The Basic Principles of Chimeric Antigen Receptor Design (original) (raw)

Skip Nav Destination

Article navigation

Issue Cover

Review| April 10 2013

Michel Sadelain;

Authors' Affiliation: Center for Cell Engineering, Molecular Pharmacology and Chemistry Program, and Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York

Corresponding Author: Michel Sadelain, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue Box 182, New York, NY 10065. Phone: 212-639-6190; Fax: 917-432-2340; E-mail: m-sadelain@ski.mskcc.org

Search for other works by this author on:

Renier Brentjens;

Authors' Affiliation: Center for Cell Engineering, Molecular Pharmacology and Chemistry Program, and Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York

Search for other works by this author on:

Isabelle Rivière

Authors' Affiliation: Center for Cell Engineering, Molecular Pharmacology and Chemistry Program, and Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York

Search for other works by this author on:

Crossmark: Check for Updates

Corresponding Author: Michel Sadelain, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue Box 182, New York, NY 10065. Phone: 212-639-6190; Fax: 917-432-2340; E-mail: m-sadelain@ski.mskcc.org

Received: November 29 2012

Revision Received: February 14 2013

Accepted: February 21 2013

Online ISSN: 2159-8290

Print ISSN: 2159-8274

©2013 American Association for Cancer Research.

2013

Cancer Discov (2013) 3 (4): 388–398.

Article history

Received:

November 29 2012

Revision Received:

February 14 2013

Accepted:

February 21 2013

Abstract

Chimeric antigen receptors (CAR) are recombinant receptors that provide both antigen-binding and T-cell–activating functions. A multitude of CARs has been reported over the past decade, targeting an array of cell surface tumor antigens. Their biologic functions have dramatically changed following the introduction of tripartite receptors comprising a costimulatory domain, termed second-generation CARs. These have recently shown clinical benefit in patients treated with CD19-targeted autologous T cells. CARs may be combined with costimulatory ligands, chimeric costimulatory receptors, or cytokines to further enhance T-cell potency, specificity, and safety. CARs represent a new class of drugs with exciting potential for cancer immunotherapy.

Significance: CARs are a new class of drugs with great potential for cancer immunotherapy. Upon their expression in T lymphocytes, CARs direct potent, targeted immune responses that have recently shown encouraging clinical outcomes in a subset of patients with B-cell malignancies. This review focuses on the design of CARs, including the requirements for optimal antigen recognition and different modalities to provide costimulatory support to targeted T cells, which include the use of second- and third-generation CARs, costimulatory ligands, chimeric costimulatory receptors, and cytokines. Cancer Discov; 3(4); 388–98. ©2013 AACR.

Introduction

Chimeric antigen receptors (CAR) are recombinant receptors for antigen, which, in a single molecule, redirect the specificity and function of T lymphocytes and other immune cells. The general premise for their use in cancer immunotherapy is to rapidly generate tumor-targeted T cells, bypassing the barriers and incremental kinetics of active immunization (1, 2). Once expressed in T cells, the CAR-modified T cells acquire supraphysiologic properties and act as “living drugs” that may exert both immediate and long-term effects. The engineering of CARs into T cells requires T cells to be cultured to allow for transduction and expansion. The transduction may use a variety of methods, but stable gene transfer is required to enable sustained CAR expression in clonally expanding and persisting T cells. In principle, any cell surface molecule can be targeted through a CAR, thus overriding tolerance to self-antigens and the antigen recognition gaps in the physiologic T-cell repertoire that limit the scope of T-cell reactivity. Various T cell subsets, as well as T-cell progenitors and other immune cells, such as natural killer cells, can be targeted with a CAR. Redirecting immune reactivity toward a chosen antigen is not the only purpose of smarter CARs, which are designed to accomplish much more than to target and initiate T-cell activation. CARs with different strengths and quality of signaling have the potential to modulate T-cell expansion and persistence as well as the strength of T-cell activation within the tumor microenvironment, features that dramatically alter the efficacy and safety of tumor-targeted T cells. In this regard, CARs provide a broader range of functional effects than transduced T-cell receptors (TCR), wherein strength of signaling, which is for the most part determined by the TCR's affinity for antigen, is the principal determinant of T-cell fate. CARs and TCRs have their respective advantages and disadvantages (1–4). Although the flexibility and “dynamic range” of CARs is attractive, current CARs are limited to recognizing cell surface antigens, whereas TCRs recognize both cell surface and intracellular proteins. CARs, however, do not require antigen processing and presentation by HLA and are therefore more broadly applicable to HLA-diverse patient populations. We discuss here the targeting and signaling properties of CARs, focusing on their effects on T-cell specificity, potency, and safety. Other general aspects of adoptive T-cell therapy that apply not only to the use of CARs but also to other T-cell therapies as well, including T-cell expansion methodologies, T-cell subset selection, and host conditioning, are beyond the scope of this review. Owing to the extraordinary potential of T-cell engineering and the modular nature of their structure, CARs are rapidly evolving and show great promise for their successful use in a wide range of immunotherapies.

CAR Targeting

CARs are recombinant receptors that typically target native cell surface antigens (4). Unlike the physiologic TCR, which engages HLA-peptide complexes, CARs engage molecules that do not require peptide processing or HLA expression to be recognized. CARs therefore recognize antigen on any HLA background, in contrast to TCRs, which need to be matched to the haplotype of the patient. Furthermore, CARs can target tumor cells that have downregulated HLA expression or proteasomal antigen processing, 2 mechanisms that contribute to tumor escape from TCR-mediated immunity (5). Another feature of the broad applicability of CARs is their ability to bind not only to proteins but also to carbohydrate and glycolipid structures, again expanding the range of potential targets. A survey of antigens targeted to date by CARs is shown in Table 1.

Table 1.

