Abstract
Neurofibromatosis Type 2 (NF2) is a tumor predisposition syndrome caused by germline inactivating mutations in the NF2 gene encoding the merlin tumor suppressor. Patients develop multiple benign tumor types in the nervous system including bilateral vestibular schwannomas. Standard treatments include surgery and radiation therapy, which may lead to loss of hearing, impaired facial nerve function and other complications. Kinase inhibitor monotherapies have been evaluated clinically for NF2 patients with limited success, and more effective non-surgical therapies are urgently needed. Schwannoma model cells treated with PI3K inhibitors upregulate activity of the focal adhesion kinase family as a compensatory survival pathway. We screened combinations of 13 clinically relevant PI3K and FAK inhibitors using human isogenic normal and merlin-deficient Schwann cell lines. The most efficacious combination was PI3K/mTOR inhibitor omipalisib with SRC/FAK inhibitor dasatinib. Sub-GI50 doses of the single drugs blocked phosphorylation of their major target proteins. The combination was superior to either single agent in promoting a G1 cell cycle arrest and produced a 44% decrease in tumor growth over a two-week period in a pilot orthotopic allograft model. Evaluation of single and combination drugs in six human primary vestibular schwannoma cell models revealed the combination was superior to the monotherapies in 3 of 6 VS samples, highlighting inter-tumor variability between patients consistent with observations from clinical trials with other molecular targeted agents. Dasatinib alone performed as well as the combination in the remaining three samples. Preclinically validated combination therapies hold promise for NF2 patients and warrants further study in clinical trials.
Keywords: Neurofibromatosis, merlin, AKT, SRC, combination
Introduction
Neurofibromatosis Type 2 (NF2) is a tumor predisposition syndrome of the nervous system. It recently was renamed NF2-related schwannomatosis (1). It is caused by pathogenic mutations in the NF2 gene that encodes the merlin tumor suppressor protein (2, 3). Individuals with NF2 develop schwannomas, meningiomas, and ependymomas throughout their nervous systems. Bilateral vestibular schwannomas (VS) on the eighth cranial nerve alone are sufficient for an NF2-schwannomatosis diagnosis (1). Although the tumors are benign and many are slow growing, over time they cause increasing loss of nerve function and can be life threatening if not removed. Current standard of care involves regular monitoring with magnetic resonance imaging, followed by surgery and/or radiotherapy when deemed necessary (4). Surgery often leaves patients with permanent disability. For VS resections, these deficits include loss of hearing, balance, and facial mobility. Over the last ten years, there have been a number of clinical trials of small molecule drugs aimed at controlling tumor growth. However, drug selection based on merlin pathway knowledge has yet to produce a Food and Drug Administration (FDA)-approved drug therapy.
Merlin plays important roles in regulating Schwann cell responses to growth factors and extracellular matrix (5). It links actin dynamics to cell cycle progression, cell differentiation and survival (6). Loss of merlin function leads to aberrant signaling in multiple receptor tyrosine kinase (RTK) activated pathways, such as the phosphoinositide-3 kinase (PI3K), mitogen-activated protein kinase (MAPK), and Hippo pathways (6-9). Merlin also regulates signaling from extracellular matrix (ECM) receptors, integrins, and cell-cell adhesion receptors, cadherins (10-12). Despite decades of research, a complete understanding of the many signaling pathways merlin controls and their effect on Schwann cell proliferation and differentiation during development and neural repair remains elusive. The complexity of the signaling pathways regulated by merlin and the ability of schwannoma cells to rewire these pathways when challenged with a single kinase inhibitor contributes to the difficulty in developing an NF2 monotherapy.
We adopted an unbiased approach to identifying drug targets and drugs that reduce viability of human schwannoma model cells. Previously, we reported results of a proof of principle, ultra-high-throughput screen of compounds in the Library of Pharmacologically Active Compounds (13). This screen identified PI3K inhibitors as an effective drug class that inhibits the well validated merlin-dependent pathway (13). As part of the NF2 Synodos Consortium, two PI3K inhibitors, omipalisib (GSK2126458), a dual PI3K/mTOR inhibitor (14), and CUDC-907, a dual PI3K/HDAC inhibitor, were identified as effective agents in a screen of 40 compounds chosen by participating NF2 clinicians. Kinome analysis of an isogenic pair of wild-type (WT) and merlin-deficient (MD) human schwannoma model cells treated with omipalisib and CUDC-907 for 24 hours revealed activation of kinases involved in integrin dependent adhesion to ECM. These were focal adhesion kinase (FAK/PTK2) and multiple members of SRC family kinases (SFK), such as Fyn, Lyn, and Src. This suggested to us that inhibition of a critical RTK-activated pathway, PI3K/mTOR, led to compensatory activation of ECM adhesion-dependent kinases to enable cell survival (15). Support for co-inhibition of these targets was provided by results of a combination high-throughput screen of the MIPE compound library in human schwannoma model cells that identified several PI3K and FAK/SFK inhibitors as top performing combinations (16). Indeed, cooperative signaling from the ErbB2/3 RTK and ß1 integrins is well-documented in Schwann cell (SC) development. (17). Here, we conducted a small combination screen of PI3K and FAK/SFK inhibitors in clinical use to evaluate their synergy and efficacy in human schwannoma models and VS cells.
