You are on page 1of 25

Marco Ruella Adoptive immunotherapy for

Michael Kalos
cancer

Authors’ address Summary: Recent clinical success has underscored the potential for
Marco Ruella1, Michael Kalos1 immunotherapy based on the adoptive cell transfer (ACT) of engi-
1
Department of Pathology and Laboratory Medicine, neered T lymphocytes to mediate dramatic, potent, and durable clinical
University of Pennsylvania, Philadelphia, PA, USA. responses. This success has led to the broader evaluation of engineered
T-lymphocyte-based adoptive cell therapy to treat a broad range of
Correspondence to: malignancies. In this review, we summarize concepts, successes, and
Michael Kalos challenges for the broader development of this promising field, focus-
Lilly Research Laboratories ing principally on lessons gleaned from immunological principles and
Eli Lilly and Company clinical thought. We present ACT in the context of integrating T-cell
450 East, 29th Street and tumor biology and the broader systemic immune response.
New York, NY 10016, USA
Tel.: +1 646-638-5095 Keywords: adoptive cell therapy, immunotherapy, chimeric antigen receptor, cancer,
e-mail: kalos_michael_d@lilly.com immune modulation, tumor microenvironment

Acknowledgements
We are grateful to the multitude of scientists who have laid
the foundations for the field, and to the patients who have Introduction
allowed, through their bravery and generosity, the clinical
development of this promising treatment modality. MR is
The principal biological role of the immune system is to
supported by funds from the “Societa Italiana di Ematologia maintain the integrity of ‘self’. This integrity is maintained
Sperimentale” (SIES), from “Associazione Italiana Pazienti Emopatici” principally by eliminating and destroying diseased and
(AIPE) and AFCRI fund number 990-9936-4-900278-
5043-2433-1410 from the University of Pennsylvania. The
infected cells, while ensuring that healthy cells and tissues
authors have read the journal’s policy on disclosure of are not targeted. A vastly complex network of events that
potential conflict of interest. MR has nothing to declare. MK bring together both innate and adaptive immune arm
is named on patents related to CART19 technology with the
potential for royalties and milestone payments from the
through both cell mediated and soluble interactions is
development and commercialization of CART19 technology. responsible for maintaining the integrity of self. The net-
work of cells includes, among others, T and B lymphocytes,
natural killer (NK) cells, antigen-presenting cells (APCs)
This article is part of a series of reviews
such as dendritic cells (DCs) and macrophages, stromal,
covering Adoptive Immunotherapy for
Cancer appearing in Volume 257 of endothelial, and epithelial cells. This network of cells is
Immunological Reviews. coordinated by a myriad of soluble factors to sense both
Video podcast available
health and damage and in turn orchestrate the immune sys-
Go to tem to clear the body of damaged cells and protect the body
www.immunologicalreviews.com to from infections. Data from patients with inherited immune
watch an interview with Guest Editor
Dr Carl June.
deficiencies have revealed the critical role for an intact
immune system for organismal health, with autoimmune
disease and increased risks of developing infections and
tumors a consequence of disruptions in immune network
functions (1, 2).
Immunological Reviews 2014
Vol. 257: 14–38
Broadly speaking, immune cells can be defined as having
Printed in Singapore. All rights reserved either effector or regulatory functions in the immune system.
The main known effector cells of the immune system are
© 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
Immunological Reviews understood to be T and NK lymphocytes, mediating,
0105-2896
© 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
14 Immunological Reviews 257/2014
Ruella & Kalos  Adoptive immunotherapy for cancer

respectively, adaptive and innate immunity. The variety of NK-dependent anti-tumor cytolytic activity, block inhibi-
cells with immune regulatory activities is considerably tory receptors, or modulate the tumor microenvironment
broader, and includes cells of lymphoid origin such as (TMA) (14–19). Unfortunately, each of these approaches
CD4+ helper and regulatory cells and B cells, and myeloid has been compromised by the impact of central and
origin such monocytes, macrophage, myeloid derived DCs, peripheral immune tolerance, which presents a fundamen-
and myeloid derived suppressor cells. Perhaps not surpris- tal impediment for the effective targeting of tumor cells
ingly given the breadth of the immune system, cells not by immune effector cells. Specifically, central tolerance, by
classically considered to be involved in the immune systems deleting the T-cell repertoire to self-antigens often overex-
such endothelial, stromal, and epithelial cells have over the pressed or aberrantly expressed by tumors, effectively
past few years been shown to have immune regulatory results in the blunting of the potent anti-tumor T cell rep-
functions (3–6). The relevance of these cell types in the reg- ertoire to such antigens. On the other hand, peripheral
ulation of a potent immune response is revealed by the fact tolerance, commonly exploited by tumors, blunts the abil-
that tumors often evolve under selective pressure by the ity of existing effector lymphocytes (T cells, NK cells) to
immune system to modulate the biology of these cells and target tumor cells by establishing an immune-suppressive
blunt anti-tumor immunity (7). state.
Abundant evidence exists to document that the immune The relevance of peripheral tolerance in blunting the anti-
system plays an essential role in controlling the growth of tumor immune response has been highlighted over the past
tumors. This evidence has been accumulated from the few years in clinical trials to evaluate antibodies which block
study of animal models with defined immune defects (8), immune-modulating molecules. In these trials, blocking the
from the increased frequency of cancers in patients with immune modulatory activity of cytotoxic T-lymphocyte anti-
naturally occurring or acquired immune deficiencies (9), gen-4 (CTLA4) (CD152) and programmed death-1 (PD1)
and also from the observation that tumors often evolve to (CD279) has resulted in potent anti-tumor activity, with
become less visible to the immune system. Early efforts to document long lasting complete responses in a subset of
target cancer through manipulating the immune system in patients (20–22); these results have led to the first-in-class
fact predated the ‘discovery’ of the immune system, most approval of an anti-CTLA4 antibody (ipilumimab/Yervoy),
famously by William Coley who applied heat killed strepto- and the likely approval of PD1-modulating agents in the
cocci (‘Coley’s toxin’) at tumor sites to induce inflamma- near future.
tion with consequent tumor regressions (10). More Overcoming central tolerance poses a unique challenge,
recently, the tuberculosis vaccine of Calmette-Guerin (BCG) since the impact of central tolerance is the lack of a potent
has been used with success to treat localized bladder neo- anti-tumor T-cell repertoire to self-antigens. One approach
plasm (11), while adjuvants based on bacterial components to overcome central tolerance is the transfer into patients of
that trigger immunity via engagement of Toll-like-receptors potent effector cells, as exemplified by the efficacy of allore-
(TLRs) on immune cells are essential components of FDA- active T cells in the context of allogeneic cell transplantation.
approved vaccine preparations. Direct evidence of the Practical limitations in terms of availability of suitable
potency of effector T cells to target and eradicate tumor donors as well as GVH-related toxicity issues have limited
cells was demonstrated through the clinical application of the broader use of allogeneic cell transplantation. One alter-
donor lymphocyte infusion (DLI) to treat leukemia after native to allogeneic cell transplantation is adoptive cell trans-
allogeneic hematopoietic stem cell transplantation (HSCT), fer (ACT), which involves the infusion into patients of
with the Graft-versus-leukemia (GVL) effect mediated by autologous lymphocytes following ex vivo expansion. Initial
alloreactive donor T cells leading to strong anti-leukemic efforts to apply ACT in tumor immunotherapy involved
responses in a significant portion of relapsing patients (12, transfer of bulk T-cell populations into patients, under the
13). premise that ex vivo manipulation of cells had the potential to
As the field of immunotherapy has unfolded over the lower activation thresholds of potential tumor-reactive cells
past two decades, a number of strategies have been evalu- as well as to expand tumor-specific T cells. The promise of
ated to target cancer. Such immunotherapeutic strategies this approach was revealed in clinical trials which showed
have included a plethora of vaccine-based strategies to that this strategy had the potential to rapidly reconstitute
trigger anti-tumor cellular and humoral immunity, anti- host immunity (23–25). Recent advances in the ability to
body-based strategies to mediate complement and effectively engineer T lymphocytes have provided an
© 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
Immunological Reviews 257/2014 15
Ruella & Kalos  Adoptive immunotherapy for cancer

unprecedented opportunity to be able to expand on the Accordingly, the principal focus in the field of ACT
potential of T-cell transfer and have ushered in what prom- shifted toward a better understanding of the properties of
ises to be a golden age for T-cell-based immunotherapy the effector cells that were important to mediate and drive
based on principles of synthetic biology (26) (Fig. 1). the observed anti-tumor activity and the development of
Indeed, clinical results from recent trials employing engi- improved approaches to isolate, manipulate, and expand
neered T cells are demonstrating the dramatic potency of such potent effector T cells.
this approach to target cancer (27–31).
This review focuses on principles of ACT in the context Immune effector cells in ACT
of recent conceptual and technical advances that are driving Two effector cell types, T lymphocytes and NK cells have
the field in this emerging era of synthetic biology. We been principally evaluated for ACT. The essential properties
describe the state of the art for the field with regard to cell of these cells that have supported their evaluation have been
types, engineering approaches, and clinical experience. the fact that these cells are capable of cytotoxic targeting,
Importantly, we focus on viewing ACT in the context of the and that these cells can be readily isolated and manipulated
systemic immune response, the biological context within ex vivo. Table 1 summarizes the advantages and disadvantages
which infused T cells are operative. for each of the effector cell types that have been evaluated
to date in ACT-based approaches against cancer.
A brief history of ACT Classical NK cells (CD3 CD56+) represent about 20–
The potential to apply ACT as a therapeutic approach was 30% of circulating lymphocytes. As part of the innate
first evaluated in rodent models over 50 years ago (32). immune system, NK cells characterized by rapid and potent
The first clinical experience in humans was reported from cytolytic activation against virus-infected or transformed
the Seattle group in 1991 in the context of cytomegalovi- cells, a potential advantage for ACT. A potential advantage
rus (CMV) prophylaxis during immune reconstitution post of NK cells is that their principle targets are cells that lack
transplantation, where CMV-specific T-cell clones were class I, often a phenotype selected for in tumor cells; on
expanded ex vivo and infused into patients at high risk the other hand the presence of class I on tumors poses
from CMV infection, resulting in the establishment of an interesting challenges for broad implementation of NK cell-
anti-CMV protective immune response (33). This seminal based therapies. In the context of ACT, classical NK cells,
report demonstrated that the fundamental requirements for both as primary cells and cell lines, have been evaluated in
successful ACT, namely ex vivo manipulation, expansion, a number of different settings and using a variety of ex vivo
and reinfusion into patients were technically feasible, and expansion protocols. Classical NK cells have been tested in
precipitated concerted efforts to apply ACT to target malig- ACT non-modified in the context of haplo-identical HSCT
nancy. Early efforts to target malignancy focused on the (37), or more recently following genetic modification to
use of tumor-infiltrating lymphocytes (TILs) pioneered by express immune-modulatory cytokines (38). Unfortunately,
the Rosenberg group at NCI (34), the use of bulk popula- despite numerous attempts, these approaches led to at best
tions of expanded lymphocytes (23), as well as the use of modest clinical activity (39, 40). A new and intriguing
tumor antigen-specific clones, isolated following consider- approach involves gene engineering of NK cells to express
able tour-de-force efforts (35, 36). These approaches revealed chimeric antigen receptors (CARs), and these exciting stud-
a number of significant points with regard to the clinical ies are now starting to be evaluated in early phase clinical
implementation of ACT: (i) T-cell- based immunothera- trials (41).
peutic approaches had the potential to effect potent anti- Smaller lymphocytes subsets that represent intermediate
tumor activity; (ii) tumor targeting strategies would need phenotypes between classical NK cells and T cells have
to incorporate and address biological considerations for also been evaluated in the context of ACT. NK T cells
both the T cell (e.g. the need for cosimulation) and the (NKT) (CD3+CD56+), iNKT cells (CD3+CD56+Va24-Ja18),
tumor (e.g. immunosuppression); and (iii) robust and and cytokine-induced killer cells (CIK) (CD3+CD56+) dis-
practical implementation of ACT to target tumors would tinguished by Lu and Negrin (42), are each ‘niche’ effec-
require significant improvements in the ability to rapidly tor lymphocyte subpopulations with potential advantages,
generate large and potent populations of tumor-specific summarized in Table 1, in the context of ACT (43). NKT
lymphocytes. cells have been shown to modulate immune responses

© 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
16 Immunological Reviews 257/2014
Ruella & Kalos  Adoptive immunotherapy for cancer

NK Treg A Anti-VEGF-R
T Anti-αvβ3 integrin B
B
CART CART homing and
trafficking
C haTCRT
A
NKT
R
T
C D
CART
Tumor cell F

Treg MDSC B
Tumor cell
CART
Lymphodepletion MDSC H Checkpoint inhibitors
Chemothererapy haTCRT Autocrine cytokine
Conditioning Anti-FAP
augmentation
Radiation
TIL
E
G Tumor cell
B haTCRT Treg CART
Treg
TAM B
Treg
Tumor cell
Treg
TIL Tumor cell depletion Tumor cell
MDSC
I B NKT
Treg
B
CART MDSC

NKT
Anti-NKG2D MDSC TAM NK

Molecules Effector cells Regulatory cells Other cells

: Myeloid-Derived Tumor : Tumor Cell


: Surface target epitope CART : CAR-engineered T cell MDSC cell
Suppressor Cell
: MHC:peptide complex
: Endothelial cell
: Endogenously processed haTCRT : Affinity-enhanced TCR- TAM : Tumor Associated
epitope engineered T cell
Macrophage
NK : Natural Killer cell
: CAR
: Tumor infiltrating
: Tumor Associated
: Endogenous TCR complex lymphocyte NKT : Natural Killer T cell
Fibroblast
: Affinity-enhanced TCR Treg : Regulatory T cell
complex B : B cell

Fig. 1. Adoptive cell transfer (ACT) approaches to effect anti-tumor immunity. (A) Engineering T cells to target tumor endothelium facilitates
the entrance of immune cells on tumor site and inhibit angiogenesis. (B) Expression of chemokines (CXCR2, CCR2B) on the surface of T cells
favors homing to tumor site. (C) chemotherapy or radiotherapy before ACT enhance T cells cytotoxicity and persistence by both target tumor and
tumor associated immunosuppressive cells [myeloid-derived suppressor cells (MDSC), Treg, tumor-associated macrophages (TAM)]. (D) Chimeric
antigen receptor engineered T cells (CART) can target TAA and kill tumor cells without the need of HLA presentation. (E) TAA that are processed
and presented in HLA on the surface of tumor cells are recognized by tumor-infiltrating lymphocytes (TIL) and haTCRT. (F) Targeting tumor
associated FAP+ fibroblasts that may suppress T-cell homing and anti-tumor activity in the tumor environment. (G) Ex vivo depletion of Tregs
reduces the number of these inhibitory cells in the infusion product. (H) Checkpoint inhibitors (e.g. anti-CTLA-4 and anti-PD-1) and the
increased expression of activating cytokines (e.g. IL-12) enhance T-cell cytotoxic function. (I) Targeting NKG2D on immune cells re-modulates
tumor environment, reducing the immunosuppressive milieu.

