+ All Categories
Home > Documents > L1 Cell Adhesion Molecule Promotes Tumorigenicity and Metastatic Potential … · Human Cancer...

L1 Cell Adhesion Molecule Promotes Tumorigenicity and Metastatic Potential … · Human Cancer...

Date post: 30-Mar-2021
Category:
Upload: others
View: 3 times
Download: 0 times
Share this document with a friend
12
Human Cancer Biology L1 Cell Adhesion Molecule Promotes Tumorigenicity and Metastatic Potential in NonSmall Cell Lung Cancer Josephine Hai 1,2 , Chang-Qi Zhu 1 , Bizhan Bandarchi 1 , Yu-Hui Wang 1 , Roya Navab 1 , Frances A. Shepherd 1,3 , Igor Jurisica 1,4 , and Ming-Sound Tsao 1,2,5 Abstract Purpose: Non–small cell lung cancer (NSCLC) is a highly metastatic cancer with limited treatment options, thus requiring development of novel targeted therapies. Our group previously identified L1 cell adhesion molecule (L1CAM) expression as a member of a prognostic multigene expression signature for NSCLC patients. However, there is little information on the biologic function of L1CAM in lung cancer cells. This study investigates the functional and prognostic role of L1CAM in NSCLC. Experimental Design: Cox proportional hazards regression analysis was done on four independent published mRNA expression datasets of primary NSCLCs. L1CAM expression was suppressed by short- hairpin RNA (shRNA)–mediated silencing in human NSCLC cell lines. Effects were assessed by examining in vitro migration and invasion, in vivo tumorigenicity in mice, and metastatic potential using an orthotopic xenograft rat model of lung cancer. Results: L1CAM is an independent prognostic marker in resected NSCLC patients, with overexpression strongly associated with worse prognosis. L1CAM downregulation significantly decreased cell motility and invasiveness in lung cancer cells and reduced tumor formation and growth in mice. Cells with L1CAM downregulation were deficient in constitutive extracellular signal–regulated kinase (Erk) activation. Ortho- topic studies showed that L1CAM suppression in highly metastatic lung cancer cells significantly decreases spread to distant organs, including bone and kidney. Conclusion: L1CAM is a novel prometastatic gene in NSCLC, and its downregulation may effectively suppress NSCLC tumor growth and metastasis. Targeted inhibition of L1CAM may be a novel therapy for NSCLC. Clin Cancer Res; 18(7); 1914–24. Ó2012 AACR. Introduction Lung cancer is one of the most lethal cancers with an estimated 1.3 million deaths per year worldwide (1). Non– small cell lung cancer (NSCLC) comprises approximately 85% of all lung cancers. Despite recent identification of many oncogenes and tumor suppressor genes important for the development and treatment of NSCLC, overall 5-year survival remains poor at 15% (2–6). Furthermore, although early-stage patients potentially are curable by surgery, approximately 30% to 60% recur postoperatively and die from metastatic disease (7). Factors contributing to early progression and metastasis in NSCLC remain largely unknown. The identification of new pathways responsible for tumor progression is critical to the development of both novel prognostic markers and therapies for NSCLC. Metastasis is associated with profound changes in cellular properties, including the disruption of cell–cell and cell– matrix adhesions and the acquisition of migratory abilities. Deregulated expression of members of all major cell adhe- sion molecule families [integrins, selectins, cadherins, and the immunoglobulin (Ig) superfamily] is frequently reported in human cancers (8). One such molecule, L1 cell adhesion molecule (L1CAM), is aberrantly expressed in several cancer types, including ovarian (9), melanoma (10), breast (11), pancreatic (12), and colon cancers (13). L1CAM is a type I membrane glycoprotein of the Ig-like superfamily, consisting of 6 Ig-like domains and 5 fibronectin-type III repeats, followed by a single-pass trans- membrane region and a highly conserved cytoplasmic tail (14). The extracellular region of L1CAM binds to a diverse range of proteins that include L1CAMs themselves, other cell surface proteins such as integrins and axonin1, and extracellular matrix proteins such as laminin and neurocan Authors' Afliations: 1 Ontario Cancer Institute/Princess Margaret Hospi- tal, University Health Network; and Departments of 2 Medical Biophysics, 3 Medicine, 4 Computer Science, and 5 Laboratory Medicine and Pathobi- ology, University of Toronto, Toronto, Ontario, Canada Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/). Corresponding Author: Ming-Sound Tsao, Room 7-613, Princess Mar- garet Hospital, 610 University Avenue, Toronto, Ontario M5G 2M9, Canada. Phone: 416-340-4737; Fax: 416-340-5571; E-mail: [email protected] doi: 10.1158/1078-0432.CCR-11-2893 Ó2012 American Association for Cancer Research. Clinical Cancer Research Clin Cancer Res; 18(7) April 1, 2012 1914 on August 24, 2021. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from Published OnlineFirst February 3, 2012; DOI: 10.1158/1078-0432.CCR-11-2893
Transcript
Page 1: L1 Cell Adhesion Molecule Promotes Tumorigenicity and Metastatic Potential … · Human Cancer Biology L1 Cell Adhesion Molecule Promotes Tumorigenicity and Metastatic Potential in

Human Cancer Biology

L1 Cell Adhesion Molecule Promotes Tumorigenicityand Metastatic Potential in Non–Small CellLung Cancer

Josephine Hai1,2, Chang-Qi Zhu1, Bizhan Bandarchi1, Yu-Hui Wang1, Roya Navab1, Frances A. Shepherd1,3,Igor Jurisica1,4, and Ming-Sound Tsao1,2,5

AbstractPurpose: Non–small cell lung cancer (NSCLC) is a highly metastatic cancer with limited treatment

options, thus requiring development of novel targeted therapies. Our group previously identified L1 cell

adhesion molecule (L1CAM) expression as a member of a prognostic multigene expression signature for

NSCLCpatients. However, there is little information on the biologic function of L1CAM in lung cancer cells.

This study investigates the functional and prognostic role of L1CAM in NSCLC.

Experimental Design: Cox proportional hazards regression analysis was done on four independent

published mRNA expression datasets of primary NSCLCs. L1CAM expression was suppressed by short-

hairpin RNA(shRNA)–mediated silencing in humanNSCLC cell lines. Effectswere assessed by examining in

vitro migration and invasion, in vivo tumorigenicity in mice, and metastatic potential using an orthotopic

xenograft rat model of lung cancer.

Results: L1CAM is an independent prognostic marker in resected NSCLC patients, with overexpression

strongly associated with worse prognosis. L1CAM downregulation significantly decreased cell motility and

invasiveness in lung cancer cells and reduced tumor formation and growth in mice. Cells with L1CAM

downregulation were deficient in constitutive extracellular signal–regulated kinase (Erk) activation. Ortho-

topic studies showed that L1CAM suppression in highly metastatic lung cancer cells significantly decreases

spread to distant organs, including bone and kidney.

Conclusion: L1CAM is a novel prometastatic gene in NSCLC, and its downregulation may effectively

suppress NSCLC tumor growth and metastasis. Targeted inhibition of L1CAM may be a novel therapy for

NSCLC. Clin Cancer Res; 18(7); 1914–24. �2012 AACR.

IntroductionLung cancer is one of the most lethal cancers with an

estimated 1.3 million deaths per year worldwide (1). Non–small cell lung cancer (NSCLC) comprises approximately85% of all lung cancers. Despite recent identification ofmany oncogenes and tumor suppressor genes important forthe development and treatment of NSCLC, overall 5-yearsurvival remains poor at 15% (2–6). Furthermore, althoughearly-stage patients potentially are curable by surgery,approximately 30% to 60% recur postoperatively and die

from metastatic disease (7). Factors contributing to earlyprogression and metastasis in NSCLC remain largelyunknown. The identification of new pathways responsiblefor tumor progression is critical to the development of bothnovel prognostic markers and therapies for NSCLC.

Metastasis is associatedwith profound changes in cellularproperties, including the disruption of cell–cell and cell–matrix adhesions and the acquisition of migratory abilities.Deregulated expression of members of all major cell adhe-sion molecule families [integrins, selectins, cadherins, andthe immunoglobulin (Ig) superfamily] is frequentlyreported in human cancers (8). One such molecule, L1 celladhesion molecule (L1CAM), is aberrantly expressed inseveral cancer types, including ovarian (9), melanoma(10), breast (11), pancreatic (12), and colon cancers(13). L1CAM is a type I membrane glycoprotein of theIg-like superfamily, consisting of 6 Ig-like domains and 5fibronectin-type III repeats, followed by a single-pass trans-membrane region and a highly conserved cytoplasmic tail(14). The extracellular region of L1CAM binds to a diverserange of proteins that include L1CAMs themselves, othercell surface proteins such as integrins and axonin1, andextracellular matrix proteins such as laminin and neurocan

Authors' Affiliations: 1Ontario Cancer Institute/Princess Margaret Hospi-tal, University Health Network; and Departments of 2Medical Biophysics,3Medicine, 4Computer Science, and 5Laboratory Medicine and Pathobi-ology, University of Toronto, Toronto, Ontario, Canada

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

Corresponding Author: Ming-Sound Tsao, Room 7-613, Princess Mar-garet Hospital, 610 University Avenue, Toronto, Ontario M5G 2M9,Canada. Phone: 416-340-4737; Fax: 416-340-5571; E-mail:[email protected]

doi: 10.1158/1078-0432.CCR-11-2893

�2012 American Association for Cancer Research.

ClinicalCancer

Research

Clin Cancer Res; 18(7) April 1, 20121914

on August 24, 2021. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst February 3, 2012; DOI: 10.1158/1078-0432.CCR-11-2893

Page 2: L1 Cell Adhesion Molecule Promotes Tumorigenicity and Metastatic Potential … · Human Cancer Biology L1 Cell Adhesion Molecule Promotes Tumorigenicity and Metastatic Potential in

(15, 16). L1CAM function has been most extensively stud-ied in the nervous system, where it is known to orchestratemorphogenetic events such as neuron–neuron adhesion,axon guidance, neurite outgrowth, neurite fasciculation,and myelination (15, 17). Consistent with these functions,mutations in human L1CAM confer a variety of neurologicdisorders and L1CAM knockout mice show defective corti-cospinal tract development (17–19).The function of L1CAM in lung cancer growth and

metastasis is unknown. In this study, we characterize tumorgrowth andmetastatic potential of L1CAM in subcutaneousmice and orthotopic nude rat models of lung cancer,respectively. We show that L1CAM plays a significant rolein stimulating cell migration, invasiveness, and systemicmetastasis in vivo, thereby establishing L1CAM as a novelbona fide therapeutic target for NSCLC.

Materials and MethodsPrognostic significance of L1CAM in NSCLC patientsThe prognostic value of L1CAM expression was assessed

in silico using published NSCLC microarray data from 4patient cohorts (Supplementary Table S1). The JBR.10 wasa phase III randomized trial of adjuvant chemotherapy(cisplatin and vinorelbine) versus observation in stage IB-II patients (20, 21). For prognostic analysis, only patientsin observation arm were analyzed. The National CancerInstitute Directors’ Challenge Consortium (DCC) cohortincluded 442 adenocarcinomas patients from 4 NorthAmerican cancer centers. Excluding patients from theJBR.10 cohort contributed to this DCC study and patientswho received adjuvant chemo/radiotherapy, expressiondata from the 311 patients were used for prognosticanalysis (22). The University of Michigan cohort consistedof 129 stage I–III squamous cell carcinomas (23). Geneexpression analyses from the above 3 cohorts were doneusing the Affymetrix U133A microarray. A final cohortincluded 172 surgically resected NSCLC patients from the

Netherlands Cancer Institute (NLCI). Their gene expres-sion profiling was conducted by Agilent 44k microarray(24). Recurrence-free survival was used as the survival endpoint for JBR.10 and NLCI data sets, and 5-year survivalwas used for DCC and Michigan data sets, in whichrecurrence-free survival was not available. The associationof the expression of L1CAM and survival was evaluatedusing Cox proportional hazards regression in SAS v9.2(SAS Institute) with gene expression as a continuousvariable.

On the basis of UCSC genome browser GRCh37/hg19assembly, L1CAM mapped to 2 Affymetrix probe sets (forboth U133A and U133 plus 2). Probe set 204584_at coversall 3 transcripts whereas 204585_s_at covers only 2 tran-scripts; thus, probe set 204584_at was used to representL1CAM expression. In the Agilent 44K,C_MaP1_0_ID41179 or A_24_P207995 represents tran-script variant 1, which is the longest transcript, and thiswas used to represent the expression of L1CAM.

Quantitative real-time PCR validationQuantitative real-time PCR (qRT-PCR) assays were done

on 156 RNA samples from the University Health Network(UHN)NSCLC patient cohort as previously described (25).mRNA copy number for each sample was quantified inHT7900 fast real-time PCR System (Applied Biosystems)and calculated by standard curve methodology. The copynumber was adjusted by the geometric mean of 4 house-keeping genes (TBP, BAT1, B2M, and ACTB) and log2-transformed (25). L1CAM primers targeting the 204584_atprobe set region were used: HsL1CAM-fw: 50-AAAGGAAA-GATTGGTTCTCCCAG -30; HsL1CAM-rev: 50-AGTAGAC-CAAGCACAGGCATACAG -30.

Generation of L1CAM short hairpin RNA gene silencingand overexpression

L1CAM gene expression was stably knockdown usingshort hairpin RNAs (shRNA) against different regions ofhuman L1CAM. All shRNA sequences were obtained fromRNAi Consortium (TRC2). The target sequences forshL1CAM5 and shL1CAM2were 50-GCCAATGCCTACATC-TACGTT-30 and 50-GCTAACCTGAAGGTTAAAGAT-30,respectively. Human full-length L1CAM cDNA plasmid,phL1A-pcDNA3, was obtained commercially (Addgene)and was subcloned into our modified Gateway recombina-tion lentiviral expression vector, pLKO.puro.DEST (26),containing a puromycin selection marker. Transduced cellswere selected by 1 mg/mL puromycin. For controls, non-specific shRNA against GFP (shGFP) was used for cell lineswith knockdowns,whereas empty vector constructwas usedfor overexpressed cell lines.

Generation of mutant L1CAM sequence resistant toshL1CAM.5

L1CAM cDNA insensitive to shL1CAM.5 was producedby insertion of 3 point mutations (C1359G, C1362T, andC1365T) at the shRNA target region by QuikChange II XLSite-directed Mutagenesis Kit (Stratagene), according to the

Translational RelevanceThirty to60%of early-stagenon–small cell lung cancer

(NSCLC) patients still develop metastatic recurrenceafter complete surgical resection. Although adjuvantchemotherapy has been shown to modestly increase thesurvival of these patients, novel targeted therapies areurgently needed to improve adjuvant treatment. Thisstudy shows that in NSCLC with high L1CAM expres-sion, tumor growth and systemicmetastasis are inhibitedby L1CAM downregulation in subcutaneous and ortho-topic models, respectively. We also confirm that L1CAMis a poor prognosticmarker for patients withNSCLC andplays a significant role in promoting invasiveness andmotility of NSCLC cells. The results of our study high-light targeting of L1CAM as a potentially importantnovel therapy for NSCLC patients.

L1CAM in Non–Small Cell Lung Cancer

www.aacrjournals.org Clin Cancer Res; 18(7) April 1, 2012 1915

on August 24, 2021. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst February 3, 2012; DOI: 10.1158/1078-0432.CCR-11-2893

Page 3: L1 Cell Adhesion Molecule Promotes Tumorigenicity and Metastatic Potential … · Human Cancer Biology L1 Cell Adhesion Molecule Promotes Tumorigenicity and Metastatic Potential in

manufacturer’s instructions. All mutations were verified byDNA sequencing. Mutant L1CAM was subcloned into alentiviral vector for subsequent stable transduction. Thefollowing primers were designed (nucleotide mismatch isunderlined):

L1mut_fw: 50 -GGGCTCTTGCTGGCCAATGCGTATATTT-ACGTTGTCC-30;L1mut_rev: 50-GGACAACGTAAATATACGCATTGGCCAG-CAAGAGCCC-30

Animals and cell linesFour-week-old male nude (CR:NIH-RNU) rats (Charles

River, Inc.) were acclimatized for 1 to 2 weeks beforeentering study protocols. Rats were housed in sterilizedcages and fed autoclaved food and water ad libitum. Severecombine immunodeficient (SCID) mice were bred on siteand obtained from the Ontario Cancer Institute (OCI;Toronto, ON) animal facility. All manipulations were car-ried out under sterile conditions in a laminar flow hood, inaccordance with procedures approved by the OCI AnimalCare Committee.