Materials and Methods
Cell culture
Human wild-type (WT; HS11) and NF2-deficient cell lines (HS01) were generated as previously described (18, 19). The NF2-deficient human SC line HS02 was generated from the parental cell line HS11 using lenti-NF2-shRNA #75 (Sigma-Mission). Primary adult Schwann cell line immortalized in vitro with telomerase reverse transcriptase (TERT) and cyclin-dependent kinase 4 (cdk4) (Dr. M Wallace, University of Florida) were used to generate NF2-deficient cell lines HS03 and HS04.3. HS03 cells were generated using lenti-NF2-shRNA #45 (Sigma-Mission). HS04.3 cells were generated using CRISPR/Cas9 with NF2sg1 (Broad Institute), Clone #3. The NF2-deficient mouse cell line, MS01, was generated as previously described (12, 20). Cells lines were authenticated by protein expression of human nuclear antigen, S100ß1, proteolipid protein and Sox10, as well as population studies using EdU incorporation and flow cytometry analysis. Cells were used between passage 6 and 18, and routinely monitored for mycoplasma, as previously described (18, 19). Vestibular schwannoma tumors and blood samples were harvested from 6 patients at the time of surgery (UM IRB #20150637) and used to create primary cell lines, as previously described (18, 21). Retrospective chart review was performed to confirm pathologic diagnosis of schwannoma (World Health Organization Grade I). DNA was extracted from blood leukocytes and tumor chunks using DNeasy Blood and Tissue Kit (Qiagen). Whole exome sequencing and multiplex ligation-dependent probe amplification was performed to detect mutations in the NF2, LTZR1, and SMARCB1 genes (20, 21).
Compounds
All compounds were purchased from SelleckChem and 10mM stocks were prepared in DMSO. For the allograft study, omipalisib and dasatinib were solubilized in 1.25% PEG, 2.5% Tween80, and 5% DMSO.
Combination screening
Combination matrix cell viability assays were conducted as previously described (16). Briefly, cells were seeded at 2,000 cells/well in 384-well plates. After overnight adhesion, cells received compounds in 0.1% DMSO and allowed to incubate for 72h at 37°C, 5% CO2. Cell viability was measured with CellTiterFluor Viability Assay (Promega) and read on a Synergy H1 plate reader (BioTek). Cell viability was normalized to DMSO-only controls. Assay performance was evaluated by calculating the Z’ score for each plate with control wells. All Z’ values were >0.5. Synergy was determined using the open access Combenefit software (22). Biological interactions were determined using the sum weighted score of two standard reference models, Loewe and Highest Single Agent (HSA). Selectivity for merlin-deficient over isogenic normal cells was calculated from change in Loewe sum synergy weigh scores (LSSWHS01/HS02 – LSSWHS11 = △LSSW).
Live-imaging and cell death assay
Cells were seeded in a 384-well CellBIND (Corning) at 1,000 cells/well with phenol red-free growth media and incubated overnight (~12-14h) at 37°C, 5% CO2. The cells were then treated with drug treatments at the concentrations listed as well as Caspase 3/7 Green Apoptosis Assay Reagent (Sartorius) at a 1:1000 dilution (n=4 wells/treatment). Cells were imaged using the Incucyte S3 live imaging system (Sartorius). Phase and green contrast images were captured every 4h for 72h. Images were analyzed using the integrated software. Primary human VS cells were seeded in 384-well CellBIND plates (Corning) at 1,000 cells/well with Schwann media and incubated overnight at 37°C and 5% CO2. Cells were then treated with 0.05% dimethyl sulfoxide (DMSO; control condition), omipalisib (2, 4, 8nM), dasatinib (200, 400, 800nM), or the combination in maintenance media consisting of Dulbecco’s modified Eagle medium (DMEM) supplemented with 10% fetal bovine serum (Seradigm) and 1% penicillin-streptomycin (n=6 per condition). At 96 hrs, the ApoTox-Glo (Promega) assay was used to detect live cell protease activity for viability and cleaved caspase-3/7 activity for apoptosis using the Glomax Discover System (Promega).
Cell cycle assay
HS01 cells were grown in a six-well CellBIND dish at 250,000 cells/well and allowed to incubate overnight (~12-14h) at 37°C, 5% CO2. Cells were then treated with drugs at the doses listed, and incubated for 24h at 37°C, 5% CO2. EdU (Click-iT™ EdU Cell Proliferation Kit, Molecular Probes, ThermoFisher) was added at 10μM to each well and incubated for 3h. Cells were lifted using 0.05% Trypsin-EDTA (Gibco) and stained with LIVE/DEAD™ Fixable Violet Cell Stain (ThermoFisher). Following staining, cells were fixed, permeabilized, and treated with the Click-iT™ reaction cocktail according to the manufacturer’s instructions. Cells were then counterstained with FxCycle Red stain and analyzed using a CytoFLEX S flow cytometer (Beckman Coulter) and CytExpert software (Beckman Coulter).
Membrane asymmetry assay
HS01 cells were grown in a six-well CellBIND dish at 250,000 cells/well and allowed to incubate overnight (~12-14h) at 37°C, 5% CO2. Cells were then treated with drugs at the doses listed, and then incubated for 72h at 37°C, 5% CO2. Cells were lifted using 0.05% Trypsin-EDTA (Gibco) and stained with Violet Ratiometric Asymmetry assay per the manufacturer’s instructions. Cells were analyzed using a CytoFLEX S flow cytometer (Beckman Coulter) and CytExpert software (Beckman Coulter).
Western blots
Protein extraction was conducted using a modified RIPA buffer (4% sodium dodecyl sulfate, 0.01% bromophenol blue, 10% glycerol, 100mmol/L dithiothreitol). Blots were incubated overnight at 4°C in a 1:1 mixture of tris buffered saline (TBS) and Odyssey blocking buffer (LI-COR Biosciences). The following antibodies were used: rabbit anti-pMLKL #17-10400 from Millipore Sigma, rabbit anti-pFAK(Y577/576) #44614G from Invitrogen, mouse anti-FAK #610087 from BD Biosciences, and rabbit anti-pAKT(T308) #2965, mouse anti-panAKT #2920, rabbit anti-pAKT(S473) #4060, rabbit anti-Src #2109, rabbit anti-pSRC(Y527) #2105, rabbit anti-EphA2 #6997, rabbit anti-pEphA2(S897) #6347, rabbit anti-RIP #3493, rabbit anti-cyclinD1 #2922, rabbit anti-p21(Waf1/Cip1) #2947 from Cell Signaling Technology. Antibodies were diluted 1:1000 unless otherwise stated. The following day, blots were incubated in a 1:1 mixture of 0.1%Tween and 0.02%SDS in TBS (TBS-TS) and Odyssey blocking buffer for 1 hour. Blots were imaged and quantified using the LI-COR Odyssey Imaging System (LI-COR Biosciences, n=3 biological replicates).
