A Phosphotyrosine Proteomic Screen Identifies Multiple Tyrosine Kinase Signaling Pathways Aberrantly...

13
Genes & Cancer 1(5) 493–505 © The Author(s) 2010 Reprints and permission: sagepub.com/journalsPermissions.nav DOI: 10.1177/1947601910375273 http://ganc.sagepub.com Introduction Malignant mesothelioma (MM) is an aggressive cancer that develops from mesothelial cells lining the pleural, perito- neal, and pericardial cavities. Approximately 2500 MM cases are diagnosed annually in the United States, and the disease is causally associated with exposure to asbestos. 1-3 Current chemotherapy regimens, which include combined treatment with cisplatin and premetrexed, only improve median survival rates by approximately 3 months when compared to single-agent premetrexed or cisplatin thera- pies. 2,3 Surgical resection, where a large portion of the dif- fuse tumor is removed, is theoretically the most effective treatment; however, microscopic portions of the tumor inevitably remain and continue to grow. 4 As a direct result of the lack of effective treatment options for MM patients, the disease prognosis remains fatal in virtually all cases. Thus, discovering new molecular targets for therapeutic intervention remains a focus of many laboratories studying MM and other refractory cancers. Our understanding of the molecular and cellular mecha- nisms that drive MM has improved in the past 2 decades. We now know that many genetic and cell signaling pertur- bations are involved in the development and progression of MM. Inactivation of tumor suppressor genes such as NF2 and CDKN2A/ARF (p16INK4A/p14ARF) occur in a large proportion of MMs. 5-9 Moreover, engineered deficiency of these tumor suppressors in knockout mice accelerates asbestos-induced MM, confirming the importance of inacti- vation of these tumor suppressors to the malignant phenotype. 10-12 Recent work has also identified cellular signaling axes that contribute to cell survival, proliferation, and chemoresistance of MM cells. Our laboratory identified the phosphatidylinositol-3 kinase (PI3K)–AKT-mammalian target of the rapamycin (mTOR) pathway as being hyper- activated in a high percentage of human and murine MMs and demonstrated that inhibiting this pathway 1 Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA, USA 2 Biochemistry and Biotechnology Facility, Fox Chase Cancer Center, Philadelphia, PA, USA 3 Department of Pathology, University of Vermont College of Medicine, Burlington, VT, USA Corresponding Author: J. R. Testa, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA Email: [email protected] A Phosphotyrosine Proteomic Screen Identifies Multiple Tyrosine Kinase Signaling Pathways Aberrantly Activated in Malignant Mesothelioma Craig W. Menges 1 ,Yibai Chen 2 , Brooke T. Mossman 3 , Jonathan Chernoff 1 , Anthony T.Yeung 2 , and Joseph R.Testa 1 Submitted 15-Mar-2010; Revised 14-May-2010; Accepted 16-May-2010 Abstract Malignant mesothelioma (MM) is a highly aggressive cancer that is refractory to all current chemotherapeutic regimens.Therefore, uncovering new rational therapeutic targets is imperative in the field. Tyrosine kinase signaling pathways are aberrantly activated in many human cancers and are currently being targeted for chemotherapeutic intervention. Thus, we sought to identify tyrosine kinases hyperactivated in MM. An unbiased phosphotyrosine proteomic screen was employed to identify tyrosine kinases activated in human MM cell lines. From this screen, we have identified novel signaling molecules, such as JAK1, STAT1, cortactin (CTTN), FER, p130Cas (BCAR1), SRC, and FYN as tyrosine phosphorylated in human MM cell lines. Additionally, STAT1 and SRC family kinases (SFK) were confirmed to be active in primary MM specimens. We also confirmed that known signal transduction pathways previously implicated in MM, such as EGFR and MET signaling axes, are coactivated in the majority of human MM specimens and cell lines tested. EGFR, MET, and SFK appear to be coactivated in a significant proportion of MM cell lines, and dual inhibition of these kinases was demonstrated to be more efficacious for inhibiting MM cell viability and downstream effector signaling than inhibition of a single tyrosine kinase. Consequently, these data suggest that tyrosine kinase inhibitor monotherapy may not represent an efficacious strategy for the treatment of MM due to multiple tyrosine kinases potentially signaling redundantly to cellular pathways involved in tumor cell survival and proliferation. Keywords tyrosine kinases, asbestos, mesothelioma, proteomics, coactivation Original Article

Transcript of A Phosphotyrosine Proteomic Screen Identifies Multiple Tyrosine Kinase Signaling Pathways Aberrantly...

Genes & Cancer1(5) 493 –505© The Author(s) 2010Reprints and permission: sagepub.com/journalsPermissions.navDOI: 10.1177/1947601910375273http://ganc.sagepub.com

Introduction

Malignant mesothelioma (MM) is an aggressive cancer that develops from mesothelial cells lining the pleural, perito-neal, and pericardial cavities. Approximately 2500 MM cases are diagnosed annually in the United States, and the disease is causally associated with exposure to asbestos.1-3 Current chemotherapy regimens, which include combined treatment with cisplatin and premetrexed, only improve median survival rates by approximately 3 months when compared to single-agent premetrexed or cisplatin thera-pies.2,3 Surgical resection, where a large portion of the dif-fuse tumor is removed, is theoretically the most effective treatment; however, microscopic portions of the tumor inevitably remain and continue to grow.4 As a direct result of the lack of effective treatment options for MM patients, the disease prognosis remains fatal in virtually all cases. Thus, discovering new molecular targets for therapeutic intervention remains a focus of many laboratories studying MM and other refractory cancers.

Our understanding of the molecular and cellular mecha-nisms that drive MM has improved in the past 2 decades. We now know that many genetic and cell signaling pertur-bations are involved in the development and progression of

MM. Inactivation of tumor suppressor genes such as NF2 and CDKN2A/ARF (p16INK4A/p14ARF) occur in a large proportion of MMs.5-9 Moreover, engineered deficiency of these tumor suppressors in knockout mice accelerates asbestos-induced MM, confirming the importance of inacti-vation of these tumor suppressors to the malignant phenotype.10-12

Recent work has also identified cellular signaling axes that contribute to cell survival, proliferation, and chemoresistance of MM cells. Our laboratory identified the phosphatidylinositol-3 kinase (PI3K)–AKT-mammalian target of the rapamycin (mTOR) pathway as being hyper-activated in a high percentage of human and murine MMs and demonstrated that inhibiting this pathway

1Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA, USA2Biochemistry and Biotechnology Facility, Fox Chase Cancer Center, Philadelphia, PA, USA3Department of Pathology, University of Vermont College of Medicine, Burlington, VT, USA

Corresponding Author:J. R. Testa, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA Email: [email protected]

A Phosphotyrosine Proteomic Screen Identifies Multiple Tyrosine Kinase Signaling Pathways Aberrantly Activated in Malignant Mesothelioma

Craig W. Menges1, Yibai Chen2, Brooke T. Mossman3,Jonathan Chernoff1, Anthony T. Yeung2, and Joseph R. Testa1

Submitted 15-Mar-2010; Revised 14-May-2010; Accepted 16-May-2010

AbstractMalignant mesothelioma (MM) is a highly aggressive cancer that is refractory to all current chemotherapeutic regimens. Therefore, uncovering new rational therapeutic targets is imperative in the field. Tyrosine kinase signaling pathways are aberrantly activated in many human cancers and are currently being targeted for chemotherapeutic intervention. Thus, we sought to identify tyrosine kinases hyperactivated in MM. An unbiased phosphotyrosine proteomic screen was employed to identify tyrosine kinases activated in human MM cell lines. From this screen, we have identified novel signaling molecules, such as JAK1, STAT1, cortactin (CTTN), FER, p130Cas (BCAR1), SRC, and FYN as tyrosine phosphorylated in human MM cell lines. Additionally, STAT1 and SRC family kinases (SFK) were confirmed to be active in primary MM specimens. We also confirmed that known signal transduction pathways previously implicated in MM, such as EGFR and MET signaling axes, are coactivated in the majority of human MM specimens and cell lines tested. EGFR, MET, and SFK appear to be coactivated in a significant proportion of MM cell lines, and dual inhibition of these kinases was demonstrated to be more efficacious for inhibiting MM cell viability and downstream effector signaling than inhibition of a single tyrosine kinase. Consequently, these data suggest that tyrosine kinase inhibitor monotherapy may not represent an efficacious strategy for the treatment of MM due to multiple tyrosine kinases potentially signaling redundantly to cellular pathways involved in tumor cell survival and proliferation.

