Experimental tumor vaccination and adoptive T-cell therapies show that interferon-γ (IFN-γ)–producing CD4+ T helper cells (Th1) can be highly effective in tumor prevention and therapy. Unexpectedly, first vaccine trials in humans revealed that tumor immune therapy may not only be protective, but, on the contrary, even promote tumor progression. Here, we analyzed T-cell immune responses to the epithelial cell adhesion molecule (EpCAM), one of the most common tumor-associated antigens (TAA) serving as immune target in colon cancer patients. Th-cell priming against EpCAM inevitably resulted in interleukin-4 (IL-4)–dominated Th2 responses, even under most stringent Th1-inducing conditions. These EpCAM-reactive Th2 cells rather promoted growth of EpCAM-expressing tumors. To analyze the role of IL-4 in tumor immune evasion, we generated EpCAM-reactive Th1 cells from IL-4.ko mice. These Th1 cells provided tumor-specific protection and established highly protective Th1 memory responses, even in naive BALB/c mice. Inhibition of tumor growth by Th1 cells resulted in intra-tumoral expression of cytokines of the IL-12 family and of IFN-γ. Preventing activation-associated death of Th1 cells further increased intratumoral IFN-γ expression and improved therapeutic efficacy. Thus, human TAA may promote tumor immune evasion by strongly favoring Th2 development.

Epithelial cell adhesion molecule (EpCAM), a human cell-surface glycoprotein, was identified as a human tumor-associated antigen (TAA) in 1979 and was recently described as cancer stem cell marker. EpCAM is a panepithelial differentiation antigen that is restricted to normal epithelia in healthy subjects, but is typically overexpressed to various degrees in most human carcinomas, making it an attractive target for tumor therapy.1  Several clinical trials with monoclonal2  or bispecific antibodies and vaccination strategies directed against EpCAM showed variable success in the treatment of carcinomas.1  Development of EpCAM-specific T cell–based strategies were suggested by in vitro studies with either chimeric T-cell receptor (TCR)–transduced primary T lymphocytes3  or TAA-specific cytotoxic T lymphocytes (CTL),4  showing cytolytic activity against EpCAM-expressing tumor cells.

T cell–based therapies focus either on active immunization5,6  or on adoptive transfer of in vitro–generated CD4+ or CD8+ T-cell lines.7,8  As vaccination is easier to perform and has long been tested, most studies focus on active immunization. For active immunization antigens are administered either with different adjuvants or using tumor antigen–loaded dendritic cells (DCs) as a “natural adjuvant.”9,10  Although such vaccination strategies are used in the clinic, the therapeutic efficiency is still limited.5,6,11  One reason might be that immunization induces efficient T-cell expansion, but T cells fail to control tumor growth.12  Moreover, it is possible that tumors themselves actively paralyze tumor immune responses as shown in various models of tumor disease.12-15  Thus, in mice that express the transgenic tumor promoter “T antigen” (Tag) and simultaneously the Tag-specific TCR, the growing tumors both paralyze and delete Tag-reactive T cells.14  Tag-specific T cells from mice with established tumors show severe defects in proliferation and cytokine production.14  Moreover, tumors can also actively induce regulatory T cells.16  Such negative regulators of protective immunity complicate active induction of therapeutically efficient tumor immune responses, even with the use of optimized DCs as an adjuvant.17 

Adoptive transfer of in vitro–generated TAA-specific T cells can circumvent many of these problems. This approach has been extensively explored for the treatment of established experimental tumors in several mouse models18-20  and even in humans.21,22  Early studies used in vitro–primed spleen cells from immunized mice or CD8+ CTLs.7,20  Subsequent studies with in vitro–generated T-cell subpopulations revealed that interferon-γ (IFN-γ)–producing CD4+ T-helper cells (Th1) are therapeutically more effective than either CD8+ CTLs or Th2 cells.18,20,23,24  In contrast to initial reports,25,26  we and others clearly showed that interleukin-4 (IL-4)–producing Th2 cells fail to provide protection against tumor cells, except if IL-4 is used for in vivo priming of DCs.27  Importantly, the studies published until now were performed by priming CD4+ T cells either against major histocompatibility complex (MHC) class II–positive tumors20  or against model antigens transfected into tumor cell lines such as ovalbumin (OVA).28 

To mimic a clinically closer situation, we immunized mice against one of the most important human TAA, EpCAM. The in vivo–primed T cells were used to generate EpCAM-reactive CD4+ T-cell lines in vitro for adoptive tumor therapy. The capacity of these T-cell lines to control the growth of EpCAM-transfected CT26 colon carcinoma cell lines (CT26-EpCAM) was analyzed. EpCAM-reactive CD4+ T cells from wild-type BALB/c mice inevitably developed an IL-4–producing Th0 or Th2 phenotype that provided no protection. Surprisingly, we were unable to generate EpCAM-reactive Th1 cells from wild-type BALB/c mice. In contrast, EpCAM-reactive Th1 cells were readily generated from IL-4.ko BALB/c mice. These Th1 cells established protective tumor immunity, even in naive BALB/c mice. Protective immunity induced intratumoral expression of antiangiogenic IFN-γ,29,30  IL-17,31  and innate cytokines of the IL-12 family.32-37  In consequence, tumor growth inhibition correlated with infiltration of CD11b+ monocytes/macrophages, T cells, and endothelial cell (EC) damage. Thus, EpCAM promotes tumor immune evasion through the induction of Th2-development.

Mice

Female wild-type BALB/c and DO11.10 TCR-transgenic mice were purchased from Charles River Laboratories (Wilmington, MA). IL-4.ko BALB/c mice were kindly provided by Dr M. Kopf (ETH, Zürich, Switzerland). The animals were maintained in the animal facilities of the Eberhard Karls University (Tübingen, Germany) under specific pathogen-free conditions. Animal experiments were approved by the Institutional Animal Care and Use Committee of the Regierungspräsidium Tübingen (HT01/03) and of the Regierung of Oberbayern (AZ 211-2531-63/98).

Cell culture, antibodies, proteins, peptides

CT26-EpCAM cells were cultured in Dulbecco modified Eagle medium (DMEM; 4.5 g glucose) supplemented with 10% (vol/vol) fetal calf serum (FCS), 100 U/mL penicillin-G, and 100 μg/mL streptomycin. Isolated T cells were cultured in DMEM supplemented with 10% (vol/vol) FCS, 1% MEM amino acids (stock solution, 50×; Biochrom AG, Berlin, Germany), 10 μM N-2-hydroxyethylpiperazine-N′-2-ethanesulfonic acid (HEPES) buffer, 1 mM sodium pyruvate, 100 U/mL penicillin-G, 100 μg/mL streptomycin, and 40 μM 2-mercaptoethanol. Cell lines were maintained at 37°C in a 7.5% CO2 humidified incubator. CT26-EpCAM cells and the murine anti–IFN-γ (XMG-1.2) antibody for in vitro culture were from R.M. (Institut für Molekulare Immunologie [IMI], München, Germany). Murine anti–IL-4 (11B11) antibody was kindly supplied by Prof E. Schmidt (Institut für Molekulargenetik [IMGB], Mainz, Germany), and EpCAM protein (s17-1A) was friendly gifted by Prof E. Kopp (Institute of Immunology [IIM], Muenchen, Germany). OVA 323-339 peptide (ISQAVHAAHAEINEAGR) was purchased from EMC Microcollections (Tübingen, Germany). OVA protein was purchased from Sigma-Aldrich (St Louis, MO).

Immunization and in vitro T-cell differentiation

Either wild-type or IL-4.ko BALB/c mice were immunized twice subcutaneously with 106 CT26-EpCAM cells dispended in 50 μL CpG-ODN1668 (200 μM, 5′-tccatgacgttcctgatgct-3′, phosphorothioate [PTO] oligonucleotide; MWG Biotech, Ebersberg, Germany). Wild-type BALB/c mice were immunized with 100 μg OVA protein suspended in water with 200 μM CpG-ODN1668. CD4+ T cells were isolated from spleen and lymph nodes (LN) 6 days after immunization using mouse anti-CD4 (L3T4) magnetic beads (MACS; Miltenyi Biotec, Gladbach, Germany). OVA- or EpCAM-reactive CD4+ T cells (1-5 × 105) were stimulated with 106 syngeneic and radiated (30 Gy) T cell–depleted splenocytes (antigen-presenting cells [APC]) 100 μg/mL OVA protein or 1 μg/mL EpCAM protein, and 0.2 μM CpG1668 and expanded for 14 days (short-term culture) with 50 U/mL IL-2 (Chiron Therapeutics, Emeryville, CA). For long-term culture, cells were restimulated with EpCAM, APCs, and CpG every 2 weeks.

