Synergistic Effects of IL12 and IL18 in Skewing Tumor-Reactive T-Cell Responses Towards a Type 1...

9
Synergistic Effects of IL-12 and IL-18 in Skewing Tumor-Reactive T-Cell Responses Towards a Type 1 Pattern Qiao Li, 1 Abbey L. Carr, 1 Elizabeth J. Donald, 1 Joseph J. Skitzki, 1 Ryugi Okuyama, 1 Lloyd M. Stoolman, 2 and Alfred E. Chang 1 Departments of 1 Surgery and 2 Pathology, University of Michigan Medical Center, Ann Arbor, Michigan Abstract We have previously described the antitumor reactivity of tumor-draining lymph node (TDLN) cells after secondary activation with antibodies. In this report, we examined the effects of interleukin (IL)-12 and IL-18 on modulating the immune function of antibody-activated murine TDLN cells. TDLN cells were activated with anti-CD3/anti-CD28 monoclo- nal antibody followed by stimulation with IL-12 and/or IL-18. IL-18 in combination with IL-12 showed a synergistic effect in augmenting IFN; and granulocyte macrophage colony-stim- ulating factor secretion, whereas IL-18 alone had minimal effect. Concurrently, IL-18 prevented IL-12–stimulated TDLN cells from producing IL-10. The IL-12/IL-18–cultured TDLN cells therefore manifested cytokine responses skewed towards a Th1/Tc1 pattern. IL-12 and IL-18 stimulated CD4 + TDLN cells and enhanced IFN; production by CD4 + cells to a greater extent than by CD8 + cells. Use of NF-KB p50 / TDLN cells suggested the involvement of NF-KB in the IL-12/IL-18 polarization effect. Furthermore, a specific NF-KB inhibitor significantly suppressed IL-12/IL-18–induced IFN; secretion, thus confirming the requirement for NF-KB activation in IL- 12/IL-18 signaling. In adoptive immunotherapy, IL-12– and IL- 18–cultured TDLN cells infiltrated pulmonary tumor nodules and eradicated established tumor metastases more efficiently than T cells generated with IL-12 or IL-18 alone. Antibody depletion revealed that both CD4 + and CD8 + cells were involved in the tumor rejection induced by IL-12/IL-18– cultured TDLN cells. These studies indicate that IL-12 and IL-18 can be used to generate potent CD4 + and CD8 + antitumor effector cells by synergistically polarizing anti- body-activated TDLN cells towards a Th1 and Tc1 phenotype. (Cancer Res 2005; 65(3): 1063-70) Introduction Ex vivo generation of large numbers of tumor-reactive effector T cells remains a critical step for the successful clinical application of adoptive immunotherapy of cancer. Accumulating literature suggest that cytokine elaboration to specific tumor stimulation is associated with the ability of effector T cells to mediate in vivo tumoricidal effects (1–7). We reported that type 1 cytokine release (i.e., IFNg) and granulocyte macrophage colony-stimulating factor (GM-CSF) correlates with in vivo tumor eradication whereas type 2 cytokine release [i.e., interleukin 10 (IL-10)] was found to suppress antitumor reactivity (3, 7). The inhibitory character of IL-10 on antitumor responses has been reported by mutiple groups (8–11). Thus, methodologies that can up-regulate type 1, whereas down-regulating type 2 cytokine responses of effector T cells should increase their therapeutic potential. The principle of T-cell activation using monoclonal antibody (mAb) ex vivo in the absence of antigen has been used to expand tumor-primed T cells contained within tumor-draining lymph nodes (TDLN) or vaccine-primed lymph nodes (VPLN). Our initial efforts involved the use of anti-CD3 mAb as a surrogate antigen to activate tumor-primed lymphoid cells, followed by expansion in IL-2 (12, 13). This approach resulted primarily in the generation of CD8 + effector cells that mediated tumor regression in vivo . Subsequent clinical studies using this method to activate VPLN cells showed that this cellular therapy can result in achieving durable tumor responses in subjects with advanced cancer (7, 14). We have extended our investigations in the animal models by examining other mAbs that deliver costimulatory signals in concert with anti-CD3 to activate tumor-primed lymphoid cells. These other antibodies have involved anti-CD28 and anti-CD137 (4-1BB; refs. 15, 16). The results of these investigations have indicated that costimulation can increase the activation of tumor-primed lym- phoid cells and their ability to mediate tumor regression in vivo . Although effector T cells can be generated through antibody activation to mediate tumor regression in animal models, clinical responses in adoptive immunotherapy have been confined to a minority of patients. One potential reason for these limited responses is that antibody activation procedures generally stimulate T cells broadly without discriminating between type 1 and type 2 cells, presumably due to the polyclonal expansion characteristics of antibodies directed to the TCR common chain (e.g., CD3 q of the TCR/CD3 complex or CD28). Thus, both type 1 cytokines (e.g., IL-2, IFNg) and type 2 cytokines (e.g., IL-4, IL-5, and IL-10) are up-regulated (17–19). Alternative protocols need to be defined that will preferentially stimulate the type 1 cytokine profile to generate more potent tumor-reactive T cells for cancer immunotherapy. Towards this end, various ex vivo strategies have been investigated using additional signaling stimuli to promote Th1/Tc1 cell proliferation and antitumor reactivity (20–23). The proinflammatory cytokine IL-18, either alone or in combination with IL-2 or IL-12, has shown immune modulatory effects in the induction of IFNg production and the promotion of different effector cells such as CD8 + cells (24), natural killer (NK) cells (25, 26), and neutrophils (27). However, its effects on type 2 cytokine secretion, on CD4 + effector cells, as well as the involved signaling molecule(s) are either not characterized or minimally documented. In this study, we used IL-18 and IL-12 as a modulatory stimulus to antibody-activated TDLN cells and presented evidence that IL-18 in concert with IL-12 could significantly inhibit IL-10 production and generate both CD4 + and CD8 + effector cells. In addition, NF nB was identified as one Requests for reprints: Qiao Li, Division of Surgical Oncology, Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI 48109-0932. Phone: 734-936- 4392; Fax: 734-647-9647; E-mail: [email protected]. D2005 American Association for Cancer Research. www.aacrjournals.org 1063 Cancer Res 2005; 65: (3). February 1, 2005 Research Article Research. on July 21, 2015. © 2005 American Association for Cancer cancerres.aacrjournals.org Downloaded from

Transcript of Synergistic Effects of IL12 and IL18 in Skewing Tumor-Reactive T-Cell Responses Towards a Type 1...