Antigens targeted by CARs

Target antigen Associated malignancy Receptor type (other specificity) In vivo studies References
α-Folate receptor Ovarian cancer scFv-FcϵRIγ Phase I (1)
Epithelial cancers scFv-41BB-CD3-ζ + (2)
CAIX Renal cell carcinoma scFv-CD4- FcϵRIγ Phase I (3–5)
Renal cell carcinoma G250-FceRIy (6–8)
CD19 B-cell malignancies scFv-CD3-ζ (EBV) (9)
B-cell malignancies scFv-CD3-ζ + (10, 11)
B cell malignancies scFv-CD28-CD3-ζ + (12–16)
Refractory follicular lymphoma scFv-CD3-ζ Phase I (17, 18)
B-cell malignancies scFv-CD28-CD3-ζ + (19–22)
ALL scFv-41BB-CD3-ζ (23)
ALL scFv-41BB-CD3-ζ + (24)
B-cell malignancies scFv-CD3-ζ (Influenza MP-1) + (25)
B-cell malignancies scFv-CD3-ζ (VZV) (26)
ALL FMC63-CD28-41BB-CD3-ζ +, − (27–29)
B-cell malignancies FMC63-41BB-CD3-ζ + (30)
Follicular lymphoma FMC63-CD28-CD3-ζ NCT00924326 (31)
B-cell malignancies FMC63-CD28-CD3-ζ NCT00924326 (32)
CLLand ALL SJ25C1-CD28-CD3-ζ (NCT00466531 NCT01044069) (33)
CLL FMC63-41BB-CD3-ζ NCT01029366 (34, 35)
Lymphoma scFv-CD3-ζ + scFv-CD28-CD3-ζ Phase I (36)
CD20 Lymphomas scFv-CD28-CD3-ζ (37)
B-cell malignancies scFv-CD4-CD3-ζ (38)
B-cell lymphomas scFv-CD3-ζ (39,40)
Mantle cell lymphoma and indolent B-cell lymphomas scFv-CD28-41BB-CD3-ζ NCT00621452 (41,42)
CD22 B-cell malignancies scFV-CD4-CD3-ζ (38)
CD23 CLL scFv-CD28-CD3-ζ + (43)
CD24 Pancreatic adenocarcinoma scFv- CD28-FcϵRIy + (44)
CD30 Lymphomas scFv-FcϵRIy (45)
Hodgkin lymphoma scFv-CD3-ζ (EBV) + (46)
scFv-CD28-CD3-ζ (EBV) + (47)
CD33 AML scFv-CD28-CD3-ζ (48)
cFv-41BB-CD3-ζ
scFv-CD28-CD3-ζ (EBV) + (49)
CD38 Non-Hodgkin lymphoma scFv-41BB-CD3-ζ + (50)
CD44v7/8 Cervical carcinoma scFv-CD8-CD3-ζ + (51)
CEA Colorectal cancer scFv-CD3-ζ + (52–56)
scFv-FcϵRIy + (55,57)
scFv-CD3ϵ (58)
scFv-CD28-CD3-ζ (59)
scFv-CD28-CD3-ζ + (60,61)
EGFRvIII Glioblastoma scFv-CD28-41BB-CD3-ζ NCT01454596 (62)
EGP-2 Multiple malignancies scFv-CD3-ζ (63)
scFv-FcϵRIy (63,64)
EGP-40 Colorectal cancer scFv-FcϵRIy (65)
EphA2 Glioblastoma scFv-CD28-CD3-ζ + (66)
Erb-B2 Breast cancer and others scFv-CD28-CD3-ζ + (67,68)
scFv-CD28-CD3-ζ (Influenza) + (69)
scFv-CD28mut.-CD3-ζ + (70)
Prostate cancer, colon cancer scFv-FcϵRIy + (71)
(72)
Various tumors scFv-CD28-41BB- CD3-ζ + (73,74)
Erb-B 2,3,4 Breast cancer and others Heregulin-CD3-ζ (75, 76)
scFv-CD3-ζ + (77)
FBP Ovarian cancer scFv-FcϵRIy + (78–80)
Ovarian cancer scFv-FcϵRIy (alloantigen) + (81)
Fetal acethylcholine receptor Rhabdomyosarcoma scFv-CD3-ζ (82)
GD2 Neuroblastoma, melanoma scFv-CD3-ζ (9,10)
scFv-CD3-ζ NCT00085930 (83, 84)
scFv-CD28-OX40-CD3-ζ −, + (74,85,86)
scFv-CD3-ζ (VZV) (26)
Ewing sarcoma scFv-CD28-CD3-ζ + (87)
GD3 Melanoma scFv-CD3-ζ, ScFv-CD3ϵ (88)
scFv-CD28-CD3-ζ + (89)
HER2 Medulloblastoma scFv-CD3-ζ + (90)
scFv-CD28-CD3-ζ + (91)
Pancreatic adenocarcinoma scFv-CD28-41BB-CD3-ζ + (44)
Glioblastoma Phase I (92)
Osteosarcoma scFv-CD28-CD3-ζ + (93)
Ovarian cancer scFv-CD28-CD3-ζ + (94)
HMW-MAA Melanoma scFv-CD3-ζ, ScFv-CD28-CD3-ζ (95)
IL-11Rα Osteosarcoma scFv-CD28-CD3-ζ + (96)
IL-13Rα2 Glioma IL-13-CD28-4-1BB-CD3-ζ + (97)
Glioblastoma IL-13-CD3-ζ + (98, 99)
Medulloblastoma IL-13-CD3-ζ + (100)
KDR Tumor neovasculature scFv-FcϵRIy (101)
κ-light chain B-cell malignancies (B-NHL, CLL) scFv-CD3-ζ + (102)
scFv-CD28-CD3-ζ + (102)
Lewis Y Various carcinomas scFv-FcϵRIy (103)
Epithelial-derived tumors scFv-CD28-CD3-ζ + (104–106)
L1-cell adhesion molecule Neuroblastoma scFv-CD3-ζ Phase I (107, 108)
MAGE-A1 Melanoma scFV-CD4-FcϵRIy (109)
scFV-CD28-FcϵRIy
Mesothelin Mesothelioma scFv-41BB-CD3-ζ + (73,110,111)
Murine CMV infected cells Murine CMV Ly49H-CD3-ζ + (112)
MUC1 Breast and ovarian cancer scFV-CD28-OX40-CD3-ζ + (113, 114)
MUC16 Ovarian cancer scFV-CD28-CD3-ζ (115)
NKG2D Ligands Myeloma, ovarian, and other tumors NKG2D-CD3-ζ + (116–121)
NY-ESO-1 (157–165) Multiple myeloma scFv-CD28-CD3-ζ + (122)
Oncofetal antigen (h5T4) Various tumors scFV-CD3-ζ (vaccination) + (123)
PSCA Prostate carcinoma 7F5-β2-CD3-ζ (124)
scFv-CD3-ζ (125)
PSMA Prostate cancer/tumor vasculature scFv-CD3-ζ + (126, 127)
Prostate/tumor vasculature scFv-CD28-CD3-ζ (128)
scFv-CD3-ζ + (129)
ROR1 B-CLL and mantle cell lymphoma scFv-CD28-CD3-ζ + (130)
Targeting via mAb IgE Various tumors FcϵRI-CD28-CD3-ζ + (131)
TAG-72 Adenocarcinomas scFv-CD3-ζ + (132, 133)
VEGF-R2 Tumor neovasculature scFv-CD3-ζ (134)
Biotinylated molecules Various tumors, ovarian cancer BBIR-z/CD28z + (135)
Target antigen Associated malignancy Receptor type (other specificity) In vivo studies References
α-Folate receptor Ovarian cancer scFv-FcϵRIγ Phase I (1)
Epithelial cancers scFv-41BB-CD3-ζ + (2)
CAIX Renal cell carcinoma scFv-CD4- FcϵRIγ Phase I (3–5)
Renal cell carcinoma G250-FceRIy (6–8)
CD19 B-cell malignancies scFv-CD3-ζ (EBV) (9)
B-cell malignancies scFv-CD3-ζ + (10, 11)
B cell malignancies scFv-CD28-CD3-ζ + (12–16)
Refractory follicular lymphoma scFv-CD3-ζ Phase I (17, 18)
B-cell malignancies scFv-CD28-CD3-ζ + (19–22)
ALL scFv-41BB-CD3-ζ (23)
ALL scFv-41BB-CD3-ζ + (24)
B-cell malignancies scFv-CD3-ζ (Influenza MP-1) + (25)
B-cell malignancies scFv-CD3-ζ (VZV) (26)
ALL FMC63-CD28-41BB-CD3-ζ +, − (27–29)
B-cell malignancies FMC63-41BB-CD3-ζ + (30)
Follicular lymphoma FMC63-CD28-CD3-ζ NCT00924326 (31)
B-cell malignancies FMC63-CD28-CD3-ζ NCT00924326 (32)
CLLand ALL SJ25C1-CD28-CD3-ζ (NCT00466531 NCT01044069) (33)
CLL FMC63-41BB-CD3-ζ NCT01029366 (34, 35)
Lymphoma scFv-CD3-ζ + scFv-CD28-CD3-ζ Phase I (36)
CD20 Lymphomas scFv-CD28-CD3-ζ (37)
B-cell malignancies scFv-CD4-CD3-ζ (38)
B-cell lymphomas scFv-CD3-ζ (39,40)
Mantle cell lymphoma and indolent B-cell lymphomas scFv-CD28-41BB-CD3-ζ NCT00621452 (41,42)
CD22 B-cell malignancies scFV-CD4-CD3-ζ (38)
CD23 CLL scFv-CD28-CD3-ζ + (43)
CD24 Pancreatic adenocarcinoma scFv- CD28-FcϵRIy + (44)
CD30 Lymphomas scFv-FcϵRIy (45)
Hodgkin lymphoma scFv-CD3-ζ (EBV) + (46)
scFv-CD28-CD3-ζ (EBV) + (47)
CD33 AML scFv-CD28-CD3-ζ (48)
cFv-41BB-CD3-ζ
scFv-CD28-CD3-ζ (EBV) + (49)
CD38 Non-Hodgkin lymphoma scFv-41BB-CD3-ζ + (50)
CD44v7/8 Cervical carcinoma scFv-CD8-CD3-ζ + (51)
CEA Colorectal cancer scFv-CD3-ζ + (52–56)
scFv-FcϵRIy + (55,57)
scFv-CD3ϵ (58)
scFv-CD28-CD3-ζ (59)
scFv-CD28-CD3-ζ + (60,61)
EGFRvIII Glioblastoma scFv-CD28-41BB-CD3-ζ NCT01454596 (62)
EGP-2 Multiple malignancies scFv-CD3-ζ (63)
scFv-FcϵRIy (63,64)
EGP-40 Colorectal cancer scFv-FcϵRIy (65)
EphA2 Glioblastoma scFv-CD28-CD3-ζ + (66)
Erb-B2 Breast cancer and others scFv-CD28-CD3-ζ + (67,68)
scFv-CD28-CD3-ζ (Influenza) + (69)
scFv-CD28mut.-CD3-ζ + (70)
Prostate cancer, colon cancer scFv-FcϵRIy + (71)
(72)
Various tumors scFv-CD28-41BB- CD3-ζ + (73,74)
Erb-B 2,3,4 Breast cancer and others Heregulin-CD3-ζ (75, 76)
scFv-CD3-ζ + (77)
FBP Ovarian cancer scFv-FcϵRIy + (78–80)
Ovarian cancer scFv-FcϵRIy (alloantigen) + (81)
Fetal acethylcholine receptor Rhabdomyosarcoma scFv-CD3-ζ (82)
GD2 Neuroblastoma, melanoma scFv-CD3-ζ (9,10)
scFv-CD3-ζ NCT00085930 (83, 84)
scFv-CD28-OX40-CD3-ζ −, + (74,85,86)
scFv-CD3-ζ (VZV) (26)
Ewing sarcoma scFv-CD28-CD3-ζ + (87)
GD3 Melanoma scFv-CD3-ζ, ScFv-CD3ϵ (88)
scFv-CD28-CD3-ζ + (89)
HER2 Medulloblastoma scFv-CD3-ζ + (90)
scFv-CD28-CD3-ζ + (91)
Pancreatic adenocarcinoma scFv-CD28-41BB-CD3-ζ + (44)
Glioblastoma Phase I (92)
Osteosarcoma scFv-CD28-CD3-ζ + (93)
Ovarian cancer scFv-CD28-CD3-ζ + (94)
HMW-MAA Melanoma scFv-CD3-ζ, ScFv-CD28-CD3-ζ (95)
IL-11Rα Osteosarcoma scFv-CD28-CD3-ζ + (96)
IL-13Rα2 Glioma IL-13-CD28-4-1BB-CD3-ζ + (97)
Glioblastoma IL-13-CD3-ζ + (98, 99)
Medulloblastoma IL-13-CD3-ζ + (100)
KDR Tumor neovasculature scFv-FcϵRIy (101)
κ-light chain B-cell malignancies (B-NHL, CLL) scFv-CD3-ζ + (102)
scFv-CD28-CD3-ζ + (102)
Lewis Y Various carcinomas scFv-FcϵRIy (103)
Epithelial-derived tumors scFv-CD28-CD3-ζ + (104–106)
L1-cell adhesion molecule Neuroblastoma scFv-CD3-ζ Phase I (107, 108)
MAGE-A1 Melanoma scFV-CD4-FcϵRIy (109)
scFV-CD28-FcϵRIy
Mesothelin Mesothelioma scFv-41BB-CD3-ζ + (73,110,111)
Murine CMV infected cells Murine CMV Ly49H-CD3-ζ + (112)
MUC1 Breast and ovarian cancer scFV-CD28-OX40-CD3-ζ + (113, 114)
MUC16 Ovarian cancer scFV-CD28-CD3-ζ (115)
NKG2D Ligands Myeloma, ovarian, and other tumors NKG2D-CD3-ζ + (116–121)
NY-ESO-1 (157–165) Multiple myeloma scFv-CD28-CD3-ζ + (122)
Oncofetal antigen (h5T4) Various tumors scFV-CD3-ζ (vaccination) + (123)
PSCA Prostate carcinoma 7F5-β2-CD3-ζ (124)
scFv-CD3-ζ (125)
PSMA Prostate cancer/tumor vasculature scFv-CD3-ζ + (126, 127)
Prostate/tumor vasculature scFv-CD28-CD3-ζ (128)
scFv-CD3-ζ + (129)
ROR1 B-CLL and mantle cell lymphoma scFv-CD28-CD3-ζ + (130)
Targeting via mAb IgE Various tumors FcϵRI-CD28-CD3-ζ + (131)
TAG-72 Adenocarcinomas scFv-CD3-ζ + (132, 133)
VEGF-R2 Tumor neovasculature scFv-CD3-ζ (134)
Biotinylated molecules Various tumors, ovarian cancer BBIR-z/CD28z + (135)