For OVA T-cell culture, CD4+ cells were isolated from DO11.10 transgenic mice as described above. OVA CD4+ T cells (2.5 × 105) were stimulated with 5 × 105 APCs, 0.2 μM CpG1668, 5 μg/mL OVA peptide, and 2 μg/mL murine anti–IL-4 (11B11) antibody.

Experimental procedures

For survival studies, CT26-EpCAM tumor cells (106) cell therapy was dispended in 100 μL PBS were injected intravenously in BALB/c mice. T-cell therapy was performed with intravenously injected (5 × 105) in vitro–cultured CD4+ cells.

To analyze the mechanisms, CT26-EpCAM tumor cells (1.5 × 106) dispended in 50 μL PBS were injected subcutaneously in the right and left flank of BALB/c mice (day 0). The mice were irradiated (2 Gy) on day 3 to induce a mild inflammatory response. On day 4, these mice received either cultured Th1 cells (5 × 106) or PBS intraperitoneally. Tumors were measured with a sliding calliper on days 10, 13, and 17 after tumor cell injection, and tumor area was calculated as product of perpendicular tumor diameters. Mice were killed at the indicated time after injection, and tumors were harvested for further analysis.

In another approach, one group of mice received 500 μg murine anti–IFN-γ antibody (XMG 1.2) intraperitoneally after irradiation on day 3 and before adoptive transfer of T cells on day 4, as indicated.

Fluorescence-activated cell-sorting analysis

For polyclonal T-cell stimulation, 106 T cells were activated with 30 ng/mL phorbol-12-myristate-13-acetate (PMA), and 1.5 mg/mL ionomycin. Cells were treated with brefeldin A (concentration as instructed; BD Biosciences, San Jose, CA), fixed in 2% (vol/vol) formaldehyde in PBS, and permeabilized with 0.5% (wt/vol) saponin in flow cytometry buffer (0.5%, wt/vol, bovine serum albumin in PBS). Cells were stained for intracellular IFN-γ (XMG-1.2; BioLegend, San Diego, CA), IL-4 (11B11; BioLegend), IL-10 (JES5-16E3; BD Pharmingen, San Diego, CA), or IL-17 (TC11-18H10; BD Biosciences) in combination with surface CD4 (RM4-5; BioLegend) for fluorescence-activated cell-sorting (FACS) analysis. Cells were acquired on the FACSCalibur (BD Biosciences) and analyzed using CellQuest Pro software (BD Biosciences).

For characterization of tumor-infiltrating cells, tumors were excised from mice on the indicated days after injection, cut with Medimax (Günter Keul, Steinfurt, Germany), digested using collagenase D (Roche, Indianapolis, IN), and single cells were stained using the following anti–mouse antibodies: fluorescein isothiocyanate (FITC)–CD4 (RM4-5; BioLegend), phycoerythrin (PE)–CD8 (Ly.2; BD Pharmingen), FITC-CD11b (Mac-1; BD Pharmingen), allophycocyanin-CD11c (N418; Caltag Laboratories, Burlingame, CA), biotinylated anti–mouse F4/80 (BM8; BMA Biomedicals, Augst, Switzerland) with a secondary PE-labeled streptavidin antibody (BD Pharmingen), and the corresponding isotype rat IgG antibodies. Cells (30 000) were acquired on the LSRII (BD Biosciences) and analyzed using FCS Express software (De Novo Software, Los Angeles, CA). Total living cells were gated by forward/sideward scatter, and infiltrating cells were calculated on the basis of 10 000 living cells.

Enzyme-linked immunosorbent assay

LN-derived T cells (0.1 × 106 cells/well) were stimulated with: (1) APCs alone (0.5 × 106 cells/well), (2) APCs and EpCAM (1 μg/mL), or (3) medium as negative control. Supernatants were then collected after 48 hours for further treatment. For detection of IFN-γ, IL-4, IL-10, and transforming growth factor-β (TGF-β) in supernatants of activated T cells and subsequent calculation of the protein content, sandwich enzyme-linked immunosorbent assays (ELISAs) were performed according to the manufacturers' instructions (IFN-γ and TGF-β; R&D Systems, Minneapolis, MN; and IL-4 and IL-10; BD Biosciences).

Quantitative real-time polymerase chain reaction

Total RNA was isolated from whole tumor tissue. Tissue was homogenized using the Mixer Mill (Retsch, Haan, Germany). RNA was purified according to the manufacturers' instructions (Macherey-Nagel, Düren, Germany) and converted to cDNA using Moloney murine leukemia virus (MMLV) reverse transcriptase (Promega, Madison, WI). Amplification was performed using the iCycler iQ instrument and the SYBR Green supermix (both from Bio-Rad Laboratories, Hercules, CA). For FoxP3, real-time polymerase chain reaction (PCR) was performed using the Light Cycler 405 and the SYBR Green mix (both from Roche). Specific primers (MWG Biotech) with the following sequences were used: hypoxanthine-guanine phosphoribosyltransferase (HPRT): 5′-GTTCTTTGCTGACCTGCTGGAT-3′ (forward), 5′-CTTAGGCTTTGTATTTGGCTTT-3′ (reverse); IL-23/p19: 5′-CATGGAGCAACTTCACACCTC-3′ (forward), 5′-GGTGATCCTCTGGCTGGA-3′ (reverse); IL-12/p35: 5′-ATGACCCTGTGCCTTGGTAG-3′ (forward), 5′-CAGATAGCCCATCACCCTGT-3′ (reverse); IFN-γ: 5′-TCAAGTGGCATAGATGTGGAAGAA-3′ (forward), 5′-TGGCTCTGCAGGATTTTCATG-3′ (reverse); intercellular adhesion molecule (ICAM): 5′-CCTGTTTCCTGCCTCTGAAG-3′ (forward), 5′-GTCTGCTGAGACCCCTCTTG-3′ (reverse); IL-6: 5′-CCGGAGAGGAGACTTCACAG-3′ (forward), 5′-CAGAATTGCCATTGCACAAC-3′ (reverse); IL-27/Ebi3: 5′-GCTCCCCTGGTTACACTGAA-3′ (forward), 5′-ACCGAGAAGCATGGCATT-3′ (reverse); IL-17: 5′-TCAGACTACCTCAACCGTTCC-3′ (forward), 5′-CTTTCCCTCCGCATTGACAC-3′ (reverse); CXCL10: 5′-TCCCTCTCGCAAGGACGGTC-3′ (forward), 5′-GTGTGTGCGTGGCTTCACTC-3′ (reverse); FoxP3: 5′-ACTCGCATGTTCGCCTACTT-3′ (forward), 5′-GTCCACACTGCTCCCTTCTC-3′ (reverse). Multiplex reactions were run in triplicates (for FoxP3, in duplicates), and samples were normalized to HPRT. The relative expression of genes of interest is represented as fold differences of the mean of treated tumor samples to the mean of control tumor samples. The expression of CXCL9, c-Myc, and CyclinD2 was measured semiquantitatively: CXCL9: 5′-TTTTCCTTTTGGGCATCATCTT-3′ (forward), 5′-AGCATCGTGCATTCCTTATCACT-3′ (reverse); c-Myc: 5′-AGCTGTTTGAAGGCTGGATT-3′ (forward), 5′-CCGCAACATAGGATGGAGAG-3′ (reverse); CyclinD2: 5′-ATTTCAAGTGCGTGCAGAAG-3′ (forward), 5′-ACACTTCTGTTCCTCACAGACCT-3′ (reverse).

