Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Aug 8.
Published in final edited form as: Curr Opin Immunol. 2009 Mar 25;21(2):215–223. doi: 10.1016/j.coi.2009.02.009

The promise and potential pitfalls of chimeric antigen receptors

Michel Sadelain 1, Renier Brentjens 1, Isabelle Rivière 1
PMCID: PMC5548385  NIHMSID: NIHMS886545  PMID: 19327974

Abstract

One important purpose of T cell engineering is to generate tumor-targeted T cells through the genetic transfer of antigen-specific receptors, which consist of either physiological, MHC-restricted T cell receptors (TCRs) or non MHC-restricted chimeric antigen receptors (CARs). CARs combine antigen-specificity and T cell activating properties in a single fusion molecule. First generation CARs, which included as their signaling domain the cytoplasmic region of the CD3ζ or Fc receptor γ chain, effectively redirected T cell cytotoxicity but failed to enable T cell proliferation and survival upon repeated antigen exposure. Receptors encompassing both CD28 and CD3ζ are the prototypes for second generation CARs, which are now rapidly expanding to a diverse array of receptors with different functional properties. First generation CARs have been tested in phase I clinical studies in patients with ovarian cancer, renal cancer, lymphoma, and neuroblastoma, where they have induced modest responses. Second generation CARs, which are just now entering the clinical arena in the B cell malignancies and other cancers, will provide a more significant test for this approach. If the immunogenicity of CARs can be averted, the versatility of their design and HLA-independent antigen recognition will make CARs tools of choice for T cell engineering for the development of targeted cancer immunotherapies.

Introduction

The advent of effective methods for gene transfer in T cells provides a new means for rapidly generating tumor-specific T cells. T cell engineering also offers a unique means to overcome or circumvent the tolerance mechanisms and immune escape stratagems used by tumors to derail or elude immune responses [13]. In principle, genetic reprogramming can be used to enforce tumor antigen recognition, to improve T cell survival, augment T cell expansion, generate memory lymphocytes and offset T cell death, anergy, and immune suppression. Furthermore, the genetic modification of T cells can be employed to enable the tracking of T cell migration in vivo and introduce into T cells a safety or recall mechanism to curb T cell responses if needed. The first objective – to afford tumor antigen recognition – is achieved by expressing antigen receptors, which consist of either physiological, MHC-restricted T cell receptors (TCRs) or non-MHC-restricted chimeric antigen receptors (CARs). The latter are the focus of this review.

The first T cell activating receptors can be traced back to the CD3ζ chain fusions that were generated to elucidate the role of the ζ chain [4,5]. These studies showed that cross-linking these fusion receptors was sufficient to provide calcium influx and T cell activation signaling including the initiation of cytotoxicity. Eshhar et al. directed such fusion receptors toward haptens by incorporating an immunoglobulin-derived scFv in the extracellular domain of the chimeric receptors, thus enabling the T cells expressing these ‘T bodies’ to lyse hapten-coated cells [6]. Several groups subsequently confirmed the ability to redirect T cell cytotoxicity using receptors encompassing different scFv’s fused to the CD3ζ or Fc receptor γ (FcRγ) cytoplasmic signaling domains. However, as reviewed below, it was not until costimulatory properties were incorporated into the next generation of CARs that a greater strength and quality of antigen-induced signaling could be provided to T cells, which then enabled T cell proliferation and survival upon repeated exposure to antigen. Following this turning point, an impressive array of rapidly developing second generation CARs has been developed and is under intense investigation. Only first generation vectors have been tested in phase I clinical trials, showing so far modest effects. Second generation CARs have just entered the clinical arena.

CARs: the rules of engagement

Most CARs utilize an antibody-derived antigen-binding motif to recognize antigen (Table 1). Others utilize receptor or ligand domains as their targeting moiety, such as heregulin [7] or IL13 [8], that bind to their cognate ligand or receptor counterpart (Table 1). In all of these instances, CARs recognize native cell-surface antigens independently of antigen processing or MHC-restricted presentation. Importantly, CARs therefore do not have to be matched to the patient HLA and can recognize tumors that have downregulated HLA expression [9,10]. The expanding range of CAR specificities is illustrated in Table 1. The cell-surface antigens targeted by CARs include proteins, carbohydrates, and glycolipids. Most current CARs incorporate an scFv derived from a murine monoclonal antibody. The scFv’s are typically cloned from hybridoma RNA, but may also be selected from phage display libraries.

Table 1.

Tumor antigens and CAR investigated in vitro and in vivo in T lymphocytes

Target antigen Associated malignancy Receptor type (other specificity) In vivo studies Reference
α-Folate receptor Ovarian cancer ScFv-FcεRIγ Phase I [41]
CAIX Renal cell carcinoma ScFv-FcεRIγ [4547]
ScFv-CD4-FcεRIγ Phase I [42,48]
CD19 B cell malignancies ScFv-CD3ζ (EBV) [32]
B cell malignancies ScFv-CD3ζ + [31,49]
B cell malignancies ScFv-CD28-CD3ζ + [25,28,44]
Refractory Follicular Lymphoma ScFv-CD3ζ Phase I [50]
CLL ScFv-CD28-CD3ζ Phase I [51]
B cell malignancies ScFv-CD28-CD3ζ + [27,52]
ALL ScFv-41BB-CD3z [53]
ALL ScFv-41BB-CD3z + [54]
B cell malignancies ScFv-CD3ζ (Influenza MP-1) + [55]
B cell malignancies ScFv-CD3ζ (VZV) [56]
CD20 Lymphomas ScFv-CD28-CD3ζ [57]
B cell malignancies ScFv-CD4-CD3ζ [58]
B cell lymphomas ScFv-CD3ζ [59,60]
Mantle cell lymphoma, ScFv-CD3ζ Phase I [43]
indolent B cell lymphomas ScFv-CD28-CD3ζ [23]
ScFv-CD28-41BB-CD3ζ [23]
CD22 B cell malignancies ScFV-CD4-CD3ζ [58]
CD30 Lymphomas ScFv-FcεRIγ [61]
Hodgkin lymphoma ScFv-CD3ζ (EBV) + [62]
CD33 AML ScFv-CD28-CD3z [63]
ScFv-41BB-CD3z
CD44v7/8 Cervical carcinoma ScFv-CD8-CD3z + [64]
CEA Colorectal cancer ScFv-CD3ζ + [6567], [68]
ScFv-FcεRIγ + [68,69]
ScFv-CD3ε [70]
ScFv-CD28-CD3ζ [71]
ScFv-CD28-CD3ζ + [72,73]
EGP-2 Multiple malignancies scFv-CD3ζ [74]
scFv-FcεRIγ [74,75]
EGP-40 Colorectal cancer scFv-FcεRIγ [76]
erb-B2 Breast and others ScFv-CD28-CD3ζ + [19,77]
ScFv-CD28-CD3ζ (Influenza) + [78]
ScFv-CD28mut.-CD3ζ + [29]
Prostate cancer ScFv-FcεRIγ + [79]
erb-B 2,3,4 Breast and others Heregulin-CD3ζ [80], [7]
ScFv-CD3ζ + [81]
FBP Ovarian cancer ScFv-FcεRIγ + [35,82,83]
ScFv-FcεRIγ (alloantigen) + [84]
Fetal acethylcholine receptor Rhabdomyosarcoma ScFv-CD3ζ [85]
GD2 Neuroblastoma ScFv-CD28 [86]
ScFv-CD3ζ [32]
ScFv-CD3ζ Phase I [37]
ScFv-CD28-OX40-CD3ζ [22]
ScFv-CD3ζ (VZV) [56]
GD3 Melanoma ScFv-CD3ζ [87]
ScFv-CD3ε [87]
Her-2 Medulloblastoma ScFv-CD3ζ + [88]
IL-13R-a2 Glioma IL-13-CD28-4-1BB-CD3ζ + [89]
Glioblastoma IL-13-CD3ζ + [8,90]
Medulloblastoma IL-13-CD3ζ + [91]
KDR Tumor neovasculature ScFv-FcεRIγ [92]
κ-light chain B cell malignancies (B-NHL, CLL) ScFv-CD3ζ + [30]
ScFv-CD28-CD3ζ + [30]
LeY Carcinomas ScFv-FcεRIγ [93]
Epithelial derived tumors ScFv-CD28-CD3ζ + [94]
L1 cell adhesion molecule Neuroblastoma ScFv-CD3ζ Phase I [95,96]
MAGE-A1 Melanoma ScFV-CD4-FcεRIγ [97]
ScFV-CD28-FcεRIγ
Murine CMV infected cells Murine CMV Ly49H-CD3ζ + [98]
MUC1 Breast, Ovary ScFV-CD28-OX40-CD3ζ + [16]
NKG2D ligands Various tumors NKG2D-CD3ζ + [99101]
Oncofetal antigen (h5T4) Various tumors ScFV-CD3ζ (vaccination) + [102]
PSCA Prostate carcinoma ScFv-b2c-CD3ζ [103]
PSMA Prostate/tumor vasculature ScFv-CD3ζ + [18,104,39]
ScFv-CD28-CD3ζ [21]
ScFv-CD3ζ + [105]
TAA targeted by mAb IgE Various tumors FcεRI-CD28-CD3ζ (+ a-TAA IgE mAb) + [106]
TAG-72 Adenocarcinomas scFv-CD3ζ + [107,108]
VEGF-R2 Tumor neovasculature scFv-CD3ζ [109]

