<<

Published OnlineFirst June 28, 2013; DOI: 10.1158/0008-5472.CAN-13-0760

Cancer Therapeutics, Targets, and Chemical Biology Research

Targeting ERBB Receptors Shifts Their Partners and Triggers Persistent ERK Signaling through a Novel ERBB/EFNB1 Complex

Paola D. Vermeer, Paul L. Colbert, Bryant G. Wieking, Daniel W. Vermeer, and John H. Lee

Abstract Most squamous cell carcinomas of the head and neck (HNSCC) overexpress ERBB1/EGFR, but EGF receptor (EGFR)-targeted therapies have yielded disappointing clinical results in treatment of this cancer. Here, we describe a novel interaction between EGFR and the EphrinB1 (EFNB1), and we show that EFNB1 phosphorylation and downstream signaling persists in the presence of . Mechanistically, cetuximab drives a shift in EGFR dimerization partners within the signaling complex, suggesting that targeted drugs may trigger partner rearrangements that allow persistent pathway activation. EFNB1 attenuation slowed tumor growth and increased survival in a murine model of HNSCC, suggesting a substantial contribution of EFNB1 signaling to HNSCC development. Together, our findings suggest that EFNB1 is part of the EGFR signaling complex and may mediate drug resistance in HNSCC as well as other solid tumors. Cancer Res; 73(18); 1–11. 2013 AACR.

Introduction ment, a process called "reverse signaling" (10). PTPN13 tran- A greater understanding of the molecular pathways driving siently interacts with phosphorylated EFNB1, shutting "reverse tumor initiation and proliferation has heralded an age of tar- signaling" off (11). Thus, in tumors with impaired PTPN13 geted therapies for cancer. The therapeutic successes of these expression or function, EFNB1 signaling may persist. In addi- drugs are satisfying from both the scientific and medical per- tion, ligands are promiscuous in their associations (12, spectives. Their failures indicate an incomplete understand- 13); for example, EFNB1 interacts with ERBB2 (14). Moreover, ing of macromolecular complexes, the oncogenic pathways EFNB1 activation correlates with phosphorylation of extracel- – they drive, their cross-talk, and drug-driven compensatory lular signal regulated kinase (ERK)1/2 (14). Together, these mechanisms. We have recently identified a novel component studies suggest that the complex consisting of ERBB2/EFNB1 of one such macromolecular complex in breast cancer (1). together with PTPN13 regulates intracellular signals critical for Here, we further characterize its role in the context of squa- epithelial tumorigenesis. mous cell carcinoma of the head and neck (HNSCC). ERBB2 belongs to the ERBB family of receptor tyrosine In human papillomavirus (HPV)-associated HNSCC, the kinases, which includes ERBB1/EGFR/HER1. More than 90% high-risk HPV type 16 (HPV16) E6 oncoprotein targets the of HNSCC cases show upregulation of ERBB1 (15, 16), which cellular phosphatase, PTPN13, for degradation (2, 3). PTPN13 may impact tumor growth and patient outcomes. ERBB1- functions as a tumor suppressor (4–9), yet the molecular targeted therapies such as cetuximab (Erbitux), a chimeric mechanisms it modulates and how they become altered in anti-ERBB1 monoclonal antibody, and (Tarceva), an cancer remain unclear. One important PTPN13 phosphatase ERBB1 kinase inhibitor, are used as therapies in patients with substrate that may influence cancer initiation and/or progres- HNSCC although with modest clinical outcomes (17). These fi fi sion is the signaling ligand EphrinB1 (EFNB1). EFNB1 belongs clinical ndings emphasize the need to better de ne compo- to a ligand family that binds and activates Eph receptor nents of macromolecular complexes active in disease, under- tyrosine kinases. Unlike most ligands, Ephrin ligands initiate stand the molecular pathways they drive, and characterize how fl their own downstream signaling following receptor engage- they in uence tumor initiation and progression. In the case of HNSCC, these types of studies may help define what role, if any, ERBB1 upregulation plays in disease initiation and/or Authors' Affiliation: Cancer Biology Research Center, Sanford Research, progression. University of South Dakota, Sioux Falls, South Dakota Our previous finding that EFNB1 associates with ERBB2 Note: Supplementary data for this article are available at Cancer Research prompted us to ask whether it associates with other ERBB Online (http://cancerres.aacrjournals.org/). family members. Here, we report that, like ERBB2, ERBB1 Corresponding Author: John H. Lee, Sanford Research, University of associates with EFNB1. Moreover, in the absence of PTPN13 South Dakota, 2301 East 60th Street North, Sioux Falls, SD 57104. Phone: function, EFNB1 phosphorylation is enhanced and ERK1/2 605-312-6103; Fax: 605-312-6201; E-mail: [email protected] signaling is potentiated. These data suggest that EFNB1 exists doi: 10.1158/0008-5472.CAN-13-0760 in a complex together with ERBB1, ERBB2, or both. A combi- 2013 American Association for Cancer Research. nation of these associations likely exists that together regulate

www.aacrjournals.org OF1

Downloaded from cancerres.aacrjournals.org on September 28, 2021. © 2013 American Association for Cancer Research. Published OnlineFirst June 28, 2013; DOI: 10.1158/0008-5472.CAN-13-0760

Vermeer et al.

