Naive and memory CD8+ T cells can undergo programmed activation and expansion in response to a short T-cell receptor stimulus, but the extent to which in vitro programming can qualitatively substitute for an in vivo antigen stimulation remains unknown. We show that self-/tumor-reactive effector memory CD8+ T cells (TEM) programmed in vitro either with peptide-pulsed antigen-presenting cells or plate-bound anti-CD3/anti-CD28 embark on a highly stereotyped response of in vivo clonal expansion and tumor destruction nearly identical to that of vaccine-stimulated TEM cells. This programmed response was associated with an interval of antigen-independent interferon-γ (IFN-γ) release that facilitated the dynamic expression of the major histocompatibility complex class I restriction element H-2Db on responding tumor cells, leading to recognition and subsequent tumor lysis. Delaying cell transfer for more than 24 hours after stimulation or infusion of cells deficient in IFN-γ entirely abrogated the benefit of the programmed response, whereas transfer of cells unable to respond to IFN-γ had no detriment to antitumor immunity. These findings extend the phenomenon of a programmable effector response to memory CD8+ T cells and have major implications for the design of current adoptive-cell transfer trials.

Adoptive cell transfer (ACT) represents a promising approach for the treatment of patients with relapsed hematologic malignancies, posttransplantation viral diseases, retroviral-induced immunodeficiency, and advanced solid cancers.1–6  In mice and humans, antigen (Ag) restimulation with vaccination after ACT significantly potentiates the in vivo antitumor efficacy of adoptively transferred self-/tumor-reactive CD8+ T cells.7–12  Efficient translation of this approach to human trials has been limited by a number of technical challenges. For example, tumor-infiltrating lymphocyte (TIL) cultures often have specificity against multiple epitopes rather than a single, defined Ag.13  In addition, TILs may contain reactivity against uncharacterized, autologous tumor-specific Ags rather than well-defined tumor-associated Ags,13–15  making the prospect of coadministering an Ag-specific vaccine impractical. Finally, nearly all T cells transferred in current human protocols contain a T-effector cell (TEFF) or T-cell effector memory (TEM) phenotypic population characterized by low or absent expression of the lymphoid trafficking molecules CD62L and CCR7.16,17  In preclinical studies, transfer of CD62LhiCCR7+ cells with a naive, central memory (TCM), or stem cell memory CD8+ T cell (TSCM) phenotype provide for superior antitumor treatment compared with TEM or TEFF cells when combined with in vivo tumor Ag vaccination.18–23  Obtaining an Ag-specific population of CD62LhiCCR7+ T cells for use in human trials has thus far proven elusive.3,24 

One potential approach to overcome these limitations could be the programming of CD8+ T cells in vitro before ACT.25  Experiments in mice have shown that application of a brief T-cell receptor (TCR) stimulus to naive CD8+ T cells is sufficient to initiate a highly stereotyped response leading to clonal proliferation,26–31  acquisition of effector functions,27–31  and ultimately formation of a memory population.27,30,31  Initiation of a programmed response with an in vitro stimulus immediately before cell infusion has the theoretical benefit of minimizing in vitro differentiation before ACT. Furthermore, a nonspecific stimulus might be used to stimulate cells with diverse or unknown specificities, such as TIL. Lastly, because programmed cells do not require further activation cues to execute an effector response, it may be possible to bypass the previous requirement to obtain TCM- or TSCM-like cells to achieve optimal antitumor treatment when coadministering a vaccine.18,23 

We sought to determine whether programming tumor-reactive CD8+ TEM cells in vitro either with peptide-pulsed Ag-presenting cells (APCs) or combined anti-CD3/anti-CD28 stimulation before ACT could substitute for in vivo vaccination using a model based on the transfer of TCR transgenic pmel-1 CD8+ T cells reactive against the shared self/tumor Ag gp100.7  Our results demonstrate that provision of a brief (< 24 hours) programming stimulus to TEM in vitro instructs cells to undergo a process of clonal expansion and acquisition of effector functions leading to the destruction of large, established tumors with near-equivalent efficacy as TEM cells restimulated in vivo with vaccination. The programmed response was dependent on an interval of Ag-independent interferon-γ (IFN-γ) release that facilitated dynamic changes in tumor surface expression of the major histocompatibility complex class I molecule H-2Db, subsequent tumor recognition, and ultimately cytolysis.

Mice and tumor cells

Pmel-1 TCR transgenic mice7  were crossed to B6.129S7-Ifngtm1Ts/J32  and B6.129S7-Ifngr1tm1Agt/J33  mice (all from The Jackson Laboratory) to derive pmel-Ifngtm1Ts/J and pmel-Ifngr1tm1Agt/J mice (henceforth described as pmel-Ifng−/− and pmel-Ifngr1−/−, respectively). Female C57BL/6 mice (The Jackson Laboratory) were used at 6 to 12 weeks of age as recipient mice in ACT experiments. All animal experiments were conducted with the approval of the National Cancer Institute Animal Use and Care Committee. B16 (H-2b), a gp100+ spontaneous murine melanoma, was obtained from the National Cancer Institute tumor repository and maintained in complete media (CM) as described previously.7  The gp100 cell lines MCA-205 and EL-4 (National Cancer Institute Tumor Repository) were also maintained in CM and used as irrelevant H-2Db targets.

In vitro programming and adoptive cell transfer

TEM cells differentiated in vitro were produced as described elsewhere.18,34–36  In vitro programming was accomplished in the following manner. In all experiments, day 7 pmel TEM cells were frozen. For TEM cells programmed with peptide stimulation, cells were thawed on days −1, −3, −5, and −7 before ACT, rested overnight in CM containing 5 ng/mL recombinant human interleukin-2 (rhIL-2; Chiron), and finally restimulated with 30 Gy irradiated C57BL/6 splenocytes pulsed with 1 μM human gp10025-33 (hgp10025-33) peptide at a ratio of pmel/irradiated-splenocytes of 1:10. All cell cultures were started with 106 pmel TEM cells per recipient mouse. The total number of cells after in vitro restimulation were adoptively transferred. For TEM cells programmed in vitro with anti-CD3 and anti-CD28 monoclonal antibodies, 24-well plates were coated with 1 μg/mL anti-CD3ϵ (145-2C11) and 0.5 μg/mL anti-CD28 (37.51; both monoclonal antibodies from BD Biosciences) diluted in 1 mL sterile phosphate-buffered saline at 4°C overnight. The plates were washed with CM, and 2 × 106 thawed pmel TEM cells were added to each well. Cells were cultured overnight at 37°C in rhIL-2–containing CM and then adoptively transferred.

