Despite the promising efficacy of adoptive cell therapies (ACT) in melanoma, complete response rates remain relatively low and outcomes in other cancers are less impressive. The immunosuppressive nature of the tumor microenvironment and the expression of immune-inhibitory ligands, such as PD-L1/CD274 by the tumor and stroma are considered key factors limiting efficacy. The addition of checkpoint inhibitors (CPI) to ACT protocols bypasses some mechanisms of immunosuppression, but associated toxicities remain a significant concern. To overcome PD-L1–mediated immunosuppression and reduce CPI-associated toxicities, we used TALEN technology to render tumor-reactive T cells resistant to PD-1 signaling. Here, we demonstrate that inactivation of the PD-1 gene in melanoma-reactive CD8+ T cells and in fibrosarcoma-reactive polyclonal T cells enhanced the persistence of PD-1 gene-modified T cells at the tumor site and increased tumor control. These results illustrate the feasibility and potency of approaches incorporating advanced gene-editing technologies into ACT protocols to silence immune checkpoints as a strategy to overcome locally active immune escape pathways. Cancer Res; 76(8); 2087–93. ©2016 AACR.

Adoptive cell therapy (ACT) using autologous tumor-infiltrating lymphocytes (TIL) or T cells redirected to the tumor by chimeric antigen receptors (CAR) can mediate substantial regression of human cancers (1). Although multiple early-phase studies using ACT have demonstrated durable responses that correlate with persistence of the transferred T cells (2, 3), their in vivo activity and potency can be thwarted by the complex immunosuppressive nature of the tumor microenvironment (4). Combination protocols incorporating checkpoint inhibitors (CPI) such as anti–CTLA-4 or anti–PD-L1/PD-1 are currently being considered (NCT02408861; NCT02210117; NCT02374242; NCT02453620), but the potential exacerbation of the toxicities associated with CPIs may limit their implementation. Advanced genetic engineering of the ACT product offers potential solution to the toxicity issues by specifically targeting tumor-reactive T cells rather than inducing systemic checkpoint blockade. The protocols used for the generation of ACT are compatible with genetic manipulation of cells before transfer, which can be used to increase the proliferative capacity of lymphocytes (5), prolong their in vivo persistence (6), improve tumor infiltration (7), and bypass tumor immune suppression.

Programmed death-1 (PD-1, CD279) is one of the key coinhibitory immune receptors expressed by activated T cells. Initially described on a T-cell hybridoma undergoing cell death (8), PD-1 is a critical regulator of T-cell responses and essential to the maintenance of peripheral self-tolerance (9). In mouse and human cancers, high levels of PD-1 are found on TILs, although its ligand (PD-L1) is upregulated by tumor and stromal cells in response to inflammatory cytokines (e.g., IFNγ; refs. 10, 11). The PDL-1–PD-1 axis plays a major role in tumor growth and immune escape through inhibition of T-cell proliferation, effector function (12), and of T-cell survival by promotion of apoptosis (13). The administration of blocking anti–PD-L1 or PDL-1 antibodies in vivo results in enhanced CD8+ T cell responses and decreases tumor burden in mouse models of cancer (14, 15). In humans, expression of PD-1 by TILs is associated with impaired effector function and/or poor outcome in several tumor types (16, 17). Direct evidence supporting the importance of this pathway in modulating antitumor immunity comes from recent clinical trials in melanoma and non–small cell lung cancer, where anti–PD-1 antibodies have improved both progression-free and overall survival (18, 19). Of relevance, systemic immunotherapy with CPIs can also induce immune-related adverse events (irAE). Specific ablation of negative regulators of T-cell function on tumor-reactive T cells used for ACT offers the potential to exploit the antitumor activity while reducing systemic toxicity.

Efficient and rapid inactivation of genes in primary antigen-specific T cells is achievable using the transient expression of transcription activator-like effector nucleases (TALEN) mRNA, which offers several safety advantages for clinical applications (20). Using two different mouse models of cancer, we defined the impact of TALEN-mediated PD-1 gene inactivation in adoptively transferred tumor-reactive CD4+ and CD8+ T cells. Our data support a model in which PD-1 gene inactivation increases persistence of T cells at the tumor site, enhancing tumor control of B16 melanoma by gp-100-reactive CD8+ T cells and mediating complete rejection of established MCA205 fibrosarcoma by endogenous CD4+ and CD8+ T cells. This work demonstrates both the feasibility and the potential potency of strategies incorporating immune checkpoint editing of tumor-reactive T cells for use in the context of adoptive T-cell therapies for cancer.

