Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2022 May 4;24(5):694-707.
doi: 10.1093/neuonc/noab244.

Anti-invasive efficacy and survival benefit of the YAP-TEAD inhibitor verteporfin in preclinical glioblastoma models

Affiliations

Anti-invasive efficacy and survival benefit of the YAP-TEAD inhibitor verteporfin in preclinical glioblastoma models

Anne Marie Barrette et al. Neuro Oncol. .

Abstract

Background: Glioblastoma (GBM) remains a largely incurable disease as current therapy fails to target the invasive nature of glioma growth in disease progression and recurrence. Here, we use the FDA-approved drug and small molecule Hippo inhibitor Verteporfin (VP) to target YAP-TEAD activity, known to mediate convergent aspects of tumor invasion/metastasis, and assess the drug's efficacy and survival benefit in GBM models.

Methods: Up to 8 low-passage patient-derived GBM cell lines with distinct genomic drivers, including 3 primary/recurrent pairs, were treated with VP or vehicle (VEH) to assess in vitro effects on proliferation, migration, invasion, YAP-TEAD activity, and transcriptomics. Patient-derived orthotopic xenograft (PDX) models were used to assess VP's brain penetrance and effects on tumor burden and survival.

Results: VP treatment disturbed YAP/TAZ-TEAD activity; disrupted transcriptome signatures related to invasion, epithelial-to-mesenchymal, and proneural-to-mesenchymal transition, phenocopying TEAD1-knockout effects; and impaired tumor migration/invasion dynamics across primary and recurrent GBM lines. In an aggressive orthotopic PDX GBM model, short-term VP treatment consistently diminished core and infiltrative tumor burden, which was associated with decreased tumor expression of Ki67, nuclear YAP, TEAD1, and TEAD-associated targets EGFR, CDH2, and ITGB1. Finally, long-term VP treatment appeared nontoxic and conferred survival benefit compared to VEH in 2 PDX models: as monotherapy in primary (de novo) GBM and in combination with Temozolomide chemoradiation in recurrent GBM, where VP treatment associated with increased MGMT methylation.

Conclusions: We demonstrate combined anti-invasive and anti-proliferative efficacy for VP with survival benefit in preclinical GBM models, indicating potential therapeutic value of this already FDA-approved drug if repurposed for GBM patients.

Keywords: Verteporfin; YAP-TEAD; invasion; migration; preclinical.

