13
Cancer Therapy: Preclinical Secretome Signature Identies ADAM17 as Novel Target for Radiosensitization of NonSmall Cell Lung Cancer Ashish Sharma, Sabine Bender, Martina Zimmermann, Oliver Riesterer, AngelaBroggini-Tenzer, and Martin N. Pruschy Abstract Purpose: Ionizing radiation (IR) induces intracellular signaling processes as part of a treatment-induced stress response. Here we investigate IR-induced ADAM17 activation and the role of ADAM17-shed factors for radiation resistance in nonsmall cell lung cancer. Experimental Design: Large-scale secretome proling was performed using antibody arrays. Secretion kinetics of ADAM17 substrates was determined using ELISA across multiple in vitro and in vivo models of nonsmall cell lung cancer. Clonogenic survival and tumor xenograft assays were performed to determine radio- sensitization by ADAM17 inhibition. Results: On the basis of a large-scale secretome screening, we investigated secretion of auto- or paracrine factors in nonsmall cell lung cancer in response to irradiation and discovered the ADAM17 network as a crucial mediator of resistance to IR. Irradiation induced a dose-dependent increase of furin-mediated cleavage of the ADAM17 proform to active ADAM17, which resulted in enhanced ADAM17 activity in vitro and in vivo. Genetic or pharmacologic targeting of ADAM17 suppressed IR-induced shedding of secreted factors, downregulated ErbB signaling in otherwise cetuximab-resistant target cells, and enhanced IR- induced cytotoxicity. The combined treatment modality of IR with the ADAM17 inhibitor TMI-005 resulted in a supra-additive antitumor response in vivo demonstrating the potential of ADAM17 targeting in combination with radiotherapy. Conclusions: Radiotherapy activates ADAM17 in nonsmall cell lung cancer, which results in shedding of multiple survival factors, growth factor pathway activation, and IR-induced treat- ment resistance. We provide a sound rationale for repositioning ADAM17 inhibitors as short-term adjuvants to improve the radiotherapy outcome of nonsmall cell lung cancer. Clin Cancer Res; 22(17); 442839. Ó2016 AACR. Introduction Radiotherapy along with chemotherapy or surgery is the primary treatment regimen for locally advanced nonsmall cell lung cancer (NSCLC). However, the response rates and clinical outcomes are still disappointing, with the 5-year survival rate being only approximately 15% (13). Ionizing radiation (IR) triggers multiple intracellular signaling processes as part of IR- induced stress responses that lead to the secretion of various para- and autocrine factors into the tumor microenvironment (46). These secreted components generate de novo resistance mechanisms during the course of the treatment and at the same time represent interesting targets to sensitize for IR-induced cell killing (79). Several studies have shown that targeting tumor- derived factors such as VEGF, PDGFA, TGFb, MMP-13, and SDF-1 counteract growth of carcinoma cells and sensitize them to irradiation (1014). Growth and survival of NSCLC cells are often dependent on ectodomain shedding which includes the proteolytic cleavage of the extracellular part of membrane proteins primarily mediated by membrane-anchored metalloproteases, and results in release of various soluble growth factors and cytokines regulating cell proliferation and migration (15, 16). ADAMs (a disintegrin and metalloproteinase) are membrane-associated metalloproteinases with modular design and complex multidomain structure (17). Most of the members of the ADAM family (13 of 21 human ADAMs) have proteolytic activity, as well as domains with adhe- sive properties and a cytoplasmic domain involved in cell signal- ing (18). They are actively associated with the process of proteo- lytic "shedding" of membrane-bound proteins and hence the rapid modulation of key signals in the tumor microenvironment. ADAM-mediated shedding is both constitutive and inducible, depending on upstream kinase activation, intracellular Ca 2þ levels, membrane lipid composition, and other experimental and natural stimuli (19, 20). Multiple resistance mechanisms in NSCLC have been linked to the EGFR, which has therefore been regarded as a promising target for a combined treatment modality. However, a recent phase III trial for patients with stage IIIA or IIIB NSCLC (RTOG 0617) demonstrated that sole addition of the EGFR-directed mAb cetux- imab to concurrent chemoradiation and consolidation treatment Laboratory for Applied Radiobiology, Department of Radiation Oncology, University Hospital Zurich, University of Zurich Zurich, Switzerland. Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/). A. Sharma and S. Bender contributed equally to this article. Corresponding Author: Martin N. Pruschy, Department of Radiation Oncology, University Hospital Zurich, Ramistr. 100, Zurich ZH-8091, Switzerland. Phone: 414-4255-8549; Fax: 414-4255-4435; E-mail: [email protected] doi: 10.1158/1078-0432.CCR-15-2449 Ó2016 American Association for Cancer Research. Clinical Cancer Research Clin Cancer Res; 22(17) September 1, 2016 4428 on March 4, 2021. © 2016 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from Published OnlineFirst April 13, 2016; DOI: 10.1158/1078-0432.CCR-15-2449

Secretome Signature Identifies ADAM17 as Novel Target for ... · Ashish Sharma, Sabine Bender, Martina Zimmermann, Oliver Riesterer, Angela Broggini-Tenzer, and Martin N. Pruschy

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Secretome Signature Identifies ADAM17 as Novel Target for ... · Ashish Sharma, Sabine Bender, Martina Zimmermann, Oliver Riesterer, Angela Broggini-Tenzer, and Martin N. Pruschy

Cancer Therapy: Preclinical

Secretome Signature Identifies ADAM17 asNovel Target for Radiosensitization ofNon–Small Cell Lung CancerAshish Sharma, Sabine Bender, Martina Zimmermann, Oliver Riesterer,Angela Broggini-Tenzer, and Martin N. Pruschy

Abstract

Purpose: Ionizing radiation (IR) induces intracellular signalingprocesses as part of a treatment-induced stress response. Here weinvestigate IR-induced ADAM17 activation and the role ofADAM17-shed factors for radiation resistance in non–small celllung cancer.

Experimental Design: Large-scale secretome profiling wasperformed using antibody arrays. Secretion kinetics of ADAM17substrateswas determined using ELISA acrossmultiple in vitro andin vivomodels of non–small cell lung cancer. Clonogenic survivaland tumor xenograft assays were performed to determine radio-sensitization by ADAM17 inhibition.

Results: On the basis of a large-scale secretome screening, weinvestigated secretion of auto- or paracrine factors in non–smallcell lung cancer in response to irradiation and discovered theADAM17 network as a crucial mediator of resistance to IR.Irradiation induced a dose-dependent increase of furin-mediated

cleavage of the ADAM17 proform to active ADAM17, whichresulted in enhanced ADAM17 activity in vitro and in vivo. Geneticor pharmacologic targeting of ADAM17 suppressed IR-inducedshedding of secreted factors, downregulated ErbB signaling inotherwise cetuximab-resistant target cells, and enhanced IR-induced cytotoxicity. The combined treatment modality of IRwith the ADAM17 inhibitor TMI-005 resulted in a supra-additiveantitumor response in vivo demonstrating the potential ofADAM17 targeting in combination with radiotherapy.

Conclusions: Radiotherapy activates ADAM17 in non–smallcell lung cancer, which results in shedding of multiple survivalfactors, growth factor pathway activation, and IR-induced treat-ment resistance. We provide a sound rationale for repositioningADAM17 inhibitors as short-term adjuvants to improve theradiotherapy outcome of non–small cell lung cancer. Clin CancerRes; 22(17); 4428–39. �2016 AACR.

IntroductionRadiotherapy along with chemotherapy or surgery is the

primary treatment regimen for locally advanced non–small celllung cancer (NSCLC). However, the response rates and clinicaloutcomes are still disappointing, with the 5-year survival ratebeing only approximately 15% (1–3). Ionizing radiation (IR)triggers multiple intracellular signaling processes as part of IR-induced stress responses that lead to the secretion of variouspara- and autocrine factors into the tumor microenvironment(4–6). These secreted components generate de novo resistancemechanisms during the course of the treatment and at the sametime represent interesting targets to sensitize for IR-induced cellkilling (7–9). Several studies have shown that targeting tumor-

derived factors such as VEGF, PDGFA, TGFb, MMP-13, andSDF-1 counteract growth of carcinoma cells and sensitize themto irradiation (10–14).