Orthotopic allograft schwannoma model
The mouse orthotopic allograft schwannoma model was generated using male and female NSG mice bred in house at 10-11 weeks of age. Care and use were approved by the University of Central Florida (UCF) Institutional Animal Care and Usage Committee (IACUC; #19-08). 5,000 luciferase-expressing merlin-deficient SC were injected into the right sciatic nerve of 10-week-old NSG mice as previously described (18). Mice were divided into treatment or control groups at 8 days post injection. Two mice failed to show sufficient tumor engraftment at this time and were excluded from the study. Control group (n=4) was treated with daily oral gavage of 1.25% PEG, 2.5% Tween80, and 5% DMSO. Mice in the treatment group (n=5) received oral daily gavage of omipalisib at 2mg/kg for the 1st week and 1mg/kg for the 2nd week, and dasatinib at 20 mg/kg. The dosage of omipalisib administered was lowered due to weight loss in one mouse that was subsequently euthanized and excluded from the study. Tumor radiance was measured at 8-, 15, and 22-days post-injection using the In Vivo Imaging System (IVIS, Caliper). All mice were euthanized after 14 days of treatment.
Statistical analysis
Significance was determined using GraphPad Prism Version 7.04 or SAS Studio (9.3M2). Two-way analysis of variance (ANOVA) and Bonferroni’s multiple comparisons post-test was used to assess treatment groups for cell cycle assay and IVIS radiance fold change of allograft tumors. A one-way ANOVA with Dunnett’s multiple comparisons post-test was used to assess significance of treatment groups compared to DMSO in western blots. Tumor weight significance between the treatment and control group was assessed using an unpaired t-test. ANOVA with Tukey post-hoc testing and Bonferroni adjustment was used to analyze viability and cleaved caspase-3/7 activity. To compare the relationship of combination therapies to monotherapies in individual primary VS cultures, 95% confidence intervals were used. Pearson’s coefficient was utilized to determine the strength and the direction of the relationship between phospho-protein expression and viability.
Data Availability
The data generated in this study are included as supplemental data files or will be made available upon request.
Results
Omipalisib and Dasatinib Synergize to Reduce Human Schwannoma Cell Viability
We tested whether co-inhibition of PI3K and FAK/SFK reduced viability of human schwannoma model cells. We selected six PI3K/mTOR and seven FAK/SFK pathway inhibitors to screen in 10x10 dose response combination matrices using human and mouse schwannoma model cell lines. Inhibitors were either FDA approved, in clinical trials, or in advanced pre-clinical development (Table S1). The initial combination screens were conducted in two human schwannoma model cells, HS01 and HS02. Biological interactions were determined using Loewe Additivity and Highest Single Agent (HSA) standard reference models. The top three combinations were assessed for selectivity in the isogenic human WT-SC line (HS11; Table S2) and screened in two additional human (HS03, HS04.3) and one mouse (MS01) schwannoma model cell lines (Table S3).
The most synergistic inhibitor combination was the PI3K/mTOR inhibitor, omipalisib with Src/Abl inhibitor, dasatinib (LSSW = 49.4 and 55.8 for HS01 and HS02, respectively; Table 1, Fig. 1A). The GI50 for omipalisib and dasatinib alone in HS01 cells were 50-80nM and >10μM, respectively (Table 1). The synergistic doses were 39-156nM for omipalisib and 156-625nM for dasatinib (Fig. 1A). At the lowest doses tested (39nM omipalisib and 156nM dasatinib), the combination produced a 44% and 49% reduction in cell viability in HS01 and HS02 cells, respectively. The combination was selective for merlin-deficient human Schwann cells over isogenic normal Schwann cells (ΔLSSW = 23.8 and 29.4 for HS01 and HS02, respectively). The low synergism scores in the HS03 and HS04.3 cells could be attributed to their immortalization with CDK4/6 and TERT. The combination was also effective in the mouse schwannoma model cell line later used in the in vivo study (MS01; Table S3).
Table 1.
Top 10 Synergic PI3K/mTOR and FAK/SRC Combinations Reducing Viability of Human Schwannoma Model Cells
| Schwannoma Model Human Cell Line | HS01 | HS02 | ||||||
|---|---|---|---|---|---|---|---|---|
| Ranking | Drug Name |
Target | GI50 (μM) |
Synergy Model | GI50 (μM) |
Synergy Model | ||
| Loewe | HSA | Loewe | HSA | |||||
| 1 | Omipalisib | p110α/β/δ/γ | 0.05 | 49.9 | 54.9 | 0.08 | 55.7 | 55.9 |
| Dasatinib | Abl/Src/c-Kit | >10 | >10 | |||||
| 2 | Pictilisib | p110α/δ | >10 | 46.1 | 49.5 | >10 | 52.9 | 53.7 |
| Dasatinib | Abl/Src/c-Kit | >10 | >10 | |||||
| 3 | Gedatolisib | p110α/γ/mTOR | 0.08 | 35.3 | 38.4 | 0.09 | 19.6 | 21.8 |
| PND-1186 | FAK | >10 | >10 | |||||
| 4 | Buparlisib | p110α/β/δ/γ | 4.3 | 33.6 | 38.1 | 4.9 | 20 | 23.8 |
| PND-1186 | FAK | >10 | >10 | |||||
| 5 | Omipalisib | p110α/β/δ/γ | 0.02 | 28.3 | 29.2 | 0.02 | 27.6 | 28.9 |
| PND-1186 | FAK | >10 | >10 | |||||
| 6 | Buparlisib | p110α/β/δ/γ | 5.3 | 27.7 | 32.9 | 3.6 | 25.4 | 27.6 |
| Dasatinib | Abl/Src/c-Kit | >10 | >10 | |||||
| 7 | Omipalisib | p110α/β/δ/γ | 0.3 | 26.1 | 29.6 | 0.33 | 15 | 21.6 |
| TAE226 | FAK | 1 | 2.1 | |||||
| 8 | Pictilisib | p110α/δ | 3 | 25.7 | 30 | >10 | 15.2 | 19.2 |
| PF-00562271 | FAK | >10 | 6.5 | |||||
| 9 | Voxtalisib | p110γ/mTOR | >10 | 23.8 | 26.7 | >10 | 21.7 | 25.15 |
| PND-1186 | FAK | >10 | >10 | |||||
| 10 | Omipalisib | p110α/β/δ/γ | 0.02 | 22.8 | 26.5 | 0.01 | 16.5 | 19.9 |
| PF-00562271 | FAK | 2.2 | 3.8 | |||||
Figure 1. Low Doses of Omipalisib and Dasatinib Synergize to Reduce Cell viability, AKT and FAK/SFK phosphorylation in HS01 cells.