Mice and cell lines

Six- to 8-week-old pmel-1/SJL TCR-transgenic, C57BL/6, and C57BL/6-SJL mice were purchased from Charles River Laboratories. All animal use was in accordance with the Home Office guidelines. The B16.BL6 melanoma cell line was obtained from Prof. James Allison (MD Anderson Cancer Center, Houston, TX), the MCA205 fibrosarcoma cell line was provided by Prof. Guido Kroemer (Institut Gustave Roussy, Villejuif, South Paris, France), EL4 thymoma cells were provided by Prof. Henning Walczak (UCL Cancer Institute, London, United Kingdom), and authentication was performed by the ATCC using short tandem repeat DNA profiles. The cells were grown in RPMI-1640 supplemented with 10% FCS, l-glutamine, penicillin/streptomycin (Sigma-Aldrich).

TALEN construction and validation

Three pairs of TALEN targeting the PD-1 murine gene were produced by Cellectis using the solid phase assembly method (21). Their activity was evaluated in 5 × 106 EL4 cells, cotransfected with 10 μg of in vitro transcribed mRNA (mMESSAGE mMACHINE T7 Kit; Ambion) from each half TALEN (Supplementary Fig. S1). Three days after transfection, locus-specific PCRs were performed on genomic DNA (Supplementary Table S1), mutations were assessed by mismatches digestion (T7) and Miseq analysis (20).

Cell culture and adoptive transfer

Splenocytes and lymph nodes cells from pmel-1/SJL transgenic mice were cultured in complete RPMI with gp100 peptide (Pepceuticals) and rhIL-2 (Preprotech) 100 UI/mL for 24 hours before selection of CD8+ T cells using the Dynabeads FlowComp Mouse CD8 Kit. TRL were selected from 100 to 150 mm3 tumors using Dynabeads FlowComp Mouse Pan T (CD90.2) and maintained in complete RPMI with rhIL-2 (1,000 UI/mL). A total of 5 × 106 Pmel-1 CD8+ T cells and MCA205 TRL/180 μL of BTX medium T were mixed with 20 μg of IVT mRNA, before electroporation using an Agile Pulse BTX system (Harvard Apparatus). C57BL/6 mice were subcutaneously implanted with either 2.5 × 105 B16 cells or 4 ×105 MCA205 cells (day 0). On day 6 for the pmel-1 model and on day 4 for the MCA205 model, total body irradiation (TBI) was performed (5 Gy) and 1 × 106 pmel-1 T cells or 1 × 106 MCA205 TRL were adoptively transferred into tumor-bearing mice (n = 5/group), followed by intraperitoneal (i.p.) injection of rhIL-2 (1.2 × 106 IU) for 3 days. Mice were sacrificed when the tumors exceeded 15 mm in diameter. Mice were treated with the anti-mouse CD8a 2.43–depleting antibody (BioXcell) and the anti-MCH class II M5/114 (BioXcell), intraperitoneally on day 0, 2, 4, 6, and 8 after ACT (10 mg/kg).

Flow cytometry

Tumors were weighed, digested in Liberase D (Roche) and mechanically disaggregated through 70-μm filters. Lymph nodes cells and lymphocytes from tumor samples, enriched on a Ficoll gradient were incubated with murine antibodies: anti–CD4-V500 (BD Horizon), anti–CD8-BV650, anti–PD-1-PE-Cy7 (Biolegend), anti–CD45.1-APC-Cy7, anti–FoxP3-AF700, anti–IFNγ-FITC (restimulation with gp100-pulsed dendritic cells for pmel cells or PMA/ionomycin for TRL), anti–KI67-PerCP-eFluor 710, Fixable Viability Dye eFluor 450 (eBioscience), anti-FLICA (Vybrant FAM Poly Caspases Assay Kit; ThermoFisher), Annexin-V–PE Apoptosis Detection Kit II (BD Pharmingen), Cell Sorting Set-up Beads (Life Technologies), and anti-human Granzyme B-APC (Invitrogen) in a blocking solution: 5% mouse serum, 5% rat serum (Thermo Scientific), 2% FCS, and 2% anti-FcR 2.4G2 (BioXcell). Samples were permeabilized using a Fixation/Permeabilization kit (eBioscience), acquired on a LSRII Fortessa and analyzed with FlowJo software (Tree Star).

