11
Microenvironment and Immunology DC-HIL/Glycoprotein Nmb Promotes Growth of Melanoma in Mice by Inhibiting the Activation of Tumor-Reactive T Cells Mizuki Tomihari 1,2 , Jin-Sung Chung 1 , Hideo Akiyoshi 1,3 , Ponciano D. Cruz, Jr. 1 , and Kiyoshi Ariizumi 1 Abstract DC-HIL/glycoprotein nmb (Gpnmb) expressed on antigen-presenting cells attenuates T-cell activation by binding to syndecan-4 (SD-4) on activated T cells. Because DC-HIL/Gpnmb is expressed abundantly by mouse and human melanoma lines, we posited that melanoma-associated DC-HIL/Gpnmb exerts similar inhibitory function on melanoma-reactive T cells. We generated small interfering RNAtransfected B16F10 melanoma cells to completely knock down DC-HIL/Gpnmb expression, with no alteration in cell morphology, melanin synthesis, or MHC class I expression. This knockdown had no effect on B16F10 proliferation in vitro or entry into the cell cycle following growth stimulation, but it markedly reduced the growth of these cells in vivo following their s.c. injection into syngeneic immunocompetent (but not immunodeficient) mice. This reduc- tion in tumor growth was due most likely to an augmented capacity of DC-HILknocked down B16F10 cells (compared with controls) to activate melanoma-reactive T cells as documented in vitro and in mice. Whereas DC-HIL knockdown had no effect on susceptibility of melanoma to killing by cytotoxic T cells, blocking SD-4 function enhanced the reactivity of CD8 + T cells to melanoma-associated antigens on parental B16F10 cells. Using an assay examining the spread to the lung following i.v. injection, DC-HILknocked down cells produced lung foci at similar numbers compared with that produced by control cells, but the size of the former foci was significantly smaller than the latter. We conclude that DC-HIL/Gpnmb confers upon melanoma the ability to downregulate the activation of melanoma-reactive T cells, thereby allowing melanoma to evade immunologic recognition and destruction. As such, the DC-HIL/SD-4 pathway is a potentially useful target for antimela- noma immunotherapy. Cancer Res; 70(14); 577887. ©2010 AACR. Introduction The success or failure of immune responses mounted against tumors is determined by the net result of competing mechanisms defending against or permitting tumor growth and metastasis. Host defense components include natural killer cells and CTLs that can lyse the tumor (1). On the other hand, tumors can use a variety of mechanisms that allow their escape from immunologic surveillance and destruction, including poorly expressed tumor-associated antigens (TAA), self-tolerance to TAA, downregulated MHC molecules, secre- tion of immunosuppressive cytokines [transforming growth factor, interleukin (IL)-10, and vascular endothelial growth factor], and deficient T-cell costimulation (2, 3). T-cell activation results from signals delivered by antigen presenting cells (APC) to the antigen-specific T-cell receptor (TCR) and to costimulatory or coinhibitory receptors on T cells. The principal costimulatory signal is transmitted by CD80 or CD86 on APC to the CD28 receptor on T cells, which amplifies TCR activation signals (4). By contrast, coinhibitory molecules inhibiting TCR signals include the following: programmed cell death-1 (PD-1) and its ligands PD-L1 (B7- H1) and PD-L2 (57), B- and T-lymphocyte attenuator and her- pesvirus entry mediator (4, 8), and Tim-3 ligand/Galectin-9 (on CD4 + T cells) and Tim-3 (on APC and Th1 effector cells; refs. 911). Like APC, tumors also may take advantage of coinhibitory ligands that compromise immunosurveillance by suppressing activation of tumor-reactive CTL, reducing susceptibility of tumors to CTL-lysis, and/or inducing their apoptosis (12, 13). Subtractive cDNA cloning of dendritic APCs (DC) led us to discover DC-HIL, a highly glycosylated (95120 kDa) type I transmembrane receptor expressed constitutively at high le- vels by many APC subsets and at lower levels by some non- lymphoid cells (14). DC-HIL was also identified by others, Authors' Affiliations: 1 Department of Dermatology, The University of Texas Southwestern Medical Center and Dermatology Section (Medical Service), Dallas Veterans Affairs Medical Center, Dallas, Texas; 2 Department of Clinical Veterinary Medicine, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan; and 3 Laboratory of Veterinary Surgery, Department of Veterinary Science, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Osaka, Japan Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/). M. Tomihari and J-S. Chung contributed equally to this work. P.D. Cruz and K. Ariizumi contributed equally to this work. Corresponding Author: Kiyoshi Ariizumi, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9069. Phone: 214-648-7552; Fax: 214-648-0280; E-mail: [email protected]. doi: 10.1158/0008-5472.CAN-09-2538 ©2010 American Association for Cancer Research. Cancer Research Cancer Res; 70(14) July 15, 2010 5778 Research. on January 8, 2020. © 2010 American Association for Cancer cancerres.aacrjournals.org Downloaded from Published OnlineFirst June 22, 2010; DOI: 10.1158/0008-5472.CAN-09-2538

Cancer Research DC-HIL/Glycoprotein Nmb Promotes Growth of ...cancerres.aacrjournals.org/content/canres/70/14/5778.full.pdf · cle. In mice, we showed the DC-HIL/SD-4 pathway to inhibit

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Cancer Research DC-HIL/Glycoprotein Nmb Promotes Growth of ...cancerres.aacrjournals.org/content/canres/70/14/5778.full.pdf · cle. In mice, we showed the DC-HIL/SD-4 pathway to inhibit

5778

Published OnlineFirst June 22, 2010; DOI: 10.1158/0008-5472.CAN-09-2538

Microenvironment and Immunology

Cancer

Research

DC-HIL/Glycoprotein Nmb Promotes Growth of Melanoma inMice by Inhibiting the Activation of Tumor-Reactive T Cells

Mizuki Tomihari1,2, Jin-Sung Chung1, Hideo Akiyoshi1,3, Ponciano D. Cruz, Jr.1, and Kiyoshi Ariizumi1

Abstract

Authors' ATexas SouService),2DepartmeAgricultureVeterinaryof Life andOsaka, Jap

Note: SupResearch O

M. Tomiha

P.D. Cruz a

CorresponSouthwestTX 75390-kiyoshi.ariiz

doi: 10.115

©2010 Am

Cancer R

Do

DC-HIL/glycoprotein nmb (Gpnmb) expressed on antigen-presenting cells attenuates T-cell activation bybinding to syndecan-4 (SD-4) on activated T cells. Because DC-HIL/Gpnmb is expressed abundantly by mouseand human melanoma lines, we posited that melanoma-associated DC-HIL/Gpnmb exerts similar inhibitoryfunction on melanoma-reactive T cells. We generated small interfering RNA–transfected B16F10 melanomacells to completely knock down DC-HIL/Gpnmb expression, with no alteration in cell morphology, melaninsynthesis, or MHC class I expression. This knockdown had no effect on B16F10 proliferation in vitro or entryinto the cell cycle following growth stimulation, but it markedly reduced the growth of these cells in vivofollowing their s.c. injection into syngeneic immunocompetent (but not immunodeficient) mice. This reduc-tion in tumor growth was due most likely to an augmented capacity of DC-HIL–knocked down B16F10 cells(compared with controls) to activate melanoma-reactive T cells as documented in vitro and in mice. WhereasDC-HIL knockdown had no effect on susceptibility of melanoma to killing by cytotoxic T cells, blocking SD-4function enhanced the reactivity of CD8+ T cells to melanoma-associated antigens on parental B16F10 cells.Using an assay examining the spread to the lung following i.v. injection, DC-HIL–knocked down cells producedlung foci at similar numbers compared with that produced by control cells, but the size of the former foci wassignificantly smaller than the latter. We conclude that DC-HIL/Gpnmb confers upon melanoma the ability todownregulate the activation of melanoma-reactive T cells, thereby allowing melanoma to evade immunologicrecognition and destruction. As such, the DC-HIL/SD-4 pathway is a potentially useful target for antimela-noma immunotherapy. Cancer Res; 70(14); 5778–87. ©2010 AACR.

Introduction

The success or failure of immune responses mountedagainst tumors is determined by the net result of competingmechanisms defending against or permitting tumor growthand metastasis. Host defense components include naturalkiller cells and CTLs that can lyse the tumor (1). On the otherhand, tumors can use a variety of mechanisms that allowtheir escape from immunologic surveillance and destruction,

ffiliations: 1Department of Dermatology, The University ofthwestern Medical Center and Dermatology Section (MedicalDallas Veterans Affairs Medical Center, Dallas, Texas;nt of Clinical Veterinary Medicine, Obihiro University ofand Veterinary Medicine, Obihiro, Japan; and 3Laboratory ofSurgery, Department of Veterinary Science, Graduate SchoolEnvironmental Sciences, Osaka Prefecture University,

an

plementary data for this article are available at Cancernline (http://cancerres.aacrjournals.org/).

ri and J-S. Chung contributed equally to this work.

nd K. Ariizumi contributed equally to this work.

ding Author: Kiyoshi Ariizumi, The University of Texasern Medical Center, 5323 Harry Hines Boulevard, Dallas,9069. Phone: 214-648-7552; Fax: 214-648-0280; E-mail:[email protected].

8/0008-5472.CAN-09-2538

erican Association for Cancer Research.

es; 70(14) July 15, 2010

Researcon Januarycancerres.aacrjournals.org wnloaded from

including poorly expressed tumor-associated antigens (TAA),self-tolerance to TAA, downregulated MHC molecules, secre-tion of immunosuppressive cytokines [transforming growthfactor, interleukin (IL)-10, and vascular endothelial growthfactor], and deficient T-cell costimulation (2, 3).T-cell activation results from signals delivered by antigen

presenting cells (APC) to the antigen-specific T-cell receptor(TCR) and to costimulatory or coinhibitory receptors onT cells. The principal costimulatory signal is transmitted byCD80 or CD86 on APC to the CD28 receptor on T cells, whichamplifies TCR activation signals (4). By contrast, coinhibitorymolecules inhibiting TCR signals include the following:programmed cell death-1 (PD-1) and its ligands PD-L1 (B7-H1) and PD-L2 (5–7), B- and T-lymphocyte attenuator and her-pesvirus entry mediator (4, 8), and Tim-3 ligand/Galectin-9(on CD4+ T cells) and Tim-3 (on APC and Th1 effector cells;refs. 9–11). Like APC, tumors also may take advantage ofcoinhibitory ligands that compromise immunosurveillanceby suppressing activation of tumor-reactive CTL, reducingsusceptibility of tumors to CTL-lysis, and/or inducing theirapoptosis (12, 13).Subtractive cDNA cloning of dendritic APCs (DC) led us to

discover DC-HIL, a highly glycosylated (95–120 kDa) typeI transmembrane receptor expressed constitutively at high le-vels by many APC subsets and at lower levels by some non-lymphoid cells (14). DC-HIL was also identified by others,

h. 8, 2020. © 2010 American Association for Cancer

Page 2: Cancer Research DC-HIL/Glycoprotein Nmb Promotes Growth of ...cancerres.aacrjournals.org/content/canres/70/14/5778.full.pdf · cle. In mice, we showed the DC-HIL/SD-4 pathway to inhibit

DC-HIL in Melanoma Immunity

Published OnlineFirst June 22, 2010; DOI: 10.1158/0008-5472.CAN-09-2538

and given a variety of names including glycoprotein nmb(Gpnmb; ref. 15), osteoactivin (16), and hematopoietic growthfactor–inducible neurokinin-1 type (17). The extracellulardomain of DC-HIL contains a cell-adhesion (RGD) motif, aproline-rich region, and an immunoglobulin-like polycystickidney disease domain shared by polycystic kidney disease–susceptible gene products (18). Recently, we found DC-HILto inhibit T-cell activation by binding syndecan-4 (SD-4) onactivated (but not resting) T cells in mice and humans(19–21). This binding attenuates strongly the T-cell responseto anti-CD3 antibody, thereby blocking the T-cell productionof proinflammatory cytokines and T-cell entry into the cell cy-cle. In mice, we showed the DC-HIL/SD-4 pathway to inhibitT cell–mediated contact hypersensitivity (19).Unlike other known coinhibitors of T-cell activation, DC-

HIL/Gpnmb is expressed constitutively and abundantly bymouse melanoma lines, a finding that prompted us to ques-tion whether DC-HIL/Gpnmb on melanoma provides resis-tance to immunologic recognition and destruction bymelanoma-reactive T cells. Our results indicate that DC-HIL/Gpnmb promotes the growth of B16F10 melanoma insyngeneic mice by inhibiting the activation of tumor-reactiveT cells, thereby making the DC-HIL/SD-4 pathway a poten-tially useful target for antimelanoma immunotherapy.

Materials and Methods

Mice and cell cultureFemale C57BL/6 mice (5- to 8-wk-old) were purchased

from Harlan Breeders; nu/nu and pmel-1 TCR transgenicmice [B6.Cg-Thy1a/Cy Tg(TcraTcrb)8Rest/J] were fromJackson Laboratory; and C57BL6-Tg(OT-1)-RAG1tm1Mom werefrom Taconic. Following NIH guidelines, these animalswere housed and cared for in the pathogen-free facilityof the Institutional Animal Care Use Center of The Univer-sity of Texas Southwestern Medical Center. All animal pro-tocols were approved by the Center. B16F10 melanomaand EL-4 T lymphoma were purchased from the AmericanType Culture Collection (ATCC) and maintained in DMEMsupplemented with 10% FCS.

Reverse transcription-PCR analysisTotal RNA (1 μg) isolated from B16F10 cells or DCs was

converted to cDNA by reverse transcriptase (Life Tech-nologies, Inc.; ref. 22). An aliquot (typically 5%) was usedfor PCR ampli f icat ion (22) using the primers : forDC-HIL, 5′-CTGCCAAGCGATTTCGTGATGTG-3′ and 5′-CCTGTCCGGGAACCTGAGATGC-3′; for PD-L1, 5′-CCCA-TACCGCAAAATCAACCAG-3′ and 5′-CAACGCCACATTTCTC‐CACATCTA-3′; for PD-L2, 5′-CACCGTGACAGCCCCTAAA-GAAGT-3′ and 5′-GGGGTCCTGATGTGGCTGGTGT-3′; forherpesvirus entry mediator, 5′-CATCTCTGCCCAGCCCTCAT-3′ and 5′-GCTATCCCAACTCCCACTATCACA-3′; for Tim-3,5′-TGCCAAGTACTCATGTTTTCAGGT-3′ and 5′-CTCT‐CCGTGGTTAGGGTTCTTG-3′; or for β-actin, 5′-GAGCGG-GAAATCGTGCGTGACATT-3′ and 5′-GAAGGTAGTTTCGTG-GATGCC-3′. Following 30 cycles of amplification, PCRproducts were separated electrophoretically on 1% agarose gel.

www.aacrjournals.org

Researcon Januarycancerres.aacrjournals.org Downloaded from

Plasmid vectorsWe constructed a small interfering RNA (siRNA)–

expressing lentivector consisting of DC-HIL–targeted shorthairpin RNA (shRNA; 58 base oligonucleotides, the sensestrand, 5′-GATCCG-GGAACTTGTCTGATGAGATCT-CGAA-AGATCTCATCAGACAAGTTCC-TTTTTG-3 ′ ; and theantisense strand, 5′-AATTCAAAAA-GGAACTTGTCTGATGA-GATCT-TTCG-AGATCTCATCAGACAAGTTCC-CG-3′, inwhich the italic nucleotides are BamHI site in the sense strandand EcoRI in the antisense strand synthesized by IntegratedDNA Technologies and ligated to the downstream of H1 RNApolymerase III promoter in pSIF1-H1-copGFP shRNA lentivec-tor (System Biosciences) that contains a cytomegalovirus-driven fluorescent copepod green fluorescent protein(copGFP). pSIF1-H1-copGFP without a shRNA insert servedas control lentivector. Packaging of pSIF constructs in pseudo-viral particles and their titration were performed according toestablished protocols (23).

Generation of stable transfectantsTo generate B16F10 cells knocked down for DC-HIL,

B16F10 cells (1 × 104) were infected with control or DC-HIL-shRNA lentivector at a multiplication of infection of20. The next day, GFP+ cells were enriched repeatedly byfluorescence-activated cell sorting (FACS) until >90% ofcells were GFP+. To minimize alterations due to in vitroexpansion, an original batch of positive cells was dividedinto several aliquots and stored in liquid nitrogen untilneeded.

Western blot analysisWhole-cell extracts were prepared from B16F10 cells by

lysis with 0.3% Triton X-100/DPBS for 15 minutes, followedby centrifugation for 20 minutes at 10,000× g (24). An aliquot(40 μg) of extract was applied to SDS/4% to 15% gradientPAGE, followed by immunoblotting using UTX-103 rabbitanti-DC-HIL monoclonal antibody (mAb; 2 μg/mL; ref. 25)or anti–β-actin antibody (Abcam). For examining theexpression of DC-HIL and gp100 in exosomes, an aliquot(20 μg) was applied to immunoblotting using UTX-103 or1 μg/mL mouse anti-human melanosome (gp100) mAb(clone HMB45, DakoCytomation, Inc.). Color was developedby horseradish peroxidase–secondary antibody (1:10,000dilution, Jackson ImmunoResearch Laboratories, Inc.) for1 hour and enhanced chemiluminescence plus system(Amersham Pharmacia Biotech).

Flow cytometryTo analyze surface expression, B16F10 cells (1 × 105) were

incubated with UTX-103 mAb, biotinylated anti-H-2Db

(eBioscience), or isotypic control antibody (each 5 μg/mL).To measure the expression of H-2Kb molecule bound to oval-bumin (OVA) peptide, B16 F10 cells were preincubated with2 μg/mL OVA257-264 (SIINFEKL) for 2 hours before stainingwith 5 μg/mL phycoerythrin (PE)-conjugated anti–H-2Kb

(Kb-SIINFEKL, eBioscience). After extensive washing, cellswere labeled fluorescently with secondary antibody [5 μg/mLPE- or FITC-anti-rabbit IgG (Jackson ImmunoResearch

Cancer Res; 70(14) July 15, 2010 5779

h. 8, 2020. © 2010 American Association for Cancer

Page 3: Cancer Research DC-HIL/Glycoprotein Nmb Promotes Growth of ...cancerres.aacrjournals.org/content/canres/70/14/5778.full.pdf · cle. In mice, we showed the DC-HIL/SD-4 pathway to inhibit

Tomihari et al.

5780

Published OnlineFirst June 22, 2010; DOI: 10.1158/0008-5472.CAN-09-2538

Laboratories, Inc.) or 5 μg/mL PE-streptavidin (BD Bio-sciences)]. Fluorescence intensity of stained cells was ana-lyzed by FACS Calibur (BD Biosciences).

MTT assayTumor cell growth in vitro was measured by the colorime-

tric MTT assay. Briefly, exponentially growing control orknockdown transfectants were washed thoroughly and incu-bated for 24 hours in serum-free media to synchronize cellgrowth. Thereafter, 1 × 104 cells per well were seeded in96-well plates (in triplicate) and cultured in complete DMEM.Cell growth was determined each day by adding 10 μL MTTReagent (from ATCC). Cells were then incubated at 37°C for2 hours, and 100 μL Detergent Reagent (ATCC) were addedand left at room temperature in the dark for 2 hours. Absor-bance was determined at 595 nm.

Cell cycle analysisB16F10 melanoma cells were seeded on a Petri dish (5 ×

105 cells/dish) and starved for 24 hours in serum-free mediato synchronize. Proliferation was stimulated by adding 10%FCS media and by culturing for 18 hours. These cells weresubjected to cell cycle analysis using bromodeoxyuridine(BrdUrd) labeling and the BrdUrd flow kit (BD Biosciences),following the manufacturer's recommendations. BrdUrdincorporation was assessed by staining with biotinylatedanti-BrdUrd antibody (BioLegend) and APC-conjugatedsteptavidin (eBioscience). BrdUrd labeling and DNA content(stained by 7-AAD) on a per cell basis were analyzed byflowcytometry.

In vivo tumor growthC57BL/6 or nu/nu mice were inoculated with control or

knockdown B16F10 transfectants by s.c. injection of 2 × 105

cells in 50 μL of sterile DPBS into the right shaved flank. Tu-mor growth was monitored daily by measuring tumor per-pendicular diameters with a metric caliper, and tumorvolume was estimated as (d2 × D × 0.5), in which d and Dare the minor and major diameters, respectively (26). Forethical reasons, animals were sacrificed when tumors grewto a volume of >5 cm3.

Exosome preparationCulture supernatant was harvested from 72-hour culture

of confluent melanoma cells (C-B16 and Kd-B16). Thesupernatant was centrifuged at 5,000× g for 30 minutesat 4°C to remove cell debris, and the obtained supernatantwas then ultracentrifugated at 100,000× g for 6 hours at4°C (27). The pellet (membrane fraction) was recoveredand measured for protein concentration and resuspendedin DPBS.

Ex vivo analyses of T cellsTo examine the capacity of B16F10 transfectants to stim-

ulate T cells, CD8+ T cells isolated from OT-I transgenicmice, using the CD8+ T-cell isolation kit (Miltenyi Biotec),were seeded at a density of 2 × 105 cells per well in 96-wellmicroplates (in triplicate) and cocultured with B16F10

Cancer Res; 70(14) July 15, 2010

Researcon Januarycancerres.aacrjournals.org Downloaded from

transfectants (2 × 104 cells/well) pulsed with 2 μg/mLOVA257-264 for 3 hours, then subsequently treated with mito-mycin C (MMC, 25 ng/mL) for 1 hour. After culturing for2 days, IL-2 and/or IFN-γ production was measured usingan ELISA kit (eBioscience). In some experiments, insteadof naive OVA-specific T cells, TAA-immunized CD8+ T cellswere used; C57BL/6 mice were immunized by footpad injec-tion of 30 μL TiterMax adjuvant (Sigma-Aldrich) containingTAA peptide mixture (hgp100 aa25-33 and TRP-2 aa180-188,each 20 μg, both from ANASPEC). Seventeen days post-immunization, cells were prepared from spleen and lymphnodes (LN) of the mice, which were then stimulated in vitrowith γ-irradiated (3,000 Gy) syngeneic spleen cells that werepulsed with the peptide mixture (2 μg/mL hgp100 and 2 μg/mLTRP-2). After 10 days in culture, CD8+ T cells were isolatedand similarly cocultured with MMC-treated B16F10 trans-fectants for 2 days. OVA-immunized CD8+ T cells also wereprepared similarly. To measure IFN-γ–producing cells,enzyme-linked immunospot (ELISPOT) method was used;B16F10 transfectants (each 2 × 105 cells) were s.c. injectedinto C57BL/6 mice. Fourteen days postinoculation, drainingLN cells were prepared and cocultured with γ-irradiatedsyngeneic spleen cells pulsed with the TAA peptide mixture(each 2 μg/mL) for 7 days. Thereafter, cells were extensivelywashed, adjusted to 2 × 106 cells/mL, 2-fold serially diluted,and seeded on a 96-well polyvinylidene difluoride membraneELISPOT plate (MultiScreenHTS-IP Filter plate). The numberof IFN-γ–producing cells was counted and calculated per2 × 106 LN cells. To examine an inhibitory effect of exo-somes on T-cell activation, CD8+ T cells (2 × 105/well) wereisolated from day 6 culture of spleen cells from pmel-1TCR transgenic mice with 1 μg/mL hgp100 peptide(EGSRNQDWL, from ANASPEC) and cocultured withMMC-treated B16F10 cells (2 × 104/well) in the presenceof indicated doses of exosomes for 2 days. IL-2 and IFN-γproduction was measured.

Cytotoxicity assayB16F10 or EL-4 tumor cells (1 × 106) were preincubated

with or without 2 μg/mL OVA257-264 or TAA peptide mixture(each 2 μg/mL) for 3 hours and then labeled with 100 μCiof Na51Cr for 1 hour. OVA-specific or TAA-immunizedT cells were added to the labeled tumor cells (1 × 104/well)at varying effector-target ratios and were incubated for4 hours. Specific 51Cr release was determined as previouslydescribed (28).

Experimental lung metastasisB16F10 transfectant cells (2 × 105) were harvested and sus-

pended in 200 μL of DPBS, and injected in the lateral tail veinof C57BL/6 mice. Lungs were harvested 11 or 14 days post-injection, and their total weight, number of metastatic foci,and melanin content were examined (29). Melanin was quan-tified in lungs as previously described (30).

Statistical analysisGroups were compared using Student's t test for both

in vitro and animal experiments.

Cancer Research

h. 8, 2020. © 2010 American Association for Cancer

Page 4: Cancer Research DC-HIL/Glycoprotein Nmb Promotes Growth of ...cancerres.aacrjournals.org/content/canres/70/14/5778.full.pdf · cle. In mice, we showed the DC-HIL/SD-4 pathway to inhibit

DC-HIL in Melanoma Immunity

Published OnlineFirst June 22, 2010; DOI: 10.1158/0008-5472.CAN-09-2538

Results

B16F10 melanoma cells express DC-HIL mRNAconstitutively at a level higher than other knowncoinhibitory ligandsTo determine the expression profile of coinhibitory ligand

genes by melanoma cells, we used reverse transcription-PCR (RT-PCR) to analyze mRNA expression of DC-HIL,PD-L1, PD-L2, herpesvirus entry mediator, and Tim-3. Bonemarrow–derived DCs served as positive control becausethese cells express all of the aforementioned ligands. DC-HIL mRNA was expressed constitutively by B16F10 melano-ma cells at a level as high as in bone marrow–derived DC(Fig. 1A); PD-L1 was present but at a much lower level, andthe other coinhibitory ligands not at all. IFN-γ treatmentdid not change the mRNA expression level of DC-HIL,whereas it upregulated mRNA expression of PD-L1 marked-ly by a level close to that of DC-HIL.

Knocked down DC-HIL expression did not changemelanoma phenotype and proliferation in vitroTo study the function of DC-HIL in melanoma, we used

siRNA to generate B16F10 cells knocked down for DC-HILexpression. We synthesized five different shRNA oligonucleo-tides and inserted each into a lentivector. The efficacy ofeach shRNA to knock down DC-HIL expression was assessedby the reduction in protein expression. The shRNA showinghighest efficacy (data not shown) was used to generate theknocked down transfectant. To avoid clonal effects, infectedB16F10 cells were enriched by repeated FACS instead of clon-ing by limiting dilution. One batch of infected B16F10 cellswas assayed by Western blotting for DC-HIL protein(Fig. 1B). Compared with control B16F10 cells (called C-B16)

www.aacrjournals.org

Researcon Januarycancerres.aacrjournals.org Downloaded from

infected with control lentivector with no shRNA sequence,DC-HIL-shRNA–infected cells (Kd-B16) showed almost noDC-HIL expression (β-actin expression was unchanged).Consistent with the very low level of total protein expres-sion, surface expression of DC-HIL was hardly detectedeven in the Kd-B16 cells treated with IFN-γ (SupplementaryFig. S1). There were no morphologic changes in Kd-B16cells, and no significant differences in the expression ofMHC class I molecules H-2Db and OVA-associated H-2Kb

between the two B16F10 transfectants (SupplementaryFig. S2). Melanin content of Kd-B16 cells was almost iden-tical to that of C-B16 cells (Supplementary Fig. S3). Finally,there was also no difference in the MTT proliferation assaybetween the two transfectants (Fig. 1C). To more preciselyanalyze growth capacity, the two B16F10 transfectants wereanalyzed for entry into the cell cycle in response to growthstimulation. Cells were synchronized by starvation andstimulated to grow by adding FCS-containing media for18 hours; cell cycle analysis was performed using BrdUrdand 7-AAD (Fig. 1D). Kd-B16 and C-B16 cells were sortedinto each cell cycle phase at close to equal frequencies.Our results indicate that knocking down DC-HIL expressionproduces no significant change in morphology, melaninsynthesis, and MHC class I expression. Moreover, theknockdown had no effect on growth in vitro nor entry intocell cycle following growth stimulation.

DC-HIL–knocked down B16F10 cells show reducedgrowth in wild-type (immunocompetent) but notimmunodeficient miceWe evaluated the tumor growth of the transfectants in syn-

geneic C57BL/6 mice (Fig. 2A). C-B16 or Kd-B16 melanomacells were inoculated s.c. into the right flank of the mice,

Figure 1. Expression of DC-HIL by B16F10 melanoma and characterization of DC-HIL–knocked down cells. Expression of DC-HIL and other coinhibitoryligands: A, total RNA was isolated from B16F10 melanoma cells nontreated or treated with IFN-γ (1,000 U/mL) plus phorbol 12-myristate 13-acetate(10 ng/mL) for 16 h or from bone marrow–derived DC and examined by RT-PCR for mRNA expression of different coinhibitory ligands and β-actin.PCR without cDNA (no cDNA) served as negative control. Characterization of DC-HIL–knocked down B16F10 cells: B16F10 cells transfected with controllentivector (C-B16) or DC-HIL–targeted shRNA-lentivector (Kd-B16) were assessed for protein expression (B), in vitro proliferation (C), and subjected tocell cycle analysis (D). B, whole-cell extracts prepared from B16F10 transfectants were assayed by Western blotting for protein expression of DC-HIL orβ-actin using UTX-103 anti–DC-HIL or anti–β-actin antibody. C, synchronized B16F10 transfectants (1 × 104 cells/well) were allowed to grow in vitro,and proliferation was measured by MTT assay; points, mean (n = 3); bars, SD. D, after starving in serum-free media, B16F10 transfectants were stimulatedfor growth by FCS and labeled with BrdUrd, and then stained with APC-anti-BrdUrd antibody and 7-AAD (total DNA content), followed by flow cytometry.Data are shown as dot plots of BrdUrd versus 7-AAD. All data are representative of at least two independent experiments.

Cancer Res; 70(14) July 15, 2010 5781

h. 8, 2020. © 2010 American Association for Cancer

Page 5: Cancer Research DC-HIL/Glycoprotein Nmb Promotes Growth of ...cancerres.aacrjournals.org/content/canres/70/14/5778.full.pdf · cle. In mice, we showed the DC-HIL/SD-4 pathway to inhibit

Tomihari et al.

Cancer Res; 70(14) July 15, 20105782

Researcon Januarycancerres.aacrjournals.org Downloaded from

Published OnlineFirst June 22, 2010; DOI: 10.1158/0008-5472.CAN-09-2538

and tumor volume was measured daily. Kd-B16 cells grew sig-nificantly slower than control cells (tumor volume of 5.08 ±0.98 cm3 for C-B16 versus 1.61 ± 0.68 cm3 for Kd-B16 at 24 dafter inoculation, P = 2 × 10−8). Mice bearing C-B16 tumorstarted to die on day 16, and all of these animals died by day26. By contrast, Kd-B16 tumor–inoculated mice started dyingon day 27, with some survivors through day 43 (Fig. 2B). Mostimportantly, Kd-B16 tumor growth in immunodeficient nu/numice was not reduced compared with controls (Fig. 2C). Wealso questioned whether concomitant inoculation of C-B16and Kd-B16 cells (mixed in equal numbers) suppresses thegrowth of s.c. injected C-B16 tumor (Fig. 2D). Indeed, thegrowth of the mixed cells was reduced significantly through-out the experiment, compared with C-B16 cells alone, but notas much as Kd-B16 alone (Fig. 2D1). Consistent with thisgrowth reduction, the survival rate of mice injected withthe mixed cells was much higher compared with mice trea-ted with C-B16 alone (Fig. 2D2). These results indicate thatreduced tumor growth produced by knocking down DC-HILrequires host immune function and that Kd-B16 cells have anadjuvant effect.To further validate our results, we examined the effect of

nonspecific siRNA on tumor growth. B16F10 cells transfectedwith nonspecific sequence (NS) siRNA (NS-B16) targeting thefirefly luciferase gene showed no change in DC-HIL protein ex-pression (Supplementary Fig. S4B). Growth of NS-B16 tumorwas similar to C-B16 by day 20 and thereafter slightly slowerthan C-B16 (Supplementary Fig. S4C). We then compared ef-fects of siRNA targeting DC-HIL versus PD-L1 gene on tumorgrowth. B16F10 cells transfected with PD-L1-siRNA showedmarkedly reduced PD-L1 expression in whole-cell extractsand on the cell surface, but β-actin and DC-HIL levels wereunchanged (Supplementary Fig. S4A and B). PD-L1–knockeddown B16F10 tumor showed growth and survival rates verysimilar to NS-B16 and C-B16, whereas in the same setting,Kd-B16 tumor had markedly reduced growth and increasedsurvival (Supplementary Fig. S4C and D). These results indi-cate that control siRNA sequences had very little to no effecton tumor growth of B16F10 cells and that knocking down DC-HIL is more effective than knocking down PD-L1 in reducingtumor growth.

DC-HIL knockdown heightens the activation ofmelanoma-reactive T cells but not susceptibility toCTL killingHaving found DC-HIL on APC to inhibit T-cell activation

after binding to SD-4 on T cells, we hypothesized thatmelanoma-associated DC-HIL exerts similar function. Wethus examined the effect of knocking down DC-HIL on theimmunogenicity of T cells for B16F10 melanoma cells. Be-cause B16F10 melanoma is inherently weak immunogenically,we used OVA as a surrogate TAA. B16F10 transfectants werepulsed with MHC class I–associated OVA peptide (OVA257-264)and then allowed to stimulate CD8+ T cells isolated from un-primed OT-I transgenic mice. T-cell activation was measuredby secretion of IL-2 and IFN-γ (Fig. 3A). Kd-B16 cells stimu-lated T cells to produce IL-2 and IFN-γ at levels 50% higherthan C-B16 cells.

Figure 2. Knockdown of DC-HIL expression reduces growth of B16F10melanoma in syngeneic wild-type (but not nu/nu) mice. Control (C-B16)or DC-HIL knockdown B16F10 cells (Kd-B16) were inoculated s.c. intothe right flank of C57BL/6 mice (12 mice/group, A and B) or nu/nu mice(5 mice/group, C), and tumor volume was measured daily. Survival rateof C57BL/6 mice injected with B16F10 transfectants is shown (B). Thetwo transfectants were also mixed (each 2 × 105 cells) and examined fortumor growth in C57BL/6 mice (n = 10; D1) and survival rate (D2).Statistical analysis (Student's t test) of these two groups on days 13 to21 indicates P = 2 × 10−8 (*) (A). P = 1.5 × 10−8 (**) and P = 0.001 (***) onday 24 document statistical significance of the mixed cells' effectcompared with C-B16 alone and Kd-B16 alone, respectively. Secondexperiment showed similar results.

Cancer Research

h. 8, 2020. © 2010 American Association for Cancer

Page 6: Cancer Research DC-HIL/Glycoprotein Nmb Promotes Growth of ...cancerres.aacrjournals.org/content/canres/70/14/5778.full.pdf · cle. In mice, we showed the DC-HIL/SD-4 pathway to inhibit

DC-HIL in Melanoma Immunity

Published OnlineFirst June 22, 2010; DOI: 10.1158/0008-5472.CAN-09-2538

We next questioned whether SD-4 is involved in the en-hanced T cell–stimulatory capacity. OVA-specific CD8+ CTLwere generated by immunizing OT-I mice with OVA peptidefollowed by in vitro reactivation, and were finally examinedby flow cytometry for SD-4 expression (Fig. 3B). All CTL ex-pressed the CD69 activation marker, and half of CD69+ CTLcoexpressed SD-4. To examine the involvement of SD-4 inT-cell immunogenicity by melanoma cells, we assayed the ac-tivation of CD8+ T cells by parental B16F10 cells in the pre-sence of soluble anti–SD-4 mAb (to block endogenous SD-4function on CTL) or control IgG (Fig. 3C). OVA-pulsed paren-tal melanoma cells stimulated CD8+ T cells to secrete IL-2 ata level similar to that in C-B16 cells. This IL-2 secretion wasenhanced by addition of anti-SD-4 mAb (but not by controlIgG) in a dose-dependent manner.

www.aacrjournals.org

Researcon Januarycancerres.aacrjournals.org Downloaded from

We next assessed the susceptibility of B16F10 transfec-tants to lysis by CTL (Fig. 3D). OVA-specific CD8+ CTL lysedOVA-pulsed C-B16 very efficiently (50% target lysis), but notunpulsed B16F10 cells. These CTL also were capable of lysingOVA-pulsed Kd-B16 cells, albeit at a lower efficiency (insig-nificant difference, P < 0.18). Thus, both control and DC-HIL–knocked down B16F10 cells seem to be similarly sensitive toCTL cytotoxicity.To ascertain the effect of knocking down DC-HIL, we used

hgp100 and TRP-2 antigen that have been used as TAA inantimelanoma immunotherapy (31, 32). CD8+ CTL were pre-pared by in vitro restimulation of LN cells isolated fromC57BL per six mice immunized with the two peptides, andwere finally cocultured with C-B16 or Kd-B16 cells (Fig. 4).Consistent with results of studies using OVA peptide, CTLs

Figure 3. Blockade of DC-HIL/SD-4 pathway leads to enhanced T-cell immunogenicity. A, OVA-specific CD8+ T cells were stimulated by coculturing withOVA-pulsed/MMC-treated B16F10 transfectants, and T-cell activation was assessed by production of IL-2 and IFN-γ. Statistical significance is P = 0.05 (*)and P = 0.01 (**), compared with the production by T cells treated with C-B16 cells. B, anti-OVA CTL were generated by immunization and subsequentin vitro stimulation, and examined by flowcytometry for surface expression of SD-4 and CD69 (activation marker). C, OVA-specific CD8+ T cells werestimulated by OVA-pulsed/MMC-treated B16F10 melanoma in the absence (none) or presence of control IgG or anti-SD-4 mAb at different doses. T-cellactivation was assessed by IL-2 production. D, anti-OVA CTL were allowed to kill target cells at varying effector-target ratios (E/T). Target cells includeOVA-pulsed Kd-B16 (• with solid lines), OVA-pulsed C-B16 (Δ with solid lines), nontreated Kd-B16 (• with dashed lines), and nontreated C-B16 cells(Δ with dashed lines). Statistical significance scores are P = 0.20 (*), P = 0.18 (**), and P = 0.58 (***). Cytotoxicity is expressed as % of lysis. All data arerepresentative of three separate experiments.

Figure 4. DC-HIL knockdown enhances the capacity to stimulate anti-TAA T cells. A, anti-hgp100/TRP-2 CTL were stimulated by C-B16 or Kd-B16melanoma cells, and IFN-γ production was measured. B, these CTL were cocultured with 51Cr-labeled B16F10 transfectants or EL-4 as H-2–mismatchedcontrol, at varying effector-target (E/T) ratios. Statistical significance scores are P = 0.59 (*), P = 0.69 (**), and P = 0.77 (***). C, 2 wk after C57BL/6mice (n = 7) were tumor challenged with B16F10 transfectants, draining LN cells were prepared from treated mice and restimulated in vitro with γ-irradiatedsyngeneic spleen cells pulsed with hgp100 and TRP-2 peptides. Finally, IFN-γ–producing cells were counted by ELISPOT per 2 × 106 LN cells.

Cancer Res; 70(14) July 15, 2010 5783

h. 8, 2020. © 2010 American Association for Cancer

Page 7: Cancer Research DC-HIL/Glycoprotein Nmb Promotes Growth of ...cancerres.aacrjournals.org/content/canres/70/14/5778.full.pdf · cle. In mice, we showed the DC-HIL/SD-4 pathway to inhibit

Tomihari et al.

5784

Published OnlineFirst June 22, 2010; DOI: 10.1158/0008-5472.CAN-09-2538

were more strongly activated by Kd-B16 cells than by controlcells (Fig. 4A). Again, there was no significant difference insusceptibility to cell lysis by hgp100/TRP-2–specific CTL(Fig. 4B). Using these TAA mouse models, we assayed IFN-γ–producing cells from the LN of mice challenged withB16F10 tumors (Fig. 4C). C57BL/6 mice were tumor chal-lenged by s.c. injection of B16F10 cells, 2 weeks after whichLN cells were prepared and restimulated in vitro with spleencells pulsed with hgp100 and TRP-2 peptides. IFN-γ–secretingcells were counted by ELISPOT assay. Mice challenged withKd-B16 cells produced a significantly higher number of IFN-γ–secreting LN cells than control cells. Altogether, theseresults indicate that knocking down DC-HIL heightens the ca-pacity of B16F10 cells to activate tumor-reactive T cells butdoes not significantly alter susceptibility to CTL lysis.

B16F10-released exosomes contain DC-HILthat inhibits T-cell activation triggered bymelanoma antigenBecause melanoma cells release exosomes (extracellular

melanosomes or microvesicles) that promote immunosup-pression (33), we examined the possibility that DC-HIL is alsopacked into exosomes that can inhibit T-cell activation(Fig. 5). Exosomes prepared from C-B16 cells containedDC-HIL protein at a high level, and those of Kd-B16 cells alsocontained it but at an extremely low level, consistent withexpression in the whole-cell extracts (Fig. 1B). By contrast,both exosome preparations expressed similar levels of themelanosomal protein gp100, indicating that similar amountsof exosomes were examined. We then questioned whetherthese exosomes inhibit activation of CD8+ T cells (expressingSD-4) by melanoma (Fig. 5B). SD-4 CD8+ T cells wereprepared by in vitro culturing spleen cells from pmel-1 TCRtransgenic mice in which all CD8+ T cells express transgenicTCR that recognizes pmel-17 (mouse homologue of humangp100) in the context of the H-2Db (34). These cells expressedSD-4 at a level as high as OT-I CD8+ T cells (data not shown).B16F10 parental cells were allowed to stimulate the pmel-1CD8+ T cells in the presence of exosomes prepared fromC-B16 or Kd-B16 cells (Fig. 5B). Addition of exosomes

Cancer Res; 70(14) July 15, 2010

Researcon Januarycancerres.aacrjournals.org Downloaded from

(200 μg/mL) from C-B16 cells led to 80% reduction in IL-2production by peml-1 T cells (50% with 100 μg/mL), whereasexosomes from Kd-B16 cells had no inhibitory effect on IL-2production. Similar results were noted for effects on IFN-γproduction (Fig. 5B).

DC-HIL knockdown does not alter the capacity ofmelanoma to spread to the lung, but it reduces growthof melanoma within lung fociWe next examined the effect of DC-HIL knockdown on

the dissemination of B16F10 melanoma to the lung followingi.v. injection into syngeneic hosts (Fig. 6). Two weeks afterinfusing B16F10 transfectants into C57BL/6 mice throughthe tail vein, mice were sacrificed and lungs were assayedfor weight, number of foci, and melanin content (exp. 1).Comparing the lungs of mice injected with C-B16 cells tothose injected with Kd-B16 cells, the latter were lighter(0.21 ± 0.02 g versus 0.20 ± 0.02 g, P = 0.23) and containedmore lung foci (2,091 ± 240 versus 2,333 ± 261, P = 0.12),although these differences were not statistically significant.However, lung foci for Kd-B16–injected mice were smaller.Because the two B16F10 transfectants contain almost identi-cal melanin content (Supplementary Fig. S2), we also evaluat-ed melanin content, and found that for Kd-B16 cells, totalmelanin content per lung was lower (846 ± 293 μg versus611 ± 112 μg, P = 0.11) and melanin per focus was significantlyless (0.40 ± 0.11 versus 0.27 ± 0.05, P = 0.03). A second exper-iment (exp. 2) reproduced the latter finding (0.68 ± 0.12 versus0.43 ± 0.21, P = 0.04). These results indicate that the effect ofknocking down DC-HIL in B16F10 cells is manifest, not in thecapacity for establishment in lung following i.v. injection, butin their potential for growth within lung foci.

Discussion

To determine whether melanoma-associated DC-HIL inhi-bits the activation of tumor-reactive T cells, we knockeddown DC-HIL on B16F10 mouse melanoma cells and analyzedits effects on tumor growth. Knocking down DC-HIL had noeffect on spontaneous growth nor on proliferation following

Figure 5. DC-HIL+ exsosomes from B16F10 cells inhibit T-cell activation triggered by TAA on melanoma. A, exosomes prepared from C-B16 or Kd-B16cells were examined by immunoblotting for expression of DC-HIL and gp100 (as a melanosomal marker). B, these exosomes were added to thecoculture of activated CD8+ T cells (from pmel-1 TCR transgenic mice) and MMC-treated B16F10 cells. T-cell activation was measured by productionof IL-2 (left) or IFN-γ (right). Columns, mean (n = 3); bars, SD. *, P > 0.001.

Cancer Research

h. 8, 2020. © 2010 American Association for Cancer

Page 8: Cancer Research DC-HIL/Glycoprotein Nmb Promotes Growth of ...cancerres.aacrjournals.org/content/canres/70/14/5778.full.pdf · cle. In mice, we showed the DC-HIL/SD-4 pathway to inhibit

DC-HIL in Melanoma Immunity

Published OnlineFirst June 22, 2010; DOI: 10.1158/0008-5472.CAN-09-2538

growth stimulation in vitro. It also had no effect on melanomagrowth in immunodeficient nude mice. However, it reducedgrowths of flank and lung B16F10 melanomas that were es-tablished, respectively, following s.c. and i.v. injections intosyngeneic immunocompetent mice. These tumor growth re-ductions are due most likely to the inhibitory function of DC-HIL on melanoma-reactive T cells because: (a) knockingdown DC-HIL in B16F10 cells enhanced the capacity of thesecells to stimulate melanoma-reactive T cells; (b) IFN-γ–secreting effector T cells were generated more in mice chal-lenged with the knocked down cells; (c) SD-4 is expressed bymelanoma-reactive CTL; and (d) parental B16F10 melanomaenhanced activation of CTL when SD-4 function is blocked.By contrast, we were unable to show any significant effect ofDC-HIL on susceptibility to CTL-mediated lysis or on the abil-ity of melanoma to become established in lung following i.v.injection. Moreover, the inhibitory function of DC-HIL maybe delivered to lymphoid organs distal to the melanoma sitethrough exosomes. Altogether, our findings indicate that DC-HIL confers on melanoma the ability to inhibit the activationof melanoma-reactive T cells, thereby compromising optimalantitumor immunity.The Gpnmb/DC-HIL gene was identified first by Weterman

and colleagues (15) using subtractive cDNA cloning betweenhighly and lowly metastatic human melanoma cell lines. Tostudy Gpnmb function, they transfected the gene into a high-ly metastatic human melanoma BLM line (originally devoidof Gpnmb expression) and established transgenic BLMclones. They reported no difference in in vitro growth be-tween transfectants and controls but did not show data intheir article (15). These results are consistent with oursshowing no effect of DC-HIL knockdown on proliferationas assessed by MTT assay and cell cycle analysis. However,

www.aacrjournals.org

Researcon Januarycancerres.aacrjournals.org Downloaded from

they also reported that two of their three transfectant clonesshowed reduced subcutaneous growth when inoculated intonude mice, and one clone exhibited reduced spontaneous me-tastasis in nude mice, suggesting a downregulatory role forGpnmb in melanoma growth and metastatic potential. Differ-ences between our and their experimental systems may ac-count for disparate implications: (a) mouse versus humanmelanoma cell lines; (b) bulk versus cloned transfectants; (c)Gpnmb protein expression was not examined in their BLMtransfectants; (d) knockdown (loss-of-function) strategy byus versus transgenic expression (gain-of-function) by them;and (e) i.v. injection of B16F10 transfectants into syngeneic im-munocompetent mice by us versus xenografts of human mel-anoma into nude mice by them (thereby excluding a role forhost immune responses in their studies). At a minimum, thesedifferences render a direct comparison of outcomes difficult.At best, our demonstration that the DC-HIL/SD-4 pathway fa-vors melanoma growth is an opportunity for investigating itspotential usefulness as a target for immunotherapy.Among coinhibitors of T-cell activation, DC-HIL most

closely resembles PD-L1 in its function (19). PD-L1 is ex-pressed by different cancers and is implicated in immuneevasion by tumors (35, 36): Transgenic expression of PD-L1into P815 mastocytoma (lacking PD-L1 expression) promotesits growth in syngeneic immunocompetent (but not nu/nu)mice by reducing stimulation of CTL and susceptibility to tu-mor lysis by CTL (13, 37). PD-L1 also is expressed constitu-tively by B16F10 melanoma but at a much lower level thanDC-HIL (37), although this expression can be upregulatedmarkedly by treatment with IFN-γ in vitro, and most recentlyshown in a subcutaneous B16F10 tumor in mice (38). How-ever, the role of the PD-L1 pathway in melanoma growth isnot clear because of two discrepant data: In mice deficient

Figure 6. Capacity of B16F10 transfectants to spread to the lung and grow in the lung foci. C57BL/6 mice (n = 6 for exp. 1 and n = 5 for exp. 2) were givenB16F10 transfectants by i.v. injection. Fourteen (exp. 1) or 11 (exp. 2) d postinjection, lungs were procured; weight, number of metastatic foci, totalmelanin content, and melanin content per focus were measured. Statistical significance (P value) is shown. Right, photographs of lungs from miceinjected with B16F10 transfectants. Scale bar, 50 mm. Images of some foci are enlarged.

Cancer Res; 70(14) July 15, 2010 5785

h. 8, 2020. © 2010 American Association for Cancer

Page 9: Cancer Research DC-HIL/Glycoprotein Nmb Promotes Growth of ...cancerres.aacrjournals.org/content/canres/70/14/5778.full.pdf · cle. In mice, we showed the DC-HIL/SD-4 pathway to inhibit

Tomihari et al.

5786

Published OnlineFirst June 22, 2010; DOI: 10.1158/0008-5472.CAN-09-2538

for PD-1 (the TCR for PD-L1), B16F10 melanoma cells grew ata rate similar to that in wild-type mice (37), whereas inocu-lation of melanoma cells into mice overexpressing PD-1markedly enhanced their growth (38). We probed the influ-ence of PD-L1 on B16F10 melanoma cells and found no sig-nificant effect on subcutaneous tumor growth and survival oftumor-bearing mice, consistent with previous work in PD-1–deficient mice, and supportive of DC-HIL being more impor-tant than PD-L1 in this setting.We showed that DC-HIL is enclosed in exosomes released

by B16F10 cells, which inhibit T-cell activation triggered by amelanoma-associated antigen pmel-17 (mouse homologue ofhuman gp100). Exosomes have been shown at abundant le-vels in body fluids of patients with melanoma (39), and theycan mediate immunosuppression by promoting differentia-tion of monocytes to myeloid suppressor cells (which facili-tate tumor growth; ref. 40) and by inhibiting T-cell function(41). It is likely that DC-HIL is involved in exosome-mediatedimmunosuppresion. Importantly, the inhibitory function ofDC-HIL can be delivered as a form of exosome to lymphoidorgans (including LN, spleen, and bone marrow) distal to themelanoma site.

Cancer Res; 70(14) July 15, 2010

Researcon Januarycancerres.aacrjournals.org Downloaded from

In summary, our findings provide a rationale basis for de-veloping treatment modalities targeting the DC-HIL/SD-4pathway to optimize immune surveillance of melanomaand control its growth.

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

Acknowledgments

We thank Irene Dougherty for the technical assistance and Marguerite Starrfor the secretary assistance.

Grant Support

VA Merit Award and NIH R01 (AI064927).The costs of publication of this article were defrayed in part by the payment

of page charges. This article must therefore be hereby marked advertisement inaccordance with 18 U.S.C. Section 1734 solely to indicate this fact.

Received 07/09/2009; revised 04/14/2010; accepted 05/05/2010; publishedOnlineFirst 06/22/2010.

References

1. Thor SP, Becker JC, Guldberg P, Zeuthen J. In situ T cells in mela-

noma. Cancer Immunol Immunother 1999;48:386–95.2. Rivoltini L, Carrabba M, Huber V, et al. Immunity to cancer: attack

and escape in T lymphocyte-tumor cell interaction. Immunol Rev2002;188:97–113.

3. Chouaib S, Asselin-Paturel C, Caignard A, Blay JY. The host-tumorimmune conflict: from immunosuppression to resistance and de-struction. Immunol Today 1997;18:493–7.

4. Carreno BM, Collins M. The B7 family of ligands and its receptors:new pathways for costimulation and inhibition of immune responses.Annu Rev Immunol 2002;20:29–53.

5. Dong H, Zhu G, Tamada K, Chen L. B7-1, a third member of the B7family, co-stimulates T-cell proliferation and interleukin-10 secretion.Nat Med 1999;5:1365–9.

6. Freeman GJ, Long AJ, Iwai Y, et al. Engagement of the PD-1 im-munoinhibitory receptor by a novel B7 family member leads to neg-ative regulation of lymphocyte activation. J Exp Med 2000;192:1027–34.

7. Latchman Y, Wood CR, Chernova T, et al. PD-L2 is a second li-gand for PD-1 and inhibits T cell activation. Nat Immunol 2001;2:261–8.

8. Carreno BM, Collins M. BTLA: a new inhibitory receptor with a B7-like ligand. Trends Immunol 2003;24:524–7.

9. Zhu C, Anderson AC, Schubart A, et al. The Tim-3 ligand galectin-9negatively regulates T helper type 1 immunity. Nat Immunol 2005;6:1245–52.

10. Sabatos CA, Chakravarti S, Cha E, et al. Interaction of Tim-3 andTim-3 ligand regulates T helper type 1 responses and induction ofperipheral tolerance. Nat Immunol 2003;4:1102–10.

11. Sanchez-Fueyo A, Tian J, Picarella D, et al. Tim-3 inhibits T helpertype 1-mediated auto- and alloimmune responses and promotes im-munological tolerance. Nat Immunol 2003;4:1093–101.

12. Hurwitz AA, Kwon ED, van EA. Costimulatory wars: the tumor men-ace. Curr Opin Immunol 2000;12:589–96.

13. Dong H, Strome SE, Salomao DR, et al. Tumor-associated B7-1 pro-motes T-cell apoptosis: a potential mechanism of immune evasion.Nat Med 2002;8:793–800.

14. Shikano S, Bonkobara M, Zukas PK, Ariizumi K. Molecular cloning ofa dendritic cell-associated transmembrane protein, DC-HIL, that pro-motes RGD-dependent adhesion of endothelial cells through recog-

nition of heparan sulfate proteoglycans. J Biol Chem 2001;276:8125–34.

15. Weterman MA, Ajubi N, van Dinter I, Degen WG, Ruitter DJ, BloemersHP. nmb, a novel gene, is expressed in low-metastatic human mela-noma cell lines and xenografts. Int J Cancer 1995;60:73–81.

16. Safadi FF, Xu J, Smock SL, Rico MC, Owen TA, Popoff SN. Cloningand characterization of osteoactivin, a novel cDNA expressed in os-teoblasts. J Cell Biol 2001;84:12–26.

17. Metz RL, Patel PS, Hameed M, Bryan M, Rameshwar P. Role of hu-man HGFIN/nmb in breast cancer. Breast Cancer Res 2007;9:R58.

18. Ponting CP, Hofmann K, Bork P. A latrophilin/CL-1-like GPS domainin polycystin-1. Curr Biol 1999;9:R585–8.

19. Chung JS, Sato K, Dougherty I, Cruz PD, Jr., Ariizumi K. DC-HIL is anegative regulator of T lymphocyte activation. Blood 2007;109:4320–7.

20. Chung JS, Dougherty I, Cruz PD, Jr., Ariizumi K. Syndecan-4mediates the coinhibitory function of DC-HIL on T cell activation.J Immunol 2007;179:5778–84.

21. Chung JS, Bonkobara M, Tomihari M, Cruz PD, Jr., Ariizumi K. TheDC-HIL/syndecan-4 pathway inhibits human allogeneic T-cell re-sponses. Eur J Immunol 2009;39:965–74.

22. Ariizumi K, Meng Y, Bergstresser PR, Takashima A. IFN-γ-dependentIL-7 gene regulation in keratinocytes. J Immunol 1995;154:6031–9.

23. Zufferey R, Nagy D, Mandel RJ, Naldini L, Trono D. Multiply attenu-ated lentiviral vector achieves efficient gene delivery in vivo. Nat Bio-technol 1997;15:871–5.

24. Sato K, Shikano S, Xia G, et al. Selective expression of vacuolar H+-ATPase subunit d2 by particular subsets of dendritic cells amongleukocytes. Mol Immunol 2006;43:1443–53.

25. Tomihari M, Hwang SH, Chung JS, Cruz PD, Jr., Ariizumi K. Gpnmbis a melanosome-associated glycoprotein that contributes to mela-nocyte/keratinocyte adhesion in a RGD-dependent fashion. Exp Der-matol 2009;18:586–95.

26. Rubinstein N, Alvarez M, Zwirner NW, et al. Targeted inhibition ofgalectin-1 gene expression in tumor cells results in heightened Tcell-mediated rejection; A potential mechanism of tumor-immuneprivilege. Cancer Cell 2004;5:241–51.

27. Araki K, Horikawa T, Chakraborty AK, et al. Small Gtpase rab3A isassociated with melanosomes in melanoma cells. Pigment Cell Res2000;13:332–6.

Cancer Research

h. 8, 2020. © 2010 American Association for Cancer

Page 10: Cancer Research DC-HIL/Glycoprotein Nmb Promotes Growth of ...cancerres.aacrjournals.org/content/canres/70/14/5778.full.pdf · cle. In mice, we showed the DC-HIL/SD-4 pathway to inhibit

DC-HIL in Melanoma Immunity

Published OnlineFirst June 22, 2010; DOI: 10.1158/0008-5472.CAN-09-2538

28. Harada M, Tamada K, Abe K, et al. Characterization of B16 melano-ma-specific cytotoxic T lymphocytes. Cancer Immunol Immunother1998;47:198–204.

29. Toura I, Kawano T, Akutsu Y, Nakayama T, Ochiai T, Taniguchi M.Cutting edge: inhibition of experimental tumor metastasis by dendrit-ic cells pulsed with α-galactosylceramide. J Immunol 1999;163:2387–91.

30. Reischer D, Heyfets A, Shimony S, Nordenberg J, Kashman Y,Flescher E. Effects of natural and novel synthetic jasmonates inexperimental metastatic melanoma. Br J Pharmacol 2007;150:738–49.

31. Overwijk WW, Tsung A, Irvine KR, et al. gp100/pmel 17 is a murinetumor rejection antigen: induction of “self”-reactive, tumoricidal Tcells using high-affinity, altered peptide ligand. J Exp Med 1998;188:277–86.

32. Mendiratta SK, Thai G, Eslahi NK, et al. Therapeutic tumor immunityinduced by polyimmunization with melanoma antigens gp100 andTRP-2. Cancer Res 2001;61:859–63.

33. Valenti R, Huber V, Iero M, Filipazzi P, Parmiani G, Rivoltini L. Tumor-released microvesicles as vehicles of immunosuppression. CancerRes 2007;67:291–5.

34. Overwijk WW, Theoret MR, Finkelstein SE, et al. Tumor regression

www.aacrjournals.org

Researcon Januarycancerres.aacrjournals.org Downloaded from

and autoimmunity after reversal of a functionally tolerant state ofself-reactive CD8+ T cells. J Exp Med 2003;198:569–80.

35. Dong H, Chen L. B7-1 pathway and its role in the evasion of tumorimmunity. J Mol Med 2003;81:281–7.

36. Okazaki T, Iwai Y, Honjo T. New regulatory co-receptors: inducibleco-stimulator and PD-1. Curr Opin Immunol 2002;14:779–82.

37. Iwai Y, Ishida M, Tanaka Y, Okazaki T, Honjo T, Minato N. Involve-ment of PD-L1 on tumor cells in the escape from host immune sys-tem and tumor immunotherapy by PD-L1 blockade. Proc Natl AcadSci U S A 2002;99:12293–7.

38. Iwai Y, Terawaki S, Honjo T. PD-1 blockade inhibits hematogenousspread of poorly immunogenic tumor cells by enhanced recruitmentof effector T cells. Int Immunol 2005;17:133–44.

39. Andre F, Schartz NEC, Movassagh M, et al. Malignant effusions andimmunogenic tumour-derived exosomes. Lancet 2002;360:295–305.

40. Valenti R, Huber V, Filipazzi P, et al. Human tumor-released micro-vesicles promote the differentiation of myeloid cells with transform-ing growth factor-β-mediated suppressive activity on T lymphocytes.Cancer Res 2006;66:9290–8.

41. Andreola G, Rivoltini L, Castelli C, et al. Induction of lymphocyte ap-optosis by tumor cell secretion of FasL-bearing microvesicles. J ExpMed 2002;195:1303–16.

Cancer Res; 70(14) July 15, 2010 5787

h. 8, 2020. © 2010 American Association for Cancer

Page 11: Cancer Research DC-HIL/Glycoprotein Nmb Promotes Growth of ...cancerres.aacrjournals.org/content/canres/70/14/5778.full.pdf · cle. In mice, we showed the DC-HIL/SD-4 pathway to inhibit

2010;70:5778-5787. Published OnlineFirst June 22, 2010.Cancer Res   Mizuki Tomihari, Jin-Sung Chung, Hideo Akiyoshi, et al.   Mice by Inhibiting the Activation of Tumor-Reactive T CellsDC-HIL/Glycoprotein Nmb Promotes Growth of Melanoma in

  Updated version

  10.1158/0008-5472.CAN-09-2538doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://cancerres.aacrjournals.org/content/suppl/2010/06/18/0008-5472.CAN-09-2538.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://cancerres.aacrjournals.org/content/70/14/5778.full#ref-list-1

This article cites 41 articles, 12 of which you can access for free at:

  Citing articles

  http://cancerres.aacrjournals.org/content/70/14/5778.full#related-urls

This article has been cited by 4 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] at

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

  Permissions

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

.http://cancerres.aacrjournals.org/content/70/14/5778To request permission to re-use all or part of this article, use this link

Research. on January 8, 2020. © 2010 American Association for Cancercancerres.aacrjournals.org Downloaded from

Published OnlineFirst June 22, 2010; DOI: 10.1158/0008-5472.CAN-09-2538