Growth and survival of NSCLC cells are often dependent onectodomain shedding which includes the proteolytic cleavage ofthe extracellular part of membrane proteins primarily mediatedby membrane-anchored metalloproteases, and results in releaseof various soluble growth factors and cytokines regulating cellproliferation and migration (15, 16). ADAMs (a disintegrin andmetalloproteinase) are membrane-associated metalloproteinaseswith modular design and complex multidomain structure (17).Most of the members of the ADAM family (13 of 21 humanADAMs) have proteolytic activity, as well as domains with adhe-sive properties and a cytoplasmic domain involved in cell signal-ing (18). They are actively associated with the process of proteo-lytic "shedding" of membrane-bound proteins and hence therapid modulation of key signals in the tumor microenvironment.ADAM-mediated shedding is both constitutive and inducible,depending on upstream kinase activation, intracellular Ca2þ

levels, membrane lipid composition, and other experimental andnatural stimuli (19, 20).

Multiple resistance mechanisms in NSCLC have been linked tothe EGFR,which has therefore been regarded as a promising targetfor a combined treatment modality. However, a recent phase IIItrial for patients with stage IIIA or IIIB NSCLC (RTOG 0617)demonstrated that sole addition of the EGFR-directedmAb cetux-imab to concurrent chemoradiation and consolidation treatment

Laboratory for Applied Radiobiology, Department of RadiationOncology, University Hospital Zurich, University of Zurich Zurich,Switzerland.

Note: Supplementary data for this article are available at Clinical CancerResearch Online (http://clincancerres.aacrjournals.org/).

A. Sharma and S. Bender contributed equally to this article.

Corresponding Author: Martin N. Pruschy, Department of Radiation Oncology,University Hospital Zurich, Ramistr. 100, Zurich ZH-8091, Switzerland. Phone:414-4255-8549; Fax: 414-4255-4435; E-mail: [email protected]

doi: 10.1158/1078-0432.CCR-15-2449

�2016 American Association for Cancer Research.

ClinicalCancerResearch

Clin Cancer Res; 22(17) September 1, 20164428

on March 4, 2021. © 2016 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst April 13, 2016; DOI: 10.1158/1078-0432.CCR-15-2449

Page 2: Secretome Signature Identifies ADAM17 as Novel Target for ... · Ashish Sharma, Sabine Bender, Martina Zimmermann, Oliver Riesterer, Angela Broggini-Tenzer, and Martin N. Pruschy

does not provide any added benefit for these patients (21). Thus,additional inhibition of the EGFR is insufficient, which might bedue to the heterogenous expression status of other ErbB receptorfamily members in NSCLC or the mutational status of EGFRdownstream signaling entities such as K-ras (22–24).

Increased ADAM17 expression in NSCLC is associated withaggressive progression and poor prognosis. ADAM17 regulatessheddingofmultiple keyoncogenic growth factors, cytokines, andadhesion molecules, including ligands of ErbB family members(15, 16, 19, 25–28). Thus, ADAM17 drives pleiotropic pathwaysand might therefore represent a more relevant target for a com-bined treatment modality in NSCLC.

Here we provide novel insights into activation of ADAM17and subsequent ligand shedding in response to irradiation. Weshow for the first time that direct targeting of ADAM17 with aclinically relevant inhibitor sensitizes lung carcinoma cells invitro and in vivo to IR and provide a sound rationale forpositioning ADAM17 inhibitors as radiosensitizers to improvethe treatment of NSCLC.

Materials and MethodsCell culture, compounds, and irradiation

The human NSCLC cell lines A549, NCI-H125, H460, Calu-3,Calu-6, and the human epidermoid carcinoma cell line A431werecultured in RPMI1640 media supplemented with 10% (v/v) FCS,1% (v/v) penicillin–streptomycin, and 1% (v/v) L-glutamine at37�C in 5% CO2. All cell culture media and supplements wereobtained from Gibco (Life Technologies). All cell lines wereauthenticated through STR sequencing done by Microsynth AG.TMI-005 was obtained from Axon Medchem (Axon 1507). Forin vitro experiments, TMI-005 was dissolved in DMSO (10mmol/L stock) and further diluted with cell culture media. N-acetyl-L-cysteine (Sigma Aldrich; A8199) was dissolved in PBS (10mmol/L stock). The furin convertase inhibitor Dec-RVKR-CMK (EnzoLife Sciences; ALX-260-022) was dissolved in PBS (5 mmol/Lstock). Cetuximab (Erbitux) was obtained from Merck KGaA (5mg/mL stock solution for infusion) andwas further diluted to 100nmol/L for in vitro experiments. Irradiation was performed using

an Xstrahl 200 kV X-Ray unit at 100 cGy/minute. To produceconditioned media, FCS-supplemented or FCS-deficient cell cul-ture media was applied to proliferating cells 1 hour prior to shamtreatment or irradiation, collected 24 hours thereafter and filteredthrough a 0.45-mm filter.

Cell proliferation and clonogenic cell survival assaysThe proliferative activity of tumor cells was assessed in 96-

well plates with the colorimetric AlamarBlue assay (BiosourceInternational). Clonogenic cell survival was determined by theability of single cells to form colonies in vitro as described inref. 29. Dose enhancement factors (DEF) were calculated usinga and b values for each survival curve. Survival data were fittedby weighted, stratified, linear regression to obtain the linear andquadratic parameters (29). The DEF values at 37% and 10%survival level were calculated using linear regression in IBMSPSS Statistics 22. All assays were repeated as independentexperiments at least thrice.

Tumor xenograft in nude mice and application of treatmentregimes

A549 (4 � 106 cells) were injected subcutaneously in the backof 4- to 6-week old CD1 athymic nudemice. Tumor volumesweredetermined from caliper measurements of tumor length (L) andwidth (l) according to the formula (L � l2)/2. Tumors wereallowed to expand to a volume of 200 mm3 (�10%) beforetreatment start. Tumors were sham irradiated or irradiated using acustomized shielding device with 3� 1Gy for 3 consecutive days.TMI-005 (dissolved in 0.9%NaCl, 0.5%methylcellulose, and 2%Tween 80) was administered orally at 25 mg/kg twice daily with10 hours apart for seven consecutive days. For combined treat-ment, (sham)-irradiation was performed 1 hour after the firstdaily TMI-005 application on day 3 to day 5 of the TMI-005schedule.

In vivo shedding experiments were performed on blood serumderived from locally irradiated A549-derived tumor xenografts. Ata tumor volume of 400 to 500mm3, tumors were sham irradiatedor irradiated with 10 Gy. IR-treated animals were sacrificed 48hours after treatment. Tumors were excised and blood serum wascollected from heart-punctured animals after euthanasia. All invivo experiments were performed according to the Guidelines fortheWelfare andUseofAnimals of theVeterin€aramtKantonZ€urich(Zurich, Switzerland).

IHCImmunohistologic endpoints were analyzed on paraffin-

embedded blocks for MIB1/Ki-67 (prediluted; Ventana-Roche,790-4286), ADAM17 (1:200; Novus Biologicals, NBP2-15281),Amphiregulin (1:250; Atlas Antibodies HPA008720), ALCAM(1:150; Novocastra Lab, NCL-CD166), CD31 (1:10; Dako,M0823) using a Discovery Immunohistochemistry Staining Sys-tem (Ventana Medical Systems).

MIB1/Ki-67–positive tumor cells and amount of vessels(CD31) were counted in at least 4 randomly chosen visual fields(magnification, 400�) in each xenograft (n ¼ 4 for each group).Viable areas of the whole tumor sections were quantified forspecific ADAM17, ALCAM, or Amphiregulin staining intensity.Each treatment group consisted of at least 4 animals and tumorsections were manually quantified for low, medium, and highstaining.

Translational Relevance

Intrinsic and acquired resistances to ionizing radiation (IR)represent a major challenge in the treatment of NSCLC. Arecent phase III trial for patients with stage IIIA or IIIB NSCLC(RTOG 0617) demonstrated that addition of the EGFR-direct-ed mAb cetuximab to concurrent chemoradiation and con-solidation treatment did not provide any added benefit. Thus,novel molecular targets for anticancer agents alone and incombination with radiotherapy are of high demand. Thesheddase ADAM17 is associated with aggressive progressionand poor prognosis in NSCLC and activates multiple ErbB-and non-ErbB–related pathways. Here we demonstrate thatgenetic and pharmacologic inhibition of ADAM17 sensitizesin vitro and in vivo NSCLC to IR. The findings of this studysuggest that clinically relevant ADAM17 inhibitors should beconsidered for the treatment of NSCLC in combination withradiotherapy.