complex intracellular signals potentiated in the absence NaF, 10 mmol/L NaPPi, 10% glycerol, and 1% Triton X-100) and of PTPN13 function. Importantly, we show that while anti- soluble assayed by BCA assay (Pierce). Equal body therapies such as cetuximab (Erbitux, anti-ERBB1) and total protein was used for immunoprecipitation and immu- (Herceptin, anti-ERBB2) potently block receptor noblot analysis by standard methodology. Antibodies for activation, they do not attenuate EFNB1 activation or ERK1/2 immunoprecipitation were as follows: anti-ERBB1 (Becton phosphorylation. Moreover, we show that these ERBB-targeted Dickinson), anti-ERBB2 (Dako), and anti-EFNB1 (Santa Cruz drugs promote the shifting of partners within ERBB/EFNB1 Biotechnology); antibodies used for immunoblot were as fol- complexes and suggest that this mechanism supports per- lows: anti-ERBB1 (Upstate), anti-ERBB2 (Invitrogen), anti- sistent EFNB1 signaling despite potent ERBB receptor block- phospho-tyrosine (Upstate), anti-glyceraldehyde-3-phosphate ade. Thus, we propose that EFNB1 activation mediates signal dehydrogenase (GAPDH; Ambion), anti-EFNB1 (Santa Cruz transduction and drug resistance. In addition, we show that Biotechnology), anti-EFNB1 (Sigma), anti-phospho-ERK1/2 knockdown of EFNB1 significantly slows tumor growth and (Cell Signaling Technology), anti-ERK1/2 (Calbiochem), anti- improves survival in a murine model of HNSCC. Together, our phospho-Tyrosine317 EFNB1 (Santa Cruz Biotechnology), findings support a tumor-suppressive role for PTPN13, suggest anti-HA (Sigma), and anti-FLAG (Sigma). that EFNB1 may be a useful therapeutic target in HNSCC and other solid tumors, and explain at least one mechanism by Immunofluorescence and proximity ligation assay which HNSCCs fail cetuximab therapy. Antibodies for immunofluorescence were as follows: anti- ERBB1 (Becton Dickinson), anti-ERBB2 (Dako), anti-HA (Sig- Materials and Methods ma), anti-EFNB1 (Santa Cruz Biotechnology), and anti-EFNB1 Cell culture (Sigma). Surface EphrinB ligands were bound by EphB1-Fc HEK293, SCC1, and SCC47 cells were maintained with (R&D Systems) on unfixed, unpermeabilized cells and detected Dulbecco's Modified Eagle Medium (DMEM) with 10% fetal with Millipore anti-human immunoglobulin G–fluorescein calf serum and 1% penicillin/streptomycin. Primary murine isothiocyanate (IgG-FITC). Antibodies used for proximity liga- tonsil epithelia (1 MTE) and human tonsil epithelia (1 HTE) tion assay (PLA) were as follows: anti-ERBB1 (Becton Dick- were maintained with Keratinocyte Serum-Free Medium. inson), anti-ERBB2 (Dako), anti-ERBB2 (Invitrogen), and anti- 1MTE stably expressing HPV16 E6 and E7 together with RAS EFNB1 (Santa Cruz Biotechnology). Standard immunofluores- and luciferase (MEERL cells) and 1 HTE stabley expressing cence and PLA protocols were followed. HPV16 E6 and E7 alone (HEE cells) were maintained with E-medium (DMEM/Hams F12, 10% fetal calf serum, 1% peni- Quantification of PLA cillin/streptomycin, 0.5 mg/mL hydrocortisone, 8.4 ng/mL PLA-positive signals (visualized as fluorescent red dots) cholera toxin, 5 mg/mL transferrin, 5 mg/mL , 1.36 ng/mL were analyzed by confocal microscopy (Olympus Fluo- tri-iodo-thyonine, and 5 ng/mL EGF). HEE cells were generated View1000, 60 oil objective, 2.5 magnified; Alexa Fluor 568 from 1 HTE cells harvested from tonsillectomies conducted for detector). At least three stacked Z series for each condition noncancerous reasons and collected under Institutional Review were analyzed, confocal IOB files converted to tiff format, and Board approval with written consent at the University of Iowa then analyzed by DuoLink Imagetool software (Olink Bio- (Iowa City, IA; ref. 3). The stable HEE cells were generated as sciences). The number of PLA-positive signals from at least previously described (3, 18). We have authenticated these cell three different Z-stacked images per condition was averaged. lines by short-tandem repeat (STR) DNA profiling and verified Experiments were repeated at least three times with similar them with the reference STR profile. Authentication was carried results. Student t test was used to analyze the data and test for out in the summer of 2012 at Genetica DNA Laboratories significance. (http://www.celllineauthentication.com/). Targeted therapy treatment Plasmids Cells were grown on 60-mm tissue culture dishes to 80% EFNB1-mutant constructs and PTPN13C/S have been previ- confluence and treated with recombinant EGF (SAFC Bio- ously described (6, 14). Wild-type (wt) human ERBB1 cDNA was sciences) in the presence or absence of cetuximab (20 mg/mL; obtained from Addgene (#11011) and cloned by PCR into Imclone), trastuzumab (10 mg/mL; Dako), or their combination pCMV-HA (Clontech). ERBB1 transmembrane mutants were for 1 hour at 37C. Control cells received either no treatment or generated from the hemagglutinin (HA)-tagged wt ERBB1 EGF alone. Cells were harvested as described earlier and construct using the QuickChange II XL Site-Directed Muta- analyzed by immunoprecipitation and Western blot analysis. genesis Kit (Agilent Technologies). For analysis by PLA, cells were seeded on 16-well chamber slides and treated as described followed by processing for PLA Transfection (as described earlier). HEK293 cells were transfected using PolyFect Transfection Reagent as per the manufacturer's instructions (Qiagen). Generating stable cell lines Cells were seeded onto 6-well tissue culture dishes and Immunoprecipitation and Western blot analysis transfected using PolyFect Transfection Reagent as describ- Cells were lysed in lysis buffer (50 mmol/L Tris–HCl pH 7.5, ed earlier. Twenty-four hours posttransfection, cells were 150 mmol/L NaCl, 5 mmol/L EDTA, 2 mN Na3VO4, 100 mmol/L placed under antibiotic selection with Zeocin (250, 500, or

OF2 Cancer Res; 73(18) September 15, 2013 Cancer Research

Downloaded from cancerres.aacrjournals.org on September 28, 2021. © 2013 American Association for Cancer Research. Published OnlineFirst June 28, 2013; DOI: 10.1158/0008-5472.CAN-13-0760

