Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Nov 24.
Published in final edited form as: Nature. 2017 May 24;545(7655):423–431. doi: 10.1038/nature22395

Therapeutic T cell engineering

Michel Sadelain 1, Isabelle Rivière 1, Stanley Riddell 2
PMCID: PMC5632949  NIHMSID: NIHMS908219  PMID: 28541315

Abstract

Genetically engineered T cells are powerful new medicines, offering hope for curative responses in patients with cancer. Chimaeric antigen receptors (CARs) are a class of synthetic receptors that reprogram lymphocyte specificity and function. CARs targeting CD19 have demonstrated remarkable potency in B cell malignancies. Engineered T cells are applicable in principle to many cancers, pending further progress to identify suitable target antigens, overcome immunosuppressive tumour microenvironments, reduce toxicities, and prevent antigen escape. Advances in the selection of optimal T cells, genetic engineering, and cell manufacturing are poised to broaden T-cell-based therapies and foster new applications in infectious diseases and autoimmunity.


Tlymphocytes develop in the thymus1, where they acquire their antigen receptor, known as the T cell receptor (TCR)2. T cells are an essential component of adaptive immunity, contributing to tumour rejection and pathogen clearance. The adoptive transfer of T cells was a pivotal experimental technique used more than half a century ago to establish that cellular components of the immune system could reject tumours3,4. Over time, adoptive transfer experiments in mice have contributed to the identification of tumour antigens and to elucidating the obstacles to establishing effective tumour immunity57. This laboratory procedure inspired clinical applications of adoptive cell transfer, including the use of autologous lymphokine-activated killer cells8 and tumour-infiltrating lymphocytes (TIL)9 to treat human solid tumours. Independently, borne out of the field of bone marrow transplantation, allogeneic donor T cells were shown to sometimes eradicate haematological malignancies via the graft-versus-leukaemia (GVL) effect10. Allogeneic T cells can also induce a devastating pathology known as graft-versus-host disease (GVHD). The dual-edged role played by T lymphocytes spurred a search to identify and separate beneficial and deleterious T cells11,12, which spawned allogeneic therapies such as donor leukocyte infusion13 and virus-specific T cell therapy1416. Altogether, these early clinical investigations eventually pointed to the need to better control the composition of therapeutic T cell products by increasing their content of tumour-specific T cells and removing T cells with harmful potential17.

A shared feature of late 20th century approaches is the focus on selecting and expanding naturally occurring T cells found in the patient or a healthy donor. The prospect of T cell engineering would singularly alter these original concepts. The emergence of replication-defective viral vectors18 provided new possibilities for cell therapies, such as the potential to genetically modify T lymphocytes19. In this perspective, it would no longer have to be a cell harvested from the patient or a donor that would be adoptively transferred, but a cell product designed and repurposed through ex vivo genetic modification20,21. Starting from easily accessible cells collected from a patient’s blood, genetic engineering provided a means to rapidly generate anti-tumour T cells for any cancer patient by introducing tumour-targeting receptors and other attributes intended to improve therapeutic efficacy and safety. Advancing therapeutic T cell engineering required progress on multiple fronts including target identification, antigen receptor isolation or design, T cell differentiation, genetic engineering, cell manufacturing sciences, and regulatory compliance. This immunotherapeutic modality thus draws not only on principles of immunology but genetics, synthetic biology, stem cell biology and a range of manufacturing technologies. The poster child for this new paradigm is CD19 CAR therapy.

Redirecting the specificity of T cells

The most natural approach to target a T cell to a chosen antigen is to express therein rearranged TCR α and β chains of defined antigen specificity, conforming to the physiological TCR–CD3 complex (Fig. 1a). This procedure was used in transgenic mice to demonstrate that the TCR was sufficient to direct antigen-specific T cell differentiation22 and later applied to human T cell clones to redirect their cytotoxicity23. The transfer of TCR genes aims to phenocopy naturally occurring T cells, thereby supplying tumour- or virus-specific T cells to individuals whose endogenous immune response is insufficient to combat the disease. Current efforts to implement this approach focus on isolating TCRs with optimal specificity and affinity24, and devising molecular strategies that eliminate potential TCR cross-reactivities25,26 and minimize αβ chain mispairing2729. TCR gene transfer however remains constrained by TCR competition for rate-limiting amounts of the signalling molecules of the CD3 complex30 (Fig. 1a), and by human leukocyte antigen (HLA) restriction, which imposes that multiple TCRs be identified for any given antigen to ensure that patients with different HLA haplotypes be eligible for therapy. These biological limitations notwithstanding, TCR gene transfer is a compelling therapeutic concept that utilizes physiological antigen recognition and T cell activation to generate antigen-specific T cells. Indeed, TCR-engineered T cells have been shown to have significant anti-tumour activity in patients with melanoma and sarcoma in small clinical trials31,32.

Figure 1. Receptors for antigen.

Figure 1

a, T cell receptor (TCR). Physiological antigen recognition is mediated by the TCR–CD3 complex, which comprises six independent gene products: the TCR α and β chains, which together bind to HLA–peptide complexes, and the CD3γ, δ, ε and ζ chains, which initiate T cell activation. b, Chimaeric antigen receptor (CAR). A prototypic second-generation CAR comprises three canonical domains for binding to antigen (unrestricted by HLA), T cell activation (commonly via the CD3ζ cytoplasmic domain) and co-stimulation (via the CD28 cytoplasmic domain in this example). The hinge and transmembrane domain also contribute to overall CAR function. c, Synthetic receptors in the extended CAR family: chimaeric co-stimulatory receptors (CCRs) provide co-stimulation in response to antigen or alternate ligands; chimaeric cytokine receptors (CyCRs) bind to one cytokine but transduce the signal of another; antigen-specific inhibitory receptors (iCARs) inhibit T cell activation in response to an antigen; synthetic Notch (synNotch) receptors induce CAR expression after antigen recognition by a chimaeric Notch receptor. The scissors indicate a cleavage site that releases a transcription factor (TF).

An alternative approach to retarget T cells rests on the design of artificial receptors for antigen33. In order to support a productive T cell response, receptors for antigen must encompass CD3 components that are sufficient to initiate T cell activation, such as the CD3 ζ chain3436. Fusion molecules that linked the ζ chain to an extracellular ligand binding domain, initially termed T bodies37 or chimaeric TCRs38, and later first-generation CARs33, are in essence a simplified TCR–CD3 complex serving as a TCR surrogate. However, experimentation in primary T cells and in transgenic mice revealed that signalling through such receptors was insufficient to direct productive immune responses3941, indicating that a better adapted and less natural design was needed to engineer sustainable T cell responses. Based on the incorporation of co-stimulatory components into antigen-specific receptors42, the design of receptors that simultaneously provided both activation and co-stimulatory signals opened a path for the generation of antigen-specific T cells capable of interleukin-2 secretion43 and, most importantly, expansion upon repeated exposure to antigen44. This receptor design, dubbed second-generation CAR33 (Fig. 1b), is the backbone of current CAR therapies.

Building up the potency of engineered T cells

The second-generation CAR design is amenable to a vast number of variations that include differences in antigen specificity, co-stimulatory signalling domain(s) and T-cell-activating components. The structural features afforded by the hinge and transmembrane regions (Fig. 1b) also affect antigen binding and signalling45. The term CAR intends to encompass this modular potential33, and over 100 different CAR specificities, predominantly using antibody single chain variable fragments (scFv) to impart antigen recognition, with at least eight co-stimulatory components, have already been reported4648.

The best studied second-generation CARs are those using CD28 or 4-1BB co-stimulatory domains48. Both these designs have been tested in clinical trials for relapsed B cell malignancies where they have induced major clinical responses (see below). These two variant CAR designs impart different functional and metabolic profiles upon the engineered T cell. CD28-based CARs promote brisk T cell proliferation, glucose metabolism, and self-limited T cell persistence, while 4-1BB-based CARs induce a less potent effector T cell but stimulate lipid oxidation and support greater T cell persistence4850. These differences illustrate the potential to differentially reprogram T cells using second-generation CARs.

The genetic optimization of T cells is not limited to the expression of a CAR or TCR to redirect specificity. The ease with which genes can be introduced into T cells enables the expression of multiple gene products to further shape the targeting and functional attributes of engineered T cells. The molecules used in conjunction with CARs for the purpose of increasing T cell potency or safety include a range of synthetic receptors, including chimaeric co-stimulatory receptors42,51,52, Notch-based receptors53, antigen-specific inhibitory receptors54 and others (Fig. 1c), and additional gene products designed to act in the tumour microenvironment55 or augment T cell safety56,57 (see below). Thus, while second-generation CARs have already demonstrated and validated the enormous potential of synthetic T cell engineering, one may anticipate a number of further advances in T cell therapy based on multiplexed enhancements.