Keywordstyrosine kinases, asbestos, mesothelioma, proteomics, coactivation

Original Article

494 Genes & Cancer / vol 1 no 5 (2010)

pharmacologically increases sensitivity to cisplatin.13 The mTOR tributary of PI3K-AKT signaling appears to be par-ticularly important in mediating survival signals in MM, especially in tumors with high baseline expression of acti-vated p70 S6-kinase,14,15 a downstream effector involved in protein translation. Other studies have shown that asbestos exposure stimulates epidermal growth factor receptor (EGFR) signaling and activation of the mitogen-activated protein kinase (MAPK) cascade in primary mesothelial cell cultures.16,17 The MAPK signaling axis itself is hyperactive in MM cells and tumors.18-21 Thus, inhibitors to the PI3K-AKT-mTOR and MAPK pathways are currently being eval-uated as potential therapies in preclinical models of MM.

The PI3K-AKT-mTOR and MAPK pathways are regu-lated by various upstream kinases, including receptor and nonreceptor tyrosine kinases.22 The tyrosine kinase family represents a large class of proteins whose aberrant activity has been linked to many human cancers.23 Recent studies have focused on identifying novel receptor and nonreceptor tyrosine kinases activated in a variety of cancers, including glioblastoma, non–small cell lung cancer, and colorectal cancer.24-27 The identified kinases, including MET, EGFR, PDGFR, IGFR, and CSF1R, are currently being evaluated preclinically as potential therapeutic targets for these can-cers.24-27 With these new findings in mind, we initiated experiments to identify tyrosine kinase family members activated in MM. Using an unbiased phosphotyrosine pro-teome screen, we identified two receptor tyrosine kinases, EGFR and MET, previously reported to be hyperactivated in MM, as well as nonreceptor tyrosine kinases SRC, FYN, JAK1, and FER; the transcription factor STAT1; and the scaffolding proteins cortactin (CTTN) and p130Cas (breast cancer anti–estrogen resistance 1, BCAR1) as tyrosine phosphorylated proteins in MM cells. We further demon-strated that STAT1 phosphorylation is relatively common and specific to MM cell lines and tumors when compared to nontransformed mesothelial cells and normal mesothelial tissue. The SRC family kinases (SFK), including SRC and FYN, were commonly activated in MM cells and primary tumor specimens, and we determined that dual tyrosine kinase inhibitor (TKI) therapy is more effective at inhibit-ing downstream effector signaling and cell viability than TKI monotherapies in MM cell lines with concomitant acti-vation of multiple tyrosine kinases. Together, these data indicate that many tyrosine kinase signaling axes are con-comitantly activated in MM cells and that mono-TKI thera-pies may not be efficacious for the treatment of MM.

ResultsPhosphotyrosine proteomic screen identifies tyrosine kinase

signaling pathways in MM. To determine if tyrosine kinases are hyperactivated in MM cells, we evaluated the relative levels of pan-phosphotyrosine proteins in human MM cell lines in comparison to normal mesothelial cells. As shown

in Figure 1A, we observed strong phosphotyrosine protein levels in the majority of MM cell lines. In contrast, normal mesothelial cells had undetectable levels of phosphotyro-sine proteins (Fig. 1A). We also observed strong phospho-tyrosine immunohistochemical (IHC) staining in MM tumors, with modest expression observed in normal meso-thelial lining from lung pleura (Fig. 1B).

From these initial experiments, we hypothesized that multiple tyrosine kinase signaling pathways may be aber-rantly activated in MM. To identify the phosphotyrosine proteins observed in the MM cell lines, we immunoprecipi-tated proteins from MM cell lysate with the same pan- phosphotyrosine antibody used for the immunoblot analysis in Figure 1A (i.e., antibody PY20—scheme shown in Fig. 1C). Proteins pulled down with the phosphotyrosine antibody or control IgG antibody were separated by SDS–gel electrophoresis and stained with Coomassie blue (Fig. 1D). All unique bands were excised from the gel, processed, and run by liquid chromatography/tandem mass spectrom-etry (LC/MS/MS) to identify specific proteins.

Proteins identified from the mass spectrometry analysis were further analyzed using criteria that ensured that (a) the protein of interest was a known phosphotyrosine accepting protein, (b) the protein had adequate peptide coverage, and (c) the protein came from a Coomassie-stained band that corresponded closely to the molecular weight of that pro-tein. Screen hits were further validated by evaluating the tyrosine phosphorylation status of the protein in MM cell lysates (Fig. 1E). Nine validated phosphotyrosine proteins identified from the proteomic analysis are shown in Table 1 and Figure 1E. Two of the proteins identified, EGFR and MET, are receptor tyrosine kinases previously reported to be aberrantly activated in MM cells,28,29 thus validating the methodology of our screen. We also identified novel nonre-ceptor tyrosine kinases SRC, FYN, FER, and JAK1. In addition to tyrosine kinases, the transcription factor Signal Transducers and Activators of Transcription 1 (STAT1) and scaffolding proteins cortactin and p130Cas were also found to be tyrosine phosphorylated in MM cells (Table 1 and Fig. 1E).

STAT1 signaling axis is aberrantly activated in MM. Tyro-sine phosphorylation of STAT1 allows nuclear transloca-tion and transcriptional activation of genes that negatively regulate cell proliferation and survival.30 STAT1 is alsoan important transcriptional regulator of inflammation. Some cancers, including MM,31 are thought to require inflammation to promote tumorigenesis. Recently, STAT1 phosphorylation was demonstrated to be required for colitis-induced gastric carcinogenesis in mice.32 Thus, STAT1 phosphorylation-mediated regulation of inflammation may be an important step in MM development, in addition to STAT1’s role as a tumor suppressor. As such, we were inter-ested in evaluating if STAT1 phosphorylation is a common event in MM.

Tyrosine kinases coactivated in mesothelioma / Menges et al. 495

We first evaluated the tyrosine phosphorylation status of STAT1 in 4 MM cell lines, Meso 6, 12, 43, and H-Meso, when compared to 2 nontransformed mesothelial cells, LP9-hTERT and 7086. Interestingly, we observed higher levels of P-STAT1 in 3 of the 4 MM cell lines in

comparison to both control cell lines (Fig. 2A). All cell lines, MM and nontransformed mesothelial cell cultures, expressed total STAT1 protein at similar levels, suggesting that STAT1 phosphorylation is specific to the MM cells. We also observed strong nuclear P-STAT1 staining in primary

ANormal

B

80100120

MW(kDa )

80100120

MW(kDa )

6 8 10 12 17 22 25 32 41 43 45 54 H-MesoMM Cell Lines

NM

MM MM

IB: PY20 IB: PY2020

30

40

5060

80

20

30

40

5060

80

Actin

EGFRMET

IP: PY20 IgG

100

JAK1CASJAK1CAS

120

EGFRMET

IP: PY20 IgG

100

JAK1CASJAK1CAS

120

DC

PY

PY

PY

p170p145

IgG PY205% input

IP

p170p145p170p145p170p145p170p145p170p145p170p145p170p145p170p145p170p145

E

170 kDa

IB: MET

IB: EGFR

Scheme for Identification of Phosphotyrosine Proteins from MM Cells

Mesothelioma Cell Lysate

60

80

SrcFyn

STAT1

FERCortactin

50

40

60

80

SrcFyn

STAT1

FERCortactin

50

40

60 kDa

60 kDa

60 kDa

80 kDa

100 kDa

IB: Fyn

IB: Src

IB: P-Src (Y416) Family

IB: FER

IB: p130Cas

Pre-clear with Protein -G Agarose

IP with anti -phosphotyrosine (PY20) or IgG control

Ran IPs on 4 -12% Bis-Tris Gel

-G

-

-

30

20

30

20

80 kDa

100 kDa

80 kDa

IB: Cortactin

IB: JAK1

IB: STAT1

Stained Gel with Brilliant Blue G Coomassie

Excised All Bands

LC/MS/MS for Protein Identification

PY

Figure 1. An unbiased phosphotyrosine proteomic screen identifies tyrosine kinases aberrantly activated in malignant mesothelioma (MM). (A) Western blot analysis of phosphotyrosine protein expression (PY20) in human MM cell lines compared to normal mesothelial cells. (B) Immunohistochemistry of phosphotyrosine protein expression in primary MMs (PY20) compared to that of normal mesothelial lining (arrows) from normal lung pleura. (C) Scheme for purifying and identifying phosphotyrosine proteins in MM cells. (D) Coomassie-stained gel of phosphotyrosine proteins immunopurified from MM cell lysates. (E) Validation of mass spectrometry queries by co-immunoprecipitation Western blot analysis.

