Immune-cell–based approaches using cytotoxic and dendritic cells are under constant scrutiny to design novel therapies for the treatment of tumors. These strategies are hampered by the lack of efficient and economical large-scale production methods for effector cells. Here we describe the propagation of large amounts of a unique population of CD4+ cytotoxic T cells, which we termed tumor killer T cells (TKTC), because of their potent and broad antitumor cell activity. With this cultivation strategy, TKTCs from peripheral blood mononuclear cells are generated within a short period of time using a pulse with a stimulating cell line followed by continuous growth in serum-free medium supplemented with a mixture of interleukin-2 and cyclosporin A. Expression and functional profiling did not allow a classification of TKTCs to any thus far defined subtype of T cells. Cytotoxic assays showed that TKTCs kill a panel of tumor targets of diverse tissue origin while leaving normal cells unaffected. Blocking experiments revealed that TKTC killing was, to a significant extent, mediated by tumor necrosis factor-related apoptosis-inducing ligand and was independent of MHC restriction. These results suggest that TKTCs have a high potential as a novel tool in the adoptive immunotherapy of cancer. (Mol Cancer Res 2009;7(3):339–53)

One of the most promising anticancer therapies is to use the patient's own immune system to eradicate tumor cells. Based on natural killer (NK) cells (1, 2), dendritic cells (3), hematopoietic stem cells (4), T cells (5) with focus on CTLs (6, 7), as well as genetically engineered T cells (8), multiple therapeutic concepts were developed for adoptive immunotherapy of a large variety of cancers like breast (9), prostate (10), and ovarian (11) cancers, as well as against melanoma (12), childhood malignancies (13), medullary thyroid carcinoma (14), or renal cell cancer (15).

The idea of adoptive immunotherapy goes back to results in the early 1980s showing that exposure of peripheral blood mononuclear cells (PBMC) to high concentrations of interleukin (IL)-2 led to the generation of lymphokine activated killer cells, which consisted mainly of NK cells and showed significant lysis of tumor cells in vitro sparing normal cells. In humans, however, lymphokine activated killer cells in combination with high-dose IL-2 gave response rates of only up to 20% (16). Further development of adoptive immunotherapy led to the use of tumor-infiltrating lymphocytes (TIL) with very promising results in mouse models (17). Since then, a huge effort has been undertaken to establish adoptive immunotherapy using TILs in humans. TIL cultures were expanded from tumors of different epithelial origin but rarely showed specific antitumor activity. On the contrary, melanoma TIL cultures could be generated more reliably, and patients who had received TILs expanded from subcutaneous tumor deposits showed response rates of up to 49% (18). Nowadays, patients with metastatic melanoma can be treated quite effectively by adoptive cell therapy using TILs, which mediate cancer regression in ∼50% of patients. Furthermore, this approach has been extended by genetic engineering of human lymphocytes to yield even higher response rates (6). However, the attempt to prolong the in vivo survival of transferred TILs by insertion of the IL-2 gene could not improve in vivo persistence or clinical effectiveness, in contrast to results from in vitro experiments (19). Nevertheless, the expansion of TILs from tumor material is labor- and time-intensive and an additional strain on the patient because tumor tissue has to be excised in a biopsy (20).

Antigen-independent activation of PBMCs derived from cancer patients [e.g., by using CD3 monoclonal antibodies (mAb; refs. 6, 21)] generated activated T cells, which exerted a certain antitumor effect when reinfused into the patient, whereby the clinical outcome was mostly poor. Furthermore, a limiting step in the cultivation of all these effector cells and, in particular, cells of T-cell origin is the demand of high amounts of IL-2 and specific culturing skills.

Among the CTLs, most are of the CD8+ phenotype that recognize antigen presented on MHC class I; however, ∼10% of human CTLs are CD4+ and recognize antigen presented via MHC class II (19, 22). Additionally, some CD4+ CTLs restricted to MHC class I have been reported (23-25). It is important to note that CD4+ CTLs are not, as previously postulated, an in vitro artifact (26) produced by prolonged activation under proliferative in vitro conditions, but increased in viral infection (27), autoimmune disorders (28, 29), or cancer (25). CD4+ CTLs seem to be different from CD4+ T cells and exhibit the phenotype of memory cells (CD11ahigh, CCR7). In addition, losses of CD27 and CD28 on the cell surface are a sign of an advanced stage of cellular differentiation (30).

Unlike conventional CD4+ T cells functioning as immune mediators by the production of cytokines, CD4+ CTLs induce apoptosis in target cells. Results from studies investigating their death mechanisms show the role of perforin as a major effector molecule (23, 29, 31-34). Furthermore, Fas ligand (32) and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) are involved. The latter is a powerful inducer of apoptosis that acts through an unusually diverse system of receptors (32, 35, 36). TRAIL is important for immune surveillance, especially in the context of cancer (32, 35, 37, 38). It is not displayed on the surface of resting peripheral blood T cells but is significantly induced by stimulation with CD3 mAbs in the presence of IFNs (39). CD4+ T-cell clones were found to constitutively express TRAIL, which was fully or at least partially responsible for cytotoxicity against certain tumor cells (39). Sato et al. (40) reported that CD4+ CTLs acting via the TRAIL pathway were able to induce apoptosis in vascular smooth muscle cells present in atherosclerotic plaques of patients with acute coronary syndromes. These cells originated from a CD4+ CD28 subset that seemed to be enriched in the patients' blood (40). TRAIL as well as agonistic antibodies specific to TRAIL receptors, death receptors DR4 and DR5, have progressed to phase I and II clinical trials as potential therapeutic agents for a variety of malignancies (41, 42). At present, the classification of CD4+ CTLs can neither be assigned to distinct types of cancer targets nor are they always restricted to MHC (23, 25, 43-45). Their cytokine pattern varies as does their way of exerting cytotoxicity (23, 25, 43-48).

Because the variety of adoptive immunotherapy against cancer is as large as the promises it holds, in-depth understanding and improvements are of high relevance. We present here a novel method that allows the generation of a subset of CD4+ cytotoxic T cells, called tumor killer T cells (TKTC). These cells induce apoptosis in tumor cells of various tissue origins and leave normal cells unaffected. The cytotoxicity could be attributed, to a significant extent, to a TRAIL-dependent mechanism. The generation procedure is relatively easy to perform and does not require autologous tumor material but only small amounts of peripheral blood. The cultivation under serum-free conditions meets the requirements of their use as a therapeutic agent. Thus, this technology together with the special features of TKTCs might be the basis by which a novel type of adoptive immunotherapy of cancer may evolve.

Large-Scale Production of TKTCs

We found that among PBMCs, a T-cell population strongly proliferated on stimulation with the EBV-transformed B cell line HB617, and that depletion of cytotoxic cells using l-leucyl-l-leucine methyl ester improved expansion of these cells. l-Leucyl-l-leucine methyl ester is a lysosomatropic compound that is converted by dipeptidyl peptidase I to metabolites that are membranolytic for cytotoxic T cells, NK cells, and lymphokine activated killer cells (47).

To prepare the culture for adoptive transfer purposes, we established serum-free conditions. A stimulating pulse with HB617 for 5 days was sufficient, and the optimal growth factors were a combination of IL-2 for continuous growth plus the immunosuppressive drug cyclosporin A. The culture was adjusted to 3 × 106 cells/mL and readjusted every 2nd day. When the culture reached a volume of 50 mL, it was transferred into a 490-cm2 roller bottle or a Roux flask to compare TKTC growth under two different conditions. The cells propagated in the Roux flasks ceased to grow after 5 weeks (data not shown). However, cultures grown in the roller bottles could be propagated up to 1010 to 1012 cells in a period of about 8 to 10 weeks (Fig. 1A shows four representative growth curves). The enormous expansion of the culture in roller bottles is probably due to an improved oxygen supply and the increased cell density at the bottom of the bottle. The growth in the roller bottle was very consistent and allowed a 1.5-fold expansion every 2nd day. After 8 to 10 weeks in exponential growth phase, the cells stopped dividing and persisted in a nonproliferating state, maintaining a relatively high viability (≥80%). After a further 2 weeks, the viability rapidly decreased and the culture had to be abandoned. In total, we generated using this method eight different TKTC cultures: five from PBMCs of healthy donors (KT-HHD1 to KT-HHD5) and three from PBMCs of cancer patients (KT-PC1, KT-PC2, and KT-HCC1).

FIGURE 1.

Growth properties and phenotype of the generated TKTC lines. A. Growth curves of TKTC lines of four different donors cultured in roller bottles. B. Representative immunofluorescence analysis of antigen expression on the surface of TKTC KT-HDD2 after 8 wk in culture using specific mAbs and flow cytometry. C. Real-time quantitative PCR analysis of expression of Foxp3 by the different TKTC lines in comparison with conventional CD4+CD25 and CD4+CD25+ Tregs. D. TKTC heterogeneity of TCR expression decreased during cultivation as shown representatively for KT-HDD2. The expression of the 22 α and the 25 β TCR chains was analyzed by reverse transcriptase PCR. After 5 wk in culture, all chains were expressed (top), whereas after 9 wk of cultivation, the TCR profile was restricted to a few α and β chains and varied between the TKTC lines (bottom). E. Conventional CD4+CD25 T cells were cocultured with the TKTC line KT-PC1 at various E/T ratios for 6 d. The suppression of conventional CD4+CD25 T cells by autologous CD4+CD25+ Tregs served as positive control. As a further control, all cell types were also cultured alone. The proliferation was determined by quantifying [3H]thymidine incorporation during the last 18 h of culture. F. The KT-PC1 line was analyzed for expression of Foxp3 at the protein level by using the AlexaFluor647-labeled anti-Foxp3 mAb. For detection of the intracellular Foxp3, the cells were permeabilized with saponin and fixed with paraformaldehyde. An AlexaFluor647-labeled isotype-matched irrelevant control mAb and an AlexaFluor647-labeled CD4 mAb served as negative and positive controls, respectively.

FIGURE 1.