Cetuximab Drives ErbB/EphrinB1 Shifting and Signaling

1,000 mg/mL; Invitrogen), clones were picked and transferred weight, and possess the HA tag (Fig. 2B). To test whether any of to 48-well dishes, maintained under selection, and passaged the ERBB1 deletions compromise its interaction with EFNB1, up to 100-mm dishes, at which time one dish was lysed for HEK293 cells were transfected, lysates immunoprecipitated for biochemical analysis and another frozen for later use. EFNB1 and immunoblotted for HA. Surprisingly, no loss of coimmunoprecipitation with any ERBB1 mutants occurred, In vivo studies suggesting that the extracellular ligand-binding domains of All animal studies were reviewed and approved by the ERBB1 are not required for its association with EFNB1 (Fig. 2B). Institutional Animal Care and Use Committee. Cells were These data are consistent with our previous finding for the trypsinized and harvested from tissue culture plates and ERBB2/EFNB1 interaction (14). EFNB1 deletion mutants and resuspended. An 18-gauge needle with 100,000 cells was wt constructs have been previously described and were all injected subcutaneously into the right hind limb of each FLAG-tagged (14). Briefly, either the entire extracellular C57Bl/6 mouse (N ¼ 5/group). C57Bl/6 male mice were 4 to domain (EFNB1-ED) or just the intracellular PDZ-binding 6 weeks of age, weighed at least 20 to 25 g, and were purchased motif (EFNB1-PDZ) of EFNB1 was deleted. HEK293 cells were from The Jackson Laboratory. Injected cells included: parental transfected with the EFNB1 constructs. Because HEK293 cells cells, nonsilencing, wt mEfnB1, or short hairpin (sh) EfnB1. express nearly undetectable endogenous ERBB1, cells were Weekly caliper measurements were taken and tumor volume also transfected with untagged wt ERBB1. Immunoprecipita- was calculated as follows: (width)2(depth). Mice were humane- tion for ERBB1 followed by Western blot analysis for FLAG ly euthanized when tumors reached 2 cm in the greatest shows no loss of coimmunoprecipitation with any of the dimension or the animal became emaciated or had functional EFNB1 mutants, suggesting that neither the extracellular impairment of the leg. domain nor the PDZ motif is required for its association with ERBB1 (Fig. 2C). Results ERBB receptor transmembrane domains mediate homo- EFNB1 associates with ERBB1 and ERBB2 and hetero-dimerization (19, 20). In fact, ERBB transmembrane To test whether ERBB1 and EFNB1 interact, two HNSCC cell domains contain two motifs: one mediating heterodimeriza- lines were processed for immunofluorescence: the SCC1 cell tion, the other homodimerization (21). To test whether line is HPV negative (HPV ), whereas the SCC47 cell line is ERBB10s transmembrane domain mediates its interaction with þ HPV-positive (HPV ). Surface, rather than total, EphrinB was EFNB1, two additional ERBB1 mutants were generated: one localized because cells robustly express intracellular EphrinB within the first transmembrane motif and another within the ligands making the distinction of surface expression difficult. second transmembrane motif. These constructs were HA- EphB1-Fc binding followed by IgG-FITC localizes surface tagged and transfected into HEK293 cells. Control cells were EphrinB ligands. Both cell lines express ERBB1 and EphrinB, transfected with wt HA-tagged ERBB1 or enhanced GFP which colocalize at cell–cell borders (Fig. 1A and B). (eGFP). Lysates were immunoprecipitated for EFNB1 and To verify that the colocalization by immunofluorescence analyzed by Western blot analysis for HA. Although the first involved ERBB1 and EFNB1 specifically, PLA and coimmuno- ERBB1 transmembrane motif mutant, T648I G652I, retains precipitation studies were conducted. Figure 1C shows positive coimmunoprecipitation with EFNB1, mutation within the PLA signals for EFNB1 and ERBB1 in SCC1 (HPV ) and SCC47 second transmembrane motif (A661I G665I) loses it (Fig. þ (HPV ) cells. The previously discovered ERBB2/EFNB1 asso- 2D). These data were verified by immunofluorescence. HEK293 ciation (14) was confirmed and the presence of ERBB1/ERBB2 cells were transfected as described and cells processed for heterodimers was determined (Fig. 1C). In addition, stained Z- immunofluorescence using an anti-HA antibody for ERBB1 stacked images in the XZ plane indicate that these interactions detection and an anti-EFNB1 antibody for EFNB1 detection. are not restricted to the cell surface but also occur within the Expression of the ERBB1 T648I G652I mutant (green) coloca- cytoplasm (Fig. 1D). These intracellular PLA signals likely lizes with endogenous EFNB1 (red), evident in the merged represent internalized complexes. Importantly, PLA quantifi- panel (yellow). Expression of the ERBB1 A661I G665I (green) cation shows that infection with HPV16 correlates with does not colocalize with EFNB1 (red; Fig. 2E). Together, these enhanced ERBB/EFNB1 interactions (Fig. 1E). EFNB1, ERBB1, data suggest that ERBB10s second transmembrane motif med- þ and ERBB2 protein levels in SCC1 (HPV ) and SCC47 (HPV ) iates the EFNB1 association. cells are very similar (Fig. 3), thus increased protein expression Expression of HPV oncogenes correlates with EFNB1 and cannot account for the enhanced interactions evident in SCC47 ERK1/2 phosphorylation in HNSCC. We and others have þ (HPV ) cells. shown that PTPN13 regulates EFNB1 phosphorylation. More- over, the loss of PTPN13 expression, as seen in basal-like breast EFNB1 and ERBB1 association is likely mediated via cancer, correlates with enhanced EFNB1 and ERK1/2 phos- their transmembrane domains phorylation (14, 22). Importantly, breast tumors compromised Because of the overwhelming expression of ERBB1 in for PTPN13 expression are associated with decreased overall HNSCCs, we focused further analysis on ERBB1 and EFNB1. survival (23). A similar dysregulation in signaling occurs in To define the domains necessary and sufficient for ERBB1/ HNSCC, both in a human cell line (SCC84) and a murine model EFNB1 association, we generated deletion mutants (Fig. 2A). of head and neck cancer (14). Thus, we wondered whether All ERBB1 mutants, including the full-length wt, were HA- EFNB10s phosphorylation state modulates its interactions with tagged; constructs express well, run at the predicted molecular ERBB receptors. In addition, given that tonsil epithelial cells

www.aacrjournals.org Cancer Res; 73(18) September 15, 2013 OF3

Downloaded from cancerres.aacrjournals.org on September 28, 2021. © 2013 American Association for Cancer Research. Published OnlineFirst June 28, 2013; DOI: 10.1158/0008-5472.CAN-13-0760

Vermeer et al.

A − B SCC1 (HPV ) SCC47 (HPV+)

EphrinB ERBB1 EphrinB ERBB1

Figure 1. ERBB1 and EFNB1 interact. A and B, en face confocal images of SCC1 (A) and SCC47 DAPI Merge DAPI Merge (B). B, cells stained for surface EphrinB ligands (green) and total C ERBB1 (red). Merged images, yellow; 40,6-diamidino-2-

) ERBB1/EFNB1 ERBB2/EFNB1 ERBB1/ERBB2 − phenylindole (DAPI; blue) nuclear counterstain. Scale bar, 10 mm. C, en face stacked confocal images of SCC1 and SCC47 cells processed for PLA for ERBB1/ EFNB1, ERBB2/EFNB1, and

) SCC1 (HPV ERBB1/ERBB2. Positive PLA + signals (red) and DAPI (blue) nuclear counterstain. Scale bar, 10 mm. D, stacked confocal images in the XZ plane taken from Z-series collected in C. SCC47 (HPV E, quantification of PLA-positive signals in C.

D E 2,000 SCC1 − + P = 0.026 * SCC1(HPV ) SCC47(HPV ) SCC47 1,600

ERBB1/EFNB1 ERBB1/EFNB1 P = 0.011 1,200 * P = 0.024 800 * ERBB2/EFNB1 ERBB2/EFNB1

400 # PLA-positive signals/cell ERBB1/ERBB2 ERBB1/ERBB2 0 ERBB1/EFNB1ERBB2/EFNB1 ERBB1/ERBB2

þ are the major site for HPV HNSCC, we tested whether ERBB/ tein kinase (MAPK) pathway (14). Similar results were evident EFNB1 interactions exist in these cells. Thus, 1 HTE and HTEs in 1 MTE cells and a previously characterized mouse model of stably expressing HEE were tested. Consistent with our pre- HPV-related HNSCC (MTEs expressing MEERL cells; Supple- vious studies, ERBB1 and ERBB2 associate with EFNB1 in both mentary Fig. S1; refs. 18, 24, 25). cell types (Fig. 3A). In addition, expression of E6 correlates with enhanced phosphorylation of EFNB1 and ERK1/2 consistent PTPN13 regulates EFNB1 phosphorylation and reverse with previous reports (2, 3). These data show that ERBB1 and signaling ERBB2 associate with EFNB1 in primary human tonsil cells as HPV16 E6 mediates the degradation of PTPN13 (2, 3). The well as those expressing HPV oncogenes. Moreover EFNB1 data suggest that enhanced EFNB1 activation and downstream associates with its ERBB partners regardless of its phosphor- signaling are a consequence of this degradation (Fig. 3A). ylation status. Importantly, expression of HPV16 oncogenes To directly test this, we conducted a series of transfections (HEE cells) enhances EFNB1 activation and subsequent ERK1/ in HEK293 cells, which express nearly undetectable ERBB 2 phosphorylation. These data are consistent with our previous receptors allowing us to control expression of the proteins findings that EFNB1 signals down the mitogen-activated pro- involved. Briefly, HEK293 cells were transfected with wt ERBB1,

OF4 Cancer Res; 73(18) September 15, 2013 Cancer Research

Downloaded from cancerres.aacrjournals.org on September 28, 2021. © 2013 American Association for Cancer Research. Published OnlineFirst June 28, 2013; DOI: 10.1158/0008-5472.CAN-13-0760