A) Drug-combination matrix of two human MD-SC lines, HS01, HS02. Left matrices are viability results expressed as % of DMSO control. Right panels show synergy matrix analyses, Loewe additivity and HSA models respectively. B) Individual dose responses from the single-point viability matrix assay. C) Western blots of whole cell lysates of HS01 cells treated for 24h as indicated. Graphs of phospho-targets are shown relative to loading control. One-way ANOVA with Dunnett’s multiple comparison's posttest. *p<0.05, **p<0.005, ***p<0.0005, ****p<0.00001, n=3.
We verified that omipalisib (4 and 16nM) and dasatinib (100 and 400nM) inhibited phosphorylation of their expected targets in HS01 cells both alone and in combination after 24 hours of treatment. Phospho-AKT (T308 and S473) decreased by 91-99% at both doses in HS01 cells treated with omipalisib alone and in combination with dasatinib (Fig. 1C). Dose-dependent decreases in phosphorylated FAK-Tyr576 and Src-Tyr527 were observed in HS01 cells treated with dasatinib alone and in combination confirming inhibition of Src/FAK (Fig. 1C). We observed that omipalisib alone increased phosphorylated Src-Tyr527 levels compared to vehicle in HS01 cells (Fig. 1C). Dasatinib produced a small decrease in total and phosphorylated EphA2-Ser897, but the reduction was not superior to omipalisib alone (Fig. S1A, S1B).
Omipalisib and Dasatinib Promote a G1 Cell Cycle Arrest in Human Schwannoma Model Cells
Cell cycle assays analyzed by flow cytometry indicated that the percentage of HS01 cells arrested in G1 of the cell cycle was higher in cultures treated with the combination than with either single drug at the same concentrations (Fig. 2A, 2B). Cyclin D1 levels were reduced in omipalisib, and combination treated cells, consistent with a G1 cell cycle arrest. A concomitant increase in p21 levels was not observed (Fig. 2C). In dasatinib treated cells, cyclin D1 levels remained consistent but an increase in p21 was observed. To further refine the combination doses, we selected sub-GI50 concentrations of each drug that synergized to reduce HS01 viability by 50% but would be ineffective alone. We tested omipalisib at 4, 8, and 16nM and dasatinib at 100, 200, and 400nM as monotherapies and in combination, respectively. Results of 72h live imaging confirmed that the combination of omipalisib and dasatinib at all doses was superior to either single agent in blocking cell proliferation as assessed by well confluence, indicating a significant cytostatic effect of the combination. Cleaved caspase-3/7 was not observed in any condition (Fig. 2D), nor was significant apoptosis or cell death observed by Violet Ratiometric flow cytometry assay (Fig. S2). Additionally, no significant modulation of necroptosis markers such as phosphorylated mixed-lineage kinase domain-like pseudokinase (pMLKL) or receptor-interacting serine/threonine protein kinase 1 (RIPK1) were observed in any condition in HS01 cells (Fig. S1). Based on this evidence, the combination of omipalisib and dasatinib produces a significant cytostatic, but not cytotoxic response in the human schwannoma model cells.
Figure 2. Omipalisib and Dasatinib in Combination Inhibit Cell Proliferation but Do Not Induce Cell Death.
A) Flow cytometry assay of EdU incorporation in HS01 cells treated for 24h with 0.1 μM omipalisib and dasatinib alone and in combination. B) Plot shows mean +/− SEM of 3 independent experiments. **p<0.01 and ****p<0.0001 determined by two-way ANOVA and Bonferroni’s multiple comparisons post-test. C) Western blots of whole cell lysates of HS01 cells treated for 24h with indicated drug (0.1 μM). D) Representative images of cell confluence (phase contrast, shown in yellow) and apoptosis (green object count, shown in pink) using live-cell imaging with indicated treatments E) Time course graphs of cell confluence and apoptosis over 72h (mean ±SEM, n=4). Untreated and DMSO treated cells initiate apoptosis at high cell density.
Inhibitor Combination Slows Intraneural Growth of Mouse Schwannoma Models Cells
We conducted a pilot in vivo drug efficacy study of the combination using an orthotopic nerve allograft schwannoma model in NSG immune-deficient mice (Fig. 3). After the 2-week regimen, the average weight of the combination-treated allografts (46.8 mg), was reduced by 61% when compared to the average weight of the vehicle treated mice (118.7 mg; p=0.04). When assessing the fold increase in radiance of the allografts over time, the combination treated mice had a 44% reduction in radiance when compared to the vehicle (p=0.09). The in vivo study had several limitations, such as small group number further exacerbated by the loss of two mice (one per group, not related to drug toxicity). This finding supports conducting a larger study to assess superiority of the combination over the single agents.
Figure 3. Pilot in vivo Allograft Study Demonstrates Efficacy of Drug Combination.
A) Bioluminescent signals (IVIS) were measured at indicated times beginning 7 days post implantation of mouse merlin-deficient SC (MS01) expressing luciferase into the right sciatic nerve of NSG mice. B) Tumor weights at conclusion of the study (mean ±SD). Significance assessed by an unpaired t-test. C) Mean fold increase in radiance in individual animals (mean ±SEM). Statistical significance determined using two-way ANOVA with Bonferroni’s multiple comparisons post-test.