NOTE: This table lists the target antigens and corresponding CARs that have been reported to date. The reference list for the table can be found in Supplementary item 1.

Abbreviations: ALL, acute lymphoblastic leukemia; AML, acute myeloid leukemia; CLL, chronic lymphocytic leukemia; CMV, cytomegalovirus; EBV, Epstein-Barr virus; EGFR, epidermal growth factor receptor; HMW-MAA, high-molecular-weight melanoma-associated antigen; mAb, monoclonal antibody; NHL, non-Hodgkin lymphoma; PSCA, prostate stem cell antigen; PSMA, prostate-specific membrane antigen.

The moieties used to bind to antigen fall in 3 general categories: (i) single-chain variable fragment (scFv) derived from antibodies; (ii) Fabs fragment antigen-binding (Fab) selected from libraries; or (iii) nature ligands that engage their cognate receptor (see Fig. 1, first-generation CARs). Successful examples in each of these categories (too many to cite) have been reported (Table 1). scFvs derived from murine immunoglobulins are commonly used, as they are easily derived from well-characterized monoclonal antibodies. However, they may prove to be more immunogenic than Fabs derived from human libraries or invariant human ligands.

Figure 1.

Figure 1. Three generations of CARs. Left, first-generation CARs, including activating receptors such as CD8/CD3-ζ fusion receptors (10) and T-bodies (14); middle, second-generation CARs providing dual signaling to direct combined activating and costimulatory signals; right, third-generation CARs comprising more complex structures with 3 or more signaling domains. mAb, monoclonal antibody.

Three generations of CARs. Left, first-generation CARs, including activating receptors such as CD8/CD3-ζ fusion receptors (10) and T-bodies (14); middle, second-generation CARs providing dual signaling to direct combined activating and costimulatory signals; right, third-generation CARs comprising more complex structures with 3 or more signaling domains. mAb, monoclonal antibody.

Figure 1.

Figure 1. Three generations of CARs. Left, first-generation CARs, including activating receptors such as CD8/CD3-ζ fusion receptors (10) and T-bodies (14); middle, second-generation CARs providing dual signaling to direct combined activating and costimulatory signals; right, third-generation CARs comprising more complex structures with 3 or more signaling domains. mAb, monoclonal antibody.

Three generations of CARs. Left, first-generation CARs, including activating receptors such as CD8/CD3-ζ fusion receptors (10) and T-bodies (14); middle, second-generation CARs providing dual signaling to direct combined activating and costimulatory signals; right, third-generation CARs comprising more complex structures with 3 or more signaling domains. mAb, monoclonal antibody.

Close modal

The rules for selecting optimal epitopes for CAR targeting are still little known. The position of the epitope and its distance to the cell surface are expected to affect the binding to the antigen and the optimal formation of T-cell target conjugates and synapses (6), but little is known about the overall rules governing the optimal epitope selection. Empirical observations indicate that the structure of the “spacer region” between an scFv and the transmembrane region (Fig. 1) can affect CAR specificity, but no definitive principles have yet emerged (7). CAR length and protrusion from the T-cell membrane are likely to affect synapse formation. The optimal affinity of CARs is also little defined. Few studies have attempted to address this question, which is of major importance in the case of TCRs (8, 9) and likely to affect CAR function as well. Informative studies comparing multiple CARs recognizing the same epitope with different affinities are still lacking. Finally, the effect of antigen density is not yet well defined. CARs typically target highly expressed antigens, but little is known about minimum thresholds. It is uncertain whether CARs are as exquisitely sensitive as TCRs (9). If not, lesser sensitivity could represent a limitation in their activity against tumors expressing low antigen levels but may also turn into an advantage where avoidance of low-level antigen expression on normal cells is desirable. Thus, the antigen-binding moiety of the CAR is not only a targeting device but also is integral to CAR function, which is not solely defined by the signaling components incorporated into the cytoplasmic domain of CAR.

CAR Signaling

The first fusion receptors shown to have T-cell–activating potential on their own were chimeric molecules between CD3-ζ or Fc receptor γ and CD8, CD4, CD25, or CD16 (Fig. 1, first-generation CARs), which were shown to initiate phosphatidylinositol and tyrosine kinase pathways together with calcium influx in human T-cell leukemias (10–13). The addition of a hapten-specific scFv derived from a murine antibody to the extracellular portion of such fusions, termed a T-body (Fig. 1, first-generation CARs), effectively redirected cytolysis by murine T-cell hybridomas (14). Although CD3-ζ chain aggregation is sufficient to enable lytic activity in CTL lines, it is important to bear in mind that the strength of signal required for cytotoxicity is lower than that needed for other T-cell functions. This likely underscores the limited therapeutic responses reported with activating receptors, the antitumoral effects of which are often confined to local administration models (15, 16) or short-term systemic models (17). In transgenic mice, T cells expressing CARs that only comprise an activation domain in their cytoplasmic domain are prone to undergoing anergy (18).

Once we could efficiently transduce human primary T cells (see below), we found that CD3-ζ CARs failed to elicit a robust cytokine response, including interleukin (IL)-2, and support T-cell expansion upon repeated exposure to antigen (19). It would take the design of a tripartite fusion receptor, possessing both activating and costimulatory properties (Fig. 1, second-generation CARs), to obtain absolute T-cell expansion of human peripheral blood T cells upon repeated exposure to antigen (20). Significantly, these essential functions cannot be investigated in leukemic or immortalized T-cell lines (21), but only in primary T cells, which CAR investigators have now solidly embraced as the gold standard for evaluating CAR function in vitro or in vivo. Eventually dubbed second-generation CARs, receptors encompassing the CD3-ζ chain, and the cytoplasmic domain of a costimulatory receptor such as CD28, 4-1BB, DAP10, OX40, or ICOS, were eventually reported (Table 1). The superior activity of dual-signaling receptors over the activating-only receptors was observed in several models using mouse or human T cells (22–24). The key attribute of dual-signaling receptors is to confer greater strength of signaling and persistence to the T cells, resulting in their overall greater potency. The enhanced persistence imparted by dual-signaling CARs has been confirmed in patients treated with a mixture of T cells transduced with either a CD28/CD3ζ or CD3ζ-only CAR (25). Second-generation CARs come in varied configurations, but exhaustive comparisons are still lacking. Some CD28 and 4-1BB–based second-generation CARs were compared in animal models, but either one proved to be superior to the other in different contexts. In one study, Carpenito and colleagues (26) found that two CD28 and 4-1BB–based CD19-specific CARs had the same therapeutic activity, but they noted that the T cells expressing the 19-BB CAR accumulated to greater levels over time, possibly in an antigen-independent fashion. This difference was not observed in another model (27). More comparative studies are needed to take into account the variability between CARs within any one given category. For example, different CD28/CD3ζ CARs differ in their ability to elicit IL-2 secretion (20, 28). Furthermore, the location of the targeted epitope, its density, the affinity of the CAR, and other topologic effects of CAR structure affect CAR signaling, as discussed above. Comparisons will thus need to include multiple representatives of the evaluated categories to reach generalizable conclusions.

A third generation of CARs, encompassing 2 costimulatory domains combined with an activation domain in their cytoplasmic domain (Fig. 1, third-generation CARs), has been described, which seems to confer yet greater potency to tumor targeted T cells in some mouse models (26, 27, 29, 30). These more complex structures warrant further study as well. A first clinical study using a CD20-specific CD28/4-1BB/CD3ζ did not show dramatic responses (31), but this early result should not in any way detract from the potential value of these “triple-decker” CARs. Overall, more investigation is needed to attain a better understanding of optimal CAR signaling to promote sustained T-cell function and survival, preventing premature death, rapid exhaustion, or undue proliferation.

Potentiation and Complementation of CAR Function

Costimulatory support can be engineered into T cells in other ways than through a CAR (Fig. 2). The coexpression of chimeric costimulatory receptors (CCR), costimulatory receptor ligands, and cytokines has been used to modulate the function and/or survival of CAR-transduced T cells.

Figure 2.