ADAM17 and Ionizing Radiation in NSCLC

www.aacrjournals.org Clin Cancer Res; 22(17) September 1, 2016 4429

on March 4, 2021. © 2016 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst April 13, 2016; DOI: 10.1158/1078-0432.CCR-15-2449

Page 3: Secretome Signature Identifies ADAM17 as Novel Target for ... · Ashish Sharma, Sabine Bender, Martina Zimmermann, Oliver Riesterer, Angela Broggini-Tenzer, and Martin N. Pruschy

Statistical analysisStatistical analyses were performed using GraphPad Prism

(version 5). If not indicated otherwise, in vitro data wereanalyzed using the unpaired Student t test and data representat least three independently performed assays, with error barsindicating SD. In vivo treatment response was evaluated by twodifferent statistical metrics. Area under the tumor volume curve(AUC) was analyzed by one-way ANOVA using the Tukey testfor pair-wise comparisons. In addition, tumor growth delayswere also calculated by Kaplan–Meier analysis with log-rank(Mantel–Cox) test for pair-wise comparisons. Immunohisto-chemical data were analyzed by one-way ANOVA on replicatetumor samples (n ¼ 4) and pair-wise analysis was performedusing the Tukey test. For all experiments, �, P < 0.05; ��, P < 0.01;���, P < 0.001.

ResultsIR induces the secretion of multiple ADAM17 substrates

A semiquantitative differential large-scale secretome analysis(>300 factors) was performed to identify auto- and paracrinefactors that are secreted in response to IR. Using several growthfactor-, chemo-, and cytokine-specific antibody arrays, multiple,differentially secreted factors were identified in conditionedmedia (CM) derived from sham irradiated and irradiated A549lung adenocarcinoma cells, respectively (Fig. 1A; SupplementaryMethods and Supplementary Table S1A). Detailed analysis of hitspointed toward a limited number of upstream effectors regulatingIR-induced secretion of biologically active factors. Interestingly,several substrates of the metalloprotease ADAM17 were upregu-lated (see Supplementary Table S1; Fig. 1A), in particular Amphir-egulin and ALCAM. Of note, lactate dehydrogenase (LDH)

Figure 1.IR induces the secretion of multiple ADAM substrates in vitro and in vivo. A, screening was performed with multiple antibody arrays (>300 factors) against CMderived from irradiated and sham-irradiated A549 cells 24 hours after irradiation (5 Gy). Scatter plots for angiogenic and secretory analytes are depicted.B, ELISA-quantified levels of ALCAM and Amphiregulin in supernatants derived from a panel of NSCLC cells and A431 cells (24 hours after sham irradiation orirradiation), unpaired Student t test. C, qRT-PCR–determined gene expression of ALCAM and Amphiregulin 24 hours after irradiation in A549 cells, one-wayANOVA test. D–F, A549-derived tumor xenografts were sham-irradiated or irradiated with 10 Gy. Tumors and blood sera were harvested 48 hours after irradiation.D, ALCAM concentration in murine blood serum derived from sham-irradiated or locally irradiated A549 tumor xenografts and quantified by ELISA; n � 4,unpaired Student t test. E, Quantification of immunohistochemical staining intensities for ALCAM and Amphiregulin in tumor sections derived from A549-derivedtumor xenografts (n ¼ 5). Whole tumor sections were quantified for specific ALCAM or Amphiregulin staining intensity. F, representative images ofimmunohistochemical staining of ALCAM and Amphiregulin in response to treatment. Scale bar, 100 mm. Data are represented as mean � SD. �, P < 0.05;�� , P < 0.01; ��� , P < 0.001.

Sharma et al.

Clin Cancer Res; 22(17) September 1, 2016 Clinical Cancer Research4430

on March 4, 2021. © 2016 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst April 13, 2016; DOI: 10.1158/1078-0432.CCR-15-2449

Page 4: Secretome Signature Identifies ADAM17 as Novel Target for ... · Ashish Sharma, Sabine Bender, Martina Zimmermann, Oliver Riesterer, Angela Broggini-Tenzer, and Martin N. Pruschy

activity, a control marker for unspecific cytolysis, was notincreased in response to irradiation (Supplementary Fig. S1A).Thus, no general protein leakage could be observed, indicatingspecific, IR-mediated secretion of these ADAM17 substrates. Onthe basis of the increased abundance of ADAM17 substrates, weinvestigated regulation of ADAM17 in response to IR in detail.

To confirm IR-enhanced ADAM17 substrate secretion in addi-tional cell lines (A549, H460, NCI-H125, CALU-3, CALU-6, allNSCLC and A431, vulval), ALCAM- and Amphiregulin-directedELISA were used to determine these ADAM17 substrates in aquantitative way in supernatants of irradiated tumor cells. Sig-nificant upregulation of both ADAM17 substrates could bedetected in the NSCLC cell lines A549 and NCI-H125, and todifferent magnitudes in the other cell lines (Fig. 1B).

Enhanced secretion of these ADAM17 substrates could bedue to IR-induced expression. However, only a marginalincrease of Amphiregulin gene expression in A549 cells couldbe detected in response to IR, while no IR-induced ALCAM geneexpression could be observed (Fig. 1C). Similar expressionprofiles could be determined in the additional lung carcinomacell line NCI-H125 and at an early time point (4 hours) afterirradiation (Supplementary Fig. S1B and S1C). Furthermore,protein levels of intracellular Amphiregulin and ALCAM inresponse to IR as determined in cell lysates did not change(Supplementary Fig. S1D).

To confirm these findings in vivo, we analyzed ALCAM secretionfrom irradiated A549-derived tumor xenografts in mice. Tumorsand blood sera were harvested 48 hours after irradiation with10 Gy. Ex vivo analysis of blood serum from mice with locallyirradiated A549-derived tumor xenografts revealed significantlyincreased levels of ALCAM in comparison with ALCAM levels inblood serum from sham-irradiatedmice (Fig. 1D).Unfortunately,Amphiregulin concentrations were below a minimal technicallydetectable concentration. In addition, we also observed increasedstaining of both ALCAM and Amphiregulin in tumor sectionsderived from locally irradiated xenografts in comparison withsham-irradiated tumors (Fig. 1E and F). These results demonstratethat IR induces the secretion of multiple ADAM17 substratesacross a panel of NSCLC cell lines and from A549-derived tumorxenografts.

IR induces posttranslational modification and activation ofADAM17

To rule out the involvement of closely related ADAM10 in theregulatory process (30, 31), both ADAM17 and ADAM10 activ-ities were determined in response to irradiation. Interestingly,irradiation only increased ADAM17 but not ADAM10 activity in adose-dependent way (Fig. 2A). ADAM17 activity steadilyincreased over 30 hours in response to irradiation (Fig. 2B). Asa control experiment, ADAM17 activity was determined in irra-diated cells pretreated with the reactive oxygen scavenger NAC.NAC completely attenuated IR-induced ADAM17 activity in bothA549 and NCI-H125 cells while having no effect on its basalactivity (Supplementary Fig. S2A). Significant induction ofADAM17 activity in response to irradiation was also observed inseveral other NSCLC cell lines. However, not all cell linesresponded to IR with increased ADAM17 activity (Fig. 2C). Ofnote, gene expression of ADAM17 or ADAM10 did not increase inresponse to IR in A549 and NCI-H125 lung carcinoma cells at the24-hour and at an early 4-hour timepoint, respectively (Fig. 2Dand Supplementary Fig. S2B and S2C).

To acquire full activity, ADAM17 requires its conversion to themature form lacking its inhibitory prodomain (18, 32). Reducedamounts of zymogen and concomitantly increased levels of themature form of ADAM17 could be detected in response toincreasing doses of IR (Fig. 2E). Likewise, densitometric analysisof the ADAM17 zymogen and active form of ADAM17 revealedthat the overall levels of ADAM17 did not change in response toincreasing doses of irradiation (Supplementary Fig. S2D). Toprobe for IR-induced ADAM17 expression and activity in vivo,A549-derived tumor xenografts were irradiated with a single doseof 10 Gy and tumors were harvested 48 hours after irradiation.Immunohistochemical staining of tumor sections did not revealan IR-induced change of the ADAM17 expression pattern. How-ever, a significant increase of ADAM17 activity was detected intissue homogenates derived from irradiated tumors in compar-ison with ADAM17 activity in sham-irradiated tumors (Fig. 2Fand 2G). These results indicate that posttranslational modifica-tion and not increased levels of ADAM17 gene expressionaccounts for IR-induced ADAM17 activity.