Ex vivo coculture assays

Where indicated, B16 was pretreated overnight with titrated doses of IFN-γ (PeproTech) before use in coculture, cytolytic, or fluorescence-activated cell sorter (FACS) analysis experiments. To assess dynamic changes in H-2Db expression on B16 in response to soluble factors produced by pmel TEM, a 24-well Transwell plate system (Corning) was used. A total of 105 B16 cells/well were plated in the lower chamber of the Transwell plate and 105 pmel or pmel-Ifng−/− TEM cells with or without restimulation with 106 irradiated C57BL/6 splenocytes pulsed with 1 μM hgp10025-33 peptide were plated in the upper chamber. B16 tumor cells were collected 48 hours later and evaluated for H-2Db expression by FACS. 51Cr-release assays were performed as described in detail elsewhere,22  except that the coculture was performed over 8 hours. Assessment of IFN-γ release by in vitro programmed pmel TEM cells was accomplished by restimulating pmel TEM with hgp10025-33 peptide-pulsed irradiated splenocytes on day 0. Cells were harvested on days 1 through 6 after restimulation, isolated with Ficoll solution (GE Healthcare), and then plated at a 1:1 ratio of 106 target-to-effector cells in an overnight coculture against indicated targets. A previously described H2-Db–restricted epitope (βgal96-103) was used as a negative control.37  Supernatants were harvested and assessed for IFN-γ release as described elsewhere.7 

In vivo tumor treatment and enumeration of adoptively transferred cells

Before ACT, all tumor-bearing recipient mice received sublethal irradiation (500 cGy) on the day of treatment. Cells prepared as described above in “In vitro programming and adoptive cell transfer” were adoptively transferred. All treated mice received rhIL-2 administered by an intraperitoneal injection twice daily at 36 μg per dose for a total of 6 doses. Where indicated, mice also received vaccination with a recombinant fowlpox virus expressing hgp100 (rFPhgp100).7  Enumeration of adoptively transferred cells was performed as described elsewhere.19,38  Experiments were performed in a blinded, randomized fashion and performed independently at least twice with similar results.

FACS analysis

B16 was stained with H-2Db–FITC (KH95; BD Biosciences) as described.7  Samples were analyzed using a FACSCalibur flow cytometer and analyzed using FlowJo software (TreeStar).

Statistics

The products of the perpendicular diameters for tumors were plotted as the mean plus or minus SEM. Tumor graphs and tissue partitioning were compared using the Wilcoxon rank-sum test. Differences in the absolute number of pmel cells after ACT and IFN-γ production were compared using a nonpaired t test. The correlation between Db expression and cytolysis was calculated using log-regression statistics after the data were projected on a semilog plot.

In vitro programming of TEM obviates the requirement for in vivo tumor-Ag vaccination

We have previously shown in the pmel-1 model that optimal treatment of large, established B16 melanoma requires the tripartite regimen of adoptively transferred antitumor CD8+ T cells, exogenous γc signaling cytokine support (eg, IL-2, IL-7, IL-15, or IL-21), and either in vivo vaccination or myeloablation with hematopoetic stem cell rescue.7,9,22,39–41  To test the hypothesis that provision of a brief (< 24 hours) programming stimulus in vitro could replace the in vivo requirement for tumor-Ag vaccination, we transferred pmel TEM cells restimulated with hgp10025-33 peptide-pulsed irradiated APCs for 24 hours before ACT. Pmel TEM cells that did not undergo in vitro programming were transferred alone or in combination with rFPhgp100 as a negative and positive control for Ag restimulation, respectively. All treated mice received exogenous IL-2. To provide for an adequate treatment window, we intentionally transferred 106 pmel TEM cells, a dose previously established to be noncurative in our model.18 

The combination of nonrestimulated pmel TEM cells and IL-2 alone caused only a modest delay in tumor growth compared with untreated controls (Figure 1A; P < .01). By contrast, provision of pmel TEM cells programmed in vitro with peptide before transfer significantly potentiated the antitumor response compared with nonrestimulated cells (P < .03). Notably, the benefit provided by in vitro programming was comparable with results obtained with the addition of rFPhgp100 vaccination to unprogrammed TEM cells (P = .6).

Figure 1

In vitro programming of TEM cells obviates the requirement for in vivo vaccination. (A-B) Sublethally irradiated WT mice bearing day 10 established subcutaneous B16 tumors were left untreated as controls (×) or received in vitro differentiated pmel TEM cells (○), pmel TEM cells plus rFPgp100 vaccination (■), or pmel TEM cells programmed in vitro with either hgp10025-33 peptide-pulsed APCs (▲) or plate-bound anti-CD3/CD28 (♦) for 24 hours before transfer. For all treatment groups receiving cell transfer, mice also received exogenous rhIL-2 (36 μg/dose × 6 doses). Results for tumor treatment experiments are presented as the mean measurements from 5 mice per group (± SEM) and are representative of 3 independent experiments per programming condition. (C) In vitro programming of TEM cells causes cells to execute a proliferative response in vivo similar to that of vaccine-stimulated TEM cells. Sublethally irradiated WT mice received nonrestimulated pmel TEM cells (○), pmel TEM cells programmed in vitro with plate-bound anti-CD3/anti-CD28 for 24 hours before cell transfer (♦), or pmel TEM cells plus rFPgp100 vaccination (■). All treated mice received exogenous rhIL-2. The percentage of adoptively transferred pmel-1 cells (identified as CD8+Vβ13+ lymphocytes/lymphocyte gate) were enumerated in the spleens of treated animals as a function of time. Each data point represents the average ± SEM of 3 independent mice per treatment group. This experiment was repeated 3 times with similar results.

Figure 1

In vitro programming of TEM cells obviates the requirement for in vivo vaccination. (A-B) Sublethally irradiated WT mice bearing day 10 established subcutaneous B16 tumors were left untreated as controls (×) or received in vitro differentiated pmel TEM cells (○), pmel TEM cells plus rFPgp100 vaccination (■), or pmel TEM cells programmed in vitro with either hgp10025-33 peptide-pulsed APCs (▲) or plate-bound anti-CD3/CD28 (♦) for 24 hours before transfer. For all treatment groups receiving cell transfer, mice also received exogenous rhIL-2 (36 μg/dose × 6 doses). Results for tumor treatment experiments are presented as the mean measurements from 5 mice per group (± SEM) and are representative of 3 independent experiments per programming condition. (C) In vitro programming of TEM cells causes cells to execute a proliferative response in vivo similar to that of vaccine-stimulated TEM cells. Sublethally irradiated WT mice received nonrestimulated pmel TEM cells (○), pmel TEM cells programmed in vitro with plate-bound anti-CD3/anti-CD28 for 24 hours before cell transfer (♦), or pmel TEM cells plus rFPgp100 vaccination (■). All treated mice received exogenous rhIL-2. The percentage of adoptively transferred pmel-1 cells (identified as CD8+Vβ13+ lymphocytes/lymphocyte gate) were enumerated in the spleens of treated animals as a function of time. Each data point represents the average ± SEM of 3 independent mice per treatment group. This experiment was repeated 3 times with similar results.

Close modal

To test whether an Ag-nonspecific programming stimulus was also sufficient to commit tumor-reactive cells to an effective antitumor program, we stimulated pmel TEM cells with plate-bound anti-CD3/anti-CD28 for 24 hours before ACT. Again, we observed that in vitro programming of pmel TEM cells could significantly augment their in vivo antitumor activity compared with nonrestimulated controls (Figure 1B; P < .05). Further, there was no significant difference in tumor treatment between mice that received anti-CD3/anti-CD28 programmed TEM cells and the positive control of TEM cells and rFPhgp100 vaccination (P > .9).