Figure 2. Strategies to provide costimulatory support to CAR-modified T cells. From upper left, physiologic costimulatory ligand display by professional or artificial antigen-presenting cells (AAPC); upper right, auto- and transcostimulation by T cells expressing costimulatory ligands; lower right, embedded costimulation provided by second- or third-generation CARs; lower left, redirected costimulation mediated by an antigen-specific chimeric costimulatory receptor. CAR-1, 2, 3, first-, second-, and third-generation CARs; HLAp, HLA-peptide complex.

Strategies to provide costimulatory support to CAR-modified T cells. From upper left, physiologic costimulatory ligand display by professional or artificial antigen-presenting cells (AAPC); upper right, auto- and transcostimulation by T cells expressing costimulatory ligands; lower right, embedded costimulation provided by second- or third-generation CARs; lower left, redirected costimulation mediated by an antigen-specific chimeric costimulatory receptor. CAR-1, 2, 3, first-, second-, and third-generation CARs; HLAp, HLA-peptide complex.

Figure 2.

Figure 2. Strategies to provide costimulatory support to CAR-modified T cells. From upper left, physiologic costimulatory ligand display by professional or artificial antigen-presenting cells (AAPC); upper right, auto- and transcostimulation by T cells expressing costimulatory ligands; lower right, embedded costimulation provided by second- or third-generation CARs; lower left, redirected costimulation mediated by an antigen-specific chimeric costimulatory receptor. CAR-1, 2, 3, first-, second-, and third-generation CARs; HLAp, HLA-peptide complex.

Strategies to provide costimulatory support to CAR-modified T cells. From upper left, physiologic costimulatory ligand display by professional or artificial antigen-presenting cells (AAPC); upper right, auto- and transcostimulation by T cells expressing costimulatory ligands; lower right, embedded costimulation provided by second- or third-generation CARs; lower left, redirected costimulation mediated by an antigen-specific chimeric costimulatory receptor. CAR-1, 2, 3, first-, second-, and third-generation CARs; HLAp, HLA-peptide complex.

Close modal

Costimulatory Ligands

The constitutive expression of costimulatory ligands on the T-cell surface (Fig. 2) provides a powerful means to potentiate CAR-targeted T cells. Several ligands for immunoglobulin (Ig) super-family and TNF receptor family costimulatory receptor, including CD80, CD86, 4-1BBL, OX40L, and CD70, have been shown to enhance T-cell proliferation and cytokine secretion upon antigen engagement (32). The combination of 2 ligands, in particular CD80 and 4-1BBL, results in sustained in vivo T-cell expansion and persistence, associated with the rejection of massive, established tumor burdens (32). Both auto- and transcostimulation have been shown to contribute to enhanced T-cell activity in this context, which may be useful to enhance adoptive cell therapies using CAR- or TCR-transduced T cells. The occurrence of costimulatory ligands found on tumor cells is also likely to influence the activity of CAR-modified T cells, whether they are activating (e.g., CD80, CD40L, 4-1BBL) or inhibitory (e.g., PD-L1).

Chimeric Costimulatory Receptors

CCRs mimic costimulatory signals but, unlike CARs, do not provide a T-cell activation signal. Their purpose is to provide costimulation, for example, a CD28-like signal (33), in the absence of the natural costimulatory ligand on the antigen-presenting cell (Fig. 2). They thus provide a means for the tumor to direct counterfeit costimulation specifically within the tumor microenvironment. CCRs targeting the glycolipid GD2, MUC16, prostate-specific membrane antigen (PSMA), and the α-folate receptor have been described, used in conjunction with a TCR or a CAR to augment T-cell reactivity against the dual-antigen expressing T cells, reinforcing T-cell activation in the absence of natural costimulatory ligands and in an antigen-dependent fashion (20, 33–35). Under particular conditions, CCRs may also be used to improve selective tumor targeting, as further discussed below.

Cytokines

Another approach to enhance the potency of CAR-targeted T cells is to further genetically modify the T cells to secrete proinflammatory or proproliferative cytokines. The purpose behind this approach is not only to provide autocrine support to enhance the function, proliferation, and/or persistence of CAR-expressing T cells but also to favorably alter the tumor microenvironment and recruit endogenous innate and cognate immune effectors. The expression of T-cell–encoded cytokines additionally aims to limit the systemic toxicity of many cytokines. Preclinical reports investigating γc cytokines or IL-12 show great promise for this approach.

T-cell–encoded IL-15 increases the viability and proliferation of human peptide-specific T cells despite withdrawal of exogenous IL-2 (36). Improved in vitro and in vivo expansion of human Epstein–Barr virus–specific cytotoxic T cells following retroviral gene transfer of the IL-2 or IL-15 cDNA has also been reported (37). The report of an isolated IL-15–modified CD8+ T-cell clone exhibiting logarithmic proliferation for over 1 year in the absence of exogenous cytokine support cautions against this approach (38), although this concern may be mitigated by using a suicide gene to potentially remove T cells via drug-induced apoptosis (37). Cytokine-modified T cells used as antigen-presenting cells to expand tumor-targeted T cells, expressing either IL-7 and IL-12 (39) or IL-21 (40), successfully expanded tumor-targeted T cells, with a more favorable central memory phenotype in the latter case. Comparisons between cytokines expressed at different levels in different assays or tumor models are complex to interpret. Nonetheless, we compared CD19 CAR-targeted human 19z1+ T cells that constitutively expressed either IL-2, IL-7, IL-15, or IL-21 under standardized conditions and found that all 4 γc cytokines enhanced tumor rejection in a xenotransplant model of human CD19+ tumor, more so in this context, with IL-7 and IL-21 than IL-2 and IL-15 (41).

In an immune-competent syngeneic tumor model, CD19-targeted, CAR-modified T cells expressing IL-12 showed greater efficacy than CAR-modified T cells alone (42). Significantly, IL-12–modified T cells eradicated CD19+ tumors in the absence of any prior conditioning and, additionally, exhibited resistance to regulatory T-cell (Treg) inhibition. In a murine melanoma model, transgenic Pmel-1 CD8+ T cells, or Pmel-1 TCR-transduced murine T cells that were modified to express IL-12, eradicated established tumors with significantly greater potency than T cells expressing the Pmel-1 TCR alone (43). Similarly improved outcomes were obtained in tumor-bearing mice treated with IL-12–secreting T cells targeted to tumor by an anti-VEGF receptor-2 CAR (44). In both latter models, the effect of IL-12 seems to act at least in part by altering myeloid cells in the tumor microenvironment (44, 45).

The titration of cytokine secretion by T cells is important because of the potential toxicity of elevated systemic levels. One may address this concern by appropriately calibrating promoter strength or through conditional cytokine release following T-cell activation using nuclear factor of activated T cells (NFAT)-inducible promoters (46). Using this approach to control cytokine secretion, 2 trials treating metastatic NY-ESO-1+ tumors with autologous TCR-targeted T cells or tumor-infiltrating T cells secreting IL-12 are under way at the National Cancer Institute (NCI; NCT01457131, NCT01236573).

CARs in the Clinic

The CD19 Paradigm

The most investigated target to date is CD19, an attractive target for CAR-based therapy, as it is present in most B-cell leukemias and lymphomas but not in any normal tissue other than the B cell lineage (47, 48). CD19+ malignancies were the first cancers to be eliminated by CAR-engineered human T cells administered intravenously to systemic tumor-bearing mice (49). Successful B-cell tumor eradication was eventually obtained with different CD19 CARs (15, 22–24), paving the way for several ongoing clinical trials. The targeting of CD19 has thus become a paradigm for evaluating CAR technology (50). We estimate that at least 50 patients with leukemia or lymphoma have been treated at the time this review is written, 28 of whom were reported from 5 centers in the past year (25, 51–56). The reported clinical outcomes were recently reviewed elsewhere (57, 58) and are briefly summarized here.

The largest series and most dramatic early results were reported from the NCI, Memorial Sloan-Kettering Cancer Center (MSKCC, New York, NY), and the Abramson Family Cancer Research Institute at the University of Pennsylvania (UPenn, Philadelphia, PA). These clinical trials followed the same overall steps, including a patient T-cell apheresis, retroviral or lentiviral CAR transduction, T-cell expansion, and host conditioning before T-cell infusion. However, the trials differ in several regards, including not only CAR design (same CD28/CD3-ζ dual-signaling domain used at the NCI and MSKCC and 4-1BB/CD3-ζ used at UPenn) but also T-cell manufacturing, conditioning chemotherapy, tumor burden, tumor chemosensitivity, and T-cell dosage, which are reviewed in detail in ref. (57) Kochendorfer and colleagues (53) and the NCI group reported on 8 patients [4 with chronic lymphocytic leukemia (CLL), 3 with follicular lymphoma, and 1 with marginal zone lymphoma] conditioned with fludarabine and cyclophosphamide, and further given IL-2 after T-cell infusion. Among the 4 patients with CLL, 1 achieved a complete response (CR) and another stable disease. Four of the 8 treated patients exhibited B-cell aplasias. Brentjens and colleagues reported on 8 patients with CLL and 1 patient with B-cell acute lymphoblastic leukemia (B-ALL), whose disease was resistant to the milder cyclophosphamide conditioning regimen used in 5 of the 8 patients with CLL (the first 3 were given T cells without any prior conditioning; no significant response was obtained). In the CLL cohort, 2 patients had stable disease and 1 patient showed a substantial lymph node reduction. None of the patients with CLL developed B-cell aplasia, in contrast with the patient with relapsed B-ALL (52). Kalos and colleagues (54, 55) treated 3 patients with bulky CLL who were conditioned with bendamustine, a highly active agent in such patients. Two of them achieved dramatic, long-lasting CRs. The reasons for the different outcomes in these 15 patients with CLL treated at 3 different centers, which include significant differences in CAR design, conditioning intensity, and the selection of chemosensitive patients, among many variables (57), still remain to be elucidated. Taken together, better responses were observed following more active conditioning, resulting in a 25% CR rate in 12 patients with CLL treated with T cells following chemotherapy conditioning. Much will undoubtedly be learned about the role of the CAR and other parameters by comparing biologic and clinical outcomes using similarly manufactured T cells in similarly selected patients.