© 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
Immunological Reviews 257/2014 17
Ruella & Kalos  Adoptive immunotherapy for cancer

Table 1. General properties of among tumor-infiltrating lymphocytes (TIL), TCR engineered T cells (TCRT), high affinity TCR engineered
T cells (haTCRT), and chimeric antigen receptor engineered T cells (CART)
Parameter TIL TCRT haTCRT CART
Tissue of origin Tumor Peripheral blood Peripheral blood Peripheral blood
Ease of generation Low Moderate* Moderate* Moderate
Efficiency of generation Moderate High High High
Ex vivo manipulation time 17–30 days 8–12 days 8–12 days 8–12 days
Genetic modification Not necessary Required Required Required
Antigens targeted All All All Surface only
Antigenic specificity Undefined Defined Defined Defined
Need for antigen processing and HLA presentation Yes Yes Yes No
Effector functionality Undefined Low High Very high
Proven clinical efficacy Melanoma Not clear Solid tumors B-cell neoplasms
Potential for Off tissue toxicity Low Low High High
Potential for Off target toxicity Low Low High Low
Feasibility for commercialization Low Low Moderate High
*
Once original TCR has been identified.

against cancer and stimulate effector cell functionality Genetically unmodified T cells
(44), and they have additionally been reported to be capa- One of the first clinical applications of ACT was the infu-
ble of overcoming peripheral tolerance (45). A potentially sion of cells termed lymphokine-induced killer cells (LIK),
important advantage of iNKT cells is the fact that they rec- which were generated from patient peripheral blood sam-
ognize a unique glycol-molecule (agal-Cer) in the context ples by exposure to high doses of interleukin-2 (IL-2).
of the CD1d molecule, which can be used to both specifi- Although positive clinical responses were reported in these
cally expand these cells ex vivo and also to stimulate their studies, these could not be dissociated from the effects of
expansion in patients (46). Additional subsets of NKT cells the co-infusion of high doses of IL-2 (51, 52). Neverthe-
have been defined, but to date have not been evaluated in less, the data generated from LIK-based strategies facilitated
ACT (47). the development of next generation approaches, specifically
cd T cells (CD3+ TCR a b , TCR c+d+), are a popula- the evaluation of lymphocytes isolated from tumor biopsies
tion of T cells very rare in peripheral blood (4–10%) but (TILs) and bulk T lymphocytes activated ex vivo prior to
substantially enriched in areas of mucosal immunity such infusion.
as gut (48). cd T cells recognize a wide variety of targets The premise behind TIL-based approaches is that T lym-
including stress-induced antigens in a classical MHC-unre- phocytes in biopsy specimens are enriched for anti-tumor
stricted manner and manifest lytic activity and pro-inflam- reactivity but have become functionally anergized, and that
matory cytokine secretion. A major population of cd T ex vivo culture of these cells results in their re-activation. TIL-
cells expresses an invariant Vc9Vd2 T-cell receptor, and has based strategies were developed principally in the context of
been shown respond to isopentenyl pyrophosphate (IPP), a melanoma, thanks to the pioneering efforts of the NCI
product of the mevalonate pathway which is dysregulated group. TILs cultured are selected for anti-tumor activity
in tumor cells. These lymphocyte subsets are currently following ex vivo culture and recognize tumors in an MHC-
tested for ACT, both unmodified following bulk expansion restricted manner via their TCR. Initial studies showed the
ex vivo (49) and also following gene engineering using feasibility of this approach with expansion of unselected
CARs (50). TILs from subcutaneous and lymph node melanomas with
abT cells (CD3+TCR a+b+) are the most abundant T-cell subsequent re-infusion together with IL-2; although the
population in the blood, the most extensively studied in response rate for these initial studies was about 30%, TIL
general, and the most robustly evaluated for ACT. Both persistence was poor (53, 54). To improve long-term per-
effector (CD8+) and helper (CD4+) T lymphocytes have sistence of infused cells, follow-up clinical studies evaluated
been evaluated in ACT in every conceivable combination, the use of non-myeloablative lymphodepleting chemother-
obtained from the peripheral blood or from tumor sites, apy (cyclophosphamide and fludarabine) prior to TIL infu-
either as bulk populations or antigen-specific clones, or fol- sion followed by high doses of IL-2, under the premise that
lowing genetic modification to express tumor re-targeting competition for niches and homeostatic cytokines were
receptors. impediments to long-term persistence of infused cells.
© 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
18 Immunological Reviews 257/2014
Ruella & Kalos  Adoptive immunotherapy for cancer

Incorporation of the lymphodepleting regimen improved Genetically retargeted T cells


clinical responses to 50%, with enhanced TIL in vivo expan- The fundamental premise behind genetic retargeting of T
sion and persistence (55, 56). In a recent update, 13% of cells is the fact that the endogenous potent tumor-specific
the patients treated with this regimen experienced complete T-cell repertoire has been compromised as a consequence of
regressions ongoing beyond 5 years (57). To evaluate the central tolerance. Indeed, comparative analyses have demon-
effect of enhanced lymphodepletion, two pilot trials were strated that TCRs against tumor have substantially lower
conducted with total body irradiation (TBI). These treat- antigen affinity (approximately 0.5 logs) compared with
ments resulted in clinical response rate of 49–72% (58). TCRs directed against virus-derived antigens (60, 61),
A major impediment with early TIL trials was that the ex providing at least partial explanation for the lack of clinical
vivo generation of TIL cultures was extremely labor intensive, efficacy of approaches directed to triggering the self-anti-
with the generation from each patient of multiple TIL sub- gen-reactive T-cell repertoire. Two related approaches have
lines, screening for tumor specific activity against autolo- been developed to genetically redirect T cells: (i) the trans-
gous or HLA-matched tumor cell lines, and long expansion fer of affinity-enhanced tumor-specific TCRs and (ii) the
cycles. As a result, more than a half of patients were transfer of synthetic CARs. Recent clinical experience has
excluded from this treatment. The development of shorter suggested that both of these approaches can mediate potent
expansion protocols and more streamlined screening anti-tumor activity.
approaches has considerably improved the broader imple- The ‘canonical’ TCR is composed of a complex of at least
mentation of this approach (59). An ongoing phase 2 study six polypeptide chains (a, b, c, d, e, and f) which assemble
is comparing CD8+ enriched short-term cultured TILs plus on the surface of T cells. The a and b chains form the pri-
high dose bolus IL-2 after non-myeloablative chemotherapy mary binding domain of the TCR, which recognizes intra-
against high dose IL-2 alone in metastatic melanoma. cellularly processed peptides presented on the surface of
The use of TIL therapy in a broader sense is currently target cells by proteins of the MHC. Accessory costimulatory
limited by a number of factors: (i) tumor specimens are not and adhesion molecules augment the strength and quality of
readily available in many cancers, especially in the metastatic the TCR-MHC peptide interaction leading to productive
setting where biopsies are not often obtainable; (ii) TILs are T-cell engagement. Initial efforts to evaluate TCR transfer
reproducibly detectable only in a minority of cancers involved transfer of self-antigen specific non-enhanced TCR
(mostly melanoma and renal cancer); (iii) expansion proto- the genetic transfer of TCR ab chains specific for self-
cols remain relatively labor-intensive, expensive, time antigens (62, 63). These pioneering efforts demonstrated
consuming, and difficult to standardize; (iv) current condi- the proof of principle that TCR-based T-cell engineering was
tioning regimens are not feasible in all patients; and (v) TIL feasible, but also highlighted the need to generate more
functionality may revert to the immune-suppressed state potent TCR to overcome the consequences of central toler-
when infused TIL re-enter TMA. ance on TCR affinity. Approaches to generate more potent
Parallel effort to TIL-based approaches involved the ex vivo TCRs have included the vaccination of human HLA trans-
expansion of bulk T lymphocytes from the peripheral blood genic mice with target tumor antigens to generate high
of cancer patients followed by subsequent re-infusion with affinity human antigen-specific TCRs, and this approach has
the support of IL-2. This approach was aimed to expand a been used to isolate high affinity TCRs against MDM-2 (64),
population of activated cells with a lowered triggering p53 (65), CEA (66), gp100 (67), and MAGE-A3 (68). More
threshold and thus with enhanced potential to be triggered recently animals transgenic for human HLA and TCR have
by tumor cells, as well as potentially trigger and expand been used to generate high-affinity human ab TCRs (69).
tumor-reactive lymphocytes rendered anergic as a result of Other approaches to generate high affinity TCRs have
peripheral tolerance. Clinical activity was observed in this involved taking advantage of gender-restricted expression of
approach, with rapid functional immune reconstitution target antigens (such as prostate, ovary) and the lack of tol-
observed in patients, and the establishment of optimized erance to these antigens in the opposite gender (70),
timelines for T-cell infusion post lymphodepletion (23). In selection of specific T-cell clones from a polyclonal pool of
a follow-up study, transfer of bulk activated T cells was graft-versus-tumor reactive T cells (71). Early clinical results
associated to the development of autologous GVHD with using affinity enhanced TCRs have highlighted the promise
rash and colitis in about 25% of treated patients, demon- of this approach with dramatic anti-tumor activity observed
strating the potency of this approach (24).
© 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
Immunological Reviews 257/2014 19
Ruella & Kalos  Adoptive immunotherapy for cancer

in a number of cases (72–74). However, as discussed fur- scFv of an antibody fused to the CD3f or less commonly, Fc
ther below, the introduction of TCRs with non-physiologi- receptor c (FcRc) signaling domains. The scFv are usually
cally enhanced affinity to self-antigens perhaps predictably derived from murine antibodies and thus have the potential
has led to increased issues related to off target toxicity in to trigger immune rejection responses by the host; current
normal tissues (66, 67, 75), an issue of considerable con- strategies involved the use of humanized or fully human
cern that has to be addressed for full implementation of this scFv to mitigate this possibility (92).
approach. Engineering the optimal signaling domain combination
Beyond TCR affinity enhancement approaches, other for maximal biological activity has been the focus of exten-
efforts to generate higher affinity tumor-specific TCRs have sive preclinical and more recently clinical activity. The most
involved efforts to increase the number of TCRs on the sur- intensively studied signaling domains have been those asso-
face of T cells (72, 73, 76), increasing introduced TCR ciated with robust effector or costimulatory activity. Initial
expression levels (77–80), and a number of strategies to (i.e. first generation) CARs contained only the CD3f chain
minimize a and b chains mispairing with endogenous signaling domain. Although such CARs were capable of
chains (81–89). Although TCR-based ACT approaches hold re-directing T cells to target antigens in vitro demonstrating
considerable promise as exemplified by early clinical trials, the proof of concept that T cells could be engineered using
they do possess certain inherent limitations. More specifi- CAR-based approaches, clinical trials based on such CARs
cally, the HLA-restricted specificity of engineered TCR limits demonstrated limited efficacy in a variety of cancers, such as
the fraction of potential patients to those expressing the rel- neuroblastoma (93), non-Hodgkin’s lymphoma (94), renal
evant HLA allele recognized by the T cells, and the fact that cancer (95), and ovarian cancer (96). A recurring theme in
tumors often downmodulate HLA alleles and/or alter prote- these studies was limited in vivo expansion, modest anti-
asomal processing pathways provides for the real possibility tumor activity, and lack of in vivo persistence.
of antigen escape variants and lack of complete responses. To overcome the limitations with first generation CARs,
CARs are synthetic receptors that combine the extracellu- second generation CARs were developed and evaluated. Sec-
lar single-chain variable fragment (scFv) of an antibody with ond generation CARs incorporate additional signaling
a transmembrane (TM) domain and intracellular signaling domains from costimulatory and accessory functional T-cell
domains derived from molecules involved in T-cell signal- molecules, in attempts to enhance in vivo functionality,
ing. Essentially all current CARs utilize the signaling domain potency, expansion capacity, and persistence. The first
from the CD3f chain and additionally can contain signaling costimulatory molecule to be included in the CAR construct
domains from molecules involved in T-cell activation or was CD28, and this led to a dramatic increase of the IL-2
costimulation. Since the scFv domain binds directly to target production and killing capacity of the T cells (97). Position-
cell surface epitopes CAR-based strategies bypass the need ing the CD28 domain 5′ to the CD3f domain was shown to
for MHC-restricted antigen presentation and are thus insen- me most effective (97, 98). Moreover, inclusion of the
sitive to tumor escape mechanism related to HLA down- CD28 domain was shown to enhance resistance to suppres-
modulation and altered processing escape-mechanisms (90). sion by regulatory T cells (99, 100). Many other signaling
CARs bypass most of the T-cell triggering limitations related domains have subsequently been evaluated preclinically,
to epitope density, since the scFv of the monoclonal anti- including 4-1BB (CD137) (101, 102), OX-40 (CD134)
body is characterized by high-affinity for the target and (103, 104), CD244 (105), CD27 (106), and inducible
additionally each surface target molecule presents a trigger- costimulator (ICOS) (98, 107). In preclinical models, sec-
ing epitope. CAR-based redirection represents a near univer- ond generation CAR T cells have been shown to mediate
sal ‘off-the-shelf method to generate large numbers of enhanced functionalities including expansion and anti-tumor
antigen-specific helper and cytotoxic T cells. As described in activity (102). CARs containing the 4-1BB domain have
more detail below, a key to this successful implementation been shown to result in improved in vivo persistence, anti-
of CAR-based approaches is the identification of antigens tumor activity and tumor localization compared with first
with surface expression on tumor cells and absent expres- generation CAR or CAR containing the CD28 domain (100,
sion on normal healthy tissues. 101). Inclusion of a CD27 signaling domain to CAR may
The concept of CARs was introduced by Eshhar and col- also enhance in vivo persistence and improve cytotoxic effect
leagues in 1989 (91). Since then, many groups have evalu- against xenogeneic tumor (106), while inclusion of the
ated the possibility to redirect T cells using antibody-based ICOS signaling domain has been shown to drive human
© 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
20 Immunological Reviews 257/2014
Ruella & Kalos  Adoptive immunotherapy for cancer