Synergistic Effects of IL-12 and IL-18 in Skewing Tumor-Reactive

T-Cell Responses Towards a Type 1 Pattern

Qiao Li,1Abbey L. Carr,

1Elizabeth J. Donald,

1Joseph J. Skitzki,

1Ryugi Okuyama,

1

Lloyd M. Stoolman,2and Alfred E. Chang

1

Departments of 1Surgery and 2Pathology, University of Michigan Medical Center, Ann Arbor, Michigan

Abstract

We have previously described the antitumor reactivity oftumor-draining lymph node (TDLN) cells after secondaryactivation with antibodies. In this report, we examined theeffects of interleukin (IL)-12 and IL-18 on modulating theimmune function of antibody-activated murine TDLN cells.TDLN cells were activated with anti-CD3/anti-CD28 monoclo-nal antibody followed by stimulation with IL-12 and/or IL-18.IL-18 in combination with IL-12 showed a synergistic effect inaugmenting IFN; and granulocyte macrophage colony-stim-ulating factor secretion, whereas IL-18 alone had minimaleffect. Concurrently, IL-18 prevented IL-12–stimulated TDLNcells from producing IL-10. The IL-12/IL-18–cultured TDLNcells therefore manifested cytokine responses skewed towardsa Th1/Tc1 pattern. IL-12 and IL-18 stimulated CD4+ TDLN cellsand enhanced IFN; production by CD4+ cells to a greaterextent than by CD8+ cells. Use of NF-KB p50�/� TDLN cellssuggested the involvement of NF-KB in the IL-12/IL-18polarization effect. Furthermore, a specific NF-KB inhibitorsignificantly suppressed IL-12/IL-18–induced IFN; secretion,thus confirming the requirement for NF-KB activation in IL-12/IL-18 signaling. In adoptive immunotherapy, IL-12– and IL-18–cultured TDLN cells infiltrated pulmonary tumor nodulesand eradicated established tumor metastases more efficientlythan T cells generated with IL-12 or IL-18 alone. Antibodydepletion revealed that both CD4+ and CD8+ cells wereinvolved in the tumor rejection induced by IL-12/IL-18–cultured TDLN cells. These studies indicate that IL-12 andIL-18 can be used to generate potent CD4+ and CD8+

antitumor effector cells by synergistically polarizing anti-body-activated TDLN cells towards a Th1 and Tc1 phenotype.(Cancer Res 2005; 65(3): 1063-70)

Introduction

Ex vivo generation of large numbers of tumor-reactive effector Tcells remains a critical step for the successful clinical application ofadoptive immunotherapy of cancer. Accumulating literaturesuggest that cytokine elaboration to specific tumor stimulation isassociated with the ability of effector T cells to mediate in vivotumoricidal effects (1–7). We reported that type 1 cytokine release(i.e., IFNg) and granulocyte macrophage colony-stimulating factor(GM-CSF) correlates with in vivo tumor eradication whereas type2 cytokine release [i.e., interleukin 10 (IL-10)] was found tosuppress antitumor reactivity (3, 7). The inhibitory character of

IL-10 on antitumor responses has been reported by mutiple groups(8–11). Thus, methodologies that can up-regulate type 1, whereasdown-regulating type 2 cytokine responses of effector T cells shouldincrease their therapeutic potential.The principle of T-cell activation using monoclonal antibody

(mAb) ex vivo in the absence of antigen has been used to expandtumor-primed T cells contained within tumor-draining lymphnodes (TDLN) or vaccine-primed lymph nodes (VPLN). Our initialefforts involved the use of anti-CD3 mAb as a surrogate antigen toactivate tumor-primed lymphoid cells, followed by expansion inIL-2 (12, 13). This approach resulted primarily in the generation ofCD8+ effector cells that mediated tumor regression in vivo .Subsequent clinical studies using this method to activate VPLNcells showed that this cellular therapy can result in achievingdurable tumor responses in subjects with advanced cancer (7, 14).We have extended our investigations in the animal models byexamining other mAbs that deliver costimulatory signals in concertwith anti-CD3 to activate tumor-primed lymphoid cells. Theseother antibodies have involved anti-CD28 and anti-CD137 (4-1BB;refs. 15, 16). The results of these investigations have indicated thatcostimulation can increase the activation of tumor-primed lym-phoid cells and their ability to mediate tumor regression in vivo .Although effector T cells can be generated through antibody

activation to mediate tumor regression in animal models, clinicalresponses in adoptive immunotherapy have been confined to aminority of patients. One potential reason for these limitedresponses is that antibody activation procedures generallystimulate T cells broadly without discriminating between type 1and type 2 cells, presumably due to the polyclonal expansioncharacteristics of antibodies directed to the TCR common chain(e.g., CD3q of the TCR/CD3 complex or CD28). Thus, both type 1cytokines (e.g., IL-2, IFNg) and type 2 cytokines (e.g., IL-4, IL-5, andIL-10) are up-regulated (17–19). Alternative protocols need to bedefined that will preferentially stimulate the type 1 cytokine profileto generate more potent tumor-reactive T cells for cancerimmunotherapy. Towards this end, various ex vivo strategies havebeen investigated using additional signaling stimuli to promoteTh1/Tc1 cell proliferation and antitumor reactivity (20–23).The proinflammatory cytokine IL-18, either alone or in

combination with IL-2 or IL-12, has shown immune modulatoryeffects in the induction of IFNg production and the promotion ofdifferent effector cells such as CD8+ cells (24), natural killer (NK)cells (25, 26), and neutrophils (27). However, its effects on type2 cytokine secretion, on CD4+ effector cells, as well as the involvedsignaling molecule(s) are either not characterized or minimallydocumented. In this study, we used IL-18 and IL-12 as amodulatory stimulus to antibody-activated TDLN cells andpresented evidence that IL-18 in concert with IL-12 couldsignificantly inhibit IL-10 production and generate both CD4+

and CD8+ effector cells. In addition, NF�nB was identified as one

Requests for reprints: Qiao Li, Division of Surgical Oncology, ComprehensiveCancer Center, University of Michigan, Ann Arbor, MI 48109-0932. Phone: 734-936-4392; Fax: 734-647-9647; E-mail: [email protected].

D2005 American Association for Cancer Research.

www.aacrjournals.org 1063 Cancer Res 2005; 65: (3). February 1, 2005

Research Article

Research. on July 21, 2015. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

of the molecular components involved in the IL-12/IL-18 signalingpathways in anti-CD3/anti-CD28 activated TDLN cells.

Materials and Methods

Mice. Female C57BL/6 (B6) mice and NF-nB p50�/� knockout mice on

B6 background were purchased from the Jackson Laboratory, Bar Harbor,

ME. They were used for experiments at age z8 weeks. The University of

Michigan Laboratory of Animal Medicine approved all protocols.Murine Tumor Cells. MCA 205 and MCA 207 murine tumors are

3-methylcholanthrene–induced fibrosarcomas that are syngeneic to B6

mice. These tumors have been previously characterized to be weakly

immunogenic with distinct tumor-specific transplantation/rejection anti-gens (28). Tumors were maintained in vivo by serial s.c. transplantation in

B6 mice and were used within the eighth transplantation generation. Tumor

cell suspensions were prepared from solid tumors by enzymatic digestion asdescribed previously (15, 16).

TDLN Preparation and T-Cell Activation and Expansion. To prepare

tumor-draining lymph nodes (TDLN), wild-type B6 mice or NF-nB p50�/�

mice were inoculated with 1 � 106 tumor cells in 0.1 mL of PBS s.c. in thelower flank. Nine days later, inguinal TDLN were removed aseptically.

Lymphoid cell suspensions were prepared as previously described (15, 16).