PubMed Disclaimer

Figures

Fig. 1
Fig. 1
Verteporfin downregulates YAP/TAZ-TEAD activity and transcriptomes related to adhesion, EMT, and invasion in GBM. (A) Immunocytochemistry analysis of subcellular expression of TAZ, YAP, TEAD1, and the TEAD1-target EGFR after 3-day treatment with Verteporfin (VP) (IC50 dose) vs vehicle (VEH) in G-13063 cells; *P = .017 (YAP), ***P = .0001 (TAZ), **P = 0.0046 (TEAD1), ***P = .00001 (EGFR) by Student t test; n = 24-51 images (dots) in 2-5 wells/condition; lines in box-and-whisker plots represent mean and bars represent min/max. See also Supplementary Figure 2. (B) Representative images of TAZ and TEAD1 from part A. Scale bars = 25 µm. (C) Representative images of EGFR from part A. Scale bars = 50 µm. (D) Volcano plot of differential expression RNA-seq data from 3 biologically distinct GBM lines (G-13063, G-16302, U87) treated with VP (IC50 dose) vs VEH for 3 days. Defined set of 574 downregulated and 164 upregulated genes are marked by blue and red dots, respectively (10% false discovery rate [FDR] adjustment). (E) TEAD-target gene expression in the differential RNA-seq dataset from (D) (*P < .05 and *'P < .1 by Student t test; n = 3 cell lines; bars represent mean ± SEM). (F) Functional enrichment analysis on the differential sets of genes from part (D) (log2-fold change >0.5 and <-0.5) against several gene set databases, HOMER tool. Shown are selected top enriched biological processes, pathways, and reactomes of downregulated (blue) or upregulated (red) VP-associated genes (see Supplementary Data 1 for full list). (G) Gene Set Enrichment Analysis (GSEA) of the predefined signature gene sets “HALLMARK EPITHELIAL MESENCHYMAL TRANSITION” (M5930) and “Multicancer Invasiveness”  in the VP/VEH rld normalized RNA-seq dataset. (H) GSEA analysis of rld normalized RNA-seq dataset from CRISPR/Cas9 TEAD1 knockout cell lines vs sham (G-13063, G-16302, G-12746, and G-13306 cell lines), phenocopying the enrichment pattern seen in VP-treated cells. Abbreviations: EMT, epithelial-mesenchymal transition; GBM, glioblastoma; NES, normalized enrichment score.
Fig. 2
Fig. 2
Verteporfin decreases tumor migration in vitro across primary and recurrent GBMs. (A) Dose-dependent effect of Verteporfin on spheroid dispersion/confluent cell migration, across 8 biologically distinct GBM lines, including 3 primary (P1-P3) and recurrent (R1-R3) pairs (n = 3 wells (dots) with multiple spheres/well, 2 independent experiments for G-13063 and G-16302; lines in box and whisker plots represent the mean spheroid area at 36 h after treatment, bars represent min/max). (B) Area of spheroid migration in representative G-13063 cells at 36 h of treatment with VEH or IC50 VP, marked by yellow dash line; inset shows spheroid at 1 h. Scale bars = 75 µm. (C) Dose-dependent effect of Verteporfin on transwell invasion. Dead cells are discarded prior to transwell plating and live cell number is measured using crystal violet (n = 3 wells (dots) per condition for each line). (D) Representative images of transwell membranes were analyzed in (C). Scale bars = 50 µm. (E) Identical transwell invasion assays to part (C) comparing effect of invasion in G-13063 cells treated with VEH, VP, or ERL (n = 3 wells (dots) per condition). (F) Representative images of transwell membranes from (E). Scale bars = 100 µm. (*P < .05, **P < .01, ***P < .001 by Student t test; dose-dependence confirmed by ANOVA (P < .05); C, E: bars represent mean ± SEM). Abbreviations: ERL, Erlotinib; GBM, glioblastoma; VEH, vehicle; VP, Verteporfin.
Fig. 3
Fig. 3
Brain accumulation and YAP-TEAD activity of Verteporfin in PDX tumors. (A) IVIS imaging in anesthetized littermate G-16302 PDX mice 2 h after VP administration on days 2 and 5 (6 mg/kg IP), measured at 605 nm/700 nm excitation/emission. On right, brain organ fluorescence at necropsy 2.5 h after treatment, day 5. Regions of interest (ROI) for intensity measurements are marked by blue lines. (B) Dose-dependent accumulation of VP-associated fluorescence in G-16302 PDX brains, normalized IVIS data over multiple experiments; ***P < .0001 (VEH) vs 6 mg/kg/10 mg/kg/50 mg/kg (VP); **P = .0011 6 mg/kg (VP) vs 10 mg/kg (VP); **P = .0069 10 mg/kg (VP) vs 50 mg/kg (VP) by Student t test; dose-dependence also significant by 1-way ANOVA, P < .0001; n = 13 (VEH), 6 (6 mg/kg VP), 9 (10 mg/kg VP),12 (50 mg/kg VP) PDX mice (dots); lines in plot represent mean. All IVIS measurements are performed using identical settings on day 2 of treatment, ~2 h after drug administration. (C) Accumulation of VP-associated autofluorescence (green) throughout the brain parenchyma, including outside cerebral vessels, assessed by epifluorescence microscopy (kidney shown as positive control and choroid plexus shows relative accumulation in vasculature (10 mg/kg intravenous VP, 2 h). Scale bars = 50 µm. (D, E) Representative immunofluorescence images of EGFR and YAP (D) and TEAD1 (E) expression in the core of G-16302 PDX tumors after VP treatment (100 mg/kg IP, 10 days). Scale bars = 50 µm. (F) Quantification of normalized intensity for TEAD1’s target EGFR, nuclear YAP, and TEAD1 in tumor core, using single confocal plane images; *P = .01 (EGFR), *P = .03 (TEAD1), ***P < .0001 (nuclear YAP) by 1-tailed unpaired Student t test, n = 6 mice (EGFR) and n = 4 mice (TEAD1) with multiple fields scored; lines in box-and-whisker plots represent mean and bars represent min/max. Abbreviations: IP, intraperitoneally; PDX, patient-derived orthotopic xenograft; VEH, vehicle; VP, Verteporfin.