T cells to a Th17 phenotype (108). Second generation CARs, CAR molecule that is mainly related to spacer regions, and
where costimulatory molecules are included in trans, have this is able to influence the entity of the activation and kill-
also been developed, enabling the ability to more precisely ing activity.
potentially modulate CAR functionality (109). Most current CAR-engineered T cells have been generated against many
clinical trials evaluate second generation CAR designs (110). different TAA for both solid and hematologic malignancies:
Third generation CARs are characterized by the addition CD19 (128–131), CD20 (94, 132), CD33 (133, 134),
of a second costimulatory domain to further improve T-cell B-cell maturation antigen (135), CD22 (127), CD23 (136),
expansion, cytotoxicity, and in vivo persistence. Third gener- CD30 (137, 138), CD38 (139), CD44v6 (140), ROR-1
ation CARs combining CD28 with 4-1BB, and CD28 with (141), j-light chain (142), Lewis Y antigen (143, 144),
OX-40 have been evaluated (111–113). Conceptually, with NKp30 (145), TAG-72 (146), CD70 (147), Carboxy-anhy-
this approach the threshold of T-cell activation is further drase-IX (CA-IX) (95, 148), human epidermal growth factor
reduced in 3rd generation CARs leading to further enhanced receptor 2 (ErbB-2/Her-2/Neu) (149–153), disialoganglio-
potency but also increased potential for low avidity off target side 2 (GD2) (154–157), GD3 (158), L1CAM (CD171)
activation. (93), VEGF-R2 (159), EGFR (107, 160, 161), MUC-1
In terms of CAR design, both the spacer/hinge and the (124), MUC-16 (162), prostate-specific membrane antigen
TM domains have the potential to influence CAR function. (PSMA) (97, 163, 164), prostate stem cell antigen (PSCA)
In fact the hinge region mediates flexibility and it is impor- (165, 166), 5T4 oncofetal antigen (h5T4) (167), NCAM
tant for the positioning of the binding scFv domain (114, (125), mesothelin (111, 168), fibroblast activating protein
115). A number of studies have shown that the TM domain (FAP), folate receptor-a (96, 169), NKG2D (170–172), IL-
is critical for effective CAR expression on the surface of the 11 receptor a-chain (173), CEA (115, 174, 175), IL-13Ra2
cell, and accordingly many different TM domains have been (117, 176), and erythropoietin-producing hepatocellular
evaluated including CD28 (116), OX40 (116), CD3f (117), carcinoma A2 (EphA2) (177). As described in more detail
FceRIc (118), CD7 (119), CD4 (102, 103), H2-Kb (120), below, a small number of CARs are currently being tested
and CD8 (101, 121). Inclusion of the CD28 TM domain in early phase clinical trials, with in some cases dramatic
was found to result in a higher expression of CAR in com- and exciting results that highlight the promise of this
parison with OX40 and CD3z TM domains (116). Other approach against cancer.
reports suggested that intracellular rather than TM domains A number of groups have initiated efforts to overcome
modulate CAR surface expression (122). With regard to the the need for patient-specific products for ACT. Attempts to
hinge region, IgG1, IgG4, IgD, and CD8 domains have been generate ‘universal donor T cells’ have involved the use of
mostly evaluated (123, 124). An additional important con- zinc-fingers nucleases to eliminate the expression of the
sideration for CAR design is the length of the hinge region, endogenous TCRs (178). Although this area of research is
which has been shown to influence the quality of the inter- still nascent and many issues will need to be identified and
action between the T cell and target, depending on the loca- resolved, it has the potential, combined with efforts to
tion of the target epitope on the target antigen (125, 126). reduce the allo-recognition of engineered T cells for exam-
Until more specific rules to guide this process are defined, ple by targeted knockdown of class I and class II loci to
this important issue will likely require empiric testing of bypass the need to generate patient-specific T cells by gener-
multiple CARs against each epitope being targeted. The rela- ating universal allogeneic TAA-specific T cells from one
tive position of the epitope in the target protein also appears donor that might be administered to multiple recipients.
to be relevant in terms of CAR engineered T cells (CART) A novel approach to simultaneously target multiple tar-
potency. For example, CD22 target epitopes that are proxi- gets on tumors is the development of bi-specific CAR. Such
mal to the cell membrane trigger more potent lytic activity moieties, coined as ‘TanCAR’ simultaneously recognize two
compared with distal epitopes (126, 127), while other stud- tumor restricted antigens with synergistic enhancement of
ies showed longer spacer domains increased the potency of effector functionality when both antigens are present on
5T4- and NCAM-specific CAR that recognized membrane target cells; T cells engineered with TanCAR retained cyto-
proximal epitopes, potentially by providing increased flexi- lytic ability with loss of one of the target molecules and
bility to the CAR (125). Indeed, the distance between the T were superior to single CAR engineered T cells in the con-
cell and the tumor cell is influenced by the combination trol of established experimental tumors in animal models
between the position of the epitope and the length of the (179). TanCAR or related approaches have the potential to
© 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
Immunological Reviews 257/2014 21
Ruella & Kalos  Adoptive immunotherapy for cancer

counteract the existence of antigen-loss variants that often engineered T cells has been shown to enhance the efficacy
exist in tumors; however, such approaches may have of low doses of MHC-restricted TILs, at least in part by
enhanced potential for off-tissue toxicity, discussed more re-programming tumor associated myeloid cells to cross-
fully below. present tumor-associated antigens (187); interestingly, this
An interesting strategy to mitigate off-tissue toxicity is to approach eliminated the need for lymphodepletion before
develop combinatorial CAR constructs that transduce an CAR therapy (188). Other approaches that involve efforts to
activating signal only when encountering a specific combi- modulation expression of cytokines, such as IL-12 by T
nation of antigens. Kloss et al. (180) recently described a cells, are also currently being evaluated (189). Clearly, the
novel approach by which T cells are co-transduced with a ability to at-will engineer T cells to express desired
CAR that provides suboptimal activation upon binding of immune-modulating soluble factors and receptors provides
one antigen and a chimeric co-stimulatory receptor (CCR) an incredibly exciting and diverse menu of opportunities to
that recognizes a second antigen. Using the prostate tumor develop more potent, and disease targeted options for ACT,
antigens PSMA and PSCA as model antigens, this group while also enabling the modulation of SAE associated with
demonstrated that combinatorial CAR-engineered T cells ACT-collateral immune activation.
eradicated tumor cells that expressed both but not individ-
ual antigens (180). T-cell subsets for ACT
A novel strategy to expand the recognition specificity Over the past few years it has become evident that T-cell
potential of CAR T cells utilizes a biotin-binding immune function may be influenced by many other parameters
receptor (BBIR), which consists of an extracellular modified beyond antigen specificity. The nature of the T cell that is
avidin domain linked to an intracellular T-cell signaling used for adoptive immunotherapy is highly correlated with
domain; target-specific antibodies can then be loaded onto clinical results (190). Initially, it was assumed that the ideal
BBIR T cells through streptavidin cross-linking (181). Devel- T cells for adoptive immunotherapy would be effector T
opment of BBIR T cells may allow the sequential or simulta- cells (Teff), due to their ability to respond robustly to anti-
neous targeting of a broad combination of distinct antigens. gen stimulation as measured by cytotoxicity. However, a
A recent report has described the development of CARs number of early clinical studies showed that highly differ-
that target the intracellular antigen WT-1 using a scFv that entiated tumor specific Teff CD8+ T cells engrafted poorly
is able to recognize MHC class I + peptide complexes and are less capable of tumor killing compared with heter-
(182). Although still extremely preliminary, this approach ogeneous population of TILs (191, 192). Retrospective
has the potential to overcome the limitation of CAR-based analyses from clinical trials revealed that patients infused
strategies to surface antigens and greatly extend the universe with less differentiated cell products had better clinical
of target antigens available for CAR-based approaches. On outcome (193), and follow-up studies in animal models
the other hand, advantages of CAR-based strategies described demonstrated that the use of a less differentiated T-cell
above and related to target epitope density and indepen- population for ACT led to better anti-tumor activity, in vivo
dence from HLA expression and processing defects will expansion and persistence (194). These results led to
likely prove to be impediments for the success of this efforts to identify sub-populations of T cells with enhanced
approach. in vivo functionality as well as develop in vitro culture
Beyond the introduction of antigen-re-targeting receptors, approaches that generated less differentiated T-cell products
gene-engineering efforts have also involved attempts to (see below). More recent studies have shown that the fre-
selectively target T cells to sites of tumor using chemokine quency of central memory T cells (Tcm) in infusion prod-
and homing receptors such as CXCR2 or CCR2B, which ucts correlates with positive clinical response (156).
when expressed on engineered T cells have been shown Furthermore, naive T cells (Tn) have shown higher anti-
facilitate the homing of T cells to tumor (137, 183–185). tumor effect and longer in vivo persistence than Tcm (195)
Engineering of T cells to express immune modulatory cyto- and antigen experienced CD8+ Tcm cells persist longer than
kines, such as for example IL-12, has been shown to effector memory T (Tem) cells (196). More recently a spe-
improve the efficacy of ACT in animal models, at least in cific T-cell subset, the stem cell memory T cells (Tscm),
part by increasing the expression of Fas within tumor-infil- has been described; Tscm have been described to be capa-
trating macrophages, DCs, and myeloid-derived suppressor ble of self-renewal and to generate potent effectors, this
cells (MDSCs) (186). Expression of single chain IL-12 by
© 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
22 Immunological Reviews 257/2014
Ruella & Kalos  Adoptive immunotherapy for cancer

population may be important for adoptive immunotherapy through their endogenous TCR (212). Clearly this approach
(197). Recent exciting work has also revealed the potential is restricted to patients that have been exposed to the
for plasticity in the Tscm repertoire, with data from animal viruses. EBV-specific T cells had been transduced with anti-
models suggesting that a very small number of the ‘right’ CD30 CARs or anti-GD2 CARs and had been preclinically
population of engineered T cells may be sufficient to medi- tested (138, 155). EBV-specific T cells engineered to express
ate potent anti-tumor activity accompanied by in vivo expan- a chimeric receptor had also been used to treat nasopharyn-
sion and differentiation into T-cell subsets (198). geal carcinoma or AML (133, 213).
Tn, Tcm, and Tscm cells express CD62L and CCR7 and
therefore have the potential to home to secondary lymphoid T-cell engineering strategies
tissues where they can interact with APCs explaining at least The development of approaches to at-will engineer T cells
in part their better clinical activity. On the other hand, prior to ACT has been a fundamental building block for the
CCR7 cells engineered to express CARs with CD28, OX40, recent success in the field. Although most preclinical and
and TCRf costimulatory domains were recently shown to clinical studies have focused on introducing TCR or CAR
be rescued from apoptosis and result in more efficient anti- re-targeting receptors into T cells (110), more recent stud-
tumor efficacy compared with CCR7+ CAR-engineered T ies have begun to evaluate the possibility to introduce T
cells (199). and other immune cell function-modulating molecules in T
A fundamental property of T cells, which mediate potent cells, such as IL-12 (188), IL-15 (214, 215), CCR2 (216),
anti-tumor activity appears to be the ability for in vivo expan- IL-2 (217). Initial successful efforts for engineering T cells
sion, differentiation, and persistence (28). A number of utilized pseudotyped c-retroviruses that were capable of
studies have shown that Teff cells possess a relatively limited infecting primary human T cells and integrating their DNA
proliferative and engraftment potential compared to Tn, in the host genome; this approach lead to high efficiency
Tcm, and Tscm, in part due to limited lifespan associated stable transduction of primary T cells that were employed
with shorter telomere length (200–203). Studies using TILs in multiple clinical trials (218, 219). Concerns about retro-
have demonstrated that a short duration in culture, a rapid virus integration site bias coupled with concerns about
doubling time and longer T-cell telomere length were corre- long-term retroviral promoter silencing led to the more
lated with better clinical outcome (53, 57, 204–206). recent development of lentivirus-based vectors for gene
Recent studies indicated that the effector-like CD27 CD8+ transfer. In addition to the fact that lentiviruses vectors
cells mediate potent protective immunity and lead to long- appear to be less susceptible to silencing by host restriction
lived memory cells (207). Finally, recent reports suggest factors and can deliver larger DNA sequences than retrovi-
that adoptively transferred Th17 cells demonstrated superior ruses (220), lentivirus vectors have been reported to be
proliferation, persistence, and anti-tumor activity compared able to infect non-cycling cells, particularly when coupled
with Th1 cells (208, 209). with cytokine ‘pre-activation’ (221, 222). From an onco-
Although data obtained in a number of studies suggest genic perspective, significant early and justified concern
that less differentiated T cells with longer telomeres, such as about the oncogenic potential of retrovirus-based gene ther-
Tn, Tcm, and Tscm, may be the preferred cells for ACT, apy as a result of the unfortunate report showing oncogenic
there is potential danger of applying reductionist reasoning transformation of stem cells due to retrovirus integration
to an inherently complex biological by selecting a particular (223, 224) was mitigated initial in animal models (223,
T-cell subtype for ACT. There is evidence that CD4+ cells 225) and subsequently in long-term longitudinal retrovirus-
are required for CD8+ memory cell formation that is import based gene-therapy studies now extending beyond 10 years,
for long-term control of the tumor (210, 211). Indeed, the which confirmed that retrovirus-based gene transfer into
most potent and durable clinical responses to date have been human T cells appears to be safe (226, 227). Although len-
obtained using bulk populations of CD8+ and CD4+ T cells tivirus-based gene transfer has not been implemented as
(27–29). long, the fact that in the natural history of HIV no T-cell
Considerable effort has been spent principally by the Bay- oncogenic events have been reported, coupled with the
lor group to evaluate the ability to isolate and engineer virus absence of any reports from to-date lentivirus-based clinical
(EBV, CMV)-specific T cells to express CARs, under the pre- trials provide reasonable assurances that this approach will
mise that these cells are inherently enriched for memory be safe from an oncogenic perspective. Novel integrating
cells and additionally can be maintained in vivo by stimulation
© 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
Immunological Reviews 257/2014 23
Ruella & Kalos  Adoptive immunotherapy for cancer

virus-based delivery systems such as foamy viruses are employed for ACT appears to be critical for in vivo efficacy.
under development (228) but beyond the scope of this An early approach, based on the pioneering work of Riddell
review. Adenovirus-based vectors such as Ad-35 vectors, and Greenberg (240), employed antibodies specific to CD3e
which result in long-term episomal transgene expression, and cross-linked using allogeneic peripheral blood mononu-
have also been evaluated in human T cells with high clear cells, with immortalized B-LCL cells added as feeder
expression efficiencies (229). cells, and exogenous IL-2. Repetitive stimulation using this
The relatively high cost for manufacture and release of approach generated large numbers of antigen-specific T cells
virus-based vectors has precipitated the development of a that were employed in early trials (192, 241–243), unfor-
variety of non-virus based gene transfer systems. Retro- tunately without significant success, in retrospect due to the
transposon systems, such as PiggyBac or Sleeping Beauty fact that this approach strongly skewed products toward
have shown preclinical promise (230–232). Novel and Teff cells (192, 244). A parallel approach, developed by
exciting approaches based on Zinc-finger nuclease, TALEN, Levine, June and colleagues, employed the use of magnetic
and CRISP/Cas9 based-technologies permit to targeted inser- beads coated with anti-CD3e and anti-CD28 antibodies to
tion of transgenes or the targeted editing of the T-cell tran- provide costimulation. This approach generates large num-
scriptome and open up the potential to be able to modulate bers of T cells with a less differentiated phenotype (245,
transgene expression and T-cell function at will (229, 231, 246), has been robustly developed (247), and is the
233, 234). approach that has been recently employed to manufacture
For some applications, permanent genomic integration of products with extensive in vivo proliferative potential, potent
new constructs may not be necessary for therapeutic efficacy anti-tumor activity, and long-term functional persistence
or may be useful to mitigate potential toxicity. A novel currently exceeding 3 years in the initial treated patients
approach that is currently being investigated to this end is (27–29, see below). Although early studies suggested that
RNA electroporation; this approach allows the rapid transfer the absolute number of the transferred T cells is almost
of the specific mRNA in T cells allowing a high expression always correlated with better clinical responses (205), this
of the specific receptor (122, 235). Since the introduced paradigm is now in question given the dramatic potency
mRNA is relatively rapidly degraded, this approach results in achieved with small numbers of cells in a recent trial (27),
‘biodegradable’ engineered T cells. Although RNA- and the discovery of T cells with stem cell properties (197,
engineered T cells have been shown to mediate potent and 248, 249). Magnetic bead-based approaches have been used
antigen specific effector functions, multiple infusions are for preselection of specific subsets of T cells, such as CD8+
required to compensate for the short in vivo half-life of these cells for TIL immunotherapy (59), CD4+ T cells for transfer
cells (236). RNA engineering has been used to deliver tran- HIV patients (250), CD25+ cells to prevent GVHD after
scripts for TCRs, CAR chemokine receptors, or cytokines allogeneic HSCT (251), or to select against particular sub-
(215, 237, 238) and is currently being evaluated in clinical sets of cells that may be deleterious for in vivo efficacy, such
trials. The biodegradable nature of this approach provides a as regulatory T cells (Tregs) (252). Based on preclinical
considerable safety advantage, in particular when evaluating data showing the enhanced persistence and anti-tumor
T cells engineered to express constructs with potential for functionality of Tcm cells (196), strategies to isolate CD8+
toxicity. RNA engineering also allows the efficient transfer Tcm cells under GMP conditions have been developed
of multiple transcripts allowing for the ability to engineer T (253). T cells may be also isolated based on for antigen
cells to co-express targeting, homing, and immune modulat- specificity, as discussed above in the context of redirecting
ing functionalities. An efficient low cost method for gene virus specific T cells to target tumors (212), and also in the
transfer to T lymphocytes that includes the use of electropo- context of utilizing virus-specific T cells to treat viral infec-
ration and a transposon-based system has recently been tions such as CMV (254, 255).
described (239). The use of artificial antigen-presenting cells (AAPCs), also
pioneered by the June group (256), to stimulate T cells
T-cell expansion strategies under more physiologic conditions provides an interesting
A fundamental requisite for current ACT strategies is the alternative approach to expand T cells for ACT. This
ability to engineer and expand T cells prior to infusion into approach, which has not yet been evaluated clinically, has
patients. As described above, the ‘quality’ of T cells the potential to provide more physiologic stimulation of