Clinical proof-of-concept: the CD19 paradigm

To successfully translate CAR therapy in the clinic, one not only needs a powerful CAR but also a suitable target, which ideally is expressed on the surface of all tumour cells and absent from all normal cells, or at least any vital cells. We identified CD19 as a promising target58 based on its cell-surface expression in most leukaemia and lymphomas and its function59,60. The targeting of CD19 was expected to induce a B cell aplasia, as eventually documented in several mouse models57,6163, but deemed to be tolerable and clinically manageable if it were limited in time. We also reasoned that B cell elimination might prevent the emergence of anti-CAR antibodies, which were eventually observed in CAR T cell trials using other CARs encompassing murine components64,65. Our studies on CD19 CARs were the first to demonstrate complete eradication of established, systemic lymphoma following a single infusion of human CAR T cells in mice58, thus providing the foundation and rationale for clinical studies. Three groups reported intriguing early results in three different B cell malignancies, diffuse large B cell lymphoma66, chronic lymphocytic leukaemia (CLL)67 and acute lymphoblastic leukaemia (ALL)68. These studies focused on relapsed, chemotherapy refractory patients, and made use of either CD28 or 4-1BB-based CARs69. Notably, despite differences in disease histology, scFv, vector utilization and manufacturing process69, all groups reported remarkably high rates of complete response, especially in ALL7175 (Table 1).

Table 1.

Clinical responses to CD19 CAR therapy

Study Disease CAR V N Cond. T cells CR rate
Davila, 2014 (ref. 72) ALL (Ad.) CD28 γRV 16 CY Auto 88%
Maude, 2014 (ref. 73) ALL (Paed.) 4-1BB LV 25 CF Auto 90%
Lee, 2015 (ref. 74) ALL (Paed.) CD28 γRV 21 CY Auto 68%
Turtle, 2016 (ref. 75) ALL (Ad.) 4-1BB LV 29 CY/CF 1:14/8 93%
Qasim, 2017 (ref. 150) ALL (Paed.) 4-1BB LV 2 IC Allo 100%
Kochenderfer, 2015 (ref. 71) NHL/CLL CD28 γRV 15 CF Auto 53%
Kochenderfer, 2016 (ref. 137) B-mix CD28 γRV 20 IC Allo 30%
Turtle, 2016 (ref. 132) NHL 4-1BB LV 32 CY/CF 1:14/8 79%

Clinical responses to CD19 CAR therapy in patients with relapsed/chemorefractory B cell malignancies. These trials make use of different CAR signalling elements (CD28 or 4-1BB), different vector types (gamma-retroviral vector (γRV) or lentiviral vector (LV)) and different conditioning regimens.

Ad., adult; Paed., paediatric; V, vector type; N, number of patients; Cond., conditioning regimen; CY, cyclophosphamide; CF, cyclophosphamide – fludarabine; IC, intense conditioning; Auto T cells, autologous T cells; Allo, allogeneic T cells; 1:1 4/8, autologous T cells with defined (1:1) CD4+ to CD8+ T cell ratio; CR rate, complete remission rate.

CD19 CAR T cells must be administered following a cytotoxic conditioning regimen that depletes endogenous lymphocytes, without which no significant benefit is obtained70,76. If conditioning is too intensive, however, toxicities such as cytokine release syndrome and neurotoxicity can be exacerbated71. Whereas cytokine release is an expected outcome of T cell activation, the mechanism of neurotoxicity remains elusive. Toxicities from CAR T cells can usually be managed successfully by administering antibodies that block the interleukin-6 receptor and corticosteroids72,77, and partly mitigated by adjusting conditioning intensity and T cell dosing. Further research is needed to improve treatment and prevention strategies for such toxicities.

It is noteworthy that complete response rates have been superior in ALL compared to CLL and lymphomas, using the same CARs (Table 1). Clinical observations point to the importance of disease location, with bone marrow disease in ALL and CLL being easier to eradicate than extra-medullary sites. Clinical data on T cell biodistribution are scarce, but suggest that CAR T cell efficacy is more likely to be limited by inhibitory molecules in the tumour microenvironment than by inadequate T cell trafficking, although it remains possible that accessibility to some tumour sites limits efficacy.

Where to apply CAR therapy

The success of engineered T cells expressing CD19 CARs is a milestone in cell therapeutics, and is one of two approaches currently revolutionizing the field of cancer immunotherapy78. The other approach uses antibodies that target immune checkpoint molecules such as CTLA-4 or PD1 to enhance priming or activity of endogenous T cell responses to tumour antigens. The efficacy of immune checkpoint blockade is often incomplete but is highest in patients with tumours that carry a high burden of somatic mutations79,80. While patients with tumours with low mutation burden are typically less responsive to checkpoint blockade, it is noteworthy that it is in one such disease, ALL, that CAR therapy has proven its efficacy (Fig. 2). CAR therapy may thus be especially valuable in low mutation cancers and in those that fail checkpoint blockade therapy (Fig. 2).

Figure 2. Where to apply CAR therapy.

Figure 2

CARs are applicable in principle to any cancer for which suitable cell-surface target antigens are identified. CARs may be especially effective in cancers with low mutation burden that elude checkpoint blockade. Adapted from ref. 175 (Nature Publishing Group), previously adapted from ref. 176 (Nature Publishing Group).

The clinical benefits induced by, not a drug or an antibody, but an engineered cell, have generated enthusiasm amongst scientists, patients and industry81. Engineered T cells are a versatile platform that can be, in principle, applied to all cancers and be used in combination with other modalities. However, extending CAR therapy to more tumours requires advances in several realms, including the identification of suitable CAR targets and the generation of T cells equipped to overcome hostile tumour microenvironments.

Identification of new targets for engineered T cells

Given the success of CD19 CARs, one may anticipate similar outcomes with other leukaemia targets and haematological malignancies. Two other targets have been recently evaluated, CD22 in ALL and BCMA in myeloma. CD22 is a B cell inhibitory receptor expressed in B cell malignancies, including pre-B ALL82. Encouraging results have been obtained with CD22 CAR T cells in chemotherapy refractory paediatric patients who relapsed after CD19 CAR therapy or blinatumomab, although with a higher rate of relapse than after CD19 CAR therapy83. BCMA is a TNF superfamily receptor member expressed in myeloma cells, normal plasma cells and a subset of B cells84. A first trial of BCMA CAR-T cells in 12 subjects with chemotherapy refractory myeloma resulted in two significant responses85.

T cells are also able to eliminate solid tumours, as exemplified in TIL therapy for melanoma, cholangiocarcinoma and colorectal cancer31,32,86. TCR-engineered T cells are likewise able to mediate tumour destruction, but the identification of suitable antigens requires further research to prevent or minimize off-tumour activity25,26, and enhance efficacy. Ideal tumour antigens to target would be mutated or uniquely expressed molecules that are present in all tumour cells. Viral antigens, cancer/testis antigens and neoantigens have the advantage of being tumour-restricted, and TCRs can be identified for a number of them. However, TCR-directed therapies remain burdened by HLA-restriction and the low frequency of shared cancer neoantigens. This approach therefore requires amassing a vast collection of validated TCRs, or high-throughput efforts to rapidly derive TCRs for personalized therapy87. Notably, CARs specific for HLA–peptide complexes offer an alternative, antibody-based approach to target neoantigens8892.

Classic CAR targets are HLA-independent and more frequently shared in their expression on solid tumours of a particular histology, but they pose a different challenge in that they are more likely to be found in normal tissues. In contrast to CD19, CD22 or BCMA, which are expressed in normal but non-vital or temporarily dispensable tissues, other commonly cited targets such as ROR1, mesothelin or prostate-specific membrane antigen (PSMA), are detected in a subset of normal tissues47,93,94. The low expression of these antigens in normal tissues may avert significant toxicity, but the toxicities encountered in clinical trials targeting carbonic anhydrase IX or carcinoembryonic antigen with T cells suggest caution for clinical efforts targeting such molecules95,96. EGFRvIII, a differentially spliced EGFR variant found in glioblastoma, offers stricter tumour specificity, but its heterogeneous expression limits its potential as a target for complete tumour eradication97. IL13Rα-2 has been successfully targeted in a single case of glioblastoma, in which local CAR T cell therapy induced a complete response lasting several months98.

Alternative targeting concepts provide potential routes to bypass these limitations, based on the therapeutic window and combinatorial antigen recognition. The therapeutic window exploits differences in antigen expression such that malignant cells that express high antigen levels are eliminated while normal cells with low levels are spared. This may be implemented by tuning receptor expression or affinity to discriminate between the high and low antigen expression profiles99101. Combinatorial antigen recognition strategies offer yet another alternative. Some are designed to avoid antigen escape102105 and others to reduce off-target effects by restricting T cell activity to tumour cells that co-express the targeted antigens51. Combinatorial targeting makes use of TCRs, CARs, split-signalling receptors49,51,106,107, sequentially acting receptors53 and inhibitory receptors54 (Fig. 1c). The discovery of suitable CAR target antigens will require deeper analysis of antigen expression in both tumours and all normal tissues, and testing in relevant model systems to determine whether therapeutic efficacy and tumour selectivity are achieved.