The NCI-H460, NCI-H125, and NCI-H1264 cell lineswere obtained from the American Type Culture Collectionand verified by short-tandem repeat (STR) polymorphismanalyses. H460SM is a clonal variant of NCI-H460 selectedfor its high metastatic ability (27). All cells were cultured inRPMI media supplemented with 10% FBS (Hyclone Eur-ope, Ltd.) and antibiotics. All cells were cultivated at 37�Cand 5% CO2.

Western blot analysisWhole-cell extracts were applied to SDS-PAGE and trans-

ferred to polyvinylidene fluoridemembranes. Primary anti-bodies used were anti-L1CAM (UJ127; Novus Biologicals),b-actin, pErk1/2, Erk1/2, pAkt, Akt, pSrc, Src (Cell Signal-ing), FAK, and pFAK (BD Transduction Laboratories). Visu-alization used horseradish peroxidase–linked anti-rabbitand anti-mouse secondary antibodies (Cell Signaling) andECL-Plus blotting substrate detection kit on a Typhoonphospho- and fluorescent imaging system 9400 (GEhealthcare).

Invasion and migration assayA total of 5 � 105 cells were seeded onto 24-well

transwell cell culture plates (BD Biosciences) fitted withmultiporous (8-mm pore size) polycarbonate membranes(Falcon). The upper chambers of the membrane werecoated with 25 mg Matrigel or 7.5 mg of collagen IV, andthe lower chambers were filled with media supplementedwith 10% FBS as a chemoattractant. After 48 hours, cellswere fixed with 0.5% glutaraldehyde and stained with0.5% crystal violet reagent. Cotton swabs were used toremove the cells from the upper surface of the membrane,leaving the migratory cells on the underside. The mem-brane was removed and mounted onto glass slides andscanned. Quantitative image analysis is described in theSupplementary Materials.

Subcutaneous tumorigenicity assayOnemillion cells were injected subcutaneously in the left

shoulder region of 6- to 8-week-old male SCIDmice (n¼ 8per cell line). Mice were examined every 2 days, and tumorlength and width were measured using calipers. Tumorvolume was calculated using the following formula (length� width2)p/6. Mice were sacrificed once the humane end-point (approximately 1.5 cm diameter) was reached. Atsacrifice, portions of tumors were snap-frozen and stored inliquid nitrogen or were fixed in 10% buffered formalin forroutine histopathologic processing.

Orthotopic model to evaluate metastatic potentialTwo groups of rats (n ¼ 13 per group) were randomly

assigned for orthotopic implantation of H460SM-shL1CAM or H460SM-shGFP control cells. Details forendobronchial tumor cell implantation in nude rats wasreported previously (28). Before tumor implantation, ani-mals received 5-Gy whole-body g-radiation using the Gam-ma Cell 40 Exactor (Nordion International Inc.; 500 rad ofwhole-body g radiation from a 127Cs source and at 120 rad/min). Cultured tumor cells were harvested by trypsinizationand adjusted to a final concentration of 1.0 � 106 cells per50 mL. Rats were anesthetized by intramuscular injection ofketamine/xylazine (110 and 12 mg/kg; CDMV, Inc.,) andendobronchially implanted with 1.0� 106 cells using a 20-gauge, 1.88-inch long Teflon catheter. The cell suspensionwas injected through a catheter passed into the distalbronchus of the right caudal lobe through a small trache-otomy incision. After withdrawal of the catheter, the tra-cheotomy was repaired with a 6–0 Proline suture (EthiconInc.,), and the incision was closed with sterile wound clips.

Assessment of metastatic potentialWhen any animal from any group succumbed to the

disease or showed terminal signs and symptoms, a singleanimal from the other group was killed, allowing a directcomparison of both groups at the same time point fortumor-associated end points. Animals that died in less than21 days after implantation were considered postanestheticor surgical related complications and were excluded fromthe final comparative analysis. All tissues were fixed in 10%buffered formalin, serially sectioned, and stained withhematoxylin and eosin for microscopic examination. Theprimary tumor weights, which reflect the tumor burden,were recorded and portions of the tumor were snap-frozenand stored in liquid nitrogen. Metastases were assessedmacroscopically and microscopically in internal organsincluding lung, kidney, brain, chest wall, and bone as wellasmediastinal lymphnodes.Organsor tissueswere countedas either positive or negative for metastasis. No assessmentof number or extent of metastasis was made.

StatisticsAll statistical analyses were carried out with SAS v9.2 (SAS

Institute) and GraphPad Prism 5.0. Statistical significancewasdeterminedusing2-tailed Student t test ormixed-modelANOVA for invasion and migration assays. Difference in

Hai et al.

Clin Cancer Res; 18(7) April 1, 2012 Clinical Cancer Research1916

on August 24, 2021. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst February 3, 2012; DOI: 10.1158/1078-0432.CCR-11-2893

Page 4: L1 Cell Adhesion Molecule Promotes Tumorigenicity and Metastatic Potential … · Human Cancer Biology L1 Cell Adhesion Molecule Promotes Tumorigenicity and Metastatic Potential in

tumor growth rates of xenografts was tested using mixed-model ANOVA. c2 and Fisher exact test was used formetastasis experiments. All biologic experiments are pre-sented as mean � SEM. Tests that produced P � 0.05 wereconsidered to be significant.

ResultsPrognostic impact of L1CAM in NSCLC patientsThe 4 published microarray studies included a total of

674 early-stage surgically resected NSCLC patients who hadnot received adjuvant chemotherapy or radiotherapy. Uni-variate survival analyses revealed that high L1CAM expres-sion was significantly associated with poorer survival inJBR.10 NSCLC cohort [HR¼ 2.41, 95% confidence interval(CI) ¼ 1.51–3.82, P ¼ 0.0002], University of Michigansquamous cell carcinoma cohort (HR ¼ 2.59, 95% CI ¼1.51–4.47, P¼ 0.0006), DCC adenocarcinoma cohort (HR¼ 1.49, 95% CI ¼ 1.03–2.17, P ¼ 0.036), and marginallyassociated in the NLCI NSCLC cohort (HR ¼ 2.53, 95% CI¼ 0.92–6.95, P ¼ 0.072; Table 1). Multivariate Cox regres-sion analysis adjusted for tumor histologic subtype, stage,age, and sex showed that L1CAM expression was an inde-pendent prognostic factor for the JBR.10 and Michiganpatient cohorts and marginally in DCC and NLCI (Table1 andSupplementary Table S2).Using aqRT-PCRonmRNAof 156 NSCLC UHN patients, we confirmed further theprognostic value of L1CAM expression (multivariate HR ¼1.32, 95% CI ¼ 1.00–1.75, P ¼ 0.053; Table 1).

Establishment of stable L1CAM suppressed andoverexpressed NSCLC cell linesWe screened L1CAM protein expression by Western blot

analysis in 12 different NSCLC cell lines and an immortal-

ized but nontumorigenic human bronchial epithelial cellline (HBE135). L1CAM expression levels varied significant-ly among NSCLC cell lines and no L1CAM was detected inHBE135 cells (Fig. 1A). Due to their high endogenous levelsof L1CAM protein, the NCI-H460 large-cell carcinoma,NCI-H125 adenosquamous carcinoma, and H460SMlarge-cell carcinoma cell lines were used for subsequentknockdown experiments, whereas NCI-H1264 adenocarci-noma cells, which contain low levels of endogenousL1CAM protein, were chosen for overexpression studies.Targeting of L1CAM expression in NCI-H460 cells using 2different shRNAs resulted in reduction of L1CAM proteinlevels by 76% and 70%, respectively (Fig. 2B). Stabletransduction of shL1CAM.5 in NCI-H125 and H460SMcells reduced L1CAM protein levels by 82% and 71%,respectively. Likewise, transduction of shL1CAM.2 target-ing a different region of L1CAM decreased protein levelsin NCI-H125 and H460SM cells by 92% and 77%,respectively (Fig. 2B). To study the consequences of over-expression, we stably transduced NCI-H1264 cells withan L1CAM expression construct, which resulted in a 10-fold increase in L1CAM levels (Fig. 2B). Stable integrationand expression of L1CAM or shRNAs in all cell lines weremonitored for more than 10 passages and did not alterthe morphology of the cells, which remained epithelial inappearance.

The role of L1CAM in migration, invasion, and Erkpathway activation

Given the known involvement of L1CAM in axon guid-ance, we first examined whether modulating endogenousL1CAM expression affected cell migration inNSCLC. In ourtime-lapse wound healing assay, knockdown of L1CAMexpression in H125 and H460SM cells significantly

Table 1. Prognostic impact of L1CAM in NSCLC patients

Cohort Tumor type Platform No. of patients HRa (95% CI) P

Univariate survival analysis L1CAM (probe set: 204584_at)JBR.10 (21) NSCLC U133A 62 2.41 (1.51–3.82) 0.0002DCC (22) ADC U133A 311 1.49 (1.03–2.17) 0.036Michigan (23) SQCC U133A 129 2.59 (1.51–4.47) 0.0006NLCI (24) NSCLC 44K 172 2.53 (0.92–6.95) 0.072UHN (25) NSCLC qRT-PCR 156 1.30 (1.00–1.69) 0.050

Multivariate survival analysis L1CAM (probe set: 204584_at)JBR.10 (21) NSCLC U133A 62 3.12 (1.52–6.40) 0.002DCC (22) ADC U133A 311 1.40 (0.96–2.03) 0.081Michigan (23) SQCC U133A 129 2.73 (1.54–4.84) 0.006NLCI (24) NSCLC 44K 172 2.53 (0.92–6.95) 0.072UHN (25) NSCLC qRT-PCR 156 1.32 (1.00–1.75) 0.053

Abbreviations: DCC, Director's Challenge Consortium adenocarcinoma; NLCI, Netherlands Cancer Institute; UHN, University HealthNetwork; NSCLC, non–small cell lung cancer; U133A, Affymetrix U133A chip; 44K, Agilent 44K gene expression array; qRT-PCR, real-time quantitative PCR, ADC, adenocarcinoma; SQCC, squamous cell carcinoma; CI, confidence interval.aHR compares the overall survival of the high-risk patient group to that of the low-risk group.

L1CAM in Non–Small Cell Lung Cancer

www.aacrjournals.org Clin Cancer Res; 18(7) April 1, 2012 1917

on August 24, 2021. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst February 3, 2012; DOI: 10.1158/1078-0432.CCR-11-2893

Page 5: L1 Cell Adhesion Molecule Promotes Tumorigenicity and Metastatic Potential … · Human Cancer Biology L1 Cell Adhesion Molecule Promotes Tumorigenicity and Metastatic Potential in

decreased the rate of lateral migration into a wound intro-duced in a confluent monolayer of cells compared withcontrols (P < 0.0001; Fig. 2A). The transmigration ability ofthese same cells was evaluated on collagen IV–coatedtranswell filters. Knockdown of L1CAM in cell lines har-boring different shRNA sequences resulted in a 70% to 80%decrease in the number of migrating cells compared withcontrols (Fig. 2B). Cell proliferation did not differ betweenLICAM knockdown and control cell lines in 10% serum,indicating that altered migratory potential was not a resultof changes in cell growth (Supplementary Fig. S1A–B).

To evaluate the role of L1CAM in invasion, we examinedthe effect of L1CAMdownregulationon the ability of cells toinvade Matrigel, a reconstituted basement membrane. Theinvasiveness of H460 and H460SM cells with reducedL1CAMexpressionwasonly 26%and32%of that of controlcells, respectively (Fig. 2C). We also conducted this assaywith H1264 cells overexpressing L1CAM to determinewhether ectopic L1CAM levels could promote cell invasion.Overexpression of L1CAM resulted in a 3-fold increase incell invasion of Matrigel, confirming that L1CAM plays apivotal role in regulating both the migration and invasive-ness of lung cancer cell lines.

As previous studies have suggested that the effects ofL1CAM on cell migration may be mediated via directintegrin interaction (29), we assayed integrin-mediatedsignaling pathways by Western blot analysis. Levels oftotal focal adhesion kinase (FAK) and activated FAK(autophosphorylation site on tyrosine-391) were deter-mined by harvesting cells after serum starvation for24 hours. No differences were detected in the levels ofFAK autophosphorylation between control and L1CAMknockdown cells (Supplementary Fig. S1C). In addition,we did not observe any differences in the level of activatedSrc (phosphotyosine-416), another downstream target ofintegrin signaling.

We next examined whether L1CAM interacts with theextracellular signal–regulated kinase (Erk) pathway inNSCLC cells, as previously reported in fibroblast and mel-anoma cells (30). After incubation in serum-free media for24hours,weobservednophosphorylated Erk1/2 (pErk1/2)in cells with L1CAM knockdown, compared with low levelsof constitutively activated Erk1/2 in control cells (Fig. 2D,compare lane 1 and 2; Supplementary Fig. S1C). Whenthese cells were stimulated with 10% serum, Erk1/2 wasactivated similarly in both control and L1CAM knockdowncells, suggesting that sustained L1CAM-dependent Erk1/2activation requires co-operation with one or more serumgrowth factors. Erk1/2 activity declined to undetectablelevels by 24hours postserum treatment in cells with L1CAMknockdown, whereas control cells expressing high levels ofendogenous L1CAM sustained pErk1/2 expression (Fig. 2D,lanes 9 and 10; Fig. 2E). Although Erk1/2 activation in bothcontrol and L1CAM knockdown cells peaked at 10minutesfollowing stimulation, pErk1/2 levels began todeclinemorerapidly after 120 minutes in cells with L1CAM knockdowncompared with the control.

To confirm that the effect of L1CAMknockdownonErk1/2 activation is not an off-target or viral effect, we transducedH125-shL1CAM cells with lentivirus-expressing mutantL1CAMcDNA,which harbors silentmutations in shL1CAMtargeting sequences, and is therefore resistant to shL1CAM-induced L1CAM degradation. Restoring L1CAM in L1CAMknockdown H125 cells reversed the loss of pErk1/2 levelsafter 24 hours of serum stimulation (Fig. 2F). These resultssuggested that L1CAM function inNSCLCmay bemediatedvia the Erk pathway.

L1CAM suppression in NSCLC cells reduces in vivotumorigenicity

Given the biologic implications of our in vitro results forthe progression of NSCLC, we next examined whether

L1CAM

β-Actin

Contro

l

shL1

CAM

.5sh

L1C

AM.2

H460

Contro

l

shL1

CAM

.5sh

L1C

AM.2

H125

Contro

l

shL1

CAM

.5sh

L1C

AM.2

H460SM

Empt

y ve

ctor

plko

.L1C

AM

H1264

200/220 kDa

46 kDa

B

AH

BE

135

Large cell Adenocarcinoma Squamous

L1CAM

β-Actin

200/220 kDa

46 kDa

1.0 0.24 0.30 1.0 0.18 0.08 1.0 0.29 0.23 1.0 9.6

H46

0

H46

0SM

H12

99

RV

H68

49

A54

9

H12

64

H19

93

H35

8

H12

5

H22

6

H52

0

MG

H7

Fold:

Figure 1. Establishment of stableL1CAM knockdown andoverexpression in NSCLC celllines. A, profiling L1CAM proteinexpression in a panel of humanNSCLC cell lines. B, whole-cellextracts from cell lines stablyexpressing shRNAs againstL1CAMor full-length L1CAMcDNAwere subjected to Western blotanalysis with anti-L1CAM antibody(UJ127) and compared with shGFPor empty vector control cell lines.The levels of b-actin were used asinternal protein loading controls.

Hai et al.

Clin Cancer Res; 18(7) April 1, 2012 Clinical Cancer Research1918

on August 24, 2021. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst February 3, 2012; DOI: 10.1158/1078-0432.CCR-11-2893

Page 6: L1 Cell Adhesion Molecule Promotes Tumorigenicity and Metastatic Potential … · Human Cancer Biology L1 Cell Adhesion Molecule Promotes Tumorigenicity and Metastatic Potential in

Figure 2. Role of L1CAM on migration, invasion, and Erk pathway in NSCLC cell lines. A, time-lapse wound-healing assay was used to monitor cell frontmigration for 45 hours in H460SM and H125 cells. The rate of wound area traveled by cells with L1CAM knockdown was compared with controls. B, cellswere seeded onto transwell membranes coated with collagen IV and the number of migrated cells were counted. C, Matrigel-coated transwell assays wereused to examine cell invasion of three different cell lines. A–C, results shown represent more than 3 biologic replicates (�, P < 0.01; ��, P < 0.001;���, P < 0.0001). D, after 24 hours serum starvation, H125-shL1CAM and control cells were stimulated with 10% serum for indicated times before harvestingto assess Erk activation. Whole-cell lysates were subjected to Western blot analysis with anti-phospho-Erk1/2 and anti-Erk1/2 as a loading control.Densitometric quantification of relative amounts of pErk1/2. One of 3 experiments with comparable results is shown. E, H125 and H460 cells weresynchronized by serumstarvation and incubated in 10%serum for 24 hours before harvesting at 80%confluence to assessErk activation. F, restoring L1CAMin L1CAM knockdown H125 cells (shL1CAM.5 þ L1mut) reversed the loss of pErk levels when cells were maintained in 10% serum for 24 hours after serumstarvation. The levels of b-actin were used as internal protein loading controls.

L1CAM in Non–Small Cell Lung Cancer

www.aacrjournals.org Clin Cancer Res; 18(7) April 1, 2012 1919

on August 24, 2021. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst February 3, 2012; DOI: 10.1158/1078-0432.CCR-11-2893

Page 7: L1 Cell Adhesion Molecule Promotes Tumorigenicity and Metastatic Potential … · Human Cancer Biology L1 Cell Adhesion Molecule Promotes Tumorigenicity and Metastatic Potential in

L1CAM contributes to tumorigenicity in vivo. We subcuta-neously injected 4 independent clones of H460-shL1CAMcell lines, representing 2 different shRNA sequences, intoSCIDmice (Fig. 3A). L1CAM downregulation in H460 cellssignificantly reduced tumor growth rate when comparedwith controls (P < 0.0001; Fig. 3A), and these results wereverified in an adenosquamous carcinoma cell line model,H125 (P < 0.0001; Fig. 3B, Supplementary Fig. S3C).Moreover, the effect could be rescued by restoring L1CAMlevels using shRNA-resistant L1CAMcDNA (P < 0.0001; Fig.3C). To confirm that both H460-shL1CAM and H125-shL1CAM cells continued to express lower L1CAM levelsin vivo compared with controls, we carried out immuno-

histochemistry, qRT-PCR, and Western blot analysis onxenograft tumors formed by these cell lines (Fig. 3D andE, Supplementary Fig. S2A–B). As expected, L1CAM-sup-pressed tumor cells showed significantly reduced L1CAMstaining compared with control tumor cells. Consistentwith our earlier in vitro results, immunohistochemistryrevealed that pErk levels were reduced in vivo in L1CAMknockdown animals (Fig. 3F).

L1CAMcontributes tometastasis in anorthotopic nuderat model

To determine whether modulating L1CAM expressioncan influence metastasis in vivo, H460SM-shL1CAM and

A

L1CAM

GAPDH

Fold:

E F

******

B

H460

Control

shL1C

AM.50

100

200

300

400

pER

K H

-Sco

res

D

***

Contro

l

shL1

CAM.5

shL1

CAM.5

+ re

scue

Contro

l

clon

e #4

clon

e #3

clon

e #4

clon

e #5

1.0 0.06 0.26 0.26 0.13

shL1CAM.5 shL1CAM.2

0 2 4 6 8 10 12 14 16 18 20 22 240.0

0.5

1.0

1.5

2.0ControlshL1CAM5.4shL1CAM2.4shL1CAM5.3shL1CAM2.5

H460

Days

Mea

n t

um

or

volu

me

(cm

3 )

Mea

n t

um

or

volu

me

(cm

3 )

Mea

n t

um

or

volu

me

(cm

3 )

0 2 4 6 8 10 12 14 16 18 20 22 240.0

0.5

1.0

1.5

2.0ControlshL1CAM.5

H125

Days

β-Actin

H46

0 co

ntro

lH

460

shL1

CA

M.5

L1CAMH & E pERK 1/2

L1CAMH & E

H12

5 co

ntro

lH

125

shL1

CA

M.5

C

15x

L1CAM

0 2 4 6 8 10 12 14 16 18 20 22 240.0

0.5

1.0

1.5

shL1CAM.5shL1CAM.5 + rescue

Control

H460

Days

***

Figure 3. L1CAM promotes tumorigenicity in vivo. A, one million tumor cells were injected into the left flanks of 6- to 8-week-old SCID mice. Knockdown ofL1CAM inH460 significantly suppressed tumor growth in 4 independent clonal cell lines (n¼8per group; ���,P<0.0001). B, knockdownof L1CAM inH125celllines suppressed tumor growth significantly (n ¼ 8 per group; ���, P < 0.0001). C, rescuing L1CAM expression in H460-shL1CAM cells restored tumorgrowth (n ¼ 5 per group; ���, P < 0.0001). D–E, representative histologic sections of xenografts from H125 control and shL1CAM, and H460 control andshL1CAM, were immunostained with L1CAM and pErk antibody. F, immunohistochemical staining of pErk was done and assigned an H-score from0 to 300, calculated by multiplying the % positive cells by the relative intensity score from 0 to 3 (���, P < 0.0001). GAPDH, glyceraldehyde-3-phosphatedehydrogenase; H&E, hematoxylin and eosin.

Hai et al.

Clin Cancer Res; 18(7) April 1, 2012 Clinical Cancer Research1920

on August 24, 2021. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst February 3, 2012; DOI: 10.1158/1078-0432.CCR-11-2893

Page 8: L1 Cell Adhesion Molecule Promotes Tumorigenicity and Metastatic Potential … · Human Cancer Biology L1 Cell Adhesion Molecule Promotes Tumorigenicity and Metastatic Potential in

control cells were endobronchially implanted into theright caudal lobe airway of nude rats. In both experimen-tal and control groups, all (13 of 13) animals developedregional mediastinal lymph node metastases (Table 2).However, H460SM-shL1CAM animals showed signifi-cantly reduced distant metastases to bone (3 of 13) andkidneys (7 of 13), but not to brain and gums, whencompared with the control group (P < 0.05; Table 2).No significant differences were noted in the weight of theprimary tumors formed by both cell lines. Overall, weobserved a significant decrease in the total number ofH460SM-shL1CAM animals developing systemic metas-tasis (8 of 13) compared with the control (P < 0.05; Table2). These results strongly suggested that L1CAM maypotentiate metastasis in lung cancer.

DiscussionTo our knowledge, this is the first study to explore the

function of L1CAM in lung cancer both in vitro and in vivo.Here we show that (i) L1CAM expression correlates withclinical outcome in early-stage NSCLC patients; (ii) sup-pression of L1CAM significantly reduces migration andinvasion in three NSCLC cell lines, whereas L1CAM over-expression enhances invasion in noninvasive cells; (iii)L1CAM induces sustained Erk activation under serum stim-ulation in vitro and in vivo; and (iv) downregulation ofL1CAM significantly retards tumor growth in SCID miceand reduces metastasis in nude rats. Collectively, theseresults are strong evidence that L1CAM plays a pivotal rolein lung cancer progression.Since its discovery in nerve cells 28 years ago, L1CAM

expression has emerged as a contributing factor in theprogression of a variety of human carcinomas (31). A recentstudy revealed that elevated L1CAM levels were associatedwith shorter survival in patients with pancreatic ductaladenocarcinoma (12). In addition, studies on resectedtumor samples from colorectal (32), ovarian (9), andmelanoma (33) cancer patients have linked L1CAM over-expression to poor prognosis. Our previous work identifiedL1CAM as a component of a 15-gene signature that predictsprognosis in NSCLC patients (20). Here we confirm theprognostic value of L1CAM expression alone in 4 indepen-dent published microarray data sets, thus establishing itsrobustness as a prognostic marker in NSCLC patients. The

result provides a rationale to further explore the role ofL1CAM in NSCLC.

Our results show that L1CAMregulates cellmigration andinvasion in vitro in 3 different histologic subtypes of NSCLCcell lines. Ectopic expression of L1CAMhas been reported tomodulate haptotactic cellmigrationonvarious extracellularmatrix (ECM) proteins in melanoma, ovarian, and coloncancer cells (30, 34, 35), and to enhance Matrigel invasionin colon and melanoma cancer cells lacking L1CAM(30, 35). Although L1CAM may play stereotyped roles inmediating neuronal migration in nontransformed cells,diversity in mechanisms underlying tumorigenicity is con-sistent with L1CAM functioning in a tissue- and context-dependent manner during tumor progression. Li and Gali-leo (36) showed that L1CAM augmented cell adhesion andmigration in breast cancer cell lines, but did not promoteMatrigel invasion. In this study, we found that suppressionof L1CAM abrogates the ability of NSCLC cells to close anartificial wound and to invade Matrigel.

The mechanisms underlying L1CAM signaling in cancerbiology remain controversial. AlthoughL1CAMmay inducean epithelial–mesenchymal transition (EMT)-like processin MCF7 breast cancer cells (37), L1CAM-mediated induc-tion of colorectal cancer cell metastasis operates via NF-kBsignaling independent of EMT changes (38). Activation ofthe Erk pathway by L1CAM has been reported previously(39) and is linked to a motile phenotype in fibroblast andmelanoma cells (30). Erk signaling is a major determinantin the control of diverse cellular processes such as prolifer-ation, survival, differentiation, and motility and is oftenupregulated in human cancers (40). Here we show thatH125 and H460 cells with L1CAM downregulation aredeficient in constitutive Erk1/2 activation upon serumstimulation, which is reversed by reintroduction ofshRNA-resistant L1CAM into L1CAM knockdown cells. Inaddition, xenograft tumors developed from L1CAM knock-down cells show drastically reduced levels of pErk1/2 invivo. These results highlight the close association of L1CAMactivity and the Erk pathway in NSCLC cells.

How L1CAM triggers intracellular Erk signaling toinduce a motile and invasive phenotype in lung cancercells is unknown. One study suggests that direct binding ofL1CAM to avb5 integrin potentiates haptotactic migrationin CHO cells via integrin-mediated signaling events (29).

Table 2. L1CAM contributes to metastasis in an orthotopic nude rat model

H460SM T wt (g),

Regional metsno. of animals Systemic mets No. of animals

cell line mean � SD LN L. Lung Bone Kidney Brain Gum Systemic mets Total

Control 7.2 � 3.0 13 13 9 (69) 13 (100) 3 (23) 4 (31) 13 (100) 13shL1CAM 6.4 � 4.1 13 9 3 (23)a 7 (54)a 0 (0) 4 (31) 8 (62)a 13

NOTE: Values in parenthesis are expressed in percentage.L. lung indicates left lung; LN, lymph nodes; mets, metastasis; T, primary tumor; wt, weight.aP < 0.05, by Fisher exact test.

L1CAM in Non–Small Cell Lung Cancer

www.aacrjournals.org Clin Cancer Res; 18(7) April 1, 2012 1921

on August 24, 2021. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst February 3, 2012; DOI: 10.1158/1078-0432.CCR-11-2893

Page 9: L1 Cell Adhesion Molecule Promotes Tumorigenicity and Metastatic Potential … · Human Cancer Biology L1 Cell Adhesion Molecule Promotes Tumorigenicity and Metastatic Potential in

Cheng and colleagues identified RanBPM as an adapterprotein directly linking L1CAM to the Erk pathway inCOS-7 cells (41). Therefore, it is conceivable that inresponse to extracellular stimuli such as serum growthfactor, L1CAM interacts with transmembrane bindingpartners or ECM receptors (e.g., RanBPM, ezrin, and avb3integrins), resulting in activation of the Erk pathway andErk-dependent gene products associated with migratoryand invasive phenotypes (38, 41, 42). Silletti and collea-gues (30) showed that sustained Erk activation by L1CAMinduces Rac1 and cathepsin activity, which directly pro-mote cell migration and invasion of melanoma cells,respectively.

Our data show that L1CAM suppression in H460 andH125 cells reduces tumor growth rate in SCID mice, whichcan be rescued by restoring L1CAM levels. A role for L1CAMin the tumorigenicity of NSCLC cells is consistent withstudies in other cancer cell types, as overexpression ofL1CAM in ovarian (34), colon (43), and pancreatic cancercells (44) augments tumor growth in mice. In addition,biweekly intraperitoneal injection of amonoclonal L1CAMantibody (chCE7) in ovarian carcinoma-bearingmice led tosignificant reduction of tumor burden (45). Similar effectswere recently reported using anti-L1CAM treatment inintrahepatic cholangiocarcinoma-bearing mice (46).

Considering that metastatic spread, not primary tumorburden, is the main cause of lung cancer death, a criticalquestion is whether L1CAM regulates lung cancer metasta-ses. Our laboratory previously reported that endobronchialimplantation of NCI-H460 cells into nude rats generates aprimary lung tumor with mediastinal lymph node spread,but rarely systemic metastases (27). Isolation and orthoto-pic reimplantation of tumor cells from these mediastinalnodes, repeated successively 4 times, produced a variant cellline designatedH460SM that spontaneouslymetastasizes tobone, kidney, brain, soft tissue, and contralateral lung.Orthotopic implantation of lung cancer cells is thought tobetter mimic the metastatic behavior of lung cancers com-paredwith subcutaneousmodels (28). Using thismodel wefound that L1CAM suppression significantly reduces met-astatic spread of H460SM cells to bone and kidney in nuderats. Proclivity of tumor metastasis for specific organs, suchas lung carcinomas for bone, is well documented (47). Ournovel finding that L1CAM mediates lung cancer metastasiscould account for the strong association between L1CAMexpression and poor prognosis of NSCLC patients. Further-more, our results showing the role of L1CAM in the growthand metastasis of NSCLC supports its potential as a ther-apeutic target for NSCLC patients in an adjuvant setting.

Considering the role of L1CAM in normal neural devel-opment, treatment of antibody-based NSCLC therapeuticstrategies at the postnatal stage is unlikely to produce severeneurotoxic effects. L1CAM knockout mice show weak hindlimb function due to abnormal corticospinal axon guidancebut are viable and have no obvious morphologic defects inmajor central and peripheral nervous system structures (17,18). Moreover, tissue-specific alternative exon splicing pro-duces functionally relevant isoforms of L1CAM. Neuronal

L1CAM differs from L1CAM found in nonneuronal cells inthat it contains 4 amino acids (RSLE) in the cytoplasmicdomain encoded by the differentially spliced exon 27 (48).Therefore, isoform-specific antibodies directed at the non-neuronal epitope of L1CAM lacking exon 2 and 27 may bean appropriate approach to inhibiting L1CAM protumori-genic function in lung cancer. A recent study showed thatcombining an L1CAM antibody with chemotherapeuticdrugs such as gemcitabine and paclitaxel augmented theresponse toward chemotherapy alone in preclinical modelsof pancreatic andovarian cancer (49). These findings under-score the potential of treatments that use L1CAMantibodiesalongside conventional NSCLC adjuvant chemotherapydrugs to improve therapeutic response.

Metastasis is the process that transforms lung cancer froma disease that is local and curable to one that is systemic andfatal. Cell migration and invasion are considered initialsteps during metastasis, consistent with our findings thatinterference with L1CAM expression suppresses bothNSCLC metastasis and cell migration and invasion. Wefurther reveal that L1CAM interacts with the Erk signalingpathway to mediate tumor growth, although the details ofthis mechanism remain to be explored. Supporting theseanimal studies is our in silico finding that L1CAMexpressionis consistently an independent prognostic factor inmultiplelarge cohorts of NSCLC patients. Taken together, our dataindicate that L1CAM is a key regulator of lung cancerprogression and a valuable prognostic and therapeutictarget in future lung cancer studies.

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

Authors' ContributionsConception and design: J. Hai, C.-Q. Zhu, R. Navab, M.-S. Tsao.Development of methodology: J. Hai, C.-Q. Zhu, M.-S. Tsao.Analysis and interpretation of data (e.g., statistical analysis, biosta-tistics, and computational analysis): J. Hai, C.-Q. Zhu, B. Bandarchi, R.Navab, F.A. Shepherd, I. Jurisica, M.-S. Tsao.Writing, review, and/or revisionof themanuscript: J.Hai, C.-Q.Zhu, F.A.Shepherd, I. Jurisica, M.-S. Tsao.Administrative, technical, or material support (i.e., reporting or orga-nizing data, constructing databases): J. Hai, C.-Q. Zhu, Y.-H. Wang, I.Jurisica, M.-S. Tsao.Study supervision: C.-Q. Zhu, I. Jurisica, M.-S. Tsao.Funding: M.-S. Tsao.

AcknowledgmentsThe authors thank Dr. Ming Li, Dr. Jiang Liu, and Emin Ibrahimov for

technical assistance and Shawna Organ and Dr. Sandy Der for helpfuldiscussions.

Grant SupportThis study was supported by the Canadian Cancer Society Research

Institute grant #020527, the Ontario Ministry of Health and Long TermCare, and the Princess Margaret Hospital Foundation. M.-S. Tsao holds theM. Qasim Choksi Chair in Lung Cancer Translational Research, F.A. Shep-herd holds the Scott Taylor Chair in Lung Cancer Research, and I. Jurisicaholds a Canada Research Chair in Integrative Cancer Informatics.

The costs of publication of this article were defrayed in part by thepayment of page charges. This article must therefore be hereby markedadvertisement in accordance with 18 U.S.C. Section 1734 solely to indicatethis fact.

Received November 13, 2011; revised January 16, 2012; accepted January29, 2012; published OnlineFirst February 3, 2012.

Hai et al.

Clin Cancer Res; 18(7) April 1, 2012 Clinical Cancer Research1922

on August 24, 2021. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst February 3, 2012; DOI: 10.1158/1078-0432.CCR-11-2893

Page 10: L1 Cell Adhesion Molecule Promotes Tumorigenicity and Metastatic Potential … · Human Cancer Biology L1 Cell Adhesion Molecule Promotes Tumorigenicity and Metastatic Potential in

References1. Ferlay J, Shin HR, Bray F, Forman D, Mathers C, Parkin DM. Estimates

ofworldwide burden of cancer in 2008:GLOBOCAN2008. Int JCancer2010;127:2893–917.

2. Henschke CI, Yankelevitz DF, Libby DM, Pasmantier MW, Smith JP,Miettinen OS. Survival of patients with stage I lung cancer detected onCT screening. N Engl J Med 2006;355:1763–71.

3. Ding L, Getz G, Wheeler DA, Mardis ER, McLellan MD, Cibulskis K,et al. Somatic mutations affect key pathways in lung adenocarcinoma.Nature 2008;455:1069–75.

4. Bass AJ, Watanabe H, Mermel CH, Yu S, Perner S, Verhaak RG, et al.SOX2 is an amplified lineage-survival oncogene in lung and esoph-ageal squamous cell carcinomas. Nat Genet 2009;41:1238–42.

5. John T, Liu G, Tsao MS. Overview of molecular testing in non-small-cell lung cancer: mutational analysis, gene copy number, proteinexpression and other biomarkers of EGFR for the prediction ofresponse to tyrosine kinase inhibitors. Oncogene 2009;28 Suppl 1:S14–23.

6. Aviel-Ronen S, Blackhall FH, Shepherd FA, Tsao MS. K-ras mutationsin non-small-cell lung carcinoma: a review. Clin Lung Cancer2006;8:30–8.

7. Goldstraw P, Crowley J, Chansky K, Giroux DJ, Groome PA, Rami-Porta R, et al. The IASLC Lung Cancer Staging Project: proposals forthe revision of the TNM stage groupings in the forthcoming (seventh)edition of the TNMClassification ofmalignant tumours. J ThoracOncol2007;2:706–14.

8. Cavallaro U, Dejana E. Adhesion molecule signalling: not always asticky business. Nat Rev Mol Cell Biol 2011;12:189–97.

9. FogelM,GutweinP,Mechtersheimer S, Riedle S, StoeckA, SmirnovA,et al. L1 expression as a predictor of progression and survival inpatients with uterine and ovarian carcinomas. Lancet 2003;362:869–75.

10. Meier F, Busch S, Gast D, Goppert A, Altevogt P, Maczey E, et al. Theadhesionmolecule L1 (CD171) promotes melanoma progression. Int JCancer 2006;119:549–55.

11. Schroder C, Schumacher U, FogelM, Feuerhake F,Muller V,Wirtz RM,et al. Expression and prognostic value of L1-CAM in breast cancer.Oncol Rep 2009;22:1109–17.

12. Tsutsumi S, Morohashi S, Kudo Y, Akasaka H, Ogasawara H, Ono M,et al. L1 Cell adhesion molecule (L1CAM) expression at the cancerinvasive front is a novel prognostic marker of pancreatic ductal ade-nocarcinoma. J Surg Oncol 2011;103:669–73.

13. Brabletz T, JungA,DagS,Hlubek F, Kirchner T. Beta-catenin regulatesthe expression of the matrix metalloproteinase-7 in human colorectalcancer. Am J Pathol 1999;155:1033–8.

14. Moos M, Tacke R, Scherer H, Teplow D, Fruh K, Schachner M. Neuraladhesionmolecule L1 as amember of the immunoglobulin superfamilywith binding domains similar to fibronectin. Nature 1988;334:701–3.

15. Grumet M, Edelman GM. Neuron-glia cell adhesion molecule interactswith neurons andastroglia via different bindingmechanisms. JCell Biol1988;106:487–503.

16. Kamiguchi H, Lemmon V. Neural cell adhesion molecule L1: signalingpathways and growth cone motility. J Neurosci Res 1997;49:1–8.

17. Cohen NR, Taylor JS, Scott LB, Guillery RW, Soriano P, Furley AJ.Errors in corticospinal axon guidance in mice lacking the neural celladhesion molecule L1. Curr Biol 1998;8:26–33.

18. Dahme M, Bartsch U, Martini R, Anliker B, Schachner M, Mantei N.Disruption of themouse L1 gene leads tomalformations of the nervoussystem. Nat Genet 1997;17:346–9.

19. Fransen E, D'Hooge R, Van CampG, VerhoyeM, Sijbers J, Reyniers E,et al. L1 knockout mice show dilated ventricles, vermis hypoplasia andimpaired exploration patterns. Hum Mol Genet 1998;7:999–1009.

20. Zhu CQ, Ding K, Strumpf D, Weir BA, Meyerson M, Pennell N, et al.Prognostic and predictive gene signature for adjuvant chemotherapyin resected non-small-cell lung cancer. J Clin Oncol 2010;28:4417–24.

21. Winton T, Livingston R, JohnsonD, Rigas J, JohnstonM, Butts C, et al.Vinorelbine plus cisplatin vs. observation in resected non-small-celllung cancer. N Engl J Med 2005;352:2589–97.

22. Shedden K, Taylor JM, Enkemann SA, Tsao MS, Yeatman TJ, GeraldWL, et al. Gene expression-based survival prediction in lung adeno-

carcinoma: a multi-site, blinded validation study. Nat Med 2008;14:822–7.

23. Raponi M, Zhang Y, Yu J, Chen G, Lee G, Taylor JM, et al. Geneexpression signatures for predicting prognosis of squamous cell andadenocarcinomas of the lung. Cancer Res 2006;66:7466–72.

24. Roepman P, Jassem J, Smit EF, Muley T, Niklinski J, van de Velde T,et al. An immune response enriched 72-gene prognostic profile forearly-stage non-small-cell lung cancer. Clin Cancer Res 2009;15:284–90.

25. Lau SK, Boutros PC, Pintilie M, Blackhall FH, Zhu CQ, Strumpf D, et al.Three-gene prognostic classifier for early-stage non small-cell lungcancer. J Clin Oncol 2007;25:5562–9.

26. Radulovich N, Leung L, TsaoMS.Modified gateway system for doubleshRNA expression andCre/lox based gene expression. BMCBiotech-nol 2011;11:24.

27. Liu J, Blackhall F, Seiden-Long I, Jurisica I, Navab R, Liu N, et al.Modeling of lung cancer by an orthotopically growing H460SM variantcell line reveals novel candidate genes for systemic metastasis. Onco-gene 2004;23:6316–24.

28. Howard RB, Mullen JB, Pagura ME, Johnston MR. Characterization ofa highly metastatic, orthotopic lung cancer model in the nude rat. ClinExp Metastasis 1999;17:157–62.

29. Mechtersheimer S, Gutwein P, Agmon-Levin N, Stoeck A, OleszewskiM, Riedle S, et al. Ectodomain shedding of L1 adhesion moleculepromotes cell migration by autocrine binding to integrins. J Cell Biol2001;155:661–73.

30. Silletti S, Yebra M, Perez B, Cirulli V, McMahon M, Montgomery AM.Extracellular signal-regulated kinase (ERK)-dependent gene expres-sion contributes to L1 cell adhesion molecule-dependent motility andinvasion. J Biol Chem 2004;279:28880–8.

31. Rathjen FG, Schachner M. Immunocytological and biochemical char-acterization of a new neuronal cell surface component (L1 antigen)which is involved in cell adhesion. EMBO J 1984;3:1–10.

32. BooYJ, Park JM,KimJ,ChaeYS,MinBW,UmJW, et al. L1 expressionas a marker for poor prognosis, tumor progression, and short survivalin patients with colorectal cancer. Ann Surg Oncol 2007;14:1703–11.

33. Thies A, Schachner M, Moll I, Berger J, Schulze HJ, Brunner G, et al.Overexpression of the cell adhesion molecule L1 is associated withmetastasis in cutaneous malignant melanoma. Eur J Cancer 2002;38:1708–16.

34. Gast D, Riedle S, Schabath H, Schlich S, Schneider A, Issa Y, et al. L1augments cell migration and tumor growth but not beta3 integrinexpression in ovarian carcinomas. Int J Cancer 2005;115:658–65.

35. Gavert N, Conacci-SorrellM,Gast D, Schneider A, Altevogt P, BrabletzT, et al. L1, a novel target of beta-catenin signaling, transforms cellsand is expressed at the invasive front of colon cancers. J Cell Biol2005;168:633–42.

36. Li Y, Galileo DS. Soluble L1CAMpromotes breast cancer cell adhesionand migration in vitro, but not invasion. Cancer Cell Int 2010;10:34.

37. Shtutman M, Levina E, Ohouo P, Baig M, Roninson IB. Cell adhesionmolecule L1 disrupts E-cadherin-containing adherens junctions andincreases scattering and motility of MCF7 breast carcinoma cells.Cancer Res 2006;66:11370–80.

38. Gavert N, Ben-Shmuel A, Lemmon V, Brabletz T, Ben-Ze'ev A. Nuclearfactor-kappaB signaling and ezrin are essential for L1-mediatedmetastasis of colon cancer cells. J Cell Sci 2010;123:2135–43.

39. Schaefer AW, Kamiguchi H, Wong EV, Beach CM, Landreth G, Lem-mon V. Activation of the MAPK signal cascade by the neural celladhesion molecule L1 requires L1 internalization. J Biol Chem1999;274:37965–73.

40. Kohno M, Pouyssegur J. Targeting the ERK signaling pathway incancer therapy. Ann Med 2006;38:200–11.

41. Cheng L, Lemmon S, Lemmon V. RanBPM is an L1-interacting proteinthat regulates L1-mediated mitogen-activated protein kinase activa-tion. J Neurochem 2005;94:1102–10.

42. EbelingO,DuczmalA, AignerS,GeigerC,SchollhammerS,KemsheadJT, et al. L1 adhesion molecule on human lymphocytes and mono-cytes: expression and involvement in binding to alpha v beta 3 integrin.Eur J Immunol 1996;26:2508–16.

L1CAM in Non–Small Cell Lung Cancer

www.aacrjournals.org Clin Cancer Res; 18(7) April 1, 2012 1923

on August 24, 2021. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst February 3, 2012; DOI: 10.1158/1078-0432.CCR-11-2893

Page 11: L1 Cell Adhesion Molecule Promotes Tumorigenicity and Metastatic Potential … · Human Cancer Biology L1 Cell Adhesion Molecule Promotes Tumorigenicity and Metastatic Potential in

43. Gast D, Riedle S, Issa Y, Pfeifer M, Beckhove P, Sanderson MP, et al.The cytoplasmic part of L1-CAM controls growth and gene expressionin human tumors that is reversed by therapeutic antibodies. Oncogene2008;27:1281–9.

44. Kiefel H, Bondong S, Erbe-Hoffmann N, Hazin J, Riedle S, Wolf J, et al.L1CAM-integrin interaction induces constitutive NF-kappaB activa-tion in pancreatic adenocarcinoma cells by enhancing IL-1betaexpression. Oncogene 2010;29:4766–78.

45. Arlt MJ, Novak-Hofer I, Gast D, Gschwend V, Moldenhauer G, Grun-berg J, et al. Efficient inhibition of intra-peritoneal tumor growth anddissemination of human ovarian carcinoma cells in nude mice by anti-L1-cell adhesion molecule monoclonal antibody treatment. CancerRes 2006;66:936–43.

46. Min JK, Kim JM, Li S, Lee JW, Yoon H, Ryu CJ, et al. L1 cell adhesionmolecule is a novel therapeutic target in intrahepatic cholangiocarci-noma. Clin Cancer Res 2010;16:3571–80.

47. Coleman RE. Skeletal complications of malignancy. Cancer 1997;80:1588–94.

48. MiuraM,KobayashiM,AsouH,UyemuraK.Molecular cloningof cDNAencoding the rat neural cell adhesion molecule L1. Two L1 isoforms inthe cytoplasmic region are produced by differential splicing. FEBSLett1991;289:91–5.

49. Schafer H, Dieckmann C, Korniienko O, Moldenhauer G, Kiefel H,Salnikov A, et al. Combined treatment of L1CAM antibodies andcytostatic drugs improve the therapeutic response of pancreatic andovarian carcinoma. Cancer Lett 2011 Dec 27 [Epub ahead of print].

Hai et al.

Clin Cancer Res; 18(7) April 1, 2012 Clinical Cancer Research1924

on August 24, 2021. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst February 3, 2012; DOI: 10.1158/1078-0432.CCR-11-2893

Page 12: L1 Cell Adhesion Molecule Promotes Tumorigenicity and Metastatic Potential … · Human Cancer Biology L1 Cell Adhesion Molecule Promotes Tumorigenicity and Metastatic Potential in

2012;18:1914-1924. Published OnlineFirst February 3, 2012.Clin Cancer Res   Josephine Hai, Chang-Qi Zhu, Bizhan Bandarchi, et al.  

Small Cell Lung Cancer−Potential in NonL1 Cell Adhesion Molecule Promotes Tumorigenicity and Metastatic

  Updated version

  10.1158/1078-0432.CCR-11-2893doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://clincancerres.aacrjournals.org/content/suppl/2012/02/03/1078-0432.CCR-11-2893.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://clincancerres.aacrjournals.org/content/18/7/1914.full#ref-list-1

This article cites 48 articles, 13 of which you can access for free at:

  Citing articles

  http://clincancerres.aacrjournals.org/content/18/7/1914.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/18/7/1914To request permission to re-use all or part of this article, use this link

on August 24, 2021. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst February 3, 2012; DOI: 10.1158/1078-0432.CCR-11-2893


Recommended