Table 1. Mass Spectrometry of Results of Proteins of Interest from Phosphotyrosine Immunoprecipitation/Coomassie Gel

Protein IdentitySwissProt

Entry Gene NameSequence

Coverage (%) Mascot Score

Epidermal growth factor receptor P00533 EGFR 13 215Hepatocyte growth factor receptor P08581 MET 9 215Proto-oncogene tyrosine-protein kinase Src P12931 SRC 15 114Signal transducer and activator of transcription 1-alpha/beta P42224 STAT1 11 81Tyrosine-protein kinase JAK1 P23458 JAK1 7 81Src substrate cortactin Q14247 CTTN 5 72Proto-oncogene tyrosine-protein kinase Fyn P06241 FYN 11 61Proto-oncogene tyrosine-protein kinase FER P16591 FER 4 52Breast cancer anti–estrogen resistance protein 1 p56945 BCAR1 2 42

496 Genes & Cancer / vol 1 no 5 (2010)

MM specimens, with no staining observed in normal meso-thelial lining (Fig. 2B).

To assess the frequency of STAT1 phosphorylation in MM, we evaluated both JAK1, a proteomic screen hit and known upstream regulator of STAT1, and STAT1 phosphor-ylation in a panel of human MM cell lines. STAT1 was phosphorylated in 8 of 13 MM cell lines tested, and JAK1 was phosphorylated in 5 of the 13 cell lines (Fig. 2C). Inter-estingly, JAK1 phosphorylation did not always correlate with STAT1 phosphorylation (e.g., Meso 6 and 22), sug-gesting that additional upstream regulators contribute to STAT1 activation (Fig. 2C). Together, these data demon-strate that the STAT1 signaling pathway is frequently acti-vated in MM cells.

Src family kinases (SFK) are hyperactivated in MM. Two SFK members, SRC and FYN, were validated as tyrosine-phosphorylated proteins in MM cells (Fig. 1). Interest-ingly, YES, another SFK member, was also identified by

mass spectrometry as a phosphotyrosine protein in MM cells, but it was ruled out by our aforementioned criteria. We have since confirmed YES phosphorylation in 4 MM cell lines using antibody arrays (data not shown). Thus, it appears that multiple SFK are strongly phosphorylated in MM cells and may represent novel therapeutic targets in this disease.

To further investigate SFK activity in MM, we used an antibody that recognizes all SFK members when tyrosine phosphorylated at residue 416, an activating phosphoryla-tion event for SFK. When comparing MM cells to non-transformed mesothelial cells by immunoblot analysis, we observed strong SFK phosphorylation in virtually all MM cell lines tested with very little P-SFK observed in control cells (Fig. 3A). Consistent with these results, we observed strong P-SFK IHC staining in MM tumor specimens and no detectable staining in normal mesothelial lining (Fig. 3B). Thus, SFK phosphorylation appears to be specific to MM cells and not to normal mesothelial cells.

A

B Normal

NM MMMM

P-STAT1

LP9 7086 6 12 43 H-Meso

P-STAT1-

STAT1

Actin

MM MM

CHuman Mesothelioma Cell Lines

P-STAT1P-STAT1

JAK1

P-JAK1-

P STAT1

6 8 10 12 17 22 25 32 41 43 45 54 H -Meso

Human Mesothelioma Cell Lines

100 kDa

100 kDa

P-STAT1- 80 kDa

80 kDa

40 kDa

STAT1

Actin

Figure 2. STAT1 is frequently phosphorylated in malignant mesothelioma (MM). (A) Western blot analysis of STAT1 phosphorylation (Y701) and total STAT1 levels in MM versus normal control cells. Actin is included as a loading control. (B) Immunohistochemistry of P-STAT1 (Y701) expression in primary MMs versus normal mesothelial lining (arrows) from lung pleura. Nuclear P-STAT1 staining is marked by arrows. (C) Immunoblot analysis of P-JAK1 (Y1022/1023), total JAK1, P-STAT1 (Y701), and total STAT1 in a panel of human MM cell lines. Actin is included as a loading control.

Tyrosine kinases coactivated in mesothelioma / Menges et al. 497

To determine if SFK activation is common in MM, we evaluated SFK phosphorylation in a panel of MM cell lines. SFK activation was observed in most (11 out 13) MM cell lines tested (Fig. 3C). SRC and FYN were also expressed in a large proportion of the MM cell lines (Fig. 3C). Taken together, these data validate SFK as tyrosine kinases that are frequently activated in MM.

EGFR and MET are coactivated in MM. EGFR and MET, 2 receptor tyrosine kinases previously implicated in MM, were also found to be tyrosine phosphorylated in MM cells from our proteomic screen (Fig. 1D,E). We evaluated the fre-quency of EGFR and MET phosphorylation in a panel of MM cell lines by immunoprecipitation/immunoblot analysis. Both EGFR and MET were coactivated in 8 of 11 cell lines tested (Fig. 4A). Consistent with this result, we also observed concomitant IHC staining for P-EGFR and P-MET in pri-mary MM samples (Fig. 4B). When comparing SFK phos-phorylation to EGFR and MET activation, we also found that half of the MM cell lines had concomitant activation of all 3 tyrosine kinases (Figs. 3B and 4A). Collectively, these data suggest that multiple tyrosine kinases that control cell prolif-eration and survival are concurrently activated in MM.

Combinatorial inhibition of EGFR, MET, and/or SFK decreases MM cell viability more effectively than mono-TKI treatments. Multiple tyrosine kinases are hyperactivated in many human cancers, and thus, small tyrosine kinase inhibitors have been exploited for the treatment of these diseases. EGFR, MET, and SFK can signal downstream to redundant signaling effectors such as PI3K-AKT-mTOR and MAPK signaling cascades. The aforementioned downstream effects are known to be hyperactivated in MM and have been shown to contribute to MM cell proliferation and sur-vival. Because EGFR, MET, and SFK can all signal to these oncogenic pathways, mono-TKI therapy against one of these tyrosine kinases (e.g., EGFR) may not be effective if other tyrosine kinases such as MET or SFK signal redun-dantly to these same cell proliferation/survival pathways.

To evaluate if targeting multiple tyrosine kinases is more efficacious at inhibiting MM cell viability than mono-TKI inhibitors, we treated 2 MM cell lines (Meso 8 and Meso 43) that exhibit concomitant activation of the aforemen-tioned tyrosine kinases with small-molecule inhibitors against EGFR (AG1478), MET (MET Kinase Inhibitor), and SFK (PP2) alone or in combination. Before conducting the experiments with various drug combinations, we first

MM MM

P-SFKP-SFK

BA

C

Normal

P-SFK

6 8 10 12 17 22 25 32 41 43 45 54 H -Meso

Human Mesothelioma Cell Lines

60 kDa

60 kDa

Fyn

Actin

Src

P-Src (Y416) Family

60 kDa

40 kDa

LP9 7086

NM MM

12 22 43P-Src (Y416)

Family

Src

Fyn

Actin

Figure 3. SFK are phosphorylated in malignant mesothelioma (MM). (A) Western blot analysis of P-SFK (Y416), SRC, and FYN levels in MM versus control mesothelial cells. Actin is included as a loading control. (B) Immunohistochemistry of P-SFK (Y416) expression in primary MMs versus normal mesothelial lining (arrows) from lung pleura. (C) Western blot analysis of P-SFK (416), SRC, and FYN expression in a panel of human MM cell lines. Actin is included as a loading control.