TDLN cells (4 � 106 cells per 2 mL per well) were activated with

1.0 Ag/mL anti-CD3 mAb (BD PharMingen, San Diego, CA) plus 1.0 Ag/mLanti-CD28 mAb (BD PharMingen) immobilized in 24-well plates (Costar,

Cambridge, MA) for 2 days. After antibody activation, the cells were

harvested and counted. The cells were then resuspended in complete mediacontaining recombinant human IL-2 (Chiron Therapeutics, Emeryville, CA)

starting at a concentration of 3 � 105 cells per mL in 6-well culture plates

(Costar) for 3 days. The concentration of IL-2 was 60 IU/mL. Recombinant

mouse IL-12 (BD PharMingen) and rmIL-18 (MBL, Watertown, MA) wereadded at the beginning of the 3-day cell expansion period, either alone or

jointly, at concentrations as indicated in the experiments. At the end of the

cell expansion in IL-2 F IL-12 and/or IL-18, cells were harvested, counted,

and used for adoptive immunotherapy or cytokine secretion assessment.Assessment of Cytokines (IFNg, GM-CSF, and IL-10). Culture super-

natants were collected at the end of cell expansion for cytokine

quantification using ELISA kits (BD PharMingen). To measure cytokinerelease in response to tumor stimulation, 1 � 106 activated and expanded

TDLN cells were cocultured with 0.25 � 106 irradiated MCA 205 in 2 mL

volumes per well for 24 hours at 37jC. The culture supernatants were thencollected and analyzed for cytokine production using ELISAs. The tumorstimulator cells were irradiated to 6,000 cGy by a 137Cs source. MCA 207

tumor cells were used for coculture as specificity controls.

CD4+ Cells and CD8+ Cell Fractionation and Fluorescence-ActivatedCell Sorting Analysis. T cells were enriched by passing TDLN cellsuspensions through nylon wool (Cellular Products, Inc., Buffalo, NY)

column. CD4+ or CD8+ T cells were positively selected from T cell–enriched

cell suspensions by treatment with super paramagnetic microbeads

conjugated with anti-mouse CD4 or anti-mouse CD8 mAbs, followed byseparation using the MACS Separator (Miltenyi Biotec. Inc., Sunnyvale, CA).

The fractionated CD4+ or CD8+ cells were confirmed by fluorescence-

activated cell sorting staining (>95% CD4+ or CD8+, respectively; data notshown). CD3, CD4, and CD8 molecules were assessed by direct

immunofluorescence assays using FITC-labeled anti-CD3, anti-CD4, anti-

CD8 mAbs or isotype-matching controls (all from BD PharMingen) and

analyzed as previously described (15, 16).Adoptive Immunotherapy. B6 mice were inoculated i.v. via tail vein with

2 � 105 tumor cells to establish pulmonary metastases. Three days after

tumor inoculation, mice were infused i.v. with cultured TDLN cells.

Commencing on the day of the cell transfer, IL-2 (30,000 IU) were given(i.p.) in 0.5 mL of PBS and continued twice daily for eight doses unless

otherwise indicated. Five mice were used in each experimental group. On

days 14 to 16, all mice were randomized and sacrificed for enumeration ofpulmonary metastatic nodules. Metastatic foci too numerous to count were

assigned an arbitrary value of >250. To deplete CD4+ and/or CD8+ T cells

in vivo , mice were given 800 Ag of anti-CD4 (GK1.5, rat IgG2b) and/or 400 Ag of

anti-CD8 (2.43, rat IgG2a; both from Ligocyte, Inc., Bozeman, MT) i.p. in 1 mLPBS immediately following cell transfer. Fluorescence-activated cell sorting

analysis of splenocytes showed complete depletion of CD4+ cells or CD8+ cells

for as long as 7 days. Therefore, in subsequent experiments the depletion

procedure was repeated once on day 8 after cell transfer. Immunocytochem-ical evaluation of adoptively transferred TDLN cells accumulating in

pulmonary tumor nodules was preformed as previously described (29).

NF-KB Inhibition. We used an intracellularly targeted peptide inhibitor

of nuclear translocation as an approach to NF-nB inhibition (30). Theinhibitor, BMS-205820, was synthesized by Bachem Bioscience, Inc., King of

Prussia, PA. BMS-205820 has the following peptide sequence PKKKRKVAA-

VALLPAVLLALLAPKKKRKV. Sequence in italic is the membrane-trans-

locating hydrophobic sequence; sequence underlined is the nuclearlocalization sequence. The two control peptides we used were SV40

(GGGPKKKRKV) and JBC (AAVALLPAVLLALLAPVQRKRQKLMP). TDLN

cells were activated with anti-CD3/anti-CD28 mAb for 2 days, washed, andthen incubated with BMS-205820 or control peptides in IL-2 containing

medium for two hours before the addition of IL-12 and IL-18.

Statistical Analysis. The significance of differences in numbers of

metastatic nodules between experimental groups was determined using thenonparametric Wilcoxon rank sum test. Two-sided P < 0.05 was considered

statistically significant between two groups. Student’s t test was used to

analyze cell expansion and cytokine release data.

Results

Synergistic Effects of IL-12 and IL-18 in Skewing Antibody-Activated TDLN Cell Responses Toward a Type 1 Pattern. Weexamined the cytokine profile released by TDLN cells stimulatedwith IL-12 and/or IL-18 following antibody activation with anti-CD3 and anti-CD28. As shown in Fig. 1, IL-12 alone enhanced IFNg,GM-CSF, and IL-10 secretion in a dose-dependent manner, whereasIL-18 alone had minimal effect on the levels of these cytokines.However, the use of IL-18 in combination with IL-12 showed asynergistic effect in augmenting IFNg and GM-CSF secretion;whereas inhibiting IL-10 production induced by IL-12 alone.Specifically, when 10 ng/mL of IL-12 and 100 ng/mL of IL-18 wereused simultaneously, both IFNg and GM-CSF production wasnotably enhanced in a synergistic fashion. When IL-12 (10 ng/mL)and IL-18 (100 ng/mL) were used separately, the sum of secretedIFNg was less then 10,000 pg/mL. However, when IL-12 (10 ng/mL)and IL-18 (100 ng/mL) were used jointly, nearly 30,000 pg/mL ofIFNg were produced, showing a synergy rather that an additiveeffect between IL-12 and IL-18. By contrast, the IL-10 secretioninduced by IL-12 exposure at different concentrations wassignificantly down-regulated when IL-18 was added. In separateexperiments, we used intermediate doses (25 and 50 ng/mL) of IL-18 between 10 and 100 ng/mL in combination with 10 ng/mL of IL-12. We found that IFNg production by TDLN cells cultured in IL-12(10 ng/mL) plus IL-18 at 10, 25, and 50 ng/mL were comparable.However, when IL-18 was used at 100 ng/mL, IFNg production wasfound to be statistically higher than any other groups, demon-strating optimal synergistic effect with IL-12 (data not shown).Jointly, these experiments confirmed that 10 ng/mL of IL-12 plus100 ng/mL of IL-18 were optimal within the concentration rangeswe tested to selectively stimulate IFNg and GM-CSF production byactivated TDLN cells without up-regulating IL-10 secretion.Therefore, these concentrations of IL-12 and IL-18 were used tostimulate the antibody-activated TDLN cells for the rest of theexperiments described in this study.We subsequently measured the cytokines released by IL-12/IL-18

stimulated TDLN cells in response to tumor antigen. IL-12/IL-18stimulated MCA 205 TDLN cells were cocultured with irradiated

