rab37 hypermethylation regulates metastasis and resistance to docetaxel … · confirmed that...

15
Translational Cancer Mechanisms and Therapy RAB37 Hypermethylation Regulates Metastasis and Resistance to Docetaxel-Based Induction Chemotherapy in Nasopharyngeal Carcinoma Yingqin Li, Xiaojing Yang, Xiaojing Du,Yuan Lei,Qingmei He, Xiaohong Hong, Xinran Tang, Xin Wen, Panpan Zhang,Ying Sun, Jian Zhang,Yaqin Wang, Jun Ma, and Na Liu Abstract Purpose: Epigenetic alterations play important roles in metastasis and drug resistance through gene regulation. How- ever, the functional features and molecular mechanisms of epigenetic changes remain largely unclear in nasopharyngeal carcinoma (NPC) metastasis. Experimental Design: Gene regulatory network analysis was used to identify metastatic-specic dysregulated genes between normal and NPC tissues and the expression was validated in published Gene-Expression Omnibus data set. The regulatory and functional role of RAB37 downregulation was examined in NPC and was validated in vitro and in vivo, and downstream target of RAB37 was explored. The clinical value of RAB37 methylation was evaluated in NPC metastasis and chemosensitivity. Results: We identied RAB37 as a specic hypermethy- lated gene that is most commonly downregulated in NPC. Moreover, RAB37 downregulation was attributed to hyper- methylation of its promoter and was signicantly associ- ated with metastasis- and docetaxel chemoresistance-relat- ed features in NPC. Ectopic RAB37 overexpression sup- pressed NPC cell metastasis and enhanced chemosensitiv- ity to docetaxel. Mechanistically, RAB37 colocalized with TIMP2, regulated TIMP2 secretion, inhibited downstream MMP2 activity, and consequently altered NPC cell metas- tasis. Furthermore, RAB37 hypermethylation was correlat- ed with poor clinical outcomes in patients with NPC. We developed a prognostic model based on RAB37 methyla- tion and N stage that effectively predicted an increased risk of distant metastasis and a favorable response to doce- taxel-containing induction chemotherapy (IC) in NPC patients. Conclusions: This study shows that RAB37 hypermethyla- tion is involved in NPC metastasis and chemoresistance, and that our prognostic model can identify patients who are at a high risk of distant metastasis and might benet from for docetaxel IC. Clin Cancer Res; 24(24); 6495508. Ó2018 AACR. Introduction Nasopharyngeal carcinoma (NPC), one of the common head and neck cancers in Southeast Asia, especially southern China, is characterized by high invasion and early metastasis (1). Due to the obscure symptoms and aggressiveness of NPC, more than 70% of patients with NPC are diagnosed with locoregionally advanced disease at the initial visit and often have poor prognoses (2, 3). According to NCCN guidelines, radiotherapy and chemotherapy are the primary treatment modalities for locoregionally advanced NPC (LA-NPC) and result in substantial improvement in locor- egional control (4, 5). However, the increased likelihood of systemic metastasis remains the major cause of treatment failure and cancer-related mortality for these patients. Accumulating studies report that induction chemotherapy (IC) might improve metastasis control in patients at a high risk of distant metastasis (68). Moreover, our phase III trial conrmed that the addition of docetaxel, cisplatin, and uorouracil (TPF) IC to concurrent chemoradiotherapy (CCRT) can signicantly improve the 3-year distant metastasis-free survival (DMFS) for LA-NPC patients (6). However, this treatment strategy only results in 7% improvement in metastasis control. This survival difference suggests the presence of unknown heterogeneity among NPC patients and highlights the urgent need for an effective strategy to identify patients at a high risk of metastasis who will benet from IC. Currently, the anatomic tumornodemetastasis (TNM) staging system is the major indicator to assess prognosis and guide therapeutic decision, but this system is not very effective (4, 9). Therefore, exploration of the molecular mechanisms and identi- cation of new biomarkers reecting tumor heterogeneity can help to provide innovative strategies to stratify precise risk and guide individual treatment for metastasis in patients with NPC. Epigenetic alterations have been shown to profoundly inu- ence the maintenance and evolution of cancer (10). DNA meth- ylation is a ubiquitous and dynamic epigenetic alteration critical for cellular development, function, and gene-expression regula- tion (11). A substantial number of studies have reported widely aberrant DNA methylation patterns in various tumor types (12). Altered DNA methylation patterns may be relevant for the Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangz- hou, Guangdong, China. Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/). Y. Li, X. Yang, X. Du, and Y. Lei contributed equally to this article. Corresponding Author: Na Liu, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, District Yuexiu, Guangzhou, Guangdong 510060, China. Phone: 86-20-873432370; Fax: 86-20-87343295; E-mail: [email protected] doi: 10.1158/1078-0432.CCR-18-0532 Ó2018 American Association for Cancer Research. Clinical Cancer Research www.aacrjournals.org 6495 on May 19, 2021. © 2018 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from Published OnlineFirst August 21, 2018; DOI: 10.1158/1078-0432.CCR-18-0532

Upload: others

Post on 19-Jan-2021

4 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: RAB37 Hypermethylation Regulates Metastasis and Resistance to Docetaxel … · confirmed that docetaxel, cisplatin, and fluorouracil (TPF) induction chemotherapy (IC) significantly

Translational Cancer Mechanisms and Therapy

RAB37 Hypermethylation Regulates Metastasisand Resistance to Docetaxel-Based InductionChemotherapy in Nasopharyngeal CarcinomaYingqin Li, Xiaojing Yang, XiaojingDu,Yuan Lei,Qingmei He, XiaohongHong, Xinran Tang,Xin Wen, Panpan Zhang, Ying Sun, Jian Zhang, Yaqin Wang, Jun Ma, and Na Liu

Abstract

Purpose: Epigenetic alterations play important roles inmetastasis and drug resistance through gene regulation. How-ever, the functional features and molecular mechanisms ofepigenetic changes remain largely unclear in nasopharyngealcarcinoma (NPC) metastasis.

Experimental Design: Gene regulatory network analysiswas used to identify metastatic-specific dysregulated genesbetween normal and NPC tissues and the expression wasvalidated in published Gene-Expression Omnibus data set.The regulatory and functional role of RAB37 downregulationwas examined in NPC and was validated in vitro and in vivo,and downstream target of RAB37 was explored. The clinicalvalue of RAB37methylation was evaluated in NPC metastasisand chemosensitivity.

Results: We identified RAB37 as a specific hypermethy-lated gene that is most commonly downregulated in NPC.Moreover, RAB37 downregulation was attributed to hyper-methylation of its promoter and was significantly associ-

ated with metastasis- and docetaxel chemoresistance-relat-ed features in NPC. Ectopic RAB37 overexpression sup-pressed NPC cell metastasis and enhanced chemosensitiv-ity to docetaxel. Mechanistically, RAB37 colocalized withTIMP2, regulated TIMP2 secretion, inhibited downstreamMMP2 activity, and consequently altered NPC cell metas-tasis. Furthermore, RAB37 hypermethylation was correlat-ed with poor clinical outcomes in patients with NPC. Wedeveloped a prognostic model based on RAB37 methyla-tion and N stage that effectively predicted an increased riskof distant metastasis and a favorable response to doce-taxel-containing induction chemotherapy (IC) in NPCpatients.

Conclusions: This study shows that RAB37 hypermethyla-tion is involved in NPC metastasis and chemoresistance, andthat our prognostic model can identify patients who are at ahigh risk of distant metastasis and might benefit from fordocetaxel IC. Clin Cancer Res; 24(24); 6495–508. �2018 AACR.

IntroductionNasopharyngeal carcinoma (NPC), one of the common head

and neck cancers in Southeast Asia, especially southern China, ischaracterized byhigh invasion and earlymetastasis (1).Due to theobscure symptoms and aggressiveness of NPC, more than 70% ofpatients with NPC are diagnosed with locoregionally advanceddisease at the initial visit and often have poor prognoses (2, 3).According to NCCN guidelines, radiotherapy and chemotherapyare the primary treatment modalities for locoregionally advancedNPC (LA-NPC) and result in substantial improvement in locor-egional control (4, 5). However, the increased likelihood of

systemic metastasis remains the major cause of treatment failureand cancer-related mortality for these patients.

Accumulating studies report that induction chemotherapy (IC)might improve metastasis control in patients at a high risk ofdistant metastasis (6–8). Moreover, our phase III trial confirmedthat the addition of docetaxel, cisplatin, and fluorouracil (TPF) ICto concurrent chemoradiotherapy (CCRT) can significantlyimprove the 3-year distant metastasis-free survival (DMFS) forLA-NPCpatients (6).However, this treatment strategy only resultsin 7% improvement inmetastasis control. This survival differencesuggests the presence of unknown heterogeneity among NPCpatients and highlights the urgent need for an effective strategyto identify patients at a high risk of metastasis who will benefitfrom IC. Currently, the anatomic tumor–node–metastasis (TNM)staging system is themajor indicator to assess prognosis and guidetherapeutic decision, but this system is not very effective (4, 9).Therefore, exploration of the molecular mechanisms and identi-fication of new biomarkers reflecting tumor heterogeneity canhelp to provide innovative strategies to stratify precise risk andguide individual treatment for metastasis in patients with NPC.