Growth properties and phenotype of the generated TKTC lines. A. Growth curves of TKTC lines of four different donors cultured in roller bottles. B. Representative immunofluorescence analysis of antigen expression on the surface of TKTC KT-HDD2 after 8 wk in culture using specific mAbs and flow cytometry. C. Real-time quantitative PCR analysis of expression of Foxp3 by the different TKTC lines in comparison with conventional CD4+CD25 and CD4+CD25+ Tregs. D. TKTC heterogeneity of TCR expression decreased during cultivation as shown representatively for KT-HDD2. The expression of the 22 α and the 25 β TCR chains was analyzed by reverse transcriptase PCR. After 5 wk in culture, all chains were expressed (top), whereas after 9 wk of cultivation, the TCR profile was restricted to a few α and β chains and varied between the TKTC lines (bottom). E. Conventional CD4+CD25 T cells were cocultured with the TKTC line KT-PC1 at various E/T ratios for 6 d. The suppression of conventional CD4+CD25 T cells by autologous CD4+CD25+ Tregs served as positive control. As a further control, all cell types were also cultured alone. The proliferation was determined by quantifying [3H]thymidine incorporation during the last 18 h of culture. F. The KT-PC1 line was analyzed for expression of Foxp3 at the protein level by using the AlexaFluor647-labeled anti-Foxp3 mAb. For detection of the intracellular Foxp3, the cells were permeabilized with saponin and fixed with paraformaldehyde. An AlexaFluor647-labeled isotype-matched irrelevant control mAb and an AlexaFluor647-labeled CD4 mAb served as negative and positive controls, respectively.

Close modal

To exclude any risk of virus transmission (hypothetical minute amounts of EBV or other unknown viruses), we performed a virus safety study to evaluate virus depletion and inactivation during the preparation of the stimulating cell line HB617, although it does not produce any detectable EBV virus (data not shown). Therefore, we mixed HB617 with two model viruses, polio and influenza. When we analyzed the virus titer after glutaric aldehyde fixation and the various washing steps, all viruses analyzed were reduced to the limit of detection of the assays. The reduction factors were 9.8 log/mL for polio and 12.8 log/mL for influenza (data not shown). Thus, we conclude that hypothetical viruses produced by HB617 cells are significantly removed and inactivated during the preparation process.

Cell Surface Phenotype of TKTCs

In all TKTC lines shown in this article, the percentage of CD4+ cells increased during cultivation, whereas the percentage of CD8+ cells decreased. After 8 weeks in culture, three of the eight TKTC lines contained basically only CD4+ cells (KT-PC1, 100%; KT-HCC1, 100%; KT-HHD2, 99%). Two lines contained ∼90% CD4+ cells (KT-HHD4, 91%; KT-PC2, 94%) and three contained ∼80% CD4+ cells (KT-HHD1, 80%; KT-HHD3, 82%; KT-HHD5, 83%); the residual cells in the latter five cultures were CD8+. The cells further expressed CD3, CD25, CD28, CD45RA, CD45RO, CD69, CD86, and MHC-II (Fig. 1B). Between the different TKTC cultures, the expression of surface markers was rather consistent. Only MHC class II displayed variation, and KT-HHD2 cells showed an extraordinarily high expression of the activation and memory marker CD45RO. In the populations containing a residual amount of CD8+ cells (KT-HHD1, KT-HHD3, KT-HHD4, and KT-HHD5), CD31 was solely expressed by the CD8+ subset (data not shown). The memory marker CD27 and chemokine receptor 7 (CCR7) were never detected. All TKTC cell lines analyzed were negative for CD56, CD57, CD161, and NKG2D at all times tested (i.e., after 4, 6, and 8 weeks in culture; Fig. 1B).

Foxp3 Expression by TKTCs

To evaluate Foxp3 expression, we performed real-time quantitative PCR. Conventional CD4+CD25 and CD4+CD25+ T regulatory lymphocytes (Treg) served as negative and positive control cells, respectively. TKTC lines expressed Foxp3 mRNA, however, at variable degrees (Fig. 1C). The highest Foxp3 mRNA expression (1.8× more than in CD4+CD25+ T lymphocytes) was found in the prostate tumor patient derived line KT-PC1, the lowest in line KT-HHD5, 16× less than in CD4+CD25+ T lymphocytes but 5× more than in CD4+CD25 T lymphocytes.

Because of its high FoxP3 mRNA expression level, we subjected the KT-PC1 line to a T-cell suppression assay using CD4+CD25+ Tregs and CD4+CD25 conventional T cells freshly isolated from human peripheral blood. Whereas the Tregs suppressed the conventional T cells in a dose-dependent manner, we did not observe any suppressive effect of the KT-PC1 line (Fig. 1E).

We then analyzed Foxp3 expression in KT-PC1 at the protein level using immunofluorescence staining and flow cytometry of permeabilized cells. In contrast to the mRNA level, the protein staining was low (Fig. 1F). We obtained the same results with TKTCs KT-HHD3 and KT-HCC1 (data not shown), indicating posttranscriptional control of Foxp3 expression in TKTCs.

TKTCs Are α/β T Cells and the Number of Expressed T-Cell Receptor Chains Decreases in the Course of Cultivation

As determined by reverse transcriptase PCR, all TKTC cultures contained exclusively α/β T cells. The expression of the T-cell receptor (TCR) chains was assessed from transcripts of each TKTC culture. Younger cultures of 5 weeks of age displayed rearrangement of all 47 TCR chains tested (Fig. 1D, top), similar to that of freshly isolated PBMCs (data not shown). After 6 to 8 weeks in culture, TKTCs lost expression of some TCR chains (Fig. 1D, bottom). Older cultures of 10 weeks of age became oligoclonal, displaying only two to five TCR chains. However, the TCR expression in oligoclonal TKTC cultures varied. In general, the faster a TKTC culture grew, the earlier the development of oligoclonality appeared.

Cytokine Profile of TKTCs

After a cultivation period of 6 weeks, we tested the supernatants of the TKTC lines for cytokine content by ELISA. As depicted in Table 1, the TKTC lines produced IL-10 and IFN-γ at high variation. IL-4 was produced by four TKTC lines (KT-HHD1, KT-HHD4, KT-HHD5, and KT-PC1) in low concentrations (3.6-37.3 pg/mL) and by others (KT-HHD2, KT-HHD4, and KT-HCC1) below the detection limit of the assay.

Table 1.

Cytokine Contents in TKTC Supernatants of 6-Wk-Old TKTC Cultures

TKTC LineIFN-γ (pg/mL)IL-4 (pg/mL)IL-10 (pg/mL)
KT-HHD1 1.4 11.9 — 
KT-HHD2 1,569.6 — 32.3 
KT-HHD3 13,798.3 — 1,644.8 
KT-HHD4 1,213.2 26.8 1,471.6 
KT-HHD5 1.5 3.6 383.7 
KT-PC1 3,236.0 37.3 663.3 
KT-HCC1 3,254.6 — 2,7720.9 
TKTC LineIFN-γ (pg/mL)IL-4 (pg/mL)IL-10 (pg/mL)
KT-HHD1 1.4 11.9 — 
KT-HHD2 1,569.6 — 32.3 
KT-HHD3 13,798.3 — 1,644.8 
KT-HHD4 1,213.2 26.8 1,471.6 
KT-HHD5 1.5 3.6 383.7 
KT-PC1 3,236.0 37.3 663.3 
KT-HCC1 3,254.6 — 2,7720.9 

TKTCs Induce Cell Death in Tumor Cells of Various Tissue Origin but Spare Normal Cells

To test the effector function of the generated cells, we analyzed the cytotoxicity of TKTCs toward a panel of tumor and normal cells and quantified the killing efficiency by flow cytometry. To distinguish the target cells in the coculture from TKTCs, we labeled them before the assay with carboxyfluorescein diacetate N-succinimidyl ester (CFSE). After the cocultivation, we stained the sample with propidium iodide (PI) to assess the percentage of dead target cells. Coincubation of fibrosarcoma Saos cells with a 10-fold excess of TKTCs led to an extensive increase in the percentage of PI+ tumor cells (Fig. 2A): 50% when incubated with the healthy donor TKTC culture KT-HHD5, and 64% by incubation with the cancer patient TKTC culture KT-PC1. The basal level of dead Saos cells was 4%. In contrast, when we exposed normal umbilical vein endothelial cells IE 104.4 to KT-HHD5 or KT-PC1, the basal level of 3% did not significantly increase but stayed low at 4% in both cases.

FIGURE 2.

Cytotoxicity of TKTCs to cancer cells versus normal cells. A. Fibrosarcoma Saos cells and normal human umbilical vein endothelial cells IE 104.4 were labeled with both CFSE and PI and incubated for 20 h with either KT-HHD5 (TKTC derived from a healthy donor) or KT-PC1 (TKTC derived from a prostate cancer patient). CFSE-positive cells were gated (top dotplots) and evaluated for PI fluorescence. B. The same assay was done by using different target cells and KT-HHD2 as TKTC effector cells. Black columns, TKTC-induced cell death; white columns, spontaneous target cell death without TKTCs. Assays were done in duplicates. MTC, medullary thyroid carcinoma; HUVEC, human umbilical vein endothelial cell IE 104.4 cells; fibroblasts, HDF5 fibroblasts; E.c. kidney, NHK13, human renal proximal tubular epithelial cells derived from kidney; ConA T blasts, concanavalin A–stimulated blast cells from a heterologous donor; n.v., no value.

FIGURE 2.

Cytotoxicity of TKTCs to cancer cells versus normal cells. A. Fibrosarcoma Saos cells and normal human umbilical vein endothelial cells IE 104.4 were labeled with both CFSE and PI and incubated for 20 h with either KT-HHD5 (TKTC derived from a healthy donor) or KT-PC1 (TKTC derived from a prostate cancer patient). CFSE-positive cells were gated (top dotplots) and evaluated for PI fluorescence. B. The same assay was done by using different target cells and KT-HHD2 as TKTC effector cells. Black columns, TKTC-induced cell death; white columns, spontaneous target cell death without TKTCs. Assays were done in duplicates. MTC, medullary thyroid carcinoma; HUVEC, human umbilical vein endothelial cell IE 104.4 cells; fibroblasts, HDF5 fibroblasts; E.c. kidney, NHK13, human renal proximal tubular epithelial cells derived from kidney; ConA T blasts, concanavalin A–stimulated blast cells from a heterologous donor; n.v., no value.

Close modal

This discrimination potential of the TKTCs was confirmed when we analyzed other normal and tumor-derived cells. As summarized in Table 2, all tumor-derived cell lines were killed by TKTCs, whereas normal cell types and lines were unaffected. There were, however, two exceptions: Burkitt's lymphoma Daudi cells and chronic myelogenous leukemia K562 cells, both classic NK cell targets, were not killed by TKTCs. Among the susceptible cells, there was a variation in the killing kinetics (Fig. 2B). The fibrosarcoma cell line Saos as well as melanoma and medullary thyroid carcinoma cell lines were faster killed by TKTC (50-70% dead cells after 20 hours) compared with HT-29 or Me180 cells (∼30% dead cells after 20 hours). These results show that TKTCs have a high capability to discriminate between normal and transformed cells.