Solid Tumors

One next frontier for CAR-based therapies is to take on solid tumors. Early attempts with first-generation CARs did not yield very encouraging data (59), although one recent study targeting the GD2 ganglioside in children with neuroblastoma showed 2 CRs in 13 patients (60). Solid tumors present a different set of challenges compared with B-cell malignancies: overall lesser sensitivity to T-cell–mediated cytotoxicity, a microenvironment that presents with an array of immunosuppressive mechanisms differing between tumor types, and a paucity of target antigens with an expression profile as favorable as that of CD19. Despite an impressive number of investigated targets (Table 1), few target candidates are tumor specific or restricted to the tumor and a “dispensable” normal cell type or a tissue that is sheltered from an immune attack. In this perspective, identifying valid targets to achieve efficacious tumor rejection while ensuring patient safety is an essential goal that requires further investigation. Nonetheless, several trials using first- and second-generation CARs are under way and listed at ClinicalTrials.gov.

CAR Safety

The two main safety concerns associated with the use of CARs are the targeted destruction of normal tissues and cytokine storms associated with large-scale immune responses. The toxicity of the different conditioning regimens used in conjunction with adoptive T-cell therapies is also a significant issue to consider but is beyond the scope of this review.

On-target, Off-tumor Responses

The immune-mediated rejection of normal tissues that express the targeted antigen is referred to as an “on-target, off-tumor” response. This occurrence is best illustrated in the B-cell aplasias induced by CD19-targeted CARs (52–54). Although B-cell aplasia can be effectively managed by administering intravenous immunoglobulin, such collateral damage may not be tolerable in many other instances. This may for example be the case for her2 (61), which is expressed at a low level in several normal tissues, including heart and pulmonary vasculature. Other examples, for which no toxicities have been reported to date, include PSMA, which is highly expressed in castrate-resistant, metastatic prostate cancer but is detected in type II astrocytes, the renal proximal tubule, and the jejunum brush border (62), ROR1, which is expressed in a subset of leukemias and lymphomas but is also detected in adipocytes (63). T cells can be very effective at destroying normal tissues that express the targeted antigen, as exemplified by the ocular and vestibular effects of MART-1–specific T cells (64) and the cholestatic effect of T cells targeted to carbonic anhydrase IX (65). Whether the very low level expression of antigens such as PSMA and ROR1 on normal tissues will expose these tissues to immune destruction is at present unknown. This problem would be easily resolved if there were truly tumor-specific cell surface molecules to target, but such molecules are so far very rare. The identification of restricted CAR targets is therefore a high priority.

Cytokine Storms

The second major concern is that of “cytokine storms” associated with intense antitumor responses mediated by large numbers of activated T cells (53–55). These events typically cause high fever and hypotension, potentially resulting in organ failure. Their management may require steroids, vasopressors, and/or supportive therapy delivered in the intensive care unit. Grupp and colleagues (66) have observed that IL-6 blockade using tocilizumab may be effective in steroid-refractory circumstances without compromising T-cell efficacy. Unlike many conventional drug-induced side effects, this toxicity cannot be controlled by simply reducing drug dosage, as proliferating T cells will increase in numbers and eventually reach critical levels at which a synchronous cytokine response exceeds tolerability. Split T-cell dosing or short-lived T cells may partially reduce this effect, but more fundamental solutions are needed to reduce and ideally prevent the occurrence of overwhelming T-cell activity.

Emerging Solutions to Improve CAR Safety

Recognizing that CAR-modified T cells are in general well tolerated, their broader use requires having solid strategies to treat or, better, prevent on-target, off-tumor effects and cytokine storms. One therapeutic option is to use suicide genes to have a means to eliminate an excessive response. Herpes simplex virus thymidine kinase (67) and inducible caspase-9 (68) are clinically tested systems that could be used to halt deleterious responses (69). The drawbacks to this approach are that it is reactive, not preventive, and that active T cells will be eliminated, possibly curtailing the therapy. A better understanding of cytokine storms may offer novel prospects for reducing toxicity without compromising therapy and limiting the use of corticosteroids (66). Significantly, we find, in patients with ALL treated with a CD19-specific CD28/CD3-ζ CAR, that stronger cytokine responses occur in those with large tumor burdens but not in those with minimal residual disease at the time of T-cell infusion, a finding that suggests that reducing tumor burden by alternative means before T-cell infusion will reduce the risk of T-cell–induced cytokine-mediated toxicity following a subsequent T-cell infusion (70). Ultimately, the design of T cells that are effective, highly tumor specific, and regulated in their maximal accumulation and activation (so as to preclude toxic cytokine elevation), will represent a valuable advance for the use of CARs. One approach to improve tumor selectivity, based on combinatorial antigen recognition, is reviewed below.

New Directions in the CAR Industry

New technologies or concepts other than the design of better CARs and their combination with costimulatory ligands, chimeric costimulatory receptors or cytokines, are emerging to broaden or improve the use of CARs. These include improved CAR delivery systems, the design of CARs that recognize intracellular antigens, and combinatorial antigen recognition to increase T-cell specificity and potency.

CAR Delivery

The mechanics of T-cell transduction are beyond the scope of this review but are briefly addressed here. CARs began to be investigated in meaningful ways when methods for the transduction of human primary T cells became available in the middle to late 1990s (71–73). For the past 15 years, virtually all CAR studies have relied on retroviral vectors, including gamma-retroviral and lentiviral vectors (74). Most current clinical trials use retroviral vectors derived from murine leukemia virus or human immune deficiency virus-1. Although retroviral vectors can induce insertional oncogenesis in hematopoietic progenitors (75, 76), T cells seem to be far less susceptible to retroviral vector–induced transformation (77–80). Transposases, which also provide random vector integration (81), are starting to be evaluated in the context of CAR therapy (82). The relative advantages and disadvantages of these different integrating systems have not yet been elucidated but will hinge on CAR expression levels, silencing over time, safety features, ease of manufacturing and usage, and cost. Although T-cell transformation secondary to insertional mutagenesis has not been reported to date, site-directed vector integration into genomic safe harbors (83) may eventually enable the achievement of long-term CAR expression without any risk of insertional oncogenesis. Alternative approaches that do not rely on transgene integration, which use RNA electroporation (84, 85) or cell surface conjugation (86), result in transient CAR expression, precluding effective T-cell persistence beyond a week or two. The usefulness of transiently CAR-expressing T cells, which would presumably require multiple infusions to provide meaningful tumor responses but may reduce destruction of normal tissues or prevent T-cell accumulation to levels that increase the risk of cytokine storms, remains to be established.

Another key aspect of CAR delivery is the addressee and identifying which T cells, expanded under which conditions, are better suited for optimal tumor eradication. As stated above, this topic is beyond the scope of this review, recognizing that different T-cell subsets (CD4+ or CD8+ αβ T cells, γδ T cells, naïve T cells, central memory T cells, effector memory T cells, virus-specific T cells, and the recently described stem-like memory T cells; refs. 87, 88) warrant further investigation to delineate whether different CAR designs are best suited for different T-cell types. CARs are also functional in Tregs (89) and in the progeny of transplanted T-cell progenitors (90).

CAR-like TCRs and TCR-like CARs

The transfer of TCRs into T cells poses two particular challenges that CARs elude: the risk of mispairing between endogenous and transduced TCR chains (91) and competition for rate-limiting CD3 complex (92), which is required for TCR signaling. Several approaches have been proposed to prevent TCR chain mispairing, including partial murinization of the constant regions, the addition of disulfide bonds, and altering the knob-in-hole directional interaction between constant regions. Another approach is to add signaling domains to the intracellular portions of the transduced TCR (93), similarly to first-generation CARs, which Sebestyén and colleagues and Roszik and colleagues showed could at once avert TCR mispairing and reduce association with CD3 (93, 94).

Conversely, HLA-peptide complexes can be targeted by antibody structures that mimic TCR recognition. CARs may be advantageous in this regard as they neither interfere nor compete with the native TCR and CD3 and can be further endowed with costimulatory capabilities. Human Fab fragments specific to peptide/MHC molecules have been derived from phage display libraries (95). Although many preferentially bind to MHC (96), some high-affinity Fabs with greater binding affinity for the peptide have been generated and shown by crystallographic analysis to have a binding footprint to MHC/peptide complexes similar to that of TCRs (97). The therapeutic potential and toxicity of these TCR-like CARs remain to be established.

Combinatorial Antigen Recognition

T cells may also be rendered more tumor-selective through combinatorial antigen recognition. We recently described a strategy that integrates combinatorial antigen recognition, split signaling, and, critically, balanced strength of T-cell activation and costimulation to generate T cells that eliminate target cells that express a combination of antigens while sparing cells that express each antigen individually (98). With this approach, T-cell activation requires TCR or CAR-mediated recognition of one antigen, whereas costimulation is independently mediated by a CCR (33) specific for a second antigen. To achieve tumor selectivity, we diminished the efficiency of T-cell activation to a level where it was ineffective without rescue provided by simultaneous CCR recognition of the second antigen (99). Novel approaches to enforce tumor specificity in the face of a dearth of truly unique target antigens are an important direction for future immunotherapies.