© 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
24 Immunological Reviews 257/2014
Ruella & Kalos  Adoptive immunotherapy for cancer

T cells preserving desired differentiation states, and has been T-cell ablation strategies
adopted by other groups (257–260). Novel strategies A concern with ACT has long been the need to ablate trans-
involving ex vivo expansion of T cells with cytokines are also ferred cells, either to counter off target toxicity. To this
currently being investigated. Expansion of CD8+ naive T end, ‘suicide gene strategies’ have been developed. Initial
cells with IL-15 provides the expansion of T cells with the suicide system involved use of the herpes simplex virus
characteristics of Tcm cells, with preserved telomere length (HSV) thymidine kinase (TK), which phosphorylates ganci-
(261); compared with T cells expanded with IL-2, the use clovir and acyclovir leading to the generation of toxic tri-
of IL-15 has been shown to support superior proliferative phosphate products and cell death (274). Perhaps
and anti-tumor activity (262). Expression of IL-15RA on predictably, HSV-TK transduction in T cells leads to immu-
CD8+ T cells has been shown to autonomously enhance the nogenic reaction with clearance of TK positive T cells (243,
viability and proliferation of primary CD8+ T cells and cyto- 275). More recently, an elegant system based on the design
toxic potential of antigen-specific CD8+ T cells (215). The and use of a caspase-9-driven apoptosis activity was devel-
combined use of IL-15 and IL-7 in the context of anti-CD3/ oped; in this approach caspase-9 intracellular signaling
anti-CD28 bead-based stimulation has been shown to domains were fused with a human FK506-binding protein
facilitate the ex vivo differentiation and expansion of gene domains; administration of FK506 (tacrolimus) leads to
modified CD8+ Tscm cells under GMP compliant conditions dimerization of caspase-9, activation of apoptotic pathways
(263). IL-21, when used for T cells expansion, leads to the and potent cell ablation (276, 277). Even more recently an
production of minimally differentiated T cells with high equally elegant system was developed that involved engi-
proliferative and anti-tumor capacity (264, 265), while neering of cells to express a truncated EGFR polypeptide
reprogramming of CD19-specific T cells with IL-21 signal- devoid of extracellular ligand-binding and intracellular sig-
ing has been shown to improve adoptive immunotherapy of naling domains but that retained binding to the anti-EGFR
B-lineage malignancies (266). IL-4 may also have a role in monoclonal antibody Erbitux, enabling both in vitro selection
T-cell expansion, as human T-cells engineered to co-express and in vivo ablation of engineered cells (278). Other
IL-4 receptor together with a CAR specific for MUC1 exhib- approaches to specifically eliminate engineered T cells
ited robust capacity to expand in vitro and effect destruction include the co-transduction of CD20 and the use of ritux-
of MUC1-expressing tumors (267). Modulation of the imab to ablate engineered cells by ADC (279).
metabolic state of T cells has been shown to influence their
phenotype and function; for example, modulation of PI3K- Target antigens for ACT
AKT-mTOR and Wnt-b-catenin pathways had been shown
The modern age of cancer immunotherapy arguably began
to control the differentiation status of T cells; co-culture of
in 1991 with the discovery by Thierry Boon and colleagues
T cells with mTor inhibitors promotes the formation of
of MART-1, the first human cancer antigen (280). Since
memory subsets, while the promotion of the Wnt pathway
then, a major area of focus in immunotherapy has been the
leads to the formation of Tscm cells with potent anti-tumor
identification and evaluation, both preclinically and clini-
activity (268).
cally, of candidate target antigens overexpressed or aber-
Modulation of substrate rigidity is another potentially
rantly expressed by tumors (281). Perhaps predictably,
promising approach to control the quality of ex vivo
initial efforts in ACT also focused on generating and utiliz-
expanded T cells. Early in vitro studies in this area have
ing T cells directed to such antigens. A plethora of clinical
revealed that modulating substrate rigidity has the potential
data have now led to the conclusion that targeting self-anti-
to impact T-cell activation and proliferation (269).
gens using ACT likely requires the engineering of enhanced
Finally, strategies that involve overexpression of genes
targeting specificity in T cells to overcome the fundamental
involved in survival such as telomerase (270), anti-apoptotic
impact of central tolerance on the endogenous T-cell reper-
genes (271), as well as the downregulation of pro apoptotic
toire (110). Indeed, an unbiased comparison of T cells spe-
molecules such as Fas (272), or dominant-negative receptors
cific for self-antigens, including cancer testis (CT) antigens,
for inhibitory molecules such as dominant negative recep-
overexpressed by tumors and T cells specific for viral anti-
tors for TGFb (273) have been evaluated preclinically.
gens has revealed that the latter have affinities for target
Despite the interesting scientific approach of these strategies,
antigens approximately one log higher compared with the
concerns regarding safety have been raised and should be
former (61). As discussed above, engineering efforts have
excluded before moving to the clinic.
© 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
Immunological Reviews 257/2014 25
Ruella & Kalos  Adoptive immunotherapy for cancer

focused on affinity enhancement of TCR (282) or the use of comprehensive and innovative preclinical testing followed by
high affinity CAR-based strategies. evaluation in clinical trials. A conceptually attractive approach
The definition of ideal target antigens is fundamentally involves the development of combinatorial activation strate-
different for TCR- and CAR- based strategies. For TCR-based gies, with T-cell activation requiring the simultaneous bind-
strategies, ideal targets will likely be tumor-specific neo- ing and triggering of multiple target unique CARs (286) and
antigens, preferably shared across a significant fraction of the use of CART ‘cocktails’ that also may help address issues
tumors to enable systematic tumor targeting, or gender- related to the escape of antigen loss variants (287).
restricted antigens that can be used to generate high affinity Animal models, extensively used to evaluate toxicity for
TCR in the non-expressing gender (70). Both of these target more traditional therapeutic agents, are considerably less
classes result in T cells with TCRs that have been shaped by useful in ACT because of lack of epitope conservation across
negative selection in the thymus and thus have minimal species and also because they fail to capture the complexity
potential for off target recognition of other tissues. In the of human T cells interacting with the broader immune sys-
case of CARs, because of the potency of this approach, ideal tem. The clearest demonstration of this is the recent report
antigens will either have a tumor-specific expression profile, of SAE using high-affinity TCR redirected T cells against
or will be uniquely expressed in non-essential tissues such MAGE-3; despite extensive preclinical testing of this affinity
as prostate, breast, and ovary, which can be removed prior enhanced receptor, ACT using cells engineered to express
to ACT to enable tumor-specific targeting. Indeed the this receptor resulted in two deaths due to cardivascular
potency of CAR-based approaches and the need for a rela- toxicity as a consequence of off-target recognition of an epi-
tively high threshold of tumor specificity has been high- tope from titin, a protein expressed in differentiated cardio-
lighted by reported which have demonstrated significant myocytes (75, 288).
serious adverse events (SAEs) when targeting antigens with Beyond targeting antigens expressed by tumors, ACT has
low-level expression in normal tissues such as CAIX and also been applied to target T cells to molecules expressed
ERBB2 (see below). If necessary, improvements can also be by tissues associated with the biology proliferation and sur-
performed on CAR–based designs to enhance affinity and vival of tumors. This effort has principally been carried out
improve potency (141). as part of efforts to target solid tumors, and had brought
Taking into account these considerations, precious few into juxtaposition the biology of T cells and tumors. Post
antigens have been identified that would qualify as ideal. In ACT T cells initially transit through the heart following the
terms of neo-antigen targets for TCR, brute force efforts natural venous drainage and very quickly are transported to
have in fact identified T cells which recognize tumor- the lungs (289). Localization to lung is a critical initial site
specific neo-antigens; however, these neo-antigens appear for T-cell trafficking due to potential low level expression
to in general result from individual tumor-specific genetic of target antigens by lung epithelium; indeed, off-target
events which result in non-self and thus non-tolerized recognition of lung epithelial tissue has been observed in a
proteins (283–285), with T cells and TCRs which are not trial with Her2-specific CART, due to low-level expression
amenable to platform development for ACT. In terms of of Her2 in lung epithelial cells (153). Rapid liver toxicity
CAR targets, the profound potency of CAR-engineered cells, has also been recorded in anti-CA IX CART trial due to the
related at least in part to the fact that each surface molecule expression of this marker in epithelium of the biliary tree
for the target protein is a triggering epitope, requires a (95). Different routes on ACT administration, such as intra-
lower threshold of expression that is considerably lower tumor or intraperitoneum, may reduce the potential toxic-
than that required for therapies; this is best exemplified by ity (242, 290), although it apparent that, as expected
the above mentioned SAE with CARs that utilized ScFv biologically, T cells subsequently redistribute to secondary
derived from clinically approved antibodies. Thus, ideal organs; indeed, analysis of tissues from post SAE biopsy of
antigens from a CAR-perspective will have no expression in the titin-related SAE described above revealed broad low-
any non-targeted tissue. Practically speaking, in the emerg- level distribution of infused T cells in the majority of
ing field of T lymphocyte engineering to manifest potent organs evaluated (75). Concern has been raised that T cells
and redirected tumor specificity, where the physiologic and may be unable to actively migrate and extravasate into
evolutionarily selected balance of recognition of self versus tumor parenchyma, limiting their effectiveness in vivo. Tar-
non-self is manipulated, the potential for off-target toxicity geting the tumor vasculature using engineered T cells is
is a risk which ultimately will need to be addressed using one possible mechanism to facilitate efflux of T cells into
© 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
26 Immunological Reviews 257/2014
Ruella & Kalos  Adoptive immunotherapy for cancer

the tumor. Systemic administration of T cells engineered (307, 308). The length of survival among patients with fol-
with CAR targeting avb3 integrin, expressed in tumor licular lymphoma correlates with the molecular features of
endothelial cells, led to extensive bleeding in tumor tissues non-malignant immune cells present in the tumor at diag-
with no evidence of damage to blood vessels in normal tis- nosis (309). The interplay of engineered T cells with these
sues; when this approach was coupled with co-delivery of cell subsets and others is likely to play an important role for
nanoparticles increased deposition of nanoparticles was the development and establishment of potent and long anti-
observed in the tumor (291). Combining ACT with agents tumor immunity, as well as in mediating the immune-based
that interfere with tumor vasculature such as avastin holds adverse events associated with strong anti-tumor responses
promise in this regard. observed in recent CAR-based trials (27–29).
At the tumor site, T cells face a dynamic and intricate Immune-suppressive mechanisms at play in the tumor
tumor micro-environment (TME) composed of a complex microenvironment involve the suppressive action of regula-
mixture of tumor cells, endothelial cells, stromal cells, fibro- tory T cells, MDSCs, TAMs and other myeloid-lineage
blasts, myeloid cells, NKT cells, T cells, and regulatory T derived suppressive cells, stromal fibroblasts and almost cer-
cells, and shaped by the selective pressure to support the tainly other cell types not yet defined. These cells inhibit T-
tumor mass both by providing trophic signals and also by cell function through the production of immunosuppressive
creating a potently anti-inflammatory and immunosuppres- cytokines and other soluble factors, as well as through the
sive milieu (292–294); these heterogeneous cell types may expression of surface molecules that bind to inhibitory
occupy up to 90% of the tumor mass (295), where they receptors such as CTLA-4, PD-1, TIM-3, BTLA, and LAG-3,
interact via soluble and cell-cell dependent mechanisms with and no doubt others, which are expressed on T cells often
each other, the tumor as well as the effector immune sys- post activation and serve to modulate the T-cell response
tem. Tumor cells can modulate their growth environment (15, 310). These suppressive cells are conditioned by the
through the activity of cytokines and chemokines leading to tumor microenvironment to become immunosuppressive
chemotactic effects on leukocytes, including monocytes and and are also stimulated to migrate in the tumor site and
macrophages, suppression of the activity of the immune proliferate (311, 312). MDSCs are directly correlated with
system, as well as regulation of neovascularization processes chronic inflammation and with the production of IL-1,IL-6,
(296, 297); accordingly, targeting cells in the micro- reactive oxygen species (ROS), and nitric oxide (NO), while
environment has the potential to fundamentally disrupt the they also promote hypoxia, lactic acid production, and
homeostatic milieu established at tumors thus increasing the adenosine accumulation which inhibit APC maturation
effectiveness of cancer-targeted therapies (298). (313, 314). The presence of TAMs is generally associated
Immune cells, both innate and adaptive, are intimately with worse prognosis, with type 2 TAMs, which produce
involved in carcinogenesis by promoting tumor transforma- immunosuppressive cytokines for Th2 responses, showing
tion as seen in chronic inflammatory diseases or tumor asso- poor antigen-presenting capacity and fail to activate T-cell-
ciated inflammation (299, 300). Tumor cells modify and mediated adaptive immunity (315, 316).
affect the behavior of tumor infiltrating cells to promote the Recent clinical experience using antibodies that block the
formation of a pro-neoplastic microenvironment (301). function of inhibitory receptors have demonstrated the
The relevance of the tumor-associated microenvironment potency of this mechanism (22, 317, 318); this exciting
for the efficacy of immunotherapeutic approaches has been line of research has led to the approval of the first in class
documented in a number of clinical settings. To enumerate ‘checkpoint inhibitor’ agent, YervoyTM, which blocks the
a few examples, the presence of TILs in tumor specimens is functional activity of CTLA4, and the likely approval in the
correlated with better clinical outcome in cancer patients near future of agents that block the post-activation inhibi-
(302, 303). Patients with cancer have elevated numbers of tory function of PD1 on T cells and PDL1/PDL2 on tumors.
Tregs in the peripheral blood and within the tumor micro- An emerging body of clinical evidence is accumulating that
environment (304), and tumor infiltration by Tregs is asso- correlate the presence of inhibitory receptors such as PD-1
ciated with poor outcome in many cancer subtypes (305, in TILs correlate with distant metastasis and poorer outcome
306). In colon cancer a T-helper 1 (Th1) immune signature in renal cancer (319) and breast cancers (320). Tregs within
is predictive of better clinical outcome compared with a the tumor microenvironment have been shown to markedly
Th17 signature (305). High frequencies of tumor-associated hinder the anti-tumor efficacy of adoptively transferred
macrophages (TAMs) are associated with poor prognosis tumor-targeted effector T cells (321), and Treg-depleting
© 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
Immunological Reviews 257/2014 27
Ruella & Kalos  Adoptive immunotherapy for cancer