Combination Therapy is More Effective at Reducing Viability of Primary Vestibular Schwannoma Cells than Omipalisib and Dasatinib Monotherapies
Multiplex ligation-dependent probe amplification analysis (MLPA) and whole exon sequencing (WES) was performed on blood leukocytes and tumors from six patients undergoing surgery for VS. Genetic testing revealed that all VS tumors contained at least one mutant NF2 allele and no LTZR1 or SMARCB1 gene mutations (Table S4). Cell viability and cleaved caspase-3/7 dependent apoptosis were assessed in the primary VS cells treated with omipalisib (4, 8, and 16nM) and dasatinib (200, 400, and 800nM) alone and in combination. Because clinical trials testing therapies for NF2-associated VS typically use a reduction of ≥20% as a threshold for response, we used this standard to assess drug response in our viability assays. When drug response was analyzed by group, the mean fold change (MFC) in viability and cleaved caspase-3/7 of all VS tumors showed that omipalisib (16nM) and dasatinib (200, 400, and 800nM) monotherapies initiated significant reductions in viability ≥20%. Overall, dasatinib was more effective than omipalisib at the monotherapy concentrations tested. (Fig. 4A). The combination therapies induced a more robust reduction in MFCs in viability at the three doses tested compared to the monotherapies alone. The highest combination dose (16nM omipalisib + 800nM dasatinib) promoted a substantial induction of cleaved caspase-3/7 as well. An equivalent increase in cleaved caspase-3/7 was not observed with either monotherapy. (Fig. 4A). When drug responses were analyzed in individual VS, we observed that each VS demonstrated greater decreases in viability with dasatinib than with omipalisib monotherapy (Fig. 4B and 4C).
Figure 4. Cell Viability and Caspase 3/7-Dependent Apoptosis in Primary Human Vestibular Schwannoma (VS) Cells Treated with Omipalisib and Dasatinib.
A) Viability and cleaved caspase-3/7 assays at 96hrs. Mean Fold Changes (MFC) were normalized to vehicle (0.05% DMSO). B) Viability for six VS samples at 96hrs. Results expressed as fold change compared to DMSO control. C) Cleaved Caspase-3/7 for six VS samples at 96hrs. Results expressed as change (△) in relative luminescence units (RLU). Line represents mean. Error bars represent standard error mean. *p<0.05. **p<0.01. ***p<0.001. n=6 replicates for each experiment.
In 3 of 6 primary VS (VSA21, VSA23, VSA26), combination therapies initiated significantly greater reductions in viability compared to their equivalent monotherapies (p<0.05). Combination therapy at the highest dose caused a detectable increase in cleaved caspase-3/7 in VSA18, VSA19, and VSA21. Western blots were conducted using protein extracts from untreated tumor chunks for 5 of 6 VS (Fig. 5); there was insufficient sample to assess VSA26. VS tumors expressed p-EphA2, p-FAK, p-AKT, and p-SRC at varying levels. There were inverse relationships between target protein expression (i.e., p-EphA2, p-Src, p-AKT and p-FAK) and cell viability in response to omipalisib and dasatinib monotherapies (Fig. S3); however, the relationship did not meet statistical significance with Pearson’s correlation coefficient.
Figure 5. Western blots for targeted pathway kinases in untreated vestibular schwannomas tumors.
Phospho-protein levels were normalized to total actin expression.
Discussion
The findings confirm results of single and combination high-throughput compound screens that identified PI3K and FAK/SFK as potential co-targets for reducing schwannoma cell viability (13, 15, 16). Omipalisib and dasatinib emerged as the most synergistic combination using multiple human and mouse schwannoma model cell lines and stringent advancement criteria. Omipalisib and dasatinib synergized at sub-GI50 concentrations and had a high selectivity for merlin-deficient schwannoma model cells over their isogenic normal Schwann cells. At sub-GI50 concentrations, the combination of omipalisib and dasatinib reduced cell confluence when compared to either drug at the same dose and decreased phosphorylation of their respective targets at 24h of treatment. Dasatinib at sub-maximal doses has been used successfully in multiple clinical trials for treatment of chronic myeloid leukemia (23, 24). Combination therapy with a drug cocktail that synergizes to produce disease control at lower doses than a monotherapy is becoming the norm for various conditions including melanoma and breast cancer, hypertension, and HIV infection (25, 26). By administering a combination of drugs at doses below the maximum tolerable dose (MTD), the risk of developing drug resistance or adverse effects seen with higher doses of monotherapies are reduced or eliminated (27-29). This study demonstrates the efficacy of this concept in human schwannoma model cells and supports further research into the use of low dose combination treatments targeting these pathways.
The combination therapy completely inhibited proliferation by promoting a strong G1 cell cycle arrest but did not induce cell death in model schwannoma cells. Thus, omipalisib and dasatinib have a cytostatic effect on the model cells rather than a cytotoxic effect as seen with CUDC907 (21). Emerging research highlights the advantages of both therapeutic strategies, and currently outlines parameters in which one strategy might be superior to the other (30). The pilot mouse schwannoma allograft model demonstrated that a two-week treatment with the combination reduced intraneural growth of mouse schwannoma model cells (MS01). Despite limitations due to small sample size, the average increase in graft size by weight for the omipalisib and dasatinib treated mice was 61% less than those from vehicle treated mice. In patient VS samples, the combination of omipalisib and dasatinib was superior to single drugs in three of the six VS, depending on the dose, whereas dasatinib alone performed statistically as well as the combination in the remaining VS samples. Overall, a greater number of VS responded to dasatinib alone than to omipalisib alone. Cleaved caspase 3/7 was detected in three of 6 VS samples treated with the combination at the highest dose. Caspase 3/7 activation was not observed in the human schwannoma model cells. This inconsistency between primary VS cells and the schwannoma model cells could be due to several reasons including: 1) the pathogenic NF2 gene mutations in VS cells compared to stable shRNAi knockdown of merlin in model cells, and 2) possible cellular stress pathways present in freshly isolated primary VS cells compared to culture-adapted schwannoma model cells (31).