Cetuximab Drives ErbB/EphrinB1 Shifting and Signaling

A 1 167 462 TM 1211 LBD1 LBD2 Full-length wild-type ERBB1 ERBB1Δ 0 LBD2 1-167 LBD1 Figure 2. ERBB1 s transmembrane ERBB1Δ 1-462 LBD2 domain mediates its association with EFNB1. A, schematic diagram of full length and mutant ERBB1 B C constructs. LBD, ligand-binding 170 kD IP: ERBB1 55 kD domain; TM, transmembrane. B, IB: HA HEK293 cells were transfected 135 kD IB: FLAG 33 kD with ERBB1 wt and mutant constructs and lysates analyzed 100 kD IP: ERBB1 170 kD by Western blot analysis as IP: EFNB1 IB: ERBB1 indicated. Untransfected cells 170 kD served as control. C, HEK293 cells IB: HA 135 kD were transfected with wt EFNB1 Control EFNB1 wt EFNB1 and mutant constructs ( - IP: EFNB1 EFNB1-ED EFNB1 EFNB1-PDZ ED, -PDZ) and lysates 55 kD analyzed by Western blot analysis IB: EFNB1 as indicated. Untransfected cells served as control. ED, extracellular Control domain; PDZ, PDZ domain. D, wt ERBB1 HEK293 cells were transfected ERBB1-LBD2ERBB1-LBD1 nonspecific band with ERBB1 transmembrane domain mutants and analyzed by Western blot analysis as indicated. D IP: EFNB1 E Cells transfected with an eGFP construct as well as untransfected IB: HA 170 kD cells served as controls. E, en face confocal images of cells DAPI ERBB1 T648I G652I transfected as in D, processed for eGFP immunofluorescence, localizing ERBB1 constructs (HA, green) and wt ERBB1 HA Untransfected endogenous EFNB1 (red). Merged Endog EFNB1 Merge panel, yellow; 40,6-diamidino-2- phenylindole (DAPI; blue) nuclear counterstain. Scale bars, 10 mm. ERBB1ERBB1 A661I G665IT648I G652IHA HA IP, immunoprecipitation; IB, immunoblot. DAPI ERBB1 A661I G665I

Endog EFNB1 Merge

wt EFNB1, and PTPN13 (either wt or a phosphatase null, ERBB1 and ERBB2 antibody blockade do not attenuate C S PTPN13 / ) and lysates analyzed by Western blot analysis. In downstream signaling eGFP-transfected controls, endogenous EFNB1 is phosphory- ERBB1- and ERBB2-targeted therapies are in clinical use for lated (Fig. 3B). The multiple bands likely represent different treatment of a variety of solid cancers including HNSCC. Our phosphorylated forms of EFNB1 as suggested by Xu and data suggest that PTPN13 modulates EFNB1-mediated signal- colleagues (26). In addition, a low level of EFNB1 coimmuno- ing and that EFNB1 associates with ERBB1 and ERBB2. To test precipitations with ERBB1 yet ERK1/2 is not phosphorylated. whether ERBB1- and/or ERBB2-targeted therapies alter EFNB1 Transfection with wt ERBB1,wtEFNB1, and wt PTPN13 activation and downstream signaling, we analyzed SCC1 þ decreases EFNB1 phosphorylation without altering its asso- (HPV ) and SCC47 (HPV ) cells. Cells were treated with EGF ciation with ERBB1; ERK1/2 remains unphosphorylated. to stimulate ERBB receptor activation and then segregated Interestingly, transfection with wt ERBB1,wtEFNB1, and into the following groups: (i) untreated (EGF alone, control), C/S PTPN13 , a phosphatase null PTPN13-mutant, increases (ii) treated with cetuximab, (iii) treated with trastuzumab, or EFNB1 phosphorylation as well as its association with ERBB1 (iv) treated with both cetuximab and trastuzumab. Unstimu- and initiates ERK1/2 phosphorylation. Together, these data lated (no EGF) and untreated cells (EGF alone) served as are consistent with those of others showing that PTPN13 control. Figure 4 shows Western blot analysis of these groups. þ regulates EFNB1 phosphorylation. In addition, the data sug- Both SCC1 (HPV ) and SCC47 (HPV ) cells show nearly un- gest that phosphorylated EFNB1 associates more readily with detectable endogenous ERBB1 or ERBB2 phosphorylation ERBB1 and that the loss of PTPN13 function potentiates not (unstimulated). In addition, and consistent with clinical data, only EFNB1 phosphorylation but also ERK1/2 signaling. cells abundantly express ERBB1 and modestly express ERBB2

www.aacrjournals.org Cancer Res; 73(18) September 15, 2013 OF5

Downloaded from cancerres.aacrjournals.org on September 28, 2021. © 2013 American Association for Cancer Research. Published OnlineFirst June 28, 2013; DOI: 10.1158/0008-5472.CAN-13-0760

Vermeer et al.

A B IP: EFNB1 IP: ERBB1 55 kD 55 kD IB: EFNB1 IB: P-tyrosine IP: ERBB1 55 kD IB: ERBB1 170 kD IB: ENFB1

IB: P-ERK1/2 40 kD

IP: ERBB2 55 kD eGFP +−− IB: EFNB1 Wt ERBB1 − ++ Wt EFNB1 − ++ IB: ERBB2 170 kD PTPN13 wt −+− PTPN13 C/S −−+

IP: EFNB1 55 kD IB:P-tyrosine Figure 3. EFNB1 associates with ERBB1 and ERBB2 and mediates ERK1/2 signaling in HTE IB: EFNB1 55 kD cells. A, 1 HTE, HTEs stably expressing HEE, were analyzed by Western blot analysis as indicated. B, HEK293 cells were transfected 40 kD IB: P-ERK1/2 as indicated and lysates analyzed by Western blot analysis as indicated. IP, immunoprecipitation; IB, immunoblot. IB: ERK1/2 40 kD

Ratio: P-ERK1/2 to 0.84 0.57 Total ERK1/2 33 kD IB: GAPDH

(15, 16, 27–31). EGF stimulation activates ERBB1 and ERBB2, data suggest that persistent signaling via EFNB1 drives ERK1/2 suggesting that at least a fraction of ERBB1 heterodimerizes phosphorylation despite blockade of ERBB1 and ERBB2. with ERBB2. Treatment with cetuximab alone potently attenu- ates EGF-mediated ERBB1 and ERBB2 phosphorylation, sug- Treatment with cetuximab or trastuzumab (together or gesting that ERBB2 phosphorylation following EGF stimula- in combination) drives the shifting of partners within tion occurs via heterodimerization with ERBB1. As expected, ERBB/EFNB1 complexes trastuzumab has no effect on ERBB1 phosphorylation but Given that EFNB1 associates with ERBB1 and ERBB2, we attenuates ERBB2 activation. Treatment with a combination wondered whether treatment with targeted drugs mediates of cetuximab and trastuzumab is not additive. In other words, changes in partner associations within these complexes that treatment with drugs in combination does not enhance the might account for sustained signaling in the presence of drug. effect of each drug when given in isolation. Importantly, To test this, the experiments carried out in Fig. 4 were dupli- despite the ability of cetuximab and trastuzumab to attenuate cated on 16-well chamber slides, cells were processed for PLA, receptor activation, neither showed an effect on ERK phos- and quantified. In SCC1 (HPV ) cells, stimulation with EGF phorylation; that is, ERK1/2 remains phosphorylated despite alone (control; light gray) results in modest association of the presence of drug. These data suggest that downstream ERBB1 with EFNB1 (Fig. 5A). ERBB2 interactions with EFNB1 signaling persists despite ERBB1 and/or ERBB2 blockade. This are more abundant in comparison, suggesting that the pre- finding is consistent with previous reports (32). Importantly, ferred partner for EFNB1 is ERBB2. In addition, ERBB1 and while EGF stimulation enhances phosphorylation of EFNB1 on ERBB2 heterodimers are in the minority. Treatment of cells tyrosine 317, neither cetuximab nor trastuzumab (alone or in with EGF þ cetuximab (black bars) leads to a significant combination) attenuate this. On the basis of our previous increase in ERBB1/EFNB1 interactions (P ¼ 0.03), a significant finding that EFNB1 signals via the MAPK pathway (14), these decrease in ERBB2/EFNB1 interactions (P ¼ 0.0001), whereas