Western blot analysis

Proteins were separated and subjected to the following antibodies: anti-TALEN (1 mg/mL), anti-GAPDH (D16H11; Cell Signaling Technology; 1:2,000), HRP-coupled secondary antibody anti-Rabbit IgG1 HRP-linked (Cell Signaling Technology).

Statistical analysis

One-way ANOVA, two-way ANOVA, or t tests with P < 0.05 were performed using Prism 6.0 software (GraphPad). Multiple comparisons were corrected with the Bonferroni coefficient and Kaplan–Meier survival curves were compared with the log-rank test.

For additional information, see Supplementary Data Section.

TALEN-mediated PD-1 gene inactivation in melanoma reactive CD8+ pmel-1 T cells increases persistence of CD8PD-1Ex2 pmel-1 cells at the tumor site

Three pairs of TALENs targeting the murine PD-1 gene were tested in EL4 cells using an optimized mRNA–electroporation protocol. Only the pair targeting the exon 2 sequence (Fig. 1A) caused detectable mismatch-identified mutagenesis (up to 27%) in the PD-1 gene, correlating with loss of PD-1 expression in up to 15% of electroporated EL4 cells (Supplementary Fig. S1). To investigate whether PD-1 inactivation of primary tumor-reactive T cells provided superior antitumor activity against the poorly immunogenic mouse melanoma B16 model, we used CD8+ T-cell receptor transgenic (TCR Tg) cells specific to the melanoma differentiation antigen gp-100 (pmel-1, Supplementary Fig. S2A; ref. 22). In vitro activated pmel-1 T cells were electroporated with control GFP mRNA (CD8wt) or PD-1–targeting TALEN mRNA (CD8PD-1Ex2), delivering a high transfection efficiency of >85% as confirmed by GFP expression (Supplementary Fig. S2B). Western blot analysis confirmed transient TALEN expression one day (d1) after transfection (Fig. 1B) and Miseq analysis of the targeted sequence showed a high frequency of mutations (53% non-homologous end joining, NHEJ; Fig. 1C). Three days after transfection, PD-1–negative cells were enriched using magnetic beads (Supplementary Fig. S2C) before ACT of 1 × 106 CD8wt or CD8PD-1Ex2 pmel-1 cells and rhIL-2 into B16 tumor-bearing mice. Significantly, enhanced tumor control was consistently observed in mice receiving CD8PD-1Ex2 cells compared with those treated with CD8wt cells (Fig. 1D). Six days after transfer in vivo, a significant enrichment of tumor-infiltrating PD-1–negative pmel-1 cells was observed in mice treated with CD8PD-1Ex2 T cells (48.2%; Fig. 1E, top), which contributed to a 2-fold increase in the total number of tumor-infiltrating pmel-1 cells (Fig. 1E, bottom). In contrast, the absolute number of T cells in draining lymph nodes (DLN) was similar between the two groups, suggesting that accumulation of CD8PD-1Ex2 T cells at the tumor site was not due to differences in engraftment nor infiltration from the DLNs (Supplementary Fig. S2D). Whereas use of RNA and electroporation to deliver TALENs is compatible with future clinical translation, it does not allow incorporation of a reporter gene to distinguish PD-1–edited cells. Nevertheless, the rate of mutagenesis, investigated ex vivo 7 days after ACT demonstrates an effective PD-1 gene editing (70%) in the PD-1–negative fraction from the CD8PD-1Ex2 group (Supplementary Fig. S2E). To investigate the mechanisms underpinning enhanced tumor control by pmel-1 cells in the CD8PD-1Ex2 group, we characterized the proliferation and effector function of CD8wt and CD8PD-1Ex2 T cells in both PD-1+ and PD-1 tumor-infiltrating pmel-1 populations. Contrary to previous work suggesting that PD-1 signaling reduces effector activity at the tumor site (23), no differences were observed in KI67, granzyme B (GZB), or IFNγ expression between PD-1+ and PD-1 cells. When we compared the adjusted percentages (percentage of cells in PD-1+ and PD-1 subpopulations) of tumor-infiltrating KI67+, GZB+, and IFNγ+ cells, we observed that the TALEN-induced PD-1 gene inactivation did not impact the proliferative or functional characteristics of pmel-1 CD8+T cells in vivo (Supplementary Fig. S2F–S2H). Instead, we consistently observed the appearance of a PD-1 pmel-1 population in the CD8PD-1Ex2 group, which contributed to an increase in the total number of KI67-, GZB-, and IFNγ-expressing pmel-1 cells in tumors from mice receiving CD8PD-1Ex2 T cells (Fig. 1F and G). Together, these data strongly suggest that, in the context of ACT, PD-1 expression primarily controls number of tumor-reactive T cells at the tumor site rather than their proliferative or functional status.