strategies combined with systemic lymphodepletion reduce tumor angiogenesis has been studied preclinically in the con-
the number of Tregs in the tumor microenvironment and text of ACT, to evaluate the potential for synergistic effects
result in higher anti-tumor activity of T cells (322, 323). between the two treatment modalities. Adoptive transfer of
Tregs have been shown to also inhibit CART cells in animal CAR T lymphocytes against VEGFR-1 delayed tumor growth
models, with prior treatment with cyclophosphamide effec- and formation and inhibited pulmonary metastasis in
tively reversing the suppression (324). xenograft models, an effect that was further enhanced by
Manipulating the microenvironment before T-cell infusion co-transfer of T lymphocytes that expressed IL-15 (331). In a
through non-myeloablative chemotherapy and lymphodeple- separate study, simultaneous targeting of tumor antigens
tion has been shown to lead to dramatic increases in the per- gp100 (PMEL), TRP-1 (TYRP1), or TRP-2 (DCT) and the
sistence of the transferred cells and robust anti-tumor activity tumor vasculature (VEGFR-2) using ACT revealed synergistic
(55, 56). The rationale of prior lymphodepletion was based effects in terms of regression of established tumors in mice
on studies in tumor models in mice that showed improved (332).
persistence and anti-tumor activity of transferred T cells fol- Tumor stromal fibroblasts (TSFs) are one of the most
lowing conditioning chemotherapy (325); in these studies, prominent cell types in the tumor microenvironment of
improved engraftment and expansion was correlated with the many human cancers such as pancreatic, gastrointestinal,
ablation of resident lymphocytes that presumably competed and breast cancers. TSFs appear to play an active role in can-
for cytokines (IL-7 and IL-15) and T-cell growth factors. In cer progression by secreting factors that enhance tumor sur-
particular, IL-15, which can provide a homeostatic growth vival, growth, angiogenesis, and metastasis, in addition to
stimulus for adoptively transferred T cells and can lead to the recruiting other tumor-promoting cell types (333, 334).
induction of a memory phenotype with enhanced effector The conversion of TSFs to a tumor-promoting state is char-
functionality (56, 323, 326), is not normally detectable, but acterized by expression of surface proteins such as fibroblast
it was detected postlymphodepletion. Lymphodepletion also activation protein (FAP), an endopeptidase with expression
results in a reduction in the number of regulatory or inhibi- largely restricted to TSFs. Accordingly, FAP has been evalu-
tory lymphocyte populations present in the tumor (327). ated as a target for ACT-based approaches. Unfortunately, as
Conditioning chemotherapy may also be useful to eliminate recently reported, although highly reactive anti-FAP CARs
or reduce myeloid suppressor cells in the tumor microenvi- had little impact on tumor progression in a variety of synge-
ronment, while APCs may be activated by lymphodepletion neic mouse tumor implantation models, high doses of FAP-
with the increase of their susceptibility to Toll-like receptor reactive T cells were observed to induce severe cachexia and
signaling (328). The importance of the lymphodepletion has dose-limiting bone toxicity (335), underscoring the previ-
been confirmed in the clinical setting where most of the ously made point about the potency and need to critically
patients not receiving lymphodepletion prior to infusion of evaluate the potential for off-target toxicity of CAR-based
CART19 did not show any objective response of their malig- approaches.
nancy (100, 329). The timing of lymphodepletion and con- ACT-based strategies that utilize engineered T cells can
ditioning relative to T-cell infusion and the particular also be applied to indirectly modulate the host immune sys-
conditioning regimen that are superior is unclear. Early infu- tem and the tumor microenvironment. For example, T cells
sion post conditioning seems to be optimal, with infusion on engineered to express an NK2GD-targeting CAR were shown
day +2 apparently superior to later time points in term of to recruit and activate host lymphocytes in a chemokine-
immune reconstitution after high dose chemotherapy and dependent manner, leading to the development of broader
stem cell infusion (24, 330). A number of regimens have host immune responses (336).
been employed for lymphodepletion, including TBI, fludara-
bine, and cyclophosphamide, high dose chemotherapy or Clinical overview of ACT
bendamustine; however, no direct comparison of these pro- As discussed above, immunotherapy using TILs has a long
tocols is available. Finally, it is possible that the optimal che- clinical history, and this approach has provided important
motherapeutic regimen will be unique to individual tumor mechanistic and clinical insights about the potency of ACT.
types with disease specific to most effectively target both the However, technical issues have prevented the broader imple-
microenvironment and the disease. mentation of this approach to target cancers. The ability to
Vasculature is another important mechanistic and structural at-will genetically engineer and ex vivo manipulate and
component of the tumor milieu; accordingly, the targeting of
© 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
28 Immunological Reviews 257/2014
Ruella & Kalos  Adoptive immunotherapy for cancer

expand potent T cells has brought about what promises to Three recent clinical trials targeting members of the
be the beginning of a golden age for ACT, with multiple MAGE family have served to highlight the previously
ongoing clinical efforts, unprecedented positive clinical theoretical safety concerns associated with the potency of
results, and a real possibility for late stage development and TCR-engineered T-cell therapy. In one trial, T cells were
regulatory approval of ACT in the next few years. Here, we engineered to express a TCR generated in HLA-A*0201 trans-
present a summary of the most salient clinical results genic mice (i.e. not sculpted by the human immune system)
obtained over the past few years that provide insights into and that recognized an epitope shared between MAGE-A3,
the promise and pitfalls this exciting treatment modality. MAGE-A9, and MAGE–A12. Of nine patients treated, five
demonstrated objective clinical responses, but three patients
TCR-redirected T cells demonstrated SAE associated with neural toxicity, including
The first successful clinical experience with TCR redirected T two deaths. Postmortem analysis revealed rare and previously
cells was in the setting of melanoma using a low affinity unrecognized expression of MAGE-A12 in brain tissue (341).
HLA-A2-restricted anti-MART1 (clone DMF4). Two of 17 Two trials which evaluated the use of affinity enhanced HLA-
patients showed an objective response with partial tumor A*01-restricted and MAGE-A3-specific TCR to target mela-
regression with no significant toxicity, with the infused T noma and myeloma respectively. The first treated patient in
cells persisting at very low levels for more than 1 year each of these trials experienced SAE associated with cardio-
(337). A subsequent trial utilizing T cells that expressed a toxicity and each patient died within 7 days of T-cell infu-
high affinity anti-MART-1 (clone DMF5) or an anti-gp100 sion (75). Extensive and detailed retrospective analysis
TCR resulted in objective responses in about 25% of the demonstrated that the affinity enhancement of the TCR
patients. This trial also demonstrated the first example of on resulted in the off target recognition of a related epitope from
target toxicity with 29/36 patients experiencing hearing the protein titin expressed in cardiomyocytes (75).
loss, erythematous skin rash, vitiligo, and/or anterior uveitis TCRs to target carcino-embryonic antigen (CEA) have
due to the destruction of normal tissues that expressed the been also recently evaluated in ACT. CEA is a glycoprotein
target antigens (67). involved in cell adhesion and is expressed mainly in gastro-
A number of clinical trials have been conducted over the intestinal tissue during fetal development. CEA is overex-
past few years to target members of the CT family of anti- pressed in different cancers, in particular in colorectal
gens. CT antigens are expressed in germ cell tissues but not cancer. An anti-CEA high affinity TCR was recently
in adult tissues, excluding the immune privileged testes, and employed in a clinical trial to treat metastatic colon cancer.
are aberrantly expressed in the cytoplasm of various solid One of three patients treated showed objective response but
tumor cells, thus representing in principle ideal targets for all patients developed severe on target toxicity with inflam-
ACT (338). NY-ESO-1 (New York-esophageal squamous cell matory colitis. Transient clinical responses as manifested by
carcinoma-1) is one such antigen, expressed in about 80% reductions in circulating levels of CEA were observed in
of synovial carcinoma, 30–40% of breast, urothelial, esoph- each of these patients (66).
ageal, thyroid, prostate, melanoma, hepatic, gastric cancers, Other TCR-based target currently being preclinically eval-
and neuroblastomas (34). In a clinical trial at NCI, 4/6 uated include the human onco-protein MDM-2 (64), the
synovial sarcoma patients and 5/11 melanoma patients p53 suppressor gene (65, 342), the tyrosinase melanocyte
showed objective clinical response following treatment with differentiation antigen, the prostate and breast cancer anti-
affinity-enhanced anti-NY-ESO-1 TCR-redirected T cells; gen TARP, the telomerase (343) and the Wilms’ tumor 1
importantly, and perhaps exceptionally - see below, no sig- antigen (WT-1) (344, 345). A recent clinical trial evaluated
nificant toxicities were recorded (31). A number of addi- the infusion of autologous T cells genetically modified for
tional ACT-based clinical trials to target NY-ESO-1 in the expression of a HA1-specific TCR to leukemia patients
multiple solid tumors are currently ongoing. relapsing after HSCT (346).
The MAGE (melanoma-associated antigen) group of CT
antigens has also been targeted in multiple ACT-based trials. CAR-redirected T cells
The MAGE family encompasses a large number of sequence- The clinical evaluation of CAR-redirected T-cell-based ACT
related members which are expressed at various frequencies has exploded over the past few years, with more than 25 clin-
in a broad range multiple solid tumor types (339, 340). ical trials of CART that are actively recruiting, as registered in

© 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
Immunological Reviews 257/2014 29
Ruella & Kalos  Adoptive immunotherapy for cancer

clinicaltrials.gov (as of 15 July 2013). Among these, half are all five treated patients, including two patients with signifi-
evaluating CART-19 in B neoplasms, three CART against Her2 cant disease burden. The CART treatment was followed by
in GBM and sarcoma, and the remaining different other tar- allogeneic cell transplant in 4/5 patients, precluding analy-
gets (GD2, j light chain, CD30, EGFR, CD33, CD138, FAP, sis of long-term efficacy for the CART cells (30). At Baylor
CEA, mesothelin). College of Medicine, six patients with relapsed or refractory
Initial clinical studies with 1st generation CART targeting NHL were treated with a combination of first- and second-
neuroblastoma, lymphoma, renal, and ovarian cancers were generation CART19 in the absence of lymphodepletion; sec-
disappointing, with limited clinical activity, lack of in vivo ond generation CARs showed enhanced persistence in this
expansion and long-term persistence in vivo (93–96). study. Despite transient stabilization of lymphoma in two
Improvements of CAR engineering led to more positive clin- patients, none showed evidence of sustained tumor regres-
ical outcomes and also revealed the potential for SAE. Two sion at the cell doses used (100). Perhaps the most dra-
studies in particular, targeting carbonic anhydrase -9 (CA- matic results to date have been reported from our group at
IX) in renal carcinoma patients using 2nd generation CARs the University of Pennsylvania in the setting of adult CLL
and ERBB2 in colon cancer using 3rd generation CARs dem- (27, 28) and pediatric ALL (29). These studies employ sec-
onstrated considerable on-target off-tissue toxicity, with ond generation CARs with 4-1BB and CD3f costimulatory
liver toxicity due to targeting of CA-IX-positive biliary epi- domains, and lentivirus-engineered T cells, expanded using
thelium (148) and death from respiratory distress syndrome anti-CD3 and -CD28 coated beads, and infused 2 days after
due to the activation of T cells presumably in response to lymphodepletion. In these studies, CART cells were able to
low expression of ERBB2 by lung epithelium (153). Other eradicate very large tumor burdens and mediate complete
notable studies include the targeting of the glycolipid anti- and ongoing complete remissions, with ongoing functional
gen GD2 on neuroblastoma with Epstein-Barr virus (EBV)- persistence of CART cells now beyond 3 years in 2/3 initial
specific T cells engineered to express 2nd generation CARs, CLL patients and 1/2 ALL patients, as revealed by ongoing
which resulted in tumor regression or necrosis in 4/8 B-cell aplasia. SAE of varying severity related to delayed
patients, with one CR and a PR, and persistence of infused tumor lysis syndrome, cytokine release syndrome, macro-
cells (155–157, 184). phage activation syndrome, and hemophagocytic lymphoh-
Most of the dramatic recent success of CART-based ACT istocytosis, all of varying severity are common features for
has been achieved in hematological tumors, in particular tar- responding patients in these trials. Elevated levels of a num-
geting the CD19 antigen which is expressed during B-cell ber of cytokines and chemokines, including IFNc, IL-6,
development from the early B-cell progenitor (pro-B cell) to MIP1a, MIp1b, MCP-1 point to the broad and systemic
the mature B cells and possibly follicular DCs and by almost immune response unleashed by this potent therapy. Toc-
all B-cell malignancies. The absence of expression of this tar- iluzimab, a humanized monoclonal antagonistic antibody
get in healthy tissues other than B cells has made it an ideal against the IL-6 receptor, has been identified and imple-
antigen to target for adoptive immunotherapy. mented as a key interventive treatment to mitigate the
Infusion of CD19-specific CART cells targeting CD19 impact of the cytokine cascade (348).
showed to be effective in acute lymphoblastic leukemia
(ALL), chronic lymphocytic leukemia (CLL), and non- Concluding thoughts
Hodgkin lymphomas (NHL) (27, 29, 30, 347). The first More than 18 years from the first clinical demonstration of
patients treated with second generation CART19 were at the the feasibility of T-cell adoptive transfer (349), the field is
NCI. These patients received anti-CD19 CARs containing poised to capitalize on the multitude of lessons learned as a
CD28 and CD3f costimulatory domains; this treatment result of combining the commitment, insights, and innova-
resulted in significant regression of tumor and a partial tions of a multitude of scientists combined with the
remission the results of the first eight patients subsequently profound bravery and generosity of patients. ACT using
enrolled in this trial were recently reported, with six of engineered T cells is demonstrating dramatic potency in
eight patients achieving an objective clinical response, clinical trials, leading to complete and durable responses in
accompanied by transient B-cell ablation (130, 347). A patients with late stage and treatment refractory disease, and
recently published trial (30) from the MSKCC group in ALL commercialization of this emerging technology is likely to
applied CART therapy as a bridge toward allogeneic trans- occur in the next few years (26).
plant; this study showed activity of infused CART19-28f in
© 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
30 Immunological Reviews 257/2014
Ruella & Kalos  Adoptive immunotherapy for cancer

In this review, we have highlighted and expanded on a resolving such issue is likely to require the application of
number of topics of direct relevance for the application of broad, integrated, and quality supported assay platforms to
engineered T cells to ACT, within space and scope con- identify relevant correlations (352). (iii) Tumor burden
straints. In this regard, a number of topics relevant to the must be considered. With ACT representing a unique para-
specific implementation of this approach were not fully digm that involves the in vivo interaction of two viable cell
developed but merit description. Ultimate effective imple- populations, with the potential for T-cell potency, expan-
mentation of T-cell-based ACT is likely to require innova- sion, and persistence paradoxically dependent on higher
tion and co-development of at least some of these areas, as tumor burdens as reflected by the more profound cytokine
summarized here: (i) the issue of targeting aggressive ver- elevations observed in the context of higher disease bur-
sus indolent disease, exemplified by T-cell inhibitory effects dens (28–30). (iv) Strategies that combine ACT with the
mediated by CLL cells (350), and the effect of indolent use of agents that impact tumor biology such as demethy-
diseases on creating an immunosuppressive microenviron- lating agents (353), tumor signaling, metabolic pathway
ment, as illustrated by the differential effect of immune and cell cycle inhibitors (354–356) will need to be further
modulating drugs, such as lenalidomide and bortezomib) investigated. The development of such combination strate-
in the indolent setting; (ii) the issue of targeting solid gies, also including checkpoint inhibitor combinations as
tumors in general as well as the differential requirements discussed above, has the potential to unleash the full-
for targeting individual tumor types such as for example breadth of the immune response against tumors with
pancreatic cancer which has been described as resistant to potentially profound anti-tumor activity. (v) Finally, issues
T-cell infiltration (351). Although the discussion above of scale-up, automation, commercialization, and intellectual
with regard to tumor microenvironment is related to this property (26, 247), addressing at large scale regulatory
topic, tumor type-specific and perhaps even patient-specific hurdles unique to cell therapy (357), and identifying and
issues related to individual tumor biology, architecture, implementing reimbursement models will increasingly need
heterogeneity, and metabolism may well impact potency to be addressed as this field further matures and is applied
and effectiveness of ACT; accessing the relevance and to treat patients at large scale.