Table 2.

Susceptibility of Different Cells and Cell Lines to TKTC Killing

Cell LineCell Type% Dead Target Cells*
Targets susceptible to TKTC killing   
    Saos Fibrosarcoma 47.7 
    MelLa Melanoma 66.0 
    MelE Melanoma 66.2 
    MelJuso Melanoma 23.0 
    DU-BZ Small-cell lung carcinoma Microscopic analysis only 
    DU-145 Prostate carcinoma Microscopic analysis only 
    Me180 Carcinoma of the uterine cervix 23.7 
    HT-29 Colon carcinoma 32.1 
    BT-20 Mamma carcinoma Microscopic analysis only 
    HEK 293 Transformed kidney cell line 75.0 
    HB617 EBV-transformed B cells 62.0 
    MTC Medullary thyroid carcinoma 53.3 
    THP-1 Acute monocytic leukemia Microscopic analysis only 
    HeLa Cervix adenocarcinoma Microscopic analysis only 
   
Targets resistant to TKTC killing   
    HDF-5 Normal diploid skin fibroblasts 5.0 
    HDF-6 Normal diploid skin fibroblasts 5.4 
    NHK-13 Normal epithelial kidney cells 2.4 
    NHK-27 Normal epithelial kidney cells 3.8 
    NHK-16 Normal epithelial kidney cells 5.1 
    ConA-1 ConA-stimulated T blasts −4.3 
    ConA-2 ConA-stimulated T blasts −12.77 
    ConA-3 ConA-stimulated T blasts 4.4 
    IE 104.4 Umbilical vein endothelial cells 0.9 
    Daudi Burkitt's lymphoma Microscopic analysis only 
    K562 Chronic myelogenous leukemia 4.7 
Cell LineCell Type% Dead Target Cells*
Targets susceptible to TKTC killing   
    Saos Fibrosarcoma 47.7 
    MelLa Melanoma 66.0 
    MelE Melanoma 66.2 
    MelJuso Melanoma 23.0 
    DU-BZ Small-cell lung carcinoma Microscopic analysis only 
    DU-145 Prostate carcinoma Microscopic analysis only 
    Me180 Carcinoma of the uterine cervix 23.7 
    HT-29 Colon carcinoma 32.1 
    BT-20 Mamma carcinoma Microscopic analysis only 
    HEK 293 Transformed kidney cell line 75.0 
    HB617 EBV-transformed B cells 62.0 
    MTC Medullary thyroid carcinoma 53.3 
    THP-1 Acute monocytic leukemia Microscopic analysis only 
    HeLa Cervix adenocarcinoma Microscopic analysis only 
   
Targets resistant to TKTC killing   
    HDF-5 Normal diploid skin fibroblasts 5.0 
    HDF-6 Normal diploid skin fibroblasts 5.4 
    NHK-13 Normal epithelial kidney cells 2.4 
    NHK-27 Normal epithelial kidney cells 3.8 
    NHK-16 Normal epithelial kidney cells 5.1 
    ConA-1 ConA-stimulated T blasts −4.3 
    ConA-2 ConA-stimulated T blasts −12.77 
    ConA-3 ConA-stimulated T blasts 4.4 
    IE 104.4 Umbilical vein endothelial cells 0.9 
    Daudi Burkitt's lymphoma Microscopic analysis only 
    K562 Chronic myelogenous leukemia 4.7 

Abbreviation: ConA, concanavalin A.

*

The percentage of dead target cells was assessed by the cytotoxity assay described in Materials and Methods. TKTC KT-HHD2 was used as effector cell line.

TKTC-Induced Cytotoxicity Is Cell Contact Dependent

To uncover the mechanisms underlying killing by TKTCs, we watched the induction of cell death by microscopy. Depending on the type of target cell, the process took place between 24 and 72 hours until all targets were eradicated. In particular, TKTCs required a longer time to exert their cytotoxicity when the targets grew in clusters, such as the HT-29 cells. The visual investigations showed a very tight contact between effector and target cells, suggesting that TKTCs kill their targets in a direct cell-to-cell contact manner (Fig. 3A). This assumption was confirmed by a transwell chamber setting, which did not lead to killing of the tumor cells (data not shown).

FIGURE 3.

TKTC-mediated cytotoxicity is cell contact dependent and results in induction of apoptosis. A. Target cells (top, fibrosarcoma Saos cells; bottom, human dermal fibroblasts HDF-5 as normal target cells) were incubated with the TKTC culture KT-HHD5 or KT-PC1 at an E/T ratio of 10:1. As control, the target cells were cultured without TKTCs (left). B. Apoptosis is induced in an E/T dose–dependent manner by TKTCs as measured by Annexin V and PI staining. KT-HHD2 cells were incubated for 20 h with PKH-26–labeled Saos cells at the indicated E/T ratios. Samples were first gated for PKH-26–positive Saos cells and then analyzed for early apoptotic (Annexin V+ PI) and late apoptotic and necrotic cells (Annexin V+ PI+). C. Cytotoxicity assay done with “old” CD4+ TKTCs. Melanoma cells MelLa were labeled with CFSE and incubated for 20 h with KT-HHD1, which had been in culture for 11 wk, at different E/T ratios. Cells were harvested and stained with PI. CFSE-positive cells were gated and evaluated for PI fluorescence.

FIGURE 3.

TKTC-mediated cytotoxicity is cell contact dependent and results in induction of apoptosis. A. Target cells (top, fibrosarcoma Saos cells; bottom, human dermal fibroblasts HDF-5 as normal target cells) were incubated with the TKTC culture KT-HHD5 or KT-PC1 at an E/T ratio of 10:1. As control, the target cells were cultured without TKTCs (left). B. Apoptosis is induced in an E/T dose–dependent manner by TKTCs as measured by Annexin V and PI staining. KT-HHD2 cells were incubated for 20 h with PKH-26–labeled Saos cells at the indicated E/T ratios. Samples were first gated for PKH-26–positive Saos cells and then analyzed for early apoptotic (Annexin V+ PI) and late apoptotic and necrotic cells (Annexin V+ PI+). C. Cytotoxicity assay done with “old” CD4+ TKTCs. Melanoma cells MelLa were labeled with CFSE and incubated for 20 h with KT-HHD1, which had been in culture for 11 wk, at different E/T ratios. Cells were harvested and stained with PI. CFSE-positive cells were gated and evaluated for PI fluorescence.

Close modal

TKTCs Induce Apoptosis in Target Cells

Next we assessed whether TKTC-mediated cytotoxicity in target cells is mediated by induction of apoptosis. For that purpose, we modified our flow cytometric cell death assay. Target cells were labeled with the membrane dye PKH-26 before exposure to effector cells. After 20 hours of exposure, cocultures were harvested and stained with Annexin V for detection of early apoptotic cells and with PI, which penetrates late apoptotic and necrotic cells. In Fig. 3B, we give an example of such an assay done on Saos cells, which were identified according to PKH-26 staining and analyzed for different stages of cell death. The analysis revealed an increase in early apoptosis to 12% at an effector/target ratio (E/T) ratio of 10:1 and 18% at an E/T of 20:1. Under the influence of TKTCs for 20 hours, the late apoptotic/necrotic Saos population reached a level of 45% at an E/T of 10:1 and 50% at an E/T of 20:1, whereas only 13% of Saos cells underwent apoptosis spontaneously.

Cytotoxic Capacity of Nonproliferating “Old” TKTCs

As mentioned above, TKTC cultures do not have the capacity to grow indefinitely but stop proliferating after an exponential growth phase over ∼8 to 10 weeks. During the following 2 weeks, persisting in a nonproliferative (potentially senescent; refs. 49, 50) state, the cells still possess cytotoxic activity. We show this function in Fig. 3C with the MelLa melanoma cell line as target and KT-HHD1 as TKTC effector. The latter had been cultured for 11 weeks before it stopped proliferation and then was kept for another week in culture. A 10-fold excess of the nonproliferating TKTCs led to 27% and a 20-fold excess to 54% PI+ MelLa cells, in contrast to a spontaneous cell death of 4% in the control assay without effector cells.

Killing of Target Cells by TKTCs Seems to Be MHC Unrestricted

To determine whether TKTC cytotoxicity is MHC restricted, we first assessed expression of MHC-I and MHC-II molecules on target cells using a panel of mAbs to different MHC class I and II epitopes (data not shown). Next, we incubated the tumor cells with those mAbs that positively reacted with the cells to block the MHC molecules and then subjected the treated cells to the flow cytometric cytotoxicity assay. The fibrosarcoma Saos cells were positive for MHC class I; however, none of the anti–MHC class I mAbs had an influence on killing of these cells by the TKTC culture KT-HHD2. In contrast, the cytotoxicity of a positive control CD8+ CTL line toward Saos cells was greatly impaired by the anti–MHC class I mAbs (Table 3). Furthermore, KT-HHD2 killing of MHC class II+ MelE melanoma cells was not influenced by three mAbs directed to different epitopes of MHC class II (data not shown). Because none of the mAbs of both the MHC class I and MHC class II panels inhibited killing, it is very likely (but not 100% proved) that TKTC killing is not restricted to MHC.

Table 3.

Effect of Anti–MHC Class I mAbs to Cytotoxicity of the TKTC Culture KT-HHD2 or of CD8+ CTLs to Fibrosarcoma Saos Cells

MHC Class I mAbKT-HHD2 (CD4+)CTL (CD8+)
MEM-119 3.9 18.8 
MEM-123 −4.2 18.9 
MEM-135 0.6 17.9 
MEM-147 0.3 37.2 
MEM-149 1.4 35.3 
MHC Class I mAbKT-HHD2 (CD4+)CTL (CD8+)
MEM-119 3.9 18.8 
MEM-123 −4.2 18.9 
MEM-135 0.6 17.9 
MEM-147 0.3 37.2 
MEM-149 1.4 35.3 

NOTE: The values represent reduction of cytotoxicity in percent.

Induction of Apoptosis by TKTCs Involves TRAIL

Next we analyzed, whether TKTCs use perforin-, Fas-, or TRAIL-based cytolytic pathways for cytotoxicity. Flow cytometry revealed a low expression of perforin in the TKTC lines (Fig. 4A). Experiments using concanamycin A to block perforin-mediated cytotoxicity mechanisms indicated that TKTC killing did not involve perforin (data not shown). Further, mAb SM1/23, the blocking mAb of Fas, was not able to reduce the cytotoxic effect of TKTCs, whereas it significantly reduced cell death induced by anti-Fas mAb CH-11 in the control cell line LN-18 (Fig. 4B). However, inhibition of TRAIL binding to its receptor resulted in a remarkable reduction of the amount of dead tumor targets. This effect was dependent on the concentration of the blocking anti-TRAIL mAb RIK-2 (Fig. 4C). Depending on the tumor target, killing was reduced by 17.4% to 46.8% (Table 4).