Figure 1.

TALEN-mediated PD-1 gene inactivation in pmel-1 CD8+T cells enhances the antitumor activity of ACT. A, schematic representation of murine Pdcd1 gene (chr. 1) and TALEN-targeted sequence (exon 2). B, TALEN protein expression at days 1, 2, and 3 after transfection. C, TALEN-induced mutagenesis (>50%) by Miseq analysis of CD8PD-1Ex2 pmel-1 T cells. NHEJ, non-homologous end joining. D, B16 tumor growth curve in mice receiving 1 × 106 CD8wt cells or CD8PD-1Ex2 ACT 6 days after tumor challenge. E, PD-1 expression on tumor-infiltrating CD8+ T cells and engraftment of transferred cells at the tumor site. F and G, proliferation (KI67) and functional analysis of tumor-infiltrating CD8wt cells or CD8PD-1Ex2 depicting GZB and IFNγ expressions. Each dot represents an individual mouse, and bar graphs/plots represent mean ± SD of two independent experiments (n = 10), two-way ANOVA (**, P < 0.01; ***, P < 0.001).

Figure 1.

TALEN-mediated PD-1 gene inactivation in pmel-1 CD8+T cells enhances the antitumor activity of ACT. A, schematic representation of murine Pdcd1 gene (chr. 1) and TALEN-targeted sequence (exon 2). B, TALEN protein expression at days 1, 2, and 3 after transfection. C, TALEN-induced mutagenesis (>50%) by Miseq analysis of CD8PD-1Ex2 pmel-1 T cells. NHEJ, non-homologous end joining. D, B16 tumor growth curve in mice receiving 1 × 106 CD8wt cells or CD8PD-1Ex2 ACT 6 days after tumor challenge. E, PD-1 expression on tumor-infiltrating CD8+ T cells and engraftment of transferred cells at the tumor site. F and G, proliferation (KI67) and functional analysis of tumor-infiltrating CD8wt cells or CD8PD-1Ex2 depicting GZB and IFNγ expressions. Each dot represents an individual mouse, and bar graphs/plots represent mean ± SD of two independent experiments (n = 10), two-way ANOVA (**, P < 0.01; ***, P < 0.001).

Close modal

PD-1 gene inactivation of polyclonal tumor-reactive lymphocytes promotes persistence of activated CD4+ and CD8+ at the tumor site

To define the therapeutic potential and functional impact of PD-1 gene inactivation in a more physiologically relevant T cell population, we performed PD-1 gene editing in polyclonal tumor-reactive lymphocytes (TRL) from C57BL/6SJL mice challenged with MCA205 cells (24). CD4+ and CD8+ T cells were isolated from the tumor (TILs) and tumor-draining lymph node (TDLN) via positive selection. After electroporation of the donor TRL, we observed a transfection efficiency >65% and we identified TALEN-induced PD-1 gene mutations by Miseq analysis in 26% of CD4+ T cells and almost 40% of CD8+ T cells (Supplementary Fig. S3A and S3B).

