Nature Neuroscience: doi: /nn Supplementary Figure 1

Similar documents
Supplementary Figure 1

Supplementary Figure 1. Basal level EGFR across a panel of ESCC lines. Immunoblots demonstrate the expression of phosphorylated and total EGFR as

Supplementary Information

- 1 - Cell types Monocytes THP-1 cells Macrophages. LPS Treatment time (Hour) IL-6 level (pg/ml)

Supplementary fig. 1. Crystals induce necroptosis does not involve caspases, TNF receptor or NLRP3. A. Mouse tubular epithelial cells were pretreated

Supplementary Fig. 1. GPRC5A post-transcriptionally down-regulates EGFR expression. (a) Plot of the changes in steady state mrna levels versus

Supplementary Fig. 1 p38 MAPK negatively regulates DC differentiation. (a) Western blot analysis of p38 isoform expression in BM cells, immature DCs

SUPPLEMENTARY INFORMATION

SUPPLEMENTARY FIGURES AND TABLES

Supplementary Figure S I: Effects of D4F on body weight and serum lipids in apoe -/- mice.

Title:Role of LPAR3, PKC and EGFR in LPA-induced cell migration in oral squamous carcinoma cells

Type of file: PDF Size of file: 0 KB Title of file for HTML: Supplementary Information Description: Supplementary Figures

Figures S1-S5, Figure Legends, Table S1 List of primers used in the study

Supporting Information. FADD regulates NF-кB activation and promotes ubiquitination of cflip L to induce. apoptosis

mtor Inhibition Specifically Sensitizes Colorectal Cancers with KRAS or BRAF Mutations to BCL-2/BCL-

PID1 increases chemotherapy-induced apoptosis in medulloblastoma and glioblastoma cells in a manner that involves NFκB

Programmed necrosis, not apoptosis, is a key mediator of cell loss and DAMP-mediated inflammation in dsrna-induced retinal degeneration

Supplementary Figure 1: Digitoxin induces apoptosis in primary human melanoma cells but not in normal melanocytes, which express lower levels of the

SHREE ET AL, SUPPLEMENTAL MATERIALS. (A) Workflow for tumor cell line derivation and orthotopic implantation.

Supplementary Information

HCC1937 is the HCC1937-pcDNA3 cell line, which was derived from a breast cancer with a mutation

ANGPTL2 increases bone metastasis of breast cancer cells through. Tetsuro Masuda, Motoyoshi Endo, Yutaka Yamamoto, Haruki Odagiri, Tsuyoshi

SUPPLEMENTARY INFORMATION

HEK293FT cells were transiently transfected with reporters, N3-ICD construct and

Figure S1. The PDE5 inhibitor sildenafil interacts with celecoxib to kill cancer cell lines. (A) Hepatoma

Intrinsic and chemo-sensitizing activity of SMAC-mimetics on high-risk childhood acute lymphoblastic leukemia

Supplemental Figure 1. Western blot analysis indicated that MIF was detected in the fractions of

Supplementary Figures

Supplementary Figure 1. A. Bar graph representing the expression levels of the 19 indicated genes in the microarrays analyses comparing human lung

Supplementary Figure 1 (Related with Figure 4). Molecular consequences of Eed deletion. (a) ChIP analysis identifies 3925 genes that are associated

Supplementary Figure 1. Establishment of prostacyclin-secreting hmscs. (a) PCR showed the integration of the COX-1-10aa-PGIS transgene into the

RAW264.7 cells stably expressing control shrna (Con) or GSK3b-specific shrna (sh-

a. b. c. d. e. f. g. h. i. j. k. l. m. n. o. p.