OF6 Cancer Res; 73(18) September 15, 2013 Cancer Research

Downloaded from cancerres.aacrjournals.org on September 28, 2021. © 2013 American Association for Cancer Research. Published OnlineFirst June 28, 2013; DOI: 10.1158/0008-5472.CAN-13-0760

Cetuximab Drives ErbB/EphrinB1 Shifting and Signaling

taining either wt murine Efnb1 (wt mEnfb1) or an short hairpin − SCC1 (HPV ) SCC47 (HPV+) RNA (shRNA)-targeting Efnb1 (shEfnb1). Transfection with a IP: ERBB1 nonsilencing shRNA (nonsilencing) as well as the parental cells IB: P-tyrosine 170 kD served as controls. Cells were ring-cloned and clones tested IB: ERBB1 170 kD biochemically. More than 50 clones were analyzed by Western IP: ERBB2 blot analysis with only modest overexpression or knockdown IB: P-tyrosine 170 kD of EFNB1 evident. However, immunofluorescence localization IB: ERBB2 170 kD of surface expression shows abundant increase in wt mEfnb1 IB: P-Tyr317 55 kD clones and decrease in shEfnb1 clones (Fig. 6A). These findings EFNB1 are consistent with our previous observations that abundant IB: P-ERK1/2 40 kD intracellular EFNB1 expression precludes Western blot anal-

IB: ERK1/2 40 kD ysis detection of surface expression changes. Thus, selection of clones was based predominantly on immunofluorescence 33 kD IB: GAPDH surface EFNB1 localization. To test the role of EFNB1 in tumor growth in vivo, 100,000 cells of each clone were injected into the hind limb of C57Bl/6 mice, N ¼ 5 mice per group. Tumor growth was measured weekly. wt mEfnb1 tumors grew signif- EGF EGF icantly faster than parental and shEfnb1 tumors (P < 0.001). However, tumor growth of the wt mEfnb1 group was not Figure 4. EFNB1 and ERK1/2 phosphorylation persist in presence of significantly different than that of the nonsilencing control cetuximab and trastuzumab therapies. SCC1 and SCC47 cells were group (P ¼ 0.42). Given that the wt mEfnb1 group was treated with EGF alone (control) or in combination with cetuximab (cetux), significantly different than the parental group, we speculate trastuzumab (traztuz), or both (cetuxþtrastuz) and analyzed by Western blot analysis as indicated. Cells alone (unstimulated) served as an that the nonsilencing construct mediates off-targets effects additional control. IP, immunoprecipitation; IB, immunoblot. that account for this difference in tumor growth with the wt mEfnb1 group. Importantly, mice bearing shEfnb1 tumors had ERBB1/ERBB2 heterodimerization is significantly enhanced significantly slower tumor growth compared with all other (P ¼ 0.04). On the other hand, treatment with EGF þ trastu- groups (P < 0.001) at all time points up to 20 days, at which time zumab (blue bars) has no effect on the number of interactions mice from the other groups reached sacrifice criteria (Fig. 6B). formed between ERBB1 and EFNB1; however, EFNB10s associ- Consistent with tumor growth, mice bearing parental tumors ation with ERBB2 decreases (P ¼ 0.03), whereas ERBB1/ERBB2 survived significantly longer than those bearing wt mEfnb1 or heterodimers significantly increase (P ¼ 0.01). Finally, treat- nonsilencing tumors (P < 0.001), whereas shEfnb1 bearing mice ment with a combination of EGF, cetuximab, and trastuzumab survived significantly longer (P < 0.001) than the other groups (dark gray bars) also results in partner shifts. ERBB1/EFNB1 (Fig. 6C). These data show that EFNB1 significantly affects associations decrease (P ¼ 0.01), whereas ERBB1/ERBB2 het- tumor growth and survival, suggesting it is a good candidate for erodimerization increases (P ¼ 0.03). Interactions between therapeutic targeting in HNSCC and other solid tumors. þ ERBB2 and EFNB1 remain unchanged. In SCC47 (HPV ) cells (Fig. 5B), treatment with targeted drugs showed similar trends Discussion as those evident with SCC1 (HPV ) cells though significance We describe a novel association in HNSCC consisting of was not reached in any condition. Nonetheless, these PLA data ERBB1 and EFNB1. ERBB2 is also a part of this complex, together with the biochemical analysis of these experiments consistent with previous findings (14). Loss of PTPN13 func- (Fig. 4) suggest that partner shifting occurs in the presence of tion increases EFNB1 phosphorylation, enhances EFNB10s targeted therapies and may explain the sustained activation interaction with ERBB1, and correlates with potentiated of EFNB1 and ERK1/2 evident in the presence of drug. ERK1/2 activation. Our data do not test whether ERBB1 directly phosphorylates EFNB1 or whether other components Loss of EFNB1 decreases tumor growth in an in vivo exist within this complex that mediate EFNB1 phosphoryla- model of HNSCC tion. However, previous studies suggest that SRC kinase The data suggest that EFNB1 associates with ERBB1 and phosphorylates EFNB1 following cognate receptor engage- ERBB2 (alone or in combination) and that PTPN13 modulates ment (10) and our previous studies support this role for SRC these interactions by regulating EFNB1 phosphorylation and (14). Whether SRC is recruited to EFNB1 when complexed to downstream signaling. Although ERBB1 and ERBB2 roles in ERBB1 remains to be tested. Regardless of the mechanism, tumor growth have been appreciated for some times, we increased EFNB1 phosphorylation correlates with ERK1/2 wanted to directly test EFNB10s function in tumor growth phosphorylation. þ using a previously described murine model of HPV HNSCC Importantly, we show that ERBB10s transmembrane domain (18, 24, 25). In these cells, HPV16 E6–mediated degradation of mediates the association with EFNB1. These data predict that PTPBL (the murine homolog of PTPN13) results in enhanced ERBB1-targeted therapies (which bind extracellular, cetuxi- EFNB1 phosphorylation and downstream ERK1/2 signaling mab, or intracellular, erlotinib, epitopes) alone will be ineffi- (14). Briefly, MTE cells stably expressing MEERL cells were cient in blocking intracellular signaling mediated by this transfected with the pcDNA3.1 Zeocin expression vector con- complex, as EFNB1 is likely impervious to them. Cetuximab's

www.aacrjournals.org Cancer Res; 73(18) September 15, 2013 OF7

Downloaded from cancerres.aacrjournals.org on September 28, 2021. © 2013 American Association for Cancer Research. Published OnlineFirst June 28, 2013; DOI: 10.1158/0008-5472.CAN-13-0760

Vermeer et al.

A 350 SCC1 (HPV−) P = 0.01* * P = 0.03 Control (EGF alone) 300 P = 0.0001 P = 0.03 Cetuximab * * P = 0.04 * Trastuzumab 250 Cetuximab+trastuzumab

200 P = 0.01 150 *

100 *

PLA-positive signals/cell Figure 5. Cetuximab and trastuzumab treatment results in P = 0.03 50 * partner shifting. A, SCC1 cells were treated with EGF alone (control; light gray) or in 0 combination with cetuximab ERBB1/EFNB1 ERBB2/EFNB1 ERBB1/ERBB2 (black), trastuzumab (blue), or both (cetuximabþtrastuzumab; B dark gray) and processed for PLA. 400 Cells alone (unstimulated) served SCC47 (HPV+) as an additional control. PLA- 350 positive signals were quantified. B, SCC47 cells were treated and 300 analyzed as described in A.