References
1. Tarella C, Gueli A, Ruella M, Cignetti A. 9. Salavoura K, Kolialexi A, Tsangaris G, Mavrou A. for pancreatic cancer. Cancer Immunol
Lymphocyte transformation and autoimmune Development of cancer in patients with primary Immunother 2013;62:949–954.
disorders. Autoimmun Rev 2013;12:802–813. immunodeficiencies. Anticancer Res 18. Sathish JG, et al. Challenges and approaches for
2. Simard EP, Pfeiffer RM, Engels EA. Cumulative 2008;28:1263–1269. the development of safer immunomodulatory
incidence of cancer among individuals with 10. Coley WB. The treatment of malignant tumors by biologics. Nat Rev Drug Discov 2013;12:306–
acquired immunodeficiency syndrome in the repeated inoculations of Erysipels, with a report 324.
United States. Cancer 2011;117:1089–1096. of ten original cases. Am J Med Sci 19. Jain RK. Normalizing tumor
3. Malhotra D, Fletcher AL, Turley SJ. Stromal and 1893;105:487–511. microenvironment to treat cancer: bench to
hematopoietic cells in secondary lymphoid 11. Lamm DL. Comparison of BCG with other bedside to biomarkers. J Clin Oncol
organs: partners in immunity. Immunol Rev intravesical agents. Urology 1991;37:30–32. 2013;31:2205–2218.
2013;251:160–176. 12. Kolb HJ, et al. Donor leukocyte transfusions for 20. Graziani G, Tentori L, Navarra P. Ipilimumab: a
4. Chauveau A, et al. Endothelial cell activation and treatment of recurrent chronic myelogenous novel immunostimulatory monoclonal antibody
proliferation modulate NKG2D activity by leukemia in marrow transplant patients. Blood for the treatment of cancer. Pharmacol Res
regulating MICA expression and shedding. J 1990;76:2462–2465. 2012;65:9–22.
Innate Immun 2013. [Epub ahead of print] 13. Gill S, Porter DL. Reduced-intensity 21. Callahan MK, Wolchok JD. At the Bedside:
5. Shen A, Baker JJ, Scott GL, Davis YP, Ho YY, hematopoietic stem cell transplants for CTLA-4- and PD-1-blocking antibodies in
Siliciano RF. Endothelial cell stimulation malignancies: harnessing the cancer immunotherapy. J Leukoc Biol
overcomes restriction and promotes productive graft-versus-tumor effect. Annu Rev Med 2013;94:41–53.
and latent HIV-1 infection of resting CD4+ T 2013;64:101–117. 22. Topalian SL, et al. Safety, activity, and immune
cells. J Virol 2013;87:9768–9779. 14. Cecco S, et al. Cancer vaccines in phase II/III correlates of anti-PD-1 antibody in cancer. N
6. Artis D. Epithelial-cell recognition of commensal clinical trials: state of the art and future Engl J Med 2012;366:2443–2454.
bacteria and maintenance of immune perspectives. Curr Cancer Drug Targets 23. Rapoport A, et al. Restoration of immunity in
homeostasis in the gut. Nat Rev Immunol 2011;11:85–102. lymphopenic individuals with cancer by
2008;8:411–420. 15. Leavy O. Therapeutic antibodies: past, present vaccination and adoptive T-cell transfer. Nat Med
7. Gorgun G, Anderson KC. Intrinsic modulation of and future. Nat Rev Immunol 2010;10:297. 2005;11:1230–1237.
lymphocyte function by stromal cell network: 16. Pardoll DM. The blockade of immune 24. Rapoport AP, et al. Rapid immune recovery and
advance in therapeutic targeting of cancer. checkpoints in cancer immunotherapy. Nat Rev graft-versus-host disease-like engraftment
Immunotherapy 2011;3:1253–1264. Cancer 2012;12:252–264. syndrome following adoptive transfer of
8. Cheon DJ, Orsulic S. Mouse models of cancer. 17. Vonderheide RH, Bajor DL, Winograd R, Evans Costimulated autologous T cells. Clin Cancer Res
Annu Rev Pathol 2011;6:95–119. RA, Bayne LJ, Beatty GL. CD40 immunotherapy 2009;15:4499–4507.

© 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
Immunological Reviews 257/2014 31
Ruella & Kalos  Adoptive immunotherapy for cancer

25. Rapoport AP, et al. Combination immunotherapy 41. Shimasaki N, Campana D. Natural killer cell repopulation with antitumor lymphocytes.
using adoptive T-cell transfer and tumor antigen reprogramming with chimeric immune Science 2002;298:850–854.
vaccination on the basis of hTERT and survivin receptors. Methods Mol Biol 2013;969:203–220. 57. Rosenberg SA, et al. Durable complete responses
after ASCT for myeloma. Blood 2011;117:788– 42. Lu PH, Negrin RS. A novel population of in heavily pretreated patients with metastatic
797. expanded human CD3+CD56+ cells derived from melanoma using T-cell transfer immunotherapy.
26. Kalos M, June CH. Adoptive T cell transfer for T cells with potent in vivo antitumor activity in Clin Cancer Res 2011;17:4550–4557.
cancer immunotherapy in the era of synthetic mice with severe combined immunodeficiency. J 58. Wrzesinski C, et al. Increased intensity
biology. Immunity 2013;39:49–60. Immunol 1994;153:1687–1696. lymphodepletion enhances tumor treatment
27. Porter DL, Levine BL, Kalos M, Bagg A, June CH. 43. Sangiolo D, Mesiano G, Carnevale-Schianca F, efficacy of adoptively transferred tumor-specific T
Chimeric antigen receptor-modified T cells in Piacibello W, Aglietta M, Cignetti A. Cytokine cells. J Immunother 2010;33:1–7.
chronic lymphoid leukemia. N Engl J Med induced killer cells as adoptive immunotherapy 59. Dudley ME, et al. CD8+ enriched “young” tumor
2011;365:725–733. strategy to augment graft versus tumor after infiltrating lymphocytes can mediate regression
28. Kalos M, et al. T cells with chimeric antigen hematopoietic cell transplantation. Expert Opin of metastatic melanoma. Clin Cancer Res
receptors have potent antitumor effects and can Biol Ther 2009;9:831–840. 2010;16:6122–6131.
establish memory in patients with advanced 44. Dhodapkar MV. Harnessing human CD1d 60. Cole DK, et al. Human TCR-binding affinity is
leukemia. Sci Transl Med 2011;3:95ra73. restricted T cells for tumor immunity: progress governed by MHC class restriction. J Immunol
29. Grupp SA, et al. Chimeric antigen and challenges. Front Biosci 2009;14:796–807. 2007;178:5727–5734.
receptor-modified T cells for acute lymphoid 45. Pilones KA, Aryankalayil J, Demaria S. Invariant 61. Aleksic M, et al. Different affinity windows for
leukemia. N Engl J Med 2013;368:1509–1518. NKT cells as novel targets for immunotherapy in virus and cancer-specific T-cell receptors:
30. Brentjens RJ, et al. CD19-Targeted T cells rapidly solid tumors. Clin Dev Immunol implications for therapeutic strategies. Eur J
induce molecular remissions in adults with 2012;2012:720803. Immunol 2012;42:3174–3179.
chemotherapy-refractory acute lymphoblastic 46. East JE, Sun W, Webb TJ. Artificial antigen 62. Dembic Z, et al. Transfer of specificity by murine
leukemia. Sci Transl Med 2013;5:177ra138. presenting cell (aAPC) mediated activation and alpha and beta T-cell receptor genes. Nature
31. Robbins PF, et al. Tumor regression in patients expansion of natural killer T cells. J Vis Exp 1986;320:232–238.
with metastatic synovial cell sarcoma and 2012;pii:4333. 63. Morgan RA, et al. High efficiency TCR gene
melanoma using genetically engineered 47. Simoni Y, Diana J, Ghazarian L, Beaudoin L, transfer into primary human lymphocytes affords
lymphocytes reactive with NY-ESO-1. J Clin Lehuen A. Therapeutic manipulation of natural avid recognition of melanoma tumor antigen
Oncol 2011;29:917–924. killer (NK) T cells in autoimmunity: are we close glycoprotein 100 and does not alter the
32. Mitchison NA. Studies on the immunological to reality? Clin Exp Immunol 2013;171:8–19. recognition of autologous melanoma antigens. J
response to foreign tumor transplants in the 48. Ferreira LM. Gammadelta T cells: innately Immunol 2003;171:3287–3295.
mouse. I. The role of lymph node cells in adaptive immune cells? Int Rev Immunol 64. Stanislawski T, et al. Circumventing tolerance to
conferring immunity by adoptive transfer. J Exp 2013;32:223–248. a human MDM2-derived tumor antigen by TCR
Med 1955;102:157–177. 49. Sakamoto M, et al. Adoptive immunotherapy for gene transfer. Nat Immunol 2001;2:962–970.
33. Greenberg PD, Reusser P, Goodrich JM, Riddell advanced non-small cell lung cancer using 65. Cohen CJ, et al. Recognition of fresh human
SR. Development of a treatment regimen for zoledronate-expanded gammadeltaTcells: a phase tumor by human peripheral blood lymphocytes
human cytomegalovirus (CMV) infection in bone I clinical study. J Immunother 2011;34:202–211. transduced with a bicistronic retroviral vector
marrow transplantation recipients by adoptive 50. Deniger DC, et al. Bispecific T-cells expressing encoding a murine anti-p53 TCR. J Immunol
transfer of donor-derived CMV-specific T cell polyclonal repertoire of endogenous gammadelta 2005;175:5799–5808.
clones expanded in vitro. Ann N Y Acad Sci T-cell receptors and introduced CD19-specific 66. Parkhurst MR, et al. T cells targeting
1991;636:184–195. chimeric antigen receptor. Mol Ther carcinoembryonic antigen can mediate regression
34. Rosenberg SA. Cell transfer immunotherapy for 2013;21:638–647. of metastatic colorectal cancer but induce severe
metastatic solid cancer–what clinicians need to 51. Law TM, et al. Phase III randomized trial of transient colitis. Mol Ther 2011;19:620–626.
know. Nat Rev Clin Oncol 2011;8:577–585. interleukin-2 with or without 67. Johnson LA, et al. Gene therapy with human and
35. Hunder NN, et al. Treatment of metastatic lymphokine-activated killer cells in the treatment mouse T-cell receptors mediates cancer
melanoma with autologous CD4+ T cells against of patients with advanced renal cell carcinoma. regression and targets normal tissues expressing
NY-ESO-1. N Engl J Med 2008;358:2698–2703. Cancer 1995;76:824–832. cognate antigen. Blood 2009;114:535–546.
36. Huang J, et al. Survival, persistence, and 52. Rosenberg SA, et al. Observations on the 68. Chinnasamy N, et al. A TCR targeting the
progressive differentiation of adoptively transferred systemic administration of autologous HLA-A*0201-restricted epitope of MAGE-A3
tumor-reactive T cells associated with tumor lymphokine-activated killer cells and recombinant recognizes multiple epitopes of the MAGE-A
regression. J Immunother 2005;28:258–267. interleukin-2 to patients with metastatic cancer. antigen superfamily in several types of cancer. J
37. Kanold J, et al. NK cell immunotherapy for N Engl J Med 1985;313:1485–1492. Immunol 2011;186:685–696.
high-risk neuroblastoma relapse after 53. Rosenberg SA, et al. Treatment of patients with 69. Li LP, et al. Transgenic mice with a diverse
haploidentical HSCT. Pediatr Blood Cancer metastatic melanoma with autologous human T cell antigen receptor repertoire. Nat
2012;59:739–742. tumor-infiltrating lymphocytes and interleukin 2. Med 2010;16:1029–1034.
38. Vivier E, Ugolini S, Blaise D, Chabannon C, J Natl Cancer Inst 1994;86:1159–1166. 70. Friedman RS, Spies AG, Kalos M. Identification of
Brossay L. Targeting natural killer cells and 54. Rosenberg SA, et al. Use of tumor-infiltrating naturally processed CD8 T cell epitopes from
natural killer T cells in cancer. Nat Rev Immunol lymphocytes and interleukin-2 in the prostein, a prostate tissue-specific vaccine
2012;12:239–252. immunotherapy of patients with metastatic candidate. Eur J Immunol 2004;34:1091–1101.
39. Miller JS, et al. Successful adoptive transfer and melanoma. A preliminary report. N Engl J Med 71. Amir AL, et al. PRAME-specific
in vivo expansion of human haploidentical NK 1988;319:1676–1680. allo-HLA-restricted T cells with potent antitumor
cells in patients with cancer. Blood 55. Dudley ME, et al. Adoptive cell therapy for reactivity useful for therapeutic T-cell receptor
2005;105:3051–3057. patients with metastatic melanoma: evaluation of gene transfer. Clin Cancer Res 2011;17:5615–
40. Rubnitz JE, et al. NKAML: a pilot study to intensive myeloablative chemoradiation 5625.
determine the safety and feasibility of preparative regimens. J Clin Oncol 72. Robbins PF, et al. Single and dual amino acid
haploidentical natural killer cell transplantation in 2008;26:5233–5239. substitutions in TCR CDRs can enhance
childhood acute myeloid leukemia. J Clin Oncol 56. Dudley ME, et al. Cancer regression and antigen-specific T cell functions. J Immunol
2010;28:955–959. autoimmunity in patients after clonal 2008;180:6116–6131.