(−) In vitro studies; (+) in vivo animal studies; (Phase I) Phase I clinical trial.

The rules for identifying the best target molecules and corresponding scFv are not yet fully elucidated. Tumor-restricted targets are preferred, but rare. Most well-defined targets are differentiation antigens or cancer/testis antigens, the selection of which depends on the level and frequency of expression on malignant tissues – including cancer stem cells – and their normal counterparts. One may reasonably assume that highly expressed tumor antigens will make better targets, especially if expression is greater on the tumor cells than on normal cells. The threshold antigen density required for optimal CAR-mediated tumor eradication is currently not known.

The optimal range of a CAR’s affinity for its target antigen has not been defined either. Whereas the physiological TCRs have affinities in the micromolar range, monoclonal antibodies and scFv’s operate in the nanomolar range. Heterodimeric TCRs, however, are coupled to the CD3 complex, a multichain complex optimized to bolster T cell activation following TCR engagement. CARs typically do not associate with the CD3 complex, and it is therefore conceivable that their higher affinity relative to TCRs is important to compensate, partly, for this disadvantage. It is noteworthy that too high an affinity can be detrimental to TCR-mediated antigen recognition [11], which highlights the importance of achieving optimal antigen receptor affinity [12]. One study on CAR affinity reveals the complex relationship been CAR density, antigen density, and CAR affinity [13]. CAR expression levels furthermore impact on T cell effector function [14].

Finally, another emerging factor in choosing an optimal scFv for making a CAR is the position of the epitope on the target molecule. The distance of the epitope to the cell surface appears to matter, as do the length and flexibility of the CAR extracellular hinge region [15,16]. These studies suggest that the CAR-antigen interaction must follow at least some of the structural requirements that apply to TCR-based immune synapses. Optimal interactions may further depend on the transmembrane and cytoplasmic components of the CAR, as well as its monomeric or dimeric structure.

Achieving meaningful signaling with second generation CARs

The first CARs were reported as receptors capable of redirecting the cytotoxic activity of CTL clones and hybridomas [17]. Noticing the lack of data on CAR-induced T cell proliferation, we set out to investigate the proliferative response of zeta chain based CARs in primary T cells. Having expressed a receptor specific for human prostate-specific membrane antigen (PSMA) in mitogen-activated peripheral blood T cells, we found that coculture with PSMA-positive LnCAP cells did allow for initiation of proliferation (typically 2–3 cell divisions), but this was soon followed by T cell death [18]. T cell death could be prevented when a costimulatory signal was coincidentally delivered, which could be achieved by expressing the CD28-ligand B7.1/CD80 on the tumor cells. CAR-transduced T cells activated in this manner were successfully reactivated by a second exposure to antigen, resulting in an absolute increase in T cell number [18]. However, most tumor cells, especially tumor cells in vivo, will not express activating costimulatory ligands such as CD80. Furthermore, it is to be expected that CAR-redirected T cells will not engage cross-presented antigen and thus will not have the benefit of dendritic cell-provided costimulation. CARs therefore had to be designed to provide a costimulatory ersatz to address their costimulatory dependence.

The emergence of CARs enabling T cells to survive repeated antigenic stimulation came with the development of CD28-CD3ζ dual-signaling receptors [1921]. These receptors increased IL-2 secretion in response to antigen and permitted absolute expansion of retargeted T cells in response to antigen in the absence of exogenous costimulation [21]. A number of CD28-CD3ζ fusion receptors have been reported (Table 2), but it is noteworthy that not all of them increase IL-2 secretion [22]. This is possibly due to the different construction designs, which utilize different domains and fusion points. Fusion to different scFv’s may also account for different signaling patterns. A side-by-side comparison of different receptors expressed at similar level in the same cells type would be needed to adequately compare different fusion receptors. Significant functional differences between CARs are likely to be found.

Table 2.

CAR cytoplasmic signaling domains investigated in T cells.

Cytoplasmic signaling domain(s) Selected references
First generation (single signaling domain) CD3 ζ [49,58,87,89,31,18,32]
FcεRIγ [69,75,76,82,92,61,35]
CD3ε [70,87]
Second generation (dual signaling domains) CD28-CD3ζ [24,73,106,72,19,110,21,111,25]
CD134-CD3 ζ [25,112,22]
CD137-CD3ζ [25,112,53]
ICOS-CD3ζ [112]
DAP10-CD3ζ [25]
Third generation (three signaling domains) CD28-CD3ζ-Lck [113,114]
CD28-CD134-CD3ζ [16,114]
CD28-CD137-CD3ζ [23]

In recent years, additional fusion receptors have been reported. Several costimulatory signaling domains, including 4-1BB, OX40, DAP10, and ICOS, have been studied (Table 2). The rationale for each one will not be reviewed here, but the general goal has been to extend the strength of signal afforded by the CAR, augment T cell effector function or extend T cell survival. Many of these newer fusions have not been investigated as extensively as theCD28-CD3ζ receptors and it is fair to say that the jury is not out yet on the relative merits of these different fusions.

More recently, triple-fusion receptors that encompass CD3ζ, CD28, and 4-1BB or OX40 signaling motifs have been reported. These receptors appear to enhance in vitro effector functions relative to the dual-fusion receptors [16,22,23], as well as the strength of PI3kinase/Akt activation initiated by contact with antigen (XS Zhong and M Sadelain, unpublished data). These are promising receptors but more studies are needed, including in vivo studies, to assess their therapeutic potential. What is clear is that second generation CARs have considerably superior signaling properties compared with their CD3ζ and FCγR forbearers, which opens up real perspectives for the therapeutic use of CARs.

The rising in vivo prowess of CARs

A subset of all described CARs has been evaluated in in vivo tumor models, investigating either murine or human T cells in xenogeneic tumor models. First and second generation CARs targeting a variety of antigens have been shown to at least delay tumor progression in some animal models (ranging from intraperitoneal cytotoxicity assays to the more convincing systemic models) or in some instances to induce durable remissions in mice bearing established systemic tumors (Table 1). Whereas some first generation CARs have been shown to induce significant responses after intravenous infusion in tumor-bearing mice, it is noteworthy that in every instance where first and second generation CARs were compared, the latter outperformed the former [19,2430]. Triple fusion receptors are active in vivo [16], but comparisons to second generation CARs have not yet been reported.

In xenogeneic models, we showed that human T cells targeted against human CD19 could eradicate systemic Raji tumors (a CD19+ Burkitt lymphoma) in SCID mice [31]. Importantly, tumor eradication was obtained following a single intravenous infusion of human T cells in the absence of post-infusion cytokine administration to support T cell function. This study also demonstrated that the in vivo activity of CAR-transduced T cells depended on the T cell expansion conditions. Thus, T cells activated in the presence of antigen and CD28-mediated costimulation were more effective than T cells expanded by OKT3 antibody and IL-2 (the latter only survived a few days in vivo). Furthermore, T cells activated by artificial antigen-presenting cells expressing the CD19 antigen and CD80 were more effective after expansion in the presence of IL-15 and IL-2 than with IL-2 alone [31]. The crucial impact of in vitro T cell activation on the outcome of adoptive T cell therapy should be borne in mind when comparing studies. CARs should thus be compared side-by-side under the same expansion conditions to permit valid receptor comparisons.