Perspectives

While a number of important biologic questions remain to be addressed, including optimizing CAR signaling, defining optimal targets, working out optimal combinatorial strategies, identifying the best and most practical processes for T-cell subset selection and T-cell manufacturing, and reducing T-cell–mediated toxicity and the toxicity of host conditioning, the first clinical successes of CAR therapy are being registered. The prospect of meeting the challenging premise of adoptive T-cell therapy—to achieve specific tumor destruction with one or few T-cell infusions and limited collateral damage to normal tissues—may be within reach. The targeting of B-cell malignancies through CD19 has emerged as the paradigm for the CAR field. At present, it also stands as an exception. The identification of safe targets in a broad range of tumor types, eventually in combinatorial fashion, and harnessing CAR technology for the treatment of solid tumors, are future challenges for all adoptive T-cell therapies including those using CARs. As this review aims to convey, many exciting strategies are in the pipeline, and as many reasons to be optimistic about the prospects for CAR therapies. Providing access to targeted T-cell therapies to all medical centers and their patients will pose additional biologic, logistical, and economic challenges, which are beyond the scope of this review. The fact that models for broad access to targeted T-cell therapies are increasingly discussed is testimony to the therapeutic potential and rising credibility of CARs.

Disclosure of Potential Conflicts of Interest

M. Sadelain and R. Brentjens have ownership interest in a patent on CAR technology. No potential conflicts of interest were disclosed by the other author.

Authors' Contributions

Conception and design: M. Sadelain

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): M. Sadelain, R. Brentjens, I. Rivière

Writing, review, and/or revision of the manuscript: M. Sadelain, R. Brentjens, I. Rivière

Grant Support

This work is supported by MSKCC's Experimental Therapeutics Center, the Terry Fox Foundation, the Alliance for Cancer Gene Therapy, the Mallah Foundation, the Majors Foundation, the Lake Road Foundation, and the Sanders Fund.

References

Sadelain

M

,

Rivière

I

,

Brentjens

R

.

Targeting tumours with genetically enhanced T lymphocytes

.

Nat Rev Cancer

2003

;

3

:

35

45

.

Ho

WY

,

Blattman

JN

,

Dossett

ML

,

Yee

C

,

Greenberg

PD

.

Adoptive immunotherapy: engineering T cell responses as biologic weapons for tumor mass destruction

.

Cancer Cell

2003

;

3

:

431

7

.

Rosenberg

SA

,

Restifo

NP

,

Yang

JC

,

Morgan

RA

,

Dudley

ME

.

Adoptive cell transfer: a clinical path to effective cancer immunotherapy

.

Nat Rev Cancer

2008

;

8

:

299

308

.

Sadelain

M

,

Brentjens

R

,

Rivière

I

.

The promise and potential pitfalls of chimeric antigen receptors

.

Curr Opin Immunol

2009

;

21

:

215

23

.

Zhou

G

,

Levitsky

H

.

Towards curative cancer immunotherapy: overcoming posttherapy tumor escape

.

Clin Dev Immunol

2012

;

2012

:

124187

.

Dustin

ML

,

Depoil

D

.

New insights into the T cell synapse from single molecule techniques

.

Nat Rev Immunol

2011

;

11

:

672

84

.

Bridgeman

JS

,

Hawkins

RE

,

Hombach

AA

,

Abken

H

,

Gilham

DE

.

Building better chimeric antigen receptors for adoptive T cell therapy

.

Curr Gene Ther

2010

;

10

:

77

90

.

Stone

JD

,

Chervin

AS

,

Kranz

DM

.

T-cell receptor binding affinities and kinetics: impact on T-cell activity and specificity

.

Immunology

2009

;

126

:

165

76

.

Edwards

LJ

,

Evavold

BD

.

T cell recognition of weak ligands: roles of signaling, receptor number, and affinity

.

Immunol Res

2011

;

50

:

39

48

.

Irving

BA

,

Weiss

A

.

The cytoplasmic domain of the T cell receptor zeta chain is sufficient to couple to receptor-associated signal transduction pathways

.

Cell

1991

;

64

:

891

901

.

Romeo

C

,

Seed

B

.

Cellular immunity to HIV activated by CD4 fused to T cell or Fc receptor polypeptides

.

Cell

1991

;

64

:

1037

46

.

Letourneur

F

,

Klausner

RD

.

T-cell and basophil activation through the cytoplasmic tail of T-cell-receptor zeta family proteins

.

Proc Natl Acad Sci U S A

1991

;

88

:

8905

9

.

Romeo

C

,

Amiot

M

,

Seed

B

.

Sequence requirements for induction of cytolysis by the T cell antigen/Fc receptor zeta chain

.

Cell

1992

;

68

:

889

97

.

Eshhar

Z

,

Waks

T

,

Gross

G

,

Schindler

DG

.

Specific activation and targeting of cytotoxic lymphocytes through chimeric single chains consisting of antibody-binding domains and the gamma or zeta subunits of the immunoglobulin and T-cell receptors

.

Proc Natl Acad Sci U S A

1993

;

90

:

720

4

.

Cooper

LJ

,

Topp

MS

,

Serrano

LM

,

Gonzalez

S

,

Chang

WC

,

Naranjo

A

, et al

T-cell clones can be rendered specific for CD19: toward the selective augmentation of the graft-versus-B-lineage leukemia effect

.

Blood

2003

;

101

:

1637

44

.

Hwu

P

,

Yang

JC

,

Cowherd

R

,

Treisman

J

,

Shafer

GE

,

Eshhar

Z

, et al

In vivo antitumor activity of T cells redirected with chimeric antibody/T-cell receptor genes

.

Cancer Res

1995

;

55

:

3369

73

.

Chmielewski

M

,

Rappl

G

,

Hombach

AA

,

Abken

H

.

T cells redirected by a CD3zeta chimeric antigen receptor can establish self-antigen-specific tumour protection in the long term

.

Gene Ther

2013

;

20

:

177

86

.

Brocker

T

,

Karjalainen

K

.

Signals through T cell receptor-zeta chain alone are insufficient to prime resting T lymphocytes

.

J Exp Med

1995

;

181

:

1653

9

.

Gong

MC

,

Latouche

JB

,

Krause

A

,

Heston

WD

,

Bander

NH

,

Sadelain

M

.

Cancer patient T cells genetically targeted to prostate-specific membrane antigen specifically lyse prostate cancer cells and release cytokines in response to prostate-specific membrane antigen

.

Neoplasia

1999

;

1

:

123

7

.

Maher

J

,

Brentjens

RJ

,

Gunset

G

,

Rivière

I

,

Sadelain

M

.

Human T-lymphocyte cytotoxicity and proliferation directed by a single chimeric TCRzeta/CD28 receptor

.

Nat Biotechnol

2002

;

20

:

70

5

.

Finney

HM

,

Lawson

AD

,

Bebbington

CR

,

Weir

AN

.

Chimeric receptors providing both primary and costimulatory signaling in T cells from a single gene product

.

J Immunol

1998

;

161

:

2791

7

.

Brentjens

RJ

,

Santos

E

,

Nikhamin

Y

,

Yeh

R

,

Matsushita

M

,

La Perle

K

, et al

Genetically targeted T cells eradicate systemic acute lymphoblastic leukemia xenografts

.

Clin Cancer Res

2007

;

13

:

5426

35

.

Milone

MC

,

Fish

JD

,

Carpenito

C

,

Carroll

RG

,

Binder

GK

,

Teachey

D

, et al

Chimeric receptors containing CD137 signal transduction domains mediate enhanced survival of T cells and increased antileukemic efficacy in vivo

.

Mol Ther

2009

;

17

:

1453

64

.

Kowolik

CM

,

Topp

MS

,

Gonzalez

S

,

Pfeiffer

T

,

Olivares

S

,

Gonzalez

N

, et al

CD28 costimulation provided through a CD19-specific chimeric antigen receptor enhances in vivo persistence and antitumor efficacy of adoptively transferred T cells

.

Cancer Res

2006

;

66

:

10995

–1

1004

.

Savoldo

B

,

Ramos

CA

,

Liu

E

,

Mims

MP

,

Keating

MJ

,

Carrum

G

, et al

CD28 costimulation improves expansion and persistence of chimeric antigen receptor-modified T cells in lymphoma patients

.

J Clin Invest

2011

;

121

:

1822

6

.

Carpenito

C

,

Milone

MC

,

Hassan

R

,

Simonet

JC

,

Lakhal

M

,

Suhoski

MM

, et al

Control of large, established tumor xenografts with genetically retargeted human T cells containing CD28 and CD137 domains

.

Proc Natl Acad Sci U S A

2009

;

106

:

3360

5

.

Zhong

XS

,

Matsushita

M

,

Plotkin

J

,

Rivière

I

,

Sadelain

M

.

Chimeric antigen receptors combining 4-1BB and CD28 signaling domains augment PI3kinase/AKT/Bcl-XL activation and CD8+ T cell-mediated tumor eradication

.

Mol Ther

2010

;

18

:

413

20

.

Pule

MA

,

Straathof

KC

,

Dotti

G

,

Heslop

HE

,

Rooney

CM

,

Brenner

MK

.

A chimeric T cell antigen receptor that augments cytokine release and supports clonal expansion of primary human T cells

.

Mol Ther

2005

;

12

:

933

41

.

Tammana

S

,

Huang

X

,

Wong

M

,

Milone

MC

,

Ma

L

,

Levine

BL

, et al

4-1BB and CD28 signaling plays a synergistic role in redirecting umbilical cord blood T cells against B-cell malignancies

.