Cancer Research

Cancer Res 2005; 65: (3). February 1, 2005 1064 www.aacrjournals.org

Research. on July 21, 2015. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

MCA 205 tumor cells. Culture supernatants after tumor cellstimulation were collected and analyzed for cytokines. As shownin Table 1, the combination of IL-12 and IL-18 resulted in anelevation of IFNg and GM-CSF secretion in response to MCA 205tumor cells by an average of 28- and 13-fold respectively in sevenindependent experiments (P < 0.05 comparing fold change with IL-12/IL-18 versus no IL-12/IL-18), whereas IL-10 productionminimallychanged by only 1.3-fold (P = 0.30 comparing fold change with IL-12/IL-18 versus no IL-12/IL-18). We also used MCA 207 tumor cells as aspecificity control. As shown in Fig. 2, the combined use of IL-12 andIL-18 in up-regulating IFNg secretion and concomitantly down-regulating IL-10 production was immunologically specific.IL-12 and IL-18 Skewed T-Cell Response toward the Type 1

Pattern by Enhancing Both Th1 and Tc1 Responses. Typically,nonactivated TDLN cells contain f50% CD3+ cells with CD4+ andCD8+ cells being 15% to 20% and 30% to 35%, respectively (15, 16).Phenotype examination revealed that at the end of IL-12 and/orIL-18 stimulation, >95% of the TDLN cells were CD3+ cellsregardless of the culture conditions (data not show). However,

IL-12 preferentially stimulated the growth of CD4+ T cells eitherby itself or in combination with IL-18. The percentage of CD4+

T cells in the final CD3+ T-cell population increased significantlyfrom f15% in the absence of IL-12/IL-18 or when IL-18 was usedalone, to f30% with the use of IL-12 alone or in combinationwith IL-18. By contrast, the percentage of CD8+ T cells declinedfrom nearly 80% to f60% under these respective conditions (datanot shown). Hence, the enhanced type 1 cytokine productioninduced by IL-12 and IL-18 seems to be associated with thepreferential expansion of CD4+ cells within the whole CD3+ TDLNcell population. Using fractionated CD4+ or CD8+ TDLN cells forcytokine secretion analysis (Fig. 3), we found that IL-12 + IL-18significantly enhanced IFNg production by CD4+ cells in a similarmanner as unfractionated TDLN cells. In addition, we also foundthat IL-18 synergistically enhanced IFNg production by CD8+

cells, although IL-12/IL-18 enhanced CD4+ cells to a much higherextent than CD8+ cells when an equal number (1 � 106) of cellswere examined (Fig. 3). Also, as seen with unfractionated TDLNcells, the IL-12–induced IL-10 production of both CD4+ and CD8+

cell subpopulations was significantly inhibited by IL-18. Theseexperiments indicate that IL-12 and IL-18 synergistically skewT-cell responses toward the type 1 pattern by enhancing both Th1and Tc1 responses, and IL-18 was capable of inhibiting both Th2and Tc2 cytokine production of IL-12–cultured TDLN cells.In vivo Antitumor Activity of IL-12/IL-18–Stimulated TDLN

Effector Cells. The in vivo antitumor reactivity of antibody-activated TDLN cells cultured in IL-12/IL-18 was examined in anadoptive immunotherapy model of mice bearing 3-day establishedMCA 205 pulmonary metastases. Varying numbers of activatedTDLN cells were transferred along with the administration of IL-2.As illustrated in Fig. 4A , the administration of IL-2 alone had notherapeutic effect compared with control mice receiving no therapy.The mean number of metastases was significantly reduced after thetransfer of TDLN cells cultured with IL-12 + IL-18 compared with anequivalent number of TDLN cells generated with IL-12 alone, IL-18alone, or with neither (P < 0.05). We examined the overallproliferation of MCA 205 TDLN cells in IL-2 with the presence orabsence of IL-12 and/or IL-18 following anti-CD3/anti-CD28activation. In several independently done experiments, there wereno significant changes in resultant total CD3+ T-cell numbers withthe use of IL-12 or IL-18 alone, or both compared with that whenIL-12 or IL-18 was not used (data not shown). These studies thereforesuggested that the use of IL-12 and IL-18 did not change the potentialfor the whole TDLN populations to proliferate, but it may result inthe generation of effector cells with increased therapeutic efficacyon a per cell basis. Additional adoptive transfer experiments weredone to identify the effector cells responsible for tumor regressionafter exposure of antibody-activated TDLN cells to IL-12 and IL-18.Groups of mice bearing 3-day established MCA 205 pulmonarymetastases received IL-12/IL-18–cultured TDLN cells followed bythe i.p injection of RIgG, anti-CD4, anti-CD8, or anti-CD4 plus anti-CD8 depletingmAbs. All mice received 3� 106 cells unless otherwiseindicated. As shown in Fig. 4B , administration of either anti-CD4 oranti-CD8 partially abrogated the antitumor activity mediated by thetransferred cells. Anti-CD4 plus anti-CD8 completely abolished thetumor regression response. These experiments indicated that bothCD4+ and CD8+ TDLN cells mediated antitumor reactivity in vivoand that they contributed additive effects.The infiltration of lung metastases by adoptively transferred

TDLN cells grown in IL-2 alone or in combination with IL-12+ IL-18 following antibody activation was investigated by

Figure 1. Synergistic polarization effects of IL-12 and IL-18 on cytokinesecretion of anti-CD3/anti-CD28 activated TDLN cells. TDLN cells wereactivated with anti-CD3 plus anti-CD28 for 2 days and then cultured in IL-2 FIL-12 and/or IL-18 for 3 days. Culture supernatants were collected at the end ofcell expansion and cytokines released into the culture media were assessed. Thedata are representitve of three independently done experiments. a, P < 0.05compared with no IL-12, no IL-18; b, P < 0.05 compared with IL-12 (1 ng/mL)alone or IL-18 (10 or 100 ng/mL) alone; c, P < 0.05 compared with IL-12(10 ng/mL) alone or IL-18 (10 or 100 ng/mL) alone; d, P < 0.05 compared withIL-12 (1 ng/mL) alone; e, P < 0.05 compared with IL-12 (10 ng/mL) alone.