Histology, immunohistochemistry, and electron microscopy

Tumors obtained from mice at day 18 after injection were either fixed in 1% (wt/vol) paraformaldehyde (PFA), embedded in paraffin, and cut into 7-μm sections or were directly embedded in tissue-freezing medium (Tissue tec; Jung/Leica, Wetzlar, Germany), snap-frozen, and cut in 10-μm cryosections. Paraffin sections were immunostained with anti–mouse Ki-67 (0.3 mg/mL; Dako, Glostrup, Denmark) and acetone-fixed cryosections with anti–mouse CD4 (0.01 mg/mL; BD Pharmingen). Positive immunostaining was detected with a streptavidin-biotin immunoperoxidase system (Vector Laboratories, Burlingame, CA). Sections were counterstained with hematoxylin (Chem Mate; Dako). Immunofluorescence staining of acetone-fixed cryosections was performed with anti–platelet/endothelial cell adhesion molecule-1 (anti–PECAM-1; BD Pharmingen) or anti–vascular endothelial growth factor receptor-2 (anti–VEGFR-2) antibody (Santa Cruz Biotechnology, Santa Cruz, CA) and a secondary Cy3-labeled antibody (Molecular Probes, Invitrogen, Carlsbad, CA). Sections were counterstained with Yopro1 (Molecular Probes, Invitrogen). Isotype-matched biotinylated rat IgG served as negative control in all stainings. For routine histology analysis, acetone-fixed cryosections were stained with hematoxylin and eosin (H&E). Digital images at the magnification indicated were obtained using an Axiovert 200 fluorescence microscope (Carl Zeiss, Jena, Germany).

Tumor samples for electron microscopy (EM) analysis were cut into small pieces and immediately fixed by immersion in 1.5% (wt/vol) glutaraldehyde in 0.1 M cacodylate buffer with 5% (wt/vol) sucrose for 2 to 4 hours. Specimens were postfixed in cacodylate buffer containing 1% (wt/vol) OsO4 for 1 hour and dehydrated in ascending series of ethanol and propyleneoxide. For contrast enhancement, they were block-stained in uranyl-acetate in 70% ethanol for 4 hours and flat-embedded in Araldite (Serva, Heidelberg, Germany). Using an ultramicrotome (Ultracut R; Leica) semi-thin sections (1 μm) and ultrathin sections (50 nm) were cut. Ultrathin sections were stained with lead citrate, mounted on copper grids, and finally analyzed with a Zeiss EM 10 electron microscope (Carl Zeiss).

Statistical analysis

Statistical analysis was performed using an unpaired Mann-Whitney test using SSPS software (SSPS, Chicago, IL). P values less than .05 were considered statistically significant.

Strong promotion of Th2 development by EpCAM in vitro and in vivo

Adoptive transfer experiments with TCR-transgenic T cells that recognize artificial TAAs from transfected tumor cell lines show that CD4+ T cells frequently provide more efficient protection than CD8+ T cells.20,23,24  We therefore tried to raise Th1 cells against EpCAM, a TAA relevant for tumor immune therapy in humans. We immunized BALB/c mice with CT26-EpCAM cells. Tumor cells were coinjected with CpG-ODN 1668 to obtain optimal in vivo priming for TAA-reactive Th1 cells. The concentration of CpG-ODN 1668 was sufficient to induce Th1 responses against Leishmania major in BALB/c mice27,38  or conventional peptide antigens.39  One week after the last boost, we isolated CD4+ T cells from spleen and LN and stimulated the T cells in vitro again with CpG-ODN 1668, 10 μg/mL 11B11 anti–IL-4 monoclonal antibody (mAb), APCs, and EpCAM protein. Surprisingly, developing CD4+ T cell lines generally resulted in either IL-4 producing Th2 or Th0 cells, with 27% of CD4+ cells producing IL-4 and 44% of CD4+ cells producing IFN-γ. Such Th2 or Th0 cells were unable to protect against intravenously injected CT26-EpCAM tumor cells (Figure 1A). Thus, the human TAA EpCAM did override the most stringent Th1 inducing signals, even the combination of anti–IL-4 mAb and CpG-ODN 1668. This strong Th2 driving force was unexpected and seems to be an unusual feature of this TAA. When T cells were primed under identical conditions with OVA, a protein that normally induces polarized Th2 responses in BALB/c mice,40  we obtained classical Th1 cells that were again approximately 40% IFN-γ+, but less than 5% were positive for IL-4.

Figure 1

Inhibition of tumor growth depends on Th1 lymphocytes. (A) EpCAM-primed Th0/Th2 cells are unable to protect against CT26-EpCAM tumor cells. (B) EpCAM-primed Th1 cells protected against CT26-EpCAM tumor cells. (C) EpCAM-primed Th1 cells protected against CT26-EpCAM tumors and A20-EpCAM tumor cells but not against A20 wild-type lymphoma. T-cell cultures and in vivo experiments were performed as described as in “Methods.”

Figure 1

Inhibition of tumor growth depends on Th1 lymphocytes. (A) EpCAM-primed Th0/Th2 cells are unable to protect against CT26-EpCAM tumor cells. (B) EpCAM-primed Th1 cells protected against CT26-EpCAM tumor cells. (C) EpCAM-primed Th1 cells protected against CT26-EpCAM tumors and A20-EpCAM tumor cells but not against A20 wild-type lymphoma. T-cell cultures and in vivo experiments were performed as described as in “Methods.”

Close modal

Interestingly, when we extended T-cell cultures directed against CT26-EpCAM for another 3 to 4 rounds of stimulation and expansion, cells progressively developed toward a Th1 phenotype with 86% of CD4+ cells producing IFN-γ and 1.5% of CD4+ cells producing IL-4. Simultaneously, these Th1 cells acquired the capacity of protecting against CT26-EpCAM tumors (Figure 1B) or EpCAM-expressing A20 lymphomas (Figure 1C). Yet, they failed to protect against A20 wild-type lymphomas, showing that the Th1 cell response was specific for the tumor antigen (Figure 1C).

Requirement of Th1 cells for the control of EpCAM-expressing carcinomas

To determine whether the IL-4 production was responsible for the lack of protection by short-term cultured T cells or whether it resulted from phenomena such as insufficient selection of T cells expressing TCR with high affinity for EpCAM, we generated Th1 cells from IL-4.ko BALB/c mice using the identical procedure. Priming and expansion of IL-4.ko T cells resulted in both Th1 and Th17 cells, 30% of the cells producing IFN-γ and 15% to 20% producing IL-17 (Figure 2A). In contrast to CD4 T cells from wild-type mice, such short-term cultured EpCAM-reactive Th1/Th17 cells provided solid protection against CT26-EpCAM tumors (Figure 2B), suggesting that IL-4 was responsible for the lack of protection. Therefore, Th1/Th17 cells were further generated from IL-4.ko mice. This also precludes that the tumor established an environment inaccessible to protective immune responses. Moreover, when analyzed for the release of immunosuppressive cytokines, CT26 cells produced no soluble TGF-β, and IL-10 production by CT26-EpCAM cells was lower than IL-10 production by A20-EpCAM lymphomas (Figure S1, available on the Blood website; see the Supplemental Materials link at the top of the online article).

Figure 2

Th1 lymphocytes establish solid tumor immunity. (A) Flow cytometric analysis of generated T cells. Cultured cells were stimulated with PMA/ionomycin, stained with surface anti-CD4, and intracellularly with anti–IFN-γ, anti–IL-4, anti–IL-10, or anti–IL-17, respectively, showing the Th1/Th17 phenotype of EpCAM-primed T cells. Representative dot plots are shown. Numbers indicate the percentage of cells in the particular fluorescence window. (B) Short-term cultured Th1 cells protect against CT26-EpCAM tumors. (C) One hundred days after the original tumor challenge, surviving mice received CT26-EpCAM or A20-EpCAM tumor cells or the parental tumors. All mice were protected against a rechallenge with CT26-EpCAM carcinoma cells, and 70% of the mice were protected against A20-EpCAM lymphoma cells, whereas all mice died with the parental A20 lymphoma cells. Mice challenged with parental CT26 cells were largely protected. Experiments were performed as described in “Methods.”

Figure 2

Th1 lymphocytes establish solid tumor immunity. (A) Flow cytometric analysis of generated T cells. Cultured cells were stimulated with PMA/ionomycin, stained with surface anti-CD4, and intracellularly with anti–IFN-γ, anti–IL-4, anti–IL-10, or anti–IL-17, respectively, showing the Th1/Th17 phenotype of EpCAM-primed T cells. Representative dot plots are shown. Numbers indicate the percentage of cells in the particular fluorescence window. (B) Short-term cultured Th1 cells protect against CT26-EpCAM tumors. (C) One hundred days after the original tumor challenge, surviving mice received CT26-EpCAM or A20-EpCAM tumor cells or the parental tumors. All mice were protected against a rechallenge with CT26-EpCAM carcinoma cells, and 70% of the mice were protected against A20-EpCAM lymphoma cells, whereas all mice died with the parental A20 lymphoma cells. Mice challenged with parental CT26 cells were largely protected. Experiments were performed as described in “Methods.”