Finally, because the expansion of responding tumor-reactive CD8+ T cells in vivo has previously been correlated with antitumor treatment efficacy in both mice and humans,17–19,38,40,42  we sought to evaluate the in vivo proliferative response of adoptively transferred in vitro–programmed CD8+ T cells compared with unprogrammed cells with or without in vivo rFPhgp100 vaccination. For groups that received pmel TEM cells programmed with anti-CD3/anti-CD28 before transfer or pmel TEM cells restimulated in vivo with rFPhgp100, we observed a massive expansion of self-/tumor-reactive CD8+ T cells in both the spleen (Figure 1C) and lymph nodes (supplemental Figure 1A, available on the Blood website; see the Supplemental Materials link at the top of the online article) of recipient mice peaking on days 5 and 6 after transfer. By contrast, pmel TEM cells that did not receive either an in vitro or in vivo Ag restimulation did not expand to any measurable extent in either tissue after adoptive transfer. A similar pattern of in vivo proliferation was observed when cells were programmed with hgp10025-33 peptide-pulsed irradiated APCs (data not shown). A comparison of the relative partitioning of transferred cells between the spleens and lymph nodes of recipient animals revealed no significant differences in the lymph node trafficking behavior of programmed compared with vaccine-stimulated pmel TEM cells (supplemental Figure 1B). Depending on the individual experiment, there were relatively more T cells in either the in vitro programmed or the in vivo restimulated pmel TEM groups; however, the overall kinetic pattern remained very similar across 3 independently performed experiments and matched previously reported results.9,18,19,38 

Taken together, these data demonstrated that provision of a 24-hour in vitro programming stimulus to tumor-reactive CD8+ TEM cells in either an Ag-specific (cognate peptide) or nonspecific (anti-CD3/CD-28) manner was sufficient to incite a program of in vivo clonal expansion and acquisition of effector functions leading to tumor destruction nearly equivalent to in vivo restimulation with vaccination.

Delayed transfer of programmed TEM cells impairs in vivo antitumor function

Naive CD8+ T cells require as little as 2 to 3 hours of TCR-based instruction to commit to a program of Ag-independent proliferation and acquisition of effector functions.26,28  However, the optimal programming duration to provide for maximal proliferation, IFN-γ release, and cytolytic function has been reported to range from 20 hours to 48 hours or longer.29,31  Further, analogous experiments have yet to be conducted using memory phenotype CD8+ T cells in the context of a secondary recall response. We therefore sought to determine the optimal timing between initiation of a programming stimulus to pmel TEM cells and ACT using tumor treatment efficacy as a read-out.

Pmel TEM cells were in vitro programmed with hgp10025-33 peptide-pulsed irradiated APCs for 12 hours, 24 hours, 3 days, 5 days, and 7 days before ACT. In each group, cultures were initiated with 106 pmel TEM cells per recipient mouse. To control for a loss of replicative potential of cells that had undergone division in vitro, we transferred the total yield of cells obtained at the conclusion of the programming stimulus. Provision of a brief (12-24 hours) programming stimulus to pmel TEM cells significantly augmented their in vivo antitumor activity compared with nonprogrammed TEM cells (Figure 2A; P < .01 for both groups). Furthermore, there were no significant differences between pmel TEM cells peptide-programmed for 12 hours and 24 hours before transfer and the positive control of pmel TEM cells restimulated in vivo with rFPgp100 vaccination (P > .3 for all comparisons). In contrast, there was a complete loss of antitumor treatment efficacy compared with nontreated controls for TEM cells transferred after restimulation on day 3 or later (data not shown; P > .05 for all comparisons).

Figure 2

Duration of in vitro programming stimulus is a critical determinant of in vivo antitumor efficacy. (A) Sublethally irradiated WT mice bearing day 10 established subcutaneous B16 tumors were left untreated as controls (×) or received pmel TEM cells (●), pmel TEM cells programmed in vitro with 1 μM hgp10025-33 peptide-pulsed irradiated APCs for indicated time points before transfer (■), or pmel TEM cells plus rFPgp100 vaccination (□). For groups receiving pmel TEM cells, mice received 106 cells. For mice receiving peptide-programmed pmel TEM cells, cultures were started with 106 cells, and the total number of cells present at the time of transfer was divided evenly between the numbers of recipient mice and transferred. For all treatment groups receiving cell transfer, mice also received exogenous rhIL-2 (36 μg/dose × 6 doses). Results for tumor treatment experiments are presented as the mean measurements from 5 mice per group (± SEM). (B) Increased duration of in vitro programming does not impair the relative efficiency of engraftment after adoptive cell transfer. pmel TEM cells were transferred without further stimulation (■) or received programming with 1 μM hgp10025-33 peptide-pulsed irradiated APCs for 24 hours (▩) or 72 hours (□) before ACT. For all groups, cultures were started with 107 pmel TEM cells, and the total number of cells present at the time of transfer were divided evenly between recipient mice and transferred. The absolute number of pmel cells (identified by CD8+tetramer+ lymphocytes) was enumerated in the spleen of recipient mice 48 hours after transfer as outlined in “In vivo tumor treatment and enumeration of adoptively transferred cells.” Each data point represents the average ± SEM of 3 independent mice per treatment group. This experiment was repeated twice with similar results. *P < .01 for the comparisons of nonstimulated pmel TEM cells and TEM cells programmed for 24 hours to TEM cells programmed for 72 hours before transfer. †P > .2 for the comparison of nonstimulated pmel TEM cells to TEM cells programmed for 24 hours before transfer.

Figure 2

Duration of in vitro programming stimulus is a critical determinant of in vivo antitumor efficacy. (A) Sublethally irradiated WT mice bearing day 10 established subcutaneous B16 tumors were left untreated as controls (×) or received pmel TEM cells (●), pmel TEM cells programmed in vitro with 1 μM hgp10025-33 peptide-pulsed irradiated APCs for indicated time points before transfer (■), or pmel TEM cells plus rFPgp100 vaccination (□). For groups receiving pmel TEM cells, mice received 106 cells. For mice receiving peptide-programmed pmel TEM cells, cultures were started with 106 cells, and the total number of cells present at the time of transfer was divided evenly between the numbers of recipient mice and transferred. For all treatment groups receiving cell transfer, mice also received exogenous rhIL-2 (36 μg/dose × 6 doses). Results for tumor treatment experiments are presented as the mean measurements from 5 mice per group (± SEM). (B) Increased duration of in vitro programming does not impair the relative efficiency of engraftment after adoptive cell transfer. pmel TEM cells were transferred without further stimulation (■) or received programming with 1 μM hgp10025-33 peptide-pulsed irradiated APCs for 24 hours (▩) or 72 hours (□) before ACT. For all groups, cultures were started with 107 pmel TEM cells, and the total number of cells present at the time of transfer were divided evenly between recipient mice and transferred. The absolute number of pmel cells (identified by CD8+tetramer+ lymphocytes) was enumerated in the spleen of recipient mice 48 hours after transfer as outlined in “In vivo tumor treatment and enumeration of adoptively transferred cells.” Each data point represents the average ± SEM of 3 independent mice per treatment group. This experiment was repeated twice with similar results. *P < .01 for the comparisons of nonstimulated pmel TEM cells and TEM cells programmed for 24 hours to TEM cells programmed for 72 hours before transfer. †P > .2 for the comparison of nonstimulated pmel TEM cells to TEM cells programmed for 24 hours before transfer.