Other factors affect CAR function and CAR comparisons. The tumor obviously makes a difference. Thus, eradication of CD19+ Raji tumor cells is easier to achieve than eradication of CD19+ pre-B acute lymphoblastic leukemia cells [25,31]. This is partly due to the different costimulatory profile of the two tumors [25,31], but additional factors probably come into play as well, including tissue tropism and intrinsic susceptibility to lysis. Finally, the mouse model also makes a difference. Tumor eradication requires fewer infused T cells in NOD/SCIDγc −/− mice than in SCID-beige mice (J Markley and M Sadelain, unpublished data). The impact of these three parameters – T cell expansion conditions, tumor characteristics, and mouse model – should be taken into account when comparing results obtained with different receptors in murine models.

Another important question is what cells are better suited for delivering CAR therapy. CARs have been investigated in bulk mouse spleen cells or bulk human peripheral blood T cells, as well as in EBV-specific T cells [32], lymphoid progenitor cells [33,34], unfractionated or Lin– Sca1+ bone marrow cells [14,35]. An important debate in adoptive cell therapies, which applies to CAR therapies like other adoptive cell therapies, is to better define the advantages and disadvantages of naïve, memory, and virus-specific T cells types [3638]. This is addressed in another article in this issue. It is clear for CAR therapy, as it is for other adoptive T cell therapies, that T cell persistence is an important factor for successful tumor eradication [27,31,39,40] and that the choice of T cell subset to utilize is one of the important aspects of this therapy.

Finally, it should be noted that other immune cell types than T cells are being investigated using CARs (Table 3).

Table 3.

CAR expression in effectors of the innate immune system

Cell type Cytoplasmic signaling domain(s) References
NK cells CD3ζ [115119]
CD28-CD3ζ [120,121]
DAP10 [116]
CD137-CD3ζ [116]
Fc-γ-receptor [119]
Cytokine-induced killer cells (CIK) CD3ζ [122,123]
DAP10 [123]
CD137-CD3ζ [123]
CD28-CD3ζ [123]
Monocytes CD64 (Fc-γ-receptor) [124]
Neutrophils Fc-γ-receptor [119]
CD3ζ [119]

Clinical studies utilizing first generation CARs

Completed clinical studies are limited to phase I studies evaluating first generation CARs targeting the folate receptor in ovarian cancer [41], carbonic anhydrase in renal cancer [42], CD20 in lymphoma, [43]) and GD2 in neuroblastoma [37]. The clinical responses have overall been very modest, with the exception of one partial response in the neuroblastoma study. Immunogenicity of CARs was observed in the first two studies, but not the latter two. The renal carcinoma study had to be halted after three patients developed unanticipated cholestasis, an on-target effect due to the high expression of the targeted antigen, carbonic anhydrase, in biliary epithelium. This study has now resumed with the added use of G250 antibody to partially mask the biliary antigen before T cell infusion (C Lamers, unpublished data). It is noteworthy that CAR-directed T cells were active against target antigen-positive T cells that are not typically picked up in imaging studies utilizing the parental monoclonal antibody, lending support to the merits of T cell based therapy.

These studies utilized first generation CARs and suboptimal T cell expansion procedures such as OKT3-mediated T cell expansion [31,37]. The field is thus keenly awaiting studies that utilize second generation CARs and improved T cell expansion procedures that provide appropriate costimulation and cytokine stimulation before T cell infusion. Several efforts are under way to address this issue [38,44]. Studies targeting CD19 are especially awaited as these hold the promise of activity in several B cell malignancies. At least seven trials targeting CD19 with second generation CARs are programmed in the US and Europe, with one having already started in chronic lymphocytic leukemia, utilizing a CD19-CD28ζ CAR [51].

Conclusion

In comparison to TCRs, CARs have two major advantages: HLA-independent recognition of antigen, which makes them broadly applicable irrespective of the patient’s HLA and enables the recognition of tumor cells that have downregulated HLA expression, and no risk of mispairing with the endogenous TCR. Their signaling capacity was once a concern, but second generation receptors go far beyond what the original zeta chain fusions could achieve, that is, redirect cytotoxicity without permitting T cell expansion and survival upon repeat antigen exposure. CARs are therefore not just an alternative to TCRs but may prove to be superior as therapeutic entities.

Their use is, however, threatened by their potential immunogenicity. Anti-CAR antibody responses have been seen with some CARs, but not universally, which suggests that CAR immunogenicity may be neither universal nor intractable. CARs encompassing humanized scFvs or scFvs derived from human monoclonal antibodies will probably reduce this concern.

The choice of a target antigen is very crucial, as for any immunotherapy. This choice will become even more crucial as potent immunotherapies such as those utilizing second and third generation CARs enter the clinic. The G250 study illustrates the ability of CAR-targeted T cells to seek out normal tissues that express high levels of the targeted antigen [42]. The targeting of CD19, a cell-surface molecule found on the majority of leukemias and lymphomas, will be an interesting case to follow as CD19 is expressed on normal proB, preB, and B cells but no other hematopoietic progenitor cell types. The several upcoming trials targeting CD19 will provide interesting variations on this one theme, as they utilize slightly different CARs, different scFvs, different vector systems, different T cell expansion methodologies, and different starting T cell subsets to treat different CD19-positive malignancies. The field of CARs is coming of age.

References and recommended reading

Papers of particular interest, published within the period of review, have been highlighted as:

• of special interest

  • 1.Sadelain M, Riviere I, Brentjens R. Targeting tumours with genetically enhanced T lymphocytes. Nat Rev Cancer. 2003;3:35–45. doi: 10.1038/nrc971. [DOI] [PubMed] [Google Scholar]
  • 2.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;3:431–437. doi: 10.1016/s1535-6108(03)00113-2. [DOI] [PubMed] [Google Scholar]
  • 3.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. doi: 10.1038/nrc2355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.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. doi: 10.1016/0092-8674(91)90314-o. [DOI] [PubMed] [Google Scholar]
  • 5.Romeo C, Amiot M, Seed B. Sequence requirements for induction of cytolysis by the T cell antigen/Fc receptor zeta chain. Cell. 1992;68:889–897. doi: 10.1016/0092-8674(92)90032-8. [DOI] [PubMed] [Google Scholar]
  • 6.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;90:720–724. doi: 10.1073/pnas.90.2.720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Altenschmidt U, Kahl R, Moritz D, Schnierle BS, Gerstmayer B, Wels W, Groner B. Cytolysis of tumor cells expressing the Neu/erbB-2, erbB-3, and erbB-4 receptors by genetically targeted naive T lymphocytes. Clin Cancer Res. 1996;2:1001–1008. [PubMed] [Google Scholar]
  • 8.Kahlon KS, Brown C, Cooper LJ, Raubitschek A, Forman SJ, Jensen MC. Specific recognition and killing of glioblastoma multiforme by interleukin 13-zetakine redirected cytolytic T cells. Cancer Res. 2004;64:9160–9166. doi: 10.1158/0008-5472.CAN-04-0454. [DOI] [PubMed] [Google Scholar]
  • 9.Seliger B, Ritz U, Ferrone S. Molecular mechanisms of HLA class I antigen abnormalities following viral infection and transformation. Int J Cancer. 2006;118:129–138. doi: 10.1002/ijc.21312. [DOI] [PubMed] [Google Scholar]
  • 10.Seliger B. Molecular mechanisms of MHC class I abnormalities and APM components in human tumors. Cancer Immunol Immunother. 2008;57:1719–1726. doi: 10.1007/s00262-008-0515-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Moore TV, Lyons GE, Brasic N, Roszkowski JJ, Voelkl S, Mackensen A, Kast WM, Le Poole IC, Nishimura MI. Relationship between CD8-dependent antigen recognition, T cell functional avidity, and tumor cell recognition. Cancer Immunol Immunother. 2009;58:719–728. doi: 10.1007/s00262-008-0594-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Richman SA, Kranz DM. Display, engineering, and applications of antigen-specific T cell receptors. Biomol Eng. 2007;24:361–373. doi: 10.1016/j.bioeng.2007.02.009. [DOI] [PubMed] [Google Scholar]
  • 13.Turatti F, Figini M, Balladore E, Alberti P, Casalini P, Marks JD, Canevari S, Mezzanzanica D. Redirected activity of human antitumor chimeric immune receptors is governed by antigen and receptor expression levels and affinity of interaction. J Immunother. 2007;30:684–693. doi: 10.1097/CJI.0b013e3180de5d90. [DOI] [PubMed] [Google Scholar]
  • 14.Papapetrou EP, Kovalovsky D, Beloeil L, Sant’angelo D, Sadelain M. Harnessing endogenous miR-181a to segregate transgenic antigen receptor expression in developing versus post-thymic T cells in murine hematopoietic chimeras. J Clin Invest. 2009;119:157–168. doi: 10.1172/JCI37216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Guest RD, Hawkins RE, Kirillova N, Cheadle EJ, Arnold J, O’Neill A, Irlam J, Chester KA, Kemshead JT, Shaw DM, et al. The role of extracellular spacer regions in the optimal design of chimeric immune receptors: evaluation of four different scFvs and antigens. J Immunother. 2005;28:203–211. doi: 10.1097/01.cji.0000161397.96582.59. [DOI] [PubMed] [Google Scholar]
  • 16.Wilkie S, Picco G, Foster J, Davies DM, Julien S, Cooper L, Arif S, Mather SJ, Taylor-Papadimitriou J, Burchell JM, et al. Retargeting of human T cells to tumor-associated MUC1: the evolution of a chimeric antigen receptor. J Immunol. 2008;180:4901–4909. doi: 10.4049/jimmunol.180.7.4901. [DOI] [PubMed] [Google Scholar]
  • 17.Eshhar Z, Bach N, Fitzer-Attas CJ, Gross G, Lustgarten J, Waks T, Schindler DG. The T-body approach: potential for cancer immunotherapy. Springer Semin Immunopathol. 1996;18:199–209. doi: 10.1007/BF00820666. [DOI] [PubMed] [Google Scholar]
  • 18.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;1:123–127. doi: 10.1038/sj.neo.7900018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Haynes NM, Trapani JA, Teng MW, Jackson JT, Cerruti L, Jane SM, Kershaw MH, Smyth MJ, Darcy PK. Single-chain antigen recognition receptors that costimulate potent rejection of established experimental tumors. Blood. 2002;100:3155–3163. doi: 10.1182/blood-2002-04-1041. [DOI] [PubMed] [Google Scholar]
  • 20.Hombach A, Heuser C, Marquardt T, Wieczarkowiecz A, Groneck V, Pohl C, Abken H. CD4+ T cells engrafted with a recombinant immunoreceptor efficiently lyse target cells in a MHC antigen- and Fas-independent fashion. J Immunol. 2001;167:1090–1096. doi: 10.4049/jimmunol.167.2.1090. [DOI] [PubMed] [Google Scholar]
  • 21.Maher J, Brentjens RJ, Gunset G, Riviere I, Sadelain M. Human T-lymphocyte cytotoxicity and proliferation directed by a single chimeric TCRzeta/CD28 receptor. Nat Biotechnol. 2002;20:70–75. doi: 10.1038/nbt0102-70. [DOI] [PubMed] [Google Scholar]
  • 22.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;12:933–941. doi: 10.1016/j.ymthe.2005.04.016. [DOI] [PubMed] [Google Scholar]
  • 23.Wang J, Jensen M, Lin Y, Sui X, Chen E, Lindgren CG, Till B, Raubitschek A, Forman SJ, Qian X, 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;18:712–725. doi: 10.1089/hum.2007.028. [DOI] [PubMed] [Google Scholar]
  • 24.Teng MW, Kershaw MH, Moeller M, Smyth MJ, Darcy PK. Immunotherapy of cancer using systemically delivered gene-modified human T lymphocytes. Hum Gene Ther. 2004;15:699–708. doi: 10.1089/1043034041361235. [DOI] [PubMed] [Google Scholar]
  • 25.Brentjens RJ, Santos E, Nikhamin Y, Yeh R, Matsushita M, La Perle K, Quintas-Cardama A, Larson SM, Sadelain M. Genetically targeted T cells eradicate systemic acute lymphoblastic leukemia xenografts. Clin Cancer Res. 2007;13:5426–5435. doi: 10.1158/1078-0432.CCR-07-0674. [DOI] [PubMed] [Google Scholar]
  • 26.Haynes NM, Trapani JA, Teng MW, Jackson JT, Cerruti L, Jane SM, Kershaw MH, Smyth MJ, Darcy PK. Rejection of syngeneic colon carcinoma by CTLs expressing single-chain antibody receptors codelivering CD28 costimulation. J Immunol. 2002;169:5780–5786. doi: 10.4049/jimmunol.169.10.5780. [DOI] [PubMed] [Google Scholar]
  • 27.Kowolik CM, Topp MS, Gonzalez S, Pfeiffer T, Olivares S, Gonzalez N, Smith DD, Forman SJ, Jensen MC, Cooper LJ. 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;66:10995–11004. doi: 10.1158/0008-5472.CAN-06-0160. [DOI] [PubMed] [Google Scholar]
  • 28.Loskog A, Giandomenico V, Rossig C, Pule M, Dotti G, Brenner MK. Addition of the CD28 signaling domain to chimeric T-cell receptors enhances chimeric T-cell resistance to T regulatory cells. Leukemia. 2006;20:1819–1828. doi: 10.1038/sj.leu.2404366. [DOI] [PubMed] [Google Scholar]
  • 29.Moeller M, Haynes NM, Trapani JA, Teng MW, Jackson JT, Tanner JE, Cerutti L, Jane SM, Kershaw MH, Smyth MJ, et al. A functional role for CD28 costimulation in tumor recognition by single-chain receptor-modified T cells. Cancer Gene Ther. 2004;11:371–379. doi: 10.1038/sj.cgt.7700710. [DOI] [PubMed] [Google Scholar]