498 Genes & Cancer / vol 1 no 5 (2010)

determined the lowest concentration needed for each small-molecule inhibitor to inhibit the activity of EGFR, MET, and SFK, respectively, in both Meso 8 and Meso 43 (Fig. 5A). Interestingly, the concentration required to inhibit phosphorylation of EGFR, MET, and SFK with AG1478, MET Kinase Inhibitor, and PP2, respectively, was consis-tently lower than the IC

50 concentration calculated for each

drug (Fig. 5A), suggesting that these drugs may inhibit cell viability through off-target effects at high concentrations. Based on the initial cell viability data, Meso 8 and 43 cells were then treated with doses of AG1478 (designated treat-ment E in Fig. 5B,C), MET Kinase Inhibitor (M), and PP2 (P) that resulted in a loss of cell viability of 20% to 40% when used as a single agent. Cells were treated with AG1478 (15 µM), MET Kinase Inhibitor (2 µM), PP2 (5 µM), or dual combinations of these inhibitors (EM, MP, EP) for 72 hours, and cell viability was evaluated by MTT assay. As shown in Figure 5B, EGFR and MET coinhibition (EM) was slightly more efficient at inhibiting cell viability com-pared to EGFR or MET alone in Meso 8 cells (P = 0.025).

Similarly, MET and SFK inhibition (MP) also inhibited cell viability more efficiently than inhibition of MET or SFK alone in both cell lines tested (Fig. 5B; Meso 8: M or P vs. MP, P = 0.025; Meso 43: M vs. MP, P = 0.012). Coinhibi-tion of EGFR and SFK was found to inhibit cell viability to the greatest extent, again more efficiently than inhibition of either EGFR or SFK alone (Fig. 5B; Meso 8: E or P vs. EP, P = 0.025; Meso 43: E or P vs. EP, P < 0.0004).

We also conducted in vitro studies of the effect of com-bining molecularly targeted drugs with two different che-motherapeutic agents. In these experiments, inhibition of EGFR, SRC, or MET showed no consistent effect on the sensitivity of mesothelioma cells to gemcitabine or doxoru-bicine (data not shown).

The tyrosine kinases EGFR, MET, and SFK can signal redundantly to downstream effector pathways such as the PI3K-AKT-mTOR and MAPK pathways that contribute to tumor cell survival and proliferation. Thus, a tumor cell with tyrosine kinases concomitantly activated may confer resis-tance to mono-TKI therapy due to redundant signaling to

P-MET

P-EGFR

A

B

P-MET

P-EGFR

Normal

EGFR

IB: EGFRIP: PY20

IB: METIP: PY20

MET

Actin

120 kDa

120 kDa

120 kDa

120 kDa40 kDa

6 8 10 12 17 22 25 32 41 43 45 54 H -Meso

Human Mesothelioma Cell Lines

P-MET

P-EGFR

MMMM

Figure 4. EGFR and MET are concomitantly activated in malignant mesothelioma (MM). (A) Immunoblot analysis of P-EGFR, total EGFR, P-MET, and total MET in a panel of MM cell lines. Actin is included as a loading control. (B) Immunohistochemistry of P-EGFR (Y845) and P-MET (Y1230/1234/1235) expression in primary MM.

499

A

Mes

o 8

0.00

0

0.20

0

0.40

0

0.60

0

0.80

0

1.00

0

1.20

0

-E

MP

EM

MP

EP

Dru

g Tr

eatm

ent

Cell Viability

Mes

o 4

3

0.00

0

0.20

0

0.40

0

0.60

0

0.80

0

1.00

0

1.20

0

-E

MP

EM

MP

EP

Dru

g Tr

eatm

ent

Cell Viability

B C

Fig

ure

5. D

ual i

nhib

ition

of c

onco

mita

ntly

act

ivat

ed t

yros

ine

kina

ses

is m

ore

effic

acio

us a

t in

hibi

ting

mal

igna

nt m

esot

helio

ma

(MM

) ce

ll vi

abili

ty a

nd d

owns

trea

m e

ffect

or s

igna

ling.

(A

) M

eso

8 an

d M

eso

43 c

ells

wer

e se

eded

on

a 96

-wel

l pla

te a

nd t

reat

ed w

ith in

crea

sing

con

cent

ratio

ns o

f AG

1478

(EG

FR in

hibi

tor)

, MET

Kin

ase

Inhi

bito

r, o

r PP

2 (S

FK in

hibi

tor)

for

72 h

ours

, and

the

n ce

ll vi

abili

ty

was

det

erm

ined

by

MT

T a

ssay

. Wes

tern

blo

t an

alys

is w

as p

erfo

rmed

to

eval

uate

a m

inim

al c

once

ntra

tion

of e

ach

drug

nee

ded

to in

hibi

t EG

FR, M

ET, a

nd S

FK p

hosp

hory

latio

n, r

espe

ctiv

ely

(24

hour

s po

sttr

eatm

ent)

. Tot

al E

GFR

, MET

, and

Src

, as

wel

l as

actin

, are

incl

uded

as

load

ing

cont

rols

. (B

) M

eso

8 an

d 43

cel

ls w

ere

seed

ed o

n a

96-w

ell p

late

and

tre

ated

or

not

with

AG

1478

(E)

, MET

Kin

ase

Inhi

bito

r (M

), PP

2 (P

), A

G14

78 +

MET

Kin

ase

Inhi

bito

r (E

M),

MET

Kin

ase

Inhi

bito

r +

PP2

(M

P), a

nd A

G14

78 +

PP2

(EP

) fo

r 72

hou

rs, f

ollo

wed

by

asse

ssm

ent

of c

ell v

iabi

lity

by M

TT

ass

ay. (

C)

Mes

o 8

and

43 c

ells

wer

e tr

eate

d w

ith t

he a

fore

men

tione

d dr

ug c

ombi

natio

ns a

nd w

ere

harv

este

d 24

hou

rs la

ter

for

Wes

tern

blo

t an

alys

is. P

-AK

T, P

-ER

K1/

2, a

nd P

-S6R

P le

vels

wer

e ev

alua

ted

to d

eter

min

e dr

ug e

ffect

s on

AK

T, M

APK

, and

mT

OR

act

ivity

, res

pect

ivel

y. T

otal

AK

T, E

RK

1/2,

and

S6R

P, a

s w

ell a

s ac

tin, a

re in

clud

ed a

s lo

adin

g co

ntro

ls.

500 Genes & Cancer / vol 1 no 5 (2010)

downstream effectors by other tyrosine kinases. To determine if dual-TKI treatment is more effective at inhibiting down-stream signaling, Meso 8 and 43 cells were treated with the aforementioned drug combinations, and immunoblot analysis with phospho-specific antibodies against AKT, ERK1/2, and S6RP was performed to determine relative effects on down-stream effector signaling. Dual inhibition of EGFR and MET, MET and SFK, and EGFR and SFK was more efficient at inhibiting AKT phosphorylation than mono-TKI treatments in Meso 8, whereas inhibition of both EGFR and SFK repressed AKT phosphorylation more than any other mono-TKI or dual-TKI treatment in Meso 43, as assessed by immu-noblot analysis (Fig. 5C). Interestingly, EGFR and SFK alone inhibited AKT phosphorylation in Meso 43 but not in Meso 8 (Fig. 5C). MET inhibition was ineffective at inhibiting AKT phosphorylation in either cell line.

Another pathway downstream of EGFR, MET, and SFK implicated in MM tumor cell survival/proliferation is the MAPK signaling cascade. To evaluate the effect of each drug combination on MAPK signaling, ERK1/2 phosphor-ylation was evaluated by Western blot analysis. In both of the MM cell lines tested, dual inhibition of EGFR/MET, MET/SFK, and EGFR/SFK was more efficient at inhibiting ERK1/2 phosphorylation than mono-TKI treatments (Fig. 5C). Dual inhibition of EGFR and SFK (EP) was the most effective treatment for inhibition of ERK1/2 phosphoryla-tion in both cell lines, similar to what was observed with regard to AKT phosphorylation (Fig. 5C).