Adapting CARs to overcome tumour microenvironments

The solid-tumour microenvironment is immunosuppressive and an obstacle for all immunotherapies108. It is comprised of multiple cell types including T cells (effector, regulatory and γδ), B cells, myeloid cells (macrophages, dendritic cells, granulocytes, monocytes), stromal cells and endothelial cells. The extracellular milieu is suboptimal for T cell function, owing to hypoxia, necrosis, acidification, nutrient shortage (glucose, glutamine, L-arginine) and an array of immunosuppressive molecules (PD-L1, IL-10, TGFβ, indolamine-2-3-dioxygenase).

The conditioning regimen usually administered before T cell infusion abates some of this resistance109, but it is not sufficient to remove or disrupt all inhibitory compartments in most instances. Additional steps need to be taken to reinforce engineered T cells to overcome micro-environmental obstacles (Fig. 3). These may consist of therapeutic combinations or the design of more efficacious T cells armed to impart resistance to inhibition.

Figure 3. Therapeutic T cell design: goals and strategies.

Figure 3

The major goals of T cell engineering address tumour targeting, T cell potency (intrinsic, that is, functionality and persistence; extrinsic: action on the tumour microenvironment), safety and cell manufacturing. Research strategies are exemplified for each one of these goals.

Small molecules that interfere with immunosuppressive cells and pathways, such as IDO inhibitors110, lenalidomide111 and adenosine antagonists112, are likely to act synergistically with engineered T cells. The Btk inhibitor ibrutinib, which is active against CLL, has also been found to improve CAR T cell function in preclinical models113. Checkpoint blockade may sustain function and persistence of engineered T cells in some instances114116, albeit not universally117. Multiple approaches have been used to interfere with immune checkpoints, including the use of blocking antibodies114,116, dominant-negative receptors114 and targeted gene disruption118.

Significantly, the latter two approaches rely on T cell engineering, thus averting the need for combining agents and the risk of cumulative toxicities. Several other approaches to adapt engineered T cells to overcome the tumour microenvironment have been recently reported (Fig. 3) and some will enter the clinic in the next year. One approach is to co-express the CAR with a cytokine, such as IL-12 or a γc cytokine62,119121. The toxicities encountered in a first trial using IL-12 caution against excessive cytokine secretion. Other approaches make use of secreted molecules to block inhibitory ligands55 or constitutive expression of co-stimulatory ligands to enhance T cell function through auto- and trans-co-stimulation122. In the latter case, the display of a ligand such as 4-1BBL serves the dual purpose of increasing intrinsic T cell persistence as well as providing targeted co-stimulation to neighbouring tumour-infiltrating T cells49.

What T cells to engineer

The anti-tumour effects of engineered T cells are predicated on their ability to migrate to tumour sites, proliferate, and mediate effector functions that destroy numerically larger tumour burdens. In addition to T cell genetic modifications that confer specificity to tumour antigens and enhance safety and efficacy, the phenotype of T cells that are engineered can affect potency123. It is now evident that the quality, efficacy, longevity and location of T cell immunity results from the diversification of naive T cells (TN) into various phenotypically distinct subsets with specific roles in protective immunity. These include memory stem (TSCM), central memory (TCM), effector memory (TEM), tissue resident memory (TRM), and highly differentiated effector (TE) T cells124. Fate mapping supports a model of progressive differentiation in which antigen-specific TN give rise to long-lived TSCM and TCM that self-renew and provide proliferating populations of shorter-lived TEM and TE cells125127 (Fig. 4a). This conceptual framework suggests that selecting less differentiated TN, TSCM or TCM subsets for genetic modification might provide cells with greater therapeutic efficacy128.

Figure 4. Cell sources for T cell engineering.

Figure 4

a, Post-thymic differentiation of memory and effector T cell subsets. T memory stem cells (TSCM) and central memory T cells (TCM) are derived following antigen activation of naive T cells (TN), and reside in low frequency in blood and higher frequency in lymphoid organs. TCM have been shown in clonogenic assays to be capable of both self-renewal and differentiation to more distal effector memory (TEM) and effector (TE) cells. TEM and TE are incapable of self-renewal, and TE, which can be present at high frequency at the peak of an immune response, are short-lived. A resident memory (TRM) subset resides in peripheral tissues but recirculates very poorly, if at all. Acquisition of effector functions increases as T cells progressively differentiate. The individual memory T cell subsets can be distinguished by the cell surface markers, which can be used for purification for genetic modification. b, Pluripotent stem-cell-derived T cells. Human T lymphocytes may be derived in culture from pluripotent stem cells. Several functionalities (cytotoxic, helper, regulatory) could be obtained in principle. αβ-like T cells normally proceed through a double-positive (CD4+CD8+) intermediate stage, which is not required for the genesis of γδ-like T cells. The stem cell platform is attractive for its ease of genetic engineering and function as a permanent T cell reservoir. T-iPS, T cell-derived induced pluripotent stem cell; eT-iPS, engineered T-iPS (here expressing a CAR).

The early clinical successes of genetically engineered T cells were accomplished by obtaining T cells from patients without regard to the phenotypic and functional heterogeneity in individual patients. Preclinical models have however shown that engineering T cells selected from TN and TCM subsets (or intermediates), or expanding TN in vitro in the presence of small molecules or cytokines that inhibit T cell differentiation, provide cell products with superior engraftment, proliferation and anti-tumour effects after adoptive transfer123,129131. These observations suggest that the potency of engineered T cells may be improved if therapeutic products were prepared from purified subsets with superior activity in preclinical models and formulated uniformly for infusion to the patient.

The concept of defining the subset formulation of engineered T cells is beginning to be explored in the clinic. The first clinical trials in which CD19 CAR T cells were engineered from selected T cell subsets and formulated in a uniform CD4/CD8 ratio demonstrated feasibility in greater than 90% of patients, including those with severe lymphopenia75,132. These studies also showed reproducible CAR T cell dose-related in vivo expansion, marked anti-tumour activity at lower cell doses, and a relationship between cell dose and toxicity that has not been clearly evident in earlier trials. Improvements in clinical cell selection methodology and use of culture conditions that retain less-differentiated phenotypes or dictate production of specific effector cytokines are areas of investigation that are likely to be implemented in future clinical applications133136.

Alternative (non-autologous) T cell sources

The successful TCR- and CAR-based therapies have to date made use of autologous T cells, which imposes individualized cell manufacturing and makes inter-patient variability unavoidable, even with selection of defined subsets. The overarching rationale for autologous approaches is to prevent a T cell attack of the recipient and rejection of the therapeutic T cells by the recipient. Immunosuppressive drugs may mitigate such complications, but are not an option because unimpeded anti-tumour function of the infused T cells is essential. The risk of exacerbating GVHD after introducing CARs in donor T cells is real, albeit variable137140, depending in part on the CAR design140.

Three strategies have been proposed to prevent GVHD. One is to transplant engineered haematopoietic stem cells or T cell precursors instead of mature T cells, thus allowing for the establishment of host tolerance. This approach however requires thymic function for positive and negative selection and yields progressive T cell reconstitution141,142. Another approach is to identify allogeneic T cells with a defined TCR specificity lacking GVHD potential143,144. VSTs may demonstrate alloreactivity in vitro but have been administered to allogeneic recipients to treat viral infections without causing serious GVHD145. However, initial clinical investigations of CAR-modified VSTs revealed limited therapeutic efficacy146, which may owe in part to the deleterious consequences of having two functioning receptors in the same T cell140. A third approach to improve safety of allogeneic T cells is to remove the TCR, which may be achieved by disruption of the TCR α or β chain147149. None of these approaches address the issue of rejection of allogeneic cells by the recipient’s immune system, and are therefore only being evaluated in heavily immunosuppressed recipients150, limiting their relevance. The latter strategies are constrained by their reliance on primary cells and the need for labour-intensive in vitro manipulations that may exhaust the replicative and engraftment potential of mature T cells123.

An alternative to manipulating mature T cells is to generate CAR T cells in vitro from pluripotent stem cells151154 (Fig. 4b). Stem cell reprogramming offers potential access to an unlimited source of therapeutic T lymphocytes and provides an excellent platform for performing additional engineering intended to enhance the therapeutic potential of induced T cells155.

How to engineer and manufacture better T cells

The genetic engineering of primary T cells began with recombinant retroviral vectors19, which remain the most used vectors in clinical studies of TCR- or CAR-modified T cells7175. Like retroviral vectors, transposons integrate semi-randomly and are thus also susceptible to variegated expression156. In hundreds of patients infused with TCR- or CAR-modified T cells, no occurrence of an oncogenic transformation has yet been reported. Alternative approaches to express the CAR rely on messenger RNA transfection, resulting in transient expression lasting only a few days157,158, and gene editing using targeted nucleases. Expressing CARs from the TCR locus affords added benefits in T cell functionality159 that warrants clinical evaluation. Targeted nucleases enabling gene ablation or addition in T cells are poised to advance therapeutic T cell engineering.

