K. et al. Inhibition of p21 activated kinase enhances tumour immune response and sensitizes pancreatic cancer to gemcitabine. Int. J. Oncol. 52, 261–269 (2018).

    CAS  PubMed  Google Scholar 

  60. Abril-Rodriguez, G. et al. PAK4 inhibition improves PD-1 blockade immunotherapy. Nat. Cancer 1, 46–58 (2019); erratum 1, 264 (2020).

  61. Wang, Y. et al. Ephrin-B2 controls VEGF-induced angiogenesis and lymphangiogenesis. Nature 465, 483–486 (2010).

    Article  CAS  PubMed  Google Scholar 

  62. Liu, Y. et al. Somatic cell type specific gene transfer reveals a tumor-promoting function for p21Waf1/Cip1. EMBO J. 26, 4683–4693 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Ciznadija, D., Liu, Y., Pyonteck, S. M., Holland, E. C. & Koff, A. Cyclin D1 and Cdk4 mediate development of neurologically destructive oligodendroglioma. Cancer Res. 71, 6174–6183 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Fan, Y. et al. Profilin-1 phosphorylation directs angiocrine expression and glioblastoma progression through HIF-1α accumulation. Nat. Cell Biol. 16, 445–456 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Chung, K. et al. Structural and molecular interrogation of intact biological systems. Nature 497, 332–337 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We are grateful to E. Holland (Fred Hutchinson Cancer Research Center) for providing the RCAS-PDGF GBM model. We thank S. Albelda and M. Leibowitz for help with CAR-T production and G. Linette for helpful discussions. This work was supported in part by the University of Pennsylvania Academic Development Fund (to Y.F.), an Abramson Cancer Center GBM-TCE Award (to Y.F., Z.A.B. and D.M.O.), a RadOnc-TCE Award (to Y.F.), an American Association for Cancer Research Judah Folkman Award (to Y.F.) and National Institutes of Health grants R01NS094533 and R01NS106108 (to Y.F.).

Author information

Authors and Affiliations

Authors

Contributions

W.M. performed the kinome screening analysis, RNA-seq and animal studies with transgenic mice. Y.W. and F.Y. conducted the mechanistic studies and CAR-T therapy work. R.Z. performed the cell function experiments. D.Z. conducted the three-dimensional vasculature imaging. M.H. generated the screening constructs. D.Z. and L.Z. contributed to the bioinformatics analysis. J.F.D. contributed to the experimental design. Z.A.B. and D.M.O. contributed to preparation of the cells derived from patients with GBM. J.A.F. helped to design the CAR-T therapy. Y.G. and Y.F. designed and co-supervised the experiments. Y.F. conceived of the ideas and wrote the manuscript. All of the authors commented on the manuscript.

Corresponding authors

Correspondence to Yanqing Gong or Yi Fan.

Ethics declarations

Competing interests

Y.F. is an inventor on a patent application covering the use of PAK4 inhibitors for vessel normalization therapy. L.Z. received research funding from AstraZeneca, Bristol-Myers Squibb/Celgene and Prelude Therapeutics.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Effects of siRNA-mediated PAK4 knockdown on EC functions.

a,b, Human GBM-derived ECs from patient #5377 were lentivirally transduced to express SMA-fLuc and CMV-rLuc, followed by transfection with an siRNA targeting PAK4 or a random sequence. a, Cell lysates were immunoblotted. This experiment was repeated independently twice with similar results. b, Four days after transfection, fLuc and rLuc bioluminescence was analyzed (n = 3 EC samples each derived from a distinct GBM tumor, mean ± SEM). Statistical analysis by two-way ANOVA. c-f, ECs isolated from human GBM tumors or normal brains were transfection with an siRNA targeting PAK4 or a random sequence. c, ECs isolated from normal human brain were subjected to proliferation analysis (n = 3 independent experiments, mean ± SEM). Statistical analysis by two-tailed Student’s t test. d, GBM ECs isolated from patient #5465 were subjected to proliferation analysis (n = 3 independent experiments, mean ± SEM). Statistical analysis by two-tailed Student’s t test. e, ECs isolated from normal human brain were subjected to migration analysis (n = 3 EC samples each derived from a distinct GBM tumor, mean ± SEM). Statistical analysis by two-tailed Students’ t-test. f, GBM ECs isolated from three patients were subjected to migration analysis (n = 3 independent assays, mean ± SEM). Statistical analysis by one-way ANOVA.

Source data

Extended Data Fig. 2 Effects of PAK4 knockout on EC proliferation and migration and mouse growth. Ten-day-old Pak4fl/fl (WT) and Cdh5-Cre;Pak4fl/fl (PAK4-∆EC) mice were treated with tamoxifen for three days.

a,b, Aortic ECs were isolated from 21-day-old mice. a, Cell proliferation was determined using a MTT-based assay (n = 5 EC samples each derived from a distinct mouse, mean ± SEM). Statistical analysis by two-tailed Student’s t test. b, Cell migration in response to FBS was measured using a transwell assay (n = 3 EC samples each derived from a distinct mouse, mean ± SEM). Statistical analysis by two-tailed Students’ t-tests. c, Animal body weight was monitored (mean ± SEM; WT group, n = 4 mice; PAK4-ΔEC group, n = 6 mice).

Source data

Extended Data Fig. 3 PAK knockout in ECs inhibits FSP-1 expression in GBM-associated ECs.