  • 30.Vera J, Savoldo B, Vigouroux S, Biagi E, Pule M, Rossig C, Wu J, Heslop HE, Rooney CM, Brenner MK, et al. T lymphocytes redirected against the kappa light chain of human immunoglobulin efficiently kill mature B lymphocyte-derived malignant cells. Blood. 2006;108:3890–3897. doi: 10.1182/blood-2006-04-017061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31•.Brentjens RJ, Latouche JB, Santos E, Marti F, Gong MC, Lyddane C, King PD, Larson S, Weiss M, Riviere I, et al. Eradication of systemic B-cell tumors by genetically targeted human T lymphocytes co-stimulated by CD80 and interleukin-15. Nat Med. 2003;9:279–286. doi: 10.1038/nm827. First demonstration of tumor eradication by systemically delivered, CD19-targeted T cells in mice. Benefit of interleukin-15 in adoptive cell therapy. [DOI] [PubMed] [Google Scholar]
  • 32.Rossig C, Bollard CM, Nuchtern JG, Rooney CM, Brenner MK. Epstein-Barr virus-specific human T lymphocytes expressing antitumor chimeric T-cell receptors: potential for improved immunotherapy. Blood. 2002;99:2009–2016. doi: 10.1182/blood.v99.6.2009. [DOI] [PubMed] [Google Scholar]
  • 33.Zakrzewski JL, Kochman AA, Lu SX, Terwey TH, Kim TD, Hubbard VM, Muriglan SJ, Suh D, Smith OM, Grubin J, et al. Adoptive transfer of T-cell precursors enhances T-cell reconstitution after allogeneic hematopoietic stem cell transplantation. Nat Med. 2006;12:1039–1047. doi: 10.1038/nm1463. [DOI] [PubMed] [Google Scholar]
  • 34.Zakrzewski JL, Suh D, Markley JC, Smith OM, King C, Goldberg GL, Jenq R, Holland AM, Grubin J, Cabrera-Perez J, et al. Tumor immunotherapy across MHC barriers using allogeneic T-cell precursors. Nat Biotechnol. 2008;26:453–461. doi: 10.1038/nbt1395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Wang G, Chopra RK, Royal RE, Yang JC, Rosenberg SA, Hwu P. A T cell-independent antitumor response in mice with bone marrow cells retrovirally transduced with an antibody/Fc-gamma chain chimeric receptor gene recognizing a human ovarian cancer antigen. Nat Med. 1998;4:168–172. doi: 10.1038/nm0298-168. [DOI] [PubMed] [Google Scholar]
  • 36.Gattinoni L, Powell DJ, Jr, Rosenberg SA, Restifo NP. Adoptive immunotherapy for cancer: building on success. Nat Rev Immunol. 2006;6:383–393. doi: 10.1038/nri1842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37•.Pule MA, Savoldo B, Myers GD, Rossig C, Russell HV, Dotti G, Huls MH, Liu E, Gee AP, Mei Z, et al. Virus-specific T cells engineered to coexpress tumor-specific receptors: persistence and antitumor activity in individuals with neuroblastoma. Nat Med. 2008;14:1264–1270. doi: 10.1038/nm.1882. Tumor regression noted in neuroblastoma patients treated with GD2 - targeted T cells. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Berger C, Jensen MC, Lansdorp PM, Gough M, Elliott C, Riddell SR. Adoptive transfer of effector CD8+ T cells derived from central memory cells establishes persistent T cell memory in primates. J Clin Invest. 2008;118:294–305. doi: 10.1172/JCI32103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Gade TP, Hassen W, Santos E, Gunset G, Saudemont A, Gong MC, Brentjens R, Zhong XS, Stephan M, Stefanski J, et al. Targeted elimination of prostate cancer by genetically directed human T lymphocytes. Cancer Res. 2005;65:9080–9088. doi: 10.1158/0008-5472.CAN-05-0436. [DOI] [PubMed] [Google Scholar]
  • 40.Stephan MT, Ponomarev V, Brentjens RJ, Chang AH, Dobrenkov KV, Heller G, Sadelain M. T cell-encoded CD80 and 4-1BBL induce auto- and transcostimulation, resulting in potent tumor rejection. Nat Med. 2007;13:1440–1449. doi: 10.1038/nm1676. [DOI] [PubMed] [Google Scholar]
  • 41.Kershaw MH, Westwood JA, Parker LL, Wang G, Eshhar Z, Mavroukakis SA, White DE, Wunderlich JR, Canevari S, Rogers-Freezer L, et al. A phase I study on adoptive immunotherapy using gene-modified T cells for ovarian cancer. Clin Cancer Res. 2006;12:6106–6115. doi: 10.1158/1078-0432.CCR-06-1183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42•.Lamers CH, Sieijfer S, Vulto AG, Kruit WH, Kliffen M, Debets R, Gratama JW, Stoter G, Oosterwijk E. 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–e22. doi: 10.1200/JCO.2006.05.9964. On-target toxicity induced by carbonic anhydrase IX-targeted T cells reacting against biliary epithelium in patients with renal cell carcinoma. [DOI] [PubMed] [Google Scholar]
  • 43.Till BG, Jensen MC, Wang J, Chen EY, Wood BL, Greisman HA, Qian X, James SE, Raubitschek A, Forman SJ, et al. Adoptive immunotherapy for indolent non-Hodgkin lymphoma and mantle cell lymphoma using genetically modified autologous CD20-specific T cells. Blood. 2008;112:2261–2271. doi: 10.1182/blood-2007-12-128843. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Hollyman D, Stefanski J, Przybylowski M, Bartido S, Borquez-Ojeda O, Taylor C, Yeh R, Capacio V, Olszewska M, Hosey J, Sadelain M, Brentjens RJ, Rivière I. Manufacturing validation of biologically functional T cells targeted to CD19 antigen for autologous adoptive cell therapy. J Immunother. 2009;32:169–180. doi: 10.1097/CJI.0b013e318194a6e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Weijtens ME, Willemsen RA, Valerio D, Stam K, Bolhuis RL. Single chain Ig/gamma gene-redirected human T lymphocytes produce cytokines, specifically lyse tumor cells, and recycle lytic capacity. J Immunol. 1996;157:836–843. [PubMed] [Google Scholar]
  • 46.Weijtens ME, Hart EH, Bolhuis RL. Functional balance between T cell chimeric receptor density and tumor associated antigen density: CTL mediated cytolysis and lymphokine production. Gene Ther. 2000;7:35–42. doi: 10.1038/sj.gt.3301051. [DOI] [PubMed] [Google Scholar]
  • 47.Weijtens ME, Willemsen RA, van Krimpen BA, Bolhuis RL. Chimeric scFv/gamma receptor-mediated T-cell lysis of tumor cells is coregulated by adhesion and accessory molecules. Int J Cancer. 1998;77:181–187. doi: 10.1002/(sici)1097-0215(19980717)77:2<181::aid-ijc2>3.0.co;2-m. [DOI] [PubMed] [Google Scholar]
  • 48.Lamers CH, Langeveld SC, Groot-van Ruijven CM, Debets R, Sleijfer S, Gratama JW. Gene-modified T cells for adoptive immunotherapy of renal cell cancer maintain transgene-specific immune functions in vivo. Cancer Immunol Immunother. 2007;56:1875–1883. doi: 10.1007/s00262-007-0330-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Cheadle EJ, Gilham DE, Hawkins RE. The combination of cyclophosphamide and human T cells genetically engineered to target CD19 can eradicate established B-cell lymphoma. Br J Haematol. 2008;142:65–68. doi: 10.1111/j.1365-2141.2008.07145.x. [DOI] [PubMed] [Google Scholar]
  • 50.Jensen MC, Popplewell L, DiGiusto DL, Kalos M, Cooper LJ, Raubitschek A, Forman SJ. A first-in-human clinical trial of adoptive therapy using CD19-specific chimeric antigen receptor re-directed T cells for recurrent/refractory follicular lymphoma. Mol Ther. 2007;15:S142. [Google Scholar]
  • 51.Brentjens RJ, Hollyman DR, Weiss M, Stefanski J, Przybylowski M, Bartido S, Borquez-Ojeda O, Taylor C, Hosey J, Heaney M, et al. A Phase I trial for the treatment of chemo-refractory chronic lymphocytic leukemia with CD19-targeted autologous T cells. Mol Ther. 2008;16:S15. [Google Scholar]