Close modal

We next evaluated the persistence of nonstimulated pmel TEM cells compared with cells that had undergone in vitro programming with peptide-pulsed irradiated APCs for either 24 hours or 72 hours before transfer to exclude the possibility that a relative detriment in engraftment efficiency was responsible for differences in tumor treatment. There was no decline in the absolute number of gp100-specific CD8+ T cells in mice that received programmed cells compared with mice that received unprogrammed TEM cells (Figure 2B). Indeed, a significantly greater number of pmel CD8+ T cells were retrieved from mice that received cells programmed for 72 hours before transfer (P < .01), correlating with a higher yield of cells in vitro after the programming step. By contrast, there was not a significant difference in the absolute number of pmel CD8+ T cells between mice that received nonstimulated TEM cells and cells programmed for 24 hours (P > .2). Collectively, these data suggested that a critical determinant of the programmed antitumor response was lost or exhausted between 24 hours and 72 hours after stimulation.

In vitro programming initiates a temporal window of Ag-independent IFN-γ release that facilitates tumor recognition

We next sought to determine what functional changes occurred in programmed pmel TEM cells 72 hours after restimulation were responsible for their loss of in vivo antitumor activity. IFN-γ production by adoptively transferred CD8+ T cells has recently been shown to be an essential mediator in the recognition and subsequent destruction of self-tissues that express gp100, such as the eye.43  Further, spontaneous IFN-γ release has been documented at early time points in naive CD8+ T cells executing an Ag-independent programmed response.17  We therefore evaluated the release of IFN-γ by tumor-reactive TEM cells as a function of time after the initiation of a programming stimulus.

Peptide-programmed pmel TEM cells were assessed for their ability to release IFN-γ in response to either B16 or hgp10025-33–pulsed target cells on days 1 through 6 after restimulation. As a specificity control, we assessed IFN-γ release in response to the gp100 tumor line EL-4 pulsed with βgal96-103, an irrelevant Db-restricted peptide. Immediately before in vitro programming, TEM cells released IFN-γ only in response to cognate peptide and, to a more limited extent, to B16 (data not shown). In contrast, pmel TEM cells released IFN-γ in an Ag-independent manner after receiving the programming stimulus (Figure 3A). This Ag-independent release of IFN-γ lasted for as long as 5 days after restimulation until ultimately fading to specificity by day 6 after restimulation such that only Ag-bearing targets (either hgp10025-33 peptide-pulsed targets or B16) were recognized with significant IFN-γ release.

Figure 3

In vitro programming of TEM cells causes an interval of Ag-independent IFN-γ release that facilitates dynamic changes in Db expression on tumor. (A) pmel TEM cells were restimulated with hgp10025-33 peptide-pulsed irradiated APCs for indicated time points, isolated using lympholyte solution, and then used in overnight coculture assays against targets, including CM (□), EL-4 pulsed with 1 μM of either βgal96-103 (○) or hgp10025-33 (●) peptides, and B16 melanoma (■). Data are mean ± SD. (B) Up-regulation of Db expression on B16 melanoma in response to titrated doses of IFN-γ. B16 melanoma cells were incubated with IFN-γ at doses representing 5-fold serial dilutions between 3375 and 5.4 ng/mL. After 48 hours, cells were harvested and evaluated by FACS analysis for Db expression. Mean fluorescence intensity (MFI) after gating on live cells versus dose of IFN-γ pretreatment is displayed. Results from 1 of 3 representative experiments are shown. (C) IFN-γ production by pmel TEM cells causes dynamic changes in the surface expression of Db on B16 melanoma in a contact-independent manner. B16 was plated at the bottom of a 24-well Transwell plate with CM only (…), and either pmel TEM (■) or pmel-Ifng1−/− TEM (▩) cells programmed for 24 hours with hgp10025-33 peptide before being plated in the top wells. After 24 hours, B16 from the bottom well was harvested and evaluated for Db expression by FACS analysis after gating for live cells. Results of a representative experiment are shown.

Figure 3

In vitro programming of TEM cells causes an interval of Ag-independent IFN-γ release that facilitates dynamic changes in Db expression on tumor. (A) pmel TEM cells were restimulated with hgp10025-33 peptide-pulsed irradiated APCs for indicated time points, isolated using lympholyte solution, and then used in overnight coculture assays against targets, including CM (□), EL-4 pulsed with 1 μM of either βgal96-103 (○) or hgp10025-33 (●) peptides, and B16 melanoma (■). Data are mean ± SD. (B) Up-regulation of Db expression on B16 melanoma in response to titrated doses of IFN-γ. B16 melanoma cells were incubated with IFN-γ at doses representing 5-fold serial dilutions between 3375 and 5.4 ng/mL. After 48 hours, cells were harvested and evaluated by FACS analysis for Db expression. Mean fluorescence intensity (MFI) after gating on live cells versus dose of IFN-γ pretreatment is displayed. Results from 1 of 3 representative experiments are shown. (C) IFN-γ production by pmel TEM cells causes dynamic changes in the surface expression of Db on B16 melanoma in a contact-independent manner. B16 was plated at the bottom of a 24-well Transwell plate with CM only (…), and either pmel TEM (■) or pmel-Ifng1−/− TEM (▩) cells programmed for 24 hours with hgp10025-33 peptide before being plated in the top wells. After 24 hours, B16 from the bottom well was harvested and evaluated for Db expression by FACS analysis after gating for live cells. Results of a representative experiment are shown.

Close modal

As previously reported by others,44,45  unmanipulated B16 spontaneously expressed very low levels of the class I restriction element, Db (Figure 3B-C). However, as has also been documented earlier, Db could be dynamically up-regulated in response to titrated doses of IFN-γ (Figure 3B).44,45  We therefore assessed whether Db on B16 could be modulated through IFN-γ produced by in vitro programmed pmel TEM cells. To evaluate the effect of soluble mediators, we used a Transwell system to physically isolate programmed TEM cells from B16. Pmel TEM cells were left unmanipulated or underwent in vitro programming with peptide. As a control, we used pmel TEM cells genetically deficient in their ability to release IFN-γ (pmel-Ifng−/−). After overnight coculture, B16 was isolated and assessed for surface expression of Db by FACS analysis. Db expression on B16 was significantly up-regulated after coculture with in vitro programmed WT but not IFN-γ–deficient TEM cells (Figure 3C). An intermediate amount of Db was expressed after coculture with nonrestimulated pmel TEM cells (data not shown). These data indicated that provision of a programming stimulus to pmel TEM cells incited an interval of Ag-independent IFN-γ release that provided for the dynamic up-regulation of Db on the surface of B16 melanoma in a contact-independent manner.

Production but not consumption of IFN-γ by programmed tumor-reactive T cells is essential for execution of the antitumor response

Based on the observation that IFN-γ can cause a marked up-regulation of Db on B16 melanoma, we next sought to determine whether these changes facilitated the recognition and ultimately destruction of tumor by pmel TEM cells both in vitro and in vivo. First, we assessed the differential ability of nonprogrammed pmel TEM cells to recognize B16 either with or without pretreatment with IFN-γ. An IFN-γ dose of 675 ng/mL was chosen because it was associated with the plateau portion of the dose-response curve for IFN-γ pretreatment and Db expression on B16 in previous titration experiments (Figure 3B). In an overnight coculture, the recognition of B16 by pmel TEM cells was significantly augmented by previous incubation with IFN-γ (Figure 4A; P < .001). Next, to test the hypothesis that increased Db expression on B16 would lead to enhanced cytolysis by pmel TEM cells, we pretreated B16 with titrated doses of IFN-γ for 48 hours before initiation of a cytolytic assay. We observed a direct correlation on a semilog plot between Db expression on B16 tumor cells and the efficiency of cytolysis (Figure 4B; R2 = 0.95, P < .001).

Figure 4

IFN-γ induces dynamic changes in Db expression on B16 melanoma leading to enhanced recognition and cytolysis. (A) IFN-γ pretreatment facilitates recognition of B16 melanoma by Pmel TEM cells. Pmel TEM cells were cocultured overnight with B16 ± pretreatment with IFN-γ (675 ng/mL). Data from a representative experiment are shown as mean IFN-γ release ± SEM. *P < .001. (B) Db expression on B16 correlates with efficiency of cytolysis by pmel TEM cells on a semilog plot. B16 cells were pretreated for 48 hours with titrated doses of IFN-γ and then used as targets in an overnight cytolytic assay with pmel TEM cells. Data from a representative experiment are shown as the mean percentage cytolysis versus Db (MFI) on B16 used as target cells. Log regression analysis was performed to determine the best line of fit.

Figure 4

IFN-γ induces dynamic changes in Db expression on B16 melanoma leading to enhanced recognition and cytolysis. (A) IFN-γ pretreatment facilitates recognition of B16 melanoma by Pmel TEM cells. Pmel TEM cells were cocultured overnight with B16 ± pretreatment with IFN-γ (675 ng/mL). Data from a representative experiment are shown as mean IFN-γ release ± SEM. *P < .001. (B) Db expression on B16 correlates with efficiency of cytolysis by pmel TEM cells on a semilog plot. B16 cells were pretreated for 48 hours with titrated doses of IFN-γ and then used as targets in an overnight cytolytic assay with pmel TEM cells. Data from a representative experiment are shown as the mean percentage cytolysis versus Db (MFI) on B16 used as target cells. Log regression analysis was performed to determine the best line of fit.

Close modal

Given the requirement for IFN-γ to facilitate tumor recognition by pmel cells in vitro, we next tested whether the augmentation of in vivo antitumor function of in vitro programmed TEM cells was the result of the release of IFN-γ. We transferred either WT or IFN-γ–deficient pmel TEM cells programmed in vitro with plate-bound anti-CD3/anti-CD28 24 hours before transfer and compared their relative in vivo antitumor efficacy. As previously shown, adoptive transfer of in vitro–programmed pmel TEM cells resulted in enhanced tumor destruction compared with unprogrammed pmel TEM cells (Figure 5). The benefit of in vitro programming was lost by transfer of pmel-Ifng−/− TEM cells such that there were no significant differences between programmed pmel-Ifng−/− cells and nonprogrammed WT pmel TEM cells (P = .35).

Figure 5

The enhanced in vivo antitumor efficacy of in vitro programmed TEM cells is dependent on IFN-γ production but independent of T-cell responsiveness to IFN-γ. WT mice bearing 10 days established B16 melanoma were sublethally irradiated and left untreated as controls (×) or received unprogrammed pmel TEM cells (●), or pmel TEM (□) cells, pmel-Ifng−/− TEM cells (■), or pmel-Ifngr1−/− TEM cells (♦) programmed in vitro with anti-CD3/anti-CD28 for 24 hours before transfer. For mice receiving programmed pmel TEM, pmel-Ifng−/− TEM cells, or pmel-Ifngr1−/− TEM cells, cultures were started with 2 × 106 cells per recipient mouse, and the total number of cells present at 24 hours were transferred. Mice treated with nonprogrammed pmel TEM cells received 2 × 106 cells. All treated mice received rhIL-2 (36 μg/dose × 6 doses). Results from different representative experiments are presented as the mean measurements from 5 mice per group (± SEM).

Figure 5

The enhanced in vivo antitumor efficacy of in vitro programmed TEM cells is dependent on IFN-γ production but independent of T-cell responsiveness to IFN-γ. WT mice bearing 10 days established B16 melanoma were sublethally irradiated and left untreated as controls (×) or received unprogrammed pmel TEM cells (●), or pmel TEM (□) cells, pmel-Ifng−/− TEM cells (■), or pmel-Ifngr1−/− TEM cells (♦) programmed in vitro with anti-CD3/anti-CD28 for 24 hours before transfer. For mice receiving programmed pmel TEM, pmel-Ifng−/− TEM cells, or pmel-Ifngr1−/− TEM cells, cultures were started with 2 × 106 cells per recipient mouse, and the total number of cells present at 24 hours were transferred. Mice treated with nonprogrammed pmel TEM cells received 2 × 106 cells. All treated mice received rhIL-2 (36 μg/dose × 6 doses). Results from different representative experiments are presented as the mean measurements from 5 mice per group (± SEM).

Close modal

Finally, we sought to establish whether IFN-γ is acting only on tumor cells or whether it may also be activating responding CD8+ T cells in an autocrine or paracrine manner. We therefore transferred programmed pmel TEM cells genetically deficient in the IFN-γ receptor (pmel-Ifngr1−/−) and compared their antitumor treatment efficacy to programmed WT pmel TEM cells. Transfer of either IFN-γ receptor–deficient or competent TEM cells resulted in significantly delayed tumor growth compared with untreated controls (Figure 5; P < .01). There was no significant difference in tumor treatment between the 2 treatment groups (P > .1), demonstrating that the effect of IFN-γ was independent of the ability of adoptively transferred CD8+ cells to perceive IFN-γ. Collectively, these data indicated that production but not consumption of IFN-γ was a critical determinant of the in vivo antitumor efficacy of in vitro–programmed TEM cells.

We found that provision of a brief programming stimulus to CD8+ TEM cells led to clonal expansion and acquisition of effector function in a manner analogous to results seen previously with programmed naive CD8+ T cells.26–30  Similar results were obtained whether an Ag-specific stimulus with cognate peptide or an Ag-nonspecific stimulus with plate-bound anti-CD3/anti-CD28 were used. Thus, Ag-experienced CD8+ T cells, like naive cells,28,29  were capable of executing a highly stereotyped effector response with as little as 12 hours of stimulation. Although work from the Schoenberger laboratory has demonstrated that programming of naive CD8+ T cells can significantly potentiate their in vivo effector function after adoptive transfer in a model of T cell–mediated autoimmune diabetes,29  a direct comparison of the relative efficacy of in vitro–programmed CD8+ T cells versus in vivo–stimulated T cells had previously not been made. Our findings demonstrated that in vitro programming of Ag-experienced CD8+ T cells could replace the requirement for in vivo vaccination with near-equivalent efficacy.

Previous inquires into the duration of a programming stimulus for naive CD8+ T cells have found that anywhere between 20 hours to more than 48 hours of stimulation are required for an optimal response29,31 ; however, requirements for programming Ag-experienced cells have not been previously evaluated. We therefore sought to determine the optimal duration between in vitro programming of TEM cells and cell transfer using tumor treatment as a read-out. We found that the enhanced antitumor efficacy of in vitro–programmed CD8+ TEM cells was entirely abrogated if cells were transferred more than 72 hours after programming. Although our experiments did not determine the minimal requirements for optimal programming of Ag-experienced cells, we found that as little as 12 hours was sufficient to significantly potentiate the in vivo antitumor response.

The loss of the programming benefit when cells were transferred more than 72 hours after stimulation was probably not the result of an impairment in proliferative potential as we transferred all cells generated in vitro after the programming stimulus. Indeed, there were a significantly greater number of adoptively transferred cells present in the spleens of mice that received TEM cells programmed for 72 hours before transfer compared with mice receiving cells programmed for 24 hours or nonprogrammed TEM cells. We found instead that a functional alteration in IFN-γ secretion was a critical determinant of the programmed antitumor response. During early time points after programming, T cells underwent an interval of Ag-independent IFN-γ release capable of inducing dynamic changes in the surface expression of the major histocompatibility complex class I restriction element Db on B16 melanoma and subsequent tumor recognition and destruction. Similar to results from Rivoltini et al using human melanoma cell lines, we observed a significant correlation between the magnitude of class I expression on tumor cells and the efficiency of cytolysis by antigen-specific T cells.46  The functional significance of IFN-γ was further confirmed by experiments using programmed CD8+ T cells genetically deficient in IFN-γ in which the antitumor benefit of programming was entirely abrogated.

These findings parallel recent results demonstrating that IFN-γ is a critical determinant in the reactivity of adoptively transferred cells against nontransformed self-tissues, such as the eye, that also express the melanocyte differentiation Ag gp100.43  Further, these data also correlate with a recently completed clinical trial from the National Cancer Institute Surgery Branch where detectable serum levels of IFN-γ could be found immediately after adoptive transfer of peripheral blood mononuclear cells transduced with TCRs against either MART-1 or gp100.47  Approximately 30% of the patients in this trial who received T cells transduced with a high-affinity receptor against the MART-1 melanoma Ag48  obtained an objective clinical response consistent with standardized oncologic criteria,49  suggesting a possible relationship between serum levels of IFN-γ and the potency of T cell–mediated antitumor treatment efficacy.

Based on these and other results in our preclinical animal model43  as well as recent clinical experience using TCR gene–transduced peripheral lymphocytes reactive against the shared melanoma/melanocyte Ags gp100 and MART-1,47  we propose a model of “immunologic sonar” in which programmed adoptively transferred cells initially release IFN-γ in an Ag-independent manner as an immunologic “ping” to facilitate target recognition. By nonspecifically releasing IFN-γ, T cells can up-regulate target Ag presentation to facilitate an “immunologic echo” or return signal. When transferred cells permeate tissues expressing cognate antigen, a virtuous cycle of positive feedback may be established in which Ag-specific T cells recognize their cognate Ag, leading to further IFN-γ release, greater target recognition, and ultimately target killing in a second phase characterized by specificity. We are currently working to confirm this model using dynamic in vivo imaging with 2-photon microscopy.

In vitro expansion techniques using beads coated with anti-CD3/anti-CD28 have successfully been used to generate large numbers of autologous and allogenic T cells for the treatment of HIV,50  refractory non-Hodgkin lymphoma,51  relapsed hematologic malignancies after stem cell transplantation,52  and other hematologic malignancies.53–55  However, accumulating evidence has demonstrated that prolonged in vitro expansion of T cells can cause a progressive loss of replicative capacity and functional exhaustion limiting their in vivo antitumor efficacy.19,56  To limit the detrimental impact of prolonged in vitro culture, we used a time-constrained exposure to anti-CD3/anti-CD28 to deliver an instructional program of expansion and acquisition of effector functions for T cells to execute in vivo after transfer. This approach not only provided for enhanced in vivo antitumor function but has the practical advantage of limiting the cost and labor of expanding large number of T cells for infusion.

In conclusion, provision of a brief programming stimulus to CD8+ TEM cells in either an Ag-specific (peptide) or nonspecific (anti-CD3/anti-CD28) manner before ACT caused cells to execute a response of in vivo clonal expansion and tumor destruction nearly equivalent to that of vaccine-stimulated TEM cells. This response was dependent on an interval of Ag-independent IFN-γ release by programmed cells that facilitates the recognition and subsequent cytolysis of B16 tumor cells. Future studies will be focused on the translation of these findings in mice to human TIL- and TCR-transduced peripheral blood mononuclear cell cultures with the goal of improving the efficacy of current adoptive cell transfer protocols.

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 authors thank S. A. Rosenberg and C. S. Hinrichs for critical review of the manuscript and T. Fojo for providing the precious resource of time to complete this project.

This work was supported in part by the intramural program of the National Cancer Institute, National Institutes of Health (Bethesda, MD) and the Howard Hughes Medical Institute (C.A.K.).

This study was done in partial fulfillment of a PhD in Biochemistry (to D.C.P.) at The George Washington University, Washington, DC.

National Institutes of Health

Contribution: C.A.K., Z.Y., and N.P.R. designed research; C.A.K., Z.Y., L.N.H., L.G., and D.C.P. performed research and collected data; C.A.K., Z.Y., and N.P.R. analyzed and interpreted data; and C.A.K., L.G., and N.P.R. wrote the manuscript.

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

Correspondence: Christopher A. Klebanoff or Nicholas P. Restifo, National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892; e-mail: klebanoc@mail.nih.gov or restifo@nih.gov.

1
Leen
 
AM
Rooney
 
CM
Foster
 
AE
Improving T cell therapy for cancer.
Annu Rev Immunol
2007
, vol. 
25
 (pg. 
243
-
265
)
2
June
 
CH
Adoptive T cell therapy for cancer in the clinic.
J Clin Invest
2007
, vol. 
117
 
6
(pg. 
1466
-
1476
)
3
Gattinoni
 
L
Powell
 
DJ
Rosenberg
 
SA
Restifo
 
NP
Adoptive immunotherapy for cancer: building on success.
Nat Rev Immunol
2006
, vol. 
6
 
5
(pg. 
383
-
393
)
4
Kennedy-Nasser
 
AA
Brenner
 
MK
T-cell therapy after hematopoietic stem cell transplantation.
Curr Opin Hematol
2007
, vol. 
14
 
6
(pg. 
616
-
624
)
5
Levine
 
BL
T lymphocyte engineering ex vivo for cancer and infectious disease.
Expert Opin Biol Ther
2008
, vol. 
8
 
4
(pg. 
475
-
489
)
6
Caligiuri
 
MA
Velardi
 
A
Scheinberg
 
DA
Borrello
 
IM
Immunotherapeutic approaches for hematologic malignancies.
Hematology Am Soc Hematol Educ Program
2004
(pg. 
337
-
353
)
7
Overwijk
 
WW
Theoret
 
MR
Finkelstein
 
SE
, et al. 
Tumor regression and autoimmunity after reversal of a functionally tolerant state of self-reactive CD8+ T cells.
J Exp Med
2003
, vol. 
198
 
4
(pg. 
569
-
580
)
8
Lou
 
Y
Wang
 
G
Lizee
 
G
, et al. 
Dendritic cells strongly boost the antitumor activity of adoptively transferred T cells in vivo.
Cancer Res
2004
, vol. 
64
 
18
(pg. 
6783
-
6790
)
9
Palmer
 
DC
Balasubramaniam
 
S
Hanada
 
K
, et al. 
Vaccine-stimulated, adoptively transferred CD8+ T cells traffic indiscriminately and ubiquitously while mediating specific tumor destruction.
J Immunol
2004
, vol. 
173
 
12
(pg. 
7209
-
7216
)
10
Wang
 
LX
Li
 
R
Yang
 
G
, et al. 
Interleukin-7-dependent expansion and persistence of melanoma-specific T cells in lymphodepleted mice lead to tumor regression and editing.
Cancer Res
2005
, vol. 
65
 
22
(pg. 
10569
-
10577
)
11
Smith
 
F
Klapper
 
J
Wunderlich
 
J
Rosenberg
 
S
Dudley
 
M
Impact of a recombinant fowlpox vaccine on the efficacy of adoptive cell therapy with tumor infiltrating lymphocytes in a patient with metastatic melanoma.
J Immunother
 
In press
12
Sikora
 
AG
Jaffarzad
 
N
Hailemichael
 
Y
, et al. 
IFN-alpha enhances peptide vaccine-induced CD8+ T cell numbers, effector function, and antitumor activity.
J Immunol
2009
, vol. 
182
 
12
(pg. 
7398
-
7407
)
13
Dudley
 
ME
Wunderlich
 
JR
Yang
 
JC
, et al. 
Adoptive cell transfer therapy following non-myeloablative but lymphodepleting chemotherapy for the treatment of patients with refractory metastatic melanoma.
J Clin Oncol
2005
, vol. 
23
 
10
(pg. 
2346
-
2357
)
14
Dudley
 
ME
Yang
 
JC
Sherry
 
R
, et al. 
Adoptive cell therapy for patients with metastatic melanoma: evaluation of intensive myeloablative chemoradiation preparative regimens.
J Clin Oncol
2008
, vol. 
26
 
32
(pg. 
5233
-
5239
)
15
Kessler
 
JH
Melief
 
CJ
Identification of T-cell epitopes for cancer immunotherapy.
Leukemia
2007
, vol. 
21
 
9
(pg. 
1859
-
1874
)
16
Powell
 
DJ
Dudley
 
ME
Robbins
 
PF
Rosenberg
 
SA
Transition of late-stage effector T cells to CD27+ CD28+ tumor-reactive effector memory T cells in humans after adoptive cell transfer therapy.
Blood
2005
, vol. 
105
 
1
(pg. 
241
-
250
)
17
Pule
 
MA
Savoldo
 
B
Myers
 
GD
, et al. 
Virus-specific T cells engineered to coexpress tumor-specific receptors: persistence and antitumor activity in individuals with neuroblastoma.
Nat Med
2008
, vol. 
14
 
11
(pg. 
1264
-
1270
)
18
Klebanoff
 
CA
Gattinoni
 
L
Torabi-Parizi
 
P
, et al. 
Central memory self-/tumor-reactive CD8+ T cells confer superior antitumor immunity compared with effector memory T cells.
Proc Natl Acad Sci U S A
2005
, vol. 
102
 
27
(pg. 
9571
-
9576
)
19
Gattinoni
 
L
Klebanoff
 
CA
Palmer
 
DC
, et al. 
Acquisition of full effector function in vitro paradoxically impairs the in vivo antitumor efficacy of adoptively transferred CD8+ T cells.
J Clin Invest
2005
, vol. 
115
 
6
(pg. 
1616
-
1626
)
20
Hinrichs
 
CS
Spolski
 
R
Paulos
 
CM
, et al. 
IL-2 and IL-21 confer opposing differentiation programs to CD8+ T cells for adoptive immunotherapy.
Blood
2008
, vol. 
111
 
11
(pg. 
5326
-
5333
)
21
Mueller
 
K
Schweier
 
O
Pircher
 
H
Efficacy of IL-2- vs IL-15-stimulated CD8 T cells in adoptive immunotherapy.
Eur J Immunol
2008
, vol. 
38
 
10
(pg. 
2874
-
2885
)
22
Klebanoff
 
CA
Finkelstein
 
SE
Surman
 
DR
, et al. 
IL-15 enhances the in vivo antitumor activity of tumor-reactive CD8+ T cells.
Proc Natl Acad Sci U S A
2004
, vol. 
101
 
7
(pg. 
1969
-
1974
)
23
Gattinoni
 
L
Zhong
 
XS
Palmer
 
DC
, et al. 
Wnt-signaling arrests effector T cell differentiation and generates CD8(+) memory stem cells.
Nat Med
2009
, vol. 
15
 
7
(pg. 
808
-
813
)
24
Klebanoff
 
CA
Gattinoni
 
L
Restifo
 
NP
CD8+ T-cell memory in tumor immunology and immunotherapy.
Immunol Rev
2006
, vol. 
211
 (pg. 
214
-
224
)
25
Hinrichs
 
CS
Gattinoni
 
L
Restifo
 
NP
Programming CD8+ T cells for effective immunotherapy.
Curr Opin Immunol
2006
, vol. 
18
 
3
(pg. 
363
-
370
)
26
Wong
 
P
Pamer
 
EG
Cutting edge: antigen-independent CD8 T cell proliferation.
J Immunol
2001
, vol. 
166
 
10
(pg. 
5864
-
5868
)
27
Kaech
 
SM
Ahmed
 
R
Memory CD8+ T cell differentiation: initial antigen encounter triggers a developmental program in naive cells.
Nat Immunol
2001
, vol. 
2
 
5
(pg. 
415
-
422
)
28
van Stipdonk
 
MJ
Lemmens
 
EE
Schoenberger
 
SP
Naive CTLs require a single brief period of antigenic stimulation for clonal expansion and differentiation.
Nat Immunol
2001
, vol. 
2
 
5
(pg. 
423
-
429
)
29
van Stipdonk
 
MJ
Hardenberg
 
G
Bijker
 
MS
, et al. 
Dynamic programming of CD8+ T lymphocyte responses.
Nat Immunol
2003
, vol. 
4
 
4
(pg. 
361
-
365
)
30
Mercado
 
R
Vijh
 
S
Allen
 
SE
Kerksiek
 
K
Pilip
 
IM
Pamer
 
EG
Early programming of T cell populations responding to bacterial infection.
J Immunol
2000
, vol. 
165
 
12
(pg. 
6833
-
6839
)
31
Prlic
 
M
Hernandez-Hoyos
 
G
Bevan
 
MJ
Duration of the initial TCR stimulus controls the magnitude but not functionality of the CD8+ T cell response.
J Exp Med
2006
, vol. 
203
 
9
(pg. 
2135
-
2143
)
32
Dalton
 
DK
Pitts-Meek
 
S
Keshav
 
S
Figari
 
IS
Bradley
 
A
Stewart
 
TA
Multiple defects of immune cell function in mice with disrupted interferon-gamma genes.
Science
1993
, vol. 
259
 
5102
(pg. 
1739
-
1742
)
33
Huang
 
S
Hendriks
 
W
Althage
 
A
, et al. 
Immune response in mice that lack the interferon-gamma receptor.
Science
1993
, vol. 
259
 
5102
(pg. 
1742
-
1745
)
34
Weninger
 
W
Crowley
 
MA
Manjunath
 
N
von Andrian
 
UH
Migratory properties of naive, effector, and memory CD8(+) T cells.
J Exp Med
2001
, vol. 
194
 
7
(pg. 
953
-
966
)
35
Manjunath
 
N
Shankar
 
P
Wan
 
J
, et al. 
Effector differentiation is not prerequisite for generation of memory cytotoxic T lymphocytes.
J Clin Invest
2001
, vol. 
108
 
6
(pg. 
871
-
878
)
36
Belz
 
GT
Bedoui
 
S
Kupresanin
 
F
Carbone
 
FR
Heath
 
WR
Minimal activation of memory CD8+ T cell by tissue-derived dendritic cells favors the stimulation of naive CD8+ T cells.
Nat Immunol
2007
, vol. 
8
 
10
(pg. 
1060
-
1066
)
37
Overwijk
 
WW
Surman
 
DR
Tsung
 
K
Restifo
 
NP
Identification of a Kb-restricted CTL epitope of beta-galactosidase: potential use in development of immunization protocols for “self” antigens.
Methods
1997
, vol. 
12
 
2
(pg. 
117
-
123
)
38
Hwang
 
LN
Yu
 
Z
Palmer
 
DC
Restifo
 
NP
The in vivo expansion rate of properly stimulated transferred CD8+ T cells exceeds that of an aggressively growing mouse tumor.
Cancer Res
2006
, vol. 
66
 
2
(pg. 
1132
-
1138
)
39
Zeng
 
R
Spolski
 
R
Finkelstein
 
SE
, et al. 
Synergy of IL-21 and IL-15 in regulating CD8+ T cell expansion and function.
J Exp Med
2005
, vol. 
201
 
1
(pg. 
139
-
148
)
40
Wrzesinski
 
C
Paulos
 
CM
Gattinoni
 
L
, et al. 
Hematopoietic stem cells promote the expansion and function of adoptively transferred antitumor CD8 T cells.
J Clin Invest
2007
, vol. 
117
 
2
(pg. 
492
-
501
)
41
Gattinoni
 
L
Finkelstein
 
SE
Klebanoff
 
CA
, et al. 
Removal of homeostatic cytokine sinks by lymphodepletion enhances the efficacy of adoptively transferred tumor-specific CD8+ T cells.
J Exp Med
2005
, vol. 
202
 
7
(pg. 
907
-
912
)
42
Dudley
 
ME
Wunderlich
 
JR
Robbins
 
PF
, et al. 
Cancer regression and autoimmunity in patients after clonal repopulation with antitumor lymphocytes.
Science
2002
, vol. 
298
 
5594
(pg. 
850
-
854
)
43
Palmer
 
DC
Chan
 
CC
Gattinoni
 
L
, et al. 
Effective tumor treatment targeting a melanoma/melanocyte-associated antigen triggers severe ocular autoimmunity.
Proc Natl Acad Sci U S A
2008
, vol. 
105
 
23
(pg. 
8061
-
8066
)
44
Lollini
 
PL
Colombo
 
MP
De
 
GC
, et al. 
In vivo reexpression of H-2 antigens in B16 melanoma cells.
Exp Clin Immunogenet
1985
, vol. 
2
 
1
(pg. 
14
-
23
)
45
Bohm
 
W
Thoma
 
S
Leithauser
 
F
Moller
 
P
Schirmbeck
 
R
Reimann
 
J
T cell-mediated, IFN-gamma-facilitated rejection of murine B16 melanomas.
J Immunol
1998
, vol. 
161
 
2
(pg. 
897
-
908
)
46
Rivoltini
 
L
Barracchini
 
KC
Viggiano
 
V
, et al. 
Quantitative correlation between HLA class I allele expression and recognition of melanoma cells by antigen-specific cytotoxic T lymphocytes.
Cancer Res
1995
, vol. 
55
 
14
(pg. 
3149
-
3157
)
47
Johnson
 
LA
Morgan
 
RA
Dudley
 
ME
, et al. 
Gene therapy with human and mouse T cell receptors mediates cancer regression and targets normal tissues expressing cognate antigen.
Blood
2009
, vol. 
114
 
3
(pg. 
535
-
546
)
48
Johnson
 
LA
Heemskerk
 
B
Powell
 
DJ
, et al. 
Gene transfer of tumor-reactive TCR confers both high avidity and tumor reactivity to nonreactive peripheral blood mononuclear cells and tumor-infiltrating lymphocytes.
J Immunol
2006
, vol. 
177
 
9
(pg. 
6548
-
6559
)
49
Therasse
 
P
Arbuck
 
SG
Eisenhauer
 
EA
, et al. 
New guidelines to evaluate the response to treatment in solid tumors: European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada.
J Natl Cancer Inst
2000
, vol. 
92
 
3
(pg. 
205
-
216
)
50
Levine
 
BL
Bernstein
 
WB
Aronson
 
NE
, et al. 
Adoptive transfer of costimulated CD4+ T cells induces expansion of peripheral T cells and decreased CCR5 expression in HIV infection.
Nat Med
2002
, vol. 
8
 
1
(pg. 
47
-
53
)
51
Laport
 
GG
Levine
 
BL
Stadtmauer
 
EA
, et al. 
Adoptive transfer of costimulated T cells induces lymphocytosis in patients with relapsed/refractory non-Hodgkin lymphoma following CD34+-selected hematopoietic cell transplantation.
Blood
2003
, vol. 
102
 
6
(pg. 
2004
-
2013
)
52
Porter
 
DL
Levine
 
BL
Bunin
 
N
, et al. 
A phase 1 trial of donor lymphocyte infusions expanded and activated ex vivo via CD3/CD28 costimulation.
Blood
2006
, vol. 
107
 
4
(pg. 
1325
-
1331
)
53
Rapoport
 
AP
Levine
 
BL
Badros
 
A
, et al. 
Molecular remission of CML after autotransplantation followed by adoptive transfer of costimulated autologous T cells.
Bone Marrow Transplant
2004
, vol. 
33
 
1
(pg. 
53
-
60
)
54
Rapoport
 
AP
Stadtmauer
 
EA
Aqui
 
N
, et al. 
Restoration of immunity in lymphopenic individuals with cancer by vaccination and adoptive T-cell transfer.
Nat Med
2005
, vol. 
11
 
11
(pg. 
1230
-
1237
)
55
Rapoport
 
AP
Stadtmauer
 
EA
Aqui
 
N
, et al. 
Rapid immune recovery and graft-vs-host disease-like engraftment-syndrome following adoptive transfer of costimulated autologous T cells.
Clin Cancer Res
2009
, vol. 
15
 
13
(pg. 
4499
-
4507
)
56
Zhou
 
J
Shen
 
X
Huang
 
J
Hodes
 
RJ
Rosenberg
 
SA
Robbins
 
PF
Telomere length of transferred lymphocytes correlates with in vivo persistence and tumor regression in melanoma patients receiving cell transfer therapy.
J Immunol
2005
, vol. 
175
 
10
(pg. 
7046
-
7052
)

Author notes

*C.A.K. and Z.Y. contributed equally to this study.

Supplemental data

Sign in via your Institution