Further investigations are warranted to determine if target phospho-protein levels can be used as reliable biomarkers for drug response and to determine how tumor heterogeneity impacts effectiveness of drug treatment. These results and the outcomes of NF2 clinical trials thus far supports development of patient-specific drug screening (18-21, 32). Previously, this lab conducted a small proof-of-principle drug study using an immortalized schwannoma cell line from a schwannomatosis patient with a germline SMARCB1 mutation (33). Generation of a patient-derived xenograft was not feasible due to slow in vivo growth but indicates that personalized models can be developed as is increasingly done for breast, prostate, and colorectal cancers (34-37).
Despite promising preclinical results, PI3K inhibitors have had limited success in clinical trials. We found that low dose omipalisib alone increased pTyr527-Src levels compared to DMSO control in model schwannoma cells, confirming our previous kinome results. This cautions against using PI3K inhibitors alone for NF2-Schwannomatosis and highlights how quickly human schwannoma cells activate compensatory cell survival kinases. Despite setbacks, the FDA has approved several PI3K inhibitors; some only when used in combination with other inhibitors or after relapse with another therapy (38). Omipalisib has high potency for all PI3K isoforms and mTOR, due to the strong interactions of a charged sulfonamide with conserved lysine residues in all p110 isoforms. It has been evaluated for pulmonary fibrosis (NCT01725139) and solid tumors (NCT00972686) but appears to no longer be under development. Dasatinib is FDA approved for use in some patients with Philadelphia chromosome-positive acute lymphoblastic leukemia and chronic myeloid leukemia. It is well tolerated and is in numerous clinical trials alone and in combination including with crizotinib (NCT01744652, NCT01644773) and erlotinib (NCT00444015, NCT00826449). We assessed the efficacy of dasatinib and saracatinib, another Src inhibitor, in combination with FDA-approved c-MET inhibitor cabozantinib (20). The combination of saracatinib and cabozantinib promoted apoptosis of mouse schwannoma cells and reduced allograft growth by 85% (20).
Although omipalisib and dasatinib as a combination therapy may not be translatable into the clinic due to toxicity, the results indicate that co-targeting FAK/SFK and PI3K is a valid strategy for NF2 schwannomas. FAK is an important cancer target and plays essential roles in SC development and has been shown to be a viable target for NF2 schwannomas (16, 39-44). FAK overexpression and increased integrin/FAK/Src/Ras signaling was reported in human schwannoma cells and contributed to cell proliferation (43). In a study of ALK- and MET-inhibitor crizotinib, FAK1 was identified as a critical effector for the drug’s mechanism of action in NF2 mouse and human schwannoma model cells (44). In a subsequent study, brigatinib, a second generation ALK inhibitor, and dasatinib were identified as one of three top performing drug combinations tested in human schwannoma cells. This combination was not as effective as brigatinib monotherapy when tested in Postn-Cre; Nf2flox/flox mice. However, this could be due to the reduced dose of brigatinib in the combination compared to the monotherapy as a result of toxicity (16). Brigatinib was found to inhibit FAK1/2, as well as FER and TNK in the human schwannoma model cells used here. Both crizotinib and brigatinib are currently in clinical trials for treatment of NF2 (NCT04283669 and NCT04374305, respectively.
PI3K is activated downstream of multiple RTKs that drive Schwann and schwannoma cell proliferation. These include: ErbB2/3, platelet-derived growth factor receptor, and c-MET (45, 46). In addition to the PI3K pathway, activation of RTKs triggers signaling from multiple parallel pathways including RAS-MAPK and SFK signaling. Thus, inhibitors of RTKs are expected to have more pleiotropic effects than inhibitors that block further downstream such as at the level of PI3K/AKT/mTOR. Indeed, we reported in mouse schwannoma models cells, that the c-MET inhibitor, cabozantinib reduced phosphorylation of the receptor as well as ERK and AKT (19). Integrin-dependent adhesion to ECM plays a critical role in SC development and is elevated in schwannomas (10). Cooperative signaling between RTK and ß1 integrins occurs in Schwann cells (10). This could explain why FAK/SFK activation occurs so readily when PI3K is inhibited and serves to block anoikis in adhesion-dependent schwannoma cells. Indeed, in silico studies revealed that among cancer cell types, those with loss of merlin are particularly sensitive to FAK inhibitors (47). Overall, a strategy that employs a monotherapy with broad downstream effects, such as brigatinib currently in phase 2 trials for NF2 patients or a dual inhibitor such as fimepinostat, that targets PI3K as well as histone deacetylase with expected broad transcriptional effects, may be preferable to highly specific mono-kinase inhibitors.
Supplementary Material
Acknowledgements
We thank Alka Mehta, Rosa Rosario, Berta Victoria, and the University of Central Florida vivarium staff for experimental support.
Funding
National Institutes of Health [R56NS102254 & R01DC017264 to C.F-V and X-Z.L., K08DC017508 to C.T.D]; Sylvester Comprehensive Cancer Center [National Institutes of Health K08DC017508 supplement to C.T.D]. This work was supported in part by the NIH/NCI Cancer Center Support Grant P30 CA008748 to Memorial Sloan Kettering Cancer Center.