FIGURE 4.

Analysis of TKTC-mediated cytotoxicity. A. Expression of intracellular perforin was assessed by permeabilization of the cells using saponin, fixation with paraformaldehyde, and indirect immunofluorescence staining. An isotype-matched irrelevant mAb and a CD3 mAb served as negative and positive controls, respectively. As representatives, the TKTC lines KT-PC1, KT-HHD3, and KT-HCC1 were used. B. Fas blocking assay. Top, CFSE-labeled Saos cells were preincubated with or without different concentrations of the blocking anti-Fas mAb SM1/23. After cocultivation with KT-HHD2 for 20 h, PI was added and CFSE/PI double-positive cells were evaluated by flow cytometry. Bottom, as a control, LN-18 cells were preincubated with or without mAb SM1/23. Thereafter, the cells were treated with or without 100 ng/mL of cytotoxic anti-Fas mAb CH-11 for 48 h. Percentages of dead cells were evaluated by flow cytometry with PI staining. C. The TKTC line KT-HHD2 was preincubated with or without different concentrations of anti-TRAIL mAb RIK-2. After cocultivation with CFSE-labeled MelE for 20 h, PI was added and CFSE/PI double-positive cells were evaluated by flow cytometry. In all assays, tumor cells were labeled with CFSE and cocultivated for 20 h with KT-HHD2 that was pretreated with anti-TRAIL mAb. Afterward, PI was added and CFSE/PI double-positive cells were evaluated by flow cytometry. All experiments were done in duplicates.

FIGURE 4.

Analysis of TKTC-mediated cytotoxicity. A. Expression of intracellular perforin was assessed by permeabilization of the cells using saponin, fixation with paraformaldehyde, and indirect immunofluorescence staining. An isotype-matched irrelevant mAb and a CD3 mAb served as negative and positive controls, respectively. As representatives, the TKTC lines KT-PC1, KT-HHD3, and KT-HCC1 were used. B. Fas blocking assay. Top, CFSE-labeled Saos cells were preincubated with or without different concentrations of the blocking anti-Fas mAb SM1/23. After cocultivation with KT-HHD2 for 20 h, PI was added and CFSE/PI double-positive cells were evaluated by flow cytometry. Bottom, as a control, LN-18 cells were preincubated with or without mAb SM1/23. Thereafter, the cells were treated with or without 100 ng/mL of cytotoxic anti-Fas mAb CH-11 for 48 h. Percentages of dead cells were evaluated by flow cytometry with PI staining. C. The TKTC line KT-HHD2 was preincubated with or without different concentrations of anti-TRAIL mAb RIK-2. After cocultivation with CFSE-labeled MelE for 20 h, PI was added and CFSE/PI double-positive cells were evaluated by flow cytometry. In all assays, tumor cells were labeled with CFSE and cocultivated for 20 h with KT-HHD2 that was pretreated with anti-TRAIL mAb. Afterward, PI was added and CFSE/PI double-positive cells were evaluated by flow cytometry. All experiments were done in duplicates.

Close modal
Table 4.

TKTCs Execute Killing Partially via TRAIL

Target% Killing without aTRAIL% Killing + aTRAILReduction (%)
MelE 54.3 36.1 33.7 
MelLa 59.3 46.6 21.0 
MelJuso 25.1 20.5 18.1 
Saos 43.4 23.1 46.8 
HT_29 26.7 22.1 17.4 
Target% Killing without aTRAIL% Killing + aTRAILReduction (%)
MelE 54.3 36.1 33.7 
MelLa 59.3 46.6 21.0 
MelJuso 25.1 20.5 18.1 
Saos 43.4 23.1 46.8 
HT_29 26.7 22.1 17.4 

NOTE: KT-HHD2 was preincubated with or without 10 μg/mL anti-TRAIL mAb RIK-2 and then added to the indicated tumor target cells. Values represent means of duplicates.

Abbreviation: aTRAIL, anti-TRAIL mAb.

TRAIL Receptor Expression on Targets and Nontargets

To analyze the TRAIL-mediated apoptosis induction in tumor cells further, we analyzed surface expression of the TRAIL receptors TRAIL-R1 and TRAIL-R2. Because TRAIL receptors were reported to be expressed in some tumor cells only on stimulation (51, 52), we also tested for intracellular TRAIL receptors. As shown in Fig. 5, we found a correlation between expression of the TRAIL receptors on tumor cells and susceptibility to TKTC killing. All five susceptible tumor cell lines tested (Saos, MelE, MelLa, MelJuso, and HT-29—staining pattern identical to that of Saos cells; not shown) expressed TRAIL receptors either on the surface or intracellularly.

FIGURE 5.

Expression of TRAIL receptors on targets and nontargets of TKTCs. Susceptible and resistant cell lines to TKTC-mediated cytotoxicity were assessed for intracellular and surface expression of TRAIL-R1 and TRAIL-R2 by using indirect immunofluorescence and flow cytometry. For total (i.e., surface and intracellular) staining, saponin was used for permeabilization and paraformaldehyde for fixation of the cells.

FIGURE 5.

Expression of TRAIL receptors on targets and nontargets of TKTCs. Susceptible and resistant cell lines to TKTC-mediated cytotoxicity were assessed for intracellular and surface expression of TRAIL-R1 and TRAIL-R2 by using indirect immunofluorescence and flow cytometry. For total (i.e., surface and intracellular) staining, saponin was used for permeabilization and paraformaldehyde for fixation of the cells.

Close modal

On the contrary, the TKTC-resistant normal human renal proximal tubular epithelial cells NHK (53) and the normal human skin fibroblasts HDF-5 (54) did not express significant levels of TRAIL receptor. Normal human umbilical vein endothelial cells IE 104.4 as well as the classic NK target cell line K562, although not susceptible to TKTC killing, expressed TRAIL receptors. However, the latter finding is in agreement with recent reports, which show that TRAIL does not promote apoptosis but rather survival and proliferation in endothelial cells (55) and promotes apoptosis in K562 cells only on sensitization by specific agents (56-58). Thus, our results indicate that the TRAIL pathway is at least partially involved in the TKTC-mediated induction of apoptosis in tumor cells.

TKTCs from Tumor Patient Spare Normal Cells in an Autologous Ex vivo Setting

It is extremely difficult to obtain tumor as well as normal tissues from tumor patients, especially from those suffering from late-stage disease. Nevertheless, we were able to grow normal fibroblasts from the prostate cancer patient from whom TKTC line KT-PC2 was derived. Because normal cells have very long population doubling times, we were limited in target cell material and could only conduct microscopic cytotoxicity assays. Figure 6 shows the experiment conducted over a period of 1 week. Over that period, KT-PC2 cells, which had been added to the patient's normal fibroblasts in an E/T ratio of 10:1, vanished, whereas the autologous fibroblasts proliferated and almost formed a confluent layer after 7 days. Thus, the TKTCs were not able to induce cell death in the autologous fibroblasts. The establishment of tumor cell lines from primary tumors is not trivial and thus hampered our attempts to perform cytotoxicity experiments on autologous tumor cell lines.

FIGURE 6.

Cytotoxicity of TKTCs toward autologous fibroblasts from a tumor patient. After adherence of normal fibroblast isolated from a tumor patient, the autologous TKTC line KT-PC2 was added at an E/T ratio of 10:1. Over a period of 166 h of coculture, an almost confluent layer of fibroblasts was established.

FIGURE 6.

Cytotoxicity of TKTCs toward autologous fibroblasts from a tumor patient. After adherence of normal fibroblast isolated from a tumor patient, the autologous TKTC line KT-PC2 was added at an E/T ratio of 10:1. Over a period of 166 h of coculture, an almost confluent layer of fibroblasts was established.

Close modal

We show here a novel method to generate large amounts of TKTCs, a CD4+ population of cytotoxic T cells from PBMCs of peripheral blood, which induce apoptosis involving TRAIL in many transformed cells of different tissue origins but leave normal cells unaffected.

We found partial inhibition of TKTC killing of tumor cells by an anti-TRAIL mAb and correlation between expression of TRAIL receptors on target cells and susceptibility to TKTC killing. Factors that influence TRAIL sensitivity in multiple cancer cells are not well defined. A new study suggests that O-glycosylation of death receptors expressed in cancer cells modulates the sensitivity to TRAIL (36). In addition to the general lack of knowledge on the TRAIL mechanism, TKTCs provide another puzzle: Although anti-TRAIL mAbs reduced significantly the cytotoxic effect of TKTCs, we were unable to show the expression of TRAIL on the surface of TKTCs by flow cytometry (data not shown). TRAIL is also not found on the surface of peripheral blood T cells but expressed by costimulation with CD3 and type I IFNs (39). Further, on the surface of T-cell lines, TRAIL was shown to be constitutively expressed at various levels, and some of these lines initially down-regulate it before reexpression on CD3 activation (59, 60). This shows specific requirements and a high dynamic of TRAIL expression in T cells on differentiation/activation. Thus, we speculate that TKTCs might need a specific interaction with the susceptible target cells for TRAIL expression.

Differences in target cell recognition involving regulation of TRAIL expression may be as well a possible explanation for the delayed kinetics of TKTC cytotoxicity and the extent of killing dependent on the type of the target cell. The latter may be also influenced by the growth characteristics of the targets such as growth in clusters, which might hamper target cell recognition. Indeed, HT-29 cells that grow in clusters are less susceptible to TKTC killing. The IFN-γ produced by TKTCs may also be involved because it was reported that IFN-γ mediates susceptibility to TRAIL killing (61, 62). Furthermore, susceptibility of target cells may be dependent on their expression and exposure of TRAIL receptors. In two tumor targets, HT-29 and Saos, TRAIL-R1 and TRAIL-R2 could only be detected intracellularly. Because TKTCs need longer time to kill HT-29 than Saos cells, one could speculate that TRAIL receptors are probably differently transported to the surface of these cells because intracellular TRAIL receptors are only transported to the cell surface on contact.