© 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
32 Immunological Reviews 257/2014
Ruella & Kalos  Adoptive immunotherapy for cancer

73. Schaft N, et al. Peptide fine specificity of 90. Vitale M, et al. HLA class I antigen 104. Hombach AA, Abken H. Costimulation by
anti-glycoprotein 100 CTL is preserved following down-regulation in primary ovary carcinoma chimeric antigen receptors revisited the T cell
transfer of engineered TCR alpha beta genes into lesions: association with disease stage. ClinCancer antitumor response benefits from combined
primary human T lymphocytes. J Immunol Res 2005;11:67–72. CD28-OX40 signalling. Int J Cancer
2003;170:2186–2194. 91. Gross G, Waks T, Eshhar Z. Expression of 2011;129:2935–2944.
74. Stone JD, Chervin AS, Aggen DH, Kranz DM. T immunoglobulin-T-cell receptor chimeric 105. Altvater B, et al. 2B4 (CD244) signaling via
cell receptor engineering. Methods Enzymol molecules as functional receptors with chimeric receptors costimulates tumor-antigen
2012;503:189–222. antibody-type specificity. Proc Natl Acad Sci USA specific proliferation and in vitro expansion of
75. Linette GP, et al. Cardiovascular toxicity and titin 1989;86:10024–10028. human T cells. Cancer Immunol Immunother
cross-reactivity of affinity enhanced T cells in my- 92. Lanitis E, et al. Redirected antitumor activity of 2009;58:1991–2001.
eloma and melanoma. Blood 2013;122:863–871. primary human lymphocytes transduced with a 106. Song DG, Ye Q, Poussin M, Harms GM, Figini
76. de Witte MA, et al. Requirements for effective fully human anti-mesothelin chimeric receptor. M, Powell DJ, Jr.. CD27 costimulation augments
antitumor responses of TCR transduced T cells. J Mol Ther 2012;20:633–643. the survival and antitumor activity of redirected
Immunol 2008;181:5128–5136. 93. Park JR, et al. Adoptive transfer of chimeric human T cells in vivo. Blood 2012;119:696–
77. Jorritsma A, et al. Selecting highly affine and antigen receptor re-directed cytolytic T 706.
well-expressed TCRs for gene therapy of lymphocyte clones in patients with 107. Shen CJ, et al. Chimeric antigen receptor
melanoma. Blood 2007;110:3564–3572. neuroblastoma. Mol Ther 2007;15:825–833. containing ICOS signaling domain mediates
78. Szymczak AL, et al. Correction of multi-gene 94. Till BG, et al. Adoptive immunotherapy for specific and efficient antitumor effect of T cells
deficiency in vivo using a single ‘self-cleaving’ indolent non-Hodgkin lymphoma and mantle against EGFRvIII expressing glioma. J Hematol
2A peptide-based retroviral vector. Nat cell lymphoma using genetically modified Oncol 2013;6:33.
Biotechnol 2004;22:589–594. autologous CD20-specific T cells. Blood 108. Paulos CM, et al. The inducible costimulator
79. Scholten KB, et al. Codon modification of T cell 2008;112:2261–2271. (ICOS) is critical for the development of human
receptors allows enhanced functional expression 95. Lamers CH, et al. Treatment of metastatic renal T(H)17 cells. Sci Transl Med 2010;2:55ra78.
in transgenic human T cells. Clin Immunol cell carcinoma with autologous T-lymphocytes 109. Topp MS, Riddell SR, Akatsuka Y, Jensen MC,
2006;119:135–145. genetically retargeted against carbonic anhydrase Blattman JN, Greenberg PD. Restoration of CD28
80. Hart DP, et al. Retroviral transfer of a dominant IX: first clinical experience. J Clin Oncol expression in CD28 CD8+ memory effector T
TCR prevents surface expression of a large 2006;24:e20–e22. cells reconstitutes antigen-induced IL-2
proportion of the endogenous TCR repertoire in 96. Kershaw MH, et al. A phase I study on adoptive production. J Exp Med 2003;198:947–955.
human T cells. Gene Ther 2008;15:625–631. immunotherapy using gene-modified T cells for 110. Gill S, Kalos M. T cell-based gene therapy of
81. Leisegang M, et al. Enhanced functionality of T ovarian cancer. Clin Cancer Res 2006;12:6106– cancer. Transl Res 2013;161:365–379.
cell receptor-redirected T cells is defined by the 6115. 111. Carpenito C, et al. Control of large, established
transgene cassette. J Mol Med 2008;86:573–583. 97. Maher J, Brentjens RJ, Gunset G, Riviere I, tumor xenografts with genetically retargeted
82. Cohen CJ, Zhao Y, Zheng Z, Rosenberg SA, Sadelain M. Human T-lymphocyte cytotoxicity human T cells containing CD28 and CD137
Morgan RA. Enhanced antitumor activity of and proliferation directed by a single chimeric domains. Proc Natl Acad Sci USA
murine-human hybrid T-cell receptor (TCR) in TCRzeta/CD28 receptor. Nat Biotechnol 2009;106:3360–3365.
human lymphocytes is associated with improved 2002;20:70–75. 112. Zhong XS, Matsushita M, Plotkin J, Riviere I,
pairing and TCR/CD3 stability. Cancer Res 98. Finney HM, Akbar AN, Lawson AD. Activation of Sadelain M. Chimeric antigen receptors
2006;66:8878–8886. resting human primary T cells with chimeric combining 4-1BB and CD28 signaling domains
83. Bendle GM, et al. Lethal graft-versus-host disease receptors: costimulation from CD28, inducible augment PI3kinase/AKT/Bcl-XL activation and
in mouse models of T cell receptor gene therapy. costimulator, CD134, and CD137 in series with CD8+ T cell-mediated tumor eradication. Mol
Nat Med 2010;16:565–570. signals from the TCR zeta chain. J Immunol Ther 2010;18:413–420.
84. Kuball J, et al. Facilitating matched pairing and 2004;172:104–113. 113. Till BG, et al. CD20-specific adoptive
expression of TCR chains introduced into human 99. Loskog A, Giandomenico V, Rossig C, Pule M, immunotherapy for lymphoma using a chimeric
T cells. Blood 2007;109:2331–2338. Dotti G, Brenner MK. Addition of the CD28 antigen receptor with both CD28 and 4-1BB
85. Sommermeyer D, Uckert W. Minimal amino acid signaling domain to chimeric T-cell receptors domains: pilot clinical trial results. Blood
exchange in human TCR constant regions fosters enhances chimeric T-cell resistance to 2012;119:3940–3950.
improved function of TCR gene-modified T cells. T regulatory cells. Leukemia 114. Bridgeman JS, Hawkins RE, Hombach AA, Abken
J Immunol 2010;184:6223–6231. 2006;20:1819–1828. H, Gilham DE. Building better chimeric antigen
86. Voss RH, et al. Molecular design of the 100. Savoldo B, et al. CD28 costimulation improves receptors for adoptive T cell therapy. Curr Gene
Calphabeta interface favors specific pairing of expansion and persistence of chimeric antigen Ther 2010;10:77–90.
introduced TCRalphabeta in human T cells. J receptor-modified T cells in lymphoma patients. J 115. Shirasu N, Shibaguci H, Kuroki M, Yamada H.
Immunol 2008;180:391–401. Clin Invest 2011;121:1822–1826. Construction and molecular characterization of
87. Ochi T, et al. Novel adoptive T-cell 101. Song DG, et al. In vivo persistence, tumor human chimeric T-cell antigen receptors specific
immunotherapy using a WT1-specific TCR vector localization, and antitumor activity of for carcinoembryonic antigen. Anticancer Res
encoding silencers for endogenous TCRs shows CAR-engineered T cells is enhanced by 2010;30:2731–2738.
marked antileukemia reactivity and safety. Blood costimulatory signaling through CD137 (4-1BB). 116. Pule MA, Straathof KC, Dotti G, Heslop HE,
2011;118:1495–1503. Cancer Res 2011;71:4617–4627. Rooney CM, Brenner MK. A chimeric T cell
88. Okamoto S, et al. Improved expression and 102. Milone MC, et al. Chimeric receptors containing antigen receptor that augments cytokine release
reactivity of transduced tumor-specific TCRs in CD137 signal transduction domains mediate and supports clonal expansion of primary human
human lymphocytes by specific silencing of enhanced survival of T cells and increased T cells. Mol Ther 2005;12:933–941.
endogenous TCR. Cancer Res 2009;69:9003– antileukemic efficacy in vivo. Mol Ther 117. Kahlon KS, Brown C, Cooper LJ, Raubitschek A,
9011. 2009;17:1453–1464. Forman SJ, Jensen MC. Specific recognition and
89. Cohen CJ, Li YF, El-Gamil M, Robbins PF, 103. Hombach AA, Heiders J, Foppe M, Chmielewski killing of glioblastoma multiforme by interleukin
Rosenberg SA, Morgan RA. Enhanced antitumor M, Abken H. OX40 costimulation by a chimeric 13-zetakine redirected cytolytic T cells. Cancer
activity of T cells engineered to express T-cell antigen receptor abrogates CD28 and IL-2 Res 2004;64:9160–9166.
receptors with a second disulfide bond. Cancer induced IL-10 secretion by redirected CD4(+) T 118. Schambach A, Swaney WP, van der Loo JC.
Res 2007;67:3898–3903. cells. Oncoimmunology 2012;1:458–466. Design and production of retro- and lentiviral

© 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
Immunological Reviews 257/2014 33
Ruella & Kalos  Adoptive immunotherapy for cancer

vectors for gene expression in hematopoietic T-cell populations for leukemia cell therapy. J T-cells expressing HER2-specific chimeric antigen
cells. Methods Mol Biol 2009;506:191–205. Immunother 2011;34:469–479. receptor. Mol Ther 2011;19:2133–2143.
119. Sinn PL, Sauter SL, McCray PB, Jr.. Gene therapy 135. Carpenter RO, et al. B-cell maturation antigen is 151. Davies DM, et al. Flexible targeting of ErbB
progress and prospects: development of a promising target for adoptive T-cell therapy of dimers that drive tumorigenesis by using
improved lentiviral and retroviral vectors–design, multiple myeloma. Clin Cancer Res genetically engineered T cells. Mol Med
biosafety, and production. Gene Ther 2013;19:2048–2060. 2012;18:565–576.
2005;12:1089–1098. 136. Giordano Attianese GM, et al. In vitro and in 152. Zhao Y, et al. A herceptin-based chimeric antigen
120. Church DN, Talbot DC. Survivin in solid tumors: vivo model of a novel immunotherapy approach receptor with modified signaling domains leads
rationale for development of inhibitors. Curr for chronic lymphocytic leukemia by anti-CD23 to enhanced survival of transduced T
Oncol Rep 2012;14:120–128. chimeric antigen receptor. Blood lymphocytes and antitumor activity. J Immunol
121. Tammana S, et al. 4-1BB and CD28 signaling 2011;117:4736–4745. 2009;183:5563–5574.
plays a synergistic role in redirecting umbilical 137. Di Stasi A, et al. T lymphocytes coexpressing 153. Morgan R, Yang J, Kitano M, Dudley M,
cord blood T cells against B-cell malignancies. CCR4 and a chimeric antigen receptor targeting Laurencot C, Rosenberg S. Case report of a
Hum Gene Ther 2010;21:75–86. CD30 have improved homing and antitumor serious adverse event following the
122. Zhao Y, et al. High-efficiency transfection of activity in a Hodgkin tumor model. Blood administration of T cells transduced with a
primary human and mouse T lymphocytes using 2009;113:6392–6402. chimeric antigen receptor recognizing ERBB2.
RNA electroporation. Mol Ther 2006;13:151– 138. Savoldo B, et al. Epstein Barr virus specific Mol Ther 2010;18:843–851.
159. cytotoxic T lymphocytes expressing the 154. Krause A, Guo HF, Latouche JB, Tan C, Cheung
123. Hombach A, Hombach AA, Abken H. Adoptive anti-CD30zeta artificial chimeric T-cell receptor NK, Sadelain M. Antigen-dependent CD28
immunotherapy with genetically engineered T for immunotherapy of Hodgkin disease. Blood signaling selectively enhances survival and
cells: modification of the IgG1 Fc ‘spacer’ 2007;110:2620–2630. proliferation in genetically modified activated
domain in the extracellular moiety of chimeric 139. Mihara K, et al. Activated T-cell-mediated human primary T lymphocytes. J Exp Med
antigen receptors avoids ‘off-target’ activation immunotherapy with a chimeric receptor against 1998;188:619–626.
and unintended initiation of an innate immune CD38 in B-cell non-Hodgkin lymphoma. J 155. Pule MA, et al. Virus-specific T cells engineered
response. Gene Ther 2010;17:1206–1213. Immunother 2009;32:737–743. to coexpress tumor-specific receptors:
124. Wilkie S, et al. Retargeting of human T cells to 140. Hekele A, et al. Growth retardation of tumors by persistence and antitumor activity in individuals
tumor-associated MUC1: the evolution of a adoptive transfer of cytotoxic T lymphocytes with neuroblastoma. Nat Med 2008;14:1264–
chimeric antigen receptor. J Immunol reprogrammed by CD44v6-specific scFv: 1270.
2008;180:4901–4909. zeta-chimera. Int J Cancer 1996;68:232–238. 156. Louis CU, et al. Antitumor activity and long-term
125. Guest RD, et al. The role of extracellular spacer 141. Hudecek M, et al. Receptor affinity and fate of chimeric antigen receptor-positive T cells
regions in the optimal design of chimeric immune extracellular domain modifications affect tumor in patients with neuroblastoma. Blood
receptors: evaluation of four different scFvs and recognition by ROR1-specific chimeric antigen 2011;118:6050–6056.
antigens. J Immunother 2005;28:203–211. receptor T cells. Clin Cancer Res 2013;19:3153– 157. Yvon E, et al. Immunotherapy of metastatic
126. James SE, et al. Antigen sensitivity of 3164. melanoma using genetically engineered
CD22-specific chimeric TCR is modulated by 142. Vera J, et al. T lymphocytes redirected against GD2-specific T cells. Clin Cancer Res
target epitope distance from the cell membrane. J the kappa light chain of human immunoglobulin 2009;15:5852–5860.
Immunol 2008;180:7028–7038. efficiently kill mature B lymphocyte-derived 158. Lo AS, Ma Q, Liu DL, Junghans RP. Anti-GD3
127. Haso W, et al. Anti-CD22-chimeric antigen malignant cells. Blood 2006;108:3890–3897. chimeric sFv-CD28/T-cell receptor zeta designer
receptors targeting B-cell precursor acute 143. Ritchie DS, et al. Persistence and efficacy of T cells for treatment of metastatic melanoma and
lymphoblastic leukemia. Blood 2013;121:1165– second generation CAR-T cell against the LeY other neuroectodermal tumors. Clin Cancer Res
1174. antigen in acute myeloid leukemia. Mol Ther 2010;16:2769–2780.
128. Brentjens RJ, et al. Genetically targeted T cells 2013;21:2122–2129. 159. Chinnasamy D, et al. Gene therapy using
eradicate systemic acute lymphoblastic leukemia 144. Peinert S, et al. Gene-modified T cells as genetically modified lymphocytes targeting
xenografts. Clin Cancer Res 2007;13:5426–5435. immunotherapy for multiple myeloma and acute VEGFR-2 inhibits the growth of vascularized
129. Porter DL, Kalos M, Zheng Z, Levine B, June C. myeloid leukemia expressing the Lewis Y syngenic tumors in mice. J Clin Invest
Chimeric antigen receptor therapy for B-cell antigen. Gene Ther 2010;17:678–686. 2010;120:3953–3968.
malignancies. J Cancer 2011;2:331–332. 145. Zhang T, Wu MR, Sentman CL. An NKp30-based 160. Zhou X, et al. Cellular immunotherapy for
130. Kochenderfer JN, et al. Eradication of B-lineage chimeric antigen receptor promotes T cell carcinoma using genetically modified
cells and regression of lymphoma in a patient effector functions and antitumor efficacy in vivo. EGFR-specific T lymphocytes. Neoplasia
treated with autologous T cells genetically J Immunol 2012;189:2290–2299. 2013;15:544–553.
engineered to recognize CD19. Blood 146. McGuinness RP, et al. Anti-tumor activity of 161. Ohno M, et al. Retrovirally engineered
2010;116:4099–4102. human T cells expressing the CC49-zeta chimeric T-cell-based immunotherapy targeting type III
131. Kebriaei P, et al. Infusing CD19-directed T cells to immune receptor. Hum Gene Ther variant epidermal growth factor receptor, a
augment disease control in patients undergoing 1999;10:165–173. glioma-associated antigen. Cancer Sci
autologous hematopoietic stem-cell transplantation 147. Shaffer DR, et al. T cells redirected against CD70 2010;101:2518–2524.
for advanced B-lymphoid malignancies. Human for the immunotherapy of CD70-positive 162. Chekmasova AA, et al. Successful eradication of
Gene Ther 2012;23:444–450. malignancies. Blood 2011;117:4304–4314. established peritoneal ovarian tumors in
132. Wang J, et al. Cellular immunotherapy for 148. Lamers CH, et al. Treatment of metastatic renal SCID-Beige mice following adoptive transfer of T
follicular lymphoma using genetically modified cell carcinoma with CAIX CAR-engineered T cells genetically targeted to the MUC16 antigen.
CD20-specific CD8+ cytotoxic T lymphocytes. cells: clinical evaluation and management of Clin Cancer Res 2010;16:3594–3606.
Mol Ther 2004;9:577–586. on-target toxicity. Mol Ther 2013;21:904–912. 163. Gong MC, Latouche JB, Krause A, Heston WD,
133. Dutour A, et al. In vitro and in vivo antitumor 149. Rainusso N, et al. Immunotherapy targeting Bander NH, Sadelain M. Cancer patient T cells
effect of anti-CD33 chimeric receptor-expressing HER2 with genetically modified T cells eliminates genetically targeted to prostate-specific
EBV-CTL against CD33 acute myeloid leukemia. tumor-initiating cells in osteosarcoma. Cancer membrane antigen specifically lyse prostate
Adv Hematol 2012;2012:683065. Gene Ther 2012;19:212–217. cancer cells and release cytokines in response to
134. Pizzitola I, et al. In vitro comparison of three 150. Nakazawa Y, et al. PiggyBac-mediated cancer prostate-specific membrane antigen. Neoplasia
different chimeric receptor-modified effector immunotherapy using EBV-specific cytotoxic 1999;1:123–127.