GBM was genetically induced, followed by implantation into WT or PAK4-ΔEC mice. Tumor sections were immunostained using anti-CD31 and anti-FSP-1 antibodies, and subjected to immunofluorescence analysis. Representative images are shown (n = 4 mice). Arrows indicated FSP-1 expression in CD31+ cells. Scale bar: 100 μm.

Extended Data Fig. 4 PAK4 knockout in ECs restores VCAM-1 expression in GBM-associated ECs.

GBM was genetically induced, followed by transplantation into WT or PAK4-ΔEC mice. Tumor sections were immunostained using anti-CD31 and anti-VCAM-1 antibodies, followed by immunofluorescence analysis. Representative images are shown (n = 4 mice). Scale bar: 100 μm.

Extended Data Fig. 5 PAK4 knockdown did not affect claudin-5 or CD31 expression in GBM ECs and normal ECs.

ECs isolated from human GBM tumor (patient #5377) or normal brain were transfected with siRNA targeting PAK4 or control sequence. Cell lysates were analyzed by immunoblot. This experiment was repeated independently twice with similar results.

Source data

Extended Data Fig. 6 PAK4 kinase activity is critical for mesenchymal-like transcriptional reprogramming and cell proliferation and migration in GBM ECs.

ECs isolated from human GBM tumors were transduced to express CRISPR targeting PAK4 or a random sequence, followed by transfection with plasmids expressing WT PAK4 or kinase-dead K350M mutant PAK4 or empty vector (EV). a, Cell lysates were analyzed by immunoblot. This experiment was repeated independently twice with similar results. b, Cells were subjected to cell proliferation analysis (n = 6 EC samples derived from different tumors, mean ± SD). Statistical analysis by two-way ANOVA. c, Cells were subjected to transwell-based cell migration analysis (mean ± SEM, n = 3 EC samples derived from different tumors for control group, and n = 6 EC samples derived from different tumors for other groups). Statistical analysis by one-way ANOVA.

Source data

Extended Data Fig. 7 PAK4 induces MEF2D phosphorylation at Ser180.

a, ECs isolated from human GBM tumor (patient #5377) were transduced to express CRISPR targeting PAK4 or a random sequence, followed by transfection with plasmids expressing WT PAK4 or kinase-dead K350M mutant PAK4 or empty vector (EV). Cell lysates were analyzed by immunoblot. b, Purified MEF2D and PAK4 proteins were incubated in kinase buffer, followed by immunoblot analysis. These experiments were repeated independently twice with similar results.

Source data

Extended Data Fig. 8 A murine Egfrviii CAR T system.

a, Schematic diagram of the mouse Egfrviii CAR T construct. b, Mouse spleen-derived T cells were transduced with MSGV retrovirus that encodes Egfrviii 139 CAR or with an empty vector, followed by flow cytometry analysis of 139 CAR expression. Representative cell sortings are shown. c, Mouse T cells expressing 139 CAR were incubated with mouse GL261 glioma cells expressing mouse Egfrviii or control WT Egfr. Cell lysis was determined by europium cytotoxicity assay (mean ± SEM, n = 3 T cell samples derived from different mice). Statistical analysis by two-way ANOVA.

Source data

Extended Data Fig. 9 Expression of Egfrviii by retroviral transduction in mouse GBM cells.

GBM was genetically engineered induced in mice. Tumor-derived spheres were transduced with retrovirus that expresses mouse Egfrviii, followed by flow cytometry analysis for Egfrviii expression. Representative cell sortings are shown.

Supplementary information

Reporting Summary

Supplementary Video 1

GBM was induced in Rosa-LSL-tdTomato;Cdh5-CreETR2 mice, followed by treatment with saline or KPT9274. Tumors were excised and imaged by light sheet microscopy (n = 3 mice). Three-dimensional images of tumor vasculature were reconvoluted.

Supplementary Video 2

GBM was induced in Rosa-LSL-tdTomato;Cdh5-CreETR2 mice, followed by treatment with saline or KPT9274. Mice were injected with Hypoxyprobe-1 (pimonidazole HCl), and brain tissues were probed with antibodies against pimonidazole adducts, followed by light sheet microscopy imaging (n = 3 mice). Three-dimensional images of the vasculature (red) and hypoxic areas (green) in tumors and normal brains were reconvoluted.

Source data

Source Data Fig. 1

Statistical source data

Source Data Fig. 1

Unprocessed western blots

Source Data Fig. 2

Statistical source data

Source Data Fig. 2

Unprocessed western blots

Source Data Fig. 3

Statistical source data

Source Data Fig. 4

Statistical source data

Source Data Fig. 4

Unprocessed western blots

Source Data Fig. 5

Statistical source data

Source Data Fig. 5

Unprocessed western blots

Source Data Fig. 6

Statistical source data

Source Data Fig. 6

Unprocessed western blots

Source Data Fig. 7

Statistical source data

Source Data Extended Data Fig. 1

Statistical source data

Source Data Extended Data Fig. 1

Unprocessed western blots

Source Data Extended Data Fig. 2

Statistical source data

Source Data Extended Data Fig. 5

Unprocessed western blots

Source Data Extended Data Fig. 6

Statistical source data

Source Data Extended Data Fig. 6

Unprocessed western blots

Source Data Extended Data Fig. 7

Unprocessed western blots

Source Data Extended Data Fig. 8

Statistical source data

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ma, W., Wang, Y., Zhang, R. et al. Targeting PAK4 to reprogram the vascular microenvironment and improve CAR-T immunotherapy for glioblastoma. Nat Cancer 2, 83–97 (2021). https://doi.org/10.1038/s43018-020-00147-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43018-020-00147-8

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer