activationoftumor-specificcd8 tcellsafterintratumoral ad5 ...ad5-mtrail (109 pfu), and/or cpg odn...

12
Activation of Tumor-Specific CD8 + T Cells after Intratumoral Ad5-TRAIL/CpG Oligodeoxynucleotide Combination Therapy Rebecca L. VanOosten and Thomas S. Griffith Department of Urology and Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa Abstract CD8 + T-cell activation via cross-presentation of antigens from apoptotic tumor cells is controversial. Dendritic cells capture naturally shed tumor antigens and cross-present them to CD8 + T cells; unfortunately, the frequency of activated CD8 + T cells is often too low to mount an effective response against the tumor. By increasing the amount of antigen for presentation, a larger T-cell response can be theoretically elicited. We used a recombinant adenovirus encoding full-length murine tumor necrosis factor–related apoptosis-inducing ligand (Ad5- mTRAIL) to induce tumor cell apoptosis, and when given intratumorally to mice bearing experimental renal cell carcinoma (Renca) tumors, Ad5-mTRAIL minimally prolonged survival and induced a low level of CTL activity. To enhance dendritic cell efficiency, an immunostimulatory CpG oligo- deoxynucleotide (CpG ODN) was combined with Ad5-mTRAIL. This combination therapy significantly augmented in vivo antigen-specific T-cell proliferation and CTL activity, as well as prolonged survival of Renca tumor-bearing mice. Interest- ingly, depletion of CD4 + or CD25 + cells before therapy further enhanced survival and in vivo CTL activity. In addition, tumor-free mice depleted of CD4 + cells were also able to reject a subsequent challenge of Renca cells, but not MHC-matched RM-11 prostate tumor cells, demonstrating the existence of immunologic memory. These results collectively show that local treatment with Ad5-mTRAIL and CpG ODN can augment tumor antigen cross-presentation resulting in T-cell prolifer- ation, enhanced CTL activity, and increased animal survival. [Cancer Res 2007;67(24):11980–90] Introduction Peptides derived from endogenously expressed proteins are presented by antigen-presenting cells (APC) in the context of MHC class I (MHC I) to CD8 + T cells, whereas peptides obtained from exogenously derived proteins are normally loaded onto MHC class II (MHC II) for presentation to CD4 + T cells. Exogenous antigens can be also loaded onto MHC I for ‘‘cross-presentation’’ to CD8 + T cells (1–3), which is believed to occur after the phagocytosis of apoptotic cells, dendritic cell sampling of live cells, or the shuttling of proteins to dendritic cells by heat shock proteins (4). Through cross-presentation, two outcomes can occur: ( a ) cross-priming and CD8 + T-cell activation, induction of cellular proliferation, and acquisition of effector function; or (b) there is no activation of CD8 + T cells with cross-tolerance, and the T cells either become anergic and/or are deleted (4). In tumor-draining lymph nodes, both cross-priming and cross- tolerance have been detected in different tumor models. In experiments where cross-priming was observed, tumor antigen– specific T-cell proliferation was detected, but the numbers of T cells proliferating were very low, and the overall effect of CD8 + T-cell activation did not inhibit tumor outgrowth (5). Furthermore, when an apoptosis-inducing agent, gemcitabine, was used as a therapy in these studies, an increase in T-cell proliferation was detected and tumor clearance was observed. This indicates that cross-priming was occurring in the presence of apoptosis, and that the cross-priming could be augmented by increasing the amount of tumor antigen available for presentation. In contrast, studies with other tumor models have reported no T-cell proliferation, and the T cells become anergic and/or deleted after time (6). This study used an adoptive transfer model of hemagglutinin-specific CD8 + T cells (clone 4 T cells; ref. 7), and the number of cells transferred altered the outcome of the experiments. In these experiments, the greater the tumor burden, the faster the CD8 + T cells became tolerized and deletion occurred. The authors suggested that when greater T-cell numbers were transferred, they underwent autoactivation; whereas when lower T-cell numbers were transferred, the T cells become tolerized due to the lack of an inflammatory environment. This titration of antigen-specific T cells could be a major underlying difference in the experiments performed by various groups studying tumor antigen cross- presentation and the induction of activation or tolerance. These conflicting results also make it important to determine if apoptosis-based immunotherapy will benefit from tumor antigen cross-priming. Tumor necrosis factor–related apoptosis inducing ligand (TRAIL) potently induces tumor cell apoptosis, but has no cyto- toxic activity against normal cells and tissues (8, 9). Previously, we described the development of a recombinant adenoviral vector encoding the TRAIL cDNA (Ad5-TRAIL) to allow for prolonged TRAIL production (10, 11). However, it remains unknown whether antigens derived from Ad5-TRAIL–generated apoptotic tumor cells can be cross-presented to CD8 + T cells, facilitating cross-priming. The experiments described in this report examine the hypothesis that increasing the amount of tumor antigen available for dendritic cells to phagocytize via Ad5-TRAIL–induced apoptosis will allow for increased cross-priming and CD8 + T-cell activation, leading to increased tumor-specific CTL activity and tumor clearance for prolonged survival. Materials and Methods Mice and reagents. BALB/c (H-2 d ) mice were purchased from The Jackson Laboratory. Clone 4 and InsHA (12) transgenic mice were obtained from Dr. Linda Sherman (The Scripps Research Institute, La Jolla, CA). h2m / mice were provided by Dr. Kevin L. Legge (University of Iowa, Iowa City, IA). CpG oligodeoxynucleotide (CpG ODN) 1826 was purchased from Requests for reprints: Thomas S. Griffith, Department of Urology, University of Iowa, 3204 MERF, 375 Newton Road, Iowa City, IA 52242-1089. Phone: 319-335-7581; Fax: 319-353-4556; E-mail: [email protected]. I2007 American Association for Cancer Research. doi:10.1158/0008-5472.CAN-07-1526 Cancer Res 2007; 67: (24). December 15, 2007 11980 www.aacrjournals.org Research Article Research. on August 16, 2020. © 2007 American Association for Cancer cancerres.aacrjournals.org Downloaded from

Upload: others

Post on 11-Jul-2020

4 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: ActivationofTumor-SpecificCD8 TCellsafterIntratumoral Ad5 ...Ad5-mTRAIL (109 pfu), and/or CpG ODN (100 Ag) were injected i.t. After another 7 days, the tumors were harvested and physically

Activation of Tumor-Specific CD8+ T Cells after Intratumoral

Ad5-TRAIL/CpG Oligodeoxynucleotide Combination Therapy

Rebecca L. VanOosten and Thomas S. Griffith

Department of Urology and Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa

Abstract

CD8+ T-cell activation via cross-presentation of antigens fromapoptotic tumor cells is controversial. Dendritic cells capturenaturally shed tumor antigens and cross-present them to CD8+

T cells; unfortunately, the frequency of activated CD8+ T cells isoften too low to mount an effective response against thetumor. By increasing the amount of antigen for presentation,a larger T-cell response can be theoretically elicited. We useda recombinant adenovirus encoding full-length murine tumornecrosis factor–related apoptosis-inducing ligand (Ad5-mTRAIL) to induce tumor cell apoptosis, and when givenintratumorally to mice bearing experimental renal cellcarcinoma (Renca) tumors, Ad5-mTRAIL minimally prolongedsurvival and induced a low level of CTL activity. To enhancedendritic cell efficiency, an immunostimulatory CpG oligo-deoxynucleotide (CpG ODN) was combined with Ad5-mTRAIL.This combination therapy significantly augmented in vivoantigen-specific T-cell proliferation and CTL activity, as wellas prolonged survival of Renca tumor-bearing mice. Interest-ingly, depletion of CD4+ or CD25+ cells before therapy furtherenhanced survival and in vivo CTL activity. In addition,tumor-free mice depleted of CD4+ cells were also able to rejecta subsequent challenge of Renca cells, but not MHC-matchedRM-11 prostate tumor cells, demonstrating the existence ofimmunologic memory. These results collectively show thatlocal treatment with Ad5-mTRAIL and CpG ODN can augmenttumor antigen cross-presentation resulting in T-cell prolifer-ation, enhanced CTL activity, and increased animal survival.[Cancer Res 2007;67(24):11980–90]

Introduction

Peptides derived from endogenously expressed proteins arepresented by antigen-presenting cells (APC) in the context of MHCclass I (MHC I) to CD8+ T cells, whereas peptides obtained fromexogenously derived proteins are normally loaded onto MHC classII (MHC II) for presentation to CD4+ T cells. Exogenous antigenscan be also loaded onto MHC I for ‘‘cross-presentation’’ to CD8+

T cells (1–3), which is believed to occur after the phagocytosis ofapoptotic cells, dendritic cell sampling of live cells, or the shuttlingof proteins to dendritic cells by heat shock proteins (4). Throughcross-presentation, two outcomes can occur: (a) cross-priming andCD8+ T-cell activation, induction of cellular proliferation, andacquisition of effector function; or (b) there is no activation ofCD8+ T cells with cross-tolerance, and the T cells either becomeanergic and/or are deleted (4).

In tumor-draining lymph nodes, both cross-priming and cross-tolerance have been detected in different tumor models. Inexperiments where cross-priming was observed, tumor antigen–specific T-cell proliferation was detected, but the numbers of Tcells proliferating were very low, and the overall effect of CD8+

T-cell activation did not inhibit tumor outgrowth (5).Furthermore, when an apoptosis-inducing agent, gemcitabine,was used as a therapy in these studies, an increase in T-cellproliferation was detected and tumor clearance was observed.This indicates that cross-priming was occurring in the presenceof apoptosis, and that the cross-priming could be augmentedby increasing the amount of tumor antigen available forpresentation. In contrast, studies with other tumor models havereported no T-cell proliferation, and the T cells become anergicand/or deleted after time (6). This study used an adoptivetransfer model of hemagglutinin-specific CD8+ T cells (clone 4T cells; ref. 7), and the number of cells transferred alteredthe outcome of the experiments. In these experiments, thegreater the tumor burden, the faster the CD8+ T cells becametolerized and deletion occurred. The authors suggested thatwhen greater T-cell numbers were transferred, they underwentautoactivation; whereas when lower T-cell numbers weretransferred, the T cells become tolerized due to the lack of aninflammatory environment. This titration of antigen-specific Tcells could be a major underlying difference in the experimentsperformed by various groups studying tumor antigen cross-presentation and the induction of activation or tolerance. Theseconflicting results also make it important to determine ifapoptosis-based immunotherapy will benefit from tumor antigencross-priming.

Tumor necrosis factor–related apoptosis inducing ligand(TRAIL) potently induces tumor cell apoptosis, but has no cyto-toxic activity against normal cells and tissues (8, 9). Previously,we described the development of a recombinant adenoviral vectorencoding the TRAIL cDNA (Ad5-TRAIL) to allow for prolongedTRAIL production (10, 11). However, it remains unknown whetherantigens derived from Ad5-TRAIL–generated apoptotic tumor cellscan be cross-presented to CD8+ T cells, facilitating cross-priming.The experiments described in this report examine the hypothesisthat increasing the amount of tumor antigen available for dendriticcells to phagocytize via Ad5-TRAIL–induced apoptosis will allowfor increased cross-priming and CD8+ T-cell activation, leading toincreased tumor-specific CTL activity and tumor clearance forprolonged survival.

Materials and Methods

Mice and reagents. BALB/c (H-2d) mice were purchased from The

Jackson Laboratory. Clone 4 and InsHA (12) transgenic mice were obtainedfrom Dr. Linda Sherman (The Scripps Research Institute, La Jolla, CA).

h2m�/� mice were provided by Dr. Kevin L. Legge (University of Iowa, Iowa

City, IA). CpG oligodeoxynucleotide (CpG ODN) 1826 was purchased from

Requests for reprints: Thomas S. Griffith, Department of Urology, University ofIowa, 3204 MERF, 375 Newton Road, Iowa City, IA 52242-1089. Phone: 319-335-7581;Fax: 319-353-4556; E-mail: [email protected].

I2007 American Association for Cancer Research.doi:10.1158/0008-5472.CAN-07-1526

Cancer Res 2007; 67: (24). December 15, 2007 11980 www.aacrjournals.org

Research Article

Research. on August 16, 2020. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 2: ActivationofTumor-SpecificCD8 TCellsafterIntratumoral Ad5 ...Ad5-mTRAIL (109 pfu), and/or CpG ODN (100 Ag) were injected i.t. After another 7 days, the tumors were harvested and physically

Coley Pharmaceutical. Anti-CD25 (PC-61) and anti-GITR (DTA-1) werepurchased from eBioscience.

Cell lines. The transplantable murine renal adenocarcinoma cell line,

Renca (13), was obtained from Dr. Robert Wiltrout (National Cancer

Institute, Frederick, MD) and maintained in RPMI 1640 supplemented with

10% fetal bovine serum, penicillin, streptomycin, sodium pyruvate,

nonessential amino acids, and HEPES (hereafter called complete RPMI).

Renca stably transfected to express hemagglutinin (RencaHA; ref. 14) was

obtained from Dr. Hyam Levitsky (Johns Hopkins University, Baltimore,

MD) and maintained in complete RPMI with 400 Ag/mL G418. The mouse

prostate tumor cell line, RM-11 (15), was obtained from Dr. Timothy Ratliff

(University of Iowa) and maintained in complete RPMI.Production of recombinant adenoviral vectors. Replication-deficient

adenovirus encoding murine tnfsf10 (Ad5-mTRAIL; ref. 10), human Flt-3L

(Ad5-Flt-3L), or h-galactosidase (Ad5-hgal) expressed from the cytomega-

lovirus promoter was made at the University of Iowa Gene Transfer Vector

Core using the RAPAd.I system (16). Recombinant adenoviruses were

screened for replication competent virus by A549 plaque assay, and virus

titer was determined by plaque assay on 293 cells.

Dendritic cell phagocytosis. Immature CD11c+ dendritic cells were

mobilized by injecting Ad5-Flt-3L [109 plaque-forming units (pfu) i.v.] intonaı̈ve BALB/c mice and isolated after 7 to 10 days from spleens using a

Miltenyi kit (Miltenyi Biotec, Inc.). Dendritic cells were then labeled with

PKH67 membrane dye (Sigma). Renca cells were labeled with PKH26membrane dye (Sigma) before induction of apoptosis by either UV irra-

diation for 5 min or Ad5-mTRAIL (300 pfu/cell) infection. Apoptosis was

allowed to occur for 16 h before adding dendritic cells, and dendritic cell

phagocytosis of the apoptotic debris lasted for 16 h. Phagocytosis wasassessed on a FACScan (Becton Dickson) at the indicated time points. To

confirm phagocytosis, irradiated Renca cells and dendritic cells were also

incubated at 4jC, which inhibits phagocytosis (17). Dendritic cell

maturation was also assessed by staining with an anti-CD80 phycoery-thrin-Cy5 antibody (eBioscience) after 24 h.

Confocal microscopy of dendritic cell phagocytosis. Samples for

phagocytosis were set up as described above. Cells were FACS ( fluores-

cence-activated cell sorting)–sorted to obtain the double positive

‘‘phagocytosing populations,’’ as well as the PKH67 only (‘‘nonphagocytos-

ing’’) population. Cells were allowed to adhere to BD BioCoat Collagen I

Chamber Slides (BD Biosciences) for 4 h at 37jC, fixed in 2% parafor-

maldehyde, and analyzed.In vitro cross-presentation. Ad5-Flt-3L–mobilized CD11c+ dendritic

cells were isolated from BALB/c or h2m�/� mice, and apoptotic cultures

were set up as described above. Phagocytosis lasted for 16 h, after which the

cultures were clarified with Fico/Lite (Atlanta Biologicals) to separateunphagocytosed apoptotic/dead cells from dendritic cells. Dendritic cells

were then cultured with carboxyfluorescein succinimidyl ester (CFSE)–

labeled (10 min at 37jC with 1 Amol/L CFSE) clone 4 T cells. The dendriticcell and clone 4 T cells were cultured in a 1:1 ratio in 96-well round-

bottomed plates for 72 to 96 h. HA518-526 peptide (IYSTVASSL; 1 Amol/L;

New England Peptide, Inc.)–pulsed dendritic cells or dendritic cells infected

with UV-inactivated influenza virus strain A/PR/8/34 H1N1 (Dr. KevinLegge, University of Iowa) mixed with clone 4 T cells served as positive

controls. After incubation, cells were stained with anti-CD8a and anti-

CD90.2 (eBioscience), and CFSE dilution was monitored by flow cytometry.

In vivo cross-presentation. InsHA mice were injected s.c. with 2 � 105

RencaHA in their hind flank. Six days after implantation, 3 � 106 CFSE-

labeled clone 4 T cells were injected i.v. On day 7, Ad5-hgal (109 pfu),

Ad5-mTRAIL (109 pfu), and/or CpG ODN (100 Ag) were injected

intratumorally (i.t.). Five days later, inguinal lymph nodes were harvestedand stained with anti-CD8a and anti-CD90.2 monoclonal antibody (mAb)

to assess CFSE dilution by flow cytometry.

In vivo cytolytic assay. InsHA mice were injected s.c. with 2 � 105

RencaHA cells in their hind flank. Six days after implantation, 3 � 106

clone 4 T cells were transferred i.v. On day 7, Ad5-hgal (109 pfu), Ad5-

mTRAIL (109 pfu), and/or CpG ODN (100 Ag) were injected i.t. Five days

later, 3 � 106 syngeneic spleen cells were labeled with 5 Amol/L CFSEhigh

and pulsed with 1 Amol/L HA518-526 peptide for 1 h at 37jC or 3 � 106

unplused cells were labeled with 0.5 Amol/L CFSElow. CFSEhigh andCFSElow cells were mixed in a 1:1 ratio, and 6 � 106 cells in total were

injected i.v. into tumor-bearing InsHA mice. Eighteen hours after transfer,

spleens were harvested and the CFSEhigh to CFSElow cell ratio was

analyzed by flow cytometry.Tumor therapy experiments. BALB/c mice were implanted s.c in the

hind flank with 2 � 105 Renca. Ad5-hgal (109 pfu), Ad5-mTRAIL (109 pfu),

and/or CpG ODN (100 Ag) were injected i.t. on day 7. In some cases, mice

were also depleted of CD8+ or CD4+ T cells with 100 Ag anti-CD8 or anti-CD4 (obtained from Dr. Timothy Ratliff, University of Iowa) 3 consecutive

days before tumor implantation and maintenance injections biweekly

throughout the experiment, CD25+ cells with anti-CD25 (500 Ag given 1 day

before tumor implantation), or GITR+ cells with anti-GITR (1 mg given 1 daybefore and 4 and 9 days after tumor implantation). CD8+, CD4+, and CD25+

cell depletion was verified by flow cytometry in sentinel mice. Tumor

outgrowth and animal survival were monitored over time. To determine thepresence of immunologic memory after Ad5-mTRAIL/CpG ODN therapy,

mice that had rejected the primary Renca challenge were rechallenged with

Renca tumor cells (2 � 105) implanted s.c. on the left flank and RM-11

tumor cells (2 � 105) implanted s.c. on the right flank. Tumor outgrowthand animal survival were monitored over time.

Assessment of tumor-infiltrating cells. BALB/c mice were implanted

s.c. in the hind flank with 2 � 105 Renca. After 7 days, Ad5-hgal (109 pfu),

Ad5-mTRAIL (109 pfu), and/or CpG ODN (100 Ag) were injected i.t.After another 7 days, the tumors were harvested and physically dissociated

to achieve a single-cell suspension, which was then labeled with FITC-

conjugated anti-CD4 and phycoerythrin-conjugated anti-CD8 mAb (eBio-science) and analyzed by flow cytometry.

Statistics. Statistical analysis was performed using ANOVA or Student’s

unpaired t test to assess differences between groups. Survival data was

analyzed using the Kaplan-Meier survival analysis and log-rank statistics. Allreported P values are two sided, and statistical significance was determined

as P < 0.05.

Results

In vitro phagocytosis of apoptotic Renca cells. Antigen cross-presentation is necessary to generate a CD8+ T-cell response toexogenously derived antigens, such as those from apoptotic cells.We hypothesize that Renca cells undergoing Ad5-mTRAIL–inducedapoptosis (18) are ingested by dendritic cell and the tumor antigenspresented to CD8+ T cells. To test this hypothesis, our experimentswill use both parental Renca tumor cells and Renca cells stablyexpressing hemagglutinin on the surface (RencaHA), making itimportant to first compare the responsiveness of both cell lines toAd5-mTRAIL. Ad5-mTRAIL induced death similarly in both Rencaand RencaHA, whereas Ad5-hgal did not induce cell death,indicating cell death was due to TRAIL expression and not simplythe result of adenoviral infection (Fig. 1A). To test our hypothesisthat apoptotic Renca cell antigens could be cross-presented, wedetermined the capacity of dendritic cell to phagocytose apoptotictumor cells in the presence of adenovirus. Thus, Renca tumorcells were labeled with PKH26, induced to undergo apoptosis withAd5-mTRAIL or UV irradiation (which yielded comparable levels ofapoptotic cells; data not shown), and then mixed with splenicCD11c+ dendritic cell labeled with PKH67. Phagocytosis wasassessed over a time course of 24 h, and the results show thatthe presence of an adenovirus in the culture did not alter the abilityof the dendritic cells to take up apoptotic tumor cells killed byAd5-mTRAIL compared with radiation-induced (adenovirus-free)apoptotic cultures (Fig. 1B). As predicted, Ad5-hgal–infected cells,which do not undergo apoptosis, were not taken up by thedendritic cells. To confirm that the method of uptake of the Rencaapoptotic bodies was phagocytosis, a coculture of irradiated Renca

Tumor Antigen Cross-Presentation after Apoptosis

www.aacrjournals.org 11981 Cancer Res 2007; 67: (24). December 15, 2007

Research. on August 16, 2020. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 3: ActivationofTumor-SpecificCD8 TCellsafterIntratumoral Ad5 ...Ad5-mTRAIL (109 pfu), and/or CpG ODN (100 Ag) were injected i.t. After another 7 days, the tumors were harvested and physically

cells and dendritic cells were incubated at 4jC (17). This treatmentprevented the uptake of apoptotic fragments. To confirm that thedendritic cells were truly engulfing the apoptotic Renca cells,dendritic cells were sorted using the same gating scheme and thenanalyzed by confocal microscopy. In the representative imagesshown, the green phagocytic cells contained red tumor cellmembranes within them (Fig. 1C and D), confirming that thedouble-positive cells measured in Fig. 1B were dendritic cells thathad specifically phagocytosed apoptotic cells.

Based on the results in Fig. 1B where dendritic cells in thepresence of adenovirus were equally capable of phagocytosingtumor cells, we next examined whether these dendritic cellswere matured by the presence of adenovirus. The dendritic cellswere stained for surface CD80 expression, which increased in thetreatment groups where virus is present compared with eitherthe irradiated cells/dendritic cells cultured at 4jC or irradiatedcells in the absence of virus (Fig. 1E). Thus, the presence of anadenovirus in these cultures does mature the dendritic cells but

Figure 1. Dendritic cells effectively phagocytose Ad5-mTRAIL–generated apoptotic bodies. A, Renca tumor cells were added to a 96-well plate at 2 � 104 per well,and the cells were infected at the indicated adenoviral concentrations (pfu/cell) for 24 h. Cells were then stained with crystal violet to measure cell death. Points, averageof three wells. B to E, 2.5 � 106 Renca tumor cells were added to six-well plates and infected with 300 pfu/cell of Ad5-mTRAIL or Ad5-hgal. UV irradiation wasadministered for 5 min to induce apoptosis. Dendritic cells (DC ) were added to plates 16 h after induction of apoptosis and allowed to phagocytize cell debris for 24 h.In some cases, cultures were placed at 4jC to inhibit phagocytosis. B, total cultures were then analyzed by flow cytometry to determine phagocytosis (double-positivepopulation). C, confocal microscopy was used to confirm FACS analysis of phagocytosis. Double-positive cells were FACS sorted and analyzed by fluorescenceon a confocal microscope. D, confocal assessment of colocalization of red and green fluorescent intensity across the cell. E, dendritic cell maturation after phagocytosiswas assessed by measuring CD80 expression by flow cytometry. Columns, average CD80 MFI after 24 h from three independent experiments; bars, SE. *, P < 0.05.All other data are representative of three independent experiments.

Cancer Research

Cancer Res 2007; 67: (24). December 15, 2007 11982 www.aacrjournals.org

Research. on August 16, 2020. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 4: ActivationofTumor-SpecificCD8 TCellsafterIntratumoral Ad5 ...Ad5-mTRAIL (109 pfu), and/or CpG ODN (100 Ag) were injected i.t. After another 7 days, the tumors were harvested and physically

does not alter their ability to phagocytose apoptotic tumorcells in vitro .Tumor antigen cross-presentation in vitro after Ad5-

mTRAIL–induced apoptosis. Delivery of trail using a recombi-nant adenoviral vector results in sustained local TRAIL expressionand increases the local TRAIL concentration at the tumor site (11).Ad5-mTRAIL is also a potentially appealing agent for initiatingcross-presentation because, in addition to inducing tumor cellapoptosis, the adenovirus stimulates dendritic cell maturation, asshown in Fig. 1. Thus, it was important to assess how effective Ad5-mTRAIL would be at stimulating dendritic cells to cross-presenttumor antigen to CD8+ T cells. To examine this in vitro , RencaHAand clone 4 T cells were used. RencaHA cells were infected withAd5-hgal, Ad5-mTRAIL, or UV irradiated to induce apoptosis, and16 h later CD11c+ dendritic cells were added to permitphagocytosis. The dendritic cell/RencaHA cultures were thenincubated with CFSE-labeled clone 4 T cells, and the percentageof proliferating cells in groups that were induced to undergoapoptosis with Ad5-mTRAIL or UV irradiation were equivalent

(Fig. 2A). As a positive control, we cultured clone 4 T cells withdendritic cells pulsed with hemagglutinin peptide or UV-inacti-vated influenza, resulting in pronounced T-cell proliferation.Conversely, clone 4 T cells cultured with dendritic cells alone,UV-irradiated Renca, or RencaHA infected with Ad5-hgal showedminimal proliferation. We confirmed that RencaHA antigens werecross-presented to clone 4 T cells by feeding apoptotic RencaHAcells (induced by either Ad5-mTRAIL or UV irradiation) to CD11c+

dendritic cells isolated from h2m�/� mice, which lack MHC Iexpression (19). In this setting, the clone 4 T cells failed toproliferate (Fig. 2A). Furthermore, we tested the ability of viableRenca or RencaHA cells to directly stimulate clone 4 T-cellproliferation. Just as we saw with the h2m�/� dendritic cells, therewas no clone 4 T-cell proliferation when cultured with Renca orRencaHA cells alone (Fig. 2B). These results collectively show thattumor cell antigen derived from apoptotic Renca cells are cross-presented to CD8+ T cells, and the presence of an adenovirus in thissystem does not alter the capacity of the dendritic cells to cross-present tumor antigen.

Figure 2. RencaHA tumor cell antigens arecross-presented to CD8+ T cells in vitro. A,2.5 � 106 Renca or RencaHA cells were infectedwith Ad5-hgal (300 pfu/cell), Ad5-mTRAIL(300 pfu/cell), or UV irradiated to induce apoptosis.Dendritic cells isolated from either BALB/c orh2m�/� mice were added to cultures 16 h afterinduction of apoptosis and allowed to phagocytizecell debris for 24 h. Dendritic cells were isolatedand then cultured at a 1:1 ratio with CFSE-labeledclone 4 T cells for 96 h, after which clone 4T-cell proliferation was determined by CFSEdilution. Control groups included clone 4 T cellscultured with dendritic cells alone UV-irradiatedRenca cells, dendritic cells pulsed with HA518-526

peptide (peptide pulsed), or dendritic cells infectedwith UV-inactivated influenza (UV Irr. Flu ).B, RencaHA cells do not directly stimulate clone4 T cell proliferation. 106 CFSE-labeled clone 4T cells were incubated alone or with Renca orRencaHA cells (1:1 ratio) for 96 h, after which clone4 T cell proliferation was determined by CFSEdilution. DI, division index as calculated by thesoftware analysis program FlowJo. All data arerepresentative of at least three independentexperiments.

Tumor Antigen Cross-Presentation after Apoptosis

www.aacrjournals.org 11983 Cancer Res 2007; 67: (24). December 15, 2007

Research. on August 16, 2020. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 5: ActivationofTumor-SpecificCD8 TCellsafterIntratumoral Ad5 ...Ad5-mTRAIL (109 pfu), and/or CpG ODN (100 Ag) were injected i.t. After another 7 days, the tumors were harvested and physically

Ad5-mTRAIL therapy must be combined with CpG ODN toshow a therapeutic effect and induce antigen-specific T cellproliferation and CTL activity. The previous experimentsindicate that Ad5-mTRAIL can generate apoptotic tumor cellsthat can be phagocytosed and cross-presented in vitro to clone 4T cells. Thus, we expected Ad5-mTRAIL administered i.t. wouldincrease animal survival. To test this, BALB/c mice were injecteds.c. with Renca in the hind flank, Ad5-hgal or Ad5-mTRAIL wasinjected i.t. 7 days later, and tumor size (Fig. 3A) and animalsurvival were monitored. Animal survival was prolonged withAd5-mTRAIL compared with untreated or Ad5-hgal–treated mice,but all the mice eventually succumbed to their tumors (Fig. 3B).Although dendritic cells were presumably cross-presenting tumorantigens from apoptotic Renca cells killed by Ad5-mTRAIL,additional priming signals may be needed to induce a more robustCD8+ T-cell response. CpG ODNs are powerful immune adjuvantsthat activate multiple immune cell populations, including dendritic

cells (20). Thus, we examined the benefits of including CpG ODNwith Ad5-mTRAIL. In vivo administration of CpG ODN maturesdendritic cells in draining lymph nodes when administered intothe footpad of BALB/c mice (Fig. 3C), as determined by the up-regulation of CD80, CD86, CD54, MHCI, and MHCII expressionlevels versus untreated groups. Consequently, we determined if thecombination of Ad5-mTRAIL and CpG ODN would prolong animalsurvival compared with single-agent therapy. Surprisingly, CpGODN treatment alone significantly enhanced animal survival overuntreated animals but still all of the animals succumbed to thetumor. The greatest increase in survival, however, was observedwhen Ad5-mTRAIL and CpG ODN were injected in combination i.t.(Fig. 3A). In fact, the combination therapy even resulted in somemice completely eliminating the tumor. Concurrently, we assessedthe recruitment of T cells into the tumor 7 days after injection ofAd5-hgal, Ad5-mTRAIL, and/or CpG ODN. There was a dramaticincrease in the percentage of both CD4+ and CD8+ cells within the

Figure 3. Ad5-mTRAIL and CpG ODN prolong survival in tumor-bearing animals. A and B, BALB/c mice were implanted s.c. with 2 � 105 Renca cells. After 7 d,mice were injected i.t. with Ad5-hgal (109 pfu), Ad5-mTRAIL (109 pfu), CpG ODN 1826 (100 Ag), or in combination. Tumor size and animal survival were monitoredand plotted over time. Untreated (n = 24) versus Ad5-mTRAIL (n = 30), P < 0.01; untreated versus CpG ODN (n = 30), P < 0.01; untreated versus Ad5-mTRAIL + CpGODN (n = 33), P < 0.01; Ad5-mTRAIL versus CpG ODN, P < 0.01; CpG ODN versus Ad5-mTRAIL + CpG ODN, P < 0.01; untreated versus Ad5-hgal (n = 24),P = 0.546; CpG ODN versus Ad5-hgal + CpG ODN (n = 20), P = 0.691. The data are cumulative of six independent experiments. C, BALB/c mice were injected withCpG ODN 1826 (50 Ag) in each hind footpad. After 48 h, popliteal lymph nodes were harvested and stained for CD80, CD86, CD54, MHC I, and MHC II expressionand analyzed by flow cytometry. D, analysis of tumor-infiltrating CD4+ or CD8+ cells 7 d after i.t. injection of Ad5-hgal (109 pfu), Ad5-mTRAIL (109 pfu), CpG ODN1826 (100 Ag), or in combination. Tumors were excised, dissociated into single cell suspensions, and labeled with FITC-conjugated anti-CD4 or phycoerythrin-conjugated anti-CD8 mAb.

Cancer Research

Cancer Res 2007; 67: (24). December 15, 2007 11984 www.aacrjournals.org

Research. on August 16, 2020. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 6: ActivationofTumor-SpecificCD8 TCellsafterIntratumoral Ad5 ...Ad5-mTRAIL (109 pfu), and/or CpG ODN (100 Ag) were injected i.t. After another 7 days, the tumors were harvested and physically

tumor only after injecting both Ad5-mTRAIL and CpG ODN(Fig. 3D).

The combination of Ad5-mTRAIL and CpG ODN led to asignificant survival advantage in Renca tumor-bearing animals;however, the experiments in Fig. 3 were not designed to directlyassess cross-presentation in vivo. To determine the effectiveness ofAd5-mTRAIL/CpG ODN therapy at stimulating a CD8+ T-cellresponse, InsHA mice implanted with RencaHA tumors weretreated with Ad5-hgal, Ad5-mTRAIL, and/or CpG ODN. One daybefore treatment, CFSE-labeled clone 4 T cells were adoptivelytransferred into the tumor-bearing InsHA mice. Five days aftertreatment, inguinal lymph nodes were harvested, and theproliferation of the adoptively transferred clone 4 T cells wasassessed by CSFE dilution. Untreated InsHA animals had a low butdetectable level of clone 4 T-cell proliferation (Fig. 4A). I.t. injectionof Ad5-mTRAIL or CpG ODN alone increased clone 4 T-cellproliferation, but the greatest level of clone 4 T-cell proliferationoccurred when the RencaHA-bearing InsHA mice received bothAd5-mTRAIL and CpG ODN. These data are consistent with thesurvival results in Fig. 3A , indicating that although there was someantigen-specific CD8+ T-cell activation and therapeutic effect wheneither Ad5-mTRAIL or CpG ODN were used alone, the greatestamount of T-cell activation and therapeutic effect was observedwhen Ad5-mTRAIL was combined with CpG ODN.

Enhancing the ability of dendritic cells to present antigen to, andinduce proliferation of, T cells is important in tumor immunology.

However, T-cell acquisition of tumor-specific cytolytic ability is alsoessential. To further investigate the induction of a tumor antigen–specific T-cell response after Ad5-mTRAIL/CpG ODN therapy, anin vivo CTL assay was performed to assess the cytolytic potential ofthe activated T cells. InsHA mice implanted with RencaHA tumorsand treated with Ad5-hgal, Ad5-mTRAIL, and/or CpG ODN i.t., andthe in vivo CTL assay was performed 5 days later. Just as observedwith the augmentation in T-cell proliferation, CTL activity doubledin mice that were given Ad5-mTRAIL and CpG ODN comparedwith mice that were untreated or given Ad5-mTRAIL alone, CpGODN alone, or Ad5-hgal and CpG ODN (Fig. 4B). Collectively, thedata in Fig. 4 show that neither therapeutic agent alone (Ad5-mTRAIL or CpG ODN) is unable to induce profound antigen-specific CD8+ T-cell proliferation nor CTL activity, but thecombination of increased apoptosis and stimulatory signals isneeded to generate a robust T-cell response.CD8+ T cells are necessary for prolonged survival of mice

treated with Ad5-mTRAIL/CpG ODN. Our in vivo data show theactivation of antigen-specific T cells and induction of CTL activity,suggesting the necessity for CD8+ T cells in the suppression oftumor outgrowth and prolongation of survival. To confirm thatAd5-mTRAIL/CpG ODN therapy was indeed augmenting the CD8+

T-cell response that prolonged survival time, CD8+ T cells weredepleted from BALB/c mice before and throughout the duration ofcombination tumor therapy. Mice depleted of CD8+ T cells andgiven Ad5-mTRAIL/CpG ODN i.t. had no survival advantage over

Figure 4. Ad5-mTRAIL and CpG ODNaugment T-cell proliferation and effectorfunction in response to RencaHA tumors.A, InsHA mice were implanted with2 � 105 RencaHA tumor cells. After 6 d,3 � 106 CFSE-labeled clone 4 T cellswere transferred i.v. into tumor-bearinganimals. One day later, mice receivedan i.t. injection of Ad5-hgal (109 pfu),Ad5-mTRAIL (109 pfu), and/or CpG ODN1826 (100 Ag). Five days after treatment,inguinal lymph nodes were harvested,stained for CD90.2 and CD8, and analyzedfor T-cell proliferation. To assess thenormal proliferation rate of the clone 4T cells, the same number of cells was alsotransferred into untreated, tumor-freeInsHA mice. Data are representativeof three independent experiments.B, InsHA mice were implanted with2 � 105 RencaHA tumor cells. After 6 d,3 � 106 clone 4 T cells were transferred i.v.into tumor-bearing animals. One day later,mice received an i.t. injection of Ad5-hgal(109 pfu), Ad5-mTRAIL (109 pfu), and/orCpG ODN 1826 (100 Ag). Five daysafter treatment, an in vivo CTL usingHA518-526 peptide-pulsed splenocytes astargets was performed to assess in vivoCTL activity. Columns, data are presentedat percent killing, are based on resultsfrom five mice per group, and arerepresentative of three independentexperiments; bars, SD. *, P < 0.05,Ad5-mTRAIL + CpG versus all othergroups.

Tumor Antigen Cross-Presentation after Apoptosis

www.aacrjournals.org 11985 Cancer Res 2007; 67: (24). December 15, 2007

Research. on August 16, 2020. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 7: ActivationofTumor-SpecificCD8 TCellsafterIntratumoral Ad5 ...Ad5-mTRAIL (109 pfu), and/or CpG ODN (100 Ag) were injected i.t. After another 7 days, the tumors were harvested and physically

untreated mice bearing Renca tumors (Fig. 5A), providing furtherevidence suggesting that Ad5-mTRAIL/CpG ODN therapy activatesantigen-specific CD8+ T cells that are critical to the generation of asuccessful antitumor immune response.CD4+ cells suppress the therapeutic potential of Ad5-TRAIL/

CpG ODN therapy. It was surprising that there was only a smallpercentage (f40%) of mice treated with Ad5-mTRAIL/CpG ODNthat completely eliminated their tumors, despite the significantincrease in CTL activity. We considered the possibility that

CD4+CD25+ regulatory T cells, which have been implicated innumerous tumor models, may be inhibiting the full therapeuticpotential of Ad5-mTRAIL/CpG ODN by suppressing the CD8+ T-cellresponse. To investigate this possibility, antibody-mediated deple-tion of CD4+ or CD25+ cells from BALB/c mice was performedbefore Renca tumor implantation, and then the mice were treatedwith Ad5-mTRAIL/CpG ODN. Interestingly, depletion of CD4+ orCD25+ cells significantly prolonged survival compared with thosemice with a full repertoire of cells (Fig. 5A). Although there was

Figure 5. Animal survival and CTL activityare suppressed by CD4+ or CD25+ cellsafter Ad5-mTRAIL/CpG ODN treatment.A, BALB/c mice were implanted s.c. with2 � 105 Renca cells. After 7 d, micewere injected i.t. with either Ad5-mTRAIL(109 pfu), CpG ODN 1826 (100 Ag), or both.Some mice received anti-CD4, anti-CD8,or anti-CD25 mAb before tumorimplantation and throughout the duration ofthe experiment (as described in Materialsand Methods) to deplete CD4+, CD8+, orCD25+ cells. Animal survival was monitoredand plotted over time. Untreated (n = 15)versus Ad5-mTRAIL + CpG ODN (n = 30),P = 0.0396; Ad5-mTRAIL + CpG ODNversus Ad5-mTRAIL + CpG ODN + CD8depletion (n = 20), P < 0.01; untreatedversus Ad5-mTRAIL + CpG ODN + CD8depletion, P = 0.646; Ad5-mTRAIL + CpGODN versus Ad5-mTRAIL + CpG ODN +CD4 depletion (n = 20), P = 0.0259;untreated versus Ad5-mTRAIL + CpG ODN+ CD4 depletion, P < 0.01; Ad5-mTRAIL +CpG ODN versus Ad5-mTRAIL + CpGODN + CD25 depletion (n = 20),P = 0.0253; Ad5-mTRAIL + CpG ODN +CD4 depletion versus Ad5-mTRAIL + CpGODN + CD25 depletion, P = 0.0646. Thedata are cumulative of four independentexperiments. B, InsHA mice wereimplanted s.c. with 2 � 105 RencaHA cells.After 5 d, 3 � 106 clone 4 T cells weretransferred into tumor-bearing animals.Two days later, mice were injected i.t. witheither Ad5-mTRAIL (109 pfu), CpG ODN1826 (100 Ag), or both. Some groups ofmice received anti-CD4, anti-CD25, oranti-GITR mAb before tumor implantationand throughout the duration of theexperiment (as described in Materials andMethods) to deplete CD4+ or CD25+ cells,or inhibit GITR+ cells. After 5 d, an in vivoCTL was performed using HA518-526

peptide-pulsed splenocytes as targets.Columns, data are presented at percentkilling and are based on results from fivemice per group; bars, SD. Data arerepresentative of three independentexperiments. *, P < 0.05. C, InsHA micewere implanted with 2 � 105 RencaHAtumor cells. Some groups of mice receivedanti-CD4, anti-CD25, or anti-GITR mAbbefore tumor implantation and throughoutthe duration of the experiment (asdescribed in Materials and Methods) todeplete CD4+ or CD25+ cells, or inhibitGITR+ cells. After 6 d, 3 � 106 CFSE-labeled clone 4 T cells were transferred i.v.into tumor-bearing animals. One daylater, mice received an i.t. injection ofAd5-mTRAIL (109 pfu) and CpGODN 1826(100 Ag). After 5 d, inguinal lymph nodeswere harvested, stained for CD90.1 andCD8, and analyzed for T-cell proliferation.The data are representative of threeindependent experiments.

Cancer Research

Cancer Res 2007; 67: (24). December 15, 2007 11986 www.aacrjournals.org

Research. on August 16, 2020. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 8: ActivationofTumor-SpecificCD8 TCellsafterIntratumoral Ad5 ...Ad5-mTRAIL (109 pfu), and/or CpG ODN (100 Ag) were injected i.t. After another 7 days, the tumors were harvested and physically

significantly enhanced survival in mice lacking CD4+ or CD25+

cells, the survival results did not indicate what effects the CD4+ orCD25+ cells were having on the CD8+ T cells. Thus, we assessedin vivo CTL activity in RencaHA-bearing InsHA mice that had beenpretreated with the anti-CD4, CD25, or GITR antibodies 5 daysbefore receiving Ad5-mTRAIL/CpG ODN. Depletion of CD4+ orCD25+ cells resulted in a statistically significant increase in CTLactivity compared with undepleted mice that received Ad5-mTRAIL/CpG ODN (Fig. 5B). Addition of the anti-GITR antibodyresulted in no alteration in CTL activity. We went on to examineclone 4 T-cell proliferation in RencaHA-bearing intact or anti-CD4,CD25, or GITR-treated InsHA mice that received Ad5-mTRAIL/CpG ODN. Despite showing increased CTL activity and survival,mice depleted of CD4+ cells did not show any increase in clone 4T-cell proliferation over undepleted mice (Fig. 5C). Similarly, therewas not an increase in clone 4 T-cell proliferation in the mice thatreceived anti-CD25. However, there was increased clone 4 T-cellproliferation in the mice that received anti-GITR mAb. GITR isexpressed at high levels on both CD4+ and CD8+ T cells afteractivation, and GITR ligation can serve as a costimulatory signal onCD8+ T cells to enhance proliferation and effector function (21, 22).These results suggest that CD4+ cells regulate the CTL activity thatis induced upon Ad5-TRAIL/CpG ODN treatment. It is possible thatthe CD4+CD25+ regulatory T-cell subset is responsible for theinhibition of the full potential of an effective T-cell response toclear Renca tumors, as indicated by an increase in CTL activity inboth groups that were depleted of either CD4+ or CD25+ cells.Mice that reject primary Renca tumors after Ad5-mTRAIL/

CpG ODN therapy also reject a secondary Renca tumorchallenge. The results showing the necessity for CD8+ T cells toreject Renca tumors after Ad5-mTRAIL/CpG ODN therapyprompted us to test for immunologic memory. To do this, micedepleted of CD4+ cells were implanted with Renca tumor cells andtreated with Ad5-mTRAIL/CpG ODN, as in Fig. 5. Mice that provedto be tumor-free after 100 days were then rechallenged with thesame dose of Renca tumor cells on the left flank or MHC-matchedRM-11 prostate tumor cells on the right flank. These mice receivedno additional therapy, and tumor outgrowth was monitored.Additionally, naı̈ve mice were similarly implanted with Renca and

RM-11 cells. Whereas the Renca cells grew progressively in thenaı̈ve mice, none of the mice that had previously rejectedestablished Renca tumors after Ad5-mTRAIL/CpG ODN treatmenthad palpable tumors (Fig. 6A). In contrast, RM-11 grew atcomparable rates in both groups (Fig. 6B), suggesting that thereis tumor-specific immunologic memory established in mice thatrejected the primary tumor after Ad5-mTRAIL/CpG ODN therapy.

Discussion

This report outlines a novel immunotherapeutic approach tocombining Ad5-mTRAIL with CpG ODN in an experimental modelof renal cell carcinoma. The benefits of combining one agent thatselectively kills tumor cells and has prolonged in vivo expressionwith another that has profound immunostimulatory capacity isextremely important, as the combination of Ad5-mTRAIL and CpGODN facilitates the switching of the tumor microenvironment fromone composed of limited antigen presentation to one with primingof tumor-specific CD8+ T cells having potent effector function.

There is controversy over the ability of antigens derived fromapoptotic cells that are cross-presented to CD8+ T cells to induceT-cell immunity versus tolerance. It makes sense that apoptoticdeath does not allow for antigens to be cross-presented to primeCD8+ T cells, because this type of death is expected to quietly clearcellular debris occurring through normal cellular turnover withoutpromoting autoimmunity (23–25). Our results are similar to thosepublished by den Brok and colleagues (26), who combinedcryoablation with CpG ODN to eradicate experimental melanomatumors. In that study, neither cryoablation nor CpG ODN therapyalone was sufficient to induce antitumor immunity, much like ourresults when Ad5-mTRAIL or CpG ODN were used individually. It isimportant to note, however, that the mechanism and specificity ofcell death in our study and the den Brok report are quite different.TRAIL only induces apoptosis in tumor cells, but cryoablationinduces a necrotic-like death in both tumor and normal cells. It stillremains to be conclusively determined whether apoptotic death isthe means by which immunologic tolerance can be induced, butthe use of a tumoricidal agent (Ad5-mTRAIL) in combination withan immunostimulatory molecule (CpG ODN) in our model system

Figure 6. Immunologic memory is presentin CD4+-depleted mice that reject primarytumor challenge after Ad5-mTRAIL/CpGODN treatment. BALB/c mice, whichreceived anti-CD4 mAb before tumorimplantation and throughout the duration ofthe experiment (as described in Materialsand Methods) to deplete CD4+ cells, wereimplanted s.c. with 2 � 105 Renca cells.After 7 d, mice were injected i.t. withAd5-mTRAIL (109 pfu) and CpG ODN1826 (100 Ag). Mice that had rejected theprimary Renca challenge and remainedtumor-free for 100 d were rechallengedwith (A) Renca tumor cells (2 � 105)implanted s.c. on the left flank and (B)RM-11 tumor cells (2 � 105) implanted s.c.on the right flank. Tumor outgrowthwas monitored over time. Data arerepresentative of two independentexperiments, consisting of five mice pergroup.

Tumor Antigen Cross-Presentation after Apoptosis

www.aacrjournals.org 11987 Cancer Res 2007; 67: (24). December 15, 2007

Research. on August 16, 2020. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 9: ActivationofTumor-SpecificCD8 TCellsafterIntratumoral Ad5 ...Ad5-mTRAIL (109 pfu), and/or CpG ODN (100 Ag) were injected i.t. After another 7 days, the tumors were harvested and physically

clearly pushes the immune system toward a priming environment.The addition of CpG ODN helps to mature dendritic cells and up-regulate costimulatory molecule expression, as well as stimulatecytokine production, at the time apoptosis is being induced sothat T-cell activation can occur with the intention of generating acell-mediated antitumor response (27). Induction of dendritic cellmaturation before antigen uptake results in better antigen cross-presentation than when the dendritic cells are matured after theytake up antigen (28), making this approach a more desirablemethod to further stimulate cross-presentation. It is reasonable tohypothesize that CpG ODN–mediated dendritic cell stimulation/maturation occurs before the generation of antigen from apoptotictumor cells when Ad5-mTRAIL and CpG ODN are administeredsimultaneously, because Ad5-mTRAIL must first infect a cell andthen have the transgene transcribed and translated before TRAIL-mediated tumor cell apoptosis can occur.

Some tumors continuously shed antigen as the result of a lowlevel of cell death, and this either leaves the immune systemtolerized or allows for a low level of T-cell proliferation, although itis not enough to inhibit tumor outgrowth (5). Our results showedthat mice bearing Renca tumors do have a low level of proliferationand CTL activity, although this does not inhibit tumor outgrowth.The absence of an increase in CTL activity upon addition of Ad5-mTRAIL alone as a therapy suggests that increasing the amount oftumor antigen available for cross-presentation is not enough tostimulate a significant response. Further, CpG ODN alone does notremarkably increase CTL activity, indicating the lack of naturalcross-priming is not a result of a lack of priming signals only, butlikely a combination of insufficient antigen levels and potentdendritic cell stimulatory signals. This confirms the theory thatalthough TRAIL is delivered as a recombinant adenovirus, theadenoviral signals for stimulating a priming environment are notsufficient and the addition of CpG ODN is necessary to tip thedynamics of the environment to one that supports T-cell activation.

The extent of antigen-specific T-cell proliferation and CTLactivity induced when combining Ad5-mTRAIL and CpG ODNsuggests this therapy is effective at stimulating endogenous T cellsto respond to tumor antigen(s) and gain effector function. Thein vitro killing of Renca tumor cells by immune effector cells(natural killer cells and CD8+ CTL) activated by immunotherapeu-tic protocols can occur by both granule and Fas-mediatedcytotoxicity (29). However, in vivo effector cell mechanisms ofkilling Renca are perforin independent (30). Although our studieswere not designed to investigate the molecules used by the effectorcells activated by the Ad5-mTRAIL/CpG ODN therapy, this is anarea of future interest. We were also surprised by the observationthat CD4+ cell depletion before Ad5-mTRAIL/CpG ODN treatmentaugmented the CD8+ T-cell response and significantly enhancedanimal survival. CD40-CD154 interactions between APC and CD4+

T cells are important in facilitating CD8+ CTL cross-priming events(31, 32). CD40 ligation on the APC increases CD80/CD86 expression(33), which is essential for proper T-cell activation. In this regard,Prilliman et al. (34) showed that CD8+ CTL cross-priming could stilloccur in the absence of CD4+ T-cell help, provided there wassufficient CD28 signaling with an agonistic mAb. The CpG ODNused in our system could substitute for the required CD4+ T-cellhelp. The report by Cho et al. (35) supports this concept, wherethey were able to induce CD8+ CTL activity in CD4 or MHC classII–deficient animals.

Despite the activation of tumor-specific CTL, the combination ofAd5-mTRAIL and CpG ODN did not always result in complete

tumor regression. Renca is a fairly aggressive tumor, and it ispossible that the slight differences in the tumor cells from expe-riment to experiment, as well as the variability in mice, result in thesingle i.t. treatment being insufficient to adequately deal with andeliminate all of the cells in the rapidly growing tumor. Additionalinjections of Ad5-mTRAIL would induce more apoptosis and furtherdebulk the rapidly growing tumor, whereas additional CpG ODNcould help maintain a strong CD8+ T cell–mediated antitumor im-mune response. Our data also implicates a suppressive effect fromCD4+CD25+ cells that restricts the full potential of the therapeuticcombination of Ad5-mTRAIL and CpG ODN. Clinically, patientswith renal cell carcinoma have elevated numbers of CD4+CD25+

regulatory T cells, and decreasing their numbers with high-doseinterleukin 2 (IL-2) or a recombinant IL-2 diphtheria toxin con-jugate enhances antitumor immunity (36, 37). Regulatory T cells canelicit suppressive activity through contact-dependent [CTLA-4,membrane-bound transforming growth factor-h (TGF-h)] orcontact-independent (soluble TGF-h, IL-10) mechanisms (38, 39).Our results show that the depletion of CD4+ or CD25+ cells resultedin increased CTL activity and prolonged animal survival aftertreatment with Ad5-mTRAIL and CpG ODN. Moreover, the animalsthat rejected the primary tumor after Ad5-mTRAIL/CpG ODNtherapy showed tumor-specific immunologic memory that wassufficient to protect against tumor rechallenge. The depletion ofCD25-expressing cells provides additional evidence to suggest thatthe cells involved in reduction of CTL activity in this experimentalsystem are indeed CD4+CD25+ regulatory T cells. Interestingly,tumor antigen–specific (clone 4) T-cell proliferation was notincreased in RencaHA-bearing mice treated with Ad5-mTRAIL/CpG ODN that also received anti-CD4 or anti-CD25 mAb, suggestingthat T-cell effector function, in this case in vivo CTL activity, is notrelated to the ability of the cells to proliferate. One or many of theimmunosuppressive mechanisms used by regulatory T cells may,therefore, be involved in this system to inhibit the full potential ofAd5-mTRAIL/CpG ODN therapy. Further characterization of theregulatory cells is under way, and it is presently unknown whetherthe regulation mediated by these cells is through a contact-dependent or contact-independent mechanism.

Nearly all DNA viruses have evolved to evade host immunedefense mechanisms. One way this can be accomplished is byreducing the genomic content of CpG motifs, yet adenoviruses donot seem to have made such genetic alterations (40). Despitehaving the expected level of genomic CpG dinucleotides, DNA fromadenovirus type 5 has weak immunostimulatory capabilities (41).In fact, type 5 adenoviral DNA contains sequences that activelysuppress the stimulatory effects of other prokaryotic DNAsequences. Our adenoviral vectors are based on the type 5 back-bone (42), raising the possibility that the immunostimulatoryeffects of the CpG ODN 1826 used in our experiments are beingsquelched by the inhibitory sequences present in the adenoviralgenome. The intended immune response is then further regulatedby the presence of CD4+ regulatory cells. Although we presentlyhave no experimental evidence to confirm or deny this hypothesis,this could also explain the observed benefit of depleting CD4+ orCD25+ cells before Ad5-mTRAIL/CpG ODN administration.

Recombinant adenoviral vectors transduce a wide range ofdividing and nondividing cell types, making this gene deliverysystem valuable as a tool for studying diseases, for vaccine therapy,and for potential clinical use (43). Wild-type adenovirus infectionsare extremely common in the general population, giving adeno-viruses a well-documented safety record (44). Due to the high

Cancer Research

Cancer Res 2007; 67: (24). December 15, 2007 11988 www.aacrjournals.org

Research. on August 16, 2020. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 10: ActivationofTumor-SpecificCD8 TCellsafterIntratumoral Ad5 ...Ad5-mTRAIL (109 pfu), and/or CpG ODN (100 Ag) were injected i.t. After another 7 days, the tumors were harvested and physically

percentage of the population that is immune to adenoviruses, onecould hypothesize two additional consequences to using anadenoviral vector in the clinical treatment of cancer. First, thepresence of neutralizing mAb could render the injected vectorinactive before it can infect its intended target. This wouldpotentially affect adenoviral vectors delivered systemically greaterthan those delivered directly into the tumor, as in our setting.Interestingly, administration of adenoviral vectors mixed with adelivery vehicle can overcome preexisting adenoviral immunity (45),making it possible to prevail over this potential obstacle. Second, theinduced primary CD8+ T cell–mediated antitumor response mayresult (in part) from the presentation of adenoviral antigens via MHCI on the tumor cells and by cross-presentation on dendritic cells,which would then be followed by the activation of adenovirus-specific CD8+ T cells and the killing of adenovirally infected tumorcells. Although a possibility in some experimental and clinicalsettings, this is not a plausible mechanism in our model becausethere were no adenoviral antigens present at the time of the secondchallenge with Renca cells in the animals that rejected the primarytumor (see Fig. 6).

In conclusion, the results outlined in this article show that Ad5-mTRAIL can induce tumor cell apoptosis, and that apoptotic

tumor cell antigens can be cross-presented to generate a CD8+

T-cell response. For this T-cell response to be significant, however,strong priming signals need to be present, which can be providedby the addition of CpG ODN. Although the results and conclusionsin this report are based on an experimental renal cell carcinomatumor system, it is tempting to speculate that Ad5-mTRAIL/CpGODN therapy could be used against other solid tumors. Theimportant benefit of this type of immunotherapy is that neitherTRAIL nor CpG ODN is directly toxic against normal cells, buteffective against tumor cells, allowing for a therapy with little sideeffects. However, caution must also be exercised as systemicautoimmune responses may develop in response to the self-antigens expressed by the tumors. Obviously, further study isneeded to address these concerns and investigate the activity ofAd5-mTRAIL/CpG ODN in other tumor models.

Acknowledgments

Received 4/26/2007; revised 9/12/2007; accepted 10/16/2007.Grant support: National Cancer Institute grant CA109446.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 in accordancewith 18 U.S.C. Section 1734 solely to indicate this fact.

Tumor Antigen Cross-Presentation after Apoptosis

www.aacrjournals.org 11989 Cancer Res 2007; 67: (24). December 15, 2007

References

1. Bevan MJ. Cross-priming for a secondary cytotoxicresponse to minor H antigens with H-2 congenic cellswhich do not cross-react in the cytotoxic assay. J ExpMed 1976;143:1283–8.2. Robinson BW, Lake RA, Nelson DJ, Scott BA, Marzo

AL. Cross-presentation of tumour antigens: evaluationof threshold, duration, distribution and regulation.Immunol Cell Biol 1999;77:552–8.3. Trinchieri G, Aden DP, Knowles BB. Cell-mediated

cytotoxicity to SV40-specific tumour-associated anti-gens. Nature 1976;261:312–4.4. van der Most RG, Currie A, Robinson BW, Lake RA.

Cranking the immunologic engine with chemotherapy:using context to drive tumor antigen cross-presentationtowards useful antitumor immunity. Cancer Res 2006;66:601–4.5. Nowak AK, Lake RA, Marzo AL, et al. Induction of

tumor cell apoptosis in vivo increases tumor antigencross-presentation, cross-priming rather than cross-tolerizing host tumor-specific CD8 T cells. J Immunol2003;170:4905–13.6. McKenna HJ, Stocking KL, Miller RE, et al. Mice

lacking flt3 ligand have deficient hematopoiesis affect-ing hematopoietic progenitor cells, dendritic cells, andnatural killer cells. Blood 2000;95:3489–97.7. Morgan DJ, Liblau R, Scott B, et al. CD8(+) T cell-

mediated spontaneous diabetes in neonatal mice.J Immunol 1996;157:978–83.8. Wiley SR, Schooley K, Smolak PJ, et al. Identification

and characterization of a new member of the TNFfamily that induces apoptosis. Immunity 1995;3:673–82.9. Pitti RM, Marsters SA, Ruppert S, Donahue CJ, Moore

A, Ashkenazi A. Induction of apoptosis by Apo-2 ligand,a new member of the tumor necrosis factor cytokinefamily. J Biol Chem 1996;271:12687–90.10. Griffith TS, Anderson RD, Davidson BL, Williams

RD, Ratliff TL. Adenoviral-mediated transfer of theTNF-related apoptosis-inducing ligand/Apo-2 ligandgene induces tumor cell apoptosis. J Immunol 2000;165:2886–94.11. Griffith TS, Broghammer EL. Suppression of tumor

growth following intralesional therapy with TRAILrecombinant adenovirus. Mol Ther 2001;4:257–66.12. Lo D, Freedman J, Hesse S, Palmiter RD, Brinster RL,

Sherman LA. Peripheral tolerance to an islet cell-specifichemagglutinin transgene affects both CD4+ and CD8+

T cells. Eur J Immunol 1992;22:1013–22.

13. Murphy GP, Hrushesky WJ. A murine renal cellcarcinoma. J Natl Cancer Inst 1973;50:1013–25.14. Morgan DJ, Kreuwel HT, Fleck S, Levitsky HI,

Pardoll DM, Sherman LA. Activation of low avidityCTL specific for a self epitope results in tumorrejection but not autoimmunity. J Immunol 1998;160:643–51.15. Elzey BD, Siemens DR, Ratliff TL, Lubaroff DM.

Immunization with type 5 adenovirus recombinant for atumor antigen in combination with recombinantcanarypox virus (ALVAC) cytokine gene delivery inducesdestruction of established prostate tumors. Int J Cancer2001;94:842–9.16. Anderson RD, Haskell RE, Xia H, Roessler BJ,

Davidson BL. A simple method for the rapid generationof recombinant adenovirus vectors. Gene Ther 2000;7:1034–8.17. Rubartelli A, Poggi A, Zocchi MR. The selective

engulfment of apoptotic bodies by dendritic cells ismediated by the a(v)h3 integrin and requires intracel-lular and extracellular calcium. Eur J Immunol 1997;27:1893–900.18. Kemp TJ, Elzey BD, Griffith TS. Plasmacytoid dendritic

cell-derived IFN-a induces TNF-related apoptosis-induc-ing ligand/Apo-2L-mediated antitumor activity by hu-man monocytes following CpG oligodeoxynucleotidestimulation. J Immunol 2003;171:212–8.19. Zijlstra M, Bix M, Simister NE, Loring JM, Raulet DH,

Jaenisch R. Beta 2-microglobulin deficient mice lackCD4�8+ cytolytic T cells. Nature 1990;344:742–6.20. Krieg A, Kline J. Immune effects and therapeutic

applications of CpG motifs in bacterial DNA. Immuno-pharmacology 2000;48:303–5.21. Ronchetti S, Zollo O, Bruscoli S, et al. GITR, a

member of the TNF receptor superfamily, is costimula-tory to mouse T lymphocyte subpopulations. Eur JImmunol 2004;34:613–22.22. Cohen AD, Diab A, Perales MA, et al. Agonist anti-

GITR antibody enhances vaccine-induced CD8(+) T-cellresponses and tumor immunity. Cancer Res 2006;66:4904–12.23. Galea-Lauri J, Wells JW, Darling D, Harrison P,

Farzaneh F. Strategies for antigen choice and primingof dendritic cells influence the polarization and efficacyof antitumor T-cell responses in dendritic cell-basedcancer vaccination. Cancer Immunol Immunother 2004;53:963–77.24. Strome SE, Voss S, Wilcox R, et al. Strategies for

antigen loading of dendritic cells to enhance the

antitumor immune response. Cancer Res 2002;62:1884–9.25. Scheffer SR, Nave H, Korangy F, et al. Apoptotic,

but not necrotic, tumor cell vaccines induce apotent immune response in vivo . Int J Cancer 2003;103:205–11.26. den Brok MH, Sutmuller RP, Nierkens S, et al. Synergy

between in situ cryoablation and TLR9 stimulationresults in a highly effective in vivo dendritic cell vaccine.Cancer Res 2006;66:7285–92.27. Sparwasser T, Koch ES, Vabulas RM, et al. Bacterial

DNA and immunostimulatory CpG oligonucleotidestrigger maturation and activation of murine dendriticcells. Eur J Immunol 1998;28:2045–54.28. Datta R, Kojima H, Banach D, et al. Activation of a

CrmA-insensitive, p35-sensitive pathway in ionizingradiation-induced apoptosis. J Biol Chem 1997;272:1965–9.29. Sayers TJ, Brooks AD, Lee JK, et al. Molecular

mechanisms of immune-mediated lysis of murine renalcancer: differential contributions of perforin-dependentversus Fas-mediated pathways in lysis by NK and T cells.J Immunol 1998;161:3957–65.30. Seki N, Brooks AD, Carter CR, et al. Tumor-specific

CTL kill murine renal cancer cells using both perforinand Fas ligand-mediated lysis in vitro , but cause tumorregression in vivo in the absence of perforin. J Immunol2002;168:3484–92.31. Zamai L, Ahmad M, Bennett IM, Azzoni L, Alnemri

ES, Perussia B. Natural killer (NK) cell-mediatedcytotoxicity: differential use of TRAIL and Fas ligandby immature and mature primary human NK cells.J Exp Med 1998;88:2375–80.32. Schoenberger SP, Toes RE, van der Voort EI, Offringa

R, Melief CJ. T-cell help for cytotoxic T lymphocytes ismediated by CD40-40L interactions. Nature 1998;393:480–3.33. McAdam AJ, Schweitzer AN, Sharpe AH. The role

of B7 co-stimulation in activation and differentiationof CD4+ and CD8+ T cells. Immunol Rev 1998;165:231–47.34. Prilliman KR, Lemmens EE, Palioungas G, et al.

Cutting edge: a crucial role for B7-28 in transmit-ting T help from APC to CTL. J Immunol 2002;169:4094–7.35. Cho HJ, Takabayashi K, Cheng PM, et al. Immunos-

timulatory DNA-based vaccines induce cytotoxic lym-phocyte activity by a T-helper cell-independentmechanism. Nat Biotechnol 2000;18:509–14.

Research. on August 16, 2020. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 11: ActivationofTumor-SpecificCD8 TCellsafterIntratumoral Ad5 ...Ad5-mTRAIL (109 pfu), and/or CpG ODN (100 Ag) were injected i.t. After another 7 days, the tumors were harvested and physically

Cancer Research

Cancer Res 2007; 67: (24). December 15, 2007 11990 www.aacrjournals.org

36. Cesana GC, DeRaffele G, Cohen S, et al. Characteriza-tion of CD4+CD25+ regulatory T cells in patients treatedwith high-dose interleukin-2 for metastatic melanoma orrenal cell carcinoma. J Clin Oncol 2006;24:1169–77.37. Dannull J, Su Z, Rizzieri D, et al. Enhancement of

vaccine-mediated antitumor immunity in cancerpatients after depletion of regulatory T cells. J ClinInvest 2005;115:3623–33.38. Knutson KL, Disis ML, Salazar LG. CD4 regulatory T

cells in human cancer pathogenesis. Cancer ImmunolImmunother 2007;56:271–85.39. Roncarolo MG, Gregori S, Battaglia M, Bacchetta R,

Fleischhauer K, Levings MK. Interleukin-10-secreting

type 1 regulatory T cells in rodents and humans.Immunol Rev 2006;212:28–50.40. Karlin S, Doerfler W, Cardon LR. Why is CpG

suppressed in the genomes of virtually all smalleukaryotic viruses but not in those of large eukaryoticviruses? J Virol 1994;68:2889–97.41. Krieg AM, Wu T, Weeratna R, et al. Sequence motifs

in adenoviral DNA block immune activation bystimulatory CpG motifs. Proc Natl Acad Sci U S A1998;95:12631–6.42. Jones N, Shenk T. Isolation of adenovirus type 5 host

range deletion mutants defective for transformation ofrat embryo cells. Cell 1979;17:683–9.

43. Tripathy SK, Black HB, Goldwasser E, Leiden JM.Immune responses to transgene-encoded proteins limitthe stability of gene expression after injection ofreplication-defective adenovirus vectors. Nat Med1996;2:545–50.44. Horwitz MS. Adenoviridae and their replication. In:

Felds BN, Knipe DM, editors. Virology. 2nd ed. NewYork: Raven Press, 1990. p. 1679–721.45. Siemens DR, Elzey BD, Lubaroff DM, et al.

Cutting edge: restoration of the ability to generateCTL in mice immune to adenovirus by delivery ofvirus in a collagen-based matrix. J Immunol 2001;166:731–5.

Research. on August 16, 2020. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 12: ActivationofTumor-SpecificCD8 TCellsafterIntratumoral Ad5 ...Ad5-mTRAIL (109 pfu), and/or CpG ODN (100 Ag) were injected i.t. After another 7 days, the tumors were harvested and physically

2007;67:11980-11990. Cancer Res   Rebecca L. VanOosten and Thomas S. Griffith  Ad5-TRAIL/CpG Oligodeoxynucleotide Combination Therapy

T Cells after Intratumoral+Activation of Tumor-Specific CD8

  Updated version

  http://cancerres.aacrjournals.org/content/67/24/11980

Access the most recent version of this article at:

   

   

  Cited articles

  http://cancerres.aacrjournals.org/content/67/24/11980.full#ref-list-1

This article cites 43 articles, 20 of which you can access for free at:

  Citing articles

  http://cancerres.aacrjournals.org/content/67/24/11980.full#related-urls

This article has been cited by 7 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. (CCC)Click on "Request Permissions" which will take you to the Copyright Clearance Center's

.http://cancerres.aacrjournals.org/content/67/24/11980To request permission to re-use all or part of this article, use this link

Research. on August 16, 2020. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from