In parallel to the generation of the donor mock-transfected (TRLwt) and PD-1–edited TRL cells (TRLPD-1Ex2), we challenged a separate group of C57BL/6 mice with MCA205 tumors. Four days later, tumor-bearing mice were treated with TBI followed by ACT with TRLwt or TRLPD-1Ex2 cells and rhIL-2. Six days after transfer, analysis of PD-1 expression on TILs from mice receiving TRLwt or TRLPD-1Ex2 revealed a 15% to 20% average increase in PD-1 CD4+ and PD-1 CD8+ T cells (Fig. 2A and B). Consistent with our findings in the TCR Tg model, a significant accumulation of PD-1 cells was observed only at the tumor site (Fig. 2C; Supplementary Fig. S3C). Although PD-1 gene inactivation did not alter TRL proliferation (KI67) or function (GZB and IFNγ; Supplementary Fig. S3D–S3F), the absolute number of PD1 TRL–expressing KI67, GZB, and IFNγ was significantly higher in mice receiving TRLPD-1Ex2 (Fig. 2D–F).

Figure 2.

Functional analysis of ACT with PD-1 gene inactivation on TRL 6 days after transfer in vivo. A, flow plots depicting PD-1 expression on TRLwt or TRLPD-1-Ex2 T cells. B, the percentage of PD-1 cells showing an average of 15% to 20% increase on TRLPD-1-Ex2 compared with TRLwt T cells. Each data point represents an individual mouse. C, absolute number of PD1+ and PD1 TRLwt and TRLPD-1-Ex2 T cells per mg of tumor. D–F, representative flow plots showing KI67, GZB, and IFNγ expressions on CD4+ and CD8+ TRLwt and TRLPD-1-Ex2 T cells and absolute number of cells per mg of tumor. Bar graphs depict mean ± SD of three independent experiments (n = 15). Nonparametric t test (B), two-way ANOVA (C, E, and F) *, P < 0.05; **, P < 0.01; ***, P < 0.001.

Figure 2.

Functional analysis of ACT with PD-1 gene inactivation on TRL 6 days after transfer in vivo. A, flow plots depicting PD-1 expression on TRLwt or TRLPD-1-Ex2 T cells. B, the percentage of PD-1 cells showing an average of 15% to 20% increase on TRLPD-1-Ex2 compared with TRLwt T cells. Each data point represents an individual mouse. C, absolute number of PD1+ and PD1 TRLwt and TRLPD-1-Ex2 T cells per mg of tumor. D–F, representative flow plots showing KI67, GZB, and IFNγ expressions on CD4+ and CD8+ TRLwt and TRLPD-1-Ex2 T cells and absolute number of cells per mg of tumor. Bar graphs depict mean ± SD of three independent experiments (n = 15). Nonparametric t test (B), two-way ANOVA (C, E, and F) *, P < 0.05; **, P < 0.01; ***, P < 0.001.

Close modal

Given that the increase in the number of PD-1 cells in mice receiving TRLPD-1Ex2 cells was not associated with increased proliferation, we evaluated whether this could be explained by a reduced rate of PD-1–induced cell death. Using Annexin-V and a fluorescent caspase substrate kit (FLICA), we observed an overall reduction in the frequency of intratumoral apoptotic cells in PD-1 compared with PD-1+ CD4+ and CD8+ T cells (Fig. 3A–D). Whereas we observed no difference in the frequency of apoptotic cells between mice treated with TRLwt and TRLPD-1Ex2, a significant increase in the absolute number of Annexin-V and FLICA TRLPD-1Ex2cells per mg of tumor was reported (Fig. 3E and F). The data suggest that instead of increasing T-cell reactivity against tumors, TRLPD-1Ex2 cells are rendered resistant to PD-1–mediated cell death at the tumor site.

Figure 3.

PD-1 regulates the survival of adoptively transferred TRL in vivo. A and C, representative histograms depicting the Annexin-V/FLICA positivity in PD-1+ and PD-1 TRL 6 days after ACT. B and D, the adjusted percentage of Annexin-V+/FLICA+ cells in PD-1+ and PD-1 subsets of CD4+ and CD8+ TRLwt and TRLPD-1Ex2 T cells. Bar graphs/plots represent mean ± SD from two independent experiments (n = 10). E and F, absolute numbers of Annexin-V/FLICA CD4+ and CD8+ TRL per mg of tumor. Two-way ANOVA (B) and multiple comparison one-way ANOVA with Bonferroni correction (C) **, P < 0.01; ***, P < 0.001.

Figure 3.

PD-1 regulates the survival of adoptively transferred TRL in vivo. A and C, representative histograms depicting the Annexin-V/FLICA positivity in PD-1+ and PD-1 TRL 6 days after ACT. B and D, the adjusted percentage of Annexin-V+/FLICA+ cells in PD-1+ and PD-1 subsets of CD4+ and CD8+ TRLwt and TRLPD-1Ex2 T cells. Bar graphs/plots represent mean ± SD from two independent experiments (n = 10). E and F, absolute numbers of Annexin-V/FLICA CD4+ and CD8+ TRL per mg of tumor. Two-way ANOVA (B) and multiple comparison one-way ANOVA with Bonferroni correction (C) **, P < 0.01; ***, P < 0.001.

Close modal

PD-1 gene inactivation drives CD4- and CD8-dependent rejection of established tumors and long-term memory

PD-1 gene inactivation significantly increased the activity of our ACT protocol (P = 0.0052), and antitumor activity was dependent on both CD8+ and CD4+ T cells as administration of depleting anti-CD8 or MHC-II–blocking antibodies ablated antitumor responses (Fig. 4A and B). No evidence of tumor recurrence was observed after MCA205 rechallenge on day 50 in mice that had previously rejected tumors (data not shown). The development of long-term memory, as demonstrated by protection against a subsequent tumor challenge, is an important feature of successful T-cell–adoptive immunotherapy (25). Our data show high levels of KI67, consistent with high proliferative capacity for both TRL and TRLPD-1Ex2 cells, which also displayed a T-bethi CD62LlowCD44hi phenotype consistent with effector memory T cells (Supplementary Fig. S4A and S4B; ref. 26). Finally, ex vivo stimulation of circulating T cells from surviving mice (40 days after tumor rejection) demonstrated high levels of IFNγ production in both TRL and TRLPD-1Ex2 groups arguing against terminal differentiation (Supplementary Fig. S4C; ref. 27).

Figure 4.

Therapeutic efficacy of ACT with PD-1 gene inactivation of TRL on MCA205 fibrosarcoma. A, MCA205 growth after ACT with TRL- and CD8-depleting or MHC-II–blocking antibodies. Two-way ANOVA *, P < 0.05; **, P < 0.01. B, treatment with TRLPD-1Ex2 resulted in insignificant prolongation of survival (n = 15). Log-rank test, P = 0.0052. Depletion of CD8+ subset or MHC-II blockade abrogates the TRLPD-1Ex2 cells induced antitumor effects (n = 10).

Figure 4.

Therapeutic efficacy of ACT with PD-1 gene inactivation of TRL on MCA205 fibrosarcoma. A, MCA205 growth after ACT with TRL- and CD8-depleting or MHC-II–blocking antibodies. Two-way ANOVA *, P < 0.05; **, P < 0.01. B, treatment with TRLPD-1Ex2 resulted in insignificant prolongation of survival (n = 15). Log-rank test, P = 0.0052. Depletion of CD8+ subset or MHC-II blockade abrogates the TRLPD-1Ex2 cells induced antitumor effects (n = 10).

Close modal

In summary, our data demonstrate the feasibility and potential clinical relevance of gene-editing approaches, conferring superior in vivo activity in the context of adoptive cell therapy protocols. Of relevance, we observed no evidence of toxicity (weight and physiologic alterations) after ACT. To the best of our knowledge, this is the first proof-of-concept study illustrating enhancement and persistence of antitumor responses using targeted genome editing of primary tumor reactive T cells. Our data indicate that the primary mechanism by which PD-1 gene inactivation affects tumor-reactive T cells is by regulating their ability to survive rather than increasing their activity on a per cell basis, which aligns with the original description of the role of PD-1 signaling in T-cell apoptosis (8). Although we investigated PD-1 as the initial proof-of-concept target, the technology and approach described potentially allow the permanent disruption of other inhibitory checkpoints (one or more), considerably advancing the design of the next generation of cancer immunotherapies.

M. Pule is a CSO at Autolus, reports receiving a commercial research grant from Cellectis, has received speakers bureau honoraria from Amgen and Roche, and has ownership interest (including patents) in Autolus. No potential conflicts of interest were disclosed by the other authors.