The genetic engineering, selection, expansion and differentiation of T cells have to be integrated within manufacturing platforms that meet biological and regulatory requirements (Fig. 3). T cell manufacturing is a complex science that must coalesce biological optima with efficacy, safety, reproducibility, traceability as well as regulatory and economic imperatives. Clinical T cell production is regulated under current good manufacturing practices (cGMP) and requires facilities and manufacturing processes that meet FDA guidelines. Streamlining the process to augment performance while reducing scale and cost is contingent upon understanding how to optimally purify or induce T cells that bestow efficient tumour eradication with limited side effects. Seemingly small process changes can significantly alter the potency and safety profile of expanded T cell populations. The manufacture of optimal cellular components will not only increase safety, efficacy and reproducibility, but also decrease the effective T cell dose and hence the scale of production75,160. The high cost and limited number of GMP antibodies available to sort specific subsets of T cells will hopefully be assuaged by new technologies such as acoustic waves that allow the capture, separation and concentration of desirable T cell fractions, microfluidics and new phase-change hydrogel substrates161164. These new platforms can potentially be combined with in vitro enrichment of desired T cell subsets using small molecules such as GSK-3β inhibitors or cytokines such as IL-7, IL-15 or IL-21123,165,166. These same principles apply to the induction of therapeutic T cells from induced pluripotent stem cells155. In order to mitigate costly cGMP operations and limit error-prone manual procedures, closed automated systems integrating cell-selection devices, microchips and bioreactors combined with biosensors for in situ monitoring, are being investigated167,168. New cell therapy platforms such as nanofluidic-based chip systems allowing individual cells to be isolated, cultured and assayed are becoming available for multiple applications including the characterization and manufacturing of T cell products. The rise of T cell therapies has incentivized the development of innovative manufacturing platforms and the establishment of standards driven by the formation of consortia such as CCRM (http://ccrm.ca), NIIMBL (http://www.niimbl.us) and the involvement of industry167,168. These initiatives will in time benefit all cell-based medicines.

Engineered T cells to fight infection and autoimmunity

The breakthroughs of T cell engineering in oncology have been remarkable, but the potential of engineered T cells extends far beyond cancer. One area awaiting further investigation is that of severe infectious diseases. Although early applications targeting HIV were not successful169, the potential of T cells to combat intractable infections is intriguing. The other realm is that of autoimmunity and transplantation tolerance170. CAR T cells can eliminate auto-reactive B cells as demonstrated in a mouse model of pemphigus vulgaris171. Engineered T regulatory cells may also be harnessed to dampen immune responses172, which may be useful in the context of GVHD, autoimmunity173 or transplantation tolerance174. This aspect of therapeutic T cell engineering is nascent and also poised for rapid development.

Perspectives

Twenty-five years of research on T cell engineering have culminated in the demonstration that genetically enhanced T cells can mediate profound responses in patients with refractory cancers. This progress was predicated on advances in cell engineering and manufacturing sciences that enabled successful clinical translation, the most striking example of which is CD19 CAR therapy. This new therapeutic modality is not yet fully mastered nor optimized. While high rates of complete response can be obtained in patients with B cell malignancies who do not have alternative treatment options, relapse may occur in up to half of patients, often owing to antigen loss variants. Despite progress in managing treatment-related toxicities, the mechanisms of severe cytokine release syndromes and neurotoxicities are poorly understood, precluding their effective prevention. There is therefore a need to further improve CAR therapies for B cell malignancies and other cancers.

A range of novel target antigens and CAR designs will soon be tested in haematological and solid cancers. The success of CD19 CAR therapy in some leukaemias and lymphomas is a good omen for tackling other haematological malignancies. There is, however, a need to identify suitable targets in solid tumours, whether neoantigens, cancer-testis antigens or non-mutated tumour-associated antigens, define the optimal T cell types for adoptive transfer, and address the immunosuppressive tumour microenvironment. New devices and processes are needed to decrease the cost of cell manufacturing. Autologous T cells are the corner stone of present cell-based cancer immunotherapy, but allogeneic cell sources, possibly including stem cell-derived, ‘off-the shelf’ T cells, may play an important role in the future.

While the recent therapeutic breakthroughs have all come to fruition in the realm of oncology, one may anticipate that T cell therapies will garner momentum in other fields. The success of CD19 CAR therapy and the rising proficiency of therapeutic T cell engineering is empowering all cell therapies, from immunotherapies to regenerative medicines.

Acknowledgments

The authors thank the National Cancer Institute for supporting their research via grants R01 CA114536; R01 CA136551; P01 CA59350 and P30 CA008748.

Footnotes

The authors declare competing financial interests: details are available in the online version of the paper.

Readers are welcome to comment on the online version of the paper.

Reviewer Information Nature thanks C. Melief, N. Restifo and the other anonymous reviewer(s) for their contribution to the peer review of this work.

Author Contributions M.S., I.R. and S.R. co-authored the review.