A third downstream effector pathway regulated by EGFR, MET, and SFK found hyperactivated in MM speci-mens is the mTOR pathway. mTOR signaling has recently been linked to MM tumor cell survival and chemoresis-tance.13-15 To evaluate the effect of the drug combination treatments on mTOR signaling, S6 Ribosomal Protein (S6RP) phosphorylation was evaluated by immunoblotting. Dual inhibition of MET/SFK and EGFR/SFK was more efficient at inhibiting S6RP phosphorylation than any other treatment (Fig. 5C). Consistent with AKT and ERK1/2 phosphorylation, dual inhibition of EGFR/SFK (EP) was the most effective treatment to inhibit mTOR signaling. Collectively, these data suggest that dual inhibition of EGFR and SFK can effectively inhibit PI3K-AKT, MAPK, and mTOR signaling in MM cells.

DiscussionRole of STAT1 activation in cancer, including MM. STAT1 is

an important mediator of the interferon gamma–induced transcriptional program.30 Upon cytokine receptor activa-tion, STAT1 is phosphorylated and homo- or heterodimer-izes before translocating to the nucleus, where it regulates the expression of genes that contribute to cellular arrest, apoptosis, and inflammation.30 STAT1 is directly phosphor-ylated by JAK1 and SRC, both identified through our screen as being activated in MM cells, as well as by

JAK2.30,33 Interestingly, JAK1 phosphorylation did not directly correlate with STAT1 phosphorylation in the panel of MM cell lines examined, suggesting that other upstream regulators such as SRC are also involved (Fig. 2C). Based on its ability to negatively regulate cell proliferation and/or survival, STAT1 is considered to be a tumor suppressor. STAT3 and STAT5, on the other hand, are considered to be oncoproteins because they positively regulate cell prolifera-tion and survival upon activation.30

Recently, however, STAT1 activation has been shown to promote radioresistance and tumorigenesis.32,34-36 Inhibit-ing STAT1 in both renal cell and squamous cell carcinomas was shown to increase radiosensitization.34,35 STAT1 was also identified as an important tumor promoter in a mouse model of leukemia.36 Moreover, both STAT1 and STAT3 were recently reported to be required for inflammation-associated gastric tumorigenesis in a mouse model of this disease.32 Thus, in certain cancers, STAT1 activation can promote tumor formation and resistance to therapies, which contrasts with its putative role as a tumor suppressor.

Our data indicate that STAT1 tyrosine phosphorylation is a common finding in MM cell lines (Fig. 2A). Further-more, IHC staining for nuclear P-STAT1 was found in MM tumors but was not detected in normal mesothelial lining (Fig. 2C). Together, these data suggest that STAT1 activa-tion may contribute to MM tumorigenesis. Interestingly, activation of STAT1-survivin signaling has been shown to be associated with a poor prognosis in MM patients.37 Asbestos, the major cause of MM, has been demonstrated to cause inflammation through the Nalp3 inflammasome in mice.38 The role of inflammation in MM progression is still unclear. However, a case could be made that if inflamma-tion drives MM development, then STAT1 signaling may be required for the development of MM, similar to its role in inflammation-associated gastric carcinogenesis.32

SFK are hyperactivated in MM. SFK, such as SRC and FYN, are nonreceptor tyrosine kinases that govern many cellular processes, including proliferation and survival.39-42 SFK can regulate both the PI3K-AKT-mTOR and MAPK pathways in various cell types.43,44 Not surprisingly, SFK have been found to be dysregulated in many human solid tumors, including breast, gastrointestinal, lung, and ovar-ian carcinomas.40,42,43,45-48 SFK activation has also been observed in leukemias, lymphomas, and myelomas.43,49 SFK inhibitors have been developed, and at least 3 such drugs are currently in phase I clinical trials.43,50,51 Having identified 2 SFK as being hyperactivated in human MM cell lines, SRC and FYN, we hypothesize that these kinases may contribute to MM cell survival and proliferation, potentially via signaling through the PI3K-AKT-mTOR and MAPK pathways. Germane to this, a recent study demonstrated that the dual SRC/BCR-ABL inhibitor dasat-inib caused cytotoxic effects in vitro against MM cell lines.52

Tyrosine kinases coactivated in mesothelioma / Menges et al. 501

We found that SRC and FYN are frequently phosphory-lated in MM. SFK activation was observed in more than 50% of the MM cell lines evaluated. Interestingly, SFK were found to be phosphorylated in MM cells that also exhibit hyperactivation of EGFR and MET. EGFR and MET can positively regulate SFK in many human cancer cells and often require SFK signaling to promote cell prolif-eration and survival.40,42,46,48,53 Conversely, SFK can posi-tively regulate EGFR and MET signaling to promote cancer cell proliferation and survival.25,54

EGFR and MET are concomitantly activated in MM. EGFR and MET are 2 receptor tyrosine kinases that have been found to be overexpressed, mutated, and hyperactivated in many human cancers. Consequently, pharmacological inhibitors and antagonistic antibodies against each receptor have been explored as potential therapies for many tumor types.55,56 EGFR and MET are independently hyperactivated in MM cells and tumors.28,29,55,57,58 The ligands for each receptor, EGF and HGF, respectively, are elevated in cells and fluids derived from pleural lavages of MM patients.59,60 EGFR and MET can signal independently to redundant pathways in MM cells, including the PI3K-AKT-mTOR and MAPK pathways. A recent independent investigation has also found coactivation of EGFR and MET in MM and dem-onstrated that dual inhibition of these kinases was more effi-cacious at inhibiting cell viability and proliferation than TKI-monotherapies directed against individual kinases.61

Coactivation of EGFR and MET has also been observed in other human tumor types. In lung adenocarcinoma cells, EGFR and MET can be concomitantly activated, with MET signaling allowing for resistance to EGFR inhibitors erlo-tinib and gefitinib by providing a redundant survival signal to the PI3K-AKT and MAPK pathways.62,63 This result is consistent with the notion that MET coactivation may allow for MM cell survival by signaling to redundant EGFR regu-lated pathways, such as the PI3K-AKT-mTOR and MAPK pathways.

In addition to their redundant roles contributing to tumor cell survival, EGFR and MET have been shown to posi-tively regulate the activity of one another in a variety of cell lines.64-67 In human glioma cells, HGF-MET signaling stim-ulated EGFR phosphorylation in a transcription-dependent manner.67 Conversely, EGFR signaling allowed for consti-tutive MET phosphorylation in human hepatoma cell lines, human epidermoid carcinoma cell lines, and mammary epi-thelial cells.64 It is not clear from our study whether EGFR activation is required for MET signaling or if MET activa-tion contributes to EGFR signaling. Determining the code-pendence of EGFR and MET signaling and evaluating the efficacy of combinatorial targeting of both receptor tyrosine kinases for the treatment of MM will be the focus of future studies.

Concomitant activation of tyrosine kinases in human cancers, including MM. Recent studies investigating the efficacy of TKIs in the treatment of glioblastoma demonstrated that more than one tyrosine kinase is often hyperactive in these tumors and that these kinases can signal downstream to redundant effector pathways that contribute to tumor cell survival.27 Importantly, concomitant activation of tyrosine kinases can allow tumor cells to acquire resistance to TKI monotherapies.27 We found that multiple tyrosine kinases, EGFR, MET, and SFK, are concomitantly activated in many MM cells lines (Figs. 3 and 4). These kinases have also been reported to be coactivated in other human cancer types.25-27,41,46,53,54,65,68 EGFR, MET and SFK can positively regulate one another and can signal downstream to the same pathways, including the PI3K-AKT-mTOR and MAPK sig-naling cascades.25-27,40,41,46,48,65,68,69 EGFR and SRC have been shown to activate MET to allow serum-independent growth in human bladder carcinoma cells.41 MET and SFK were recently shown to compensate for loss of EGFR acti-vation in breast cancer cells, providing redundant survival signaling and allowing tumor cells to escape targeted ther-apy against EGFR.54 Furthermore, MET was found to be activated in glioblastoma cells with hyperactive EGFRvIII receptor, and coactivation of MET by EGFRvIII was found to be important for tumor cell viability.65 Dual MET/EGFR combinatorial inhibition was shown to decrease erlotinib-resistant lung cancer cell survival both in vitro and in vivo.62 Thus, EGFR, MET, and SFK represent tyrosine kinases that are often concomitantly activated in human cancers and can signal redundantly to regulate tumor cell survival and resis-tance to single TKI therapies.