Targeting of ADAM17 activity sensitizes NSCLC cells to IRDeregulated shedding of ADAM17 substrates might affect the

response of NSCLC cells to IR. We therefore targeted ADAM17with the ADAM17 inhibitor TMI-005 (Apratastat, 25mmol/L) andtwo different ADAM17-directed siRNAs (si-ADAM17#1, 2). Cel-lular pretreatment with TMI-005 and si-ADAM17#2 stronglydownregulated ADAM17 activity, and both targeting approachessignificantly decreased the proliferative activity of A549 cells(Fig. 3A). si-ADAM17#1 was overall less potent than si-ADAM17#2 to downregulate ADAM17 expression, its shedding,andproliferative activity inA549 cells (Fig. 3AandSupplementaryFig. S3A–S3C). To quantify the effect of reduced ADAM17 activityon cellular radiosensitivity, we determined clonogenicity ofNSCLC cells in siRNA- and TMI-005–pretreated A549 and NCI-H125 cells in response to irradiation. Cellular pretreatment withTMI-005 and si-ADAM17#2, respectively, sensitized both A549andNCI-H125 cells to IR (Fig. 3B andC). TheDEFs for TMI-005 at37% cell survival were 1.40� 0.24 and 1.45� 0.26 for A549 andNCI-H125 cells, respectively.

Several ligands of the EGFR are shed by ADAM17, which resultsin EGF-receptor tyrosine kinase (RTK) activation (33, 34). Wetherefore determined the potency of direct EGFR-targeting withthe mAb cetuximab in comparison with ADAM17-targeting as aradiosensitizing strategy. Cetuximab strongly abrogated short-term EGF-mediated activation of the EGF-RTK demonstrating itsEGF-RTK–directed inhibitory activity (Supplementary Fig. S3D).Interestingly, cetuximab did not reduce short-term proliferativeactivity when used as single treatment modality (Fig. 3A) and didnot affect clonogenicity on combined treatment with increasingdoses of IR in A549 cells (Fig. 3D). Complementary clonogenicsurvival experiments were performed with recombinant Amphir-egulin. While recombinant Amphiregulin could activate the EGF-RTK, it did not rescue clonogenic survival of both ADAM17 wild-type and ADAM17 knockdown A549 cells in response to irradi-ation (Supplementary Fig. S3E and S3F).

The proprotein convertase furin mediates IR-induced ADAM17activation

The proprotein convertase furin proteolytically activatesADAM17 and ADAM10 (35, 36). IR-induced ADAM17 conver-sion and activity was determined in A549 cells pretreated with the

ADAM17 and Ionizing Radiation in NSCLC

www.aacrjournals.org Clin Cancer Res; 22(17) September 1, 2016 4431

on March 4, 2021. © 2016 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst April 13, 2016; DOI: 10.1158/1078-0432.CCR-15-2449

Page 5: Secretome Signature Identifies ADAM17 as Novel Target for ... · Ashish Sharma, Sabine Bender, Martina Zimmermann, Oliver Riesterer, Angela Broggini-Tenzer, and Martin N. Pruschy

irreversible furin inhibitor decanoyl-RVKR-chloromethylketone(Dec-RVKR-CMK). Dec-RVKR-CMK counteracted IR-inducedconversion of pro-ADAM17 to active ADAM17 and kept ADAM17activity at basal level in irradiated cells (Fig. 4A and B). The furininhibitor abrogated IR-enhanced ADAM17 activity also in NCI-H125 cells (Supplementary Fig. S4A). Furthermore, Dec-RVKR-CMK sensitized A549 cells to IR and reduced clonogenicity in asupra-additive way (Fig. 4C). To detect a putative upstream IR-regulatedmechanism, furin gene expression in response to IRwasdetermined by qRT-PCR. Interestingly, increased IR dose–depen-dent furin gene expression was observed in A549 cells already 4hours after irradiation (Fig. 4D).

To exclude the possibility that furin inhibitor–based radio-sensitization is due to reduced ADAM10 conversion, radiosensi-tization of A549 cells was also determined in cells pretreated withtwo different ADAM10-directed siRNAs. Knockdown of ADAM10or pretreatment with the specific ADAM10 inhibitor GI254023did only minimally reduce the proliferative activity of A549 cellsand did not alter clonogenic survival in response to IR (Supple-

mentary Figs. S4B–S4D). These results corroborate that IR spe-cifically activates ADAM17, most probably due to IR-enhancedfurin expression, and that targeting of ADAM17 enhances IR-induced cytotoxicity.

ADAM17 mediates prosurvival ligand shedding anddownstream signaling in response to IR

To determine downstream effects of ADAM17 activity,ADAM17-mediated ligand shedding was analyzed in irradiatedcells pretreated with either the ADAM17 inhibitor TMI-005 or theADAM17-directed siRNA. Treatment of A549 andNCI-H125 cellswith TMI-005 significantly reduced the basal and IR-inducedconcentrations of the secreted ADAM17-specific substrates,ALCAM and Amphiregulin. Likewise, IR-induced ALCAM andAmphiregulin levels were downregulated in supernatants derivedfromA549 cells, pretreatedwith theADAM17-directedADAM17#2siRNA (Fig. 5A and B).

The EGFR-directedmAb cetuximab did not sensitize A549 cellsfor IR. Nevertheless, we used the EGFR phosphorylation status at

Figure 2.IR induces posttranslational modification and activation of ADAM17 in vitro and in vivo. A, ADAM10 and ADAM17 activities in A549 cells determined 24 hoursafter irradiation with increasing doses of IR. B, IR-induced ADAM17 activity in A549 cells determined at different time points after irradiation, unpairedStudent t test. C, IR-induced ADAM17 activity in a panel of NSCLC cell lines and A431 cells, 24 hours after irradiation, unpaired Student t test. D, qRT-PCR–based quantification of ADAM10 and ADAM17 gene expression 24 hours after irradiation in A549 cells, one-way ANOVA. E, Western blots of ADAM17 zymogenand active form in A549-derived cell lysates 24 hours after irradiation with increasing doses of IR. F, ADAM17 activity quantified in tumor lysates derivedfrom A549-tumor xenografts in response to sham irradiation or irradiation with 10 Gy. Tumors were harvested 48 hours after irradiation; n � 4, unpaired Student ttest. G, representative images of ADAM17-immunohistochemical staining in response to sham-irradiation or irradiation with 10 Gy. Scale bar, 100 mm.Data are represented as mean � SD. � , P < 0.05; �� , P < 0.01.

Sharma et al.

Clin Cancer Res; 22(17) September 1, 2016 Clinical Cancer Research4432

on March 4, 2021. © 2016 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst April 13, 2016; DOI: 10.1158/1078-0432.CCR-15-2449

Page 6: Secretome Signature Identifies ADAM17 as Novel Target for ... · Ashish Sharma, Sabine Bender, Martina Zimmermann, Oliver Riesterer, Angela Broggini-Tenzer, and Martin N. Pruschy

tyrosine 1173 (Y1173) as surrogate marker to probe for autocrinereceptor activation in response to irradiation and ADAM17 inhi-bition. Enhanced EGFR phosphorylation was observed in cells 24hours after irradiation. On the other hand, the EGFR phosphor-ylation status was strongly reduced at the 24-hour timepoint insham-irradiated and irradiated cells pretreated with TMI-005 butonly minimally in cells pretreated with cetuximab (Fig. 5C).Enhanced EGFR phosphorylation over time was observed incontrol but not in ADAM17-directed siRNA knockdown cellscomparing the EGFR phosphorylation status 1, 4, and 24 hoursafter irradiation (Fig. 5D). These results correlate with IR-enhanced conditioning of the media and accumulation of auto-crine ligands. To corroborate that ADAM17 regulates an autocrineloop, na€�ve A549 cells were stimulated with conditioned media(CM) derived from irradiated and sham-irradiated, control, andADAM17-directed siRNA knockdown cells, respectively (Fig. 5E).A partially enhanced EGFR phosphorylation status was observedin cells that were short-time incubated with CM derived fromirradiated cells in comparison with cells that were short-time

incubated with CM derived from sham-irradiated cells. Stimula-tion of na€�ve A549 cells with CM derived from sham-irradiatedand irradiated, but ADAM17 knockdown, cells resulted in lowestEGFR activation levels. Collectively, these results indicate thatADAM17 strongly regulates an autocrine growth factor loop andsuggest that sustained and IR-activated ADAM17 substrate shed-ding might contribute to radiation resistance.

We next assessed the relevance of ADAM17 inhibition forradiation-induced cell killing and investigated different modesof cell death augmented by targeting ADAM17 in combinationwith IR. Microscopic evaluation of the cellular morphology ofA549 cells in response to treatment with IR and ADAM17inhibition revealed a flat and enlarged phenotype indicativefor treatment-induced senescence (data not shown). A dose-dependent increase of senescence-associated b-galactosidasewas determined in irradiated A549 cells. The amount ofb-galactosidase–positive cells was only minimally increased inADAM17 knockdown cells and in cells pretreated with TMI-005. Interestingly though, irradiation of TMI-005–pretreated or

Figure 3.Targeting of ADAM17 activity reduces the proliferative activity of NSCLC cells and sensitizes for IR. A, short-term proliferative activity of A549 cells pretreatedwith the ADAM17 inhibitor TMI-005, cetuximab, or transfected with ADAM17- or luciferase-directed siRNA. A549 (B) and NCI-H125 (C) cells were pretreatedwith TMI-005 (25 mmol/L) for 1 hour or transfected with ADAM17- or luciferase-directed siRNA (A549 cells, middle), and respective clonogenic cell survivalwas determined 7–10 days after treatment with increasing doses of IR. D, clonogenic cell survival of A549 cells pretreated with cetuximab (100 nmol/L) for 1 hourand irradiated with increasing doses of IR. Data are represented as mean � SD. � , P < 0.05; �� , P < 0.01; ��� , P < 0.001. Significance was measured by theunpaired Student t test.