References

  • 1.Miller JF. Immunological function of the thymus. Lancet. 1961;2:748–749. doi: 10.1016/s0140-6736(61)90693-6. [DOI] [PubMed] [Google Scholar]
  • 2.Jorgensen JL, Reay PA, Ehrich EW, Davis MM. Molecular components of T-cell recognition. Annu Rev Immunol. 1992;10:835–873. doi: 10.1146/annurev.iy.10.040192.004155. [DOI] [PubMed] [Google Scholar]
  • 3.Mitchison NA. Studies on the immunological response to foreign tumor transplants in the mouse. I The role of lymph node cells in conferring immunity by adoptive transfer. J Exp Med. 1955;102:157–177. doi: 10.1084/jem.102.2.157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Klein G, Sjogren HO, Klein E, Hellstrom KE. Demonstration of resistance against methylcholanthrene-induced sarcomas in the primary autochthonous host. Cancer Res. 1960;20:1561–1572. [PubMed] [Google Scholar]
  • 5.Greenberg PD. Adoptive T cell therapy of tumors: mechanisms operative in the recognition and elimination of tumor cells. Adv Immunol. 1991;49:281–355. doi: 10.1016/s0065-2776(08)60778-6. [DOI] [PubMed] [Google Scholar]
  • 6.Melief CJ. Tumor eradication by adoptive transfer of cytotoxic T lymphocytes. Adv Cancer Res. 1992;58:143–175. doi: 10.1016/s0065-230x(08)60294-8. [DOI] [PubMed] [Google Scholar]
  • 7.Old LJ. Tumor immunology: the first century. Curr Opin Immunol. 1992;4:603–607. doi: 10.1016/0952-7915(92)90034-c. [DOI] [PubMed] [Google Scholar]
  • 8.Rosenberg SA, Lotze MT. Cancer immunotherapy using interleukin-2 and interleukin-2-activated lymphocytes. Annu Rev Immunol. 1986;4:681–709. doi: 10.1146/annurev.iy.04.040186.003341. [DOI] [PubMed] [Google Scholar]
  • 9.Rosenberg SA, Spiess P, Lafreniere R. A new approach to the adoptive immunotherapy of cancer with tumor-infiltrating lymphocytes. Science. 1986;233:1318–1321. doi: 10.1126/science.3489291. [DOI] [PubMed] [Google Scholar]
  • 10.Weiden PL, et al. Antileukemic effect of graft-versus-host disease in human recipients of allogeneic-marrow grafts. N Engl J Med. 1979;300:1068–1073. doi: 10.1056/NEJM197905103001902. [DOI] [PubMed] [Google Scholar]
  • 11.Martin PJ, Kernan NA. In: Graft vs. Host Disease: Immunology, Pathophysiology, and Treatment. Burkoff SJ, Deeg HJ, Ferrara J, Atkinson K, editors. Marcel Dekker; New York: 1990. pp. 371–387. [Google Scholar]
  • 12.Riddell SR, et al. Restoration of viral immunity in immunodeficient humans by the adoptive transfer of T cell clones. Science. 1992;257:238–241. doi: 10.1126/science.1352912. [DOI] [PubMed] [Google Scholar]
  • 13.Kolb HJ, et al. Graft-versus-leukemia effect of donor lymphocyte transfusions in marrow grafted patients. Blood. 1995;86:2041–2050. [PubMed] [Google Scholar]
  • 14.Riddell SR, Greenberg PD. Principles for adoptive T cell therapy of human viral diseases. Annu Rev Immunol. 1995;13:545–586. doi: 10.1146/annurev.iy.13.040195.002553. [DOI] [PubMed] [Google Scholar]
  • 15.Papadopoulos EB, et al. Infusions of donor leukocytes to treat Epstein–Barr virus-associated lymphoproliferative disorders after allogeneic bone marrow transplantation. N Engl J Med. 1994;330:1185–1191. doi: 10.1056/NEJM199404283301703. [DOI] [PubMed] [Google Scholar]
  • 16.Heslop HE, Brenner MK, Rooney CM. Donor T cells to treat EBV-associated lymphoma. N Engl J Med. 1994;331:679–680. doi: 10.1056/NEJM199409083311017. [DOI] [PubMed] [Google Scholar]
  • 17.Sadelain M. In: Encyclopedia of Immunobiology. Ratcliffe MJH, editor. Vol. 4. Elsevier Academic Press; 2016. [Google Scholar]
  • 18.Miller AD. Retrovirus packaging cells. Hum Gene Ther. 1990;1:5–14. doi: 10.1089/hum.1990.1.1-5. [DOI] [PubMed] [Google Scholar]
  • 19.Sadelain M, Mulligan RC. International Congress of Immunology, editor. 8th International Congress of Immunology. Springer-Verlag; Budapest; Hungary: 1992. [Google Scholar]
  • 20.Sadelain M, Rivière 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]
  • 21.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]
  • 22.Dembić Z, et al. Transfer of specificity by murine alpha and beta T-cell receptor genes. Nature. 1986;320:232–238. doi: 10.1038/320232a0. [DOI] [PubMed] [Google Scholar]
  • 23.Clay TM, et al. Efficient transfer of a tumor antigen-reactive TCR to human peripheral blood lymphocytes confers anti-tumor reactivity. J Immunol. 1999;163:507–513. [PubMed] [Google Scholar]
  • 24.Stone JD, Harris DT, Kranz DM. TCR affinity for p/MHC formed by tumor antigens that are self-proteins: impact on efficacy and toxicity. Curr Opin Immunol. 2015;33:16–22. doi: 10.1016/j.coi.2015.01.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Morgan RA, et al. Cancer regression and neurological toxicity following anti-MAGE-A3 TCR gene therapy. J Immunother. 2013;36:133–151. doi: 10.1097/CJI.0b013e3182829903. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Cameron BJ, et al. Identification of a Titin-derived HLA-A1-presented peptide as a cross-reactive target for engineered MAGE A3-directed T cells. Sci Transl Med. 2013;5:197ra103. doi: 10.1126/scitranslmed.3006034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Voss RH, et al. Molecular design of the Cαβ interface favors specific pairing of introduced TCRαβ in human T cells. J Immunol. 2008;180:391–401. doi: 10.4049/jimmunol.180.1.391. [DOI] [PubMed] [Google Scholar]
  • 28.Bialer G, Horovitz-Fried M, Ya’acobi S, Morgan RA, Cohen CJ. Selected murine residues endow human TCR with enhanced tumor recognition. J Immunol. 2010;184:6232–6241. doi: 10.4049/jimmunol.0902047. [DOI] [PubMed] [Google Scholar]
  • 29.Stone JD, et al. A novel T cell receptor single-chain signaling complex mediates antigen-specific T cell activity and tumor control. CII. 2014;63:1163–1176. doi: 10.1007/s00262-014-1586-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Ochi T, et al. Novel adoptive T-cell immunotherapy using a WT1-specific TCR vector encoding silencers for endogenous TCRs shows marked antileukemia reactivity and safety. Blood. 2011;118:1495–1503. doi: 10.1182/blood-2011-02-337089. [DOI] [PubMed] [Google Scholar]
  • 31.Robbins PF, et al. A pilot trial using lymphocytes genetically engineered with an NY-ESO-1-reactive T-cell receptor: long-term follow-up and correlates with response. Clin Cancer Res. 2015;21:1019–1027. doi: 10.1158/1078-0432.CCR-14-2708. TCR-engineered T cells induce antigen-specific responses in patients with melanoma or sarcoma. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Rapoport AP, et al. NY-ESO-1-specific TCR-engineered T cells mediate sustained antigen-specific antitumor effects in myeloma. Nat Med. 2015;21:914–921. doi: 10.1038/nm.3910. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Sadelain M, Brentjens R, Rivière I. The promise and potential pitfalls of chimeric antigen receptors. Curr Opin Immunol. 2009;21:215–223. doi: 10.1016/j.coi.2009.02.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.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]
  • 35.Romeo C, Seed B. Cellular immunity to HIV activated by CD4 fused to T cell or Fc receptor polypeptides. Cell. 1991;64:1037–1046. doi: 10.1016/0092-8674(91)90327-u. [DOI] [PubMed] [Google Scholar]
  • 36.Letourneur F, Klausner RD. T-cell and basophil activation through the cytoplasmic tail of T-cell-receptor zeta family proteins. Proc Natl Acad Sci USA. 1991;88:8905–8909. doi: 10.1073/pnas.88.20.8905. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.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 USA. 1993;90:720–724. doi: 10.1073/pnas.90.2.720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Brocker T, Peter A, Traunecker A, Karjalainen K. New simplified molecular design for functional T cell receptor. Eur J Immunol. 1993;23:1435–1439. doi: 10.1002/eji.1830230705. [DOI] [PubMed] [Google Scholar]
  • 39.Brocker T, Karjalainen K. Signals through T cell receptor-zeta chain alone are insufficient to prime resting T lymphocytes. J Exp Med. 1995;181:1653–1659. doi: 10.1084/jem.181.5.1653. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Gong MC, et al. 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]
  • 41.Brocker T. Chimeric Fv-zeta or Fv-epsilon receptors are not sufficient to induce activation or cytokine production in peripheral T cells. Blood. 2000;96:1999–2001. [PubMed] [Google Scholar]
  • 42.Krause A, et al. 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]
  • 43.Hombach A, et al. 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]
  • 44.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;20:70–75. doi: 10.1038/nbt0102-70. Human T cells expressing a second-generation CAR expand and remain functional upon repeated exposure to antigen. [DOI] [PubMed] [Google Scholar]
  • 45.Jensen MC, Riddell SR. Designing chimeric antigen receptors to effectively and safely target tumors. Curr Opin Immunol. 2015;33:9–15. doi: 10.1016/j.coi.2015.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Hinrichs CS, Restifo NP. Reassessing target antigens for adoptive T-cell therapy. Nat Biotechnol. 2013;31:999–1008. doi: 10.1038/nbt.2725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Morello A, Sadelain M, Adusumilli PS. Mesothelin-targeted CARs: driving T cells to solid tumors. Cancer Discov. 2016;6:133–146. doi: 10.1158/2159-8290.CD-15-0583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.van der Stegen SJ, Hamieh M, Sadelain M. The pharmacology of second-generation chimeric antigen receptors. Nat Rev Drug Discov. 2015;14:499–509. doi: 10.1038/nrd4597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Zhao Z, et al. Structural design of engineered costimulation determines tumor rejection kinetics and persistence of CAR T cells. Cancer Cell. 2015;28:415–428. doi: 10.1016/j.ccell.2015.09.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Kawalekar OU, et al. Distinct signaling of coreceptors regulates specific metabolism pathways and impacts memory development in CAR T cells. Immunity. 2016;44:380–390. doi: 10.1016/j.immuni.2016.01.021. [DOI] [PubMed] [Google Scholar]