Using a phosphotyrosine proteomic screen, we were able to identify multiple tyrosine kinases aberrantly acti-vated in MM cells and tumor specimens. Because these various tyrosine kinases are coactivated in MM cells, we hypothesize that TKI monotherapies will be ineffective in the treatment of MM. Consistent with this notion, single-agent erlotinib therapy had no efficacy in a phase II clinical trial with MM patients.20 We had previously proposed that resistance to erlotinib in MM patients may be linked to other mechanisms that allow for continual signaling through the PI3K-AKT-mTOR pathway in the presence of erlo-tinib.20 Data presented here demonstrate that inhibition of multiple tyrosine kinases is more effective at inhibiting cell viability and downstream effector signaling pathways in MM cell lines. Collectively, these data suggest that combi-natorial TKI therapy might represent a promising approach for the treatment of MM.

Materials and MethodsCell culture. Human MM cell lines were established from

surgically explanted primary tumors as described previously70

502 Genes & Cancer / vol 1 no 5 (2010)

and grown in RPMI 1640 with 10% fetal bovine serum (FBS), supplemented with L-glutamine and penicillin/streptomycin. Nontransformed mesothelial cell culture LP9 (AG# 7086—referred to as 7086 herein) and derivative cell line LP9-hTERT were grown in M199/MCDB106 + 15% fetal calf serum (FCS) + 0.4 mg/mL hydrocortisone + 10 ng/mL EGF. The 7086 and LP9-hTERT cells used for Western blot analy-sis were at passage 6 or earlier.

Western blot analysis and co-immunoprecipitation. MM cells were trypsinized and harvested by centrifugation, washed in phosphate-buffered saline (PBS), and trans-ferred to lysis buffer (50 mM Tris-Cl [pH 8.0], 150 mM NaCl, 1% Triton X-100 containing proteinase inhibitor cocktail and phosphatase inhibitor cocktail II; Sigma Aldrich, St. Louis, MO). The cell lysates were clarified by centrifugation at 14,000 g for 10 minutes, and the protein concentrations of the whole-cell extracts were determined using a Bio-Rad protein assay kit (Bio-Rad, Hercules, CA). Then, 50 µg of whole-cell extract for each cell line was separated on a 4% to 12% Bis-Tris NuPAGE gel and transferred to nitrocellulose for immunoblotting. All NuPAGE products and gels were purchased from Invitro-gen (Carlsbad, CA). Primary antibodies were used for Western blot analysis at a dilution of 1:1000, with incuba-tion overnight. Anti-MET (C12), anti-EGFR (1005), anti-p130Cas (M-72), anti-cortactin (A-4), anti-p-Tyr (PY20), anti-β-actin (I-19), anti-P-ERK (E4), and anti-c-SRC (B-12) were purchased from Santa Cruz Biotechnology (Santa Cruz, CA); anti-phospho (P)–SRC (Y416) family, anti-P-JAK1 (Y1022/1023), anti-JAK1, anti-STAT1, anti-EGFR (Y845), anti-P-Tyr-100, anti-P-AKT (S473), anti-P-S6RP (S235/236), anti-S6RP, anti-ERK1/2, and anti-Fyn were purchased from Cell Signaling Technology (Beverly, MA); anti-P-MET (pYpYpY1230/1234/1235) was purchased from Invitrogen, and anti-P-STAT1 (Y701) was a gift from G. Rall (Fox Chase Cancer Center). For co-immunoprecip-itation of phosphotyrosine proteins from MM cell lysates, agarose-conjugated anti-P-Tyr (PY20) was employed (Santa Cruz Biotechnology).

Immunohistochemistry. An MM tissue microarray (TMA) was prepared by the Histopathology Facility at Fox Chase Cancer Center in accordance with guidelines of our Institu-tional Review Board (protocol 00-812). The TMA included 15 MM specimens spotted in duplicate along with 4 normal lung tissue samples and 2 normal kidney tissue samples used as controls. TMAs were deparaffinized and rehydrated prior to antibody staining. Anti-P-STAT1 (Y701—dilution 1:50), anti-P-EGFR (Y845—dilution 1:10), anti-P-MET (pYpYpY1230/1234/1235—dilution 1:50), and anti-P-SRC fam-ily (Y416—dilution 1:50) were incubated overnight with TMAs after antigen retrieval with 10 mM sodium citrate buffer (pH 6.0). Anti-P-Tyr-100 (dilution 1:100) was incu-bated overnight after antigen retrieval with 1 mM EDTA

(pH 8.0). TMAs were counterstained with hematoxylin. All images were taken on a Nikon Eclipse E600 microscope equipped with a Nikon Digital Camera DXM1200 at a mag-nification of 40x. All images were acquired using Nikon ACT-1 version 2 software.

Scaled-up phosphotyrosine immunoprecipitation. Of total protein lysate, 20 mg from human MM cell line Meso 8 was used to immunoprecipitate phosphotyrosine- containing proteins or nonspecific proteins (control IgG). Agarose-conjugated anti-phosphotyrosine (PY20) anti-body and control mouse IgG was allowed to incubate with the lysate overnight while rotating at 4°C. Immunoprecipi-tated proteins were centrifuged and washed 6 times with lysis buffer before being reduced and denatured in NuPAGE LDS Sample Buffer (Invitrogen). Immunoprecipitated pro-teins were separated on a 4% to 12% NuPAGE Bis-Tris Gel and subsequently stained for 30 minutes with Brilliant Coomassie G Blue stain (Sigma Aldrich). The gel was washed in distilled water overnight while rotating at room temperature.

Mass spectrometry analysis. Gel bands containing proteins were digested with trypsin and analyzed by nano LC/MS/MS on a QSTAR XL mass spectrometer (Applied Biosys-tems/MDS Sciex, Foster City, CA) as previously described in detail.71 For discovery of more proteins and peptides in each gel slice and to overcome the possibility of false nega-tives due to undersampling of coeluting peptides, an exclu-sion list composed of the peptides sequenced in the first LC/MS/MS run was assembled and used to direct another LC/MS/MS run to sequence only new peptides in the same sample. The two peak lists were combined for database searching for protein identification. The mass spectrum was processed by Analyst QS and then submitted to SwissProt protein database, release 54.1, using MASCOT 2.2 (Matrix Sciences, London, UK). Fixed modification of carbamido-methylcysteine, variable oxidation of methionine, and one trypsin miss were allowed for protein identification in LC/MS/MS. The protein identifications in Table 1 required more than one peptide for each protein. False discovery rate due to coincidence in the database was less than 3.5% for individual peptides as judged by hits at a decoy database containing randomized sequences in each entry.

MTT cell viability assay. Meso 8 or Meso 43 MM cells were each seeded on a 96-well plate, with 5000 cells per well. After 24 hours, various drug concentrations and com-binations were added to individual wells. MTT reagent was added to the plate 72 hours posttreatment, and absorbance was determined at 490 nm. EGFR inhibitor AG1478, MET Kinase Inhibitor, and SFK inhibitor PP2 were purchased from Calbiochem (EMD, Darmstadt, Germany). P values were determined using the Wilcoxon test (one-sided); P values of ≤0.05 were considered statistically significant.

Tyrosine kinases coactivated in mesothelioma / Menges et al. 503

Declaration of Conflicting Interests

The author(s) declared no potential conflicts of interest with respect to the authorship and/or publication of this article.