IL-12 and IL-18 Interactions involving Antitumor Immunity

www.aacrjournals.org 1065 Cancer Res 2005; 65: (3). February 1, 2005

Research. on July 21, 2015. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

immunocytochemistry to localize labeled TDLN cells in tissuesections (29). Briefly, TDLN cells were labeled with CFSEimmediately before infusion into animals bearing 10-day oldMCA 205 metastases. Ten-day metastases were used since theseare large enough to clearly visualize infiltration by the adoptivelytransferred cells. In contrast, preliminary studies of 3-daymetastases (not shown) indicated that these lesions were toosmall for unequivocal microscopic identification. Twenty-fourhours after infusion, lungs were harvested, sectioned andexamined for TDLN cell infiltration as described (29). Represen-tative sections from animals treated with TDLN cells cultured inIL-2 alone (Fig. 5A) or in IL-2 + IL-12 + IL-18 (Fig. 5B) areshown. In both cases, infused TDLN cells were observed to: (1)attach to venules, (2) mix with host leukocytes in perivenularcollections, and (3) infiltrate tumor nodules. Quantitativecomparison of TDLN cell trafficking into tumor noduleswas beyond the scope of this study but is under activeinvestigation.The Synergistic Effect of IL-12 and IL-18 in Skewing T-Cell

Responses toward the Type 1 Pattern Was NF-KB Dependent.To evaluate potential signaling pathways involved in the IL-12/IL-18 skewing effect, we examined the role of the transcription factorNF-nB. We used NF-nB p50�/� knockout mice and compared theimmune response of IL-12/IL-18–cultured NF-nB p50�/� TDLNcells with similarly treated wild-type TDLN cells. Using B6 NF-nBp50�/� knockout mice as TDLN donors, NF-nB p50�/� TDLN cellswere induced. The wild-type TDLN or NF-nB p50�/� TDLN cellswere then activated by anti-CD3/anti-CD28 followed by cell culturein IL-2 F IL-12 and/or IL-18. Supernatants at the end of cell culturewere collected to examine the IFNg and GM-CSF production (Fig. 6,T cells alone). The IL-12 and IL-18 induced IFNg and GM-CSFsecretion seen with wild-type TDLN cells was nearly absent insimilarly cultured NF-nB p50�/� TDLN cells. In addition, IL-12/IL-18–stimulated TDLN cells were cocultured with irradiated MCA205 tumor cells to assess cytokine secretion in response to tumorantigen (Fig. 6, T cells + MCA 205). In this setting, MCA 205-stimulated NF-nB p50�/� TDLN cells produced comparable levelsof IFNg (f1,000 pg/mL) and GM-CSF (f200 pg/mL) comparedwith wild-type TDLN cells when cultured in IL-2 alone afterantibody activation (open bars). This indicated that the TDLN cells

from NF-nB p50�/� knockout mice were generally responsive tothe effects of anti-CD3/anti-CD28 and IL-2 in vitro . However, theenhanced IFNg and GM-CSF production by IL-12/IL-18–stimulatedwild-type TDLN cells in response to MCA 205 tumor antigen wasminimal for similarly stimulated NF-nB p50�/� TDLN cells. Inadoptive transfer experiments, IL-12 + IL-18–cultured NF-nB p50�/�

TDLN cells mediated tumor regression, but the efficacy wassignificantly decreased when compared with IL-12 + IL-18–culturedwild-type TDLN cells (Fig. 7A). Collectively, these experimentsindicated the requirement of NF-nB in IL-12/IL-18–inducedimmune modulation.We proceeded to use an intracellularly targeted peptide inhibitor

(BMS-205820) of nuclear translocation as an approach to NF-nBinhibition in order to confirm that the role of NF-nB was less globalbut more specific for IL-12/IL-18. The inhibitor was used after thecompletion of anti-CD3/anti-CD28 activation but right before theadditionof IL-12/IL-18. As shown inFig. 7B , BMS-205820, at 5Amol/L,significantly inhibited IL-12/IL-18–induced IFNg secretion to alevel comparable to that achieved without the use of those cytokines.Control peptides SV40 or JBC had no effects.

Discussion

Ex vivo antibody activation of T cells is a viable approach forthe cellular treatment of cancer. However, the nondiscriminatingcharacter of antibodies in activating both type 1 and type 2responses may result in the generation of nontherapeutic effectorcells. We evaluated the immunomodulatory properties of IL-12and IL-18 in shifting the differentiation of antibody-activated TDLNcells in vitro . We have found that IL-12 and IL-18 synergisticallypotentiated IFNg and GM-CSF production of antibody-activatedTDLN cells whileminimally effecting IL-10 production. The IL-12/IL-18 skewing of effector T cells towards a type 1 response resulted inmore potent antitumor reactivity in vivo . IL-12 and IL-18 stimulatedCD4+ TDLN cells that could mediate tumor regression independentof CD8+ cells. The synergistic effect of IL-12 and IL-18 in skewing T-cell responses towards the type 1 phenotype was found to be NF-nBdependent. These results have potentially broad applications for thegeneration of effector T cells for clinical therapy.The use of IL-12 and/or IL-18 as a therapeutic agent for cancer,

autoimmune diseases, and infectious diseases has been explored by

Table 1. Cytokine secretion (pg/mL) in response to tumor antigen by anti-CD3/anti-CD28 activated TDLN cells that werecultured in IL-12 (10 ng/mL) + IL-18 (100 ng/mL) compared with no IL-12/no IL-18

Experiment IFNg GM-CSF IL-10

No IL-12,

no IL-18

IL-12 +

IL-18

*Fold change No IL-12,

no IL-18

IL-12 +

IL-18

Fold change No IL-12,

no IL-18

IL-12 +

IL-18

Fold change

1 322 10,377 32 5 105 21 452 295 0.72 49 780 16 5 132 26 9 10 1.1

3 1,401 17,034 12 n.d. n.d. n.d. 194 437 2.3

4 201 6,996 35 56 114 2 276 397 1.4

5 1,269 57,105 45 5 68 14 377 574 1.56 1,845 76,324 41 80 169 2 160 234 1.5

7 9,022 118,306 13 n.d. n.d. n.d. 487 432 0.9

Mean 28c

13c

1.3c

*Fold change was calculated by dividing the concentration when both IL-12 and IL-18 were used by the concentration when neither was used.cFold change comparing ‘‘IL-12+IL-18’’ versus ‘‘No IL-12/ No IL-18’’: P = 0.048 for IFNg, P = 0.002 for GM-CSF, P = 0.300 for IL-10.

Cancer Research

Cancer Res 2005; 65: (3). February 1, 2005 1066 www.aacrjournals.org

Research. on July 21, 2015. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

other investigators (27, 31–34). A recent study showed that IL-12and IL-18 are central mediators orchestrating Th1 and/or Th2immune responses to respiratory syncytial viral infection (34).Osaki et al. reported that the combination of IL-12 and IL-18administration resulted in a more potent antitumor responsecompared with each cytokine alone (33). However, they also foundthat administration of IL-12 and IL-18 was associated with lethalorgan damage, attributed in part to extremely high levels of host-induced IFNg production. In a different approach, the IL-18 genehas been transferred into dendritic cells, either alone (35, 36) orsimultaneously with the IL-12 gene (24), resulting in more effectiveantitumor responses associated with dendritic cell–based vaccines.As revealed by our results, the skewing of the immune response toa Th1/Tc1 reaction seems to be an important mechanism as to whythe combination of IL-12 and IL-18 can enhance antitumorreactivity. To document the levels of IL-12 and IL-18 that might

normally be present, we collected culture supernatants at varioustime points (days 1, 2, and 4) during standard TDLN cell activationwith anti-CD3/anti-CD28 and expansion in IL-2. IL-18 was notdetectable at any time points, whereas IL-12 was detected atmarginal levels (about 0.1 ng/mL, data not shown) compared withthe amount (10 ng/mL) we used.The mechanisms for the synergistic effects of IL-12 and IL-18 have

not been clearly defined. In the present study, the polarization effectof IL-12 and IL-18 in skewing tumor-reactive TDLN cell responsestoward the type 1 pattern was found to be NF-nB dependent. NF-nB isan inducible transcription factor affecting cells of the immunesystem (37–39). NF-nB is expressed as a cytosolic protein thattranslocates to the nucleus following cell activation, where itregulates the expression of a number of genes whose products areinvolved in inflammation and lymphocyte activation (30). In thisstudy, the TDLN cells fromNF-nB p50�/� knockoutmice were found