  • 51.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. 2013;31:71–75. doi: 10.1038/nbt.2459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Prosser ME, Brown CE, Shami AF, Forman SJ, Jensen MC. Tumor PD-L1 co-stimulates primary human CD8+ cytotoxic T cells modified to express a PD1:CD28 chimeric receptor. Mol Immunol. 2012;51:263–272. doi: 10.1016/j.molimm.2012.03.023. [DOI] [PubMed] [Google Scholar]
  • 53.Roybal KT, et al. Precision tumor recognition by T cells with combinatorial antigen-sensing circuits. Cell. 2016;164:770–779. doi: 10.1016/j.cell.2016.01.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Fedorov VD, Themeli M, Sadelain M. PD-1- and CTLA-4-based inhibitory chimeric antigen receptors (iCARs) divert off-target immunotherapy responses. Sci Transl Med. 2013;5:215ra172. doi: 10.1126/scitranslmed.3006597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Jackson H, et al. Using CAR T Cells to Deliver Immune Modulating Agents Directly to the Tumor Microenvironment. The New York Academy of Sciences; 2016. [Google Scholar]
  • 56.Zhou X, et al. Inducible caspase-9 suicide gene controls adverse effects from alloreplete T cells after haploidentical stem cell transplantation. Blood. 2015;125:4103–4113. doi: 10.1182/blood-2015-02-628354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Paszkiewicz PJ, et al. Targeted antibody-mediated depletion of murine CD19 CAR T cells permanently reverses B cell aplasia. J Clin Invest. 2016;126:4262–4272. doi: 10.1172/JCI84813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Brentjens RJ, 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. Human T cells engineered to express a CD19-specific CAR eradicate systemic B cell malignancies in mice. [DOI] [PubMed] [Google Scholar]
  • 59.Engel P, et al. Abnormal B lymphocyte development, activation, and differentiation in mice that lack or overexpress the CD19 signal transduction molecule. Immunity. 1995;3:39–50. doi: 10.1016/1074-7613(95)90157-4. [DOI] [PubMed] [Google Scholar]
  • 60.Rickert RC, Rajewsky K, Roes J. Impairment of T-cell-dependent B-cell responses and B-1 cell development in CD19-defficient mice. Nature. 1995;376:352–355. doi: 10.1038/376352a0. [DOI] [PubMed] [Google Scholar]
  • 61.Kochenderfer JN, Yu Z, Frasheri D, Restifo NP, Rosenberg SA. Adoptive transfer of syngeneic T cells transduced with a chimeric antigen receptor that recognizes murine CD19 can eradicate lymphoma and normal B cells. Blood. 2010;116:3875–3886. doi: 10.1182/blood-2010-01-265041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Pegram HJ, et al. Tumor-targeted T cells modified to secrete IL-12 eradicate systemic tumors without need for prior conditioning. Blood. 2012;119:4133–4141. doi: 10.1182/blood-2011-12-400044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Davila ML, Kloss CC, Gunset G, Sadelain M. CD19 CAR-targeted T cells induce long-term remission and B cell aplasia in an immunocompetent mouse model of B cell acute lymphoblastic leukemia. PLoS One. 2013;8:e61338. doi: 10.1371/journal.pone.0061338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Lamers CH, et al. Immune responses to transgene and retroviral vector in patients treated with ex vivo-engineered T cells. Blood. 2011;117:72–82. doi: 10.1182/blood-2010-07-294520. [DOI] [PubMed] [Google Scholar]
  • 65.Maus MV, et al. T cells expressing chimeric antigen receptors can cause anaphylaxis in humans. Cancer Immunol Res. 2013;1:26–31. doi: 10.1158/2326-6066.CIR-13-0006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Kochenderfer JN, 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;116:4099–4102. doi: 10.1182/blood-2010-04-281931. First report of a clinical response to CD19 CAR therapy in non-Hodgkin lymphoma. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Kalos M, 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. doi: 10.1126/scitranslmed.3002842. First report of clinical response to CD19 in CAR therapy in chronic lymphocytic leukaemia. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Brentjens RJ, et al. CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia. Sci Transl Med. 2013;5:177ra38. doi: 10.1126/scitranslmed.3005930. First report of clinical responses to CD19 CAR therapy in acute lymphoblastic leukaemia. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Sadelain M. CAR therapy: the CD19 paradigm. J Clin Invest. 2015;125:3392–3400. doi: 10.1172/JCI80010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Brentjens RJ, 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;118:4817–4828. doi: 10.1182/blood-2011-04-348540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Kochenderfer JN, et al. Chemotherapy-refractory diffuse large B-cell lymphoma and indolent B-cell malignancies can be effectively treated with autologous T cells expressing an anti-CD19 chimeric antigen receptor. J Clin Oncol. 2015;33:540–549. doi: 10.1200/JCO.2014.56.2025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Davila ML, et al. Efficacy and toxicity management of 19–28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci Transl Med. 2014;6:224ra25. doi: 10.1126/scitranslmed.3008226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Maude SL, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med. 2014;371:1507–1517. doi: 10.1056/NEJMoa1407222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Lee DW, et al. T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial. Lancet. 2015;385:517–528. doi: 10.1016/S0140-6736(14)61403-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Turtle CJ, et al. CD19 CAR-T cells of defined CD4+:CD8+ composition in adult B cell ALL patients. J Clin Invest. 2016;126:2123–2138. doi: 10.1172/JCI85309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Savoldo B, et al. CD28 costimulation improves expansion and persistence of chimeric antigen receptor-modified T cells in lymphoma patients. J Clin Invest. 2011;121:1822–1826. doi: 10.1172/JCI46110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Lee DW, et al. Current concepts in the diagnosis and management of cytokine release syndrome. Blood. 2014;124:188–195. doi: 10.1182/blood-2014-05-552729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Couzin-Frankel J. Cancer immunotherapy. Science. 2013;342:1432–1433. doi: 10.1126/science.342.6165.1432. [DOI] [PubMed] [Google Scholar]
  • 79.Rizvi NA, et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science. 2015;348:124–128. doi: 10.1126/science.aaa1348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Snyder A, et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N Engl J Med. 2014;371:2189–2199. doi: 10.1056/NEJMoa1406498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.June CH, Riddell SR, Schumacher TN. Adoptive cellular therapy: a race to the finish line. Sci Transl Med. 2015;7:280ps7. doi: 10.1126/scitranslmed.aaa3643. [DOI] [PubMed] [Google Scholar]
  • 82.Haso W, et al. Anti-CD22-chimeric antigen receptors targeting B-cell precursor acute lymphoblastic leukemia. Blood. 2013;121:1165–1174. doi: 10.1182/blood-2012-06-438002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Anti-CD22 CAR therapy leads to ALL remissions. Cancer Discov. 2017;7:120. doi: 10.1158/2159-8290.CD-NB2017-001. http://dx.doi.org/10.1158/2159-8290.CD-NB2017-001. [DOI] [PubMed] [Google Scholar]
  • 84.Carpenter RO, et al. B-cell maturation antigen is a promising target for adoptive T-cell therapy of multiple myeloma. Clin Cancer Res. 2013;19:2048–2060. doi: 10.1158/1078-0432.CCR-12-2422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Ali SA, et al. T cells expressing an anti-B-cell maturation antigen chimeric antigen receptor cause remissions of multiple myeloma. Blood. 2016;128:1688–1700. doi: 10.1182/blood-2016-04-711903. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Robbins PF, et al. Tumor regression in patients with metastatic synovial cell sarcoma and melanoma using genetically engineered lymphocytes reactive with NY-ESO-1. J Clin Oncol. 2011;29:917–924. doi: 10.1200/JCO.2010.32.2537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Lu YC, Robbins PF. Targeting neoantigens for cancer immunotherapy. Int Immunol. 2016;28:365–370. doi: 10.1093/intimm/dxw026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Stewart-Jones G, et al. Rational development of high-affinity T-cell receptor-like antibodies. Proc Natl Acad Sci USA. 2009;106:5784–5788. doi: 10.1073/pnas.0901425106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Zhang G, et al. Anti-melanoma activity of T cells redirected with a TCR-like chimeric antigen receptor. Sci Rep. 2014;4:3571. doi: 10.1038/srep03571. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Oren R, et al. Functional comparison of engineered T cells carrying a native TCR versus TCR-like antibody-based chimeric antigen receptors indicates affinity/avidity thresholds. J Immunol. 2014;193:5733–5743. doi: 10.4049/jimmunol.1301769. [DOI] [PubMed] [Google Scholar]
  • 91.Inaguma Y, et al. Construction and molecular characterization of a T-cell receptor-like antibody and CAR-T cells specific for minor histocompatibility antigen HA-1H. Gene Ther. 2014;21:575–584. doi: 10.1038/gt.2014.30. [DOI] [PubMed] [Google Scholar]
  • 92.Maus MV, et al. An MHC-restricted antibody-based chimeric antigen receptor requires TCR-like affinity to maintain antigen specificity. Mol Ther. 2017;3:16023. doi: 10.1038/mto.2016.23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Balakrishnan A, et al. Analysis of ROR1 protein expression in human cancer and normal tissues. Clin Cancer Res. 2016 doi: 10.1158/1078-0432.CCR-16-2083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.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;3:81–85. [PubMed] [Google Scholar]