Footnotes
Conflict of Interest Statement
The authors declare no potential conflicts of interest
References
- 1.Plotkin SR, Messiaen L, Legius E, Pancza P, Avery RA, Blakeley JO, et al. Updated diagnostic criteria and nomenclature for neurofibromatosis type 2 and schwannomatosis: An international consensus recommendation. Genetics in Medicine. 2022;24(9):1967–77. [DOI] [PubMed] [Google Scholar]
- 2.Rouleau GA, Merel P, Lutchman M, Sanson M, Zucman J, Marineau C, et al. Alteration in a new gene encoding a putative membrane-organizing protein causes neuro-fibromatosis type 2. Nature. 1993;363(6429):515–21. [DOI] [PubMed] [Google Scholar]
- 3.Trofatter JA, MacCollin MM, Rutter JL, Murrell JR, Duyao MP, Parry DM, et al. A novel moesin-, ezrin-, radixin-like gene is a candidate for the neurofibromatosis 2 tumor suppressor. Cell. 1993;72(5):791–800. [DOI] [PubMed] [Google Scholar]
- 4.Yao L, Alahmari M, Temel Y, Hovinga K. Therapy of Sporadic and NF2-Related Vestibular Schwannoma. Cancers (Basel). 2020;12(4). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Ramesh V. Merlin and the ERM proteins in Schwann cells, neurons and growth cones. Nature Reviews Neuroscience. 2004;5(6):462–70. [DOI] [PubMed] [Google Scholar]
- 6.Petrilli AM, Fernández-Valle C. Role of Merlin/NF2 inactivation in tumor biology. Oncogene. 2016;35(5):537–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Cui Y, Ma L, Schacke S, Yin JC, Hsueh Y-P, Jin H, et al. Merlin cooperates with neurofibromin and Spred1 to suppress the Ras–Erk pathway. Human Molecular Genetics. 2020;29(23):3793–806. [DOI] [PubMed] [Google Scholar]
- 8.Morrison H, Sperka T, Manent J, Giovannini M, Ponta H, Herrlich P. Merlin/Neurofibromatosis Type 2 Suppresses Growth by Inhibiting the Activation of Ras and Rac. Cancer Research. 2007;67(2):520–7. [DOI] [PubMed] [Google Scholar]
- 9.Mindos T, Dun XP, North K, Doddrell RD, Schulz A, Edwards P, et al. Merlin controls the repair capacity of Schwann cells after injury by regulating Hippo/YAP activity. J Cell Biol. 2017;216(2):495–510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Fernandez-Valle C, Tang Y, Ricard J, Rodenas-Ruano A, Taylor A, Hackler E, et al. Paxillin binds schwannomin and regulates its density-dependent localization and effect on cell morphology. Nature Genetics. 2002;31(4):354–62. [DOI] [PubMed] [Google Scholar]
- 11.Sparrow N, Manetti ME, Bott M, Fabianac T, Petrilli A, Bates ML, et al. The Actin-Severing Protein Cofilin Is Downstream of Neuregulin Signaling and Is Essential For Schwann Cell Myelination. The Journal of Neuroscience. 2012;32(15):5284–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Thaxton C, Bott M, Walker B, Sparrow NA, Lambert S, Fernandez-Valle C. Schwannomin/merlin promotes Schwann cell elongation and influences myelin segment length. Mol Cell Neurosci. 2011;47(1):1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Petrilli AM, Fuse MA, Donnan MS, Bott M, Sparrow NA, Tondera D, et al. A chemical biology approach identified PI3K as a potential therapeutic target for neurofibromatosis type 2. Am J Transl Res. 2014;6(5):471–93. [PMC free article] [PubMed] [Google Scholar]
- 14.Knight SD, Adams ND, Burgess JL, Chaudhari AM, Darcy MG, Donatelli CA, et al. Discovery of GSK2126458, a Highly Potent Inhibitor of PI3K and the Mammalian Target of Rapamycin. ACS Medicinal Chemistry Letters. 2010;1(1):39–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Allaway R, Angus SP, Beauchamp RL, Blakeley JO, Bott M, Burns SS, et al. Traditional and systems biology based drug discovery for the rare tumor syndrome neurofibromatosis type 2. PLoS One. 2018;13(6):e0197350. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Chang L-S, Oblinger JL, Smith AE, Ferrer M, Angus SP, Hawley E, et al. Brigatinib causes tumor shrinkage in both NF2-deficient meningioma and schwannoma through inhibition of multiple tyrosine kinases but not ALK. PLOS ONE. 2021;16(7):e0252048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Manetti ME, Geden S, Bott M, Sparrow N, Lambert S, Fernandez-Valle C. Stability of the tumor suppressor merlin depends on its ability to bind paxillin LD3 and associate with β1 integrin and actin at the plasma membrane. Biol Open. 2012;1(10):949–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Fuse MA, Dinh CT, Vitte J, Kirkpatrick J, Mindos T, Plati SK, et al. Preclinical assessment of MEK1/2 inhibitors for neurofibromatosis type 2-associated schwannomas reveals differences in efficacy and drug resistance development. Neuro Oncol. 2019;21(4):486–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Petrilli AM, Garcia J, Bott M, Klingeman Plati S, Dinh CT, Bracho OR, et al. Ponatinib promotes a G1 cell-cycle arrest of merlin/NF2-deficient human schwann cells. Oncotarget. 2017;8(19):31666–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Fuse MA, Plati SK, Burns SS, Dinh CT, Bracho O, Yan D, et al. Combination therapy with c-Met and Src inhibitors induces caspase-dependent apoptosis of merlin-deficient Schwann cells and suppresses growth of schwannoma cells. Molecular cancer therapeutics. 2017;16(11):2387–98. [DOI] [PubMed] [Google Scholar]
- 21.Huegel J, Dinh CT, Martinelli M, Bracho O, Rosario R, Hardin H, et al. CUDC907, a dual phosphoinositide-3 kinase/histone deacetylase inhibitor, promotes apoptosis of NF2 Schwannoma cells. Oncotarget. 2022;13:890–904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Di Veroli GY, Fornari C, Wang D, Mollard S, Bramhall JL, Richards FM, et al. Combenefit: an interactive platform for the analysis and visualization of drug combinations. Bioinformatics. 2016;32(18):2866–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Murai K, Ureshino H, Kumagai T, Tanaka H, Nishiwaki K, Wakita S, et al. Low-dose dasatinib in older patients with chronic myeloid leukaemia in chronic phase (DAVLEC): a single-arm, multicentre, phase 2 trial. The Lancet Haematology. 2021;8(12):e902–e11. [DOI] [PubMed] [Google Scholar]