Supplementary Materials for

Supplementary Materials for

Supplementary Figure 1. Expression of CUGBP1 in non-parenchymal liver cells treated with TGF-β

Pharmacologic inhibition of histone demethylation as a therapy for pediatric brainstem glioma

Supplementary Figure 1. IL-12 serum levels and frequency of subsets in FL patients. (A) IL-12

SUPPLEMENTARY FIGURES

Page 39 of 44. 8h LTA & AT h PepG & AT h LTA

Supplementary material. Supplementary Figure legends

X P. Supplementary Figure 1. Nature Medicine: doi: /nm Nilotinib LSK LT-HSC. Cytoplasm. Cytoplasm. Nucleus. Nucleus

L1 on PyMT tumor cells but Py117 cells are more responsive to IFN-γ. (A) Flow

Supplementary Information

Supplementary Figure 1. Repression of hepcidin expression in the liver of mice treated with

Supplementary Figure 1: si-craf but not si-braf sensitizes tumor cells to radiation.

Supplementary Materials

ERK1/2/MAPK pathway-dependent regulation of the telomeric factor TRF2

Supplementary Figure 1. BMS enhances human T cell activation in vitro in a

Supplementary Figure 1 IMQ-Induced Mouse Model of Psoriasis. IMQ cream was

Supplementary Figure 1: Tissue of Origin analysis on 152 cell lines. (a) Heatmap representation of the 30 Tissue scores for the 152 cell lines.

Supplementary Figure 1:

Fang et al. NMuMG. PyVmT unstained Anti-CCR2-PE MDA-MB MCF MCF10A

Supplementary Appendix

Complexity of intra- and inter-pathway loops in colon cancer and melanoma: implications for targeted therapies

SUPPLEMENTARY INFORMATION. Involvement of IL-21 in the epidermal hyperplasia of psoriasis

A263 A352 A204. Pan CK. pstat STAT3 pstat3 STAT3 pstat3. Columns Columns 1-6 Positive control. Omentum. Rectosigmoid A195.

Supplementary Figure 1. mrna targets were found in exosomes and absent in free-floating supernatant. Serum exosomes and exosome-free supernatant were

Nature Medicine: doi: /nm.4078

Supplementary Figure 1. IDH1 and IDH2 mutation site sequences on WHO grade III

(A) RT-PCR for components of the Shh/Gli pathway in normal fetus cell (MRC-5) and a

Supplementary Information

Bmi-1 regulates stem cell-like properties of gastric cancer cells via modulating mirnas

Supplementary Figure 1.TRIM33 binds β-catenin in the nucleus. a & b, Co-IP of endogenous TRIM33 with β-catenin in HT-29 cells (a) and HEK 293T cells

Supplementary Figures

Supplemental Figures:

SUPPLEMENTARY INFORMATION

SUPPLEMENTARY INFORMATION

Supplementary Materials for

Integrin CD11b negatively regulates TLR-triggered inflammatory responses by. activating Syk and promoting MyD88 and TRIF degradation via cbl-b

Intracellular MHC class II molecules promote TLR-triggered innate. immune responses by maintaining Btk activation

Supplementary Figures

Supplementary Figure 1. Deletion of Smad3 prevents B16F10 melanoma invasion and metastasis in a mouse s.c. tumor model.

Doctoral Degree Program in Marine Biotechnology, College of Marine Sciences, Doctoral Degree Program in Marine Biotechnology, Academia Sinica, Taipei,

p47 negatively regulates IKK activation by inducing the lysosomal degradation of polyubiquitinated NEMO

EPIGENETIC RE-EXPRESSION OF HIF-2α SUPPRESSES SOFT TISSUE SARCOMA GROWTH

Supplementary Figure 1 ITGB1 and ITGA11 increase with evidence for heterodimers following HSC activation. (a) Time course of rat HSC activation

Effective Targeting of Quiescent Chronic Myelogenous

Pro-apoptotic signalling through Toll-like receptor 3 involves TRIF-dependent

Supplementary Figure 1. Supernatants electrophoresis from CD14+ and dendritic cells. Supernatants were resolved by SDS-PAGE and stained with

Irf1 fold changes (D) 24h 48h. p-p65. t-p65. p-irf3. t-irf3. β-actin SKO TKO 100% 80% 60% 40% 20%

Supporting Information

Supplementary Data. Supplementary Methods:

Soft Agar Assay. For each cell pool, 100,000 cells were resuspended in 0.35% (w/v)

Supplementary Methods: IGFBP7 Drives Resistance to Epidermal Growth Factor Receptor Tyrosine Kinase Inhibition in Lung Cancer