  • 95.Lamers CH, 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–e22. doi: 10.1200/JCO.2006.05.9964. [DOI] [PubMed] [Google Scholar]
  • 96.Parkhurst MR, et al. T cells targeting carcinoembryonic antigen can mediate regression of metastatic colorectal cancer but induce severe transient colitis. Mol Ther. 2011;19:620–626. doi: 10.1038/mt.2010.272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Johnson LA, et al. Rational development and characterization of humanized anti-EGFR variant III chimeric antigen receptor T cells for glioblastoma. Sci Transl Med. 2015;7:275ra22. doi: 10.1126/scitranslmed.aaa4963. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Brown CE, et al. Regression of glioblastoma after chimeric antigen receptor T-Cell Therapy. N Engl J Med. 2016;375:2561–2569. doi: 10.1056/NEJMoa1610497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Hudecek M, et al. Receptor affinity and extracellular domain modifications affect tumor recognition by ROR1-specific chimeric antigen receptor T cells. Clin Cancer Res. 2013;19:3153–3164. doi: 10.1158/1078-0432.CCR-13-0330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Caruso HG, et al. Tuning sensitivity of CAR to EGFR density limits recognition of normal tissue while maintaining potent antitumor activity. Cancer Res. 2015;75:3505–3518. doi: 10.1158/0008-5472.CAN-15-0139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Liu X, et al. Affinity-tuned ErbB2 or EGFR chimeric antigen receptor T cells exhibit an increased therapeutic index against tumors in mice. Cancer Res. 2015;75:3596–3607. doi: 10.1158/0008-5472.CAN-15-0159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.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. doi: 10.2217/imt.10.81. [DOI] [PubMed] [Google Scholar]
  • 103.Wilkie S, et al. Dual targeting of ErbB2 and MUC1 in breast cancer using chimeric antigen receptors engineered to provide complementary signaling. J Clin Immunol. 2012;32:1059–1070. doi: 10.1007/s10875-012-9689-9. [DOI] [PubMed] [Google Scholar]
  • 104.Zah E, Lin MY, Silva-Benedict A, Jensen MC, Chen YY. ADDENDUM: T cells expressing CD19/CD20 bispecific chimeric antigen receptors prevent antigen escape by malignant B cells. Cancer Immunol Res. 2016;4:639–641. doi: 10.1158/2326-6066.CIR-16-0108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Hegde M, et al. Tandem CAR T cells targeting HER2 and IL13Rα2 mitigate tumor antigen escape. J Clin Invest. 2016;126:3036–3052. doi: 10.1172/JCI83416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Wu CY, Roybal KT, Puchner EM, Onuffer J, Lim WA. Remote control of therapeutic T cells through a small molecule-gated chimeric receptor. Science. 2015;350:aab4077. doi: 10.1126/science.aab4077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Sun J, Sadelain M. The quest for spatio-temporal control of CAR T cells. Cell Res. 2015;25:1281–1282. doi: 10.1038/cr.2015.131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Mellman I, Coukos G, Dranoff G. Cancer immunotherapy comes of age. Nature. 2011;480:480–489. doi: 10.1038/nature10673. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Wrzesinski C, et al. Increased intensity lymphodepletion enhances tumor treatment efficacy of adoptively transferred tumor-specific T cells. J Immunother. 2010;33:1–7. doi: 10.1097/CJI.0b013e3181b88ffc. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Munn DH. Blocking IDO activity to enhance anti-tumor immunity. Front Biosci (Elite Ed) 2012;4:734–745. doi: 10.2741/e414. [DOI] [PubMed] [Google Scholar]
  • 111.Ramsay AG, Clear AJ, Fatah R, Gribben JG. Multiple inhibitory ligands induce impaired T-cell immunologic synapse function in chronic lymphocytic leukemia that can be blocked with lenalidomide: establishing a reversible immune evasion mechanism in human cancer. Blood. 2012;120:1412–1421. doi: 10.1182/blood-2012-02-411678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Young A, Mittal D, Stagg J, Smyth MJ. Targeting cancer-derived adenosine: new therapeutic approaches. Cancer Discov. 2014;4:879–888. doi: 10.1158/2159-8290.CD-14-0341. [DOI] [PubMed] [Google Scholar]
  • 113.Ruella M, et al. Kinase inhibitor ibrutinib to prevent cytokine-release syndrome after anti-CD19 chimeric antigen receptor T cells for B-cell neoplasms. Leukemia. 2017;31:246–248. doi: 10.1038/leu.2016.262. [DOI] [PubMed] [Google Scholar]
  • 114.Cherkassky L, et al. Human CAR T cells with cell-intrinsic PD-1 checkpoint blockade resist tumor-mediated inhibition. J Clin Invest. 2016;126:3130–3144. doi: 10.1172/JCI83092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Chong EA, et al. PD-1 blockade modulates chimeric antigen receptor (CAR) modified T cells and induces tumor regression: refueling the CAR. Blood. 2017;129:1039–1041. doi: 10.1182/blood-2016-09-738245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.John LB, et al. Anti-PD-1 antibody therapy potently enhances the eradication of established tumors by gene-modified T cells. Clin Cancer Res. 2013;19:5636–5646. doi: 10.1158/1078-0432.CCR-13-0458. [DOI] [PubMed] [Google Scholar]
  • 117.Condomines M, et al. Tumor-targeted human T cells expressing CD28-based chimeric antigen receptors circumvent CTLA-4 inhibition. PLoS One. 2015;10:e0130518. doi: 10.1371/journal.pone.0130518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Menger L, et al. TALEN-mediated inactivation of PD-1 in tumor-reactive lymphocytes promotes intratumoral T-cell persistence and rejection of established tumors. Cancer Res. 2016;76:2087–2093. doi: 10.1158/0008-5472.CAN-15-3352. [DOI] [PubMed] [Google Scholar]
  • 119.Hoyos V, et al. Engineering CD19-specific T lymphocytes with interleukin-15 and a suicide gene to enhance their anti-lymphoma/leukemia effects and safety. Leukemia. 2010;24:1160–1170. doi: 10.1038/leu.2010.75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.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;115:3508–3519. doi: 10.1182/blood-2009-09-241398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Chinnasamy D, et al. Local delivery of interleukin-12 using T cells targeting VEGF receptor-2 eradicates multiple vascularized tumors in mice. Clin Cancer Res. 2012;18:1672–1683. doi: 10.1158/1078-0432.CCR-11-3050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Stephan MT, et al. 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]
  • 123.Gattinoni L, Klebanoff CA, Restifo NP. Paths to stemness: building the ultimate antitumour T cell. Nat Rev Cancer. 2012;12:671–684. doi: 10.1038/nrc3322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Farber DL, Yudanin NA, Restifo NP. Human memory T cells: generation, compartmentalization and homeostasis. Nat Rev Immunol. 2014;14:24–35. doi: 10.1038/nri3567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Buchholz VR, et al. Disparate individual fates compose robust CD8+ T cell immunity. Science. 2013;340:630–635. doi: 10.1126/science.1235454. [DOI] [PubMed] [Google Scholar]
  • 126.Gerlach C, et al. Heterogeneous differentiation patterns of individual CD8+ T cells. Science. 2013;340:635–639. doi: 10.1126/science.1235487. [DOI] [PubMed] [Google Scholar]
  • 127.Graef P, et al. Serial transfer of single-cell-derived immunocompetence reveals stemness of CD8+ central memory T cells. Immunity. 2014;41:116–126. doi: 10.1016/j.immuni.2014.05.018. [DOI] [PubMed] [Google Scholar]
  • 128.Busch DH, Fräßle SP, Sommermeyer D, Buchholz VR, Riddell SR. Role of memory T cell subsets for adoptive immunotherapy. Semin Immunol. 2016;28:28–34. doi: 10.1016/j.smim.2016.02.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Gattinoni L, et al. Acquisition of full effector function in vitro paradoxically impairs the in vivo antitumor efficacy of adoptively transferred CD8+ T cells. J Clin Invest. 2005;115:1616–1626. doi: 10.1172/JCI24480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Berger C, et al. 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]
  • 131.Hinrichs CS, et al. Adoptively transferred effector cells derived from naive rather than central memory CD8+ T cells mediate superior antitumor immunity. Proc Natl Acad Sci USA. 2009;106:17469–17474. doi: 10.1073/pnas.0907448106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Turtle CJ, et al. Immunotherapy of non-Hodgkin’s lymphoma with a defined ratio of CD8+ and CD4+ CD19-specific chimeric antigen receptor-modified T cells. Sci Transl Med. 2016;8:355ra116. doi: 10.1126/scitranslmed.aaf8621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Stemberger C, et al. Novel serial positive enrichment technology enables clinical multiparameter cell sorting. PLoS One. 2012;7:e35798. doi: 10.1371/journal.pone.0035798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.van der Waart AB, et al. Inhibition of Akt signaling promotes the generation of superior tumor-reactive T cells for adoptive immunotherapy. Blood. 2014;124:3490–3500. doi: 10.1182/blood-2014-05-578583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Kagoya Y, et al. BET bromodomain inhibition enhances T cell persistence and function in adoptive immunotherapy models. J Clin Invest. 2016;126:3479–3494. doi: 10.1172/JCI86437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Sabatino M, et al. Generation of clinical-grade CD19-specific CAR-modified CD8+ memory stem cells for the treatment of human B-cell malignancies. Blood. 2016;128:519–528. doi: 10.1182/blood-2015-11-683847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Kochenderfer JN, et al. Donor-derived CD19-targeted T cells cause regression of malignancy persisting after allogeneic hematopoietic stem cell transplantation. Blood. 2013;122:4129–4139. doi: 10.1182/blood-2013-08-519413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Brudno JN, et al. Allogeneic T cells that express an anti-CD19 chimeric antigen receptor induce remissions of B-cell malignancies that progress after allogeneic hematopoietic stem-cell transplantation without causing graft-versus-host disease. J Clin Oncol. 2016;34:1112–1121. doi: 10.1200/JCO.2015.64.5929. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Jacoby E, et al. Murine allogeneic CD19 CAR T cells harbor potent antileukemic activity but have the potential to mediate lethal GVHD. Blood. 2016;127:1361–1370. doi: 10.1182/blood-2015-08-664250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Ghosh A, et al. Donor CD19 CAR T cells exert potent graft-versus-lymphoma activity with diminished graft-versus-host activity. Nat Med. 2017;23:242–249. doi: 10.1038/nm.4258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Yang L, Baltimore D. Long-term in vivo provision of antigen-specific T cell immunity by programming hematopoietic stem cells. Proc Natl Acad Sci USA. 2005;102:4518–4523. doi: 10.1073/pnas.0500600102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Zakrzewski JL, 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]