  • 52.Singh H, Manuri PR, Olivares S, Dara N, Dawson MJ, Huls H, Hackett PB, Kohn DB, Shpall EJ, Champlin RE, et al. Redirecting specificity of T-cell populations for CD19 using the Sleeping Beauty system. Cancer Res. 2008;68:2961–2971. doi: 10.1158/0008-5472.CAN-07-5600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Imai C, Mihara K, Andreansky M, Nicholson IC, Pui CH, Geiger TL, Campana D. Chimeric receptors with 4-1BB signaling capacity provoke potent cytotoxicity against acute lymphoblastic leukemia. Leukemia. 2004;18:676–684. doi: 10.1038/sj.leu.2403302. [DOI] [PubMed] [Google Scholar]
  • 54.Huang X, Guo H, Kang J, Choi S, Zhou TC, Tammana S, Lees CJ, Li ZZ, Milone M, Levine BL, et al. Sleeping Beauty transposon-mediated engineering of human primary T cells for therapy of CD19+ lymphoid malignancies. Mol Ther. 2008;16:580–589. doi: 10.1038/sj.mt.6300404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Cooper LJ, Al-Kadhimi Z, Serrano LM, Pfeiffer T, Olivares S, Castro A, Chang WC, Gonzalez S, Smith D, Forman SJ, et al. Enhanced antilymphoma efficacy of CD19-redirected influenza MP1-specific CTLs by cotransfer of T cells modified to present influenza MP1. Blood. 2005;105:1622–1631. doi: 10.1182/blood-2004-03-1208. [DOI] [PubMed] [Google Scholar]
  • 56.Landmeier S, Altvater B, Pscherer S, Eing BR, Kuehn J, Rooney CM, Juergens H, Rossig C. Gene-engineered varicellazoster virus reactive CD4+ cytotoxic T cells exert tumor-specific effector function. Cancer Res. 2007;67:8335–8343. doi: 10.1158/0008-5472.CAN-06-4426. [DOI] [PubMed] [Google Scholar]
  • 57.Yu K, Hu Y, Tan Y, Shen Z, Jiang S, Qian H, Liang B, Shan D. Immunotherapy of lymphomas with T cells modified by anti-CD20 scFv/CD28/CD3zeta recombinant gene. Leuk Lymphoma. 2008;49:1368–1373. doi: 10.1080/10428190802064958. [DOI] [PubMed] [Google Scholar]
  • 58.James SE, Greenberg PD, Jensen MC, Lin Y, Wang J, Till BG, Raubitschek AA, Forman SJ, Press OW. Antigen sensitivity of CD22-specific chimeric TCR is modulated by target epitope distance from the cell membrane. J Immunol. 2008;180:7028–7038. doi: 10.4049/jimmunol.180.10.7028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Jensen MC, Cooper LJ, Wu AM, Forman SJ, Raubitschek A. Engineered CD20-specific primary human cytotoxic T lymphocytes for targeting B-cell malignancy. Cytotherapy. 2003;5:131–138. doi: 10.1080/14653240310001028. [DOI] [PubMed] [Google Scholar]
  • 60.Wang J, Press OW, Lindgren CG, Greenberg P, Riddell S, Qian X, Laugen C, Raubitschek A, Forman SJ, Jensen MC. Cellular immunotherapy for follicular lymphoma using genetically modified CD20-specific CD8+ cytotoxic T lymphocytes. Mol Ther. 2004;9:577–586. doi: 10.1016/j.ymthe.2003.12.011. [DOI] [PubMed] [Google Scholar]
  • 61.Hombach A, Muche JM, Gerken M, Gellrich S, Heuser C, Pohl C, Sterry W, Abken H. T cells engrafted with a recombinant anti-CD30 receptor target autologous CD30(+) cutaneous lymphoma cells. Gene Ther. 2001;8:891–895. doi: 10.1038/sj.gt.3301467. [DOI] [PubMed] [Google Scholar]
  • 62.Savoldo B, Rooney CM, DiStasi A, Abken H, Hombach A, Foster AE, Zhang L, Heslop HE, Brenner MK, Dotti G. Epstein Barr virus specific cytotoxic T lymphocytes expressing the anti-CD30zeta artificial chimeric T-cell receptor for immunotherapy of Hodgkin disease. Blood. 2007;110:2620–2630. doi: 10.1182/blood-2006-11-059139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Finney HM, Akbar AN, Lawson AD. Activation of resting human primary T cells with chimeric receptors: costimulation from CD28, inducible costimulator, CD134, and CD137 in series with signals from the TCRzeta chain. J Immunol. 2004;172:104–113. doi: 10.4049/jimmunol.172.1.104. [DOI] [PubMed] [Google Scholar]
  • 64.Dall P, Herrmann I, Durst B, Stoff-Khalili MA, Bauerschmitz G, Hanstein B, Niederacher D. In vivo cervical cancer growth inhibition by genetically engineered cytotoxic T cells. Cancer Immunol Immunother. 2005;54:51–60. doi: 10.1007/s00262-004-0569-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Beecham EJ, Ortiz-Pujols S, Junghans RP. Dynamics of tumor cell killing by human T lymphocytes armed with an anti-carcinoembryonic antigen chimeric immunoglobulin T-cell receptor. J Immunother. 2000;23:332–343. doi: 10.1097/00002371-200005000-00006. [DOI] [PubMed] [Google Scholar]
  • 66.Beecham EJ, Ma Q, Ripley R, Junghans RP. Coupling CD28 co-stimulation to immunoglobulin T-cell receptor molecules: the dynamics of T-cell proliferation and death. J Immunother. 2000;23:631–642. doi: 10.1097/00002371-200011000-00004. [DOI] [PubMed] [Google Scholar]
  • 67.Hombach A, Schneider C, Sent D, Koch D, Willemsen RA, Diehl V, Kruis W, Bolhuis RL, Pohl C, Abken H. An entirely humanized CD3 zeta-chain signaling receptor that directs peripheral blood t cells to specific lysis of carcinoembryonic antigen-positive tumor cells. Int J Cancer. 2000;88:115–120. [PubMed] [Google Scholar]
  • 68.Haynes NM, Snook MB, Trapani JA, Cerruti L, Jane SM, Smyth MJ, Darcy PK. Redirecting mouse CTL against colon carcinoma: superior signaling efficacy of single-chain variable domain chimeras containing TCR-zeta vs Fc epsilon RI-gamma. J Immunol. 2001;166:182–187. doi: 10.4049/jimmunol.166.1.182. [DOI] [PubMed] [Google Scholar]
  • 69.Darcy PK, Haynes NM, Snook MB, Trapani JA, Cerruti L, Jane SM, Smyth MJ. Redirected perforin-dependent lysis of colon carcinoma by ex vivo genetically engineered CTL. J Immunol. 2000;164:3705–3712. doi: 10.4049/jimmunol.164.7.3705. [DOI] [PubMed] [Google Scholar]
  • 70.Nolan KF, Yun CO, Akamatsu Y, Murphy JC, Leung SO, Beecham EJ, Junghans RP. Bypassing immunization: optimized design of ‘designer T cells’ against carcinoembryonic antigen (CEA)-expressing tumors, and lack of suppression by soluble CEA. Clin Cancer Res. 1999;5:3928–3941. [PubMed] [Google Scholar]
  • 71.Hombach A, Wieczarkowiecz A, Marquardt T, Heuser C, Usai L, Pohl C, Seliger B, Abken H. Tumor-specific T cell activation by recombinant immunoreceptors: CD3 zeta signaling and CD28 costimulation are simultaneously required for efficient IL-2 secretion and can be integrated into one combined CD28/CD3 zeta signaling receptor molecule. J Immunol. 2001;167:6123–6131. doi: 10.4049/jimmunol.167.11.6123. [DOI] [PubMed] [Google Scholar]
  • 72.Gyobu H, Tsuji T, Suzuki Y, Ohkuri T, Chamoto K, Kuroki M, Miyoshi H, Kawarada Y, Katoh H, Takeshima T, et al. Generation and targeting of human tumor-specific Tc1 and Th1 cells transduced with a lentivirus containing a chimeric immunoglobulin T-cell receptor. Cancer Res. 2004;64:1490–1495. doi: 10.1158/0008-5472.can-03-2780. [DOI] [PubMed] [Google Scholar]
  • 73.Sasaki T, Ikeda H, Sato M, Ohkuri T, Abe H, Kuroki M, Onodera M, Miyamoto M, Kondo S, Nishimura T. Antitumor activity of chimeric immunoreceptor gene-modified Tc1 and Th1 cells against autologous carcinoembryonic antigen-expressing colon cancer cells. Cancer Sci. 2006;97:920–927. doi: 10.1111/j.1349-7006.2006.00271.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Ren-Heidenreich L, Mordini R, Hayman GT, Siebenlist R, LeFever A. Comparison of the TCR zeta-chain with the FcR gamma-chain in chimeric TCR constructs for T cell activation and apoptosis. Cancer Immunol Immunother. 2002;51:417–423. doi: 10.1007/s00262-002-0301-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Ren-Heidenreich L, Hayman GT, Trevor KT. Specific targeting of EGP-2+ tumor cells by primary lymphocytes modified with chimeric T cell receptors. Hum Gene Ther. 2000;11:9–19. doi: 10.1089/10430340050016111. [DOI] [PubMed] [Google Scholar]