Close modal

Th1/Th17 cells establish solid tumor immunity in wild-type mice

Tumor rejection by Th1/Th17 cells may cause epitope spreading and thus newly prime naive T cells against glycoproteins that are expressed by the target tissue. To determine the effects of tumor rejection by EpCAM-reactive Th1/Th17 cells on the establishment of tumor immunity in wild-type mice, mice were rescued from CT26-EpCAM tumors by treatment with EpCAM-reactive Th1/Th17 cells from IL-4.ko mice. Subsequently, mice were challenged with a second tumor on day 100 after original tumor challenge. Surviving mice received one of the following tumors: CT26-EpCAM or A20-EpCAM to analyze reactivity against EpCAM, or parental CT26 tumors or A20 lymphomas to determine specificity. After rejection of the original CT26-EpCAM carcinoma, 100% of mice were protected against CT26-EpCAM carcinoma and 70% against A20-EpCAM lymphoma (Figure 2C). Thus, Th1 cells from IL-4.ko mice established a solid and persistent Th1 memory response against EpCAM-expressing tumors that resisted the Th2-driving properties of the EpCAM protein.

Importantly, the tumor responses were still highly specific, as all mice receiving the parental A20 lymphomas ultimately died (Figure 2C). In sharp contrast, mice challenged with the parental CT26 carcinoma were largely protected. These data suggest that rejection of CT26-EpCAM tumors resulted in functional relevant epitope spreading that may have contributed to the protective immunity against the parental CT26 carcinoma cell line.

Relation of intratumoral cytokine expression and tumor rejection in vivo

To investigate the therapeutic mechanisms underlying the Th1 cell-mediated immunotherapy, we used a subcutaneous CT26-EpCAM tumor model, as this allows direct analysis of both the tumor and the inflammatory environment. We subcutaneously injected 1.5 × 106 CT26-EpCAM cells into the right and left flank of syngeneic BALB/c mice on day 0. On day 4, mice received either 5 × 106 EpCAM-reactive Th1 cells or PBS (control). To determine the impact of these EpCAM-reactive Th1 cells on CT26-EpCAM tumor growth, we measured the tumor size and calculated the tumor growth (Figure 3A). Until day 10, the Th1 cell–treated group and the PBS-treated group showed similar tumor growth dynamics. From day 13 on, the Th1 cell–treated group showed a significant inhibition of tumor growth (Figure 3A).

Figure 3

Determination of the therapeutic effect of adoptively transferred tumor-reactive Th1 cells on tumor growth. (A) Tumors from Th1 cell–treated mice (d10, n = 34; d17, n = 34; d13, n = 24) and PBS-treated mice (d10, n = 22; d17, n = 22; d13, n = 12) were measured with a sliding caliper, and tumor area was calculated as product of perpendicular tumor diameters (*P < .05). Data revealed an inhibitory effect of the adoptive Th1-cell transfer on tumor growth from day 13 on. Error bars denote SEM. (B,C) Adoptive transfer of tumor-reactive Th1 cells leads to inhibition of tumor growth. (B) The difference in tumor weight on day 10 (n = 10) and day 13 (n = 10) after tumor cell injection is shown from 1 experiment of 3. Δ indicates reduction or increase of tumor weight in Th1 cell–treated mice compared with PBS-treated mice. (C) The inhibition of tumor growth is shown by analyzing the tumor weight of Th1 cell–treated and PBS-treated mice on day 11 (n = 22) and day 18 (n = 34) after tumor cell injection (*P < .05). Data are the mean of 2 experiments. (D) OVA-primed Th1 cells were injected in CT26-EpCAM tumor-bearing mice. No difference in tumor weight (n = 12) was observed. Δ indicates increase of tumor weight in the Th1 cell–treated group compared with the PBS-treated group. Experiments were performed as described in “Methods.”

Figure 3

Determination of the therapeutic effect of adoptively transferred tumor-reactive Th1 cells on tumor growth. (A) Tumors from Th1 cell–treated mice (d10, n = 34; d17, n = 34; d13, n = 24) and PBS-treated mice (d10, n = 22; d17, n = 22; d13, n = 12) were measured with a sliding caliper, and tumor area was calculated as product of perpendicular tumor diameters (*P < .05). Data revealed an inhibitory effect of the adoptive Th1-cell transfer on tumor growth from day 13 on. Error bars denote SEM. (B,C) Adoptive transfer of tumor-reactive Th1 cells leads to inhibition of tumor growth. (B) The difference in tumor weight on day 10 (n = 10) and day 13 (n = 10) after tumor cell injection is shown from 1 experiment of 3. Δ indicates reduction or increase of tumor weight in Th1 cell–treated mice compared with PBS-treated mice. (C) The inhibition of tumor growth is shown by analyzing the tumor weight of Th1 cell–treated and PBS-treated mice on day 11 (n = 22) and day 18 (n = 34) after tumor cell injection (*P < .05). Data are the mean of 2 experiments. (D) OVA-primed Th1 cells were injected in CT26-EpCAM tumor-bearing mice. No difference in tumor weight (n = 12) was observed. Δ indicates increase of tumor weight in the Th1 cell–treated group compared with the PBS-treated group. Experiments were performed as described in “Methods.”

Close modal

On day 18 or the indicated time, we determined tumor weight and analyzed in these tumors cytokine expression profiles, growth pattern by histology, and ultrastructure by EM.

Adoptive Th1-cell transfer inhibited tumor growth that was first detectable on day 13 (Figure 3A,B) and reached up to 84% reduction of tumor weight by day 18 (Figure 3C). Importantly, only EpCAM-reactive Th1 cells provided protection, while BALB/c mice receiving 5 × 106 OVA-reactive Th1 cells remained fully susceptible to CT26-EpCAM cells (Figure 3D). As expected from these findings, quantitative mRNA analysis showed no significant increase of FoxP3 mRNA inside Th1 cell–treated tumors (Figure S2).

Quantitative mRNA analysis of tumor samples revealed that the onset of tumor growth inhibition on day 13 coincided with the strongest increase in cytokine expression in tumors of Th1 cell–treated mice as compared with controls (Figure 4A). Th1/Th17 cell–associated genes, namely IL-23/p19, IL-12/p35, IFN-γ, IL-27/Ebi3, IL-6, and ICAM were 3- to 10-fold up-regulated specifically in tumors of Th1 cell–treated mice (Figure 4A), with the most prominent increase for IL-23/p19. Surprisingly, we observed no obvious difference neither for the IFN-γ–induced chemokines, CXCL9 and CXCL10, nor for the IFN-γ–sensitive regulators of cell proliferation and differentiation, c-Myc or cyclin D2. Moreover, angiogenesis-associated genes such as VEGF, placenta growth factor (PIGF), angiopoietin-1 (Ang-1), and Ang-2 were poorly regulated (data not shown). As up-regulation of IL-23 and IL-6 in tumors of Th1 cell–treated mice should provide an environment favorable for the survival and proliferation of IL-17–producing cells,41  we analyzed the expression of IL-17 in these tumors. In agreement with this hypothesis, expression of IL-17A mRNA was highest (> 10-fold) again on day 13, the time when IL-23/p19 and IL-6 peaked (Figure 4B). These data suggest that infiltrating Th1 and Th17 cells might act in concert with cytokines of the IL-12 family to exert relevant tumor control, especially as IFN-γ and IL-12 are both well known to exert strong inhibition of tumor growth, in vitro and in vivo.28-30,35 

Figure 4

Gene expression analysis of tumors by PCR at different points in time. (A,B) In real-time PCR analysis, multiplex reactions were run in triplicates, and samples were normalized to the housekeeping gene HPRT. (A) Relative expression of genes of interest is represented as fold differences of the mean of Th1 cell–treated mice (d10, n = 10; d11, n = 10; d13, n = 10; d18, n = 9) over PBS controls (d10, n = 10; d11, n = 10; d13, n = 8; d18, n = 9), with highest levels on day 13 (*P < .05). (B) IL-17 expression in tumors of Th1 cell–treated mice (d10, n = 10; d13, n = 8; d18, n = 9) and control mice (d10, n = 10; d13, n = 7; d18, n = 12) on days 10, 13, and 18, showing an increase of IL-17 in tumors of Th1 cell–treated mice from day 13 on (*P < .05).

Figure 4

Gene expression analysis of tumors by PCR at different points in time. (A,B) In real-time PCR analysis, multiplex reactions were run in triplicates, and samples were normalized to the housekeeping gene HPRT. (A) Relative expression of genes of interest is represented as fold differences of the mean of Th1 cell–treated mice (d10, n = 10; d11, n = 10; d13, n = 10; d18, n = 9) over PBS controls (d10, n = 10; d11, n = 10; d13, n = 8; d18, n = 9), with highest levels on day 13 (*P < .05). (B) IL-17 expression in tumors of Th1 cell–treated mice (d10, n = 10; d13, n = 8; d18, n = 9) and control mice (d10, n = 10; d13, n = 7; d18, n = 12) on days 10, 13, and 18, showing an increase of IL-17 in tumors of Th1 cell–treated mice from day 13 on (*P < .05).

Close modal

Enhanced tumor rejection by prevention of activation-associated Th-1 cell death

To prevent activation-associated death of the transferred Th1 cells from IL-4.ko mice, we treated the tumor-bearing mice with 500 μg anti–IFN-γ mAb, 1 day before the adoptive transfer of EpCAM-reactive Th1 cells.42  If the immune-mediated tumor rejection relies on the recipient's T cells, such a treatment should promote Th2 development and abrogate tumor protection by the Th1 cells. In contrast, if the therapy primarily relies on the transferred Th1 cells from IL-4.ko mice, the anti–IFN-γ mAb should protect those transferred Th1 cells from suicide and thus strengthen the protective effect against the tumor.42  Indeed, on day 17, inhibition of tumor growth was even more efficient in mice where activation-associated cell death of Th1 cells was prevented by anti–IFN-γ mAb (Figure 5A) than in mice treated with Th1 cells alone. Reduced tumor growth in anti–IFN-γ–treated mice was associated with a 3-fold stronger up-regulation of intratumoral IFN-γ mRNA as compared with mice receiving Th1 cells only (Figure 5B). Most other cytokines remained comparable between the group receiving both, Th1 cells and anti–IFN-γ mAb, and the mice treated with Th1 cells only (Figure 5B).

Figure 5

Combined Th1-cell and anti–IFN-γ treatment leads to further reduction of tumor growth. (A) On day 3 after injection of CT26-EpCAM tumor cells, mice were irradiated, treated with 500 μg anti–IFN-γ antibody, and on day 4 EpCAM-reactive Th1 cells were injected. Tumor weight of PBS-treated (n = 18), Th1 cell–treated (n = 10), and combined-treated (n = 8) mice was analyzed on day 17. Δ, reduction of tumor weight of Th1 cell–treated mice compared with PBS controls (*P < .05). (B) Real-time PCR data of tumors. Multiplex reactions were run in triplicates, and samples were normalized to the housekeeping gene HPRT. Relative expression of genes of interest is represented as fold differences of the mean of Th1 cell-treated mice (n = 7) or combined-treated mice (Th1 cells and anti–IFN-γ, n = 6) over PBS controls (n = 14) on day 17 after tumor cell injection. A strong up-regulation of IFN-γ was found in tumors treated with both Th1 cells and anti–IFN-γ mAb (*P < .05). Experiments were performed as described in “Methods.”

Figure 5

Combined Th1-cell and anti–IFN-γ treatment leads to further reduction of tumor growth. (A) On day 3 after injection of CT26-EpCAM tumor cells, mice were irradiated, treated with 500 μg anti–IFN-γ antibody, and on day 4 EpCAM-reactive Th1 cells were injected. Tumor weight of PBS-treated (n = 18), Th1 cell–treated (n = 10), and combined-treated (n = 8) mice was analyzed on day 17. Δ, reduction of tumor weight of Th1 cell–treated mice compared with PBS controls (*P < .05). (B) Real-time PCR data of tumors. Multiplex reactions were run in triplicates, and samples were normalized to the housekeeping gene HPRT. Relative expression of genes of interest is represented as fold differences of the mean of Th1 cell-treated mice (n = 7) or combined-treated mice (Th1 cells and anti–IFN-γ, n = 6) over PBS controls (n = 14) on day 17 after tumor cell injection. A strong up-regulation of IFN-γ was found in tumors treated with both Th1 cells and anti–IFN-γ mAb (*P < .05). Experiments were performed as described in “Methods.”

Close modal

Selective tumor infiltration by CD4+ T cells

As inflammatory cytokines were up-regulated in tumors of mice treated with Th1 cells, we investigated the potential source of these cytokines by characterizing the cellular infiltrate using FACS analysis and immunohistochemistry. Analyzing tumors of Th1 cell–treated mice showed a strong increase in homogeneously distributed CD4+ T cells (Figure 6Aii), peaking on day 18 with a 16-fold increase (Table 1). In contrast, tumors of the control group had only few CD4+ T cells (Figure 6Ai and Table 1). Besides CD4+ T cells, FACS analysis showed a relative 3-fold increase of CD11b+ cells in tumors of Th1 cell–treated mice at day 18. F4/80+ macrophages or CD11c+ infiltrating DCs were not detectable in any of the analyzed tumors (Table 1). These data suggest that infiltrating CD4+ T cells and cells of the monocyte/macrophage lineage were involved in arresting tumor growth. We found no evidence for infiltrating CD8+ T cells in the Th1 cell–treated mice. On day 18, we even detected 5 times more CD8+ T cells in tumors of untreated mice than in tumors of Th1 cell–treated mice (Table 1). Despite the significant difference in tumor size, we found only minor differences in tumor morphology (Figure 6Aiii,iv) or proliferation markers such as the proliferation-associated nuclear antigen Ki-67 (Figure 6Av,vi,B).

Figure 6

Immunohistochemical analysis and EM of tumors. Serial sections of representative tumors (n = 8) of Th1 cell–treated and control mice from day 18. (A) Tumors were stained with monoclonal anti-CD4 antibody (i,ii) showing a strong increase in CD4+ cells in tumors of Th1 cell–treated mice. indicates CD4+ cells. Staining with H&E (iii,iv) showed no major differences in the morphology of the tumors. Necrosis (N) was detected in tumors of Th1 cell–treated and PBS-treated mice. Ki-67 staining (v,vi) showed no obvious differences in the proliferation rate of the tumors. indicates Ki-67–positive cells. Pictures, ×100 magnification. Inlays, ×200 original magnification. (B) Quantitative analysis of (Av,vi). Ki-67+ cells of 500 total cells in tumor sections (n = 60) of Th1 cell–treated and PBS-treated mice were counted. Error bars denote SEM. (C) Tumors were stained with monoclonal anti–PECAM-1 antibody (i-iv) showing homogenous vascularization in both groups. However, minor changes in vessel morphology (white arrows) and a minor decrease of tumor vessel density were detectable in tumors of Th1 cell–treated mice (ii,iv) compared with control mice (i,iii). Staining of tumors with monoclonal anti–VEGFR-2 antibody (v,vi) revealed no obvious change in the receptor expression pattern between both groups. Pictures, ×200 original magnification. (D) EM of blood vessels. (i) Blood vessels with a bulged EC body (E), tight junctions (), and the basal lamina (labeled with *). (ii) Blood lacuna without limiting ECs, but with extracellular matrix–like collagen fibrils (*) that are located between tumor cells and blood cells. (iii) Blood vessel with leukocyte transendothelial migration. (iv) Higher magnification of panel iii, showing a transcellularly transmigrated leukocyte (L) still covered by a basal lamina (). (v) Obliterated vessel. The left arrow labels a tight junction; the right arrow, the rest of the lumen. (vi) Vessels in adjacent muscle of Th1 cell–treated tumors appear normal. In summary, blood vessels of PBS-treated (i,ii) tumors were seen as normal tumor vessels. In contrast, degenerated ECs were observed more frequently in tumors of Th1 cell–treated mice (iii-v). In addition, strong inflammatory processes (iii,iv) and more obliterated vessels (v) were seen in tumors of Th1 cell–treated mice. Experiments were performed as described in “Methods.”

Figure 6

Immunohistochemical analysis and EM of tumors. Serial sections of representative tumors (n = 8) of Th1 cell–treated and control mice from day 18. (A) Tumors were stained with monoclonal anti-CD4 antibody (i,ii) showing a strong increase in CD4+ cells in tumors of Th1 cell–treated mice. indicates CD4+ cells. Staining with H&E (iii,iv) showed no major differences in the morphology of the tumors. Necrosis (N) was detected in tumors of Th1 cell–treated and PBS-treated mice. Ki-67 staining (v,vi) showed no obvious differences in the proliferation rate of the tumors. indicates Ki-67–positive cells. Pictures, ×100 magnification. Inlays, ×200 original magnification. (B) Quantitative analysis of (Av,vi). Ki-67+ cells of 500 total cells in tumor sections (n = 60) of Th1 cell–treated and PBS-treated mice were counted. Error bars denote SEM. (C) Tumors were stained with monoclonal anti–PECAM-1 antibody (i-iv) showing homogenous vascularization in both groups. However, minor changes in vessel morphology (white arrows) and a minor decrease of tumor vessel density were detectable in tumors of Th1 cell–treated mice (ii,iv) compared with control mice (i,iii). Staining of tumors with monoclonal anti–VEGFR-2 antibody (v,vi) revealed no obvious change in the receptor expression pattern between both groups. Pictures, ×200 original magnification. (D) EM of blood vessels. (i) Blood vessels with a bulged EC body (E), tight junctions (), and the basal lamina (labeled with *). (ii) Blood lacuna without limiting ECs, but with extracellular matrix–like collagen fibrils (*) that are located between tumor cells and blood cells. (iii) Blood vessel with leukocyte transendothelial migration. (iv) Higher magnification of panel iii, showing a transcellularly transmigrated leukocyte (L) still covered by a basal lamina (). (v) Obliterated vessel. The left arrow labels a tight junction; the right arrow, the rest of the lumen. (vi) Vessels in adjacent muscle of Th1 cell–treated tumors appear normal. In summary, blood vessels of PBS-treated (i,ii) tumors were seen as normal tumor vessels. In contrast, degenerated ECs were observed more frequently in tumors of Th1 cell–treated mice (iii-v). In addition, strong inflammatory processes (iii,iv) and more obliterated vessels (v) were seen in tumors of Th1 cell–treated mice. Experiments were performed as described in “Methods.”

Close modal

Enhanced EC death after Th1-cell therapy

As the antiangiogenic cytokines IL-12, IFN-γ, and IL-27 were up-regulated in tumors of Th1 cell-treated mice and as tumor growth depends on neovessel formation, we investigated the effects of Th1 cell therapy on tumor angiogenesis. Immunohistologic staining with a mAb specific for the endothelial antigen PECAM-1 showed no gross differences between the 2 groups (Figure 6Ci-iv). However, changes in vessel morphology, such as clumpy and curved structures, appeared in tumors of Th1 cell–treated mice (Figure 6Cii,iv arrows). The expression of the most important receptor of angiogenesis, VEGFR-2, was not affected by the Th1-cell therapy (Figure 6Cv,vi).

We therefore performed EM to study ultrastructural changes. Tumors of Th1 cell–treated mice revealed major EC damage such as cell death, cytoplasma-enriched endothelia, and vessel obliteration (Figure 6Diii,v), demonstrating severe impairment of tumor vessels in Th1 cell–treated mice. This impaired vessel structure was associated with enhanced diapedesis as sign of inflammation (Figure 6Diii,iv). Tumors of PBS-treated mice showed a vascular bed typical for normal tumor tissue (Figure 6Di,ii).43  Vessels of adjacent muscle tissue remained unchanged in Th1 cell–treated mice (Figure 6Dvi), showing that the Th1 cell–mediated effects on the vasculature were restricted to the tumor tissue.

Currently, various immune therapies are developed against solid human cancer. While antibody-based immune therapies show first success in humans, the therapeutic success of T cell–based clinical trials, such as active immunization or adoptive T-cell transfer, was only successful in single patients.22  In some trials, tumor immune therapy did not only fail to establish protection but even promoted tumor development.17,44  The mechanisms underlying these failures remain enigmatic. Indirect data from humans suggest that protective immunity was associated with the development of a Th1 immunity, while tumor promotion occurred in patients that developed a Th2 immunity.17,44  These findings are in agreement with various experimental data from mice showing that only Th1 cells are capable of providing protective tumor immunity, while Th2 responses fail to protect.20,24,29 

Using a clinically relevant model antigen, EpCAM, here we were unable to establish protective tumor immunity in normal BALB/c mice, even in the presence of the TLR9-ligand CpG-ODN and anti–IL-4 mAb.45  Characterizing these inefficient immune responses, we found that EpCAM-reactive Th cells inevitably developed an IL-4–producing Th2 or Th0 phenotype, even under most stringent Th1 cell-inducing conditions. As IL-4 is required for the differentiation of naive Th cells toward a Th2 phenotype during the period of T-cell priming, the combination of anti–IL-4 mAb and CpG-ODN is most efficient in preventing Th-2 cell differentiation under most conditions, even in vivo.38,45  Besides IL-4, specific sets of accessory molecules such as Jagged, certain C-type lectin receptors, parasite-derived proteoglycans, or eosinophil-derived neurotoxin46  can promote Th2 differentiation. Such stimuli may either activate GATA3 through alternative activation of STAT6 or other signaling pathways that involve modulation of the TCR-signaling pathway.47  The mechanisms underlying the potent Th2-inducing properties of EpCAM remain enigmatic.

To determine whether the failure of raising protective immune responses against CT26-EpCAM was secondary to the Th2/Th0 phenotype of the transferred T cells, we established EpCAM-reactive Th1 cells from syngeneic IL-4.ko BALB/c mice. Using the same protocol of in vivo priming followed by one round of in vitro stimulation, we generated EpCAM-reactive, IL-4–deficient Th1-cell lines. Surprisingly, these Th1 cells did not only specifically protect naive BALB/c mice against EpCAM-expressing tumors, but even established protective immunity against subsequent tumor challenges in normal BALB/c mice. This proves that EpCAM-induced IL-4 directly abrogated the capacity of CD4+ T cells to establish protective tumor immunity. Moreover, the data underlined the high therapeutic efficacy of adoptively transferred Th1 cells, even against tumors that normally induce tumor immune evasion through the induction of Th2 responses.

EpCAM-reactive Th1 cells protected against both intravenously and subcutaneously applied carcinomas. To analyze the mechanisms underlying this Th1 cell–mediated control of EpCAM-expressing, MHC class II–negative carcinoma cells, we focused on subcutaneously injected tumors. This model provides direct access to the tumor and thus allows to investigate the tumor-host interactions over time. In agreement with previous reports,24,29  we found that the Th1-cell therapy induced the expression of antiangiogenic factors, while we found no evidence for a significant suppression of VEGF. The Th1-cell therapy strongly induced the expression of proinflammatory and antiangiogenic cytokines, namely IL-12, IL-23, IL-27, and IFN-γ. Expression of all 4 cytokines peaked exactly at the initial inhibition of tumor growth. Consistent with these findings, we found a 3-fold increase of CD11b+ monocytes/macrophages in tumors of Th1 cell–treated mice. Tumor-infiltrating macrophages that are activated by IFN-γ–producing Th1 cells (M1 phenotype) possess strong cytotoxic properties against tumor cells and produce proinflammatory cytokines such as IL-23 and IL-12.35  While the antiangiogenic role of IFN-γ and IL-12 is well established,29,30,32,33  the role of IL-23 in either promoting or inhibiting tumor development is ambiguous. While transfection of tumor cells with IL-23 or systemic application of IL-23 promote tumor rejection and establish protective immunity,36,37  IL-23.ko mice are resistant to chemically induced tumor induction.48  This role of IL-23 in antitumor immunity is reminiscent of the double-edged role of tumor necrosis factor (TNF).24 

Analyzing the tumor microenvironment at various times of tumor growth inhibition, we only found infiltrating CD4+ T cells but no evidence for CTL-mediated tumor cell killing or induction of regulatory T cells. In 2 additional model tumors, transplanted A20 lymphomas and endogenously growing islet carcinomas, we extensively investigated the role of CD8+ T cells and CTL. Both experiments showed, that the therapeutic outcome of the adoptive Th1-cell transfer was not attenuated by the depletion of CD8+ T cells.20,24  Th1-cell transfer differs from conditions of active immunization.15 

As tumors of either untreated or treated mice showed equal numbers of Ki-67–positive cells, it is unlikely that the Th1 cells directly inhibited the proliferation of tumor cells.24  The intra-tumoral cytokine expression at the time of growth arrest together with the EC damage uncovered by EM strongly suggest that the antiangiogenic properties of Th1 cells significantly contributed to the T cell–mediated inhibition of tumor growth. EM revealed EC death, and cytoplasma-enriched ECs were frequent in tumors of Th1 cell–treated mice. Importantly, ECs in adjacent muscle tissue were not affected, showing that the antiangiogenic properties of the Th1 cell therapy were restricted to the tumor tissue. Together, the data suggest that the combined action of IFN-γ, IL-12, and IL-27 inhibited tumor growth by severe EC damage. Th1 cells can impair tumor angiogenesis either directly by inhibiting EC proliferation through IFN-γ or indirectly through induction of antiangiogenic chemokines.24  Similar to the model described by Qin and Blankenstein,29  IFN-γ seems to directly inhibit tumor angiogenesis in this model. Importantly, this may also be relevant for the human situation, where adoptive transfer of a Th1-cell clone resulted in the rapid involution of metastases without causing a cytokine release syndrome.49 

As prevention of activation-associated Th1-cell death further enhanced the efficiency of adoptive T-cell therapy with Th1 cells, the findings reported here strongly support the increasing importance of TAA-reactive CD4+ T cells in tumor therapy. The different tumor models using either injected tumor cell lines or endogenously growing tumors suggest that Th1 cells may inhibit tumor development and seeding of metastases through different modes of action. Yet, all studies unanimously show that proper Th1-cell differentiation, ie, a strong IFN-γ production and the absence of IL-4, is crucial for the establishment of protective tumor immune responses. Thus, human tumor vaccine approaches should focus on the use of TAA that allow appropriate induction of tumor-reactive Th1 responses.

The online version of this article contains a data supplement.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

The skillful help Gabi Frommer-Kästle with electron microscopy is gratefully acknowledged. We thank G. Riethmüller, P. Kufer, and E. Kopp for helpful discussions and providing EpCAM.

This work was supported by the Deutsche Forschungsgemeinschaft (SFB 685), the Wilhelm Sander-Stiftung (2005.043), the Deutsche Krebshilfe (10 7128), and the Comprehensive Cancer Center Tübingen.

Contribution: A.Z., R.H., and H.B. performed and analyzed the experiments and prepared the manuscript; S.W. technically supported the performance of the experiments; H.W. performed and analyzed the electron microscopy; R.M. and M.R. designed the experiments, interpreted the data, and prepared the manuscript; and all authors checked the final version of the manuscript.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Martin Röcken, Department of Dermatology, University Medical Center, Eberhard Karls University Tübingen, Liebermeisterstr 25, 72076 Tübingen, Germany; e-mail: mrocken@med.uni-tuebingen.de; or Ralph Mocikat, Institut für Molekulare Immunologie, Helmholtz-Zentrum München, Marcchioninistr 25, 81377 München, Germany; e-mail: mocikat@helmholtz-muenchen.de.

1
Chaudry
 
MA
Sales
 
K
Ruf
 
P
Lindhofer
 
H
Winslet
 
MC
EpCAM an immunotherapeutic target for gastrointestinal malignancy: current experience and future challenges.
Br J Cancer
2007
, vol. 
96
 (pg. 
1013
-
1019
)
2
Riethmuller
 
G
Schneider-Gadicke
 
E
Schlimok
 
G
, et al. 
Randomised trial of monoclonal antibody for adjuvant therapy of resected Dukes' C colorectal carcinoma. German Cancer Aid 17-1A Study Group.
Lancet
1994
, vol. 
343
 (pg. 
1177
-
1183
)
3
Daly
 
T
Royal
 
RE
Kershaw
 
MH
, et al. 
Recognition of human colon cancer by T cells transduced with a chimeric receptor gene.
Cancer Gene Ther
2000
, vol. 
7
 (pg. 
284
-
291
)
4
Trojan
 
A
Witzens
 
M
Schultze
 
JL
, et al. 
Generation of cytotoxic T lymphocytes against native and altered peptides of human leukocyte antigen-A*0201 restricted epitopes from the human epithelial cell adhesion molecule.
Cancer Res
2001
, vol. 
61
 (pg. 
4761
-
4765
)
5
Pardoll
 
DM
Cancer vaccines.
Nat Med
1998
, vol. 
4
 (pg. 
525
-
531
)
6
Lollini
 
P-L
Cavallo
 
F
Nanni
 
P
Forni
 
G
Vaccines for tumour prevention.
Nat Rev Cancer
2006
, vol. 
6
 (pg. 
204
-
216
)
7
Hanson
 
HL
Donermeyer
 
DL
Ikeda
 
H
, et al. 
Eradication of established tumors by CD8+ T cell adoptive immunotherapy.
Immunity
2000
, vol. 
13
 (pg. 
265
-
276
)
8
June
 
CH
Principles of adoptive T cell cancer therapy.
J Clin Invest
2007
, vol. 
117
 (pg. 
1204
-
1212
)
9
Schuler
 
G
Steinman
 
RM
Dendritic cells as adjuvants for immune-mediated resistance to tumors.
J Exp Med
1997
, vol. 
186
 (pg. 
1183
-
1187
)
10
Rosenberg
 
SA
Yang
 
JC
Restifo
 
NP
Cancer immunotherapy: moving beyond current vaccines.
Nat Med
2004
, vol. 
10
 (pg. 
909
-
915
)
11
Powell
 
DJ
Dudley
 
ME
Hogan
 
KA
Wunderlich
 
JR
Rosenberg
 
SA
Adoptive transfer of vaccine-induced peripheral blood mononuclear cells to patients with metastatic melanoma following lymphodepletion.
J Immunol
2006
, vol. 
177
 (pg. 
6527
-
6539
)
12
Willimsky
 
G
Blankenstein
 
T
Sporadic immunogenic tumours avoid destruction by inducing T-cell tolerance.
Nature
2005
, vol. 
437
 (pg. 
141
-
146
)
13
Adam
 
C
King
 
S
Allgeier
 
T
, et al. 
DC-NK cell cross talk as a novel CD4+ T-cell–independent pathway for antitumor CTL induction.
Blood
2005
, vol. 
106
 (pg. 
338
-
344
)
14
Forster
 
I
Hirose
 
R
Arbeit
 
JM
Clausen
 
BE
Hanahan
 
D
Limited capacity for tolerization of CD4+ T cells specific for a pancreatic β cell neo-antigen.
Immunity
1995
, vol. 
2
 (pg. 
573
-
585
)
15
Mocikat
 
R
Braumuller
 
H
Gumy
 
A
, et al. 
Natural killer cells activated by MHC class I(low) targets prime dendritic cells to induce protective CD8 T cell responses.
Immunity
2003
, vol. 
19
 (pg. 
561
-
569
)
16
Wang
 
HY
Wang
 
RF
Regulatory T cells and cancer.
Curr Opin Immunol
2007
, vol. 
19
 (pg. 
217
-
223
)
17
Schadendorf
 
D
Ugurel
 
S
Schuler-Thurner
 
B
, et al. 
Dacarbazine (DTIC) versus vaccination with autologous peptide-pulsed dendritic cells (DC) in first-line treatment of patients with metastatic melanoma: a randomized phase III trial of the DC study group of the DeCOG.
Ann Oncol
2006
, vol. 
17
 (pg. 
563
-
570
)
18
Daniel
 
D
Chiu
 
C
Giraudo
 
E
, et al. 
CD4+ T cell-mediated antigen-specific immunotherapy in a mouse model of cervical cancer.
Cancer Res
2005
, vol. 
65
 (pg. 
2018
-
2025
)
19
Gattinoni
 
L
Powell
 
DJ
Rosenberg
 
SA
Restifo
 
NP
Adoptive immunotherapy for cancer: building on success.
Nat Rev Immunol
2006
, vol. 
6
 (pg. 
383
-
393
)
20
Egeter
 
O
Mocikat
 
R
Ghoreschi
 
K
Dieckmann
 
A
Rocken
 
M
Eradication of disseminated lymphomas with CpG-DNA activated T helper type 1 cells from nontransgenic mice.
Cancer Res
2000
, vol. 
60
 (pg. 
1515
-
1520
)
21
Mackinnon
 
S
Thomson
 
K
Verfuerth
 
S
Peggs
 
K
Lowdell
 
M
Adoptive cellular therapy for cytomegalovirus infection following allogeneic stem cell transplantation using virus-specific T cells.
Blood Cells Mol Dis
2008
, vol. 
40
 (pg. 
63
-
67
)
22
Rosenberg
 
SA
Restifo
 
NP
Yang
 
JC
Morgan
 
RA
Dudley
 
ME
Adoptive cell transfer: a clinical path to effective cancer immunotherapy.
Nat Rev Cancer
2008
, vol. 
8
 (pg. 
299
-
308
)
23
Perez-Diez
 
A
Joncker
 
NT
Choi
 
K
, et al. 
CD4 cells can be more efficient at tumor rejection than CD8 cells.
Blood
2007
, vol. 
109
 (pg. 
5346
-
5354
)
24
Muller-Hermelink
 
N
Braumuller
 
H
Pichler
 
B
, et al. 
TNFR1 signaling and IFN-γ signaling determine whether T cells induce tumor dormancy or promote multistage carcinogenesis.
Cancer Cell
2008
, vol. 
13
 (pg. 
507
-
518
)
25
Nishimura
 
T
Iwakabe
 
K
Sekimoto
 
M
, et al. 
Distinct role of antigen-specific T helper type 1 (Th1) and Th2 cells in tumor eradication in vivo.
J Exp Med
1999
, vol. 
190
 (pg. 
617
-
628
)
26
Knutson
 
KL
Disis
 
ML
Tumor antigen-specific T helper cells in cancer immunity and immunotherapy.
Cancer Immunol Immunother
2005
, vol. 
54
 (pg. 
721
-
728
)
27
Biedermann
 
T
Zimmermann
 
S
Himmelrich
 
H
, et al. 
IL-4 instructs TH1 responses and resistance to Leishmania major in susceptible BALB/c mice.
Nat Immunol
2001
, vol. 
2
 (pg. 
1054
-
1060
)
28
Li
 
Z
Pradera
 
F
Kammertoens
 
T
Li
 
B
Liu
 
S
Qin
 
Z
Cross-talk between T cells and innate immune cells is crucial for IFN-γ-dependent tumor rejection.
J Immunol
2007
, vol. 
179
 (pg. 
1568
-
1576
)
29
Qin
 
Z
Blankenstein
 
T
CD4+ T cell-mediated tumor rejection involves inhibition of angiogenesis that is dependent on IFN γ receptor expression by nonhematopoietic cells.
Immunity
2000
, vol. 
12
 (pg. 
677
-
686
)
30
Beatty
 
GL
Paterson
 
Y
IFN-γ-dependent inhibition of tumor angiogenesis by tumor-infiltrating CD4+ T cells requires tumor responsiveness to IFN-γ.
J Immunol
2001
, vol. 
166
 (pg. 
2276
-
2282
)
31
Aggarwal
 
S
Gurney
 
AL
IL-17: prototype member of an emerging cytokine family.
J Leukoc Biol
2002
, vol. 
71
 (pg. 
1
-
8
)
32
Sgadari
 
C
Angiolillo
 
AL
Tosato
 
G
Inhibition of angiogenesis by interleukin-12 is mediated by the interferon-inducible protein 10.
Blood
1996
, vol. 
87
 (pg. 
3877
-
3882
)
33
Strasly
 
M
Cavallo
 
F
Geuna
 
M
, et al. 
IL-12 Inhibition of endothelial cell functions and angiogenesis depends on lymphocyte-endothelial cell cross-talk.
J Immunol
2001
, vol. 
166
 (pg. 
3890
-
3899
)
34
Shimizu
 
M
Shimamura
 
M
Owaki
 
T
, et al. 
Antiangiogenic and antitumor activities of IL-27.
J Immunol
2006
, vol. 
176
 (pg. 
7317
-
7324
)
35
Trinchieri
 
G
Pflanz
 
S
Kastelein
 
RA
The IL-12 family of heterodimeric cytokines: new players in the regulation of T cell responses.
Immunity
2003
, vol. 
19
 (pg. 
641
-
644
)
36
Kaiga
 
T
Sato
 
M
Kaneda
 
H
Iwakura
 
Y
Takayama
 
T
Tahara
 
H
Systemic administration of IL-23 induces potent antitumor immunity primarily mediated through Th1-type response in association with the endogenously expressed IL-12.
J Immunol
2007
, vol. 
178
 (pg. 
7571
-
7580
)
37
Lo
 
C-H
Lee
 
S-C
Wu
 
P-Y
, et al. 
Antitumor and antimetastatic activity of IL-23.
J Immunol
2003
, vol. 
171
 (pg. 
600
-
607
)
38
Zimmermann
 
S
Egeter
 
O
Hausmann
 
S
, et al. 
CpG oligodeoxynucleotides trigger protective and curative Th1 responses in lethal murine leishmaniasis.
J Immunol
1998
, vol. 
160
 (pg. 
3627
-
3630
)
39
Vabulas
 
RM
Pircher
 
H
Lipford
 
GB
Hacker
 
H
Wagner
 
H
CpG-DNA activates in vivo T cell epitope presenting dendritic cells to trigger protective antiviral cytotoxic T cell responses.
J Immunol
2000
, vol. 
164
 (pg. 
2372
-
2378
)
40
Schipf
 
A
Heilmann
 
A
Boue
 
L
Mossmann
 
H
Brocker
 
T
Rocken
 
M
Th2 cells shape the differentiation of developing T cell responses during interactions with dendritic cells in vivo.
Eur J Immunol
2003
, vol. 
33
 (pg. 
1697
-
1706
)
41
Veldhoen
 
M
Hocking
 
RJ
Atkins
 
CJ
Locksley
 
RM
Stockinger
 
B
TGFβ in the context of an inflammatory cytokine milieu supports de novo differentiation of IL-17–producing T cells.
Immunity
2006
, vol. 
24
 (pg. 
179
-
189
)
42
Berner
 
V
Liu
 
H
Zhou
 
Q
, et al. 
IFN-γ mediates CD4+ T-cell loss and impairs secondary antitumor responses after successful initial immunotherapy.
Nat Med
2007
, vol. 
13
 (pg. 
354
-
360
)
43
Carmeliet
 
P
Jain
 
RK
Angiogenesis in cancer and other diseases.
Nature
2000
, vol. 
407
 (pg. 
249
-
257
)
44
Schultz
 
ES
Schuler-Thurner
 
B
Stroobant
 
V
, et al. 
Functional analysis of tumor-specific Th cell responses detected in melanoma patients after dendritic cell-based immunotherapy.
J Immunol
2004
, vol. 
172
 (pg. 
1304
-
1310
)
45
Himmelrich
 
H
Launois
 
P
Maillard
 
I
, et al. 
In BALB/c mice, IL-4 production during the initial phase of infection with Leishmania major is necessary and sufficient to instruct Th2 cell development resulting in progressive disease.
J Immunol
2000
, vol. 
164
 (pg. 
4819
-
4825
)
46
MacDonald
 
AS
Maizels
 
RM
Alarming dendritic cells for Th2 induction.
J Exp Med
2008
, vol. 
205
 (pg. 
13
-
17
)
47
Ouyang
 
W
Lohning
 
M
Gao
 
Z
, et al. 
Stat6-independent GATA-3 autoactivation directs IL-4-independent Th2 development and commitment.
Immunity
2000
, vol. 
12
 (pg. 
27
-
37
)
48
Langowski
 
JL
Zhang
 
X
Wu
 
L
, et al. 
IL-23 promotes tumour incidence and growth.
Nature
2006
, vol. 
442
 (pg. 
461
-
465
)
49
Hunder
 
NN
Wallen
 
H
Cao
 
J
, et al. 
Treatment of metastatic melanoma with autologous CD4+ T cells against NY-ESO-1.
N Engl J Med
2008
, vol. 
358
 (pg. 
2698
-
2703
)

Author notes

*A.Z., R.H., and H.B. contributed equally to this work.

Supplemental data

Sign in via your Institution