Hum Gene Ther

2010

;

21

:

75

86

.

Wang

J

,

Jensen

M

,

Lin

Y

,

Sui

X

,

Chen

E

,

Lindgren

CG

, et al

Optimizing adoptive polyclonal T cell immunotherapy of lymphomas, using a chimeric T cell receptor possessing CD28 and CD137 costimulatory domains

.

Hum Gene Ther

2007

;

18

:

712

25

.

Till

BG

,

Jensen

MC

,

Wang

J

,

Qian

X

,

Gopal

AK

,

Maloney

DG

, et al

CD20-specific adoptive immunotherapy for lymphoma using a chimeric antigen receptor with both CD28 and 4-1BB domains: pilot clinical trial results

.

Blood

2012

;

119

:

3940

50

.

Stephan

MT

,

Ponomarev

V

,

Brentjens

RJ

,

Chang

AH

,

Dobrenkov

KV

,

Heller

G

, et al

T cell-encoded CD80 and 4-1BBL induce auto- and transcostimulation, resulting in potent tumor rejection

.

Nat Med

2007

;

13

:

1440

9

.

Krause

A

,

Guo

HF

,

Latouche

JB

,

Tan

C

,

Cheung

NK

,

Sadelain

M

.

Antigen-dependent CD28 signaling selectively enhances survival and proliferation in genetically modified activated human primary T lymphocytes

.

J Exp Med

1998

;

188

:

619

26

.

Wilkie

S

,

van Schalkwyk

MC

,

Hobbs

S

,

Davies

DM

,

van der Stegen

SJ

,

Pereira

AC

, et al

Dual targeting of ErbB2 and MUC1 in breast cancer using chimeric antigen receptors engineered to provide complementary signaling

.

J Clin Immunol

2012

;

32

:

1059

70

.

Duong

CP

,

Westwood

JA

,

Berry

LJ

,

Darcy

PK

,

Kershaw

MH

.

Enhancing the specificity of T-cell cultures for adoptive immunotherapy of cancer

.

Immunotherapy

2011

;

3

:

33

48

.

Hsu

C

,

Hughes

MS

,

Zheng

Z

,

Bray

RB

,

Rosenberg

SA

,

Morgan

RA

.

Primary human T lymphocytes engineered with a codon-optimized IL-15 gene resist cytokine withdrawal-induced apoptosis and persist long-term in the absence of exogenous cytokine

.

J Immunol

2005

;

175

:

7226

34

.

Quintarelli

C

,

Vera

JF

,

Savoldo

B

,

Giordano Attianese

GM

,

Pule

M

,

Foster

AE

, et al

Co-expression of cytokine and suicide genes to enhance the activity and safety of tumor-specific cytotoxic T lymphocytes

.

Blood

2007

;

110

:

2793

802

.

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

.

Foster

AE

,

Leen

AM

,

Lee

T

,

Okamura

T

,

Lu

A

,

Vera

J

, et al

Autologous designer antigen-presenting cells by gene modification of T lymphocyte blasts with IL-7 and IL-12

.

J Immunother

2007

;

30

:

506

16

.

Kaka

AS

,

Shaffer

DR

,

Hartmaier

R

,

Leen

AM

,

Lu

A

,

Bear

A

, et al

Genetic modification of T cells with IL-21 enhances antigen presentation and generation of central memory tumor-specific cytotoxic T-lymphocytes

.

J Immunother

2009

;

32

:

726

36

.

Markley

JC

,

Sadelain

M

.

IL-7 and IL-21 are superior to IL-2 and IL-15 in promoting human T cell-mediated rejection of systemic lymphoma in immunodeficient mice

.

Blood

2010

;

115

:

3508

19

.

Pegram

HJ

,

Lee

JC

,

Hayman

EG

,

Imperato

GH

,

Tedder

TF

,

Sadelain

M

, et al

Tumor-targeted T cells modified to secrete IL-12 eradicate systemic tumors without need for prior conditioning

.

Blood

2012

;

119

:

4133

41

.

Kerkar

SP

,

Muranski

P

,

Kaiser

A

,

Boni

A

,

Sanchez-Perez

L

,

Yu

Z

, et al

Tumor-specific CD8 +T cells expressing interleukin-12 eradicate established cancers in lymphodepleted hosts

.

Cancer Res

2010

;

70

:

6725

34

.

Chinnasamy

D

,

Yu

Z

,

Kerkar

SP

,

Zhang

L

,

Morgan

RA

,

Restifo

NP

, et al

Local delivery of interleukin-12 using T cells targeting VEGF receptor-2 eradicates multiple vascularized tumors in mice

.

Clin Cancer Res

2012

;

18

:

1672

83

.

Kerkar

SP

,

Goldszmid

RS

,

Muranski

P

,

Chinnasamy

D

,

Yu

Z

,

Reger

RN

, et al

IL-12 triggers a programmatic change in dysfunctional myeloid-derived cells within mouse tumors

.

J Clin Invest

2011

;

121

:

4746

57

.

Ponomarev

V

,

Doubrovin

M

,

Lyddane

C

,

Beresten

T

,

Balatoni

J

,

Bornman

W

, et al

Imaging TCR-dependent NFAT-mediated T-cell activation with positron emission tomography in vivo

.

Neoplasia

2001

;

3

:

480

8

.

Li

YS

,

Wasserman

R

,

Hayakawa

K

,

Hardy

RR

.

Identification of the earliest B lineage stage in mouse bone marrow

.

Immunity

1996

;

5

:

527

35

.

Li

YS

,

Hayakawa

K

,

Hardy

RR

.

The regulated expression of B lineage associated genes during B cell differentiation in bone marrow and fetal liver

.

J Exp Med

1993

;

178

:

951

60

.

Brentjens

RJ

,

Latouche

JB

,

Santos

E

,

Marti

F

,

Gong

MC

,

Lyddane

C

, et al

Eradication of systemic B-cell tumors by genetically targeted human T lymphocytes co-stimulated by CD80 and interleukin-15

.

Nat Med

2003

;

9

:

279

86

.

Kohn

DB

,

Dotti

G

,

Brentjens

R

,

Savoldo

B

,

Jensen

M

,

Cooper

LJ

, et al

CARs on track in the clinic

.

Mol Ther

2011

;

19

:

432

8

.

Kochenderfer

JN

,

Wilson

WH

,

Janik

JE

,

Dudley

ME

,

Stetler-Stevenson

M

,

Feldman

SA

, et al

Eradication of B-lineage cells and regression of lymphoma in a patient treated with autologous T cells genetically engineered to recognize CD19

.

Blood

2010

;

116

:

4099

102

.

Brentjens

RJ

,

Rivière

I

,

Park

JH

,

Davila

ML

,

Wang

X

,

Stefanski

J

, et al

Safety and persistence of adoptively transferred autologous CD19-targeted T cells in patients with relapsed or chemotherapy refractory B-cell leukemias

.

Blood

2011

;

118

:

4817

28

.

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

.

Kalos

M

,

Levine

BL

,

Porter

DL

,

Katz

S

,

Grupp

SA

,

Bagg

A

, et al

T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia

.

Sci Transl Med

2011

;

3

:

95ra73

.

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

.

Jensen

MC

,

Popplewell

L

,

Cooper

LJ

,

DiGiusto

D

,

Kalos

M

,

Ostberg

JR

, et al

Antitransgene rejection responses contribute to attenuated persistence of adoptively transferred CD20/CD19-specific chimeric antigen receptor redirected T cells in humans

.

Biol Blood Marrow Transplant

2010

;

16

:

1245

56

.

Davila

ML

,

Brentjens

R

,

Wang

X

,

Rivière

I

,

Sadelain

M

.

How do CARs work? Early insights from recent clinical studies targeting CD19

.

Oncoimmunology

2012

;

1

:

1

7

.

Cooper

LJ

,

Jena

B

,

Bollard

CM

.

Good T cells for bad B cells

.

Blood

2012

;

119

:

2700

2

.

Kershaw

MH

,

Westwood

JA

,

Parker

LL

,

Wang

G

,

Eshhar

Z

,

Mavroukakis

SA

, et al

A phase I study on adoptive immunotherapy using gene-modified T cells for ovarian cancer

.

Clin Cancer Res

2006

;

12

:

6106

15

.

Louis

CU

,

Savoldo

B

,

Dotti

G

,

Pule

M

,

Yvon

E

,

Myers

GD

, et al

Antitumor activity and long-term fate of chimeric antigen receptor-positive T cells in patients with neuroblastoma

.

Blood

2011

;

118

:

6050

6

.

Morgan

RA

,

Yang

JC

,

Kitano

M

,

Dudley

ME

,

Laurencot

CM

,

Rosenberg

SA

.

Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2

.

Mol Ther

2010

;

18

:

843

51

.

Silver

DA

,

Pellicer

I

,

Fair

WR

,

Heston

WD

,

Cordon-Cardo

C

.

Prostate-specific membrane antigen expression in normal and malignant human tissues

.

Clin Cancer Res

1997

;

3

:

81

5

.

Hudecek

M

,

Schmitt

TM

,

Baskar

S

,

Lupo-Stanghellini

MT

,

Nishida

T

,

Yamamoto

TN

, et al

The B-cell tumor-associated antigen ROR1 can be targeted with T cells modified to express a ROR1-specific chimeric antigen receptor

.

Blood

2010

;

116

:

4532

41

.

Seaman

BJ

,

Guardiani

EA

,

Brewer

CC

,

Zalewski

CK

,

King

KA

,

Rudy

S

, et al

Audiovestibular dysfunction associated with adoptive cell immunotherapy for melanoma

.

Otolaryngol Head Neck Surg

2012

;

147

:

744

9

.

Lamers

CH

,

Sleijfer

S

,

Vulto

AG

,

Kruit

WH

,

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

.

Grupp

SA

,

Porter

DL

,

Teachey

D

,

Barrett

DM

,

Chew

A

,

Suppa

E

, et al

CD19-Redirected chimeric antigen receptor T (CART19) cells induce a cytokine release syndrome (CRS) and induction of treatable macrophage activation syndrome (MAS) that can be managed by the IL-6 antagonist tocilizumab (toc) [abstract]

. In:

Proceedings of the 54th ASH Annual Meeting and Exposition; 2012 Dec 8–11

;

Atlanta, GA. Washington, DC

:

ASH

;

2012

.

Abstract nr 2604

.

Lupo-Stanghellini

MT

,

Provasi

E

,

Bondanza

A

,

Ciceri

F

,

Bordignon

C

,

Bonini

C

.

Clinical impact of suicide gene therapy in allogeneic hematopoietic stem cell transplantation

.

Hum Gene Ther

2010

;

21

:

241

50

.

Di Stasi

A

,

Tey

SK

,

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

.

Sadelain

M

.

Eliminating cells gone astray

.

N Engl J Med

2011

;

365

:

1735

7

.

Brentjens

RJ

,

Davila

ML

,

Riviére

I

,

Park

J

,

Wang

X

,

Cowell

LG

, et al

CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia

.

Sci Transl Med

2013

;

5

:

177ra38

.

Mavilio

F

,

Ferrari

G

,

Rossini

S

,

Nobili

N

,

Bonini

C

,

Casorati

G

, et al

Peripheral blood lymphocytes as target cells of retroviral vector-mediated gene transfer

.

Blood

1994

;

83

:

1988

97

.

Bunnell

BA

,

Muul

LM

,

Donahue

RE

,

Blaese

RM

,

Morgan

RA

.

High-efficiency retroviral-mediated gene transfer into human and nonhuman primate peripheral blood lymphocytes

.

Proc Natl Acad Sci U S A

1995

;

92

:

7739

43

.

Gallardo

HF

,

Tan

C

,

Ory

D

,

Sadelain

M

.

Recombinant retroviruses pseudotyped with the vesicular stomatitis virus G glycoprotein mediate both stable gene transfer and pseudotransduction in human peripheral blood lymphocytes

.

Blood

1997

;

90

:

952

7

.

Suerth

JD

,

Schambach

A

,

Baum

C

.

Genetic modification of lymphocytes by retrovirus-based vectors

.

Curr Opin Immunol

2012

;

24

:

598

608

.

Kustikova

OS

,

Schiedlmeier

B

,

Brugman

MH

,

Stahlhut

M

,

Bartels

S

,

Li

Z

, et al

Cell-intrinsic and vector-related properties cooperate to determine the incidence and consequences of insertional mutagenesis

.

Mol Ther

2009

;

17

:

1537

47

.

Rivière

I

,

Dunbar

CE

,

Sadelain

M

.

Hematopoietic stem cell engineering at a crossroads

.

Blood

2012

;

119

:

1107

16

.

Newrzela

S

,

Cornils

K

,

Li

Z

,

Baum

C

,

Brugman

MH

,

Hartmann

M

, et al

Resistance of mature T cells to oncogene transformation

.

Blood

2008

;

112

:

2278

86

.

Recchia

A

,

Bonini

C

,

Magnani

Z

,

Urbinati

F

,

Sartori

D

,

Muraro

S

, et al

Retroviral vector integration deregulates gene expression but has no consequence on the biology and function of transplanted T cells

.

Proc Natl Acad Sci U S A

2006

;

103

:

1457

62

.

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

.

Scholler

J

,

Brady

TL

,

Binder-Scholl

G

,

Hwang

WT

,

Plesa

G

,

Hege

KM

, et al

Decade-long safety and function of retroviral-modified chimeric antigen receptor T cells

.

Sci Transl Med

2012

;

4

:

132ra53

.

Huang

X

,

Wilber

A

,

McIvor

RS

,

Zhou

X

.

DNA transposons for modification of human primary T lymphocytes

.

Methods Mol Biol

2009

;

506

:

115

26

.

Jin

Z

,

Maiti

S

,

Huls

H

,

Singh

H

,

Olivares

S

,

Mates

L

, et al

The hyperactive Sleeping Beauty transposase SB100X improves the genetic modification of T cells to express a chimeric antigen receptor

.

Gene Ther

2011

;

18

:

849

56

.

Sadelain

M

,

Papapetrou

EP

,

Bushman

FD

.

Safe harbours for the integration of new DNA in the human genome

.

Nat Rev Cancer

2012

;

12

:

51

8

.

Zhao

Y

,

Zheng

Z

,

Cohen

CJ

,

Gattinoni

L

,

Palmer

DC

,

Restifo

NP

, et al

High-efficiency transfection of primary human and mouse T lymphocytes using RNA electroporation

.

Mol Ther

2006

;

13

:

151

9

.

Birkholz

K

,

Hombach

A

,

Krug

C

,

Reuter

S

,

Kershaw

M

,

Kampgen

E

, et al

Transfer of mRNA encoding recombinant immunoreceptors reprograms CD4+ and CD8+ T cells for use in the adoptive immunotherapy of cancer

.

Gene Ther

2009

;

16

:

596

604

.

Urbanska

K

,

Lanitis

E

,

Poussin

M

,

Lynn

RC

,

Gavin

BP

,

Kelderman

S

, et al

A universal strategy for adoptive immunotherapy of cancer through use of a novel T-cell antigen receptor

.

Cancer Res

2012

;

72

:

1844

52

.

Gattinoni

L

,

Lugli

E

,

Ji

Y

,

Pos

Z

,

Paulos

CM

,

Quigley

MF

, et al

A human memory T cell subset with stem cell-like properties

.

Nat Med

2011

;

17

:

1290

7

.

Cieri

N

,

Camisa

B

,

Cocchiarella

F

,

Forcato

M

,

Oliveira

G

,

Provasi

E

, et al

IL-7 and IL-15 instruct the generation of human memory stem T cells from naive precursors

.

Blood

2013

;

121

:

573

84

.

Lee

JC

,

Hayman

E

,

Pegram

HJ

,

Santos

E

,

Heller

G

,

Sadelain

M

, et al

In vivo inhibition of human CD19-targeted effector T cells by natural T regulatory cells in a xenotransplant murine model of B cell malignancy

.

Cancer Res

2011

;

71

:

2871

81

.

Zakrzewski

JL

,

Suh

D

,

Markley

JC

,

Smith

OM

,

King

C

,

Goldberg

GL

, et al

Tumor immunotherapy across MHC barriers using allogeneic T-cell precursors

.

Nat Biotechnol

2008

;

26

:

453

61

.

Bendle

GM

,

Linnemann

C

,

Hooijkaas

AI

,

Bies

L

,

de Witte

MA

,

Jorritsma

A

, et al

Lethal graft-versus-host disease in mouse models of T cell receptor gene therapy

.

Nat Med

2010

;

16

:

565

70

.

Okamoto

S

,

Mineno

J

,

Ikeda

H

,

Fujiwara

H

,

Yasukawa

M

,

Shiku

H

, et al

Improved expression and reactivity of transduced tumor-specific TCRs in human lymphocytes by specific silencing of endogenous TCR

.

Cancer Res

2009

;

69

:

9003

11

.

Sebestyen

Z

,

Schooten

E

,

Sals

T

,

Zaldivar

I

,

San Jose

E

,

Alarcon

B

, et al

Human TCR that incorporate CD3zeta induce highly preferred pairing between TCRalpha and beta chains following gene transfer

.

J Immunol

2008

;

180

:

7736

46

.

Roszik

J

,

Sebestyen

Z

,

Govers

C

,

Guri

Y

,

Szoor

A

,

Palyi-Krekk

Z

, et al

T-cell synapse formation depends on antigen recognition but not CD3 interaction: studies with TCR:zeta, a candidate transgene for TCR gene therapy

.

Eur J Immunol

2011

;

41

:

1288

97

.

Denkberg

G

,

Reiter

Y

.

Recombinant antibodies with T-cell receptor-like specificity: novel tools to study MHC class I presentation

.

Autoimmun Rev

2006

;

5

:

252

7

.

Hulsmeyer

M

,

Chames

P

,

Hillig

RC

,

Stanfield

RL

,

Held

G

,

Coulie

PG

, et al

A major histocompatibility complex-peptide-restricted antibody and t cell receptor molecules recognize their target by distinct binding modes: crystal structure of human leukocyte antigen (HLA)-A1-MAGE-A1 in complex with FAB-HYB3

.

J Biol Chem

2005

;

280

:

2972

80

.

Stewart-Jones

G

,

Wadle

A

,

Hombach

A

,

Shenderov

E

,

Held

G

,

Fischer

E

, et al

Rational development of high-affinity T-cell receptor-like antibodies

.

Proc Natl Acad Sci U S A

2009

;

106

:

5784

8

.

Kloss

CC

,

Condomines

M

,

Cartellieri

M

,

Bachmann

M

,

Sadelain

M

.

Combinatorial antigen recognition with balanced signaling promotes selective tumor eradication by engineered T cells

.

Nat Biotechnol

2012

;

31

:

71

5

.

Hanada

K

,

Restifo

NP

.

Double or nothing on cancer immunotherapy

.

Nat Biotechnol

2013

;

31

:

33

4

.

©2013 American Association for Cancer Research.

2013

Supplementary data