Figure 2. The synergistic effect of IL-18 inup-regulating IFNg secretion, whereasdown-regulating IL-10 secretion ofantibody-activated TDLN cells was tumorantigen specific. After antibody activation,MCA 205 TDLN cells were stimulated in IL-2containing medium with either IL-12 (10 ng/mL)or IL-18 (100 ng/mL) alone or with both. Thecells were then stimulated with irradiatedMCA 205 tumor cells for 24 hours. Culturesupernatants after tumor cell stimulation werecollected and analyzed for IFNg and IL-10secretion. MCA 207 tumor cells were used asa specificity control. The data arerepresentative of four independentexperiments. *, P < 0.05 compared with anyother groups; **, P < 0.05 compared withIL-12 (10 ng/mL) alone.

Figure 3. Cytokine secretion of IL-12 and/orIL-18 cultured CD4+ or CD8+ MCA205 TDLNcells. CD4+ or CD8+ T cells were positivelyselected as described in the Methods section,and activated by anti-CD3/anti-CD28 mAbsfollowed by cell culture in IL-2 with or withoutthe presence of IL-12 and/or IL-18 asindicated. Cytokine secretion of 1 � 106 ofeach cell group in response to MCA205tumor stimulation was analyzed. Representativeof two independently done experiments. *,P < 0.05 compared with any other groupsof CD4+ or CD8+ cells respectively; **,P < 0.05 compared with IL-12 (10 ng/mL)alone for CD4+ or CD8+ cells.

IL-12 and IL-18 Interactions involving Antitumor Immunity

www.aacrjournals.org 1067 Cancer Res 2005; 65: (3). February 1, 2005

Research. on July 21, 2015. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

responsive to the effects of anti-CD3/anti-CD28 and IL-2 in vitro.However, these cells were not responsive to IL-12/IL-18 in inducingIFNg secretion or in augmenting their therapeutic efficacy. The useof NF-nB–specific inhibitor BMS-205820 confirmed that the role ofNF-nB was less global but more specific for IL-12/IL-18.CD8+ T cell responses have been reported when IL-12 + IL-18 was

used to induce antitumor responses (24). It is noteworthy that inthis study we were able to generate CD4+ T-cell responses inaddition to CD8+ cells with the combination of antibody activationfollowed by IL-12/IL-18 culture. We have previously shown thatboth CD4+ and CD8+ tumor-reactive cells can be generated fromTDLN after anti-CD3/anti-CD28 activation (15, 19). The subsequentexposure of these activated cells to IL-12 + IL-18 further increasedthe proliferation of CD4+ cells. In addition, the skewing of cytokineresponse to tumor antigen was more pronounced in the CD4+

subpopulation than in CD8+ cells.Previous studies by Plautz et al. showed that cultured TDLN cells

must infiltrate pulmonary nodules to suppress tumor growth (40).

Immunohistochemical localization of the TDLN cells in the currentstudy showed active migration of infused cells into pulmonarytumor nodules whether the TDLN cells were cultured with orwithout IL-12 + IL-18. Because TDLN cells cultured with IL-12 +IL-18 increased tumor antigen–specific IFNg and GM-CSFsecretion in vitro , it is likely that these cytokines contributed tothe enhanced therapeutic efficacy in vivo .Son et al. has reported that cell culture in IL-2 and IL-18 resulted

in enhanced NK activity (25). The activation method we used forTDLN cells involved expansion in IL-2. However, NK cells werefound to be dispensable in the tumor rejection response mediatedby the adoptively transferred TDLN cells cultured in IL-12/IL-18.NK cells comprised f5% of the freshly harvested whole TDLN cellpopulations. After anti-CD3/anti-CD28 activation followed byIL-12/IL-18 polarization, the percentage of NK cells was <2% (datanot shown). We depleted NK cells in vivo after the adoptive transferof antibody-activated and IL-12/IL-18 cultured TDLN cells andfound no significant decrease in the therapeutic efficacy (data not

Figure 4. IL-12/IL-18 polarized effector cells mediated tumor regression in vivo .A, TDLN cells cultured with IL-12 + IL-18 showed more potent therapeuticefficacy. B6 mice with 3-day established MCA 205 pulmonary metastases weretreated by adoptive transfer of MCA 205 TDLN cells activated with anti-CD3/anti-CD28 mAbs followed by cell culture in IL-2 F IL-12 and/or IL-18. Differentnumbers of TDLN cells were transferred. IL-2 (30,000 IU) was given i.p. bid foreight doses after cell infusion. Data are representative of four independentexperiments. *, P < 0.05 compared with any other group when an equal numberof cells were transferred. B, IL-12/IL-18 cultured CD4+ and CD8+ TDLN cellsindependently mediated tumor regression. Immediately following cell transfer asin A , groups of mice receiving IL-12/IL-18 cultured TDLN cells were given anti-CD4 and/or anti-CD8 mAbs to deplete T-cell subpopulations in vivo . RIgG wasgiven to a separate group of animals as a control for the depleting antibodies.Data are representative of three independent experiments. *, P < 0.05 comparedwith any other groups except for RIgG control; **, P < 0.05 compared with the notreatment groups.

Figure 5. IL-12 and IL-18-polarized TDLN cells accumulated in pulmonaryMCA205 tumor nodules. TDLN cells were cultured either in IL-2 alone (A) or inIL-2+IL-12+IL-18 (B ) following antibody activation. The lymphoblasts were thenlabeled with CFSE and infused (i.v.) into mice with 10-day pulmonary MCA 205metastases. After 24 hours, the lungs were processed as described in Materialsand Methods. Examples of infiltration TDLN cells (black arrows ) and tumor nuclei(white arrows ) are shown.

Cancer Research

Cancer Res 2005; 65: (3). February 1, 2005 1068 www.aacrjournals.org

Research. on July 21, 2015. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

shown). On the other hand, depletion of both CD4+ and CD8+ cellsfollowing adoptive transfer completely abrogated the antitumorreactivity. Hence, it was unlikely that either host NK cells ortransferred NK cells played a significant role in the regression oftumor in these experiments.During the generation of antitumor effector T cells, it is crucial

to identify in vitro correlates of T-cell function that are associatedwith in vivo therapeutic efficacy. We have determined theimportance of IFNg secretion in mediating the antitumor effectsof the TDLN cells both in animal studies (3, 16) and in clinical trials(7). In an animal study, we examined the TCR Vh repertoire usageof TDLN cells with respect to IFNg release and therapeutic efficacy.Enrichment of Vh subsets of TDLN cells by in vitro activation withanti-Vh mAb revealed that Vh8+ cells released high amounts ofIFNg in response to tumor and mediated potent tumor regressionin vivo . Depletion of Vh8+ subset from the whole TDLN poolconfirmed that IFNg production correlated with in vivo tumoreradication (3). In a separate study, we found that co-stimulation ofTDLN cells through newly induced 4-1BB and CD3/CD28 signalingcan significantly increase antitumor reactivity. The augmentedantitumor effect through 4-1BB and CD28 costimulation wasdependent on IFNg production, because tumor regression wasabrogated by IFNg neutralization after adoptive transfer of 4-1BB/CD28-costimulated TDLN cells (16). We have also shown that IFNgsecretion by the activated vaccine-primed lymph node cellscorrelates with tumor response in a phase II adoptive cellular trial

in patients with advanced renal cell cancers. We found that theIFNg: IL-10 ratio of cytokine released by effector T cells in responseto tumor antigen was associated with clinical outcomes. Specifi-cally, activated T cells that have an increased IFN-g/IL-10 ratiocorrelated with tumor response (7). Therefore, the use of polarizingreagents to modulate T-cell function towards the type 1 responseand/or down-regulate type 2 responses may offer a rationalstrategy in generating effector cells for cellular therapy.The findings in this study indicate that proinflammatory

cytokines IL-12 and IL-18 promote the differentiation of Th1 andTc1 effector cells. Moreover, the observations that IL-18 signifi-cantly inhibited both Th2 and Tc2 cytokine production of IL-12–cultured TDLN cells represent novel findings. The mechanismsinvolved in this phenomenon remain to be defined.

Figure 6. IL-12/IL-18-induced skewing of antibody-activated TDLN cellstowards the type 1 pattern was NF-nB dependent. TDLN induced from wild-type or NF-nB p50�/� KO B6 mice were designated as wt TDLN or NF-nB�/�

TDLN respectively. The wild-type TDLN or NF-nB�/� TDLN cells were thenactivated with anti-CD3/anti-CD28 followed by culture in IL-2 F IL-12 and/orIL-18. Supernatants at the end of cell culture were collected to examine IFNg

and GM-CSF secretion (T cells alone). The cultured MCA205 TDLN cells werestimulated with MCA 205 tumor cells to assess cytokine secretion in response totumor antigen (T cells + MCA 205). These findings were reproduced in a secondindependent experiment. *P < 0.05 compared with all other groups (T cellsalone). **, P < 0.05 compared with all other groups (T cells + MCA 205).

Figure 7. NF-nB dependent IL-12 and IL-18 interactions in TDLN cells.A, generation of potent therapeutic effector cells with IL-12 + IL-18 requiredNF-nB. Wt TDLN and NF-nB p50�/� TDLN cells were activated withanti-CD3/anti-CD28 followed by culturing in IL-2 and IL-12 +IL-18. B6 mice with3-day established MCA 205 pulmonary metastases were treated by adoptivetransfer of 1 � 106 activated and cultured TDLN cells. IL-2 was given i.p. bid foreight doses after cell transfer. Data are representative of two independentexperiments. *, P < 0.05 compared with ‘‘No treatment’’ or ‘‘IL-2 only’’ groups. **,P < 0.01 compared with ‘‘Wt TDLN No IL-12/IL-18’’ or ‘‘NF-nB�/� TDLN IL-12 +IL-18’’ groups. B, NF-nB inhibitor BMS-205820 abrogated IL-12/IL-18-inducedIFNg production. Wild-type TDLN cells were activated with anti-CD3/anti-CD28mAb for 2 days, washed, and then incubated with BMS-205820, SV40 and JBCfor 2 hours in IL-2 containing medium before addition of IL-12 and IL-18 for cellculture. Culture supernatants were then collected for IFNg assay. Similar resultswere obtained in three independent experiments. *, P < 0.05 compared with allother groups except for the group without the use of IL-12/IL-18.

IL-12 and IL-18 Interactions involving Antitumor Immunity

www.aacrjournals.org 1069 Cancer Res 2005; 65: (3). February 1, 2005

Research. on July 21, 2015. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Acknowledgments

Received 2/23/2004; revised 11/8/2004; accepted 11/22/2004.Grant support: NIH grants CA82529 and CA69102, the Gillson Longenbaugh

Foundation, and the Harshman Family.

The costs of publication of this article were defrayed in part by the payment of pagecharges. This article must therefore be hereby marked advertisement in accordancewith 18 U.S.C. Section 1734 solely to indicate this fact.

We thank Kerry Odneal for her excellent assistance in the preparation of this

article.

References1. Barth RJ, Mule JJ, Speiss PJ, Rosenberg SA. Interferon g

and tumor necrosis factor have a role in tumorregressions mediated by murine CD8+ tumor-infiltratinglymphocytes. J Exp Med 1991;173:647–58.

2. Schwartzentruber DJ, Solomon D, Rosenberg SA,Topalian SL. Characterization of lymphocytes infiltrat-ing human breast cancer: specific immune reactivitydetected by measuring cytokine secretion. J Immunoth1992;12:1–12.

3. Aruga A, Aruga E, Tanigawa K, Bishop DK, Sondak VK,Chang AE. Type 1 versus type 2 cytokine release by VhT cell subpopulations determines in vivo antitumorreactivity: IL-10 mediates a suppressive role. J Immunol1997;159:664–73.

4. Dobrzanski MJ, Reome JB, Dutton RW. Therapeuticeffects of tumor-reactive type 1 and type 2 CD8+ T cellsubpopulations in established pulmonary metastases.J Immunol 1999;162:6671–80.

5. Nakajima C, Uekusa Y, Iwasaki, et al. A role ofinterferon-g in tumor immunity: T cells with thecapacity to reject tumor cells are generated but fail tomigrate to tumor sites in IFN-g-deficient mice. CancerRes 2001;61:3399–405.

6. Segal JG, Lee NC, Tsung YL, Norton JA, Tsung K. Therole of IFN-g in rejection of established tumors byIL-12: source of production and target. Cancer Res 2002;62:4696–703.

7. Chang AE, Li Q, Jiang G, Sayre DM, Braun TM, Redman BG.Phase II trial of autologous tumor vaccination, anti-CD3-activated vaccine-primed lymphocytes, and inter-leukin-2 in stage IV renal cell cancer. J Clin Oncol2003;21:884–90.

8. Zhu LX, Sharma S, Gardner B, et al. IL-10 mediatessigma receptor-dependent suppression of antitumorimmunity. J Immunol 2003;170:3585–91.

9. Halak BK, Maguire HC, Lattime EC. Tumor-inducedinterleukin-10 inhibits type 1 immune responsesdirected at a tumor antigen as well as a non-tumorantigen present at the tumor site. Cancer Res1999;59:911–7.

10. Igietseme JU, Ananaba GA, Bolier J, et al. Suppressionof endogenous IL-10 gene expression in dendritic cellsenhances antigen presentation for specific Th1 induc-tion: potential for cellular vaccine development.J Immunol 2000;164:4212–9.

11. Yang AS, Lattime EC. Tumor-induced interleukin 10suppresses the ability of splenic dendritic cells tostimulate CD4 and CD8 T-cell responses. Cancer Res2003;63:2150–7.

12. Yoshizawa H, Chang AE, Shu S. Specific adoptiveimmunotherapy mediated by tumor-draining lymphnode cells sequentially activated with anti-CD3 andIL-2. J Immunol 1991;147:729–37.