250

200

150

PLA-positive signals/cell 100

50

0 ERBB1/EFNB1 ERBB2/EFNB1 ERBB1/ERBB2

binding site on ERBB1 has been solved (33). The extracellular bination of these targeted therapies showed no ill effect on domain of ERBB1 is subdivided into four subdomains (I–IV; EFNB1 phosphorylation or downstream signaling. The findings þ ref. 34). EGF binds epitopes on domains I and III, whereas were strikingly similar in both HPV and HPV cells suggest- cetuximab binds only to domain III, yet overlaps with the ing that HPV status does not alter drug efficacy. However, the domain III epitope bound by EGF. Thus, cetuximab binding is PLA data show that HPV infection promotes ERBB/EFNB1 sufficient to sterically inhibit EGF binding. ERBB10s extracel- interactions (Fig. 1E), whereas analysis of these interactions in lular domains do not mediate its interaction with EFNB1, the context of drug treatment showed no significant differ- þ strongly suggesting that cetuximab binding will not inhibit it. ences in the HPV cells (Fig. 5B). We speculate this is a Trastuzumab binds an epitope within the juxtamembrane reflection of the increased number of associations due to HPV region of extracellular domain IV of ERBB2 (35). Our previous infection in these cells and suggest that a maximal threshold of findings show that ERBB20s extracellular domains do not total ERBB/EFNB1 interactions per cell may exist such that mediate its association with EFNB1 (14), and therefore, tras- once this threshold is reached, conditions that would other- tuzumab binding likely will not affect this interaction. wise drive significant changes become difficult to discern. In In cancers with decreased or lost PTPN13 function, ERBB/ addition, we noted a higher variability in the number of EFNB1 associations and signaling may be potentiated. We interactions for some conditions, which also affected attain- tested these possibilities and found that indeed, while treat- ment of statistical significance. Taken together, however, these ment with cetuximab potently blocked ERBB1 activation, data explain, at least in part, the modest clinical benefits EFNB1 phosphorylation persisted as did that of ERK1/2. evident with cetuximab in the HNSCC setting. In addition, the Trastuzumab treatment yielded similar results and the com- data suggest that dual targeting of ERBB1 and ERBB2 likely will

OF8 Cancer Res; 73(18) September 15, 2013 Cancer Research

Downloaded from cancerres.aacrjournals.org on September 28, 2021. © 2013 American Association for Cancer Research. Published OnlineFirst June 28, 2013; DOI: 10.1158/0008-5472.CAN-13-0760

Cetuximab Drives ErbB/EphrinB1 Shifting and Signaling

only a minority of patients, whereas most showed de novo or A acquired trastuzumab resistance (38). fi Parental wt mEfnb1 The nding that targeted drugs promote shifting of partners within ERBB/EFNB1 complexes emphasizes the complexity of macromolecular complexes, their roles in disease and suggests that targeted drugs may drive the progression of tumors. As with other solid tumors, it is increasingly evident that targeting a single element lacks long-term clinical efficacy. These find- Nonsilencing sh Efnb1 ings strongly show that silencing or eliminating a single oncogenic driver merely imposes evolutionary forces that promote alternative mechanisms to maintain, or even poten- 10 um tiate, the cancerous phenotype. This may be best exemplified by non–small cell lung cancer in which patients with classic ERBB1 mutations (L858R and exon 19 deletions) initially B 3,000 Parental respond to ERBB1-targeted therapies, yet eventually acquire ) Nonsilencing 3 2,500 wt mEfnb1 sh Efnb1 a secondary mutation, T790M, which confers resistance (39). 2,000 Multipronged approaches for effective treatment have been proposed for several cancers (40–43). Our current findings that 1,500 targeted therapies drive shifting of partners within a multi- 1,000 kinase/ligand/phosphatase complex strongly support these 500 proposals. Along these lines, the use of in com-

Tumor volume (mm bination with trastuzumab shows promise for patients with 0 * HER2 breast cancer (44, 45). Pertuzumab binds an extracellular 010203040 Time (d) epitope within domain II of ERBB2 and blocks dimerization with other ERBB family members (46). Our data show that in C SCC1 cells, ERBB1/2 heterodimers are in the minority and only 1.0 Parental Nonsilencing become a major dimer group following cetuximab treatment wt mEfnb1 0.8 sh Efnb1 (Fig. 5A). The current study did not evaluate ERBB3 or ERBB4, therefore it is unclear what role they may play in the multi- 0.6 kinase/ligand/phosphatase complex described. However, giv- en our finding that the transmembrane domain of ERBB1 0.4 Survival mediates the association with EFNB1, it seems unlikely that 0.2 pertuzumab will abrogate ERBB/EFNB1 interactions. The role of ERBB transmembrane domains in mediating dimer forma- 0.0 tion is well established (47–49). Our finding that the trans- 0 102030405060 membrane also is critical for EFNB1 associations, strongly Time (d) suggests that small molecules able to abolish transmembrane interactions may effectively block dimer associations of mul- Figure 6. Knockdown of Efnb1 slows tumor growth and prolongs survival tiple signaling molecules not limited to the ERBB family. Such in vivo. A, MTE cells stably expressing MEERL (parental), overexpressing drugs may have potent clinical efficacy. Our data also suggest Efnb1 (wt mEfnb1), shRNA knocking down EphrinB1 (shEfnb1), or a that EFNB1-targeted therapy may be of clinical value. In all, the nonsilencing shRNA control (nonsilencing) were analyzed by 0 data suggest that blocking formation of the complex as immunofluorescence for surface EFNB1 (green); 4 ,6-diamidino-2- phenylindole (DAPI; blue) nuclear counterstain. Scale bars, 10 mm. B, opposed to blocking individual components within the com- tumor volume as a function of time for C57Bl/6 mice injected with stable plex may provide better tumor growth control and clinical clones described in A. C, Kaplan–Meier survival plot of mice in B. ^, one outcomes. mouse that was censored from the data. This mouse had an open sore In summary, the present findings are consistent with pub- that met the criterion for euthanasia before hitting maximal allowed tumor volume. lished data and support a tumor-suppressive function for PTPN13 (2, 14). These studies confirm our previous findings that EFNB1 associates with members of the ERBB receptor not yield better results than either drug alone. This has in fact tyrosine kinases. For those cancers with upregulation of ERBB1 been shown for HNSCC cell lines (36). Moreover, a randomized (such as HNSCC), its association with EFNB1 may significantly phase II study using , a dual ERBB1 and ERBB2 affect tumorigenesis. Moreover, acquisition of PTPN13 loss-of- inhibitor, showed no significant difference in objective function mutations or its decreased expression (due to HPV response rate between lapatinib and placebo (37). These find- infection or epigenetic silencing) may further enhance ERBB1/ ings are not unique to HNSCC but seem to be the rule rather EFNB1–mediated signals. Our study suggests that EFNB1 is an than the exception in a majority of solid tumors. For example, important target for therapeutic intervention and that com- in a subset of molecularly classified breast tumors that over- binatorial targeted therapy approaches must include ERBB1, expressed ERBB2, trastuzumab therapy proved beneficial for ERBB2, as well as EFNB1 and possibly SRC for quenching

www.aacrjournals.org Cancer Res; 73(18) September 15, 2013 OF9

Downloaded from cancerres.aacrjournals.org on September 28, 2021. © 2013 American Association for Cancer Research. Published OnlineFirst June 28, 2013; DOI: 10.1158/0008-5472.CAN-13-0760

Vermeer et al.

complex mediated oncogenic signals. Although these studies Writing, review, and/or revision of the manuscript: P.D. Vermeer, D.W. Vermeer, J.H. Lee focus on HNSCC, EFNB1 complexes with ERBB2 in breast Administrative, technical, or material support (i.e., reporting or orga- cancer where loss of PTPN13 correlates with decreased overall nizing data, constructing databases): P.D. Vermeer, D.W. Vermeer survival (14) and thus, we speculate that a similar complex Study supervision: P.D. Vermeer, J.H. Lee exists in breast tumors (and possibly other solid tumors), Acknowledgments suggesting that a multitargeted drug approach may be bene- The authors thank Catherine Christopherson for excellent administrative ficial in this setting as well. assistance and Nichole Haag for technical assistance.