Supplemental Materials for. Effects of sphingosine-1-phosphate receptor 1 phosphorylation in response to. FTY720 during neuroinflammation

SUPPLEMENTARY INFORMATION

Supplementary Figure S1. PTPN2 levels are not altered in proliferating CD8+ T cells. Lymph node (LN) CD8+ T cells from C57BL/6 mice were stained with

Ephrin receptor A2 is an epithelial cell receptor for Epstein Barr virus entry

Targeted mass spectrometry (LC/MS/MS) for Olaparib pharmacokinetics. For LC/MS/MS of Olaparib pharmacokinetics metabolites were extracted from

Supplemental information

Islet viability assay and Glucose Stimulated Insulin Secretion assay RT-PCR and Western Blot

Supplementary Figure 1. Validation of astrocytes. Primary astrocytes were

Supplementary Figure 1: Expression of NFAT proteins in Nfat2-deleted B cells (a+b) Protein expression of NFAT2 (a) and NFAT1 (b) in isolated splenic

Supplementary Materials for

Supplementary Figures

Antitumor Activity of CUDC-305, a Novel Oral HSP90 Inhibitor, in Solid and Hematological Tumor Xenograft Models

Supplemental Figure 1. Signature gene expression in in vitro differentiated Th0, Th1, Th2, Th17 and Treg cells. (A) Naïve CD4 + T cells were cultured

SUPPLEMENTARY MATERIAL

BRaf V600E cooperates with Pten silencing to elicit metastatic melanoma (Nature Genetics Supplementary Information)

Transcription:

Supplementary Figure 1 EGFR inhibition activates signaling pathways (a-b) EGFR inhibition activates signaling pathways (a) U251EGFR cells were treated with erlotinib (1µM) for the indicated times followed by Western blot with the indicated antibodies. (b) GBM9 neurospheres were treated with afatinib (100nM) for the indicated

time points followed by Western blot with the indicated antibodies. (c-g) EGFR inhibition decreases pstat3 and pakt activation. Glioma cells were treated with high dosage of erlotinib (10µM) or afatinib (10µM). Western blot showing that pstat3 and pakt decrease upon treatment with erlotinib or afatinib. Western blots are representative of at least three independent replicates. (h-j) perk is activated in U87EGFRwt and blocked by cetuximab. (h) U87EGFRwt cells were cultured in medium with (10%) or without serum in DMEM. perk is activated under both conditions. (i) Cells were serum starved and treated overnight with cetuximab (100 µg/ml). Cetuximab treatment blocks both EGFR and ERK activation. (j) Cells were treated wtith EGF (50 ng/ml) for 15 min. EGF increases perk level in U87EGFRwt cells. SF:Serum free. Western blots are representative of at least three independent replicates. Full-length blots are presented in Supplementary Figure 15.

Supplementary Figure 2

Intracellular signaling networks and biological responses to EGFR inhibition in glioma cells. (a-b) EGFR inhibition does not activate NF- B transcriptional activity. (a) A luciferase reporter assay shows that EGFR inhibition with erlotinib does not result in an increase in NF- B reporter activity in GBM9 neurospheres and U87EGFRwt cells. Erlotinib was used for 24h at a concentration of 100 nm for GBM9 neurospheres and 1uM for U87EGFRwt cells. GBM9: Ctrl vs erlotinib: P =0.88, t=0.16, d.f.=4; U87EGFRwt: Ctrl vs erlotinib: P =0.60, t=0.57, d.f.=4. (b) As a positive control we used LPS (1µg/ml) which activates the NF- B reporter. Renilla luciferase was used as an internal control. Ctrl vs LPS: P =0.0012, t=8.27, d.f.=4. Data are presented as mean±s.e.m; **P < 0.01 from two-tailed unpaired Student's t-test ( n = 3 biologically independent experimental replicates). n.s. not significant. (c-d) Upregulation of TNF in response to EGFR inhibition in multiple patient derived samples and established cell lines. (c) SK987, SK748. SK1422, and GBM622 are patient derived primary GBM cells cultured as neurospheres and treated with erlotinib (100nM) for 24h followed by extraction of RNA and qrt-pcr for TNF. SB19vIII and U251EGFRwt cells are established GBM cell lines that were treated with erlotinib (1µM) for 24h followed by qrt-pcr for TNF. SB19vIII: Ctrl vs Erlotinib: P =0.0014,t=7.88, d.f.=4; SK987: Ctrl vs erlotinib: P =0.0024,t=6.83, d.f.=4; SK748: Ctrl vs Erlotinib: P =0.0019, t=7.32, d.f.=4; SK1422: Ctrl vs erlotinib: P =0.0030,t=6.45, d.f.=4; GBM622: Ctrl vs erlotinib: P =0.21,t=1.5, d.f.=4; U251EGFRwt: Ctrl vs erlotinib: P =0.0005, t=10.4, d.f.=4. Data are presented as mean±s.e.m; **P < 0.01, ***P<0.001 from two-tailed unpaired Student's t-test ( n = 3 biologically independent experimental replicates). n.s. not significant. (d) Western blot showing EGFR levels in various neurospheres and cell lines. No increase in TNF is detected in GBM622 cells expressing a very low level of EGFR. Western blots are representative of at least three independent replicates. Fulllength blots are presented in Supplementary Figure 15. (e) Downregulation of TNFR1 in response to EGFR inhibition in patient derived primary GBM neurosphere cultures and established cell lines. GBM9, SK987 and U87EGFRwt cells were treated with erlotinib for the indicated time points followed by Western blot with TNFR1 antibodies. Erlotinib was used at a concentration of 100 nm for GBM9 and SK987 and 1uM for U87EGFRwt cells. Western blots are representative of at least three independent replicates. (f) EGFR inhibition induced effects on cell viability are influenced by specific inhibitors of downstream signaling pathways. In this experiment primary GBM9 neurospheres were exposed to various inhibitors alone or in combination with erlotinib (100 nm) for 72h followed by Alamarblue Cell Viability assay. The concentrations of the various drugs were: SB203850 (10uM), SP600125 (1uM), U0126 (1uM), BMS-345541 (10uM), XL765 (10uM), Necrostatin-1 (300nM). Erlotinib vs erlotinib +SP600125: P =0.0001,t=14.96, d.f.=4; Erlotinib vs erlotinib +U0126: P =0.0018, t=7.42, d.f.=4. Data are presented as mean±s.e.m; **P < 0.01, ***P<0.001 from two-tailed unpaired Student's t- test ( n = 3 biologically independent experimental replicates). (g-i) Western blot showing efficient silencing of JNK1/2, Axl and TNFR1. For cell viability experiments involving sirna knockdown of JNK1/2, Axl and TNFR1, we confirmed silencing by Western blot. Western blots are representative of at least three independent replicates. Full-length blots are presented in Supplementary Figure 15.

Supplementary Figure 3 EGFR inhibition induces a biologically significant upregulation of TNF in glioma cells.

(a-b) Thalidomide blocks EGFR inhibition induced upregulation of TNF. Patient derived GBM9 neurospheres or established GBM cell line U87EGFRwt cells were treated with erlotinib with or without thalidomide (1µM) followed by collection of supernatants and TNF ELISA. (a) Erlotinib vs erlotinib +thalidomide: P =0.0015, t=7.77, d.f.=4. (b) Erlotinib vs erlotinib +thalidomide: P =0.0008, t=8.98, d.f.=4. Data are presented as mean±s.e.m; **P < 0.01, ***P<0.001 from two-tailed unpaired Student's t-test ( n = 3 biologically independent experimental replicates). (c-d) A neutralizing antibody to TNF sensitizes cells to EGFR inhibition. Patient derived GBM9 neurospheres or established GBM cell line U87EGFRwt cells were exposed to erlotinib plus either normal mouse IgG (Ctrl) or a neutralizing antibody to TNF. The TNF neutralizing antibody sensitizes cells to EGFR inhibition as shown by the Alamarblue Cell Viability assay. (c) Erlotinib vs erlotinib+tnf Ab: P =0.0013,t=8.07, d.f.=4. (d) Erlotinib vs erlotinib+tnf Ab: P =0.0012, t=8.13, d.f.=4. Data are presented as mean±s.e.m; **P < 0.01 from two-tailed unpaired Student's t-test ( n =3 biologically independent experimental replicates). (e-f) Thalidomide sensitizes patient derived primary GBM9 and GBM39 neurospheres to EGFR inhibiton. GBM9 or GBM39 neurospheres were exposed to EGFR inhibition using afatinib (100nM) and thalidomide (1µM) alone or in combination concurrently for 72h followed by Alamarblue Cell Viability Assay. DMSO was used as a control (Ctrl). (e) Afatinib vs afatinib+thalidomide: P =0.0001,t=14.47, d.f.=4. (f) Afatinib vs afatinib+thalidomide: P =0.0005, t=10.4, d.f.=4. Data are presented as mean±s.e.m; ***P<0.001 from two-tailed unpaired Student's t-test ( n =3 biologically independent experimental replicates).

Supplementary Figure 4 Inhibition of JNK and ERK sensitizes glioma cells to EGFR inhibition. (a-b) GBM9 cells were treated with erlotinib and/or JNK inhibitor SP600125 (1µM), ERK inhibitor U0126 (1µM) for 72 hours followed by Annexin-FACS assay. Unstained cells represent viable cells. Annexin positive cells are undergoing apoptosis. PI (Propidium iodide)

positive cells are undergoing late apoptosis. In Control cells are treated with control vehicle (DMSO). The combination treatment decreases the number of viable cells and there is an increase in double stained cells (Annexin+PI Positive). (b) Unstained: Erlotinib vs Erlotinib+SP600125: P =0.0008, t=9.10, d.f.=4; Erlotinib vs erlotinib+u0126: P =0.0036, t=6.14, d.f.=4. Annexin+PI: Erlotinib vs erlotinib+sp600125: P =0.0003, t=11.37, d.f.=4; Erlotinib vs erlotinib+u0126: P =0.0021,t=7.04, d.f.=4. (c-d) A similar experiment was conducted in U87EGFR cells with a similar result. (d) Unstained: Erlotinib vs erlotinib+sp600125: P =0.0045, t=5.77, d.f.=4; Erlotinib vs erlotinib+u0126: P =0.0013,t=8.06, d.f.=4. Annexin+PI: Erlotinib vs erlotinib+sp600125: P =0.0017, t=7.53, d.f.=4; Erlotinib vs erlotinib+u0126: P =0.0006, t=10.04, d.f.=4. Data are presented as mean±s.e.m; **P<0.01, ***P<0.001 using two-tailed unpaired Student's t-test ( n =3 biologically independent experimental replicates).

Supplementary Figure 5 Axl and TNF inhibition renders glioma cells sensitive to EGFR inhibition.

(a-b) GBM9 cells were treated with erlotinib and/or Axl inhibitor R428 (1µM), Thalidomide (1µM) for 72 hours followed by Annexin- FACS assay. Unstained cells represent viable cells. Annexin positive cells are undergoing apoptosis. PI (Propidium iodide) positive cells are undergoing late apoptosis. In Control cells are treated with control vehicle (DMSO). The combination treatment decreases the number of viable cells and there is an increase in both PI and double stained cells (Annexin+PI Positive). (b) Unstained: Erlotinib vs erlotinib+r428: P =0.0051,t=5.58, d.f.=4; Erlotinib vs erlotinib+thalidomide: P =0.0009,t=8.75, d.f.=4. Annexin+PI: Erlotinib vs erlotinib+r428: P =0.015, t=4.10, d.f.=4; Erlotinib vs erlotinib+thalidomide: P =0.0011,t=8.32, d.f.=4. (c-d) A similar experiment was conducted in U87EGFR cells with a similar result. (d) Unstained: Erlotinib vs erlotinib+r428: P =0.0045, t=5.71, d.f.=4; Erlotinib vs erlotinib+thalidomide: P =0.0036, t=6.13, d.f.=4. Annexin+PI: Erlotinib vs erlotinib+r428: P =0.0014, t=7.83, d.f.=4; Erlotinib vs erlotinib+thalidomide: P =0.0022, t=6.98, d.f.=4. Data are presented as mean±s.e.m; *P<0.05, **P<0.01, ***P<0.001 using two-tailed unpaired Student's t-test ( n =3 biologically independent experimental replicates).

Supplementary Figure 6 Etanercept renders glioma cells sensitive to EGFR inhibition.

(a-b) GBM9 cells were treated with erlotinib and/or enbrel (1µM) for 72 hours followed by Annexin-FACS assay. Unstained cells represent viable cells. Annexin positive cells are undergoing apoptosis. PI (Propidium iodide) positive cells are undergoing late apoptosis. In Control cells are treated with control vehicle (DMSO). The combination treatment decreases the number of viable cells and there is an increase in double stained cells (Annexin+PI Positive). (b) Unstained: Erlotinib vs erlotinib+enbrel: P =0.0008, t=9.11, d.f.=4; Annexin+PI: Erlotinib vs erlotinib+enbrel: P=0.0012, t=8.18, d.f.=4. (c-d) A similar experiment was conducted in U87EGFRwt cells with a similar result. (d) Unstained: Erlotinib vs erlotinib+enbrel: P =0.0006, t=9.6, d.f.=4; Annexin+PI: Erlotinib vs erlotinib+enbrel: P =0.0003, t=11.34, d.f.=4. Data are presented as mean±s.e.m; **P<0.01, ***P<0.001 using two-tailed unpaired Student's t-test ( n =3 biologically independent experimental replicates).

Supplementary Figure 7 Combination treatment increases caspase 3/7 activity in glioma cells.

(a) GBM9 cells were treated by erlotinib and/or SP600125, U1026 for 72 hours followed by Caspase-Glo 3/7 Assay. Caspase3/7 activity was evaluated in GBM9 cells exposed to erlotinib plus SP600125. Similar results were obtained using a combination of erlotinib and U1026. Erlotinib vs erlotinib+sp600125: P =0.001, t=8.66, d.f.=4; Erlotinib vs erlotinib+u0126: P =0.0002, t=13.24, d.f.=4. Similar procedures were performed in b-f and similar results were obtained. (b) Erlotinib vs erlotinib+r428: P =0.0004, t=10.76, d.f.=4; Erlotinib vs erlotinib+thalidomide: P=0.0004, t=10.64, d.f.=4. (c) Erlotinib vs erlotinib+sp600125: P<0.0001, t=21.41, d.f.=4; Erlotinib vs erlotinib+u1026: P =0.0007, t=9.49, d.f.=4. (d) Erlotinib vs erlotinib+r428: P=0.0001, t=14.8, d.f.=4; Erlotinib vs erlotinib+t: P <0.0001, t=16.77, d.f.=4. (e) Erlotinib vs erlotinib+enbrel: P =0.0096, t=8.72, d.f.=4. (f) Erlotinib vs erlotinib+enbrel: P =0.0017, t=7.43, d.f.=4. Data are presented as mean±s.e.m; **P<0.01, ***P<0.001, ****P<0.0001 from two-tailed unpaired Student's t-test ( n =3 biologically independent experimental replicates).

Supplementary Figure 8 Combination treatment inhibits glioma cell proliferation.

(a) GBM9 cells were plated in 6 well plated and treated with drugs as indicated. Cell proliferation was measured through cell counting using the trypan blue dye exclusion test. A combination of erlotinib and SP600125 (SP) significantly reduces cell growth after 72 hours. Similar results were shown using a combination of erlotinb and U1026 (U). Erlotinib vs erlotinib+sp: P =0.0016, t=7.60, d.f.=4; Erlotinib vs erlotinib+u: P =0.0012, t=8.24, d.f.=4. Similar procedures were performed in b-f and similar results were obtained. (b) Erlotinib vs erlotinib+r428: P =0.0008, t=9.24, d.f.=4; Erlotinib vs erlotinib+t: P =0.0004, t=11.18, d.f.=4. (c) Erlotinib vs erlotinib+sp: P =0.0004, t=10.78, d.f.=4; Erlotinib vs erlotinib+u: P =0.0014, t=7.85, d.f.=4. (d) Erlotinib vs erlotinib+r428: P =0.001, t=8.62, d.f.=4; Erlotinib vs erlotinib+t: P =0.0003, t=11.58, d.f.=4. (e) Erlotinib vs erlotinib+enbrel: P =0.0014, t=7.85, d.f.=4. (f) Erlotinib vs erlotinib+enbrel: P =0.0005, t=10.37, d.f.=4. SP: SP600125; U:U0126; T: Thalidomide. Data are presented as mean±s.e.m; **P<0.01, ***P<0.001 from two-tailed unpaired Student's t-test ( n =3 biologically independent experimental replicates).

Supplementary Figure 9

TNF inhibition sensitizes mouse tumors to EGFR inhibition with afatinib in vivo. (a) Combined treatment of afatinib and thalidomide prolonged survival and suppressed tumor growth in an orthotropic model. Kaplan- Maier survival curves were calculated using GraphPad Prism 7. Statistical significance verified by the log rank test, P=0.0015,**P<0.01. (b) Representative bioluminescence images from afatinib and afatinib plus thalidomide group at day 1, 10 and 20 post-treatment. Since all mice in vehicle and thalidomide group died within 20 days after transplant, images at day 20 post-treatment were not available. (c-d) Body weight of xenograft mice were monitored regularly and recorded every 2 days. No significant change of body weight was observed in subcutaneous mouse model between groups. (e) Body weight of orthotopic models. Body weight losses are found in vehicle, erlotinib and thalidomide group but not in combination therapy (erlotinib+thalidomide). Erlotinib vs erlotinib+thalidmoide: P<0.0001, t=6.32, d.f.=14; (f) Similar results were obtained in orthotopic models treated by afatinib and/or thalidmide. Afatinib vs afatinib+thalidomide: P =0.0003, t=4.79, d.f.=14.

Supplementary Figure 10 Erlotinib treatment activates Axl JNK ERK signaling in orthotopic models; this is suppressed by TNF inhibition. (a) Immunostainig of paxl, pjnk and perk proteins from a representative brain section from vehicle, erlotinb and erlotinib plus

thalidomide group. Cells with brown staining are considered as positive. Erlotinib group shows higher expression of paxl, pjnk and perk compared to vehicle group, whereas thaldimide inhibits erlotinib induced paxl-pjnk-perk activation. (b-d) Semi-quantitative analysis of paxl, pjnk and perk immunostaining. Four random fields in 3 tissue blocks at x200 magnification were scored semiquantitatively as: 0=No positive staining; 1=1-25% tumor cells stained, 2=26%-75% tumor cells stained and 3=>75% tumor cells stained. Differences between treatment groups were analyzed by the Mann-Whitney U test (n=12). (b) paxl: Erlotinib vs. vehicle, p=0.0062, U=26; Erlotinib vs. erlotinib+thalidomide, p=0.0093, U=28. (c) pjnk: Erlotinib vs. vehicle, p=0.022, U=32; Erlotinib vs. erlotinib+thalidomide, p=0.014, U=29.5. (d) perk: Erlotinib vs. vehicle, p=0.0040, U=23.5; Erlotinib vs. erlotinib+thalidomide, p=0.0026, U=21.

Supplementary Figure 11 Full-length western blots. Full-length Western blots for cropped images in Fig. 1.

Supplementary Figure 12

Full-length western blots and DNA agarose gel. Full-length Western blots for cropped images in Fig. 2 and Fig. 3f, 3h, 3i.

Supplementary Figure 13

Full-length western blots. Full-length Western blots for cropped images in Fig. 4g, 4h, 4i.

Supplementary Figure 14

Full-length western blots. Full-length Western blots for cropped images in Fig. 6j-l and Fig. 7f, 7g.

Supplementary Figure 15

Full-length western blots. Full-length Western blots for cropped images in Supplementary Fig. 1 and 2d,e,g i.