13. Yoshizawa H, Chang AE, Shu S. Cellular interactionsin effector cell generation and tumor regressionmediated by anti-CD3/interleukin 2 activated tumordraining lymph node cells. Cancer Res 1992;52:1129–36.

14. Chang AE, Aruga A, Cameron MJ, et al. Adoptiveimmunotherapy with vaccine-primed lymph node cellssecondarily activated with anti-CD3 and interleukin-2.J Clin Oncol 1997;15:796–807.

15. Li Q, Yu B, Grover A, Zeng X, Takeshita N, Chang AE.Therapeutic effects of tumor reactive CD4+ cellsgenerated from tumor-primed lymph nodes usinganti-CD3/anti-CD28 monoclonal antibodies. J Immu-noth 2002;25:304–13.

16. Li Q, Carr A, Ito F, Teitz-Tennenbaum S, Chang AE.Polarization effects of 4-1BB during CD28 costimulationin generating tumor-reactive T cells for cancer immu-notherapy. Cancer Res 2003;63:2546–52.

17. Kagamu H, Shu S. Purification of L-selectinlow cellspromotes the generation of highly potent CD4 anti-tumor effector T lymphocytes. J Immunol 1998;160:3444–52.

18. Chirathaworn C, Kohlmeier JE, Tibbetts SA, RumseyLM, Chan MC, Benedict SH. Stimulation throughintercellular adhesion molecule-1 provides a secondsignal for T cell activation. J Immunol 2002;168:5530–7.

19. Li Q, Furman SA, Bradford CA, Chang AE. Expandedtumor-reactive CD4+ T cells responses to humancancers induced by secondary anti-CD3/anti-CD28activation. Clin Cancer Res 1999;5:461–9.

20. Hart-Meyers J, Ryu A, Monney L, et al. Cutting Edge:CD94/NKG2 is expressed on Th1 but not Th2 cells andcostimulates Th1 effector functions. J Immunol 2002;169:5382–6.

21. Hou W, Wu Y, Sun S, et al. Pertussis toxin enhancesTh1 responses by stimulation of dendritic cells.J Immunol 2003;170:1728–36.

22. Hodge JW, Sabzevari H, Yafal A, Gritz L, Lorenz M,Schlom J. A triad of costimulatory molecules synergizeto amplify T-cell activation. Cancer Res 1999;59:5800–7.

23. Kemp R, Ronchese F. Tumor-specific Tc1, but notTc2, cells deliver protective antitumor immunity.J Immunol 2001;167:6497–502.

24. Tatsumi T, Huang J, Gooding WE, et al. Intratumoraldelivery of dendritic cells engineered to secrete bothinterleukin (IL)-12 and IL-18 effectively treats local anddistant disease in association with broadly reactive Tc1-type immunity. Cancer Res 2003;63:6378–86.

25. Son Y, Dallal RM, Mailliard RB, Egawa S, Jonak ZL,Lotze MT. Interleukin-18 (IL-18) synergizes with IL-2to enhance cytotoxicity, interferon-g production, andexpansion of natural killer cells. Cancer Res 2001;61:884–8.

26. Baxevanis CN, Gritzapis AD, Papamichail M. In vivoantitumor activity of NKT cells activated by the

combination of IL-12 and IL-18. J Immunol 2003;171:2953–9.

27. Leung BP, Culshaw S, Gracie JA, et al. A role for IL-18in neutrophil activation. J Immunol 2001;167:2879–86.

28. Barth RJ, Bock SN, Mule JJ, Rosenberg SA. Uniquemurine tumor-associated antigens identified by tumorinfiltrating lymphocytes. J Immunol 1990;144:1531–7.

29. Skitzi J, Craig RA, Okuyama R, Knibbs RN, McDonagh K,Chang AE, Stoolman LM. Donor cell cycling, trafficking,and accumulation during adoptive immunotherapy formurine lung metastases. Cancer Res 2004;64:2183–91.

30. Fujihara SM, Cleaveland JS, Grosmaire LS, et al. Ad-amino acid peptide inhibitor of NF-nB nuclearlocalization is efficacious in models of inflammatorydisease. J Immunol 2000;165:1004–12.

31. Smyth MJ, Taniguchi M, Street S. The anti-tumoractivity of IL-12: mechanisms of innate immunity thatare model and dose dependent. J Immunol 2000;165:2665–70.

32. Halin C, Gafner V, Villani ME, et al. Synergistictherapeutic effects of a tumor targeting antibodyfragment, fused to interleukin 12 and to tumor necrosisfactor. Cancer Res 2003;63:3202–10.

33. Osaki T, Peron JM, Cai Q, et al. IFN-g-inducingfactor/IL-18 administration mediates IFN-g-and IL-12-independent antitumor effects. J Immunol 1998;160:1742–9.

34. Wang SZ, Bao YX, Rosenberget CL, Tesfaigzi Y,Stark JM, Harrod KS. IL-12 and IL-18 modulate inflam-matory and immune responses to respiratory syncytialvirus infection. J Immuno 2004;173:4040–9.

35. Tatsumi T, Gambotto A, Robbins PD, Storkus WJ.Interleukin 18 gene transfer expands the repertoire ofantitumor Th1-type immunity elicited by dendritic cell-based vaccines in association with enhanced therapeu-tic efficacy. Cancer Res 2002;62:5853–8.

36. Ju DW, Tao Q, Lou G, et al. Interleukin 18 transfectionenhances antitumor immunity induced by dendriticcell-tumor cell conjugates. Cancer Res 2001;61:3735–40.

37. Grilli M, Chiu J, Lenardo MJ. NF-nB and Re1:participants in a multiform transcriptional regulatorysystem. Int Rev Cytol 1993;143:1–62.

38. Muller JM, Ziegler-Heitbrock H, Baeuerle PA. Nuclearfactor nB, a mediator of lipopolysaccharide effects.Immunobiol 1993;187:233–56.

39. Corn RA, Aronica MA, Zhang F, et al. T cell-intrinsicrequirement for NF-nB induction in postdifferentiationIFN-g production and clonal expansion in a Th1response. J Immunol 2003;171:1816–24.

40. Mukai S, Kjaergaard J, Shu S, Plautz GE. Infiltrationof tumors by systemically transferred tumor-reactiveT lymphocytes is required for antitumor efficacy.Cancer Res 1999;59:5245–9.

Cancer Research

Cancer Res 2005; 65: (3). February 1, 2005 1070 www.aacrjournals.org

Research. on July 21, 2015. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

2005;65:1063-1070. Cancer Res   Qiao Li, Abbey L. Carr, Elizabeth J. Donald, et al.   Tumor-Reactive T-Cell Responses Towards a Type 1 PatternSynergistic Effects of IL-12 and IL-18 in Skewing

  Updated version

  http://cancerres.aacrjournals.org/content/65/3/1063

Access the most recent version of this article at:

   

   

  Cited articles

  http://cancerres.aacrjournals.org/content/65/3/1063.full.html#ref-list-1

This article cites 39 articles, 36 of which you can access for free at:

  Citing articles

  http://cancerres.aacrjournals.org/content/65/3/1063.full.html#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

  [email protected] at

To request permission to re-use all or part of this article, contact the AACR Publications

Research. on July 21, 2015. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from