Disclosure of Potential Conflicts of Interest Grant Support No potential conflicts of interest were disclosed. This work was supported by R01 DE018386 NIH/National Institute of Dental and Craniofacial Research (J.H. Lee, B.G. Wieking, and D.W. Vermeer) and 8P20GM103548 Center of Biomedical Research Excellence NIH/Research Cen- Authors' Contributions ters in Minority Institutions and Institutional Development Award (P.D. Verm- Conception and design: P.D. Vermeer, P.L. Colbert, J.H. Lee eer, P.L. Colbert, and B.G. Wieking). Development of methodology: P.D. Vermeer, P.L. Colbert, D.W. Vermeer The costs of publication of this article were defrayed in part by the payment of Acquisition of data (provided animals, acquired and managed patients, page charges. This article must therefore be hereby marked advertisement in provided facilities, etc.): P.D. Vermeer, P.L. Colbert, B.G. Wieking, D.W. accordance with 18 U.S.C. Section 1734 solely to indicate this fact. Vermeer Analysis and interpretation of data (e.g., statistical analysis, biostatistics, Received March 13, 2013; revised May 16, 2013; accepted June 10, 2013; computational analysis): P.D. Vermeer, P.L. Colbert, D.W. Vermeer, J.H. Lee published OnlineFirst June 28, 2013.

References 1. Sorani MD. Informatics technology mimics ecology: dense, mutualistic 14. Vermeer PD, Bell M, Lee K, Vermeer DW, Wieking BG, Bilal E, et al. collaboration networks are associated with higher publication rates. ErbB2, EphrinB1, Src kinase and PTPN13 signaling complex regulates PLoS ONE 2012;7:e30463. MAP kinase signaling in human cancers. PLoS ONE 2012;7:e30447. 2. Spanos WC, Hoover A, Harris GF, Wu S, Strand GL, Anderson ME, 15. Rubin Grandis J, Melhem MF, Gooding WE, Day R, Holst VA, Wagener et al. The PDZ binding motif of human papillomavirus type 16 E6 MM, et al. Levels of TGF-alpha and EGFR protein in head and neck induces PTPN13 loss, which allows anchorage-independent squamous cell carcinoma and patient survival. J Natl Cancer Inst growth and synergizes with ras for invasive growth. J Virol 2008; 1998;90:824–32. 82:2493–500. 16. Grandis JR, Tweardy DJ. Elevated levels of transforming 3. Spanos WC, Geiger J, Anderson ME, Harris GF, Bossler AD, Smith RB, alpha and epidermal messenger RNA are early et al. Deletion of the PDZ motif of HPV16 E6 preventing immortalization markers of carcinogenesis in head and neck cancer. Cancer Res 1993; and anchorage-independent growth in human tonsil epithelial cells. 53:3579–84. Head Neck 2008;30:139–47. 17. Lee J, Moon C. Current status of experimental therapeutics for head 4. Wang Z, Shen D, Parsons DW, Bardelli A, Sager J, Szabo S, et al. and neck cancer. Exp Biol Med 2011;236:375–89. Mutational analysis of the tyrosine phosphatome in colorectal cancers. 18. Hoover AC, Spanos WC, Harris GF, Anderson ME, Klingelhutz AJ, Lee Science 2004;304:1164–6. JH. The role of human papillomavirus 16 E6 in anchorage-independent 5. Yeh SH, Wu DC, Tsai CY, Kuo TJ, Yu WC, Chang YS, et al. Genetic and invasive growth of mouse tonsil epithelium. Arch Otolaryngol Head characterization of fas-associated phosphatase-1 as a putative tumor Neck Surg 2007;133:495–502. suppressor on 4q21.3 in hepatocellular carcinoma. 19. Mendrola JM, Berger MB, King MC, Lemmon MA. The single trans- Clin Cancer Res 2006;12:1097–108. membrane domains of ErbB receptors self-associate in cell mem- 6. Hoover AC, Strand GL, Nowicki PN, Anderson ME, Vermeer PD, branes. J Biol Chem 2002;277:4704–12. Klingelhutz AJ, et al. Impaired PTPN13 phosphatase activity in spon- 20. Escher C, Cymer F, Schneider D. Two GxxxG-like motifs facilitate taneous or HPV-induced squamous cell carcinomas potentiates onco- promiscuous interactions of the human ErbB transmembrane gene signaling through the MAP kinase pathway. Oncogene 2009;28: domains. J Mol Biol 2009;389:10–6. 3960–70. 21. Gerber D, Sal-Man N, Shai Y. Two motifs within a transmembrane 7. Niu J, Huang YJ, Wang LE, Sturgis EM, Wei Q. Genetic polymorphisms domain, one for homodimerization and the other for heterodimeriza- in the PTPN13 gene and risk of squamous cell carcinoma of head and tion. J Biol Chem 2004;279:21177–82. neck. Carcinogenesis 2009;30:2053–8. 22. Palmer A, Zimmer M, Erdmann KS, Eulenburg V, Porthin A, Heumann 8. Glondu-Lassis M, Dromard M, Lacroix-Triki M, Nirde P, Puech C, Knani R, et al. EphrinB phosphorylation and reverse signaling: regulation by D, et al. PTPL1/PTPN13 regulates breast cancer cell aggressiveness Src kinases and PTP-BL phosphatase. Mol Cell 2002;9:725–37. through direct inactivation of Src kinase. Cancer Res 2010;70: 23. Revillion F, Puech C, Rabenoelina F, Chalbos D, Peyrat JP, Freiss G. 5116–26. Expression of the putative tumor suppressor gene PTPN13/PTPL1 is 9. Mita Y, Yasuda Y, Sakai A, Yamamoto H, Toyooka S, Gunduz M, et al. an independent prognostic marker for overall survival in breast cancer. Missense polymorphisms of PTPRJ and PTPN13 affect sus- Int J Cancer 2009;124:638–43. ceptibility to a variety of human cancers. J Cancer Res Clin Oncol 2010; 24. Williams R, Lee DW, Elzey BD, Anderson ME, Hostager BS, Lee JH. 136:249–59. Preclinical models of HPVþ and HPV HNSCC in mice: an immune 10. Pasquale EB. Eph receptors and in cancer: bidirectional clearance of HPVþ HNSCC. Head Neck 2009;31:911–8. signalling and beyond. Nat Rev Cancer 2010;10:165–80. 25. Spanos WC, Nowicki P, Lee DW, Hoover A, Hostager B, Gupta A, et al. 11. Erdmann KS. The protein tyrosine phosphatase PTP-basophil/baso- Immune response during therapy with cisplatin or radiation for human phil-like. Interacting proteins and molecular functions. Eur J Biochem papillomavirus-related head and neck cancer. Arch Otolaryngol Head 2003;270:4789–98. Neck Surg 2009;135:1137–46. 12. Lee HS, Mood K, Battu G, Ji YJ, Singh A, Daar IO. Fibroblast growth 26. Xu Z, Lai KO, Zhou HM, Lin SC, Ip NY. Ephrin-B1 reverse signaling factor receptor-induced phosphorylation of ephrinB1 modulates its activates JNK through a novel mechanism that is independent of interaction with Dishevelled. Mol Biol Cell 2009;20:124–33. tyrosine phosphorylation. J Biol Chem 2003;278:24767–75. 13. Tanaka M, Kamata R, Sakai R. Phosphorylation of ephrin-B1 via 27. Silva SD, Cunha IW, Younes RN, Soares FA, Kowalski LP, Graner E. the interaction with claudin following cell-cell contact formation. ErbB receptors and fatty acid synthase expression in aggressive head EMBO J 2005;24:3700–11. and neck squamous cell carcinomas. Oral Dis 2010;16:774–80.