  • 143.Melenhorst JJ, et al. Allogeneic virus-specific T cells with HLA alloreactivity do not produce GVHD in human subjects. Blood. 2010;116:4700–4702. doi: 10.1182/blood-2010-06-289991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.O’Reilly RJ, Prockop S, Hasan AN, Koehne G, Doubrovina E. Virus-specific T-cell banks for ‘off the shelf’ adoptive therapy of refractory infections. Bone Marrow Transplant. 2016;51:1163–1172. doi: 10.1038/bmt.2016.17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Leen AM, et al. Multicenter study of banked third-party virus-specific T cells to treat severe viral infections after hematopoietic stem cell transplantation. Blood. 2013;121:5113–5123. doi: 10.1182/blood-2013-02-486324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Cruz CR, et al. Infusion of donor-derived CD19-redirected virus-specific T cells for B-cell malignancies relapsed after allogeneic stem cell transplant: a phase 1 study. Blood. 2013;122:2965–2973. doi: 10.1182/blood-2013-06-506741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Provasi E, et al. Editing T cell specificity towards leukemia by zinc finger nucleases and lentiviral gene transfer. Nat Med. 2012;18:807–815. doi: 10.1038/nm.2700. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Torikai H, et al. A foundation for universal T-cell based immunotherapy: T cells engineered to express a CD19-specific chimeric-antigen-receptor and eliminate expression of endogenous TCR. Blood. 2012;119:5697–5705. doi: 10.1182/blood-2012-01-405365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Berdien B, Mock U, Atanackovic D, Fehse B. TALEN-mediated editing of endogenous T-cell receptors facilitates efficient reprogramming of T lymphocytes by lentiviral gene transfer. Gene Ther. 2014;21:539–548. doi: 10.1038/gt.2014.26. [DOI] [PubMed] [Google Scholar]
  • 150.Qasim W, et al. Molecular remission of infant B-ALL after infusion of universal TALEN gene-edited CAR T cells. Sci Transl Med. 2017;9:eaaj2013. doi: 10.1126/scitranslmed.aaj2013. Talen-mediated TCR deletion enables CD19 CAR therapy with donor T cells in allogeneic recipients. [DOI] [PubMed] [Google Scholar]
  • 151.Timmermans F, et al. Generation of T cells from human embryonic stem cell-derived hematopoietic zones. J Immunol. 2009;182:6879–6888. doi: 10.4049/jimmunol.0803670. [DOI] [PubMed] [Google Scholar]
  • 152.Vizcardo R, et al. Regeneration of human tumor antigen-specific T cells from iPSCs derived from mature CD8+ T cells. Cell Stem Cell. 2013;12:31–36. doi: 10.1016/j.stem.2012.12.006. [DOI] [PubMed] [Google Scholar]
  • 153.Nishimura T, et al. Generation of rejuvenated antigen-specific T cells by reprogramming to pluripotency and redifferentiation. Cell Stem Cell. 2013;12:114–126. doi: 10.1016/j.stem.2012.11.002. [DOI] [PubMed] [Google Scholar]
  • 154.Themeli M, et al. Generation of tumor-targeted human T lymphocytes from induced pluripotent stem cells for cancer therapy. Nat Biotechnol. 2013;31:928–933. doi: 10.1038/nbt.2678. Human CAR T cells generated in vitro from pluripotent stem cells induce tumour regression in mice. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Themeli M, Rivière I, Sadelain M. New cell sources for T cell engineering and adoptive immunotherapy. Cell Stem Cell. 2015;16:357–366. doi: 10.1016/j.stem.2015.03.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Singh H, Huls H, Kebriaei P, Cooper LJ. A new approach to gene therapy using Sleeping Beauty to genetically modify clinical-grade T cells to target CD19. Immunol Rev. 2014;257:181–190. doi: 10.1111/imr.12137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Zhao Y, et al. High-efficiency transfection of primary human and mouse T lymphocytes using RNA electroporation. Mol Ther. 2006;13:151–159. doi: 10.1016/j.ymthe.2005.07.688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Birkholz K, 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. doi: 10.1038/gt.2008.189. [DOI] [PubMed] [Google Scholar]
  • 159.Eyquem J, et al. Targeting a CAR to the TRAC locus with CRISPR/Cas9 enhances tumour rejection. Nature. 2017;543:113–117. doi: 10.1038/nature21405. CRISPR–Cas9-mediated CAR delivery to the TCR locus delays T cell differentiation and exhaustion of CAR T cells resulting in improved CAR T cell therapeutic efficacy. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Wang X, Rivière I. Manufacture of tumor- and virus-specific T lymphocytes for adoptive cell therapies. Cancer Gene Ther. 2015;22:85–94. doi: 10.1038/cgt.2014.81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Chiou PY, Ohta AT, Wu MC. Massively parallel manipulation of single cells and microparticles using optical images. Nature. 2005;436:370–372. doi: 10.1038/nature03831. [DOI] [PubMed] [Google Scholar]
  • 162.Kennedy DR, Gerhardson T, Sporbert B, Mealey D, Rust MJ, Lipkens B. The Concentration and separation of blood components using acoustic radiation force. Blood. 2013;122:3665. [Google Scholar]
  • 163.Urbansky A, Lenshof A, Dykes J, Laurell T, Scheding S. Separation of lymphocyte populations from peripheral blood progenitor cell products using affinity bead acoustophoresis. Blood. 2014;124:315–315. doi: 10.1002/cyto.a.22507. [DOI] [PubMed] [Google Scholar]
  • 164.Jesuraj NJ, et al. A novel phase-change hydrogel substrate for t cell activation promotes increased expansion of CD8+ cells expressing central memory and naive phenotype markers. Blood. 2016;128:3368–3368. [Google Scholar]
  • 165.Cieri N, et al. IL-7 and IL-15 instruct the generation of human memory stem T cells from naive precursors. Blood. 2013;121:573–584. doi: 10.1182/blood-2012-05-431718. [DOI] [PubMed] [Google Scholar]
  • 166.Chapuis AG, et al. T-cell therapy using interleukin-21-primed cytotoxic T-cell lymphocytes combined with cytotoxic T-cell lymphocyte antigen-4 blockade results in long-term cell persistence and durable tumor regression. J Clin Oncol. 2016;34:3787–3795. doi: 10.1200/JCO.2015.65.5142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Kaiser AD, et al. Towards a commercial process for the manufacture of genetically modified T cells for therapy. Cancer Gene Ther. 2015;22:72–78. doi: 10.1038/cgt.2014.78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Roh KH, Nerem RM, Roy K. Biomanufacturing of therapeutic cells: state of the art, current challenges, and future perspectives. Annu Rev Chem Biomol Eng. 2016;7:455–478. doi: 10.1146/annurev-chembioeng-080615-033559. [DOI] [PubMed] [Google Scholar]
  • 169.Deeks SG, et al. A phase II randomized study of HIV-specific T-cell gene therapy in subjects with undetectable plasma viremia on combination antiretroviral therapy. Mol Ther. 2002;5:788–797. doi: 10.1006/mthe.2002.0611. [DOI] [PubMed] [Google Scholar]
  • 170.DuPage M, Bluestone JA. Harnessing the plasticity of CD4+ T cells to treat immune-mediated disease. Nat Rev Immunol. 2016;16:149–163. doi: 10.1038/nri.2015.18. [DOI] [PubMed] [Google Scholar]
  • 171.Ellebrecht CT, et al. Reengineering chimeric antigen receptor T cells for targeted therapy of autoimmune disease. Science. 2016;353:179–184. doi: 10.1126/science.aaf6756. CAR T cells can treat autoimmunity in a murine model of pemphigus vulgaris. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Lee JC, 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;71:2871–2881. doi: 10.1158/0008-5472.CAN-10-0552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Blat D, Zigmond E, Alteber Z, Waks T, Eshhar Z. Suppression of murine colitis and its associated cancer by carcinoembryonic antigen-specific regulatory T cells. Mol Ther. 2014;22:1018–1028. doi: 10.1038/mt.2014.41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Boardman DA, et al. Expression of a chimeric antigen receptor specific for donor HLA class I enhances the potency of human regulatory T cells in preventing human skin transplant rejection. Am J Transplant. 2017;17:931–943. doi: 10.1111/ajt.14185. [DOI] [PubMed] [Google Scholar]
  • 175.Alexandrov LB, et al. Signatures of mutational processes in human cancer. Nature. 2013;500:415–421. doi: 10.1038/nature12477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Lawrence MS, et al. Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature. 2013;499:214–218. doi: 10.1038/nature12213. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES