Adoptive transfer of T cells genetically engineered to express a tumor-targeting chimeric antigen receptor (CAR) or T-cell receptor (TCR) can mediate cancer regression in some patients. CARs are synthetic single-chain proteins that use antibody domains to target cell surface antigens. TCRs are natural heterodimeric proteins that can target intracellular antigens through recognition of peptides bound to human leukocyte antigens. CARs have shown promise in B-cell malignancies and TCRs in melanoma, but neither approach has achieved clear success in an epithelial cancer. Treatment of epithelial cancers may be particularly challenging because of a paucity of target antigens expressed by carcinomas and not by important healthy tissues. In addition, epithelial cancers may be protected by inhibitory ligands and soluble factors in the tumor microenvironment. One strategy to overcome these negative regulators is to modulate expression of T-cell genes to enhance intrinsic T-cell function. Programmable nucleases, which can suppress inhibitory genes, and inducible gene expression systems, which can enhance stimulatory genes, are entering clinical testing. Other work is delineating whether control of genes for immune checkpoint receptors (e.g., PDCD1, CTLA4) and cytokine and TCR signaling regulators (e.g., CBLB, CISH, IL12, IL15) can increase the antitumor activity of therapeutic T cells. Clin Cancer Res; 22(7); 1559–64. ©2016 AACR.

No potential conflicts of interest were disclosed.

The following editor(s) reported relevant financial relationships: P.S. Steeg reports receiving commercial research grants from Genentech.

The members of the planning committee have no real or apparent conflicts of interest to disclose.

Upon completion of this activity, the participant should have a better understanding of antigen receptor gene therapy, the adoptive transfer of T cells genetically engineered to express a tumor-targeting chimeric antigen receptor or T-cell receptor, and the areas under investigation that can improve the function of adoptively transferred T cells.

This activity does not receive commercial support.

Antigen receptor gene therapy, the adoptive transfer of T cells genetically engineered to express a tumor-targeting chimeric antigen receptor (CAR) or T-cell receptor (TCR), is an emerging class of cancer treatments that may hold promise for wide-ranging cancers. This type of treatment generally is performed by first harvesting mononuclear cells from the peripheral blood of the patient through a leukapheresis procedure. T cells from the leukapheresis product are then transduced with a viral vector other gene transfer platform encoding a CAR or TCR. The genetically engineered T cells are expanded in the laboratory and administered to the patient intravenously. Lymphocyte-depleting chemotherapy may be given prior to cell infusion to enhance engraftment and function of the engineered T cells. T-cell infusion may be followed by systemic administration of cytokines, such as interleukin-2, to support the T cells.

CARs are synthetic molecules composed of antibody single-chain variable fragments (ScFv) that bind to a target tumor antigen, and domains from CD3 signaling chains and T-cell costimulatory receptor molecules that provide intracellular signaling (Fig. 1). Investigators from several medical centers have reported remarkable results with CARs targeting CD19 in diverse types of B-cell malignancies (1–6). TCRs are natural heterodimeric proteins composed of an α- and β-chain (Fig. 1). Antigen recognition is mediated by the complementarity determining regions of the receptor. Signaling is accomplished by endogenous CD3 molecules that associate with the TCR α- and β-chains to form the TCR complex. Certain TCR gene therapies appear to have activity in melanoma, with the most promising approach targeting the cancer germline antigen, cancer/testis antigen 1 (alternative name NY-ESO-1; ref.7).

Figure 1.

TCRs and CARs recognize tumor cells through distinct mechanisms. Genetically engineered TCRs complex with endogenous CD3 units to form structures with recognition and signaling capability. They signal with a physiologic CD3 signal, and costimulation requires engagement of a separate coreceptor. TCRs recognize a peptide–HLA complex on the cell surface. The peptide in this complex is derived from a protein that is processed by intracellular machinery. Thus, TCRs can recognize intracellular antigens, but they also require antigen processing and presentation by the tumor cell, and their antigen recognition is HLA restricted. CARs are synthetic proteins that do not exist in nature. They are composed of an antibody-derived recognition unit linked to a signaling domain from the CD3 molecule and a costimulatory domain from a costimulatory receptor. CARs recognize cell surface but not intracellular antigens. They do not require antigen processing or presentation by the tumor, and their target recognition is not HLA restricted.

Figure 1.

TCRs and CARs recognize tumor cells through distinct mechanisms. Genetically engineered TCRs complex with endogenous CD3 units to form structures with recognition and signaling capability. They signal with a physiologic CD3 signal, and costimulation requires engagement of a separate coreceptor. TCRs recognize a peptide–HLA complex on the cell surface. The peptide in this complex is derived from a protein that is processed by intracellular machinery. Thus, TCRs can recognize intracellular antigens, but they also require antigen processing and presentation by the tumor cell, and their antigen recognition is HLA restricted. CARs are synthetic proteins that do not exist in nature. They are composed of an antibody-derived recognition unit linked to a signaling domain from the CD3 molecule and a costimulatory domain from a costimulatory receptor. CARs recognize cell surface but not intracellular antigens. They do not require antigen processing or presentation by the tumor, and their target recognition is not HLA restricted.

Close modal

TCRs but not CARs can target intracellular antigens

The antigen recognition mechanisms of CARs and TCRs differ in crucial ways that affect which antigens they can target and how tumors might evade attack. CARs, through their ScFv domain, recognize membrane-bound cell surface antigens (Fig. 1). In contrast, TCRs recognize peptides that are generated by intracellular processing of proteins and presented to T cells by histocompatibility complex (MHC) molecules (Fig. 1). CAR recognition of targets is not MHC restricted; all tumors with the target antigen, regardless of human leukocyte antigen (HLA) haplotype, can be targeted. Because CARs recognize antigens that are not presented by MHC molecules, tumors cannot evade recognition through downregulation or loss of antigen processing and presentation genes such as transporter associated with antigen processing, proteasome subunits, or HLA molecules. However, CARs can bind to soluble antigens, which, if present in blood, can inhibit the intended recognition of membrane-bound molecules. In addition, most CARs have some degree of inherent ScFv clustering that can trigger tonic signaling, premature T-cell exhaustion, and loss of antitumor activity (8). In contrast, TCRs are not inhibited by soluble antigen because their targets are presented by MHC molecules, and they do not exhibit substantial tonic signaling because they are composed of physiologic TCR signaling proteins (Figure 1).

Advantages to targeting tumor-specific antigens

Antigen receptor gene therapy is based on the infusion of high numbers of T cells directed against a tumor antigen. The strength of this T-cell attack can be further augmented by host conditioning with chemotherapy that decreases negative regulatory cells and increases homeostatic cytokines (9, 10). The potency of the treatment can be additionally increased by the administration of exogenous cytokines (10). The intent of the treatment is to completely eliminate every cell expressing the target antigen with a single infusion of T cells, and in some cases this goal appears to be achieved. However, the ability of antigen receptor gene therapy to destroy target cells located throughout the body has important consequences for target antigen selection.

A review of clinical experience with various target antigens is informative. CD19 (a B-cell antigen) CAR therapy has been reported to eliminate not only malignant B cells but also normal B cells, which have broad tissue distribution (3). Melanoma antigen recognized by T cells 1 (MART1) and gp100 (melanocyte antigens) TCR therapy were shown to mediate not only melanoma regression but also sometimes-severe injury to melanocytes populating skin, eyes, and ears, which cause rash and vision and hearing loss (11). Carcinoembryonic antigen TCR therapy, which targeted an antigen expressed by both healthy colonic epithelial cells and colon cancers, was found to induce severe diarrhea that led to discontinuation of a clinical trial after three patients (12). Finally, carbonic anhydrase IX (CAIX) CAR therapy, which was directed against an antigen present on the surface of renal cell carcinomas and biliary epithelium, was found to cause cholangitis by CAR-mediated recognition of CAIX on the epithelial cells of normal bile ducts (13, 14). From these studies it is evident that antigen receptor gene therapy can induce destruction of diverse target cells regardless of their location. One implication of this finding is that, for treatments that are intended to induce complete regression of metastatic epithelial tumors, it may be important to target antigens that are expressed by tumors but not by vital healthy tissues.