  • 76.Daly T, Royal RE, Kershaw MH, Treisman J, Wang G, Li W, Herlyn D, Eshhar Z, Hwu P. Recognition of human colon cancer by T cells transduced with a chimeric receptor gene. Cancer Gene Ther. 2000;7:284–291. doi: 10.1038/sj.cgt.7700121. [DOI] [PubMed] [Google Scholar]
  • 77.Moeller M, Haynes NM, Kershaw MH, Jackson JT, Teng MW, Street SE, Cerutti L, Jane SM, Trapani JA, Smyth MJ, et al. Adoptive transfer of gene-engineered CD4+ helper T cells induces potent primary and secondary tumor rejection. Blood. 2005;106:2995–3003. doi: 10.1182/blood-2004-12-4906. [DOI] [PubMed] [Google Scholar]
  • 78.Murphy A, Westwood JA, Brown LE, Teng MW, Moeller M, Xu Y, Smyth MJ, Hwu P, Darcy PK, Kershaw MH. Antitumor activity of dual-specific T cells and influenza virus. Cancer Gene Ther. 2007;14:499–508. doi: 10.1038/sj.cgt.7701034. [DOI] [PubMed] [Google Scholar]
  • 79.Pinthus JH, Waks T, Kaufman-Francis K, Schindler DG, Harmelin A, Kanety H, Ramon J, Eshhar Z. Immunogene therapy of established prostate tumors using chimeric receptor-redirected human lymphocytes. Cancer Res. 2003;63:2470–2476. [PubMed] [Google Scholar]
  • 80.Muniappan A, Banapour B, Lebkowski J, Talib S. Ligand-mediated cytolysis of tumor cells: use of heregulin-zeta chimeras to redirect cytotoxic T lymphocytes. Cancer Gene Ther. 2000;7:128–134. doi: 10.1038/sj.cgt.7700100. [DOI] [PubMed] [Google Scholar]
  • 81.Altenschmidt U, Klundt E, Groner B. Adoptive transfer of in vitro-targeted, activated T lymphocytes results in total tumor regression. J Immunol. 1997;159:5509–5515. [PubMed] [Google Scholar]
  • 82.Hwu P, Yang JC, Cowherd R, Treisman J, Shafer GE, Eshhar Z, Rosenberg SA. In vivo antitumor activity of T cells redirected with chimeric antibody/T-cell receptor genes. Cancer Res. 1995;55:3369–3373. [PubMed] [Google Scholar]
  • 83.Parker LL, Do MT, Westwood JA, Wunderlich JR, Dudley ME, Rosenberg SA, Hwu P. Expansion and characterization of T cells transduced with a chimeric receptor against ovarian cancer. Hum Gene Ther. 2000;11:2377–2387. doi: 10.1089/104303400750038480. [DOI] [PubMed] [Google Scholar]
  • 84.Kershaw MH, Westwood JA, Hwu P. Dual-specific T cells combine proliferation and antitumor activity. Nat Biotechnol. 2002;20:1221–1227. doi: 10.1038/nbt756. [DOI] [PubMed] [Google Scholar]
  • 85.Gattenlohner S, Marx A, Markfort B, Pscherer S, Landmeier S, Juergens H, Muller-Hermelink HK, Matthews I, Beeson D, Vincent A, et al. Rhabdomyosarcoma lysis by T cells expressing a human autoantibody-based chimeric receptor targeting the fetal acetylcholine receptor. Cancer Res. 2006;66:24–28. doi: 10.1158/0008-5472.CAN-05-0542. [DOI] [PubMed] [Google Scholar]
  • 86.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;188:619–626. doi: 10.1084/jem.188.4.619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Yun CO, Nolan KF, Beecham EJ, Reisfeld RA, Junghans RP. Targeting of T lymphocytes to melanoma cells through chimeric anti-GD3 immunoglobulin T-cell receptors. Neoplasia. 2000;2:449–459. doi: 10.1038/sj.neo.7900108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Ahmed N, Ratnayake M, Savoldo B, Perlaky L, Dotti G, Wels WS, Bhattacharjee MB, Gilbertson RJ, Shine HD, Weiss HL, et al. Regression of experimental medulloblastoma following transfer of HER2-specific T cells. Cancer Res. 2007;67:5957–5964. doi: 10.1158/0008-5472.CAN-06-4309. [DOI] [PubMed] [Google Scholar]
  • 89.Chang L, Chang WC, McNamara G, Aguilar B, Ostberg JR, Jensen MC. Transgene-enforced co-stimulation of CD4+ T cells leads to enhanced and sustained anti-tumor effector functioning. Cytotherapy. 2007;9:771–784. doi: 10.1080/14653240701656079. [DOI] [PubMed] [Google Scholar]
  • 90.Lazovic J, Jensen MC, Ferkassian E, Aguilar B, Raubitschek A, Jacobs RE. Imaging immune response in vivo: cytolytic action of genetically altered T cells directed to glioblastoma multiforme. Clin Cancer Res. 2008;14:3832–3839. doi: 10.1158/1078-0432.CCR-07-5067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Stastny MJ, Brown CE, Ruel C, Jensen MC. Medulloblastomas expressing IL13Ralpha2 are targets for IL13-zetakine+ cytolytic T cells. J Pediatr Hematol Oncol. 2007;29:669–677. doi: 10.1097/MPH.0b013e3181468c68. [DOI] [PubMed] [Google Scholar]
  • 92.Kershaw MH, Westwood JA, Zhu Z, Witte L, Libutti SK, Hwu P. Generation of gene-modified T cells reactive against the angiogenic kinase insert domain-containing receptor (KDR) found on tumor vasculature. Hum Gene Ther. 2000;11:2445–2452. doi: 10.1089/10430340050207939. [DOI] [PubMed] [Google Scholar]
  • 93.Mezzanzanica D, Canevari S, Mazzoni A, Figini M, Colnaghi MI, Waks T, Schindler DG, Eshhar Z. Transfer of chimeric receptor gene made of variable regions of tumor-specific antibody confers anticarbohydrate specificity on T cells. Cancer Gene Ther. 1998;5:401–407. [PubMed] [Google Scholar]
  • 94.Westwood JA, Smyth MJ, Teng MW, Moeller M, Trapani JA, Scott AM, Smyth FE, Cartwright GA, Power BE, Honemann D, et al. Adoptive transfer of T cells modified with a humanized chimeric receptor gene inhibits growth of Lewis-Y-expressing tumors in mice. Proc Natl Acad Sci U S A. 2005;102:19051–19056. doi: 10.1073/pnas.0504312102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Gonzalez S, Naranjo A, Serrano LM, Chang WC, Wright CL, Jensen MC. Genetic engineering of cytolytic T lymphocytes for adoptive T-cell therapy of neuroblastoma. J Gene Med. 2004;6:704–711. doi: 10.1002/jgm.489. [DOI] [PubMed] [Google Scholar]
  • 96.Park JR, Digiusto DL, Slovak M, Wright C, Naranjo A, Wagner J, Meechoovet HB, Bautista C, Chang WC, Ostberg JR, et al. Adoptive transfer of chimeric antigen receptor re-directed cytolytic T lymphocyte clones in patients with neuroblastoma. Mol Ther. 2007;15:825–833. doi: 10.1038/sj.mt.6300104. [DOI] [PubMed] [Google Scholar]
  • 97.Willemsen RA, Ronteltap C, Chames P, Debets R, Bolhuis RL. T cell retargeting with MHC class I-restricted antibodies: the CD28 costimulatory domain enhances antigen-specific cytotoxicity and cytokine production. J Immunol. 2005;174:7853–7858. doi: 10.4049/jimmunol.174.12.7853. [DOI] [PubMed] [Google Scholar]
  • 98.Iizuka K, Nakajima C, Iizuka YM, Takase M, Kato T, Noda S, Tanaka K, Kanagawa O. Protection from lethal infection by adoptive transfer of CD8 T cells genetically engineered to express virus-specific innate immune receptor. J Immunol. 2007;179:1122–1128. doi: 10.4049/jimmunol.179.2.1122. [DOI] [PubMed] [Google Scholar]
  • 99.Barber A, Zhang T, Sentman CL. Immunotherapy with chimeric NKG2D receptors leads to long-term tumor-free survival and development of host antitumor immunity in murine ovarian cancer. J Immunol. 2008;180:72–78. doi: 10.4049/jimmunol.180.1.72. [DOI] [PubMed] [Google Scholar]
  • 100.Zhang T, Barber A, Sentman CL. Chimeric NKG2D modified T cells inhibit systemic T-cell lymphoma growth in a manner involving multiple cytokines and cytotoxic pathways. Cancer Res. 2007;67:11029–11036. doi: 10.1158/0008-5472.CAN-07-2251. [DOI] [PubMed] [Google Scholar]