Based on expression profiling and functional characteristics, TKTCs cannot be assigned to any known subtype of T cells. Although the expression profile of TKTCs (i.e., up-regulation of CD45RO, down-regulation of CD28 together with the lack of CD27 expression) resembles an effector memory phenotype (CD45RO+ CD27 CD28 CCR7; refs. 63, 64), they coexpress also the CD45RA isoform of the tyrosine phosphatase CD45, which is a feature of continuously cycling cells (63) retaining their functional program after activation (65). CD45RA+ CD45RO+ double-positive T cells can be isolated from human peripheral blood, where they show an intermediate phenotype for CD31, CD62L, CD58, and CD95, which are known to be differentially expressed on unprimed versus primed cells (66). However, TKTCs are negative for CD31 and CD62L.

CD4+ TKTC cultures do express on their surface at low density CD25 molecules, the IL-2 receptor α chain and one of the hallmarks of Tregs. Tregs can be subdivided into CD25high and CD25low cells. CD25high cells represent about 2% to 3% of the total CD4+ T-cell population, and at least in in vitro settings exhibit the characteristics postulated for Tregs initially identified in the mouse (67). Whereas the CD25high cells seem to be rather homogeneous in terms of their surface antigen expression, CD25low cells are more heterogeneous, which we could also somehow detect for TKTCs, in particular for MHC class II and CD45RO. However, CD69 (an antigen indicating recent activation) is not expressed on Tregs, but TKTCs were determined to be CD69+. Therefore, TKTCs seem to belong to a different cell subset. This assumption is further supported by the fact that Tregs can only be expanded to high numbers on antigenic stimulation in combination with IL-15, compared with activation in combination with IL-2 (68). Moreover, Tregs preferentially kill targets using a perforin-dependent mechanism (69), whereas TKTCs barely express perforin (Fig. 4A) and TKTC killing was not blocked by concanamycin A, which should block perforin-mediated cytotoxicity. We also evaluated Foxp3 expression. In contrast to mice, human Foxp3 is not restricted to Tregs but rather a T-cell activation marker probably counterbalancing the stimulation process for maintaining homeostasis (70). All TKTC lines tested expressed Foxp3 mRNA, but at variable degrees from very low levels to high expression (16× less up to 1.8× more than CD4+CD25+ Treg lymphocytes that served as positive control). Strikingly, in the mRNA high lines, Foxp3 expression was low at the protein level, suggesting posttranscriptional control of Foxp3 in these TKTCs. Finally, we tested the TKTC line KT-PC1 in a T-cell suppression assay, because it showed the highest Foxp3 mRNA expression. As shown in Fig. 1E, this line did not show any suppressive capacity.

TKTCs can also be clearly distinguished from natural killer T (NKT) cells. Although killing of TKTC, similar to that of CD4+ NKT cells, is MHC independent (71), in contrast to NKT and NKT-like cells, TKTCs do not express the typical NK cell markers CD56, CD57, CD161, and NKG2D. Furthermore, classic NKT cells are characterized by the expression of distinct TCR chains, namely Vα24 and Jα18 coexpressed with Vβ11. However, the TKTC lines are not restricted to the NKT TCRs but vary in their TCR expressions.

The fact that TKTCs secrete IFN-γ rather than IL-4 leads to the assumption that this cell type might be related rather to T cells of the Th1 type than to Th2 or Tregs. In addition, TKTCs also produce IL-10. During many infections, IL-10–producing effector Th1 cells are generated and thought to represent a regulatory mechanism to dampen exaggerated inflammation (72). Thus, it will be a future goal to investigate whether the IFN-γ/IL-10 double-producing TKTCs could both exert antitumor capacity and, via secretion of IL-10, locally control the defense mechanism thereby preventing systemic inflammation.

During the first few weeks of a TKTC culture, the permanent presence of IL-2 as well as components of HB617 might lead to an unspecific activation and proliferation of all T cells in the culture. However, to cause distinct T-cell clones to proliferate for 2 months and beyond, a stronger activation signal must be provided. Whether this signal is due to specific antigen stimulation or not, we tested the TKTC lines for TCR clonality by PCR. TKTC cultures of 4 to 6 weeks of age showed polyclonality. Because TCR diversity diminished in the course of cultivation (e.g., KT-PC1 turned out to be oligoclonal and KT-HCC1 was even monoclonal), it is tempting to speculate that a specific kind of signal leads to the activation and proliferation of TKTCs. However, the V segment usage varied between the TKTC cultures: Oligoclonal cultures expressed different TCR-α and TCR-β chains.

It should be noted that the stimulating cell line HB617 is EBV+. It is well documented that CD4+ CTLs are increased in EBV infection (27). In fact, patients experience a primary burst of CD4+ effector T cells, which are then down-regulated. EBV-specific CD4+ effector T cells stay enriched in the CD27+CD28+ compartment during the primary and persistent phases of infection (30, 73). Furthermore, many of these T cells coexpress CCR7 as opposed to TKTCs, which clearly were confirmed to be CD27 and CCR7-. Unlike EBV-associated CD4+ CTLs described in the literature, which gain cytotoxic potential by the acquisition of lytic granules containing granzymes as they lose CD27 expression and acquisition of perforin as soon as they are CD28 (30), TKTCs exert cytotoxicity, at least in part, by the TRAIL pathway. Moreover, EBV-specific CD4+ and CD8+ effector T cells are MHC restricted (30, 74), whereas our data indicate that TKTC cytotoxicity is rather MHC independent.

In summary, we provide a relatively simple and cheap protocol to generate large amounts of a novel type of CD4+ CTLs, termed TKTCs, with broad tumor specificity. Whether this population exists in vivo and/or is expanded under certain pathologic conditions remains to be elucidated in next studies. However, we cannot grow TKTCs in vitro without depletion of monocytes, NK cells, and suppressor T cells by using l-leucyl-l-leucine methyl ester. Thus, based on these in vitro studies, we suggest that in vivo the generation of this subset is likely to be impaired and controlled by other cells of the immune system. The charm of this particular T-cell subset for potential tumor patient therapy is given by the possibility to generate it in large amounts from small samples of peripheral blood and its broad killing of tumor cells involving the TRAIL pathway. Many tumors manage to develop resistance to conventional therapy on treatment. However, TRAIL is able to bypass this antiapoptotic effect in some tumor systems (75). Therefore, research groups investigating antitumor immunotherapies started to focus on TRAIL (76). In addition, the fact that TKTC killing is probably MHC unrestricted might be an additional advantage in patients whose tumor has down-regulated MHC expression to escape immune surveillance. These characteristics of TKTCs together with the technology to generate them serum-free under good manufacturing practice conditions prove this novel cell type as a potential new tool for adoptive immunotherapy against cancer.

Preparation of TKTCs

Heparinized human whole blood donations were purchased from the Austrian Red Cross for the preparation of five TKTC cultures from healthy human donors, designated KT-HHD1 to KT-HHD5. Heparinized whole blood from one colorectal carcinoma patient as well as from two prostate adenocarcinoma patients was obtained using sodium heparin–coated Vacutainer vials (Becton Dickinson) for the establishment of tumor patient TKTC cultures designated KT-HCC1 (killer T-human colorectal carcinoma patient 1) and KT-PC1 and KT-PC2 (killer T-prostate carcinoma patients 1 and 2). PBMCs were isolated by Ficoll-Paque (GE Healthcare) gradient centrifugation. Monocytes, NK cells, and cytotoxic T cells were eliminated by incubation with l-leucyl-l-leucine methyl ester hydrochloride (NovaBiochem) for 20 min at room temperature as described (47). Remaining cells were stimulated with the lymphoblastoid cell line HB617 (fixed with 0.02% glutaraldehyde for 10 min at room temperature) in vitro for 5 d and then propagated in serum-free medium (Aim-V, Life Technologies, Inc.) supplemented with 360 IU/mL IL-2 (Proleukin, Chiron Corp.) and 12.5 ng/mL cyclosporine A (Novartis Pharma). The cell number was adjusted to 3 × 106/mL every other day, and the population doubling time approached 3 d. TKTCs were propagated in Roux flasks until they had reached a volume of 50 mL. The cell number was upscaled in roller bottles rotating at 1 rpm at 37°C and 5% CO2.

Cell Lines and Culture Conditions

The stimulator cell line HB617 and the target cell lines Daudi (American Type Culture Collection), K562, and Saos were cultivated in X-Vivo 20 medium (Cambrex Bio Science) and passaged every 3 to 4 d. All melanoma cell lines—MelE, MelLa, and MelJuso—were cultivated in RPMI 1640 (Biochrom AG)/4 mmol/L l-glutamine/10% FCS (Hyclone) and passaged every 3 to 4 d at a split ratio of 1:10. The colon carcinoma cell line HT-29 (American Type Culture Collection) was cultivated in D-MEM/Ham's F-12 1:1 (Biochrom)/4 mmol/L l-glutamine/10% FCS and passaged every 3 to 4 d. The uterine cervix carcinoma cell line Me180 (American Type Culture Collection) was propagated in RPMI 1640/4 mmol/L l-glutamine/10% FCS and passaged every 3 to 4 d. LN-18 were grown in D-MEM (Biochrom)/4 mmol/L l-glutamine/5% FCS and passaged every 3 to 4 d, 1:5. The medullary thyroid carcinoma cell line (kindly provided by R. Pfragner, Institute of Pathophysiology, Medical University of Graz, Graz, Austria) was cultivated in Ham's F-12 (Biochrom)/4 mmol/L l-glutamine/10% FCS and passaged every 3 to 4 d at a split ratio of 1:1 or 1:2 as described (77). Target cells were split at a ratio of 1:5, unless indicated otherwise.

Normal human umbilical vein endothelial cells IE 104.4 were isolated using standard procedures and propagated in M199 (Biochrom)/15% FCS/10% endothelial cell growth supplement with a subculture ratio of 1:2 every 3 to 4 d (78). Normal human kidney epithelial cells NHK-13, NHK-27, and NHK-16 were cultivated in serum-free medium and passaged every 3 to 4 d at a split ratio of 1:2 as described (79). Normal human fibroblasts HDF-5 and HDF-6 were isolated from skin biopsies (28) and cultivated in D-MEM/Ham's F12 (1:1)/4 mmol/L l-glutamine/10% FCS and passaged at a split ratio of 1:2 every 3 to 4 d (54). Concanavalin A–stimulated blast cells were generated by culture of freshly isolated PBMCs in RPMI 1640/4 mmol/L l-glutamine/10% FCS supplemented with 5 μg/mL concanavalin A (Sigma-Aldrich; ref. 80) and 10 ng/mL IL-2 for 5 d. All cells were cultivated in a humidified atmosphere at 37°C and 5% CO2.

Virus Safety Study

Polio as a small, nonenveloped, robust virus and influenza representing medium to large, enveloped viruses were chosen as model viruses based on the recommendation of the Committee for Proprietary Medicinal Products (81). To show virus removal and inactivation in the course of the fixation and washing steps of HB617, the protocol for TKTC preparation was done as described above using HB617 cell cultures mixed with polio or influenza virus, respectively, before the centrifugation/washing step, before fixation, and before washing after fixation. After addition of fixed HB617 to PBMCs on day 1, virus was measured and virus reduction factors were calculated for each individual step as described earlier (81), which resulted in the cumulated reduction factor.

Antibodies

The following mouse anti-human mAbs were kindly provided by Vaclav Horejsi (Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic): MEM-57 (CD3ε; IgG2a), MEM-115 (CD4; IgG2a), MEM-31 (CD8; IgG2a), MEM-174 (CD11b; IgG2a), MEM-18 (CD14; IgG1), MEM-168 (CD16; IgM), WIN-19 (CD19), MEM-97 (CD20; IgG1), MEM-181 (CD25; IgG1), MEM-56 (CD45RA; IgG2b), MEM-28 (CD45; IgG1), MEM-102 (CD48; IgG1), MEM-188 (CD56; IgG2a), MEM-43/5 (CD59; IgG2b), MEM-75 (CD71; IgG1), MEM-119 (MHC-I; IgM), MEM-123 (MHC-I; IgG3), MEM-135 (MHC-I; IgG1), MEM-147 (MHC-I α chain; IgG1), MEM-149 (MHC-I; IgG1), MEM-32B (MHC-II DR dimer; IgG1), MEM-136 (MHC-II DR, DP, DQ β chain; IgG1), and MEM-137 (MHC-II DR α chain; IgG1). The mouse anti-human mAb 248.23.2 (CD28; IgM) was a kind gift from U. Moebius (University of Heidelberg, Heidelberg, Germany), mouse anti-human mAb UCHL-1 (CD45RO; IgG2a) from Peter Beverley (Jenner Institute, Compton, United Kingdom), and mouse anti-human mAb FN50 (CD69; IgG1) from Steinar Funderud (Norwegian Radium Hospital, Oslo, Norway). Other mAbs were obtained from the 6th Human Leukocyte Differentiation Antigen workshop: Dreg56 (CD62L; IgG1) and BU63 (CD86; IgG1). Mouse anti-human mAbs to TRAP1 (CD154; IgG1), RIK-2 (TRAIL; IgG1), TRAIL-R1 (DR4), TRAIL-R2 (DR5), perforin (δG9; IgG2b, phycoerythrin labeled), and Foxp3 (259D/C7; IgG1, AlexaFluor647 labeled) were from Becton Dickinson Biosciences Pharmingen; mAb DX2 (CD95; IgG1) was from Sigma-Aldrich. The blocking mAb to human Apo-1/Fas (SM1/23) was purchased from Bender MedSystems. The apoptosis-inducing mAb to human Fas (CH-11) was purchased from Immunotech. The CD3 mAb OKT3 was from Orthoclone.

Immunophenotyping

For cell surface immunofluorescence staining, cells were pelleted and resuspended in staining buffer (PBS/10% FCS/0.02% NaN3). After blocking (10% human AB serum) for 10 min on ice, cells were seeded into a 96-well round-bottomed plate and titrated concentrations of primary mAbs were added. After a 45-min incubation on ice, cells were washed twice with staining buffer and resuspended in 10 μg/mL FITC-conjugated goat anti-mouse F(ab′)2 antibody fragments (Sigma-Aldrich) diluted in staining buffer. The cells were left for 30 min on ice and then washed twice with buffer. Pellets were resuspended in PBS. Before measurement, PI (Sigma-Aldrich) was added to a final concentration of 250 ng/mL for exclusion of dead cells.

For intracellular immunofluorescence staining and flow cytometric analysis, the cells were permeabilized with 0.3% saponin for 10 min at room temperature and fixed with 1% paraformaldehyde in PBS for 15 min. Then the cells were incubated for 20 min with the primary mAbs. In case of indirect staining, the cells were washed and incubated for another 20 min with the fluorophore-conjugated secondary antibody.

Samples were measured on a FACSCalibur (Becton Dickinson) in a standard three-color setup. Analysis was done using the CellQuest and FlowJo softwares.

Real-time Quantitative PCR

For quantitative real-time PCR analysis, RNA was extracted using TRI Reagent (Sigma-Aldrich) according to the manufacturer's instructions. cDNA was prepared with oligo-dT primers (VBC) and StrataScript reverse transcriptase (Stratagene). mRNA levels were quantified via the LightCycler instrument (Roche Diagnostics GmbH). Amplification was done in a total volume of 10 μL for 50 cycles of 5 s at 95°C and 15 s at 72°C. The product was detected using SYBR Green I dye (Roche Diagnostics). The relative expression of the samples was determined by normalizing the expression of each target to elongation factor 1α (EF1a) and then comparing this normalized value to the normalized expression in a reference sample (CD4+CD25 cells) to calculate the fold change value. Primers were designed so that amplicons spanned intron/exon boundaries to minimize amplification of genomic DNA. Primer sequences were as follows: EF1a, 5′-GTGCTAACATGCCTTGGTTC-3′ and 5′-AGAACACCAGTCTCCACTCG-3′; Foxp3, 5′-GAAACAGCACATTCCCAGAGTTC-3′ and 5′-ATGGCCCAGCGGATGAG-3′.

Isolation of CD4+CD25 and CD4+CD25+ T Lymphocytes

PBMCs were isolated from blood of healthy donors by standard density-gradient centrifugation using Lymphoprep (Nycomed). CD4+ T cells were purified from the PBMCs by depletion of CD8-, CD14-, CD16-, CD19-, CD20-, and CD56-positive cells using mouse anti-human mAbs (CD8 mAb MEM-87, CD14 mAb MEM-18, CD16 mAb MEM-154, CD19 mAb WIJ09, CD20 mAb MEM-97, and CD56 mAb MEM-188) and magnetic sorting using goat anti-mouse IgG–coated microbeads (Miltenyi Biotec). The resulting CD4+ T-cell population was separated into conventional CD4+CD25 T cells and CD4+CD25+ Tregs by using anti-CD25 magnetic microbeads (Miltenyi Biotec).

T-Cell Suppression Assay

For the suppression assay, we incubated either autologous CD4+CD25+ Tregs or TKTCs together with CD4+CD25 conventional T cells (5 × 104 per well) in 96-well flat-bottomed tissue culture plates (Nunc) at the E/T ratios 1:1, 1:2, or 1:4. We induced the proliferation of the CD4+CD25 T cells by adding irradiated non–T cells (1 × 105 per well) and the CD3 mAb OKT3 at a final concentration of 1 μg/mL. The stimulatory allogeneic non–T cells were prepared by depleting T cells from PBMCs using rosetting with neuraminidase (Sigma-Aldrich)–treated sheep erythrocytes (Dade Behring) and irradiation of the obtained cell population twice at 3,000 rad. The assay was done in triplicates at a final volume of 200 μL. After 6 d of incubation, proliferation was determined by quantifying [3H]thymidine (1 μCi/well) incorporation during the last 18 h of culture. Results are presented as mean ± SD. The medium used throughout was RPMI 1640 (Sigma-Aldrich) supplemented with 5% FCS (Life Technologies), 2 mmol/L glutamine, 100 units/mL penicillin, and 100 μg/mL streptomycin (all Invitrogen).

Cytokine Analysis

Six weeks after isolation from whole blood, TKTC cultures were adjusted to 3 × 106 cells/mL and supernatants were collected 48 h later by centrifugation of cells at 170 × g for 10 min. Samples were stored at −80°C until analyzed. Concentrations of IL-4, IL-10, and IFN-γ were determined by ELISA using 96-well microplates (Corning 9018) coated with catching antibody (diluted in PBS to 5 μg/mL, 100 μL/well) at 4°C over night. Plates were washed and blocked with PBS/2% bovine serum albumin (150 μL/well) for 1 h at 37°C. Standards as well as samples were diluted in four steps at a ratio of 1:4 in RPMI 1640/10% FCS and pipetted into the precoated plates (70 μL/well). After 90 min at 37°C, plates were washed again and biotinylated secondary antibody was applied. After 60 min at 37°C, the plates were washed, the streptavidin-alkaline phosphatase conjugate (Chemicon) was added (1:2,000), and incubation was continued for 25 min at room temperature. After another washing step, substrate was added and incubated for about 30 min at room temperature until a yellow staining of all standard dilutions appeared. Plates were measured at 405 nm (reference wavelength, 650 nm). Values were assessed in duplicates. All standards, coating, and secondary antibodies were purchased from R&D Systems.

T-Cell Receptor Analysis

Total RNA was isolated with Trizol reagent (Invitrogen) and 2.5 μg were reverse transcribed with Superscript II (Invitrogen) following the manufacturer's instructions. One hundredth of the resulting cDNA was used as template in a standard 50-μL PCR reaction. 5′ primers specific for 22 Vα and 25 Vβ region genes (all of which were amplified with the same 3′ Cα and Cβ region primers) were designed according to sequences of T-Cell Receptor Typing Amplimer Kits (Clontech).

Cytotoxicity Assays

For microscopic evaluation of TKTC cytotoxicity, target cells were seeded into 24-well plates. After adherence, effector cells were added at an E/T ratio of 10:1.

For the flow cytometric cytotoxicity assays, the target cells were labeled with CFSE (Sigma-Aldrich) according to the manufacturer's instructions. Briefly, target cells were washed twice in HBSS, and then 106 cells were pelleted and resuspended in HBSS/10 μmol/L CFSE and incubated at 37° C for 20 min in the dark. The staining reaction was stopped by addition of 5 mL Aim-V/10% FCS. After exposure to TKTCs for 20 h in cell culture plates at various E/T ratios, the coculture was harvested, pelleted, resuspended in PBS, and stained with PI at 250 ng/mL for 5 min before acquisition. For analysis, CFSE-positive target cells were gated and evaluated for percent PI-positive cells, which were regarded as late apoptotic or necrotic. All samples were analyzed using a FACSCalibur. All assays using tumor cell targets were done in AIM-V (for serum-free target cells) or AIM-V supplemented with 10% FCS and, if necessary, growth supplements (for targets growing in serum-containing medium). Normal cell targets were cultured in the corresponding medium.

Apoptosis Assay

Induction of apoptosis was evaluated by a flow cytometric assay adapted from Fischer et al. (82). Briefly, target cells were labeled with the membrane dye PKH-26 (Sigma-Aldrich) according to the manufacturer's instructions for distinction between target and effector cells. After exposure to TKTCs for 20 h at various E/T ratios, the coculture was harvested. For that purpose, coculture supernatants (containing TKTCs and cell debris) were transferred into a 10-mL vial; remaining adherent cells were trypsinized and added to the supernatants. The mixture was centrifuged, washed in cold PBS, and costained with Annexin V-FITC (Roche) and PI for discrimination between viable, apoptotic, and late apoptotic/necrotic cells. Media were used as described above (cytotoxicity assays). For analysis, PKH-26+ target cells were gated and evaluated for percent Annexin V+PI cells, which were in the early apoptotic state, as well as percent Annexin V+PI+ cells, which were regarded as late apoptotic or necrotic. All samples were analyzed using a FACSCalibur.

Blocking Assays

These assays are based on the flow cytometric cytotoxicity assay described above using CFSE and PI. TKTCs were preincubated with anti-Fas mAb SM1/23 or anti-TRAIL mAb RIK-2 at a concentration of 1, 10, or 100 μg/mL for 1 h at 37°C before their addition to the targets. Blocking antibodies were maintained in cultures throughout the whole experiment. As positive control of the FAS blocking mAb, 2 × 105 LN-18 cells were preincubated for 1 h at 37°C in the absence and in the presence of 1 μg/mL blocking anti-FAS SM1/23 mAb. Thereafter, the cells were treated with or without 100 ng/mL of cytotoxic anti-Fas CH-11 mAb for 48 h. After detachment and centrifugation, the percentages of dead cells were evaluated by PI staining and flow cytometric analysis on a FACSCalibur. To test for MHC restriction, targets were seeded into tissue culture plates. After adherence, MHC-I or MHC-II blocking mAbs were added according to the expression of their corresponding epitopes on the target cells at a concentration of 5 or 50 μg/mL and preincubated for 1 h before TKTC addition. After 20 h of cocultivation, all cultures were harvested and the percentage of PI positive (PI+) cells was assessed by flow cytometry. Results are presented as percent killing reduction [%PI+ cells of the control assay (without blocking agent) were taken as 100% killing] in the presence of blocking agent. Each experiment was done in duplicate; results are presented as mean ± SD.

Dr. Hermann Katinger is the founder and CEO of Polymun Scientific Immunbiologische Forschung GmbH. Thomas Hemetsberger is employed by Polymun Scientific Immunbiologische Forschung GmbH.

Grant support: Polymun Scientific Immunbiologische Forschung GmbH.

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

We thank Thomas Sterovsky for excellent technical support; Vaclav Horejsi for providing a huge panel of invaluable mAbs and Gerhard Zlabinger (Institute of Immunology, Medical University of Vienna, Vienna, Austria) for assisting and measuring the cytokines; and David Bernhard (Institute of Experimental Pathophysiology and Immunology, Medical University Innsbruck, Innsbruck, Austria) for providing CH-11 mAb.