Targeting tumor-specific antigens that are shared between patients

Few antigens are shared by the tumors of different patients and not expressed by healthy tissues (15). Cancer germline antigens are expressed in development but silenced in nongermline adult tissues, and reexpressed by some cancers. Successful treatment of some patients with melanoma or synovial sarcoma with TCR gene therapy targeting the germline cancer antigen NY-ESO-1 has been reported (7). Mutated gene products can also be attractive targets, especially when they are oncogenic drivers. Certain oncogenic Kirsten rat sarcoma viral oncogene homolog (KRAS) mutations occur at relatively high frequency in pancreatic, colorectal, lung, endometrial, ovarian, and prostate cancers. A TCR recognizing an HLA-A11 restricted epitope of the G12D KRAS variant has been described and may permit the first KRAS-targeted TCR gene therapy (16). Perhaps the most attractive targets for T-cell therapy in solid tumors are the viral antigens of tumors such as cervical, oropharyngeal, and anal cancer [caused by human papillomavirus (HPV)] or undifferentiated nasopharygeal carcinoma [uNPC; caused by Epstein–Barr virus (EBV)]. In a recent clinical trial, adoptively transferred tumor-infiltrating T cells from cultures selected for their recognition of HPV oncoproteins mediated durable, complete tumor regression in some patients with HPV+ cervical cancer (17). The magnitude of HPV reactivity of the infused cells correlated with objective tumor response, but “bystander” cells with non-HPV specificities were also infused and may have played a role in the clinical responses. A TCR targeting an HLA-A2-restricted epitope of HPV-16 E6 has been identified, and T cells expressing this receptor demonstrate recognition of cervical and oropharyngeal cancer cell lines (18). Results of an ongoing clinical trial of TCR gene therapy with this receptor in cervical, oropharyngeal, anal, vaginal, vulvar, and penile cancers will more directly interrogate the HPV-16 E6 oncoprotein as a therapeutic target (NCT02280811). As with HPV, EBV can drive oncogenesis, and adoptive transfer of EBV-specific T cells has been studied and clinical activity reported in hematologic malignancies (19, 20) and possibly undifferentiated nasopharyngeal carcinoma (21).

Inhibition of negative regulators of T-cell function

Effective treatment of metastatic epithelial cancers by CAR or TCR genetically engineered T cells may require enhancement of T-cell function. Enhanced T-cell function can be achieved through reduced expression or blockade of inhibitory molecules, or through increased expression of stimulatory molecules. Decreased gene expression can be accomplished by strategies that target either genomic DNA or mRNA. Emerging genome editing technologies can precisely introduce indels to prevent gene expression, or replace nucleotide sequences to modify gene expression (Fig. 2; ref.22). At least four types of programmable nucleases for genome editing have been described: meganucleases, zinc finger nucleases (ZFN), transcription activator-like effector nucleases (TALEN), and CRISPR-associated nuclease Cas9. Although programmable nuclease technologies hold great promise for manipulating gene expression in therapeutic T cells, their clinical development is in its infancy. A small clinical trial has tested ZFN editing of CCR5 for the treatment of patients with human immunodeficiency virus (HIV), suggesting the clinical feasibility of this approach in humans (23). Recombinant Cas9 protein complexed with an in vitro transcribed single-guide RNA (RNPs) has been reported to efficiently edit primary human CD4+ T-cell CXCR4 and PD-1 genes (24). A megaTAL nuclease introduced with adeno-associated virus-mediated delivery of a CCR5-targeting template has also been described to efficiently modify CCR5 in human T cells (25). Genome editing strategies have the advantage that they can completely eliminate expression of a functional gene product in some cells but the disadvantages that the platforms for high-efficiency editing and scaled up clinical application may require further development.

Figure 2.

Emerging technologies for gene regulation and potential target genes for modified expression in therapeutic T cells. Expression of genes with inhibitory effects on T-cell function can be reduced or eliminated from some cells by RNA silencing or gene editing technologies. Programmable nucleases (light blue), such as CRISPR-Cas9, TALENs, ZFNs, and meganucleases, can edit a particular gene, changing the genomic sequence and permanently preventing expression. Inhibitory RNAs (magenta) can target an mRNA transcript for reduced expression. The target genes for these systems may be inhibitory receptors (green), such as PD-1 or CTLA-4, or antagonists of TCR or cytokine signals, such as CISH, CBL-B, or SHP-1. Other technologies, such as ribozyme switches, transcriptional activators, and dimerizing receptors, use small molecules to activate expression of genes that stimulate T-cell function, such as cytokines or engineered stimulatory receptors. Small-molecule dimerizers can also be used as safety switches to induce cell death in the event of life-threatening T cell–mediated toxicity (i.e., iCaspase 9).

Figure 2.

Emerging technologies for gene regulation and potential target genes for modified expression in therapeutic T cells. Expression of genes with inhibitory effects on T-cell function can be reduced or eliminated from some cells by RNA silencing or gene editing technologies. Programmable nucleases (light blue), such as CRISPR-Cas9, TALENs, ZFNs, and meganucleases, can edit a particular gene, changing the genomic sequence and permanently preventing expression. Inhibitory RNAs (magenta) can target an mRNA transcript for reduced expression. The target genes for these systems may be inhibitory receptors (green), such as PD-1 or CTLA-4, or antagonists of TCR or cytokine signals, such as CISH, CBL-B, or SHP-1. Other technologies, such as ribozyme switches, transcriptional activators, and dimerizing receptors, use small molecules to activate expression of genes that stimulate T-cell function, such as cytokines or engineered stimulatory receptors. Small-molecule dimerizers can also be used as safety switches to induce cell death in the event of life-threatening T cell–mediated toxicity (i.e., iCaspase 9).

Close modal

Another approach to inhibiting gene expression is to increase degradation of a target mRNA through RNA interference with short hairpin RNA or artificial microRNA (Fig. 2). These technologies are easily adapted to clinical application by integrating expression of the targeting RNA into established clinical gene transfer systems. They also have the advantage that tandem hairpin designs may permit simultaneous targeting of multiple genes. A disadvantage is that this approach can decrease but not completely eliminate expression of a gene.

Potential targets for gene silencing or knockdown, or mAb blockade

A host of molecules have been reported to inhibit T-cell function, some of which have been studied in mouse models or clinical trials of T-cell-based cancer therapy. Much of this work has centered on inhibitory receptors expressed by T cells. mAbs that block interactions of the inhibitory receptor programmed death 1 (PD-1), with its ligands, programmed death ligand 1 (PD-L1) and programmed death ligand 2 (PD-L2), have clinical activity in melanoma, non–small cell lung cancer, renal cell carcinoma, urothelial cancer, head and neck squamous cell carcinoma, and other tumors (26). PD-1 axis blockade with mAbs also has been reported to improve adoptive T-cell therapy in mouse models of CAR and TCR therapy (27–29). Hence, PD-1 is an attractive molecule to target in combination with antigen receptor gene therapy. Another T-cell inhibitory receptor that has been targeted in cancer therapy is CTL antigen 4 (CTLA-4). Inhibition of CTLA-4 binding to its ligands, CD80 and CD86, can induce regression of melanoma and renal cell carcinoma (30, 31). A clinical trial for melanoma that combines CTLA-4 blockade with tumor-infiltrating lymphocyte (TIL) infusion is ongoing (NCT01701674); tumor response in 5 of 11 patients has been reported (32). Checkpoint blockade with the combination of anti-PD-1 and anti-CTLA-4 mAbs has greater clinical activity than blockade with either agent alone in melanoma (33). Dual PD-1 and CTLA-4 blockade combined with adoptive T-cell therapy is a potentially interesting area for further exploration (34). Further study in animal models and in clinical trials will be required to determine the optimal combinations of inhibitory receptors to antagonize. Emerging data also support strategies to inhibit intrinsic regulators of TCR and cytokine signaling, such as Src Homology Region 2 Domain-Containing Phosphatase 1 (SHP-1; refs.35, 36), cytokine inducible SH-2-Containing Protein (CISH; ref. 37), or E3 ubiquitin-protein ligase CBL-B (38, 39).

Controlled overexpression of genes that stimulate T-cell function

The function of antitumor T cells for adoptive transfer may be improved by transgenic expression of molecules that enhance T-cell activation and proliferation. It may be important to have an element of control over the timing and magnitude of expression of these molecules and the survival of the cells that express them. For example, constitutive IL15 transgene expression enhances the antitumor function of T cells in a mouse model of TCR gene therapy, but some mice die from delayed hyperproliferation of the infused cells (40), and human T cells transduced to constitutively express IL15 can display uncontrolled proliferation in vitro (41). Thus, although transgenic IL15 expression can improve T-cell function, its expression may need to be controlled. As another example of increased treatment risk from transgenic cytokine expression, in a clinical trial of TIL for melanoma in which T cells were transduced to express single-chain IL12 under a nuclear factor of the activated T-cell (NFAT) promoter, tumor regression occurred in some patients, but IL12-related toxicities prevented further development of the approach (42).

Technologies to modulate transgene expression or activation, or to terminate T-cell survival in vivo, might be required. One system for controlling T-cell stimulatory signals is to administer T cells that express an engineered costimulatory receptor that is reversibly dimerized by a small molecule (i.e., rimiducid) that is given systemically to the patient (Fig. 2; ref. 43). Another strategy is to use the same type of system as a “suicide gene” to induce cell death through dimerization of inducible caspase 9 (iCasp9; Fig. 2). This approach was reported to eliminate donor-derived iCasp9-engineered T cells in patients with graft-versus-host disease in a stem cell transplantation clinical trial (44). Other technologies in which transgene expression can be controlled by small molecules include the RheoSwitch inducible promotor system (45) and synthetic ribozyme switches; these technologies require vetting in clinical trials but have the potential to allow titration of transgene expression in vivo (Fig. 2; refs. 46, 47).

Few tumor-specific antigens are present on the surface of epithelial cancer cells and thus accessible to antibody or CAR therapy. TCR therapy can reach the larger pool of intracellular antigens but is limited by HLA restriction and the potential for tumor escape through defects in antigen processing and presentation. The identification and targeting of carefully selected epithelial tumor antigens is crucial for advancement of the field. Successful treatments may also require enhancement of T-cell function through genetic manipulation of intrinsic T-cell signals. Technologies for control of gene expression in T cells are evolving rapidly and might soon provide the decisive steps forward that are needed.

1.
Lee
DW
,
Kochenderfer
JN
,
Stetler-Stevenson
M
,
Cui
YK
,
Delbrook
C
,
Feldman
SA
, et al
T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial
.
Lancet
2015
;
385
:
517
28
.
2.
Kochenderfer
JN
,
Dudley
ME
,
Kassim
SH
,
Somerville
RPT
,
Carpenter
RO
,
Stetler-Stevenson
M
, et al
Chemotherapy-refractory diffuse large B-cell lymphoma and indolent B-cell malignancies can be effectively treated with autologous T cells expressing an anti-CD19 chimeric antigen receptor
.
J Clin Oncol
2015
;
33
:
540
9
.
3.
Kochenderfer
JN
,
Dudley
ME
,
Feldman
SA
,
Wilson
WH
,
Spaner
DE
,
Maric
I
, et al
B-cell depletion and remissions of malignancy along with cytokine-associated toxicity in a clinical trial of anti-CD19 chimeric-antigen-receptor-transduced T cells
.
Blood
2012
;
119
:
2709
20
.
4.
Garfall
AL
,
Maus
MV
,
Hwang
W-T
,
Lacey
SF
,
Mahnke
YD
,
Melenhorst
JJ
, et al
Chimeric antigen receptor T cells against CD19 for multiple myeloma
.
N Engl J Med
2015
;
373
:
1040
7
.
5.
Grupp
SA
,
Kalos
M
,
Barrett
D
,
Aplenc
R
,
Porter
DL
,
Rheingold
SR
, et al
Chimeric antigen receptor-modified T cells for acute lymphoid leukemia
.
N Engl J Med
2013
;
368
:
1509
18
.
6.
Porter
DL
,
Levine
BL
,
Kalos
M
,
Bagg
A
,
June
CH
. 
Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia
.
N Engl J Med
2011
;
365
:
725
33
.
7.
Robbins
PF
,
Kassim
SH
,
Tran
TLN
,
Crystal
JS
,
Morgan
RA
,
Feldman
SA
, et al
A pilot trial using lymphocytes genetically engineered with an NY-ESO-1-reactive T-cell receptor: long-term follow-up and correlates with response
.
Clin Cancer Res
2015
;
21
:
1019
27
.
8.
Long
AH
,
Haso
WM
,
Shern
JF
,
Wanhainen
KM
,
Murgai
M
,
Ingaramo
M
, et al
4-1BB costimulation ameliorates T cell exhaustion induced by tonic signaling of chimeric antigen receptors
.
Nat Med
2015
;
21
:
581
90
.
9.
Hinrichs
CS
,
Rosenberg
SA
. 
Exploiting the curative potential of adoptive T-cell therapy for cancer
.
Immunol Rev
2014
;
257
:
56
71
.
10.
Klebanoff
CA
,
Gattinoni
L
,
Palmer
DC
,
Muranski
P
,
Ji
Y
,
Hinrichs
CS
, et al
Determinants of successful CD8+ T-cell adoptive immunotherapy for large established tumors in mice
.
Clin Cancer Res
2011
;
17
:
5343
52
.
11.
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
.
12.
Parkhurst
MR
,
Yang
JC
,
Langan
RC
,
Dudley
ME
,
Nathan
D-AN
,
Feldman
SA
, et al
T cells targeting carcinoembryonic antigen can mediate regression of metastatic colorectal cancer but induce severe transient colitis
.
Mol Ther
2010
;
19
:
620
6
.
13.
Lamers
CHJ
,
Sleijfer
S
,
Vulto
AG
,
Kruit
WHJ
,
Kliffen
M
,
Debets
R
, et al
Treatment of metastatic renal cell carcinoma with autologous T-lymphocytes genetically retargeted against carbonic anhydrase IX: first clinical experience
.
J Clin Oncol
2006
;
24
:
e20
2
.
14.
Lamers
CH
,
Sleijfer
S
,
van Steenbergen
S
,
van Elzakker
P
,
van Krimpen
B
,
Groot
C
, et al
Treatment of metastatic renal cell carcinoma with CAIX CAR-engineered T cells: clinical evaluation and management of on-target toxicity
.
Mol Ther
2013
;
21
:
904
12
.
15.
Hinrichs
CS
,
Restifo
NP
. 
Reassessing target antigens for adoptive T-cell therapy
.
Nat Biotechnol
2013
;
31
:
999
1008
.
16.
Wang
QJ
,
Yu
Z
,
Griffith
K
,
Hanada
K
,
Restifo
NP
,
Yang
JC
. 
Identification of T-cell receptors targeting KRAS-mutated human tumors
.
Cancer Immunol Res
2016
;
4
:
204
14
.
17.
Stevanović
S
,
Draper
LM
,
Langhan
MM
,
Campbell
TE
,
Kwong
ML
,
Wunderlich
JR
, et al
Complete regression of metastatic cervical cancer after treatment with human papillomavirus-targeted tumor-infiltrating T cells
.
J Clin Oncol
2015
;
33
:
1543
50
.
18.
Draper
LM
,
Kwong
MLM
,
Gros
A
,
Stevanović
S
,
Tran
E
,
Kerkar
S
, et al
Targeting of HPV-16+ epithelial cancer cells by TCR gene engineered T cells directed against E6
.
Clin Cancer Res
2015
;
21
:
4431
9
.
19.
Bollard
CM
,
Gottschalk
S
,
Torrano
V
,
Diouf
O
,
Ku
S
,
Hazrat
Y
, et al
Sustained complete responses in patients with lymphoma receiving autologous cytotoxic T lymphocytes targeting Epstein-Barr virus latent membrane proteins
.
J Clin Oncol
2014
;
32
:
798
808
.
20.
Heslop
HE
,
Slobod
KS
,
Pule
MA
,
Hale
GA
,
Rousseau
A
,
Smith
CA
, et al
Long-term outcome of EBV-specific T-cell infusions to prevent or treat EBV-related lymphoproliferative disease in transplant recipients
.
Blood
2010
;
115
:
925
35
.
21.
Louis
CU
,
Straathof
K
,
Bollard
CM
,
Ennamuri
S
,
Gerken
C
,
Lopez
TT
, et al
Adoptive transfer of EBV-specific T cells results in sustained clinical responses in patients with locoregional nasopharyngeal carcinoma
.
J Immunother
2010
;
33
:
983
90
.
22.
Cox
DBT
,
Platt
RJ
,
Zhang
F
. 
Therapeutic genome editing: prospects and challenges
.
Nat Med
2015
;
21
:
121
31
.
23.
Tebas
P
,
Stein
D
,
Tang
WW
,
Frank
I
,
Wang
SQ
,
Lee
G
, et al
Gene editing of CCR5 in autologous CD4 T cells of persons infected with HIV
.
N Engl J Med
2014
;
370
:
901
10
.
24.
Hendel
A
,
Bak
RO
,
Clark
JT
,
Kennedy
AB
,
Ryan
DE
,
Roy
S
, et al
Chemically modified guide RNAs enhance CRISPR-Cas genome editing in human primary cells
.
Nat Biotechnol
2015
;
33
:
985
9
.
25.
Sather
BD
,
Ibarra
GSR
,
Sommer
K
,
Curinga
G
,
Hale
M
,
Khan
IF
, et al
Efficient modification of CCR5 in primary human hematopoietic cells using a megaTAL nuclease and AAV donor template
.
Sci Transl Med
2015
;
7
:
307ra156
307ra156
.
26.
Topalian
SL
,
Drake
CG
,
Pardoll
DM
. 
Immune checkpoint blockade: a common denominator approach to cancer therapy
.
Cancer Cell
2015
;
27
:
450
61
.
27.
John
LB
,
Devaud
C
,
Duong
CPM
,
Yong
CS
,
Beavis
PA
,
Haynes
NM
, et al
Anti-PD-1 antibody therapy potently enhances the eradication of established tumors by gene-modified T cells
.
Clin Cancer Res
2013
;
19
:
5636
46
.
28.
Blake
SJP
,
Ching
ALH
,
Kenna
TJ
,
Galea
R
,
Large
J
,
Yagita
H
, et al
Blockade of PD-1/PD-L1 promotes adoptive T-cell immunotherapy in a tolerogenic environment
.
PLoS One
2015
;
10
:
e0119483
.
29.
Peng
W
,
Liu
C
,
Xu
C
,
Lou
Y
,
Chen
J
,
Yang
Y
, et al
PD-1 blockade enhances T-cell migration to tumors by elevating IFN-γ inducible chemokines
.
Cancer Res
2012
;
72
:
5209
18
.
30.
Phan
GQ
,
Yang
JC
,
Sherry
RM
,
Hwu
P
,
Topalian
SL
,
Schwartzentruber
DJ
, et al
Cancer regression and autoimmunity induced by cytotoxic T lymphocyte-associated antigen 4 blockade in patients with metastatic melanoma
.
Proc Natl Acad Sci U S A
2003
;
100
:
8372
7
.
31.
Blansfield
JA
,
Beck
KE
,
Tran
K
,
Yang
JC
,
Hughes
MS
,
Kammula
US
, et al
Cytotoxic T-lymphocyte-associated antigen-4 blockage can induce autoimmune hypophysitis in patients with metastatic melanoma and renal cancer
.
J Immunother
2005
;
28
:
593
8
.
32.
Prabhakaran
S
,
Woods
D
,
Royster
E
,
Zager
J
,
Sondak
V
,
Pilon-Thomas
S
, et al
Pilot trial of ipilimumab and adoptive tumor-infiltrating lymphocyte (TIL) cell therapy
.
Ann Surg Oncol
2015
;
22
:
523
.
33.
Larkin
J
,
Chiarion-Sileni
V
,
Gonzalez
R
,
Grob
JJ
,
Cowey
CL
,
Lao
CD
, et al
Combined nivolumab and ipilimumab or monotherapy in untreated melanoma
.
N Engl J Med
2015
;
373
:
23
34
.
34.
Baksh
K
,
Weber
J
. 
Immune checkpoint protein inhibition for cancer: preclinical justification for CTLA-4 and PD-1 blockade and new combinations
.
Semin Oncol
2015
;
42
:
363
77
.
35.
Hebeisen
M
,
Baitsch
L
,
Presotto
D
,
Baumgaertner
P
,
Romero
P
,
Michielin
O
, et al
SHP-1 phosphatase activity counteracts increased T cell receptor affinity
.
J Clin Invest
2013
;
123
:
1044
56
.
36.
Stromnes
IM
,
Fowler
C
,
Casamina
CC
,
Georgopolos
CM
,
McAfee
MS
,
Schmitt
TM
, et al
Abrogation of SRC homology region 2 domain-containing phosphatase 1 in tumor-specific T cells improves efficacy of adoptive immunotherapy by enhancing the effector function and accumulation of short-lived effector T cells invivo
.
J Immunol
2012
;
189
:
1812
25
.
37.
Palmer
DC
,
Guittard
GC
,
Franco
Z
,
Crompton
JG
,
Eil
RL
,
Patel
SJ
, et al
Cish actively silences TCR signaling in CD8+ T cells to maintain tumor tolerance
.
J Exp Med
2015
;
212
:
2095
113
.
38.
Lutz-Nicoladoni
C
,
Wallner
S
,
Stoitzner
P
,
Pircher
M
,
Gruber
T
,
Wolf
AM
, et al
Reinforcement of cancer immunotherapy by adoptive transfer of cblb-deficient CD8+ T cells combined with a DC vaccine
.
Immunol Cell Biol
2012
;
90
:
130
4
.
39.
Stromnes
IM
,
Blattman
JN
,
Tan
X
,
Jeevanjee
S
,
Gu
H
,
Greenberg
PD
. 
Abrogating Cbl-b in effector CD8(+) T cells improves the efficacy of adoptive therapy of leukemia in mice
.
J Clin Invest
2010
;
120
:
3722
34
.
40.
Klebanoff
CA
,
Finkelstein
SE
,
Surman
DR
,
Lichtman
MK
,
Gattinoni
L
,
Theoret
MR
, et al
IL-15 enhances the invivo antitumor activity of tumor-reactive CD8+ T cells
.
Proc Natl Acad Sci U S A
2004
;
101
:
1969
74
.
41.
Hsu
C
,
Jones
SA
,
Cohen
CJ
,
Zheng
Z
,
Kerstann
K
,
Zhou
J
, et al
Cytokine-independent growth and clonal expansion of a primary human CD8+ T-cell clone following retroviral transduction with the IL-15 gene
.
Blood
2007
;
109
:
5168
77
.
42.
Zhang
L
,
Morgan
RA
,
Beane
JD
,
Zheng
Z
,
Dudley
ME
,
Kassim
SH
, et al
Tumor-infiltrating lymphocytes genetically engineered with an inducible gene encoding interleukin-12 for the immunotherapy of metastatic melanoma
.
Clin Cancer Res
2015
;
21
:
2278
88
.
43.
Narayanan
P
,
Lapteva
N
,
Seethammagari
M
,
Levitt
JM
,
Slawin
KM
,
Spencer
DM
. 
A composite MyD88/CD40 switch synergistically activates mouse and human dendritic cells for enhanced antitumor efficacy
.
J Clin Invest
2011
;
121
:
1524
34
.
44.
Di Stasi
A
,
Tey
S-K
,
Dotti
G
,
Fujita
Y
,
Kennedy-Nasser
A
,
Martinez
C
, et al
Inducible apoptosis as a safety switch for adoptive cell therapy
.
N Engl J Med
2011
;
365
:
1673
83
.
45.
Komita
H
,
Zhao
X
,
Katakam
AK
,
Kumar
P
,
Kawabe
M
,
Okada
H
, et al
Conditional interleukin-12 gene therapy promotes safe and effective antitumor immunity
.
Cancer Gene Ther
2009
;
16
:
883
91
.
46.
Win
MN
,
Smolke
CD
. 
A modular and extensible RNA-based gene-regulatory platform for engineering cellular function
.
Proc Natl Acad Sci U S A
2007
;
104
:
14283
8
.
47.
Chen
YY
,
Jensen
MC
,
Smolke
CD
. 
Genetic control of mammalian T-cell proliferation with synthetic RNA regulatory systems
.
Proc Natl Acad Sci U S A
2010
;
107
:
8531
6
.