ADAM17 and Ionizing Radiation in NSCLC

www.aacrjournals.org Clin Cancer Res; 22(17) September 1, 2016 4433

on March 4, 2021. © 2016 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst April 13, 2016; DOI: 10.1158/1078-0432.CCR-15-2449

Page 7: Secretome Signature Identifies ADAM17 as Novel Target for ... · Ashish Sharma, Sabine Bender, Martina Zimmermann, Oliver Riesterer, Angela Broggini-Tenzer, and Martin N. Pruschy

ADAM17 knockdown cells strongly increased the amount ofb-galactosidase–expressing cells (Fig. 5F and SupplementaryFig. S5A), indicating that ADAM17 increases the threshold forIR-induced senescence. The same senescent phenotype wasobserved in ADAM17-inhibited, irradiated NCI-H125 cells(Supplementary Fig. S5B). Inhibition of ADAM17 by eithertargeting approach did not enhance the overall minimalamount of IR-induced apoptosis in these NSCLC cells (Sup-plementary Fig. S5C). In contrast to ADAM17 targeting, cetux-imab did not further promote the IR-induced senescence phe-notype, which correlates with lack of radiosensitization bycetuximab at the level of the quantitative clonogenic cellsurvival assay (Fig. 5G, see also Fig. 3D).

Effect of TMI-005 and IR alone and in combination on thegrowth of A549-derived tumor xenografts

To determine the efficacy of ADAM17 inhibition in combina-tion with IR in vivo, immunocompromised mice with A549-derived tumor xenograftswere treatedwith theADAM17 inhibitorTMI-005 (administered orally at 25 mg/kg twice daily for sevenconsecutive days, day 1–7) and locally irradiated during 3 con-secutive days (1Gy, days 3–5; Fig. 6A). Treatmentwith TMI-005orIR alone resulted in a minimal treatment response, whereascombined treatment exerted a strong supra-additive tumorgrowth delay. The combined treatment regimen of TMI-005 andIR resulted in a statistically significant reduction of the area underthe tumor volume curve (AUC) in these NSCLC tumors (AUC

Figure 4.IR-induced ADAM17 activity is abrogated by the furin inhibitor Dec-RVKR-CMK. A–C, A549 cells were preincubated for 1 hour with the furin inhibitor Dec-RVKR-CMK (50 mmol/L) followed by irradiation with the indicated doses of IR. A, ADAM17 protein levels were investigated by Western blotting, 24 hours afterirradiation. Quantification of band intensities from 3 independent experiments is depicted in the right panel.B, cell lysateswere probed for ADAM17 activity, 24 hoursafter irradiation. C, clonogenic cell survival of A549 cells was determined 7–10 days after treatment according to A. Data are represented as mean� SD. � , P < 0.05;�� , P < 0.01; ��� , P < 0.001. Significance was measured by the unpaired Student t test. D, qRT-PCR–based quantification of furin gene expression 4 hoursafter irradiation in A549 cells; one-way ANOVA.

Sharma et al.

Clin Cancer Res; 22(17) September 1, 2016 Clinical Cancer Research4434

on March 4, 2021. © 2016 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst April 13, 2016; DOI: 10.1158/1078-0432.CCR-15-2449

Page 8: Secretome Signature Identifies ADAM17 as Novel Target for ... · Ashish Sharma, Sabine Bender, Martina Zimmermann, Oliver Riesterer, Angela Broggini-Tenzer, and Martin N. Pruschy

Figure 5.ADAM17 mediates prosurvival ligand shedding and downstream signaling in response to IR (A–D) A549 or NCI-H125 cells were pretreated with TMI-005(25 mmol/L) for 1 hour or A549 cells were transfected with ADAM17- or luciferase-directed siRNA (middle), and irradiated with the indicated doses of IR. A and B,secretion of ALCAM (A) and Amphiregulin (B) into the cell culture media was measured by ELISA, 24 hours after irradiation. C, EGFR phosphorylationstatus in control, TMI-005, or cetuximab-pretreated A549 cells. Levels of phospho-EGFR (Y1173), EGFR, ADAM17, and b-actin were analyzed in whole cell lysates24 hours after irradiation by Western blotting. (Continued on the following page.)

ADAM17 and Ionizing Radiation in NSCLC

www.aacrjournals.org Clin Cancer Res; 22(17) September 1, 2016 4435

on March 4, 2021. © 2016 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst April 13, 2016; DOI: 10.1158/1078-0432.CCR-15-2449

Page 9: Secretome Signature Identifies ADAM17 as Novel Target for ... · Ashish Sharma, Sabine Bender, Martina Zimmermann, Oliver Riesterer, Angela Broggini-Tenzer, and Martin N. Pruschy

control 26,596 � 3,986 vs. TMI-005 plus IR 13,091 � 3,196; P <0.001; Fig. 6B). Furthermore, the absolute growth delay to triplethe initial tumor volume (200 mm3) was most enhanced inresponse to the combined treatment modality when comparedwith the absolute tumor growth delay in response to TMI-005 orIR alone [13.0 � 2.1 days (TMI-005 plus IR) vs. 3.76 � 2.18 days(TMI-005) and3.09�2.17days (IR;P<0.001 for TMI-005plus IRversus control and eachmonotherapy)], respectively, suggesting apositive interaction between TMI-005 and IR (Fig. 6C). Tumor

tissue staining intensity of both ALCAM and Amphiregulin wassignificantly reduced on TMI-005 treatment, demonstrating thepotency of the ADAM17 inhibitor (Fig. 6D and F). Irradiationalone only minimally reduced tumor proliferation, which mightbe due to the minimal treatment regimen and correlated with thesmall tumor growth delay induced by IR alone. In comparisonwith control tumors, the number of MIB-1–positive cells wassignificantly reduced to 55%and57% in tumors treatedwith TMI-005 alone and in combination with IR, respectively (control vs.

(Continued.) D, EGFR-phosphorylation status (Y1173) in cell lysates derived from si-luciferase- or si-ADAM17–transfected A549 cells 1, 4, and 24 hours afterirradiation and analyzed by Western blotting. E, EGFR-phosphorylation status in na€�ve A549 cells stimulated with CM collected from sham-irradiated or irradiated(10 Gy), si-luciferase or si-ADAM17#2 transfected A549 cells 24 hours after IR. Na€�ve A549 cells were incubated with CM for 1 hour, followed by whole celllysate analysis and Western blotting with the respective antibodies. F and G, quantification of senescence-associated b-galactosidase (b-gal) staining of sham-irradiated and irradiated A549 cells pretreated with TMI-005 (25 mmol/L; F), cetuximab (100 nmol/L; G), or transfected with ADAM17- or luciferase-directedsiRNA, 3 days after irradiation (H). Data are represented as mean � SD. � , P < 0.05; �� , P < 0.01; ��� , P < 0.001. Significance was measured by the unpairedStudent t test.

Figure 6.Effect of TMI-005 and IR alone and in combination on the growth of A549-derived tumor xenografts. Schematic illustration of the in vivo experimentalsetup and treatment regimen (A). B, tumor growth delay of A549-derived tumor xenografts in response to combined treatment with TMI-005 (25 mg/kg, twicedaily for 7 days) and IR (3 � 1 Gy); n � 6, data is represented as mean � SEM, one-way ANOVA Tukey test. C, Kaplan–Meier survival analysis for A549 tumorsreaching 600 mm3 tumor volume after treatment with TMI-005 or IR or their combination; n � 6, log-rank (Mantel–Cox) test for pair-wise comparisons. D–E,immunohistochemical staining intensities for ALCAM, Amphiregulin, and Ki-67 on tumor tissue from A549-derived tumor xenografts treated with TMI-005and IR alone or in combination, data is represented as mean � SD; one-way ANOVA on replicate tumor samples (n ¼ 4); pair-wise analysis using the Tukey test.F, representative images of immunohistochemical staining of ADAM17, ALCAM, Amphiregulin, and Ki-67 in response to treatments. Scale bar, 100 mm. ��, P < 0.01;��� , P < 0.001.

Sharma et al.

Clin Cancer Res; 22(17) September 1, 2016 Clinical Cancer Research4436

on March 4, 2021. © 2016 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst April 13, 2016; DOI: 10.1158/1078-0432.CCR-15-2449

Page 10: Secretome Signature Identifies ADAM17 as Novel Target for ... · Ashish Sharma, Sabine Bender, Martina Zimmermann, Oliver Riesterer, Angela Broggini-Tenzer, and Martin N. Pruschy

TMI-005 plus IR; P < 0.001; Fig. 6E and F). Furthermore, nochanges could be observed at the level of the tumor vasculature inresponse to treatment (Supplementary Fig. S6A).

Analysis of tissuemorphology and integrity of coirradiated skinadjacent to the tumor did not reveal any normal tissue toxicities inresponse to the different treatmentmodalities, and determinationof bodyweight did not show anyweight loss with TMI-005 duringcourse of the treatment and follow-up period. Furthermore,primary endothelial cells (HUVEC) and primary pulmonaryfibroblasts were less sensitive to ADAM17 inhibition, and onlytumor cells were sensitized to IR by TMI-005 (SupplementaryFig. S6B–S6D).

Collectively, these results indicate that combined treatment ofTMI-005 and IR exerts a supra-additive tumor growth delay inthese NSCLC cell–derived tumor xenografts and leads to bettertumor control with minimal normal tissue toxicity.

DiscussionIncreased expression of ADAM17 significantly correlates with

enhanced aggression of NSCLC on the clinical level (27, 28), andADAM17-shedded substrates are cytoprotective against differentantitumor agents, as demonstrated inmultiple tumor cell systems(4, 13, 37). Here, we identified that IR promotes furin-mediatedactivation of ADAM17 with increased shedding of ADAM17substrates and thereby contributes to an IR-induced stressresponse in NSCLC. Subsequently, direct inhibition of ADAM17lowered an intrinsic and treatment-induced threshold and strong-ly enhanced the efficacy of IR in vitro and in vivo. These resultsrepresent a promising rationale for the clinical assessment of acombined treatment modality of ADAM17 inhibitors with irra-diation inNSCLC patients. We identified ADAM17 as direct targetfor radiosensitization based on our semiquantitative antibodyarray approach, screening for factors differentially secreted inresponse to irradiation. These factors were quantitatively validat-ed by ELISA across a panel of NSCLC cell lines and couldsubsequently also bemonitored in the blood serum derived frommice with locally irradiated tumors. As such, serum detection ofADAM17 substrates in irradiated tumor patients might also serveas a relevant, minimally invasive endpoint on the clinical level.

An increase of specific ADAM17 substrates after irradiation waspreviously detected on the preclinical and clinical level withoutinvestigating the underlying mechanism mediating radiation-induced secretion. For example, upregulated CXCL16 levels wereobserved in breast cancer cell lines in response to IR (38) andincreased TGF-a levelswere detected after irradiation inhormone-refractory prostate cancer patients (4). Increased ADAM17 sub-strate secretion in response to irradiation was also reported fromlung and prostate carcinoma cells by Chung and colleagues,investigating in vitro a signaling and radiosensitizing link betweenMEK1/2 inhibition and reduced EGF ligand secretion (39).Importantly, we demonstrated here in vitro and in vivo that IR-enhanced secretion of ADAM17 substrates is primarily driven byIR-dependent, posttranslational activation of ADAM17 and notby IR-increased expression of the respective ADAM17 substrates.

The exact regulatory steps required for ADAM17 activation orfor any ADAMprotease remain poorly understood and posttrans-lational modification by an either Furin- and/or MAP/ERK-kinase-upstream mechanism might be stimuli- and cell sys-tem–dependent. ADAM17 activity is also increased on cellulartreatment with chemotherapeutic agents (40, 41) and ADAM17

activation by the antimetabolite 5-FU and the topoisomerase Iinhibitor SN-38 primarily occurs in a Kras-mutated backgroundand via the MAP/ERK kinase pathway. Our results demonstratethat irradiation promotes ADAM17 activation, but in a furin-mediated way and interestingly, in both Kras-wild-type (NCI-H125, CALU-3) and Kras-mutated NSCLC cell lines (A549,H460). More importantly, we demonstrate the potency of directpharmacologic targeting of ADAM17 to sensitize for IR andindependent of the Kras status (A549, NCI-H125).

Inhibition of ADAM17 significantly reduces the secretion ofmultiple ADAM17 substrates. Insofar, ADAM17 is a promisingtarget as inhibition of its proteolytic activity not only affects asingle receptor tyrosine kinase, for example, the EGFR, and itsdownstream signal transduction cascades, but deregulates mul-tiple auto- and paracrine-controlled processes. Only recentlynovel ADAM17-directed inhibitory antibodies have been devel-oped and demonstrated broad antitumor potency both in EGFRligand–dependent and -independent tumor cells (42, 43). WhileEGFR-directed agents, also in combination with radiotherapy, area promising treatment strategy, many tumors rapidly acquireresistance during the course of treatment. Alternative conceptssuggest combination therapies of EGFR inhibition in combina-tion with inhibitors for other HER family receptors (44, 45). Amajor advantage of targeting ADAM17 includes its broad spec-trumof downregulated substrates for all different HER family andother RTKs, which might increase the potency of direct ADAM17targeting as compared with the inhibition of individual receptors.

A recent phase III clinical trial for locally advanced NSCLCinvestigated both radiotherapy dose escalation and the combineduse of cetuximab, which both failed to further improve treatmentoutcome (21). Lack of responsiveness to EGFR inhibition mightbe linked to the Kras mutational status; however, the specific roleof Kras in NSCLC for the sensitivity to EGFR inhibitors alone andin combination with radiotherapy is still controversial (46–49).We used the EGFR phosphorylation status as surrogate marker todetermine shedding of ADAM17-mediated EGFR ligands. Inter-estingly, irradiation induced prolonged activation of EGF-RTKactivity, which correlated with increased IR-mediated ADAM17activation. EGF-RTK activity was strongly counteracted by phar-macologic inhibition of ADAM17 or in ADAM17 knockdowncells with subsequent downregulation of EGFR ligand shedding.In contrast, the EGFR-directed antibody cetuximab only partiallyreduced prolonged, IR-dependent RTK activation and did notsensitize for IR. Thus, ADAM17 might be a superior target forradiosensitization due to the inhibition of multiple downstreamprocesses, but also pharmacodynamically due to sustained inhi-bition of ligand shedding and thereby reducing an IR-induced,acquired resistance mechanism. As NSCLC is also treated withradiochemotherapy regimens including classic chemotherapeu-tics, for example, cisplatinum and taxanes, it will be important toprobe in detail inhibition of these pleiotropic effects by ADAM17inhibition in combination with chemoradiation for NSCLC.

IR-induced ADAM17 activity and subsequent shedding ofADAM17 ligands are part of prosurvival responses. Shed ligands,such as HER family receptor ligands, ALCAM and other chemo-kines, might directly increase the cellular capacity to cope withIR-inducedDNAdamage,may influence themigratory capacity oftarget cells, or may attract immune cells to the tumor microen-vironment, respectively. At this stage, we have not evaluatedthe relevance of the individual shed ligands for an increasedtreatment threshold acting in an auto- and paracrine way in vivo.

ADAM17 and Ionizing Radiation in NSCLC

www.aacrjournals.org Clin Cancer Res; 22(17) September 1, 2016 4437

on March 4, 2021. © 2016 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst April 13, 2016; DOI: 10.1158/1078-0432.CCR-15-2449

Page 11: Secretome Signature Identifies ADAM17 as Novel Target for ... · Ashish Sharma, Sabine Bender, Martina Zimmermann, Oliver Riesterer, Angela Broggini-Tenzer, and Martin N. Pruschy

Furthermore, the relevance of differential ADAM17 activities,differentially expressed substrates, and the absolute amounts andinterplay between ligands, shed to different extents from differenttumors cells, will also require in-depth investigations. The supra-additive effect of ADAM17 inhibitors on clonogenic survival ofirradiated cells in vitro indicates a mechanism for radiosensitiza-tion that is at least in part based on an autocrine loop. Further-more, we cannot dissect the impact of basal and radiation-induced ligand shedding for the ADAM17 substrate–mediatedtreatment threshold. However, limited downregulation of radia-tion-induced shedding by the less potent ADAM-directed si-ADAM17#1 suggest that already minor changes of ADAM17activity directly affect an ADAM17-regulated survival and treat-ment threshold.

Our in vivo results demonstrate the potency of the combinedtreatment modality of the ADAM17 inhibitor TMI-005 and IR.TMI-005 significantly reduced the tumor proliferative capacityand exerted a supra-additive tumor growth delay in combinationwith IR. TMI-005, also known as Apratastat, is an orally bioavail-able small-molecular inhibitor of ADAM17, which was initiallydesigned for the treatment of rheumatoid arthritis as it inhibitsTNFa release (50, 51). Adequate ADAM17-inhibitory clinicaldosing for TMI-005 was established in a long-term, randomizedphase II study with up to 390 rheumatoid arthritis patients (52).Although, the clinical program with TMI-005 and other closelyrelated derivatives were stopped due to lack of efficacy in rheu-matoid arthritis, related to constitutive activation of the TNFareceptor on immunologic cells, but not due to toxicity reasons(50, 53). On the basis of its low toxicity profile and targetrelevance independent of TNFa, TMI-005 and other new classesof ADAM17 inhibitors have thus a strong rational for reposition-ing in NSCLC as part of a combined treatment modality withradiotherapy.

Disclosure of Potential Conflicts of InterestNo potential conflicts of interest were disclosed.

Authors' ContributionsConceptionanddesign:A. Sharma, S. Bender, A. Broggini-Tenzer,M.N. PruschyDevelopment of methodology: A. Sharma, S. Bender, A. Broggini-Tenzer,M.N. PruschyAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): A. Sharma, S. Bender, M. ZimmermannAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis):A. Sharma, S. Bender,O. Riesterer, A. Broggini-Tenzer,M.N. PruschyWriting, review, and/or revision of the manuscript: A. Sharma, S. Bender,O. Riesterer, A. Broggini-Tenzer, M.N. PruschyAdministrative, technical, or material support (i.e., reporting or organizingdata, constructing databases): O. Riesterer, A. Broggini-Tenzer, M.N. PruschyStudy supervision: M.N. Pruschy

AcknowledgmentsThe authors thank Kristian Ikenberg and the Department of Pathology,

University Hospital Zurich, for excellent support, the Biologisches Zentrallaborof the University Hospital Zurich for animal housing, and Janosch Ott fortechnical support during animal experiments.

Grant SupportThis work was supported in part by grants from the Swiss Cancer League

(KLS-02788-02-2011), the Swiss National Science Foundation (144060), theVontobel Stiftung, the Fonds für Medizinische Forschungen, the HartmannMueller Foundation (1797), and the KFSP Tumor Oxygenation of theUniversity of Zurich.

The costs of publication of this articlewere defrayed inpart by the payment ofpage charges. This article must therefore be hereby marked advertisement inaccordance with 18 U.S.C. Section 1734 solely to indicate this fact.

Received October 13, 2015; revised March 11, 2016; accepted April 4, 2016;published OnlineFirst April 13, 2016.

References1. Moding EJ, KastanMB, KirschDG. Strategies for optimizing the response of

cancer and normal tissues to radiation. Nat Rev Drug Discov 2013;12:526–42.

2. Pfister DG, Johnson DH, Azzoli CG, Sause W, Smith TJ, Baker S Jr, et al.American Society of Clinical Oncology treatment of unresectable non-small-cell lung cancer guideline: update 2003. J Clin Oncol 2004;22:330–53.

3. Solda F, Lodge M, Ashley S, Whitington A, Goldstraw P, Brada M. Stereo-tactic radiotherapy (SABR) for the treatment of primary non-small cell lungcancer; systematic review and comparisonwith a surgical cohort. RadiotherOncol 2013;109:1–7.

4. Hagan M, Yacoub A, Dent P. Ionizing radiation causes a dose-dependentrelease of transforming growth factor alpha in vitro from irradiatedxenografts and during palliative treatment of hormone-refractory prostatecarcinoma. Clin Cancer Res 2004;10:5724–31.

5. Shan YX, Jin SZ, Liu XD, Liu Y, Liu SZ. Ionizing radiation stimulatessecretion of pro-inflammatory cytokines: dose-response relationship,mechanisms and implications. Radiat Environ Biophys 2007;46:21–9.

6. Shen CJ, Sharma A, Vuong DV, Erler JT, Pruschy M, Broggini-Tenzer A.Ionizing radiation induces tumor cell lysyl oxidase secretion. BMC Cancer2014;14:532.

7. Broggini-Tenzer A, Sharma A, Nytko KJ, Bender S, Vuong V, Orlowski K,et al. Combined treatment strategies for microtubule stabilizing agent-resistant tumors. J Natl Cancer Inst 2015;107pii:dju504.

8. Karar J, Maity A. Modulating the tumor microenvironment to increaseradiation responsiveness. Cancer Biol Ther 2009;8:1994–2001.

9. Verheij M, Vens C, van Triest B. Novel therapeutics in combination withradiotherapy to improve cancer treatment: rationale,mechanisms of actionand clinical perspective. Drug Resist Updat 2010;13:29–43.

10. Abdollahi A, Li M, Ping G, Plathow C, Domhan S, Kiessling F, et al.Inhibition of platelet-derived growth factor signaling attenuates pulmo-nary fibrosis. J Exp Med 2005;201:925–35.

11. Biswas S, Guix M, Rinehart C, Dugger TC, Chytil A, Moses HL, et al.Inhibition of TGF-beta with neutralizing antibodies prevents radiation-induced acceleration of metastatic cancer progression. J Clin Invest 2007;117:1305–13.

12. Flechsig P, Hartenstein B, Teurich S, Dadrich M, Hauser K, Abdollahi A,et al. Loss of matrix metalloproteinase-13 attenuates murine radiation-induced pulmonary fibrosis. Int J Radiat Oncol Biol Phys 2010;77:582–90.

13. Gorski DH, Beckett MA, Jaskowiak NT, Calvin DP, Mauceri HJ, SalloumRM, et al. Blockage of the vascular endothelial growth factor stress responseincreases the antitumor effects of ionizing radiation. Cancer Res 1999;59:3374–8.

14. Wang SC, Yu CF, Hong JH, Tsai CS, Chiang CS. Radiation therapy-inducedtumor invasiveness is associated with SDF-1-regulated macrophage mobi-lization and vasculogenesis. PLoS One 2013;8:e69182.

15. Hynes NE, Schlange T. Targeting ADAMS and ERBBs in lung cancer. CancerCell 2006;10:7–11.

16. Pruessmeyer J, Martin C, Hess FM, Schwarz N, Schmidt S, Kogel T, et al. Adisintegrin and metalloproteinase 17 (ADAM17) mediates inflammation-induced shedding of syndecan-1 and -4 by lung epithelial cells. J Biol Chem2010;285:555–64.

17. Seals DF, Courtneidge SA. The ADAMs family of metalloproteases: multi-domain proteins with multiple functions. Genes Dev 2003;17:7–30.

18. GoozM. ADAM-17: the enzyme that does it all. Crit Rev BiochemMol Biol2010;45:146–69.

Clin Cancer Res; 22(17) September 1, 2016 Clinical Cancer Research4438

Sharma et al.

on March 4, 2021. © 2016 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst April 13, 2016; DOI: 10.1158/1078-0432.CCR-15-2449

Page 12: Secretome Signature Identifies ADAM17 as Novel Target for ... · Ashish Sharma, Sabine Bender, Martina Zimmermann, Oliver Riesterer, Angela Broggini-Tenzer, and Martin N. Pruschy

19. Mochizuki S,Okada Y. ADAMs in cancer cell proliferation and progression.Cancer Sci 2007;98:621–8.

20. Saftig P, Reiss K. The "A Disintegrin And Metalloproteases" ADAM10 andADAM17: novel drug targets with therapeutic potential? Eur J Cell Biol2011;90:527–35.

21. Bradley JD, Paulus R, Komaki R,Masters G, BlumenscheinG, Schild S, et al.Standard-dose versus high-dose conformal radiotherapy with concurrentand consolidation carboplatin plus paclitaxel with or without cetuximabfor patientswith stage IIIA or IIIB non-small-cell lung cancer (RTOG0617):a randomised, two-by-two factorial phase 3 study. Lancet Oncol2015;16:187–99.

22. JackmanD, PaoW, Riely GJ, Engelman JA, KrisMG, Janne PA, et al. Clinicaldefinition of acquired resistance to epidermal growth factor receptortyrosine kinase inhibitors in non-small-cell lung cancer. J Clin Oncol2010;28:357–60.

23. Mendelsohn J, Baselga J. Epidermal growth factor receptor targeting incancer. Semin Oncol 2006;33:369–85.

24. Riely GJ, KrisMG, Zhao B, Akhurst T,MiltonDT,Moore E, et al. Prospectiveassessment of discontinuation and reinitiation of erlotinib or gefitinib inpatients with acquired resistance to erlotinib or gefitinib followed by theaddition of everolimus. Clin Cancer Res 2007;13:5150–5.

25. Baumgart A, Seidl S, Vlachou P, Michel L, Mitova N, Schatz N, et al.ADAM17 regulates epidermal growth factor receptor expression throughthe activation of Notch1 in non-small cell lung cancer. Cancer Res 2010;70:5368–78.

26. Murphy G. The ADAMs: signalling scissors in the tumour microenviron-ment. Nat Rev Cancer 2008;8:929–41.

27. Duffy MJ, McKiernan E, O'Donovan N, McGowan PM. Role of ADAMs incancer formation and progression. Clin Cancer Res 2009;15:1140–4.

28. Ni SS, Zhang J, Zhao WL, Dong XC, Wang JL. ADAM17 is overexpressed innon-small cell lung cancer and its expression correlates with poor patientsurvival. Tumour Biol 2013;34:1813–8.

29. Franken NA, Rodermond HM, Stap J, Haveman J, van Bree C. Clonogenicassay of cells in vitro. Nat Protoc 2006;1:2315–9.

30. Rosso O, Piazza T, Bongarzone I, Rossello A, Mezzanzanica D, Canevari S,et al. The ALCAM shedding by the metalloprotease ADAM17/TACE isinvolved in motility of ovarian carcinoma cells. Mol Cancer Res 2007;5:1246–53.

31. Sahin U, Weskamp G, Kelly K, Zhou HM, Higashiyama S, Peschon J, et al.Distinct roles for ADAM10 and ADAM17 in ectodomain shedding of sixEGFR ligands. J Cell Biol 2004;164:769–79.

32. Xu P, Liu J, Sakaki-Yumoto M, Derynck R. TACE activation by MAPK-mediated regulation of cell surface dimerization and TIMP3 association.Sci Signal 2012;5:ra34.

33. Borrell-PagesM, Rojo F, Albanell J, Baselga J, Arribas J. TACE is required forthe activation of the EGFR by TGF-alpha in tumors. EMBO J 2003;22:1114–24.

34. Sahin U, Blobel CP. Ectodomain shedding of the EGF-receptor ligandepigen is mediated by ADAM17. FEBS Lett 2007;581:41–4.

35. Anders A, Gilbert S, Garten W, Postina R, Fahrenholz F. Regulation of thealpha-secretase ADAM10 by its prodomain and proprotein convertases.FASEB J 2001;15:1837–9.

36. Tellier E, Canault M, Rebsomen L, Bonardo B, Juhan-Vague I, Nalbone G,et al. The shedding activity of ADAM17 is sequestered in lipid rafts. ExpCellRes 2006;312:3969–80.

37. Hobor S, Van Emburgh BO, Crowley E, Misale S, Di Nicolantonio F,Bardelli A. TGFalpha and amphiregulin paracrine network promotes

resistance to EGFR blockade in colorectal cancer cells. Clin Cancer Res2014;20:6429–38.

38. Matsumura S, Demaria S. Up-regulation of the pro-inflammatory chemo-kine CXCL16 is a common response of tumor cells to ionizing radiation.Radiat Res 2010;173:418–25.

39. Chung EJ, Urick ME, Kurshan N, Shield W, Asano H III, Smith PD, et al.MEK1/2 inhibition enhances the radiosensitivity of cancer cells by down-regulating survival and growth signals mediated by EGFR ligands. Int JOncol 2013;42:2028–36.

40. Kyula JN, Van Schaeybroeck S, Doherty J, Fenning CS, Longley DB,Johnston PG. Chemotherapy-induced activation of ADAM-17: a novelmechanism of drug resistance in colorectal cancer. Clin Cancer Res 2010;16:3378–89.

41. Van Schaeybroeck S, Kyula JN, FentonA, FenningCS, Sasazuki T, ShirasawaS, et al. Oncogenic Kras promotes chemotherapy-induced growth factorshedding via ADAM17. Cancer Res 2011;71:1071–80.

42. Caiazza F, McGowan PM, Mullooly M, Murray A, Synnott N, O'DonovanN, et al. Targeting ADAM-17 with an inhibitory monoclonal antibody hasantitumour effects in triple-negative breast cancer cells. Br J Cancer2015;112:1895–903.

43. Rios-Doria J, Sabol D, Chesebrough J, Stewart D, Xu L, Tammali R, et al. AMonoclonal Antibody to ADAM17 Inhibits Tumor Growth by InhibitingEGFR and Non-EGFR-Mediated Pathways. Mol Cancer Ther 2015;14:1637–49.

44. Huang S, Li C, Armstrong EA, Peet CR, Saker J, Amler LC, et al. Dualtargeting of EGFR and HER3 with MEHD7945A overcomes acquiredresistance to EGFR inhibitors and radiation. Cancer Res 2013;73:824–33.

45. Iida M, Brand TM, Starr MM, Huppert EJ, Luthar N, Bahrar H, et al.Overcoming acquired resistance to cetuximabbydual targetingHER familyreceptors with antibody-based therapy. Mol Cancer 2014;13:242.

46. Choi EJ, Ryu YK, KimSY,WuHG,Kim JS, Kim IH, et al. Targeting epidermalgrowth factor receptor-associated signaling pathways in non-small celllung cancer cells: implication in radiation response. Mol Cancer Res 2010;8:1027–36.

47. Dittmann K, Mayer C, Rodemann HP. Inhibition of radiation-inducedEGFR nuclear import by C225 (Cetuximab) suppresses DNA-PK activity.Radiother Oncol 2005;76:157–61.

48. Mukohara T, Engelman JA,HannaNH, YeapBY, Kobayashi S, LindemanN,et al. Differential effects of gefitinib and cetuximab on non-small-cell lungcancers bearing epidermal growth factor receptor mutations. J Natl CancerInst 2005;97:1185–94.

49. Wild R, Fager K, Flefleh C, Kan D, Inigo I, Castaneda S, et al. Cetuximabpreclinical antitumor activity (monotherapy and combination based) isnot predicted by relative total or activated epidermal growth factor receptortumor expression levels. Mol Cancer Ther 2006;5:104–13.

50. Moss ML, Sklair-Tavron L, Nudelman R. Drug insight: tumor necrosisfactor-converting enzyme as a pharmaceutical target for rheumatoid arthri-tis. Nat Clin Pract Rheumatol 2008;4:300–9.

51. Talele TT, Khedkar SA, Rigby AC. Successful applications of computer aideddrug discovery: moving drugs from concept to the clinic. Curr Top MedChem 2010;10:127–41.

52. ThabetMM,Huizinga TW.Drug evaluation: apratastat, a novel TACE/MMPinhibitor for rheumatoid arthritis. Curr Opin Investig Drugs 2006;7:1014–9.

53. Shu C, Zhou H, Afsharvand M, Duan L, Zhang H, Noveck R, et al.Pharmacokinetic-pharmacodynamic modeling of apratastat: a popula-tion-based approach. J Clin Pharmacol 2011;51:472–81.

www.aacrjournals.org Clin Cancer Res; 22(17) September 1, 2016 4439

ADAM17 and Ionizing Radiation in NSCLC

on March 4, 2021. © 2016 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst April 13, 2016; DOI: 10.1158/1078-0432.CCR-15-2449

Page 13: Secretome Signature Identifies ADAM17 as Novel Target for ... · Ashish Sharma, Sabine Bender, Martina Zimmermann, Oliver Riesterer, Angela Broggini-Tenzer, and Martin N. Pruschy

2016;22:4428-4439. Published OnlineFirst April 13, 2016.Clin Cancer Res   Ashish Sharma, Sabine Bender, Martina Zimmermann, et al.  

Small Cell Lung Cancer−Radiosensitization of Non Secretome Signature Identifies ADAM17 as Novel Target for

  Updated version

  10.1158/1078-0432.CCR-15-2449doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://clincancerres.aacrjournals.org/content/suppl/2016/04/13/1078-0432.CCR-15-2449.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://clincancerres.aacrjournals.org/content/22/17/4428.full#ref-list-1

This article cites 52 articles, 21 of which you can access for free at:

  Citing articles

  http://clincancerres.aacrjournals.org/content/22/17/4428.full#related-urls

This article has been cited by 2 HighWire-hosted articles. Access the articles at:

   

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

  Subscriptions

Reprints and

  [email protected]

To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at

  Permissions

  Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

.http://clincancerres.aacrjournals.org/content/22/17/4428To request permission to re-use all or part of this article, use this link

on March 4, 2021. © 2016 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst April 13, 2016; DOI: 10.1158/1078-0432.CCR-15-2449