1
Terme M, Ullrich E, Delahaye NF, Chaput N, Zitvogel L. Natural killer cell-directed therapies: moving from unexpected results to successful strategies.
Nat Immunol
2008
;
9
:
486
–94.
2
Motohashi S, Nakayama T. Clinical applications of natural killer T cell-based immunotherapy for cancer.
Cancer Sci
2008
;
99
:
638
–45.
3
Ballestrero A, Boy D, Moran E, Cirmena G, Brossart P, Nencioni A. Immunotherapy with dendritic cells for cancer.
Adv Drug Deliv Rev
2008
;
60
:
173
–83.
4
Lundqvist A, Childs R. Allogeneic hematopoietic cell transplantation as immunotherapy for solid tumors: current status and future directions.
J Immunother
2005
;
28
:
281
–8.
5
June CH. Adoptive T cell therapy for cancer in the clinic.
J Clin Invest
2007
;
117
:
1466
–76.
6
Rosenberg SA, Restifo NP, Yang JC, Morgan RA, Dudley ME. Adoptive cell transfer: a clinical path to effective cancer immunotherapy.
Nat Rev Cancer
2008
;
8
:
299
–308.
7
Xue SA, Stauss HJ. Enhancing immune responses for cancer therapy.
Cell Mol Immunol
2007
;
4
:
173
–84.
8
Berger C, Berger M, Feng J, Riddell SR. Genetic modification of T cells for immunotherapy.
Expert Opin Biol Ther
2007
;
7
:
1167
–82.
9
Zhou J, Zhong Y. Breast cancer immunotherapy.
Cell Mol Immunol
2004
;
1
:
247
–55.
10
McNeel DG. Cellular immunotherapies for prostate cancer.
Biomed Pharmacother
2007
;
61
:
315
–22.
11
Chu CS, Kim SH, June CH, Coukos G. Immunotherapy opportunities in ovarian cancer.
Expert Rev Anticancer Ther
2008
;
8
:
243
–57.
12
Katipamula R, Markovic SN. Emerging therapies for melanoma.
Expert Rev Anticancer Ther
2008
;
8
:
553
–60.
13
Cooper LJ. Adoptive cellular immunotherapy for childhood malignancies.
Bone Marrow Transplant
2008
;
41
:
183
–92.
14
Stift A, Sachet M, Yagubian R, et al. Dendritic cell vaccination in medullary thyroid carcinoma.
Clin Cancer Res
2004
;
10
:
2944
–53.
15
Wysocki PJ, Zolnierek J, Szczylik C, Mackiewicz A. Recent developments in renal cell cancer immunotherapy.
Expert Opin Biol Ther
2007
;
7
:
727
–37.
16
Rosenberg SA, Lotze MT, Muul LM, et al. Observations on the systemic administration of autologous lymphokine-activated killer cells and recombinant interleukin-2 to patients with metastatic cancer.
N Engl J Med
1985
;
313
:
1485
–92.
17
Rosenberg SA, Spiess P, Lafreniere R. A new approach to the adoptive immunotherapy of cancer with tumor-infiltrating lymphocytes.
Science
1986
;
233
:
1318
–21.
18
Dudley ME, Rosenberg SA. Adoptive cell transfer therapy.
Semin Oncol
2007
;
34
:
524
–31.
19
Heemskerk B, Liu K, Dudley ME, et al. Adoptive cell therapy for patients with melanoma, using tumor-infiltrating lymphocytes genetically engineered to secrete interleukin-2.
Hum Gene Ther
2008
;
19
:
496
–510.
20
Dudley ME, Wunderlich JR, Shelton TE, Even J, Rosenberg SA. Generation of tumor-infiltrating lymphocyte cultures for use in adoptive transfer therapy for melanoma patients.
J Immunother
2003
;
26
:
332
–42.
21
Curti BD, Ochoa AC, Powers GC, et al. Phase I trial of anti-CD3-stimulated CD4+ T cells, infusional interleukin-2, and cyclophosphamide in patients with advanced cancer.
J Clin Oncol
1998
;
16
:
2752
–60.
22
Murphy K, Travers P, Walport M. Janeway's immunobiology. 7th ed. New York, Oxford: Garland Science Publishing; 2007.
23
Echchakir H, Bagot M, Dorothee G, et al. Cutaneous T cell lymphoma reactive CD4+ cytotoxic T lymphocyte clones display a Th1 cytokine profile and use a fas-independent pathway for specific tumor cell lysis.
J Invest Dermatol
2000
;
115
:
74
–80.
24
Nishimura M, Avichezer D, Custer M, et al. MHC class I-restricted recognition of a melanoma antigen by a human CD4+ tumor infiltrating lymphocyte.
Cancer Res
1999
;
59
:
6230
–8.
25
Semino C, Ferlazzo G, Ratto G, Melioli G. Cytotoxic properties of CD4+ T-cell clones which lyse HLA class II negative autologous non-small-cell lung cancer cells.
Cell Immunol
1999
;
196
:
87
–94.
26
Fleischer B. Acquisition of specific cytotoxic activity by human T4+ T lymphocytes in culture.
Nature
1984
;
308
:
365
–7.
27
Appay V, Zaunders J, Papagno L, et al. Characterization of CD4+ CTLs ex vivo.
J Immunol
2002
;
168
:
5954
–8.
28
Doyle A, Griffith JB. Cell and tissue culture: laboratory procedures in biotechnology. West Sussex: John Wiley & Sons, Inc. 1998.
29
Namekawa T, Wagner U, Goronzy J, Weyand C. Functional subsets of CD4 T cells in rheumatoid synovitis.
Arthritis Rheum
1998
;
41
:
2108
–16.
30
Appay V. The physiological role of cytotoxic CD4+ T-cells: the holy grail?
Clin Exp Immunol
2004
;
138
:
10
–3.
31
Duftner C, Goldberger C, Falkenbach A, et al. Prevalence, clinical relevance and characterization of circulating cytotoxic CD4+CD28 T cells in ankylosing spondylitis.
Arthritis Res. Ther.
2003
;
5
:
R292
–300.
32
Gagnon S, Ennis F, Rothman A. Bystander target cell lysis and cytokine production by dengue virus-specific human CD4+ cytotoxic T-lymphocyte clones.
J Virol
1999
;
73
:
3623
–9.
33
Wiley S, Schooley K, Smolak P, et al. Identification and characterization of a new member of the TNF family that induces apoptosis.
Immunity
1995
;
3
:
673
–82.
34
Yasukawa M, Ohminami H, Arai J, Kasahara Y, Ishida Y, Fujita S. Granule exocytosis, and not the fas/fas ligand system, is the main pathway of cytotoxicity mediated by alloantigen-specific CD4+ as well as CD8+ cytotoxic T lymphocytes in humans.
Blood
2000
;
95
:
2352
–5.
35
Almasan A, Ashkenazi A. Apo2L/TRAIL: apoptosis signalling, biology, and potential for cancer therapy.
Cytokine Growth Factor Rev
2003
;
14
:
337
–48.
36
Wagner KW, Punnoose EA, Januario T, et al. Death-receptor O-glycosylation controls tumor-cell sensitivity to the proapoptotic ligand Apo2L/TRAIL.
Nat Med
2007
;
13
:
1070
–7.
37
Gomez-Benito M, Balsas P, Bosque A, Anel A, Marzo I, Naval J. Apo2L/TRAIL is an indirect mediator of apoptosis induced by interferon-α in human myeloma cells.
FEBS Lett
2005
;
579
:
6217
–22.
38
Seki N, Hayakawa Y, Brooks A, et al. Tumor necrosis factor-related apoptosis-inducing ligand-mediated apoptosis is an important endogenous mechanism for resistance to liver metastases in murine renal cancer.
Cancer Res
2003
;
63
:
207
–13.
39
Kayagaki N, Yamaguchi N, Nakayama M, Eto H, Okumura K, Yagita H. Type I interferons (IFNs) regulate tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) expression on human T cells: A novel mechanism for the antitumor effects of type I IFNs.
J Exp Med
1999
;
189
:
1451
–60.
40
Sato K, Niessner A, Kopecky S, Frye R, Goronzy J, Weyand C. TRAIL-expressing T cells induce apoptosis of vascular smooth muscle cells in the atherosclerotic plaque.
J Exp Med
2006
;
203
:
239
–50.
41
Elrod HA, Sun SY. Modulation of death receptors by cancer therapeutic agents.
Cancer Biol Ther
2007
;
7
:
163
–73.
42
Henson ES, Johnston JB, Gibson SB. The role of TRAIL death receptors in the treatment of hematological malignancies.
Leuk Lymphoma
2008
;
49
:
27
–35.
43
Dorothee G, Vergnon I, Menez J, et al. Tumor-infiltrating CD4+ T lymphocytes express APO2 ligand (APO2L)/TRAIL upon specific stimulation with autologous lung carcinoma cells: role of IFN-α on APO2L/TRAIL expression and -mediated cytotoxicity.
J Immunol
2002
;
169
:
809
–17.
44
Yamaguchi Y, Miyahara E, Ohshita A, et al. Locoregional immunotherapy of malignant effusion from colorectal cancer using the streptococcal preparation OK-432 plus interleukin-2: induction of autologous tumor-reactive CD4+ Th1 killer lymphocytes.
Br J Cancer
2003
;
89
:
1876
–84.
45
Zennadi R, Abdel-Wahab Z, Seigler H, Darrow T. Generation of melanoma-specific, cytotoxic CD4+ T helper 2 cells: requirement of both HLA-DR15 and Fas antigens on melanomas for their lysis by Th2 cells.
Cell Immunol
2001
;
210
:
96
–105.
46
Porakishvili N, Roschupkina T, Kalber T, et al. Expansion of CD4+ T cells with a cytotoxic phenotype in patients with B-chronic lymphocytic leukaemia (B-CLL).
Clin Exp Immunol
2001
;
126
:
29
–36.
47
Thiele D, Lipsky P. Tumor cell lysis by T cells distinct from NK cells and alloantigen-specific cytotoxic T cells.
Clin Immunol Immunopathol
1988
;
49
:
405
–23.
48
VanLith M, Kohlgraf K, Sivinski C, Tempero R, Hollingsworth M. MUC1-specific anti-tumor responses: molecular requirements for CD4-mediated responses.
Int Immunol
2002
;
14
:
873
–82.
49
Effros RB. From Hayflick to Walford: the role of T cell replicative senescence in human aging.
Exp Gerontol
2004
;
39
:
885
–90.
50
Pawelec G, Akbar A, Caruso C, Effros R, Grubeck-Loebenstein B, Wikby A. Is immunosenescence infectious?
Trends Immunol
2004
;
25
:
406
–10.
51
Bild AH, Mendoza FJ, Gibson EM, et al. MEKK1-induced apoptosis requires TRAIL death receptor activation and is inhibited by AKT/PKB through inhibition of MEKK1 cleavage.
Oncogene
2002
;
21
:
6649
–56.
52
Marini P, Schmid A, Jendrossek V, et al. Irradiation specifically sensitises solid tumour cell lines to TRAIL mediated apoptosis.
BMC Cancer
2005
;
5
:
5
.
53
Wieser M, Stadler G, Jennings P, et al. hTERT alone immortalizes epithelial cells of renal proximal tubules without changing their functional characteristic.
Am J Physiol Renal Physiol
2008
;
295
:
1365
–75.
54
Voglauer R, Grillari J, Fortschegger K, et al. Establishment of human fibroma cell lines from a MEN1 patient by introduction of either hTERT or SV40 early region.
Int J Oncol
2005
;
26
:
961
–70.
55
Secchiero P, Gonelli A, Carnevale E, et al. TRAIL promotes the survival and proliferation of primary human vascular endothelial cells by activating the Akt and ERK pathways.
Circulation
2003
;
107
:
2250
–6.
56
Iacomino G, Medici MC, Russo GL. Valproic acid sensitizes K562 erythroleukemia cells to TRAIL/Apo2L-induced apoptosis.
Anticancer Res
2008
;
28
:
855
–64.
57
Hietakangas V, Poukkula M, Heiskanen KM, Karvinen JT, Sistonen L, Eriksson JE. Erythroid differentiation sensitizes K562 leukemia cells to TRAIL-induced apoptosis by down-regulation of c-FLIP.
Mol Cell Biol
2003
;
23
:
1278
–91.
58
Jia L, Patwari Y, Kelsey SM, Newland AC. Trail-induced apoptosis in type I leukemic cells is not enhanced by overexpression of bax.
Biochem Biophys Res Commun
2001
;
283
:
1037
–45.
59
Wang R, Zhang L, Zhang X, et al. Differential regulation of the expression of CD95 ligand, receptor activator of nuclear factor-κB ligand (RANKL), TNF-related apoptosis-inducing ligand (TRAIL), and TNF-α during T cell activation.
J Immunol
2001
;
166
:
1983
–90.
60
Wendling U, Walczak H, Dorr J, et al. Expression of TRAIL receptors in human autoreactive and foreign antigen-specific T cells.
Cell Death Differ
2000
;
7
:
637
–44.
61
Park S, Billiar T, Seol D. IFN-γ inhibition of TRAIL-induced IAP-2 up-regulation, a possible mechanism of IFN-γ-enhanced TRAIL-induced apoptosis.
Biochem Biophys Res Commun
2002
;
291
:
233
–6.
62
Wang S, Boonman Z, Li H, et al. Role of TRAIL and IFN-γ in CD4+ T cell-dependent tumor rejection in the anterior chamber of the eye.
J Immunol
2003
;
171
:
2789
–96.
63
LaSalle J, Hafler D. The Coexpression of CD45RA and CD45RO Isoforms on T Cells during the S/G2/M stages of Cell Cycle.
Cell Immunol
1991
;
138
:
197
–206.
64
Pages F, Berger A, Camus M, et al. Effector memory T cells, early metastasis, and survival in colorectal cancer.
N Engl J Med
2005
;
353
:
2654
–66.
65
Morimoto C, Schlossman S. P. Rambotti Lecture. Human naive and memory T cells revisited: new markers (CD31 and CD27) that help define CD4+ T cell subsets.
Clin Exp Rheumatol
1993
;
11
:
241
–7.
66
Hamann D, Baars P, Hooibrink B, van Lier R. Heterogeneity of the human CD4+ T-cell population: two distinct CD4+ T-cell subsets characterized by coexpression of CD45RA and CD45RO isoforms.
Blood
1996
;
88
:
3513
–21.
67
Baecher-Allan C, Wolf E, Hafler D. Functional analysis of highly defined, FACS-isolated populations of human regulatory CD4+CD25+ T cells.
Clin Immunol
2005
;
115
:
10
–8.
68
Koenen H, Fasse E, Joosten I. IL-15 and cognate antigen successfully expand de novo-induced human antigen-specific regulatory CD4+ T cells that require antigen-specific activation for suppression.
J Immunol
2003
;
171
:
6431
–41.
69
Grossman W, Verbsky J, Barchet W, Colonna M, Atkinson J, Ley T. Human T regulatory cells can use the perforin pathway to cause autologous target cell death.
Immunity
2004
;
21
:
589
–601.
70
Wang J, Ioan-Facsinay A, van der Voort EI, Huizinga TW, Toes RE. Transient expression of FOXP3 in human activated nonregulatory CD4+ T cells.
Eur J Immunol
2007
;
37
:
129
–38.
71
Godfrey DI, MacDonald HR, Kronenberg M, Smyth MJ, Van Kaer L. NKT cells: what's in a name?
Nat Rev Immunol
2004
;
4
:
231
–7.
72
Trinchieri G. Interleukin-10 production by effector T cells: Th1 cells show self control.
J Exp Med
2007
;
204
:
239
–43.
73
Amyes E, Hatton C, Montamat-Sicotte D, et al. Characterization of the CD4+ T cell response to Epstein-Barr Virus during primary and persistent infection.
J Exp Med
2003
;
198
:
903
–11.
74
Appay V, Dunbar P, Callan M, et al. Memory CD8+ T cells vary in differentiation phenotype in different persistent virus infections.
Nat Med
2002
;
8
:
379
–85.
75
Walczak H, Bouchon A, Stahl H, Krammer PH. Tumor necrosis factor-related apoptosis-inducing ligand retains its apoptosis-inducing capacity on Bcl-2- or Bcl-xL-overexpressing chemotherapy-resistant tumor cells.
Cancer Res
2000
;
60
:
3051
–7.
76
Cretney E, Takeda K, Smyth MJ. Cancer: novel therapeutic strategies that exploit the TNF-related apoptosis-inducing ligand (TRAIL)/TRAIL receptor pathway.
Int J Biochem Cell Biol
2007
;
39
:
280
–6.
77
Stadler G, Wieser M, Streubel B, et al. Low telomerase activity: Possible role in the progression of human medullary thyroid carcinoma.
Eur J Cancer
2008
;
44
:
866
–75.
78
Chang MW, Grillari J, Mayrhofer C, et al. Comparison of early passage, senescent and hTERT immortalized endothelial cells.
Exp Cell Res
2005
;
309
:
121
–36.
79
Wieser M, Stadler G, Böhm E, et al. Nuclear flow fish: a novel technique for telomere dynamics.
Exp Gerontol
2006
;
41
:
230
–5.
80
Kataoka T, Shinohara N, Takayama H, et al. Concanamycin A, a powerful tool for characterization and estimation of contribution of perforin- and Fas-based lytic pathways in cell-mediated cytotoxicity.
J Immunol
1996
;
156
:
3678
–86.
81
EECRegulatoryDocument. Committee for Proprietary Medicinal Products: Ad Hoc Working Party on Biotechnology/Pharmacy and Working Party on Safety Medicines. Note for guidance. Validation of virus removal and inactivation procedures.
Biologicals
1991
;
19
:
247
–51.
82
Fischer K, Andreesen R, Mackensen A. An improved flow cytometric assay for the determination of cytotoxic T lymphocyte activity.
J Immunol Methods
2002
;
259
:
159
–69.