  • 101.Barber A, Zhang T, DeMars LR, Conejo-Garcia J, Roby KF, Sentman CL. Chimeric NKG2D receptor-bearing T cells as immunotherapy for ovarian cancer. Cancer Res. 2007;67:5003–5008. doi: 10.1158/0008-5472.CAN-06-4047. [DOI] [PubMed] [Google Scholar]
  • 102.Jiang HR, Gilham DE, Mulryan K, Kirillova N, Hawkins RE, Stern PL. Combination of vaccination and chimeric receptor expressing T cells provides improved active therapy of tumors. J Immunol. 2006;177:4288–4298. doi: 10.4049/jimmunol.177.7.4288. [DOI] [PubMed] [Google Scholar]
  • 103.Morgenroth A, Cartellieri M, Schmitz M, Gunes S, Weigle B, Bachmann M, Abken H, Rieber EP, Temme A. Targeting of tumor cells expressing the prostate stem cell antigen (PSCA) using genetically engineered T-cells. Prostate. 2007;67:1121–1131. doi: 10.1002/pros.20608. [DOI] [PubMed] [Google Scholar]
  • 104.Dobrenkov K, Olszewska M, Likar Y, Shenker L, Gunset G, Cai S, Pillarsetty N, Hricak H, Sadelain M, Ponomarev V. Monitoring the efficacy of adoptively transferred prostate cancer-targeted human T lymphocytes with PET and bioluminescence imaging. J Nucl Med. 2008;49:1162–1170. doi: 10.2967/jnumed.107.047324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Ma Q, Safar M, Holmes E, Wang Y, Boynton AL, Junghans RP. Anti-prostate specific membrane antigen designer T cells for prostate cancer therapy. Prostate. 2004;61:12–25. doi: 10.1002/pros.20073. [DOI] [PubMed] [Google Scholar]
  • 106.Teng MW, Kershaw MH, Jackson JT, Smyth MJ, Darcy PK. Adoptive transfer of chimeric Fcepsilon RI gene-modified human T cells for cancer immunotherapy. Hum Gene Ther. 2006;17:1134–1143. doi: 10.1089/hum.2006.17.1134. [DOI] [PubMed] [Google Scholar]
  • 107.McGuinness RP, Ge Y, Patel SD, Kashmiri SV, Lee HS, Hand PH, Schlom J, Finer MH, McArthur JG. Anti-tumor activity of human T cells expressing the CC49-zeta chimeric immune receptor. Hum Gene Ther. 1999;10:165–173. doi: 10.1089/10430349950018968. [DOI] [PubMed] [Google Scholar]
  • 108.Patel SD, Ge Y, Moskalenko M, McArthur JG. Anti-Tumor CC49-zeta CD4 T cells possess both cytolytic and helper functions. J Immunother. 2000;23:661–668. doi: 10.1097/00002371-200011000-00007. [DOI] [PubMed] [Google Scholar]
  • 109.Niederman TM, Ghogawala Z, Carter BS, Tompkins HS, Russell MM, Mulligan RC. Antitumor activity of cytotoxic T lymphocytes engineered to target vascular endothelial growth factor receptors. Proc Natl Acad Sci U S A. 2002;99:7009–7014. doi: 10.1073/pnas.092562399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Hombach A, Sent D, Schneider C, Heuser C, Koch D, Pohl C, Seliger B, Abken H. T-cell activation by recombinant receptors: CD28 costimulation is required for interleukin 2 secretion and receptor-mediated T-cell proliferation but does not affect receptor-mediated target cell lysis. Cancer Res. 2001;61:1976–1982. [PubMed] [Google Scholar]
  • 111.Westwood JA, Smyth MJ, Teng MW, Moeller M, Trapani JA, Scott AM, Smyth FE, Cartwright GA, Power BE, Honemann D, et al. Adoptive transfer of T cells modified with a humanized chimeric receptor gene inhibits growth of Lewis-Y-expressing tumors inmice. Proc Natl Acad Sci U S A. 2005;102:19051–19056. doi: 10.1073/pnas.0504312102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Finney HM, Akbar AN, Lawson AD. Activation of resting human primary T cells with chimeric receptors: costimulation from CD28, inducible costimulator, CD134, and CD137 in series with signals from the TCR zeta chain. J Immunol. 2004;172:104–113. doi: 10.4049/jimmunol.172.1.104. [DOI] [PubMed] [Google Scholar]
  • 113.Geiger TL, Nguyen P, Leitenberg D, Flavell RA. Integrated src kinase and costimulatory activity enhances signal transduction through single-chain chimeric receptors in T lymphocytes. Blood. 2001;98:2364–2371. doi: 10.1182/blood.v98.8.2364. [DOI] [PubMed] [Google Scholar]
  • 114.Nguyen P, Geiger TL. Antigen-specific targeting of CD8+ T cells with receptor-modified T lymphocytes. Gene Ther. 2003;10:594–604. doi: 10.1038/sj.gt.3301932. [DOI] [PubMed] [Google Scholar]
  • 115.Daldrup-Link HE, Meier R, Rudelius M, Piontek G, Piert M, Metz S, Settles M, Uherek C, Wels W, Schlegel J, et al. In vivo tracking of genetically engineered, anti-HER2/neu directed natural killer cells to HER2/neu positive mammary tumors with magnetic resonance imaging. Eur Radiol. 2005;15:4–13. doi: 10.1007/s00330-004-2526-7. [DOI] [PubMed] [Google Scholar]
  • 116.Imai C, Iwamoto S, Campana D. Genetic modification of primary natural killer cells overcomes inhibitory signals and induces specific killing of leukemic cells. Blood. 2005;106:376–383. doi: 10.1182/blood-2004-12-4797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Muller T, Uherek C, Maki G, Chow KU, Schimpf A, Klingemann HG, Tonn T, Wels WS. Expression of a CD20-specific chimeric antigen receptor enhances cytotoxic activity of NK cells and overcomes NK-resistance of lymphoma and leukemia cells. Cancer Immunol Immunother. 2008;57:411–423. doi: 10.1007/s00262-007-0383-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Uherek C, Tonn T, Uherek B, Becker S, Schnierle B, Klingemann HG, Wels W. Retargeting of natural killer-cell cytolytic activity to ErbB2-expressing cancer cells results in efficient and selective tumor cell destruction. Blood. 2002;100:1265–1273. [PubMed] [Google Scholar]
  • 119.Roberts MR, Cooke KS, Tran AC, Smith KA, Lin WY, Wang M, Dull TJ, Farson D, Zsebo KM, Finer MH. Antigen-specific cytolysis by neutrophils and NK cells expressing chimeric immune receptors bearing zeta or gamma signaling domains. J Immunol. 1998;161:375–384. [PubMed] [Google Scholar]
  • 120.Kruschinski A, Moosmann A, Poschke I, Norell H, Chmielewski M, Seliger B, Kiessling R, Blankenstein T, Abken H, Charo J. Engineering antigen-specific primary human NK cells against HER-2 positive carcinomas. Proc Natl Acad Sci U S A. 2008;105:17481–17486. doi: 10.1073/pnas.0804788105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Pegram HJ, Jackson JT, Smyth MJ, Kershaw MH, Darcy PK. Adoptive transfer of gene-modified primary NK cells can specifically inhibit tumor progression in vivo. J Immunol. 2008;181:3449–3455. doi: 10.4049/jimmunol.181.5.3449. [DOI] [PubMed] [Google Scholar]
  • 122.Marin V, Dander E, Biagi E, Introna M, Fazio G, Biondi A, D’Amico G. Characterization of in vitro migratory properties of anti-CD19 chimeric receptor-redirected CIK cells for their potential use in B-ALL immunotherapy. Exp Hematol. 2006;34:1219–1229. doi: 10.1016/j.exphem.2006.05.004. [DOI] [PubMed] [Google Scholar]
  • 123.Marin V, Kakuda H, Dander E, Imai C, Campana D, Biondi A, D’Amico G. Enhancement of the anti-leukemic activity of cytokine induced killer cells with an anti-CD19 chimeric receptor delivering a 4-1BB-zeta activating signal. Exp Hematol. 2007;35:1388–1397. doi: 10.1016/j.exphem.2007.05.018. [DOI] [PubMed] [Google Scholar]
  • 124.Biglari A, Southgate TD, Fairbairn LJ, Gilham DE. Human monocytes expressing a CEA-specific chimeric CD64 receptor specifically target CEA-expressing tumour cells in vitro and in vivo. Gene Ther. 2006;13:602–610. doi: 10.1038/sj.gt.3302706. [DOI] [PubMed] [Google Scholar]

RESOURCES