Acknowledgments and Funding

The following Fox Chase Cancer Center shared facilities were used in this study: Biotechnology, Cell Culture, Histopathology, and Biostatistics & Bioinformatics Facilities.

Grant support: This work was supported by the Mesothelioma Applied Research Foundation, National Cancer Institute (grant numbers CA-114047, CA-009035, and CA-06927), Local No. 14 Mesothelioma Fund of the International Association of Heat and Frost Insulators & Allied Workers in memory of Hank Vaughan and Alice Haas, Pew Charitable Trust, Kresge Foundation, and Pennsylvania Department of Health. The Department of Health disclaims responsibility for any analyses, interpretations, or conclusions.

References

1. Bridda A, Padoan I, Mencarelli R, Frego M. Peritoneal mesothelioma:

a review. MedGenMed 2007;9:32.

2. Ellis P, Davies AM, Evans WK, Haynes AE, Lloyd NS. The use of

chemotherapy in patients with advanced malignant pleural mesothe-

lioma: a systematic review and practice guideline. J Thorac Oncol

2006;1:591-601.

3. Janne PA, Wozniak AJ, Belani CP, et al. Pemetrexed alone or in com-

bination with cisplatin in previously treated malignant pleural meso-

thelioma: outcomes from a phase IIIB expanded access program. J

Thorac Oncol 2006;1:506-12.

4. Ismail-Khan R, Robinson LA, Williams CC Jr, Garrett CR, Bepler

G, Simon GR. Malignant pleural mesothelioma: a comprehensive

review. Cancer Control 2006;13:255-63.

5. Bianchi AB, Mitsunaga SI, Cheng JQ, et al. High frequency of

inactivating mutations in the neurofibromatosis type 2 gene (NF2)

in primary malignant mesotheliomas. Proc Natl Acad Sci U S A

1995;92:10854-8.

6. Cheng JQ, Jhanwar SC, Klein WM, et al. p16 alterations and dele-

tion mapping of 9p21-p22 in malignant mesothelioma. Cancer Res

1994;54:5547-51.

7. Hirao T, Bueno R, Chen CJ, Gordon GJ, Heilig E, Kelsey KT. Altera-

tions of the p16(INK4) locus in human malignant mesothelial tumors.

Carcinogenesis 2002;23:1127-30.

8. Pei J, Kruger WD, Testa JR. High-resolution analysis of 9p loss in

human cancer cells using single nucleotide polymorphism-based map-

ping arrays. Cancer Genet Cytogenet 2006;170:65-8.

9. Sekido Y, Pass HI, Bader S, et al. Neurofibromatosis type 2 (NF2)

gene is somatically mutated in mesothelioma but not in lung cancer.

Cancer Res 1995;55:1227-31.

10. Altomare DA, Vaslet CA, Skele KL, et al. A mouse model reca-

pitulating molecular features of human mesothelioma. Cancer Res

2005;65:8090-5.

11. Jongsma J, van Montfort E, Vooijs M, et al. A conditional mouse

model for malignant mesothelioma. Cancer Cell 2008;13:261-71.

12. Altomare DA, Menges CW, Pei J, et al. Activated TNF-α/NF-κB

signaling via down-regulation of Fas-associated factor 1 in asbestos-

induced mesotheliomas from Arf knockout mice. Proc Natl Acad Sci

U S A 2009;106:3420-5.

13. Altomare DA, You H, Xiao GH, et al. Human and mouse mesothe-

liomas exhibit elevated AKT/PKB activity, which can be targeted

pharmacologically to inhibit tumor cell growth. Oncogene 2005;24:

6080-9.

14. Barbone D, Yang TM, Morgan JR, Gaudino G, Broaddus VC. Mam-

malian target of rapamycin contributes to the acquired apoptotic resis-

tance of human mesothelioma multicellular spheroids. J Biol Chem

2008;283:13021-30.

15. Wilson SM, Barbone D, Yang TM, et al. mTOR mediates survival sig-

nals in malignant mesothelioma grown as tumor fragment spheroids.

Am J Respir Cell Mol Biol 2008;39:576-83.

16. Pache JC, Janssen YM, Walsh ES, et al. Increased epidermal growth

factor-receptor protein in a human mesothelial cell line in response to

long asbestos fibers. Am J Pathol 1998;152:333-40.

17. Zanella CL, Posada J, Tritton TR, Mossman BT. Asbestos causes

stimulation of the extracellular signal-regulated kinase 1 mitogen-

activated protein kinase cascade after phosphorylation of the epider-

mal growth factor receptor. Cancer Res 1996;56:5334-8.

18. de Melo M, Gerbase MW, Curran J, Pache JC. Phosphorylated extra-

cellular signal-regulated kinases are significantly increased in malig-

nant mesothelioma. J Histochem Cytochem 2006;54:855-61.

19. Frey AB, Wali A, Pass H, Lonardo F. Osteopontin is linked to p65 and

MMP-9 expression in pulmonary adenocarcinoma but not in malig-

nant pleural mesothelioma. Histopathology 2007;50:720-6.

20. Garland LL, Rankin C, Gandara DR, et al. Phase II study of erlotinib

in patients with malignant pleural mesothelioma: a Southwest Oncol-

ogy Group Study. J Clin Oncol 2007;25:2406-13.

21. Vintman L, Nielsen S, Berner A, Reich R, Davidson B. Mitogen-acti-

vated protein kinase expression and activation does not differentiate

benign from malignant mesothelial cells. Cancer 2005;103:2427-33.

22. Blume-Jensen P, Hunter T. Oncogenic kinase signalling. Nature

2001;411:355-65.

23. Traxler P. Tyrosine kinases as targets in cancer therapy: successes and

failures. Expert Opin Ther Targets 2003;7:215-34.

24. Dierck K, Machida K, Voigt A, et al. Quantitative multiplexed pro-

filing of cellular signaling networks using phosphotyrosine-specific

DNA-tagged SH2 domains. Nat Methods 2006;3:737-44.

25. Emaduddin M, Bicknell DC, Bodmer WF, Feller SM. Cell growth,

global phosphotyrosine elevation, and c-Met phosphorylation through

Src family kinases in colorectal cancer cells. Proc Natl Acad Sci

U S A 2008;105:2358-62.

26. Rikova K, Guo A, Zeng Q, et al. Global survey of phosphotyro-

sine signaling identifies oncogenic kinases in lung cancer. Cell

2007;131:1190-203.

27. Stommel JM, Kimmelman AC, Ying H, et al. Coactivation of receptor

tyrosine kinases affects the response of tumor cells to targeted thera-

pies. Science 2007;318:287-90.

28. Jagadeeswaran R, Ma PC, Seiwert TY, et al. Functional analysis of

c-Met/hepatocyte growth factor pathway in malignant pleural meso-

thelioma. Cancer Res 2006;66:352-61.

504 Genes & Cancer / vol 1 no 5 (2010)

29. Okuda K, Sasaki H, Kawano O, et al. Epidermal growth factor recep-

tor gene mutation, amplification and protein expression in malignant

pleural mesothelioma. J Cancer Res Clin Oncol 2008;134:1105-11.

30. Calo V, Migliavacca M, Bazan V, et al. STAT proteins: from nor-

mal control of cellular events to tumorigenesis. J Cell Physiol

2003;197:157-68.

31. Bianchi C, Bianchi T. Malignant mesothelioma: global incidence and

relationship with asbestos. Ind Health 2007;45:379-87.

32. Ernst M, Najdovska M, Grail D, et al. STAT3 and STAT1 mediate

IL-11-dependent and inflammation-associated gastric tumorigenesis

in gp130 receptor mutant mice. J Clin Invest 2008;118:1727-38.

33. Cirri P, Chiarugi P, Marra F, et al. c-Src activates both STAT1 and

STAT3 in PDGF-stimulated NIH3T3 cells. Biochem Biophys Res

Commun 1997;239:493-7.

34. Hui Z, Tretiakova M, Zhang Z, et al. Radiosensitization by inhib-

iting STAT1 in renal cell carcinoma. Int J Radiat Oncol Biol Phys

2009;73:288-95.