Fig. 4
Fig. 4
Verteporfin decreases infiltrative tumor burden in aggressive PDX tumors. (A) Experimental setup and representative histological sections of infiltrative G-16302 PDX tumors at 4-week post-transplantation (wk pt) treated with vehicle or Verteporfin (100 mg/kg IP, days 21-30). Tumors are stained with human nuclear antigen (HNA). “Core” is defined as tumor area near the injection site with highest tumor density (>100 HNA+/HNA− cells) and “infiltrative edge” (IE) is defined as adjacent tumor area of lower tumor density (~50-100 HNA+/HNA− cells) and confluent tumor spread, marked by yellow lines. Scale bar = 1 mm. (B) Quantification of PDX tumor burden area within the tumor “core” and “infiltrative edge” in 8 serial histological sections per mouse, 4-wk pt and VEH/VP treatment and 3-wk pt with no treatment (NT) (*P = .0161; n = 11 mice (dots) per condition from 2 combined experiments, normalized to average VEH tumor burden area; similar results observed in 3 independent experiments). (C, D) Representative histological images (C) and quantifications (D) of Ki67 tumor core intensity normalized to HNA (10 mg/kg IP VP administration days 1-28; *P = .027; n = 6 (VP) and n = 4 (VEH)-treated G-16302 PDX mice (dots)). Scale bars = 100 µm. (E, F) Representative immunofluorescence images (E) and quantification (F) of CDH2 expression in VP/VEH-treated PDX tumors (10 mg/kg IP days 1-28 and 100 mg/kg IP days 21-30; *P = .0136; n = 4 G-16302 PDX mice (dots) per condition). Scale bars = 30 µm. (G, H) Representative images (G) and quantifications (H) of single migratory HNA+ tumor cells (annotated in yellow) away from the core and infiltrative edge in PDX mice with early Verteporfin treatment regimen (10 mg/kg IP days 1-28) (*P = .04; n = 6 G-16302 PDX mice (dots) per condition, section with largest tumor burden annotated for each). Scale bars = 1 mm. See also Supplementary Figure 5F. For parts B, D, F, and H, lines in box-and-whisker plots represent mean and bars represent min/max; P-values determined by Student t test. Abbreviations: IP, intraperitoneally; PDX, patient-derived orthotopic xenograft; VEH, vehicle; VP, Verteporfin.
Fig. 5
Fig. 5
Verteporfin survival studies in primary and recurrent GBM models. (A) Experimental setup (top) and Kaplan-Meier survival analysis (bottom) of Verteporfin treatment alone or in combination with standard chemoradiation therapy, Temozolomide plus Radiation (TMZ + RT) in the recurrent GBM G-16302 PDX model, gender-matched (VP = 10 mg/kg IP beginning on day 14, TMZ = 5 mg/kg days 21-22, two fractions of whole brain 3Gy RT days 21-22). Combined VP + TMZ + RT treatment confers survival benefit over vehicle (*P = .02 VP + TMZ + RT vs VEH; P = .72 VP vs VEH; P = .55 TMZ + RT vs VEH; P = .76 TMZ + RT vs VP; P = .26 VP + TMZ + RT vs VP; P = .23 VP + TMZ + RT vs TMZ + RT; chi-square log-rank test; n = 9 mice/treatment condition, median survival 45.5/40.5/42.5/62 days for VEH/VP/TMZ + RT/VP + TMZ + RT, respectively). (B) DNA methylation analysis of human MGMT comparing cell line (G-16302, n = 3 technical replicates (dots)) and the survival cohorts of G-16302 PDX tumors at end stage (n = 5 PDX mice (dots) per treatment group, *P < .05 by Student t test). (C) Representative hematoxylin and eosin (H&E) images of tumor necrosis in PDX tumors of TMZ + RT and TMZ + RT + VP-treated mice. Scale bar = 1 mm. T, tumor; N, necrosis. (D) Pathological scoring of percentage necrosis stratified into 2 groups based on longest and shortest survival (n = 10 mice (dots) per treatment group, 2-4 sections per mouse, *P < .05 by Student t test, lines in box-and-whisker plots represent mean, bars represent min/max; ns, not significant). (E) Experimental setup (top) and Kaplan-Meier survival analysis (bottom) of Verteporfin vs vehicle treatment in G-13063 orthotopic PDX mice (*P = .02 by chi-square log-rank test; n = 9 mice/treatment condition, median survival 171/211 days for VEH/VP, respectively; tumors confirmed in all mice included in the analysis). Abbreviations: GBM, glioblastoma; PDX, patient-derived orthotopic xenograft; RT, radiation therapy; TMZ, Temozolomide; VEH, vehicle; VP, Verteporfin.

Comment in

References

    1. Brat DJ, Castellano-Sanchez AA, Hunter SB, et al. Pseudopalisades in glioblastoma are hypoxic, express extracellular matrix proteases, and are formed by an actively migrating cell population. Cancer Res. 2004;64(3):920–927. - PubMed
    1. Xie Q, Mittal S, Berens ME. Targeting adaptive glioblastoma: an overview of proliferation and invasion. Neuro Oncol. 2014;16(12):1575–1584. - PMC - PubMed
    1. Cooper LA, Gutman DA, Chisolm C, et al. The tumor microenvironment strongly impacts master transcriptional regulators and gene expression class of glioblastoma. Am J Pathol. 2012;180(5):2108–2119. - PMC - PubMed
    1. Darmanis S, Sloan SA, Croote D, et al. Single-cell RNA-seq analysis of infiltrating neoplastic cells at the migrating front of human glioblastoma. Cell Rep. 2017;21(5):1399–1410. - PMC - PubMed
    1. Zhong J, Paul A, Kellie SJ, Neill GMO. Mesenchymal migration as a therapeutic target in glioblastoma. J Oncol. 2010;2010:430142. - PMC - PubMed

Publication types