OF10 Cancer Res; 73(18) September 15, 2013 Cancer Research

Downloaded from cancerres.aacrjournals.org on September 28, 2021. © 2013 American Association for Cancer Research. Published OnlineFirst June 28, 2013; DOI: 10.1158/0008-5472.CAN-13-0760

Cetuximab Drives ErbB/EphrinB1 Shifting and Signaling

28. Sheikh Ali MA, Gunduz M, Nagatsuka H, Gunduz E, Cengiz B, Fukush- 38. Vu T, Claret FX. Trastuzumab: updated mechanisms of action and ima K, et al. Expression and mutation analysis of epidermal growth resistance in breast cancer. Front Oncol 2012;2:62. factor receptor in head and neck squamous cell carcinoma. Cancer Sci 39. Gazdar AF. Activating and resistance mutations of EGFR in non–small- 2008;99:1589–94. cell lung cancer: role in clinical response to EGFR 29. Lee CS, Redshaw A, Boag G. receptor inhibitors. Oncogene 2009;28(Suppl 1):S24–31. immunoreactivity in human laryngeal squamous cell carcinoma. 40. Oxnard GR. Strategies for overcoming acquired resistance to epider- Pathology 1997;29:251–4. mal growth factor receptor: targeted therapies in lung cancer. Arch 30. Xia W, Lau YK, Zhang HZ, Liu AR, Li L, Kiyokawa N, et al. Strong Pathol Lab Med 2012;136:1205–9. correlation between c-erbB-2 overexpression and overall survival of 41. Hurvitz SA, Hu Y, O'Brien N, Finn RS. Current approaches and future patients with oral squamous cell carcinoma. Clin Cancer Res 1997;3: directions in the treatment of HER2-positive breast cancer. Cancer 3–9. Treat Rev 2013;39:219–29. 31. Cavalot A, Martone T, Roggero N, Brondino G, Pagano M, Cortesina G. 42. Rabinowits G, Haddad RI. Overcoming resistance to EGFR inhibitor in Prognostic impact of HER-2/neu expression on squamous head and head and neck cancer: a review of the literature. Oral Oncol 2012; neck carcinomas. Head Neck 2007;29:655–64. 48:1085–9. 32. Mandic R, Rodgarkia-Dara CJ, Zhu L, Folz BJ, Bette M, Weihe E, et al. 43. Pegram M. Can we circumvent resistance to ErbB2-targeted agents Treatment of HNSCC cell lines with the EGFR-specific inhibitor cetux- by targeting novel pathways? Clin Breast Cancer 2008;8(Suppl 3): imab (Erbitux) results in paradox phosphorylation of tyrosine 1173 in S121–30. the receptor. FEBS Lett 2006;580:4793–800. 44. Baselga J, Cortes J, Kim SB, Im SA, Hegg R, Im YH, et al. Pertuzumab 33. Li S, Schmitz KR, Jeffrey PD, Wiltzius JJ, Kussie P, Ferguson KM. plus trastuzumab plus docetaxel for metastatic breast cancer. N Engl Structural basis for inhibition of the epidermal growth factor receptor J Med 2012;366:109–19. by cetuximab. Cancer Cell 2005;7:301–11. 45. Keating GM. Pertuzumab: in the first-line treatment of HER2-positive 34. Citri A, Yarden Y. EGF-ERBB signalling: towards the systems level. Nat metastatic breast cancer. Drugs 2012;72:353–60. Rev Mol Cell Biol 2006;7:505–16. 46. Fuentes G, Scaltriti M, Baselga J, Verma CS. Synergy between tras- 35. Cho HS, Mason K, Ramyar KX, Stanley AM, Gabelli SB, Denney DW Jr, tuzumab and pertuzumab for human epidermal growth factor 2 (Her2) et al. Structure of the extracellular region of HER2 alone and in complex from colocalization: an in silico based mechanism. Breast Cancer Res with the Herceptin Fab. Nature 2003;421:756–60. 2011;13:R54. 36. Kondo N, Tsukuda M, Sakakibara A, Takahashi H, Hyakusoku H, 47. Prakash A, Janosi L, Doxastakis M. GxxxG motifs, phenylalanine, and Komatsu M, et al. Combined molecular targeted drug therapy for cholesterol guide the self-association of transmembrane domains of EGFR and HER-2 in head and neck squamous cell carcinoma cell ErbB2 receptors. Biophys J 2011;101:1949–58. lines. Int J Oncol 2012;40:1805–12. 48. Prakash A, Janosi L, Doxastakis M. Self-association of models of 37. Del Campo JM, Hitt R, Sebastian P, Carracedo C, Lokanatha D, transmembrane domains of ErbB receptors in a lipid bilayer. Biophys J Bourhis J, et al. Effects of lapatinib monotherapy: results of a rando- 2010;99:3657–65. mised phase II study in therapy-naive patients with locally advanced 49. Cymer F, Schneider D. Transmembrane helix-helix interactions squamous cell carcinoma of the head and neck. Br J Cancer 2011;105: involved in ErbB receptor signaling. Cell Adh Migr 2010;4:299– 618–27. 312.

www.aacrjournals.org Cancer Res; 73(18) September 15, 2013 OF11

Downloaded from cancerres.aacrjournals.org on September 28, 2021. © 2013 American Association for Cancer Research. Published OnlineFirst June 28, 2013; DOI: 10.1158/0008-5472.CAN-13-0760

Targeting ERBB Receptors Shifts Their Partners and Triggers Persistent ERK Signaling through a Novel ERBB/EFNB1 Complex

Paola D. Vermeer, Paul L. Colbert, Bryant G. Wieking, et al.

Cancer Res Published OnlineFirst June 28, 2013.

Updated version Access the most recent version of this article at: doi:10.1158/0008-5472.CAN-13-0760

Supplementary Access the most recent supplemental material at: Material http://cancerres.aacrjournals.org/content/suppl/2013/06/28/0008-5472.CAN-13-0760.DC1

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Subscriptions Department at [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://cancerres.aacrjournals.org/content/early/2013/09/06/0008-5472.CAN-13-0760. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from cancerres.aacrjournals.org on September 28, 2021. © 2013 American Association for Cancer Research.