35. Khodarev NN, Minn AJ, Efimova EV, et al. Signal transducer and

activator of transcription 1 regulates both cytotoxic and prosurvival

functions in tumor cells. Cancer Res 2007;67:9214-20.

36. Kovacic B, Stoiber D, Moriggl R, et al. STAT1 acts as a tumor pro-

moter for leukemia development. Cancer Cell 2006;10:77-87.

37. Kothmaier H, Quehenberger F, Halbwedl I, et al. EGFR and PDGFR

differentially promote growth in malignant epithelioid mesothelioma

of short and long term survivors. Thorax 2008;63:345-51.

38. Dostert C, Petrilli V, Van Bruggen R, Steele C, Mossman BT, Tschopp

J. Innate immune activation through Nalp3 inflammasome sensing of

asbestos and silica. Science 2008;320:674-7.

39. Roskoski R Jr. Src kinase regulation by phosphorylation and dephos-

phorylation. Biochem Biophys Res Commun 2005;331:1-14.

40. Rahimi N, Hung W, Tremblay E, Saulnier R, Elliott B. c-Src kinase

activity is required for hepatocyte growth factor-induced motility and

anchorage-independent growth of mammary carcinoma cells. J Biol

Chem 1998;273:33714-21.

41. Yamamoto N, Mammadova G, Song RX, Fukami Y, Sato K. Tyrosine

phosphorylation of p145met mediated by EGFR and Src is required

for serum-independent survival of human bladder carcinoma cells. J

Cell Sci 2006;119:4623-33.

42. Zhang J, Kalyankrishna S, Wislez M, et al. SRC-family kinases are

activated in non-small cell lung cancer and promote the survival of

epidermal growth factor receptor-dependent cell lines. Am J Pathol

2007;170:366-76.

43. Alvarez RH, Kantarjian HM, Cortes JE. The role of Src in solid and

hematologic malignancies: development of new-generation Src inhib-

itors. Cancer 2006;107:1918-29.

44. Johnson FM, Gallick GE. SRC family nonreceptor tyrosine kinases

as molecular targets for cancer therapy. Anticancer Agents Med Chem

2007;7:651-9.

45. Han LY, Landen CN, Trevino JG, et al. Antiangiogenic and antitu-

mor effects of SRC inhibition in ovarian carcinoma. Cancer Res

2006;66:8633-9.

46. Herynk MH, Zhang J, Parikh NU, Gallick GE. Activation of Src by

c-Met overexpression mediates metastatic properties of colorectal car-

cinoma cells. J Exp Ther Oncol 2007;6:205-17.

47. Hiscox S, Morgan L, Green TP, Barrow D, Gee J, Nicholson RI.

Elevated Src activity promotes cellular invasion and motility in

tamoxifen resistant breast cancer cells. Breast Cancer Res Treat

2006;97:263-74.

48. Mao W, Irby R, Coppola D, et al. Activation of c-Src by receptor tyro-

sine kinases in human colon cancer cells with high metastatic poten-

tial. Oncogene 1997;15:3083-90.

49. Summy JM, Gallick GE. Treatment for advanced tumors: SRC

reclaims center stage. Clin Cancer Res 2006;12:1398-401.

50. Cortes J, Rousselot P, Kim DW, et al. Dasatinib induces complete

hematologic and cytogenetic responses in patients with imatinib-

resistant or -intolerant chronic myeloid leukemia in blast crisis. Blood

2007;109:3207-13.

51. Quintas-Cardama A, Kantarjian H, Jones D, et al. Dasatinib (BMS-

354825) is active in Philadelphia chromosome-positive chronic

myelogenous leukemia after imatinib and nilotinib (AMN107) ther-

apy failure. Blood 2007;109:497-9.

52. Tsao AS, He D, Saigal B, et al. Inhibition of c-Src expression and acti-

vation in malignant pleural mesothelioma tissues leads to apoptosis,

cell cycle arrest, and decreased migration and invasion. Mol Cancer

Ther 2007;6:1962-72.

53. Fu YN, Yeh CL, Cheng HH, et al. EGFR mutants found in non-small

cell lung cancer show different levels of sensitivity to suppression of

Src: implications in targeting therapy. Oncogene 2008;27:957-65.

54. Mueller KL, Hunter LA, Ethier SP, Boerner JL. Met and c-Src cooper-

ate to compensate for loss of epidermal growth factor receptor kinase

activity in breast cancer cells. Cancer Res 2008;68:3314-22.

55. Toschi L, Janne PA. Single-agent and combination therapeutic strate-

gies to inhibit hepatocyte growth factor/MET signaling in cancer. Clin

Cancer Res 2008;14:5941-6.

56. Zhang H, Berezov A, Wang Q, et al. ErbB receptors: from oncogenes

to targeted cancer therapies. J Clin Invest 2007;117:2051-8.

57. Janne PA, Taffaro ML, Salgia R, Johnson BE. Inhibition of epidermal

growth factor receptor signaling in malignant pleural mesothelioma.

Cancer Res 2002;62:5242-7.

58. Mukohara T, Civiello G, Davis IJ, et al. Inhibition of the met receptor

in mesothelioma. Clin Cancer Res 2005;11:8122-30.

59. Adamson IY, Bakowska J. KGF and HGF are growth factors for

mesothelial cells in pleural lavage fluid after intratracheal asbestos.

Exp Lung Res 2001;27:605-16.

60. Cesario A, Catassi A, Festi L, et al. Farnesyltransferase inhibitors

and human malignant pleural mesothelioma: a first-step comparative

translational study. Clin Cancer Res 2005;11:2026-37.

61. Kawaguchi K, Murakami H, Taniguchi T, et al. Combined inhibition

of MET and EGFR suppresses proliferation of malignant mesothe-

lioma cells. Carcinogenesis 2009;30:1097-105.

62. Tang Z, Du R, Jiang S, et al. Dual MET-EGFR combinatorial inhibi-

tion against T790M-EGFR-mediated erlotinib-resistant lung cancer.

Br J Cancer 2008;99:911-22.

63. Yano S, Wang W, Li Q, et al. Hepatocyte growth factor induces gefi-

tinib resistance of lung adenocarcinoma with epidermal growth factor

receptor-activating mutations. Cancer Res 2008;68:9479-87.

64. Bonine-Summers AR, Aakre ME, Brown KA, et al. Epidermal growth

factor receptor plays a significant role in hepatocyte growth factor

Tyrosine kinases coactivated in mesothelioma / Menges et al. 505

mediated biological responses in mammary epithelial cells. Cancer

Biol Ther 2007;6:561-70.

65. Huang PH, Mukasa A, Bonavia R, et al. Quantitative analysis of

EGFRvIII cellular signaling networks reveals a combinatorial

therapeutic strategy for glioblastoma. Proc Natl Acad Sci U S A

2007;104:12867-72.

66. Jo M, Stolz DB, Esplen JE, Dorko K, Michalopoulos GK, Strom SC.

Cross-talk between epidermal growth factor receptor and c-Met signal

pathways in transformed cells. J Biol Chem 2000;275:8806-11.

67. Reznik TE, Sang Y, Ma Y, et al. Transcription-dependent epidermal

growth factor receptor activation by hepatocyte growth factor. Mol

Cancer Res 2008;6:139-50.

68. Guo A, Villen J, Kornhauser J, et al. Signaling networks assembled

by oncogenic EGFR and c-Met. Proc Natl Acad Sci U S A 2008;105:

692-7.

69. Popsueva A, Poteryaev D, Arighi E, et al. GDNF promotes tubulo-

genesis of GFRalpha1-expressing MDCK cells by Src-mediated

phosphorylation of Met receptor tyrosine kinase. J Cell Biol 2003;

161:119-29.

70. Taguchi T, Jhanwar SC, Siegfried JM, Keller SM, Testa JR. Recurrent

deletions of specific chromosomal sites in 1p, 3p, 6q, and 9p in human

malignant mesothelioma. Cancer Res 1993;53:4349-55.

71. Ke E, Patel BB, Liu T, et al. Proteomic analyses of pancreatic cyst

fluids. Pancreas 2009;38:e33-42.