Epigenetic alterations have been shown to profoundly influ-ence the maintenance and evolution of cancer (10). DNA meth-ylation is a ubiquitous and dynamic epigenetic alteration criticalfor cellular development, function, and gene-expression regula-tion (11). A substantial number of studies have reported widelyaberrant DNA methylation patterns in various tumor types (12).Altered DNA methylation patterns may be relevant for the

Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology inSouth China, Collaborative Innovation Center of Cancer Medicine, GuangdongKey Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangz-hou, Guangdong, China.

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

Y. Li, X. Yang, X. Du, and Y. Lei contributed equally to this article.

Corresponding Author: Na Liu, Sun Yat-sen University Cancer Center, 651Dongfeng Road East, District Yuexiu, Guangzhou, Guangdong 510060, China.Phone: 86-20-873432370; Fax: 86-20-87343295; E-mail: [email protected]

doi: 10.1158/1078-0432.CCR-18-0532

�2018 American Association for Cancer Research.

ClinicalCancerResearch

www.aacrjournals.org 6495

on May 19, 2021. © 2018 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 21, 2018; DOI: 10.1158/1078-0432.CCR-18-0532

Page 2: RAB37 Hypermethylation Regulates Metastasis and Resistance to Docetaxel … · confirmed that docetaxel, cisplatin, and fluorouracil (TPF) induction chemotherapy (IC) significantly

dysregulation of specific genes or signaling pathways, leading totumor initiation, progression, and treatment resistance (13).Thus, there is a strong rationale for evaluating the interactionand enrichment of dysmethylated genes involved in identifiedfeatures and signaling pathways. Although several abnormallymethylated genes have been identified in NPC (14–16), theirinteraction and contribution to metastasis and chemoresistanceremain elusive.

Here, using gene regulatory network analysis, we identifyRAB37, a member of RAS superfamily, as a specific hyper-methylated gene that is consistently downregulated in NPC.RAB37 hypermethylation contributes to its decreased expres-sion and is relevant to metastasis- and chemoresistance-relatedproperties in NPC. Restored RAB37 expression inhibits metas-tasis through the tissue inhibitor of the metalloproteinase 2(TIMP2)/matrix metalloproteinase 2 (MMP2) axis andenhances sensitivity to docetaxel in NPC cells. Moreover,RAB37 hypermethylation is correlated with unfavorable sur-vival and, when combined with advanced N stage, can effec-tively identify patients who are at a high risk for distantdissemination and are likely to benefit from docetaxel-basedIC in NPC. Therefore, this study highlights the oncogenic roleof RAB37 hypermethylation and provides innovative strategiesto identify patients at a high risk for metastasis who will benefitfrom docetaxel IC, thereby providing novel perspectives for theprecise treatment of NPC.

Materials and MethodsMethylation microarray analysis

Our previous Infinium Human Methylation 450 K BeadChipmicroarray data were reanalyzed to identify gene interactionnetworks between NPC samples and normal nasopharyngealepithelial tissues (GSE52068). We used GenomeStudio Methyl-ation Module V.1.9.0 to extract image intensities and the algo-rithm in "methylumi" and "lumi" packages in R Bioconductor fornormalization and background correction. TheDNAmethylationlevels for per-probe loci were determined using b-values of

normalized signal intensities. Using the "IMA" Bioconductorpackage, we filtered significantly different methylation loci withthe criteria of single-nucleotide polymorphism probes, P � 0.01in 12 of 48 samples and probes located in the X and Y chromo-somes. Finally, 473,788 CpG sites were subjected to furtheranalysis. The nonparametric Wilcoxon rank-sum test with falsediscovery rate (FDR) adjustment was used to identify significantlydifferent DNA methylation loci.

"GRENITS" package from R Bioconductor was used tocluster differentially dysmethylated loci into causal interac-tion networks from methylation microarray data in groups ofNPC and normal tissues with fully Bayesian spline autore-gression (17). Differential loci with interaction network prob-ability >0.99 between groups were filtered for further func-tion-associated network analysis using Cytoscape software.The final differential loci involved in functional network wereclustered and visualized by heat map analyses (P < 0.05 andDb � |0.2|).

Clinical specimensFor RAB37 promoter methylation level analysis, we ob-

tained 8 freshly frozen NPC specimens and 8 normal naso-pharyngeal epithelial specimens from Sun Yat-sen UniversityCancer Center (Guangzhou, China). For methylation leveland survival analysis, we collected a total of 110 paraffin-embedded NPC specimens from Sun Yat-sen University CancerCenter between March 2004 and April 2007. None of thepatients had received radiotherapy or chemotherapy beforebiopsy, and all patients were diagnosed with nonmetastaticNPC. The 7th edition of the AJCC Cancer Staging Manualwas used to reclassify TNM stages (18). The median follow-upperiod was 102.95 months (range, 6.9–133.0 months). Thepathologic types were all WHO III. All patients received radio-therapy, and patients with stage III–IV NPC received IC plusconcurrent platinum-based chemotherapy or concurrent che-motherapy alone. The detailed clinicopathologic characteris-tics are shown in Supplementary Table S3. This study has beenapproved by the Institutional Ethical Review Boards of the SunYat-sen University Cancer Center and conducted in accordancewith the Declaration of Helsinki. Written informed consentsobtained from all patients.

Propensity score matching analysis was used to adjust forpatient selection bias and create two clinicopathologic matchedcohorts with or without docetaxel IC. The propensity score wascalculated with host and tumor factors, including age, sex, T stage,N stage, WHOpathologic type, Epstein–Barr virus antigen immu-noglobulins (VCA-IgA and EA-IgA) and radiotherapy methodsusing the caliper algorithm with 0.05.

DNA extraction and bisulfite pyrosequencing analysisGenomic DNA was extracted from all tissues and cells using an

AllPrep RNA/DNAMini Kit (Qiagen), QIAamp DNA FFPE TissueKit (Qiagen), or EZ1 DNA tissue Kit (Qiagen), respectively.Bisulfite modification of DNA was conducted using an EpiTectBisulfite Kit (Qiagen). The bisulfite pyrosequencing primers weredesigned with PyroMark Assay Design Software 2.0 (Qiagen) andlisted as follow: Forward: 50-TTAGGGGTAAGTAAGAGATTA-TAGGGG-30, Reverse: 50-AATCTACCTAAAAAAACCAACAACC-TCC-30, and sequencing: 50-GGTTTAGGGGTTGGAT-30. The Pyro-Mark Q96 ID System and software (Qiagen) were applied toquantify the methylation level. The percentage of individual CpG

Translational Relevance

Metastasis is the major reason of treatment failure in naso-pharyngeal carcinoma (NPC). Our phase III clinical trialconfirmed that docetaxel, cisplatin, and fluorouracil (TPF)induction chemotherapy (IC) significantly improved the dis-tantmetastasis control inNPC.However, this pattern providesonly results in 7% improvement in metastasis control, whichpromotes the urgent need to develop a better patient stratifi-cation to efficiently identify patients at high risk of metastasisfor IC. Here, we identified RAB37 as a specific hypermethylat-ed gene that is consistently downregulated, contributing tometastasis and docetaxel-based chemoresistance in NPC. Inpatients with NPC, RAB37 hypermethylation was correlatedwith poor clinical outcomes. We developed a prognosticmodel based on RAB37 methylation and N stage that effec-tively predicted an increased risk of distant metastasis and afavorable response to docetaxel-contained IC in patients withNPC, thereby facilitating the development of novel predictiveand therapeutic strategies against NPC metastasis.

Li et al.

Clin Cancer Res; 24(24) December 15, 2018 Clinical Cancer Research6496

on May 19, 2021. © 2018 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 21, 2018; DOI: 10.1158/1078-0432.CCR-18-0532

Page 3: RAB37 Hypermethylation Regulates Metastasis and Resistance to Docetaxel … · confirmed that docetaxel, cisplatin, and fluorouracil (TPF) induction chemotherapy (IC) significantly

methylation was calculated by average methylation level. Formethylation and survival analysis, cutoff methylation value with5.8% was determined by receiver-operating characteristic (ROC)curve analysis (19).

Cell cultureNP69, a human immortalized nasopharyngeal epithelial cell

line, was maintained in Keratinocyte/serum-free medium (Invi-trogen) supplemented with bovine pituitary extract (BD Bios-ciences). Human NPC cell lines (CNE2, CNE1, SUNE1, HONE1,HNE1, and C666-1) were cultured in RPMI 1640 (Invitrogen)supplemented with 10% fetal bovine serum (FBS, Gibco). NP69and all NPC cell lines, which had been authenticated, werebountifully provided by Dr. Mu-sheng Zeng (Sun Yat-sen Uni-versity Cancer Center). 293FT cells obtained from ATCC weregrown in DMEM (Invitrogen) with 10% FBS. All the cells wereauthenticated using short-tandem repeat profiling, tested formycoplasma contamination, and cultured for less than 2 months.

RNA extraction and real-time PCRTotal RNA from cultured cells and clinical samples were iso-

lated with TRIzol reagent (Invitrogen). Total RNA was reversetranscribed using random primers and M-MLV reverse transcrip-tase (Promega). SYBR Green–based (Invitrogen) real-time PCRanalysis was carried out in a CFX96 Touch sequence detectionsystem (Bio-Rad). Real-time PCR primers for RAB37 and TIMP2were as follows: Forward: 50-TGCTGCTAGGCAACAAGG-30,Reverse: 50-GCCAGAAAGGCTAACTCCA-30; Forward: 50- AGATG-TAGTGATCAGGGCCA-30, Reverse: 50-AGGCCCTTTGAACAT-CTTTATCTG-30. GAPDH was regarded as an endogenous controlfor all the genes. The 2�DDCT equation was utilized to calculate therelative gene expression (20).

Western blotting assayTotal protein was obtained using RIPA buffer (Beyotime Bio-

technology) containing EDTA-free Protease Inhibitor Cocktail(Roche). Protein extracts were separated via 10%–12% SDS–PAGE and transferred to polyvinylidene fluoride membranes(Merck Millipore). The membranes were blocked in 5% nonfatmilk and incubated with primary antibodies targeting RAB37(1:400; Proteintech, 13051-1-AP), TIMP2 (1:600; Proteintech,17353-1-AP), a-tubulin (1:500; Sigma-Aldrich, T3526), andGAPDH (1: 5,000; Abcam, ab70699) overnight at 4�C. Secondaryantibody incubation was performed using horseradish peroxi-dase-conjugated antibodies (anti-mouse or anti-rabbit; 1:2,000;Proteintech) at room temperature. Finally, the target proteinbands were detected using an enhanced chemiluminescencesystem (Thermo Fisher Scientific).

Gene-expression profiling and gene-expression profilinginteractive analysis (GEPIA)

Gene-expression profiles of NPC were derived from Gene-Expression Omnibus (GEO; http://www.ncbi.nlm.nih.gov/geo/)under the series accession numbers GSE12452, GSE53819, andGSE64634. GEPIA (21) was conducted to examine the expressionlevel ofRAB37 indifferent cancer types and corresponding normaltissues (http://gepia.cancer-pku.cn/index.html) based on TheCancer Genome Atlas (TCGA; ref. 22) and GTEx data (23, 24).Comparison was performed with Student exact t tests to identifydifferentially expressed genes between cancer and normal groupswith P value less than 0.05.

Gene set enrichment analysis (GSEA) and STRING analysisGene-expression profiles of 31 NPC specimens (GSE12452)

were used to conduct GSEA to identify gene signatures betweengroups with high and low RAB37 expression. We used theMolecular Signatures Database (MSigDB) C2 collection ofchemical and genetic perturbations (n ¼ 3,409 gene sets) andthe GSEA algorithm (http://software.broadinstitute.org/gsea/msigdb; ref. 25) to rank genes with expression levels correlatedwith RAB37 expression levels across NPC samples. GSEA resultsare shown using normalized enrichment scores, accounting forthe size anddegree towhich a gene set is overrepresented at the topor bottom of the ranked list of genes (P < 0.05 and FDR � 0.25).An enrichment heat map and a STRING network (https://string-db.org/) were used for visualization of the GSEA results with thetop 100 ranked genes (50with positive score and 50with negativescore) in RAB37-low vs. RAB37-high groups.

Oligonucleotides, plasmids, and stable cell linesEffective siRNA oligonucleotides targeting RAB37 and TIMP2

were obtained fromGenePharma. The sequences were as follows:siRAB37-1, 50-CCATGCTTATTACAGAGAT-30; siRAB37-2, 50-GCGTCACCCATGCTTATTA-30; siTIMP2-1, 50-GAGATCAAGCA-GATAAAGA-30; and siTIMP2-2, 50-GGAAAGAAGGAATATCTCA-30. The pSin-EF2-puro-vector and TIMP2 plasmids were pur-chased from Vigene Biosciences. The sequence of the humanRAB37 gene was synthesized and cloned into the lentiviral plas-mid pSin-EF2-puromycin (Addgene). The primers used foramplification were as follows: RAB37-Forward, 50-CGGGATC-CATGACGGGCACGCCAGGCGCCGTT-30, RAB37-Reverse1, 50-TGGGACGTCGTATGGGTACATGAAGGAGCAGCAGCT-30 andRAB37-Reverse2, 50-GGAATTCTCAAGCGTAGTCTGGGACGTC-GTATG-30. Lentivirus packaging expression plasmids werecotransfected into 293FT cells to generate stably transfected celllines. The lentiviral particles were subsequently harvested andinfected into NPC cells 48 hours later. Stable clones were thenselected using 0.5 mg/mL puromycin (Sigma-Aldrich), and real-time RT-PCR or Western blotting assays were used to validatethe infection efficiency.

Wound-healing, Transwell migration, and invasion assaysFor wound-healing assay, transfected cells were seeded in 6-

well plates and starved for 24 hours. Confluent monolayers werescratched using a 10-mL tip, and images were captured at 0 hourand 36 hours. For migration and invasion assays, 5 � 104 or 1 �105 transfected cells were resuspended in serum-freemedium andplated in the upper transwell chamber (Corning) with 8-mmporesize membrane with or without Matrigel (BD Biosciences). Then,medium supplemented with 10% FBS was placed in the lowerchamber. After 12 or 24 hours of incubation, the cells thatmigrated or invaded through the upper membrane were fixed,stained with hematoxylin, and counted using an invertedmicroscope.

Docetaxel treatmentFor chemotherapy, 5 � 104 SUNE1 and HONE1 cell lines

stably expressing RAB37 or vector were seeded in 96-wellplates. The next day, the cells were treated with or without doc-etaxel for 48 hours at the dose of 0, 2.5, 5, 10, 20, and 40 mg/mL.Cell viability was measured, and the concentration of 50%inhibition of cell growth (IC50) was calculated using SPSS22.0 software (IBM).

Epigenetic Silencing of RAB37 Promotes NPC Metastasis

www.aacrjournals.org Clin Cancer Res; 24(24) December 15, 2018 6497

on May 19, 2021. © 2018 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 21, 2018; DOI: 10.1158/1078-0432.CCR-18-0532

Page 4: RAB37 Hypermethylation Regulates Metastasis and Resistance to Docetaxel … · confirmed that docetaxel, cisplatin, and fluorouracil (TPF) induction chemotherapy (IC) significantly

Mass spectrometry and coimmunoprecipitation (co-IP) assayFor immunoprecipitation (IP) assays, SUNE1 or HONE1 cells

were lysed with IP lysis buffer. Primary anti-HA (2 mg; Sigma-Aldrich, H6908) or anti-IgG (negative control, 3 mg; Proteintech,30000-0-AP) antibodies were incubated with the lysates over-night at 4�C. Protein A/G Sepharose beads (Thermo Scientific)were used to recover the immune complexes, which were thenwashed and collected after being isolated. Mass spectrometry wasperformed by Huijun Biotechnology. For co-IP assay, Westernblotting was performed to determine protein levels. Cells weretrypsinized and rinsed in PBS and then lysed with RIPA lysisbuffer. The protein extracts were subsequently incubated usinganti-RAB37 and anti-TIMP2 antibodies, followed by precipitationwith protein A agarose (Invitrogen) and immobilized on proteinG Sepharose beads. The precipitated proteins were separated anddetected by Western blotting using rabbit antibodies. Finally, theblots were visualized using a chemiluminescence system.

Immunofluorescence stainingFor immunofluorescence staining assays, cells were grown on

coverslips (Thermo Fisher Scientific). After 24 hours, cells werefixed with 2% paraformaldehyde, permeabilized in 0.5% TritonX-100 inPBS, and incubatedwithprimary anti-E-cadherin (1:100;BD Biosciences, 610181), anti-Vimentin (1:100; Proteintech,10366-1-AP), anti-RAB37 (1: 200; Proteintech, 13051-1-AP), andanti-TIMP2 (1: 200; Proteintech, 17353-1-AP) antibodies over-night at 4�C. The coverslips were incubated with Alexa Fluor 488or 594 goat IgG secondary antibody (1:1,000; Life Technologies;A-11008 or A-11001) and counterstained with 40,6-diamidino-2-phenylindole (DAPI). Images were captured using a confocallaser-scanning microscope (Olympus FV1000).

Gelatin zymography and ELISA assayThe level of MMP2 in the medium was tested using gelatin

zymography assays. Briefly, conditioned medium (CM) was col-lected and analyzed using 10% SDS–PAGE containing 0.1%gelatin. After electrophoresis, the gelwaswashedwith 2.5%TritonX-100 at roomtemperature for 1hour to remove SDSand renatureMMP2 and then incubated at 37�C overnight in the developingbuffer. The gel was subsequently stained with 0.25% CoomassieBlue R-250 for 2 hours and destained in 50%methanol and 10%glacial acetic acid to reveal zones with gelatinase activity. Clearbands indicate proteolytic activity. ELISAwas performedusing theTIMP2 ELISA Kit according to the manufacturer's instructions(JianglaiBio).

In vivo lung metastasis modelFemale BALB/c nude mice (4–6 weeks old, 15–18 g) were

purchased from Beijing Vital River Laboratory Co., Ltd. For thelung metastasis model, SUNE1 cells that stably overexpressedthe vector or RAB37 were resuspended in PBS, and 2 � 105 cellswere injected into the tail veins of mice (n ¼ 8 per group). Eightweeks later, the mice were sacrificed, and lung metastaticcolonies were quantified. For the lung metastasis model fol-lowing docetaxel treatment, HONE1 cells with or withoutRAB37 overexpression (2 � 105 cells in 100-mL PBS) wereinjected into the tail veins of mice. Two weeks later, 100-mLsaline or docetaxel at a dose of 2 mg/g were delivered viaintraperitoneal injection, once per week for 3 weeks. Threeweeks later, the mice were sacrificed for further analysis. Allanimal experiments were performed according to protocols

provided by the Institutional Animal Care and Use Committeeof Sun Yat-sen University Cancer Center (approval number:L102012017002J). The experimental methods complied withthe Declaration of Helsinki.

IHC stainingIHC was performed to detect the expression levels of RAB37

and TIMP2 on paraffin-embedded sections prepared from in vivoexperiments. Briefly, the sections were deparaffinized, rehydrated,subjected to endogenous peroxidase activity blocking, antigenretrieval, nonspecific binding blocking, and incubated with pri-mary antibodies (RAB37, 1:200, Proteintech, 13051-1-AP;TIMP2, 1:200, Proteintech, 17353-1-AP) at 4�C overnight. Allsections were scored and validated by the two experiencedpathologists.

Statistical analysisAll data are presented as the mean � SD from at least three

independent experiments. Two-tailed Student t tests were used forcomparisons between groups. Comparisons among categoricalvariables were performed with c2 and Fisher exact tests. TheKaplan–Meier method and log-rank test were, respectively, usedto construct survival curves and compare the differences. Inde-pendent prognostic factors were determined by multivariateanalysis using a Cox proportional hazards regression model. Pvalues < 0.05 were considered statistically significant. Statisticalanalyses were performed using SPSS 22.0 software (IBM).

Data availabilityThemicroarray data sets used in this paper have been deposited

at Gene-Expression Omnibus (http://www.ncbi.nlm.nih.gov/geo/) under the series accession numbers GSE52068, GSE12452,GSE53819, and GSE64634. Gene-expression profiling interactiveanalysis in different cancer types and corresponding normaltissues was deposited at GEPIA based on TCGA and GTExdata (http://www.ncbi.nlm.nih.gov/geo/). The authenticity ofthis article has been validated by uploading the key raw dataonto the Research Data Deposit public platform (http://www.researchdata.org.cn), with the approval RDD number asRDDB2018000395.

ResultsRAB37 promoter is hypermethylated in NPC

The regulatory network constructed by genomic or epigeneticaberrations has been revealed to reflect the core molecularmechanisms underlying tumorigenesis and progression (26–28).Previously, our methylation microarray study (GSE52068)identified 473,788 differential methylated gene CpG locusbetween normal human nasopharyngeal (n ¼ 24) and NPCtumor (n¼ 24) samples (15). However, the regulatory interactionamong these dysmethylated loci remains unclear. Here, using asemiparametric Bayesian model, we found differentially coregu-latory methylation loci in networks between NPC and normaltissues with the criteria of interaction link probability > 0.99(Supplementary Table S1). To further evaluate function-associ-ated networks, 19 methylation loci were identified to be involvedin regulatory network interactions (Supplementary Fig. S1). Inter-estingly, these methylation loci could distinguish NPC tissuesfrom normal tissues and clustered together in hierarchical clusteranalysis (P < 0.05 and Db � |0.2|, Fig. 1A). To identify genes for

Li et al.

Clin Cancer Res; 24(24) December 15, 2018 Clinical Cancer Research6498

on May 19, 2021. © 2018 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 21, 2018; DOI: 10.1158/1078-0432.CCR-18-0532

Page 5: RAB37 Hypermethylation Regulates Metastasis and Resistance to Docetaxel … · confirmed that docetaxel, cisplatin, and fluorouracil (TPF) induction chemotherapy (IC) significantly

Figure 1.

RAB37 promoter is hypermethylated in NPC tissues. A, Heat map clustering of dysmethylated genes involved in protein–protein interaction networksbetween NPC (n ¼ 24) and normal nasopharyngeal tissues (n ¼ 24). B, Schematic location of CpG islands (blue area) within the RAB37 promoter region. Thetranscriptional start site is indicated with an arrow. Regions analyzed by bisulfite pyrosequencing are listed (bottom). C–E, Bisulfite pyrosequencing analysisand quantitative methylation levels of freshly frozen normal nasopharyngeal epithelial tissues (n ¼ 8) and NPC tissues (n ¼ 8) (C, D) and immortal NP69 andNPC cell lines (E). Cell line methylation results are presented as the mean (n ¼ 3) � SD. Student t test, �� , P < 0.01.

Epigenetic Silencing of RAB37 Promotes NPC Metastasis

www.aacrjournals.org Clin Cancer Res; 24(24) December 15, 2018 6499

on May 19, 2021. © 2018 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 21, 2018; DOI: 10.1158/1078-0432.CCR-18-0532

Page 6: RAB37 Hypermethylation Regulates Metastasis and Resistance to Docetaxel … · confirmed that docetaxel, cisplatin, and fluorouracil (TPF) induction chemotherapy (IC) significantly

which the altered methylation status was causally linked toexpression dysregulation, we examined the mRNA expression ofmethylated genes in public data sets from the GEO. Notably, 10genes were differentially expressed in NPC in at least one data set,but only RAB37 was significantly downregulated in all three datasets (GSE12452, GSE53819, and GSE64634; SupplementaryTable S2).

To further validate the dysregulated methylation status ofRAB37, we performed bisulfite pyrosequencing analysis in othernormal (n¼ 8) andNPC tissues (n¼ 8). Figure 1B shows the CpGislands and bisulfite pyrosequencing regions in the RAB37 pro-moter. Compared with those in normal tissues, the methylationlevels of RAB37 were significantly upregulated in NPC tissues(Fig. 1C and D, P < 0.01). Consistently, the RAB37 methylationlevel was particularly higher in NPC cell lines than in immortal-ized normal human nasopharyngeal epithelial cells (Fig. 1E,P < 0.05). Collectively, these data suggest that RAB37 is hyper-methylated in NPC.

RAB37 promoter hypermethylation contributes to itsdownregulation in NPC

To investigate the causal link between RAB37 promoter meth-ylation and RAB37 downregulation, we first examined RAB37expression by quantitative RT-PCR andWestern blotting assays intissues and cell lines. The results showed that compared with thatin normal epithelial samples, RAB37 was notably downregulatedin freshly frozen NPC samples (Fig. 2A, P < 0.01). Similarly, theRAB37 expression level was significantly lower in NPC cell linesthan in immortalized NP69 cells (Fig. 2B, P < 0.01). To furtherverify these findings, we performed external validation usingpublic GEO database and demonstrated that RAB37 was alsonotably downregulated in patients with NPC in three data sets(GSE12452, GSE53819, and GSE64634; Figure 2C, Supplemen-tary Fig. S2, and Supplementary Table S2, P < 0.05). Moreover,RAB37 expression levels were broadly downregulated acrossdifferent cancer types, including urothelial bladder carcinoma,lymphoma, glioma and lung carcinoma, based onTCGA (22) andGTEx data (23, 24) by using GEPIA tool (Fig. 2D, P < 0.05). Takentogether, these results indicate that RAB37 is downregulated inNPC and potentially in multiple other tumor types, includinglymphoma, glioma, bladder, and lung carcinoma.

Next, we investigated whether RAB37 promoter hypermethyla-tion leads to RAB37 expression reduction. To this end, bothNP69andNPCcellswere treatedwithorwithout 5-aza-20-deoxycytidine(DAC, an inhibitor of DNA methyltransferase) for 72 hours.Following DAC treatment, RAB37 promoter methylation levelswere found to be significantly lower in NPC cells than in NP69cells (Fig. 2E; Supplementary Fig. S3, P < 0.05). Conversely, themRNA and protein levels of RAB37 were obviously increased inNPC cells followingDAC treatment (Fig. 2F, P < 0.01). Thus, thesefindings illustrate that RAB37 downregulation results from itspromoter hypermethylation in NPC.

RAB37 downregulation is associated with metastasis andchemoresistance in NPC

To understand the potential impact of RAB37 downregulationon the NPC phenotype, we performed GSEA and compared geneprofiles of NPC samples with high and low RAB37 levels inthe GSE12452 data set to identify biological differences betweenthe two subgroups (Fig. 3A). GSEA revealed that comparedwith RAB37-high NPCs, RAB37-low NPCs were significantly

enriched in gene sets related to metastasis (Bidus_Metastasis_Up,Alonso_Metastasis_Up, Rickman_Metastasis_Up, Jaeger_Metas-tasis_Up, and Zucchi_Metastasis_Up) and chemoresistance fea-tures (Honma_Docetaxel_resistance; Fig. 3B). To obtain furtherinsights into the biological pathways involved in NPC pathogen-esis, STRING analysis showed that the top-ranked gene signaturesfrom GSEA were clustered in network and enriched in cell metas-tasis and chemoresistance pathways in patients with low RAB37expression (Supplementary Fig. S4). These data suggest thatRAB37 may be an important modulator of NPC metastasis andchemoresistance.

RAB37 suppresses metastasis and docetaxel resistance in NPCTo functionally validate the above findings, we transfected

RAB37-encoding plasmids into SUNE1 and HONE1 cells toexplore cell motility and invasion ability (SupplementaryFig. S5). Wound-healing assays showed that NPC cells withRAB37 overexpression migrated less than the control cells(Fig. 3C, P < 0.01). In transwell assays, ectopic RAB37 expres-sion significantly inhibited the migratory and invasive abilitiesof NPC cells (Fig. 3D and E, P < 0.01). In contrast, knockdownof RAB37 expression using target siRNA transfection markedlyincreased migration and invasion in immortalized NP69 cells(Fig. 3F, P < 0.01). In addition, we investigated the effects ofRAB37 on cell viability and colonization. The results showedthat RAB37 overexpression had no effects on NPC cell prolif-eration in vitro (Supplementary Fig. S6, P > 0.05). Moreover,DAC treatment significantly suppressed NPC cell proliferation,migration, and invasion, whereas only the inhibitory effects ofmigration and invasion were significantly sensitized by RAB37overexpression (Supplementary Fig. S7, P < 0.05). Altogether,these data suggest that RAB37 inhibits NPC cell migration andinvasion in vitro.

Furthermore, we investigated whether RAB37 is involved inresistance to docetaxel, which we had previously demonstrated ina randomized controlled phase III clinical trial to be an effectiveIC agent in improving DMFS. Indeed, RAB37 overexpressionimpaired the survival of NPC cells (Fig. 3G and H, P < 0.01).The IC50 for docetaxel strikingly decreased to 13.1 mg/mL and1.9 mg/mL in RAB37-overexpressing SUNE1 and HONE1 cells,respectively, compared with that in vector-transfected cells(25.3 and 10.5 mg/mL, Fig. 3G and H). Therefore, RAB37 sup-presses NPC cell migration and invasion and enhances sensitivityto docetaxel chemotherapy in vitro.

RAB37 regulates TIMP2 secretion and inhibits downstreamMMP2

To explore the mechanism underlying the RAB37-mediatedsuppressive effects in NPC, we sought to identify molecularpathways associated with RAB37 dysregulation. GSEA showedthat gene sets related to epithelial-to-mesenchymal transition(EMT; Sarrio_Epithelial_Mesenchymal_Transition_Up) and GOMicrotubule Cytoskeleton Organization and Protein Secretionfeatures were correlated with RAB37 downregulation (Fig. 4A–C).Strikingly, the morphology of NPC cells overexpressing RAB37changed from a spindle shape or elongatedmesenchymal form toan epithelial-like form (Fig. 4D), suggesting that RAB37 functionsto inhibit EMT. In line with these findings, immunofluorescencestaining showed that ectopic RAB37 expression markedlyincreased the protein levels of epithelial markers (E-cadherin)but decreased the protein levels of mesenchymal markers

Li et al.

Clin Cancer Res; 24(24) December 15, 2018 Clinical Cancer Research6500

on May 19, 2021. © 2018 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 21, 2018; DOI: 10.1158/1078-0432.CCR-18-0532

Page 7: RAB37 Hypermethylation Regulates Metastasis and Resistance to Docetaxel … · confirmed that docetaxel, cisplatin, and fluorouracil (TPF) induction chemotherapy (IC) significantly

Figure 2.

RAB37 hypermethylation is associated with its downregulation in NPC. A and B, Real-time PCR analysis and representative Western blot of RAB37 expressionin freshly frozen normal nasopharyngeal epithelial tissues (n ¼ 16) and NPC tissues (n ¼ 16) (A) and immortal NP69 and NPC cell lines (B). Cell linemethylation results are presented as the mean (n ¼ 3) � SD. Student t test, �� , P < 0.01. C, Expression of RAB37 in an independent GEO set of 31 NPC and10 normal tissues (GSE12452). Student t test, �� , P <0.01.D,RAB37 expression level in different cancer based on TCGA andGTEx databases. Student t test; � , P <0.05;�� , P < 0.01. BLCA, urothelial bladder carcinoma; DLBC, diffuse large B-cell lymphoma; GBM, glioblastoma; LGG, lower grade glioma; and LUSC, lungsquamous cell carcinoma. E and F, Methylation levels (E) and RAB37 expression (F) in NP69 and NPC cell lines with or without 5-aza-20-deoxycytidine (DAC)treatment. Mean (n ¼ 3) � SD. Student t test, � , P < 0.05; �� , P < 0.01.

Epigenetic Silencing of RAB37 Promotes NPC Metastasis

www.aacrjournals.org Clin Cancer Res; 24(24) December 15, 2018 6501

on May 19, 2021. © 2018 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 21, 2018; DOI: 10.1158/1078-0432.CCR-18-0532

Page 8: RAB37 Hypermethylation Regulates Metastasis and Resistance to Docetaxel … · confirmed that docetaxel, cisplatin, and fluorouracil (TPF) induction chemotherapy (IC) significantly

(Vimentin, Fig. 4E) in NPC cells. Together, these results indicatethat RAB37 suppresses EMT in NPC cells.

To determine whether RAB37 regulates protein secretion, wenext performed IP plus mass spectrometry in SUNE1 andHONE1 cells with stable RAB37 overexpression. The resultsshowed that TIMP2 had the highest interaction score withRAB37 (Fig. 4F; Supplementary Data 1–2). TIMP2 is a memberof the TIMP family, a secreted protein that inhibits MMPs andextracellular matrix turnover, thereby decreasing cell motility,migration, and invasion (29). However, the regulatory inter-action between RAB37 and TIMP2 has not been reported. Co-IPassay results verified that RAB37 could physically interact with

TIMP2 in NPC cells (Fig. 4G). To address whether RAB37regulates TIMP2 secretion, we determined the expression levelsof TIMP2 in CM from control and ectopic RAB37-expressingcells. Concordant increase in TIMP2 expression was observed inthe CM of NPC cells overexpressing RAB37 (Fig. 4H). Immu-nofluorescence staining revealed that endogenous RAB37 colo-calized with TIMP2 in NPC cells (Fig. 4I). Moreover, DACtreatment significantly upregulated the secretion TIMP2 levelsin NPC cells (Supplementary Fig. S8, P < 0.05). Importantly,RAB37 overexpression notably decreased the level of activeMMP2 in CM (Fig. 4H). Thus, RAB37 promotes TIMP2 secre-tion and inhibits downstream MMP2.

Figure 3.

RAB37 downregulation is correlated with metastasis- and chemoresistance-associated features in NPC. A, Using GSEA, heat map showing differentially enrichedsignatures between groups with low (green, n ¼ 15) and high (yellow, n ¼ 16) RAB37 expression based on GSE12452 gene-expression data. B, Metastasis-and docetaxel resistance-related biological functions enriched in response to low RAB37 expression. C–E, Representative images and quantification of wound-healing (C) andmigration and invasion transwell (D,E) assays inNPC cells transfectedwithRAB37 expressionplasmids or control vector.Mean (n¼ 3)�SD. Student ttest, �� , P < 0.01. F, Representative images and quantification of transwell assays in immortalized NP69 cells transfected with RAB37-targeted or control siRNAs.Mean (n ¼ 3) � SD. Student t test, �� , P < 0.01. G and H, Dose–response curve for docetaxel treatment in SUNE1 (E) and HONE1 (F) cells transfected withRAB37 expression plasmids or vector. The representations of RAB37 and vector groupswere relative to its own control (RAB37 overexpression or vector in 0 mg/mLdocetaxel), respectively. The comparison was performed between the groups with RAB37 and vector at the same docetaxel concentration. Mean (n ¼ 4) � SD.Student t test, � , P < 0.05; �� , P < 0.01. Scale bar, 100 mm.

Li et al.

Clin Cancer Res; 24(24) December 15, 2018 Clinical Cancer Research6502

on May 19, 2021. © 2018 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 21, 2018; DOI: 10.1158/1078-0432.CCR-18-0532

Page 9: RAB37 Hypermethylation Regulates Metastasis and Resistance to Docetaxel … · confirmed that docetaxel, cisplatin, and fluorouracil (TPF) induction chemotherapy (IC) significantly

Figure 4.

RAB37 regulates EMT and TIMP2 secretion.A–C, EMT- (A), microtubule cytoskeleton- (B) and protein secretion-related (C) signaling pathways enriched in responseto lowRAB37 expression.D,Representative imagesofNPCcellswith orwithoutRAB37overexpression.E, Immunofluorescence imagesof E-cadherin andVimentin inNPC cells with or without RAB37 overexpression. F, Staining of RAB37-associated proteins. Two RAB37-specific bands (red box) were excised andanalyzed by mass spectrometry in NPC cells overexpressing RAB37. G, Western blot analysis of RAB37 and TIMP2 from co-IP assays using anti-HA antibodies.H, Western blot analysis of RAB37 and TIMP2 in CM, and proteolytic activity of MMP2 in CM. I, Immunofluorescence images of RAB37 and TIMP2 in NPC cellswith or without RAB37 overexpression. Data are representative of three independent experiments. Scale bar, 50 mm.

Epigenetic Silencing of RAB37 Promotes NPC Metastasis

www.aacrjournals.org Clin Cancer Res; 24(24) December 15, 2018 6503

on May 19, 2021. © 2018 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 21, 2018; DOI: 10.1158/1078-0432.CCR-18-0532

Page 10: RAB37 Hypermethylation Regulates Metastasis and Resistance to Docetaxel … · confirmed that docetaxel, cisplatin, and fluorouracil (TPF) induction chemotherapy (IC) significantly

RAB37 suppresses NPC cell metastasis by regulating TIMP2secretion

We next investigated whether TIMP2 is required for RAB37-mediated inhibitory effects onNPC cells. TIMP2-targeting siRNAswere transfected into SUNE1 andHONE1 cells with stable RAB37overexpression. The results showed that the knockdown of TIMP2expression significantly abrogated the RAB37-mediated inhibi-tion of migration and invasion (Fig. 5A and B, P < 0.05). Notably,TIMP2 downregulation resulted in decreased secretion of TIMP2in CM and thus reversed the RAB37-mediated attenuation in thelevel of MMP2 in CM (Fig. 5C; Supplementary Fig. S9). Together,these data demonstrate that RAB37 inhibits NPC cell migrationand invasion by regulating TIMP2 secretion in vitro.

To further evaluate the effect of RAB37 on NPC metastasis invivo, we used lung metastasis colonization models. The mice inRAB37 overexpression group displayed significantly fewer lungmetastatic nodules than did the mice in the control group (Fig.5D, P < 0.01). Hematoxylin and eosin staining confirmed thatmice with RAB37 overexpression exhibited remarkably fewer andsmaller metastatic tumors in the lungs (Fig. 5E, P < 0.01).Moreover, we constructed xenograft lung metastasis colonizationmodel to determine the effects of RAB37 on docetaxel chemo-sensitivity in NPC cell metastasis. Docetaxel treatment signifi-cantly reduced lung metastatic tumors in mice, which was furthersensitized by RAB37 overexpression (Supplementary Fig. S10,P < 0.05). Of note, concordantly high expression of RAB37 andTIMP2 was confirmed in oligometastatic lung nodules in theRAB37-overexpressing group in vivo, suggesting that RAB37 andTIMP2 expression was pathologically relevant (Fig. 5F). Hence,RAB37 suppresses NPC cell metastasis by regulating TIMP2 secre-tion both in vitro and in vivo.

RAB37 hypermethylation is associated with adverse prognosisfor distant metastasis in patients with NPC

To assess the clinical relevance of our findings, we evaluated therelation betweenRAB37methylation status and clinicopathologicparameters in 110 patients with NPC. Bisulfite pyrosequencinganalysis revealed that the RAB37 promoter was differentiallymethylated in NPC specimens. With a cutoff value of 5.8%, pati-ents were divided into high RAB37 methylation (methylationlevel�5.8) or low RAB37methylation groups (methylation level<5.8%). As shown in Supplementary Table S3, no significantcorrelation was found between RAB37 methylation status andpatient age, sex, viral capsid antigen immunoglobulin A (VCA-IgA), early antigen immunoglobulin A (EA-IgA), or clinical stage.However, RAB37 methylation status was significantly correlatedwith death (P ¼ 0.010) and distant metastasis (P ¼ 0.009;Supplementary Table S3). Kaplan–Meier survival analysisshowed that patients with high RAB37 methylation levels hadsignificantly shorter overall survival (OS; 5-year OS; 76.7% vs.100%, P ¼ 0.010) and DMFS rates (5-year DMFS; 75.5% vs.95.8%, P ¼ 0.010; Fig. 6A and B) than patients with low RAB37methylation levels.

Furthermore, multivariate analysis revealed that the RAB37methylation status [OS, hazard ratio (HR), 4.265; 95%confidenceinterval (CI), 1.291–14.10; P¼ 0.017; DMFS, HR, 8.316; 95%CI,1.119–61.774; P ¼ 0.038] and N stage (OS, HR, 2.409; 95% CI,1.036–5.601; P¼ 0.041; DMFS,HR, 3.365; 95%CI, 1.303–8.695;P ¼ 0.012) were independent prognostic indicators for OS andDMFS (Supplementary Table S4). These findings suggest thatRAB37hypermethylation is clinically associatedwith unfavorable

outcomes for metastasis and may serve as an independent prog-nostic predictor for patients with NPC.

Prognostic model combining RAB37 methylation and N stagefor predicting NPC metastasis and docetaxel IC benefit

To provide a clinically applicable method to stratify patientsat various risk of distant metastasis, we built a predictive modelbased on RAB37 methylation status and N stage. According tothis model, patients were classified into the following threeseparate groups: 23 (20.9%) patients in the low-risk group (lowRAB37 methylation and early N stage), 72 (65.5%) patients inthe intermediate-risk group (high RAB37 methylation oradvanced N stage), and 15 (13.6%) patients in the high-riskgroup (high RAB37methylation and advanced N stage). Impor-tantly, patients in the three groups were at significantly differentrisk of distant metastasis, and the combination of high RAB37methylation or/and advanced N stage identified patients withnotably high risk of distant metastasis (Fig. 6C; SupplementaryFig. S11, P ¼ 0.002). The data indicate that the combination ofthe RAB37 methylation status and N stage is more precise thanclinical stage alone for predicting distant metastasis in NPC.

Furthermore, we evaluated the clinical value of the RAB37methylation status in the prediction of sensitivity to docetaxel-containing IC. In combined intermediate- and high-risk groups,patients who received docetaxel IC experienced significantly bet-ter control of distant metastasis than patients who did not receivedocetaxel IC (5-yearDMFS; 81.9%vs. 50%,P¼ 0.047, Fig. 6D). Incontrast, patients in the low-risk group did not benefit fromdocetaxel-containing IC (Supplementary Fig. S12). These findingssuggest that our prognostic model based on the RAB37 methyl-ation status and N stage is effective in stratifying patients who areat a high risk of distant metastasis and likely to benefit fromdocetaxel-containing IC.

DiscussionThe present study highlights the oncogenic role of RAB37

hypermethylation in NPC with broad biological and clinicalrelevance. We demonstrated that NPCs with methylation-medi-ated RAB37 downregulation are enriched in metastasis- andchemoresistance-related pathways. Moreover, reversal of RAB37overexpression can suppress NPC cell metastasis by regulatingTIMP2 secretion and can increase docetaxel chemosensitivity,offering opportunities for novel therapeutic intervention. Fur-thermore, we provided evidence that our prognostic model com-bining RAB37 hypermethylation status with advanced N stageeffectively identified patient at a high risk for distant metastasiswho are likely to benefit from docetaxel-based IC, representing apromising strategy for the treatment of NPC metastasis.

Metastasis is the major reason of therapeutic failure and deathin patients with cancer (30, 31). At present, the prognosis andtreatment decisions for patients with metastatic NPC mainlydepend on clinical TNM stage (2). Although patients with thesame stage of LA-NPC receive similar therapy, they eventuallydisplay dramatic differences in clinical outcomes, with approxi-mate 30% distant dissemination rate (5). Hence, heterogeneityexists in patients with NPC, and the traditional TNM stage is notsufficient to stratify patients at individual risk of metastasis.Substantial evidence confirms that epigenetic alterations, partic-ularly DNA methylation, play profound roles in cancer develop-ment, progression, metastasis, and drug resistance (32–34).

Li et al.

Clin Cancer Res; 24(24) December 15, 2018 Clinical Cancer Research6504

on May 19, 2021. © 2018 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 21, 2018; DOI: 10.1158/1078-0432.CCR-18-0532

Page 11: RAB37 Hypermethylation Regulates Metastasis and Resistance to Docetaxel … · confirmed that docetaxel, cisplatin, and fluorouracil (TPF) induction chemotherapy (IC) significantly

Figure 5.

RAB37 suppresses NPC cell metastasis through regulating TIMP2. SUNE1 and HONE1 cells overexpressing RAB37 were transfected with either siTIMP2 ornegative control (NC). A and B, Transwell migration (A) or invasion (B). Representative images (left) and quantification (right) of transwell assays. Mean (n ¼ 3)� SD. Student t test, � , P < 0.05; �� , P < 0.01. C,Western blot analysis of RAB37 and TIMP2 in CM, and proteolytic activity of MMP2 in CM. � , P < 0.01. D and E, SUNE1cells (2 � 105 cells in 100-mL PBS) were injected into the tail veins of mice. Representative images and quantification of macroscopic (D) and microscopicmetastatic nodules (E) in the lungs of mice. Mean (n ¼ 8) � SD. Student t test, �� , P < 0.01. F, IHC images of RAB37 and TIMP2 in lung metastatic noduleswith or without RAB37 overexpression. Scale bar, 100 mm.

Epigenetic Silencing of RAB37 Promotes NPC Metastasis

www.aacrjournals.org Clin Cancer Res; 24(24) December 15, 2018 6505

on May 19, 2021. © 2018 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 21, 2018; DOI: 10.1158/1078-0432.CCR-18-0532

Page 12: RAB37 Hypermethylation Regulates Metastasis and Resistance to Docetaxel … · confirmed that docetaxel, cisplatin, and fluorouracil (TPF) induction chemotherapy (IC) significantly

In this study, a group of dysmethylated genes were identifiedto closely interact with each other and to preferably distinguishNPC tissues from normal tissues. Based on the regulatory roleof DNA methylation in gene expression, significant RAB37downregulation in NPC was validated across three public GEOdata sets and shown to result from promoter hypermethylation,consistent with the findings in lung cancer and esophagealsquamous cell carcinoma (ESCC; refs. 35–37). Moreover,TCGA and GTEx data analysis confirmed the downregulatedRAB37 expression in a wide range of cancer types, includinglymphoma, glioma, urothelial bladder carcinoma, and lungsquamous cell carcinoma. These results indicate that RAB37might function as a pan-cancer suppressor. However, the rolesand mechanisms of RAB37 remain undefined in NPC. GSEAdemonstrated that metastasis-related pathways were enrichedin response to RAB37 downregulation. Functionally, RAB37suppressed NPC metastasis in vitro and in vivo, consistent withthe inhibitory effects of RAB37 in other cancer types (35–37).

Therefore, RAB37 hypermethylation contributes to its down-regulation and leads to metastasis in NPC.

RAB37 is a member of the RAB GTPase family of proteins thatare critical regulators for membrane vesicle trafficking (36–38).Dysregulation of the RAB pathway is associated with a wide rangeof human diseases, including cancer (39–42). However, there islimited knowledge regarding the roles and mechanisms ofRAB37 in NPC. Our study demonstrated that RAB37 not onlyplays highly important roles in metastasis and EMT but alsoinhibits docetaxel-based chemoresistance in NPC, which providenew insights into the biological and pathologic roles of RAB37.Bioinformatics analysis revealed thatRAB37 alterations are highlyrelevant to microtubule cytoskeleton organization and proteinsecretion pathways in NPC, which have been verified to be im-portant drivers of tumor progression, metastasis, and chemother-apy resistance (43–48). Based on mass spectrometry data, weconfirmed that TIMP2 has the highest score and is the onlymember of the secreted TIMP protein family that physically

Figure 6.

RAB37 hypermethylation is associated with poor survival and docetaxel-based IC sensitivity in patients with NPC. A and B, Kaplan–Meier analysis of overallsurvival (OS, A) and distant metastasis-free survival (DMFS, B) according to the RAB37 methylation status in 110 patients with NPC. C, Kaplan–Meieranalysis, based on the prognostic model, of DMFS in low-risk (low RAB37 methylation and early N stage), intermediate-risk (high RAB37 methylationor advanced N stage), and high-risk (high RAB37 methylation and advanced N stage) patients with NPC. D, DMFS in intermediate- and high-riskpatients treated with IC with and without docetaxel. P values were determined using the log-rank test.

Li et al.

Clin Cancer Res; 24(24) December 15, 2018 Clinical Cancer Research6506

on May 19, 2021. © 2018 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 21, 2018; DOI: 10.1158/1078-0432.CCR-18-0532

Page 13: RAB37 Hypermethylation Regulates Metastasis and Resistance to Docetaxel … · confirmed that docetaxel, cisplatin, and fluorouracil (TPF) induction chemotherapy (IC) significantly

interacts with RAB37 in NPC. Furthermore, overexpressed RAB37exerts its suppressive effects through the regulation of TIMP2secretion and downstream MMP2 inhibition in NPC cells, whichis in line with previous findings in other cancer types (33, 34),thereby uncovering the role of a novel RAB37/TIMP2/MMP2 axisin tumor biology.

Accumulating evidence on epigenetic aberrations in cancerhas emphasized their crucial roles in diagnosis, prognosis, andthe prediction of therapy response (10, 31). Here, we foundthat patients with RAB37 hypermethylation exhibited poorsurvival, which is similar to the progression-free survival ten-dency observed in lung cancer and ESCC patients (32–34).Both univariate and multivariate analyses showed that theRAB37 methylation status and N stage were independent prog-nostic factors for OS and DMFS. The prognostic model com-bining RAB37 methylation with N stage could accurately strat-ify patients at risk of metastasis. In addition, we showed thatdocetaxel-based IC is beneficial for the control of distantmetastasis in patients classified into the intermediate- tohigh-risk groups by our model but not in patients in thelow-risk group. IC has advantages of acceptable toxicity andearly elimination of micrometastases. Our prospective phase IIIstudy confirmed that the addition of TPF IC to CCRT caneffectively reduce distant metastasis and prolong survival.However, the criteria for enrollment mainly depend on TNMstage (4), and the precise candidates exactly benefiting fromTPF IC remain undefined. Therefore, we conclude that patientsin the intermediate- to high-risk groups might be cured bydocetaxel-based IC, whereas low-risk patients may be candi-dates for less aggressive therapeutic strategies that preventtumor metastasis. Our study may thus improve predictionstrategies for evaluating individualized risk of distant metasta-sis for IC treatment.

In conclusion, we identified a key oncogenic role of RAB37hypermethylation in NPC metastasis via the RAB37/TIMP2/MMP2 axis. We also highlighted the effective value of an RAB37methylation-based prognostic model and confirmed docetaxel-

based IC as an effective treatment for patients with NPC at ahigh risk of metastasis (with RAB37 hypermethylation or/andadvanced N stage), thereby facilitating the development of novelpredictive and therapeutic strategies against NPC metastasis.

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

Authors' ContributionsConception and design: Y. Li, X. Du, N. LiuDevelopment of methodology: X. Yang, X. Tang, P. ZhangAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): Y. Li, X. Yang, X. Du, Y. Lei, Q. He, X. HongAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis): Y. Li, X. Yang, X. Du, Y. Lei, X. Hong, X. Tang, X. Wen,Y. Sun, J. Zhang, Y. Wang, J. Ma, N. LiuWriting, review, and/or revision of the manuscript: Y. Li, X. Du, Y. Sun, J. Ma,N. LiuAdministrative, technical, or material support (i.e., reporting or organizingdata, constructing databases): Y. Li, X. Du, N. LiuStudy supervision: N. Liu

AcknowledgmentsWe would like to thank Hanqi Yin (Guangdong Longsee Biomedical Cor-

poration, China) for bioinformatics and statistical consultation.This study was supported by grants from the National Natural Science

Foundation of China (81703026 and 81773229), the Natural Science Foun-dation of Guangdong Province (2017A030312003 and 2017A030313881), theHealth and Medical Collaborative Innovation Project of Guangzhou City,China (201803040003), the Innovation Team Development Plan of the Min-istry of Education (IRT_17R110), the Overseas Expertise Introduction Projectfor Discipline Innovation (111 Project, B14035), and the Young TeachersCultivation Project of Sun Yat-sen University (16ykpy21).

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 February 13, 2018; revised June 30, 2018; accepted August 17, 2018;published first August 21, 2018.

References1. ChenW, Zheng R, Baade PD, Zhang S, ZengH, Bray F, et al. Cancer statistics

in China, 2015. CA Cancer J Clin 2016;66:115–32.2. Chua ML, Wee JT, Hui EP, Chan AT. Nasopharyngeal carcinoma. Lancet

2016;387:1012–24.3. Lai SZ, Li WF, Chen L, Luo W, Chen YY, Liu LZ, et al. How does intensity-

modulated radiotherapy versus conventional two-dimensional radiother-apy influence the treatment results in nasopharyngeal carcinoma patients?Int J Radiat Oncol Biol Phys 2011;80:661–8.

4. Lee AW, Ma BB, Ng WT, Chan AT. Management of nasopharyngealcarcinoma: current practice and future perspective. J Clin Oncol 2015;33:3356–64.

5. Chen YP, Wang ZX, Chen L, Liu X, Tang LL, Mao YP, et al. A Bayesiannetwork meta-analysis comparing concurrent chemoradiotherapy fol-lowed by adjuvant chemotherapy, concurrent chemoradiotherapy aloneand radiotherapy alone in patients with locoregionally advanced naso-pharyngeal carcinoma. Ann Oncol 2014;26:205–11.

6. Sun Y, Li WF, Chen NY, Zhang N, Hu GQ, Xie FY, et al. Inductionchemotherapy plus concurrent chemoradiotherapy versus concurrent che-moradiotherapy alone in locoregionally advanced nasopharyngeal carci-noma: a phase 3, multicentre, randomised controlled trial. Lancet Oncol2016;17:1509–20.

7. Cao SM, Yang Q, Guo L, Mai HQ, Mo HY, Cao KJ, et al. Neoadjuvantchemotherapy followed by concurrent chemoradiotherapy versus concur-rent chemoradiotherapy alone in locoregionally advanced nasopharyngeal

carcinoma: aphase IIImulticentre randomised controlled trial. Eur JCancer2017;75:14–23.

8. Peng H, Chen L, Zhang J, Li WF, Mao YP, Zhang Y, et al. Inductionchemotherapy improved long-term outcomes of patients with locoregion-ally advancednasopharyngeal carcinoma: a propensitymatched analysis of5-year survival outcomes in the era of intensity-modulated radiotherapy. JCancer 2017;8:371–7.

9. ZhangY, LiWF,MaoYP, ZhouGQ,PengH, SunY, et al. Establishment of anintegrated model incorporating standardised uptake value and N-classifi-cation for predicting metastasis in nasopharyngeal carcinoma. Oncotarget2016;7:13612–20.

10. Flavahan WA, Gaskell E, Bernstein BE. Epigenetic plasticity and the hall-marks of cancer. Science 2017;357:pii: eaal2380.

11. Heyn H, Esteller M. DNA methylation profiling in the clinic: applicationsand challenges. Nat Rev Genet 2012;13:679–92.

12. Teschendorff AE, ReltonCL. Statistical and integrative system-level analysisof DNA methylation data. Nat Rev Genet 2018;19:129–47.

13. Lyko F. TheDNAmethyltransferase family: a versatile toolkit for epigeneticregulation. Nat Rev Genet 2017;2:81–92.

14. Li LL, Shu XS, Wang ZH, Cao Y, Tao Q. Epigenetic disruption of cellsignaling in nasopharyngeal carcinoma. Chin J Cancer 2011;30:231–9.

15. Jiang W, Liu N, Chen XZ, Sun Y, Li B, Ren XY, et al. Genome-wideidentification of a methylation gene panel as a prognostic biomarker innasopharyngeal carcinoma. Mol Cancer Ther 2015;14:2864–73.

Epigenetic Silencing of RAB37 Promotes NPC Metastasis

www.aacrjournals.org Clin Cancer Res; 24(24) December 15, 2018 6507

on May 19, 2021. © 2018 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 21, 2018; DOI: 10.1158/1078-0432.CCR-18-0532

Page 14: RAB37 Hypermethylation Regulates Metastasis and Resistance to Docetaxel … · confirmed that docetaxel, cisplatin, and fluorouracil (TPF) induction chemotherapy (IC) significantly

16. Ren X, Yang X, Cheng B, Chen X, Zhang T, He Q, et al. HOPX hypermethy-lation promotes metastasis via activating SNAIL transcription in nasopha-ryngeal carcinoma. Nat Commun 2017;8:14053.

17. Morrissey ER, Juarez MA, Denby KJ, Burroughs NJ. Inferring the time-invariant topology of a nonlinear sparse gene regulatory network usingfully Bayesian spline autoregression. Biostatistics 2011;12:682–94.

18. AminMB, Edge S, Greene F, Byrd DR. AJCC cancer stagingmanual (8th ed)Springer: New York2017.

19. Zweig MH, Campbell G. Receiver-operating characteristic (ROC) plots: afundamental evaluation tool in clinical medicine. Clin Chem1993;39:561–77.

20. Livak KJ, Schmittgen TD. Analysis of relative gene expression data usingreal-time quantitative PCR and the 2(-Delta Delta C (T))Method.Methods2001;25:402–8.

21. Tang Z, Li C, Kang B, Gao G, Li C, Zhang Z. GEPIA: a web server for cancerand normal gene expression profiling and interactive analyses. NucleicAcids Res 2017;45:W98–102.

22. Weinstein JN, Collisson EA, Mills GB, Shaw KR, Ozenberger BA, Ellrott K,et al. The Cancer Genome Atlas pan-cancer analysis project. Nat Genet2013;45:1113–20.

23. Chang K, Creighton CJ, Davis C, Donehower L, Drummond J, Wheeler D,et al. The Genotype-Tissue Expression (GTEx) project. Nat Genet2013;45:580–5.

24. Ardlie KG, Deluca DS, Segr�e AV, Sullivan TJ, Young TR, Gelfand ET, et al.The Genotype-Tissue Expression (GTEx) pilot analysis: multitissue generegulation in humans. Science 2015;348:648–60.

25. SubramanianA, TamayoP,Mootha VK,Mukherjee S, Ebert BL,GilletteMA,et al. Gene set enrichment analysis: a knowledge-based approach forinterpreting genome-wide expression profiles. Proc Natl Acad Sci U S A2005;102:15545–50.

26. Mine KL, ShulzhenkoN, Yambartsev A, RochmanM, SansonGF, LandoM,et al. Gene network reconstruction reveals cell cycle and antiviral genes asmajor drivers of cervical cancer. Nat Commun 2013;4:1806.

27. Mitra K, Carvunis AR, Ramesh SK, Ideker T. Integrative approaches forfinding modular structure in biological networks. Nat Rev Genet2013;14:719–32.

28. Califano A, Alvarez MJ. The recurrent architecture of tumour initiation,progression and drug sensitivity. Nat Rev Cancer 2017;17:116–30.

29. Sato T, Koike L,Miyata Y,HirataM,Mimaki Y, Sashida Y, et al. Inhibition ofactivator protein-1 binding activity and phosphatidylinositol 3-kinasepathway by nobiletin, a polymethoxy flavonoid, results in augmentationof tissue inhibitor of metalloproteinases-1 production and suppression ofproduction of matrix metalloproteinases-1 and -9 in human fibrosarcomaHT-1080 cells. Cancer Res 2002;62:1025–9.

30. HanahanD,Weinberg RA. Hallmarks of Cancer: TheNext Generation. Cell2011;144:646–74.

31. Steeg PS. Targeting metastasis. Nat Rev Cancer 2016;16:201–18.

32. Dawson MA, Kouzarides T. Cancer epigenetics: from mechanism to ther-apy. Cell 2012;150:12–27.

33. Lappalainen T, Greally JM. Associating cellular epigenetic models withhuman phenotypes. Nat Rev Genet 2017;18:441–51.

34. DawsonMA. The cancer epigenome: concepts, challenges, and therapeuticopportunities. Science 2017;355:1147–52.

35. WuCY, TsengRC,HsuHS,WangYC,HsuMT. Frequent down-regulation ofhRAB37 inmetastatic tumor by genetic and epigeneticmechanisms in lungcancer. Lung Cancer 2009;63:360–7.

36. Tsai CH,ChengHC,WangYS, LinP, Jen J, Kuo IY, et al. SmallGTPaseRab37targets tissue inhibitor of metalloproteinase 1 for exocytosis and thussuppresses tumour metastasis. Nat Commun 2014;5:4804.

37. Tzeng HT, Tsai CH, Yen YT, Cheng HC, Chen YC, Pu SW, et al. Dysregula-tion of Rab37-mediated cross-talk between cancer cells and endothelialcells via thrombospondin-1 promotes tumor neovasculature and metas-tasis. Clin Cancer Res 2017;23:2335–45.

38. Sheng Y, Song Y, Li Z, Wang Y, Lin H, Cheng H, et al. RAB37 interactsdirectly with ATG5 and promotes autophagosome formation via regulat-ing ATG5-12-16 complex assembly. Cell Death Differ 2018;25:918–34.

39. Ljubicic S, Bezzi P, Brajkovic S, Nesca V, Guay C, Ohbayashi N, et al. TheGTPase Rab37 participates in the control of insulin exocytosis. Plos One2013;8:e68255.

40. Winslow AR, Chen CW, Corrochano S, Acevedo-Arozena A, Gordon DE,Peden AA, et al. Alpha-synuclein impairs macroautophagy: implicationsfor Parkinson's disease. J Cell Biol 2010;190:1023–37.

41. Cheng KW, Lahad JP, Kuo WL, Lapuk A, Yamada K, Auersperg N, et al. TheRAB25 small GTPase determines aggressiveness of ovarian and breastcancers. Nat Med 2004;10:1251–6.

42. Chehna-Patel N, Sachdeva G, Gajbhiye R, Warty N, Khole V. "Spot"-tingdifferences between the ectopic and eutopic endometrium of endometri-osis patients. Fertil Steril 2010;94:1964–71, 1971.

43. Kalluri R. The biology and function of fibroblasts in cancer. Nat Rev Cancer2016;16:582–98.

44. Britschgi A, Andraos R, Brinkhaus H, Klebba I, Romanet V, Muller U, et al.JAK2/STAT5 inhibition circumvents resistance to PI3K/mTOR blockade: arationale for cotargeting these pathways inmetastatic breast cancer. CancerCell 2012;22:796–811.

45. Ramel D,Wang X, Laflamme C,Montell DJ, Emery G. Rab11 regulates cell-cell communication during collective cell movements. Nat Cell Biol2013;15:317–24.

46. Even-Ram S, Doyle AD, Conti MA, Matsumoto K, Adelstein RS, YamadaKM. Myosin IIA regulates cell motility and actomyosin-microtubule cross-talk. Nat Cell Biol 2007;9:299–309.

47. Besson A, Assoian RK, Roberts JM. Regulation of the cytoskeleton: anoncogenic function for CDK inhibitors? Nat Rev Cancer 2004;4:948–55.

48. Dexter DL, Leith JT. Tumor heterogeneity and drug resistance. J Clin Oncol1986;4:244–57.

Clin Cancer Res; 24(24) December 15, 2018 Clinical Cancer Research6508

Li et al.

on May 19, 2021. © 2018 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 21, 2018; DOI: 10.1158/1078-0432.CCR-18-0532

Page 15: RAB37 Hypermethylation Regulates Metastasis and Resistance to Docetaxel … · confirmed that docetaxel, cisplatin, and fluorouracil (TPF) induction chemotherapy (IC) significantly

2018;24:6495-6508. Published OnlineFirst August 21, 2018.Clin Cancer Res   Yingqin Li, Xiaojing Yang, Xiaojing Du, et al.   CarcinomaDocetaxel-Based Induction Chemotherapy in Nasopharyngeal

Hypermethylation Regulates Metastasis and Resistance toRAB37

  Updated version

  10.1158/1078-0432.CCR-18-0532doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://clincancerres.aacrjournals.org/content/suppl/2018/08/21/1078-0432.CCR-18-0532.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://clincancerres.aacrjournals.org/content/24/24/6495.full#ref-list-1

This article cites 47 articles, 10 of which you can access for free at:

  Citing articles

  http://clincancerres.aacrjournals.org/content/24/24/6495.full#related-urls

This article has been cited by 1 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/24/24/6495To request permission to re-use all or part of this article, use this link

on May 19, 2021. © 2018 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 21, 2018; DOI: 10.1158/1078-0432.CCR-18-0532