Conception and design: L. Menger, L. Poirot, M. Pule, K.S. Peggs, S.A. Quezada

Development of methodology: L. Menger, A. Sledzinska, F. Arce Vargas, L. Poirot, M. Pule

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): L. Menger, A. Sledzinska, K. Bergerhoff, K.S. Peggs

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): L. Menger, J. Herrero, K.S. Peggs

Writing, review, and/or revision of the manuscript: L. Menger, A. Sledzinska, L. Poirot, M. Pule, J. Herrero, K.S. Peggs, S.A. Quezada

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): L. Menger, F. Arce Vargas, J. Smith

Study supervision: K.S. Peggs, S.A. Quezada

S.A. Quezada is funded by a CRUK Career Development Fellowship and a Cancer Research Institute Investigator Award. K.S. Peggs receives funding from CRUK, Leukemia and Lymphoma Research. This work was funded in part by an EU FP7 grant and undertaken at UCL Hospitals/UCL with support from the Department of Health and CRUK funding schemes for National Institute for Health Research Biomedical Research Centers and Experimental Cancer Medicine Centers. The research leading to these results has received funding from the European Union Seventh Framework Programme under grant amendment no 602239 ATECT.

1.
Rosenberg
SA
,
Restifo
NP
. 
Adoptive cell transfer as personalized immunotherapy for human cancer.
Science
2015
;
348
:
62
8
.
2.
Dudley
ME
,
Wunderlich
JR
,
Robbins
PF
,
Yang
JC
,
Hwu
P
,
Schwartzentruber
DJ
, et al
Cancer regression and autoimmunity in patients after clonal repopulation with antitumor lymphocytes.
Science
2002
;
298
:
850
4
.
3.
Johnson
LA
,
Morgan
RA
,
Dudley
ME
,
Cassard
L
,
Yang
JC
,
Hughes
MS
, et al
Gene therapy with human and mouse T-cell receptors mediates cancer regression and targets normal tissues expressing cognate antigen.
Blood
2009
;
114
:
535
46
.
4.
Gill
S
,
June
CH
. 
Going viral: chimeric antigen receptor T-cell therapy for hematological malignancies.
Immunol Rev
2015
;
263
:
68
89
.
5.
Liu
K
,
Rosenberg
SA
. 
Transduction of an IL-2 gene into human melanoma-reactive lymphocytes results in their continued growth in the absence of exogenous IL-2 and maintenance of specific antitumor activity.
J Immunol
2001
;
167
:
6356
65
.
6.
Kalbasi
A
,
Shrimali
RK
,
Chinnasamy
D
,
Rosenberg
SA
. 
Prevention of interleukin-2 withdrawal-induced apoptosis in lymphocytes retrovirally cotransduced with genes encoding an antitumor T-cell receptor and an antiapoptotic protein.
J Immunother
2010
;
33
:
672
83
.
7.
Craddock
JA
,
Lu
A
,
Bear
A
,
Pule
M
,
Brenner
MK
,
Rooney
CM
, et al
Enhanced tumor trafficking of GD2 chimeric antigen receptor T cells by expression of the chemokine receptor CCR2b.
J Immunother
2010
;
33
:
780
8
.
8.
Ishida
Y
,
Agata
Y
,
Shibahara
K
,
Honjo
T
. 
Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death.
EMBO J
1992
;
11
:
3887
95
.
9.
Nishimura
H
,
Okazaki
T
,
Tanaka
Y
,
Nakatani
K
,
Hara
M
,
Matsumori
A
, et al
Autoimmune dilated cardiomyopathy in PD-1 receptor-deficient mice.
Science
2001
;
291
:
319
22
.
10.
Gros
A
,
Robbins
PF
,
Yao
X
,
Li
YF
,
Turcotte
S
,
Tran
E
, et al
PD-1 identifies the patient-specific CD8(+) tumor-reactive repertoire infiltrating human tumors.
J Clin Invest
2014
;
124
:
2246
59
.
11.
Taube
JM
,
Anders
RA
,
Young
GD
,
Xu
H
,
Sharma
R
,
McMiller
TL
, et al
Colocalization of inflammatory response with B7-h1 expression in human melanocytic lesions supports an adaptive resistance mechanism of immune escape.
Sci Transl Med
2012
;
4
:
127ra137
.
12.
Tseng
SY
,
Otsuji
M
,
Gorski
K
,
Huang
X
,
Slansky
JE
,
Pai
SI
, et al
B7-DC, a new dendritic cell molecule with potent costimulatory properties for T cells.
J Exp Med
2001
;
193
:
839
46
.
13.
Dong
H
,
Strome
SE
,
Salomao
DR
,
Tamura
H
,
Hirano
F
,
Flies
DB
, et al
Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion.
Nat Med
2002
;
8
:
793
800
.
14.
Blank
C
,
Brown
I
,
Peterson
AC
,
Spiotto
M
,
Iwai
Y
,
Honjo
T
, et al
PD-L1/B7H-1 inhibits the effector phase of tumor rejection by T cell receptor (TCR) transgenic CD8+ T cells.
Cancer Res
2004
;
64
:
1140
5
.
15.
Barber
DL
,
Wherry
EJ
,
Masopust
D
,
Zhu
B
,
Allison
JP
,
Sharpe
AH
, et al
Restoring function in exhausted CD8 T cells during chronic viral infection.
Nature
2006
;
439
:
682
7
.
16.
Thompson
RH
,
Dong
H
,
Lohse
CM
,
Leibovich
BC
,
Blute
ML
,
Cheville
JC
, et al
PD-1 is expressed by tumor-infiltrating immune cells and is associated with poor outcome for patients with renal cell carcinoma.
Clin Cancer Res
2007
;
13
:
1757
61
.
17.
Ahmadzadeh
M
,
Johnson
LA
,
Heemskerk
B
,
Wunderlich
JR
,
Dudley
ME
,
White
DE
, et al
Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired.
Blood
2009
;
114
:
1537
44
.
18.
Robert
C
,
Schachter
J
,
Long
GV
,
Arance
A
,
Grob
JJ
,
Mortier
L
, et al
Pembrolizumab versus ipilimumab in advanced melanoma.
N Engl J Med
2015
;
372
:
2521
32
.
19.
Brahmer
J
,
Reckamp
KL
,
Baas
P
,
Crino
L
,
Eberhardt
WE
,
Poddubskaya
E
, et al
Nivolumab versus docetaxel in advanced squamous-cell non–small cell lung cancer.
N Engl J Med
2015
;
373
:
123
35
.
20.
Menger
L
,
Gouble
A
,
Marzolini
MA
,
Pachnio
A
,
Bergerhoff
K
,
Henry
JY
, et al
TALEN-mediated genetic inactivation of the glucocorticoid receptor in cytomegalovirus-specific T cells.
Blood
2015
;
126
:
2781
9
.
21.
Daboussi
F
,
Leduc
S
,
Marechal
A
,
Dubois
G
,
Guyot
V
,
Perez-Michaut
C
, et al
Genome engineering empowers the diatom Phaeodactylum tricornutum for biotechnology.
Nat Commun
2014
;
5
:
3831
.
22.
Overwijk
WW
,
Tsung
A
,
Irvine
KR
,
Parkhurst
MR
,
Goletz
TJ
,
Tsung
K
, et al
gp100/pmel 17 is a murine tumor rejection antigen: induction of self-reactive, tumoricidal T cells using high-affinity, altered peptide ligand.
J Exp Med
1998
;
188
:
277
86
.
23.
Keir
ME
,
Butte
MJ
,
Freeman
GJ
,
Sharpe
AH
. 
PD-1 and its ligands in tolerance and immunity.
Annu Rev Immunol
2008
;
26
:
677
704
.
24.
Kjaergaard
J
,
Shu
S
. 
Tumor infiltration by adoptively transferred T cells is independent of immunologic specificity but requires downregulation of L-selectin expression.
J Immunol
1999
;
163
:
751
9
.
25.
Wang
LX
,
Kjaergaard
J
,
Cohen
PA
,
Shu
S
,
Plautz
GE
. 
Memory T cells originate from adoptively transferred effectors and reconstituting host cells after sequential lymphodepletion and adoptive immunotherapy.
J Immunol
2004
;
172
:
3462
8
.
26.
Seder
RA
,
Ahmed
R
. 
Similarities and differences in CD4+ and CD8+ effector and memory T-cell generation.
Nat Immunol
2003
;
4
:
835
42
.
27.
Paley
MA
,
Kroy
DC
,
Odorizzi
PM
,
Johnnidis
JB
,
Dolfi
DV
,
Barnett
BE
, et al
Progenitor and terminal subsets of CD8+ T cells cooperate to contain chronic viral infection.
Science
2012
;
338
:
1220
5
.