- 24.Naqvi K, Jabbour E, Skinner J, Anderson K, Dellasala S, Yilmaz M, et al. Long-term follow-up of lower dose dasatinib (50 mg daily) as frontline therapy in newly diagnosed chronic-phase chronic myeloid leukemia. Cancer. 2020;126(1):67–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Satti J. The emerging low-dose therapy for advanced cancers. Dose Response. 2009;7(3):208–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Kamat AA, Kim TJ, Landen CN Jr., Lu C, Han LY, Lin YG, et al. Metronomic chemotherapy enhances the efficacy of antivascular therapy in ovarian cancer. Cancer Res. 2007;67(1):281–8. [DOI] [PubMed] [Google Scholar]
- 27.Hryniuk W, Bush H. The importance of dose intensity in chemotherapy of metastatic breast cancer. J Clin Oncol. 1984;2(11):1281–8. [DOI] [PubMed] [Google Scholar]
- 28.Kaplan LD, Straus DJ, Testa MA, Von Roenn J, Dezube BJ, Cooley TP, et al. Low-dose compared with standard-dose m-BACOD chemotherapy for non-Hodgkin's lymphoma associated with human immunodeficiency virus infection. National Institute of Allergy and Infectious Diseases AIDS Clinical Trials Group. N Engl J Med. 1997;336(23):1641–8. [DOI] [PubMed] [Google Scholar]
- 29.Munker S, Gerken M, Fest P, Ott C, Schnoy E, Fichtner-Feigl S, et al. Chemotherapy for metastatic colon cancer: No effect on survival when the dose is reduced due to side effects. BMC Cancer. 2018;18(1):455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Anttila JV, Shubin M, Cairns J, Borse F, Guo Q, Mononen T, et al. Contrasting the impact of cytotoxic and cytostatic drug therapies on tumour progression. PLoS Comput Biol. 2019;15(11):e1007493. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Evans DG. Neurofibromatosis type 2 (NF2): a clinical and molecular review. Orphanet J Rare Dis. 2009;4:16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Misztal C, Bracho O, Bas E, Estivill M, Ivan ME, Morcos J, et al. Effect of AR42 in Primary Vestibular Schwannoma Cells and a Xenograft Model of Vestibular Schwannoma. Otol Neurotol. 2022;43(6):694–701. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Allaf A, Victoria B, Rosario R, Misztal C, Humayun Gultekin S, Dinh CT, et al. WP1066 induces cell death in a schwannomatosis patient-derived schwannoma cell line. Cold Spring Harb Mol Case Stud. 2022;8(4). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Zanella ER, Grassi E, Trusolino L. Towards precision oncology with patient-derived xenografts. Nat Rev Clin Oncol. 2022;19(11):719–32. [DOI] [PubMed] [Google Scholar]
- 35.Yang YS, Liu CY, Wen D, Gao DZ, Lin S, He HF, et al. Recent advances in the development of transplanted colorectal cancer mouse models. Transl Res. 2022;249:128–43. [DOI] [PubMed] [Google Scholar]
- 36.Genta S, Coburn B, Cescon DW, Spreafico A. Patient-derived cancer models: Valuable platforms for anticancer drug testing. Front Oncol. 2022;12:976065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Adamiecki R, Hryniewicz-Jankowska A, Ortiz MA, Li X, Porter-Hansen BA, Nsouli I, et al. In Vivo Models for Prostate Cancer Research. Cancers (Basel). 2022;14(21). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Peng Y, Wang Y, Zhou C, Mei W, Zeng C. PI3K/Akt/mTOR Pathway and Its Role in Cancer Therapeutics: Are We Making Headway? Front Oncol. 2022;12:819128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Wu Y, Li N, Ye C, Jiang X, Luo H, Zhang B, et al. Focal adhesion kinase inhibitors, a heavy punch to cancer. Discov Oncol. 2021;12(1):52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Fernandez-Valle C, Wood PM, Bunge MB. Localization of focal adhesion kinase in differentiating Schwann cell/neuron cultures. Microsc Res Tech. 1998;41(5):416–30. [DOI] [PubMed] [Google Scholar]
- 41.Chen LM, Bailey D, Fernandez-Valle C. Association of beta 1 integrin with focal adhesion kinase and paxillin in differentiating Schwann cells. J Neurosci. 2000;20(10):3776–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Grove M, Brophy PJ. FAK is required for Schwann cell spreading on immature basal lamina to coordinate the radial sorting of peripheral axons with myelination. J Neurosci. 2014;34(40):13422–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Ammoun S, Flaiz C, Ristic N, Schuldt J, Hanemann CO. Dissecting and targeting the growth factor-dependent and growth factor-independent extracellular signal-regulated kinase pathway in human schwannoma. Cancer Res. 2008;68(13):5236–45. [DOI] [PubMed] [Google Scholar]
- 44.Troutman S, Moleirinho S, Kota S, Nettles K, Fallahi M, Johnson GL, et al. Crizotinib inhibits NF2-associated schwannoma through inhibition of focal adhesion kinase 1. Oncotarget. 2016;7(34):54515–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Dilwali S, Roberts D, Stankovic KM. Interplay between VEGF-A and cMET signaling in human vestibular schwannomas and schwann cells. Cancer Biol Ther. 2015;16(1):170–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Lallemand D, Manent J, Couvelard A, Watilliaux A, Siena M, Chareyre F, et al. Merlin regulates transmembrane receptor accumulation and signaling at the plasma membrane in primary mouse Schwann cells and in human schwannomas. Oncogene. 2009;28(6):854–65. [DOI] [PubMed] [Google Scholar]
- 47.Shapiro IM, Kolev VN, Vidal CM, Kadariya Y, Ring JE, Wright Q, et al. Merlin deficiency predicts FAK inhibitor sensitivity: a synthetic lethal relationship. Sci Transl Med. 2014;6(237):237ra68. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
The data generated in this study are included as supplemental data files or will be made available upon request.