© 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
34 Immunological Reviews 257/2014
Ruella & Kalos  Adoptive immunotherapy for cancer

164. Ma Q, Safar M, Holmes E, Wang Y, Boynton AL, 179. Grada Z, et al. TanCAR: a novel bispecific antitumor immunity. Proc Natl Acad Sci USA
Junghans RP. Anti-prostate specific membrane chimeric antigen receptor for cancer 2009;106:17469–17474.
antigen designer T cells for prostate cancer immunotherapy. Mol Ther Nucleic Acids 2013;2: 195. Hinrichs CS, et al. Human effector CD8+ T cells
therapy. Prostate 2004;61:12–25. e105. derived from naive rather than memory subsets
165. Katari UL, et al. Engineered T cells for pancreatic 180. Kloss CC, Condomines M, Cartellieri M, possess superior traits for adoptive
cancer treatment. HPB 2011;13:643–650. Bachmann M, Sadelain M. Combinatorial antigen immunotherapy. Blood 2011;117:808–814.
166. Morgenroth A, et al. Targeting of tumor cells recognition with balanced signaling promotes 196. Berger C, Jensen MC, Lansdorp PM, Gough M,
expressing the prostate stem cell antigen (PSCA) selective tumor eradication by engineered T cells. Elliott C, Riddell SR. Adoptive transfer of effector
using genetically engineered T-cells. Prostate Nat Biotechnol 2013;31:71–75. CD8+ T cells derived from central memory cells
2007;67:1121–1131. 181. Urbanska K, et al. A universal strategy for establishes persistent T cell memory in primates.
167. Jiang HR, Gilham DE, Mulryan K, Kirillova N, adoptive immunotherapy of cancer through use J Clin Invest 2008;118:294–305.
Hawkins RE, Stern PL. Combination of of a novel T-cell antigen receptor. Cancer Res 197. Gattinoni L, et al. A human memory T cell
vaccination and chimeric receptor expressing T 2012;72:1844–1852. subset with stem cell-like properties. Nat Med
cells provides improved active therapy of tumors. 182. Dao T, et al. Targeting the intracellular WT1 2011;17:1290–1297.
J Immunol 2006;177:4288–4298. oncogene product with a therapeutic 198. Stemberger C, Neuenhahn M, Gebhardt FE,
168. Tchou J, et al. Mesothelin, a novel immunotherapy human antibody. Sci Transl Med Schiemann M, Buchholz VR, Busch DH. Stem
target for triple negative breast cancer. Breast 2013;5:176ra133. cell-like plasticity of naive and distinct memory
Cancer Res Treat 2012;133:799–804. 183. Kershaw MH, et al. Redirecting migration of T CD8+ T cell subsets. Semin Immunol
169. Kandalaft LE, Powell DJ, Jr., Coukos G. A phase I cells to chemokine secreted from tumors by 2009;21:62–68.
clinical trial of adoptive transfer of folate genetic modification with CXCR2. Hum Gene 199. Hombach AA, Chmielewski M, Rappl G, Abken
receptor-alpha redirected autologous T cells for Ther 2002;13:1971–1980. H. Adoptive immunotherapy with redirected T
recurrent ovarian cancer. J Transl Med 184. Craddock JA, et al. Enhanced tumor trafficking of cells produces CCR7- cells that are trapped in the
2012;10:157. GD2 chimeric antigen receptor T cells by periphery and benefit from combined
170. Spear P, Barber A, Rynda-Apple A, Sentman CL. expression of the chemokine receptor CCR2b. J CD28-OX40 costimulation. Hum Gene Ther
NKG2D CAR T-cell therapy inhibits the growth Immunother 2010;33:780–788. 2013;24:259–269.
of NKG2D ligand heterogeneous tumors. 185. Moon EK, et al. Expression of a functional CCR2 200. Klebanoff CA, et al. Central memory self/
Immunol Cell Biol 2013;91:435–440. receptor enhances tumor localization and tumor tumor-reactive CD8+ T cells confer superior
171. Barber A, Meehan KR, Sentman CL. Treatment of eradication by retargeted human T cells antitumor immunity compared with effector
multiple myeloma with adoptively transferred expressing a mesothelin-specific chimeric memory T cells. Proc Natl Acad Sci USA
chimeric NKG2D receptor-expressing T cells. antibody receptor. Clin Cancer Res 2005;102:9571–9576.
Gene Ther 2011;18:509–516. 2011;17:4719–4730. 201. Roberts AD, Ely KH, Woodland DL. Differential
172. Zhang T, Barber A, Sentman CL. Generation 186. Kerkar SP, et al. Collapse of the tumor stroma is contributions of central and effector memory T
of antitumor responses by genetic modification triggered by IL-12 induction of Fas. Mol Ther cells to recall responses. J Exp Med
of primary human T cells with a chimeric 2013;21:1369–1377. 2005;202:123–133.
NKG2D receptor. Cancer Res 2006;66:5927– 187. Kerkar SP, et al. IL-12 triggers a programmatic 202. Sallusto F, Lenig D, Forster R, Lipp M,
5933. change in dysfunctional myeloid-derived cells Lanzavecchia A. Two subsets of memory T
173. Huang G, Yu L, Cooper LJ, Hollomon M, Huls within mouse tumors. J Clin Invest lymphocytes with distinct homing potentials and
H, Kleinerman ES. Genetically modified T cells 2011;121:4746–4757. effector functions. Nature 1999;401:708–712.
targeting interleukin-11 receptor alpha-chain kill 188. Pegram HJ, et al. Tumor-targeted T cells 203. Fritsch RD, Shen X, Sims GP, Hathcock KS,
human osteosarcoma cells and induce the modified to secrete IL-12 eradicate systemic Hodes RJ, Lipsky PE. Stepwise differentiation of
regression of established osteosarcoma lung tumors without need for prior conditioning. CD4 memory T cells defined by expression of
metastases. Cancer Res 2012;72:271–281. Blood 2012;119:4133–4141. CCR7 and CD27. J Immunol 2005;175:6489–
174. Chmielewski M, et al. T cells that target 189. Zhang L, et al. Evaluation of gamma-retroviral 6497.
carcinoembryonic antigen eradicate orthotopic vectors that mediate the inducible expression of 204. Schwartzentruber DJ, et al. In vitro predictors of
pancreatic carcinomas without inducing IL-12 for clinical application. J Immunother therapeutic response in melanoma patients
autoimmune colitis in mice. Gastroenterology 2012;35:430–439. receiving tumor-infiltrating lymphocytes and
2012;143:e1092. 190. Klebanoff CA, Gattinoni L, Restifo NP. Sorting interleukin-2. J Clin Oncol 1994;12:1475–1483.
175. Emtage PC, Lo AS, Gomes EM, Liu DL, through subsets: which T-cell populations 205. Besser MJ, et al. Clinical responses in a phase II
Gonzalo-Daganzo RM, Junghans RP. mediate highly effective adoptive study using adoptive transfer of short-term
Second-generation anti-carcinoembryonic antigen immunotherapy? J Immunother 2012;35:651– cultured tumor infiltration lymphocytes in
designer T cells resist activation-induced cell 660. metastatic melanoma patients. Clin Cancer Res
death, proliferate on tumor contact, secrete 191. Yee C, et al. Melanocyte destruction after 2010;16:2646–2655.
cytokines, and exhibit superior antitumor activity antigen-specific immunotherapy of melanoma: 206. Itzhaki O, et al. Establishment and large-scale
in vivo: a preclinical evaluation. Clin Cancer Res direct evidence of t cell-mediated vitiligo. J Exp expansion of minimally cultured “young” tumor
2008;14:8112–8122. Med 2000;192:1637–1644. infiltrating lymphocytes for adoptive transfer
176. Kong S, et al. Suppression of human glioma 192. Yee C, et al. Adoptive T cell therapy using therapy. J Immunother 2011;34:212–220.
xenografts with second-generation IL13R-specific antigen-specific CD8+ T cell clones for the 207. Olson JA, McDonald-Hyman C, Jameson SC,
chimeric antigen receptor-modified T cells. Clin treatment of patients with metastatic melanoma: Hamilton SE. Effector-like CD8<sup>+</sup> T
Cancer Res 2012;18:5949–5960. in vivo persistence, migration, and antitumor cells in the memory population mediate potent
177. Chow KK, et al. T cells redirected to EphA2 for effect of transferred T cells. Proc Natl Acad Sci protective immunity. Immunity 2013;38:1250–
the immunotherapy of glioblastoma. Mol Ther USA 2002;99:16168–16173. 1260.
2013;21:629–637. 193. Gattinoni L, Klebanoff CA, Restifo NP. Paths to 208. Martin-Orozco N, et al. T helper 17 cells
178. Torikai H, et al. A foundation for universal T-cell stemness: building the ultimate antitumour T promote cytotoxic T cell activation in tumor
based immunotherapy: T cells engineered to cell. Nat Rev Cancer 2012;12:671–684. immunity. Immunity 2009;31:787–798.
express a CD19-specific chimeric-antigen-receptor 194. Hinrichs CS, et al. Adoptively transferred effector 209. Kryczek I, et al. Human TH17 cells are
and eliminate expression of endogenous TCR. cells derived from naive rather than central long-lived effector memory cells. Sci Transl Med
Blood 2012;119:5697–5705. memory CD8+ T cells mediate superior 2011;3:104ra100.

© 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
Immunological Reviews 257/2014 35
Ruella & Kalos  Adoptive immunotherapy for cancer

210. Sun JC, Bevan MJ. Defective CD8 T cell memory 227. Muul LM, et al. Persistence and expression of the adoptively transferred CD20/CD19-specific
following acute infection without CD4 T cell adenosine deaminase gene for 12 years and chimeric antigen receptor redirected T cells in
help. Science 2003;300:339–342. immune reaction to gene transfer components: humans. Biol Blood Marrow Transplant
211. Sun JC, Bevan MJ. Cutting edge: long-lived CD8 long-term results of the first clinical gene therapy 2010;16:1245–1256.
memory and protective immunity in the absence trial. Blood 2003;101:2563–2569. 244. Gattinoni L, et al. Acquisition of full effector
of CD40 expression on CD8 T cells. J Immunol 228. Williams DA. Foamy virus vectors come of age. function in vitro paradoxically impairs the in
2004;172:3385–3389. Mol Ther 2008;16:635–636. vivo antitumor efficacy of adoptively transferred
212. Brenner MK, Heslop HE. Adoptive T cell therapy 229. Perez EE, et al. Establishment of HIV-1 resistance CD8+ T cells. J Clin Invest 2005;115:1616–
of cancer. Curr Opin Immunol 2010;22:251– in CD4+ T cells by genome editing using 1626.
257. zinc-finger nucleases. Nat Biotechnol 245. Porter DL, et al. A phase 1 trial of donor
213. Louis CU, et al. Adoptive transfer of EBV-specific 2008;26:808–816. lymphocyte infusions expanded and activated ex
T cells results in sustained clinical responses in 230. Wilson MH, Coates CJ, George AL, Jr.. PiggyBac vivo via CD3/CD28 costimulation. Blood
patients with locoregional nasopharyngeal transposon-mediated gene transfer in human 2006;107:1325–1331.
carcinoma. J Immunother 2010;33:983–990. cells. Mol Ther 2007;15:139–145. 246. Levine BL, et al. Antiviral effect and ex vivo
214. Hoyos V, et al. Engineering CD19-specific T 231. Hackett PB, Largaespada DA, Cooper LJ. A CD4+ T cell proliferation in HIV-positive patients
lymphocytes with interleukin-15 and a suicide transposon and transposase system for human as a result of CD28 costimulation. Science
gene to enhance their anti-lymphoma/leukemia application. Mol Ther 2010;18:674–683. 1996;272:1939–1943.
effects and safety. Leukemia 2010;24:1160– 232. Manuri PV, et al. piggyBac transposon/ 247. Levine BL, June CH. Perspective: assembly line
1170. transposase system to generate CD19-specific T immunotherapy. Nature 2013;498:S17.
215. Rowley J, Monie A, Hung CF, Wu TC. cells for the treatment of B-lineage malignancies. 248. Turtle CJ, Swanson HM, Fujii N, Estey EH,
Expression of IL-15RA or an IL-15/IL-15RA Hum Gene Ther 2010;21:427–437. Riddell SR. A distinct subset of self-renewing
fusion on CD8+ T cells modifies adoptively 233. Reyon D, Tsai SQ, Khayter C, Foden JA, Sander human memory CD8+ T cells survives cytotoxic
transferred T-cell function in cis. Eur J Immunol JD, Joung JK. FLASH assembly of TALENs for chemotherapy. Immunity 2009;31:834–844.
2009;39:491–506. high-throughput genome editing. Nat Biotechnol 249. Buchholz VR, Neuenhahn M, Busch DH. CD8+ T
216. Asai H, et al. Co-introduced functional CCR2 2012;30:460–465. cell differentiation in the aging immune system:
potentiates in vivo anti-lung cancer functionality 234. Tebas P, et al. Antiviral effects of autologous CD4 until the last clone standing. Curr Opin Immunol
mediated by T cells double gene-modified to T cells genetically modified with a conditionally 2011;23:549–554.
express WT1-specific T-cell receptor. PLoS ONE replicating lentiviral vector expressing long 250. Levine BL. T lymphocyte engineering ex vivo for
2013;8:e56820. antisense to HIV. Blood 2013;121:1524–1533. cancer and infectious disease. Expert Opin Biol
217. Kang TH, et al. Tumor-targeted delivery of IL-2 235. Barrett DM, et al. Treatment of advanced Ther 2008;8:475–489.
by NKG2D leads to accumulation of leukemia in mice with mRNA engineered T cells. 251. Samarasinghe S, et al. Functional characterization
antigen-specific CD8+ T cells in the tumor loci Human Gene Ther 2011;22:1575–1586. of alloreactive T cells identifies CD25 and CD71
and enhanced anti-tumor effects. PLoS ONE 236. Zhao Y, et al. Multiple injections of as optimal targets for a clinically applicable
2012;7:e35141. electroporated autologous T cells expressing a allodepletion strategy. Blood 2010;115:396–407.
218. Rosenberg SA, Kochenderfer JN. Personalized cell chimeric antigen receptor mediate regression of 252. Touil S, et al. Depletion of T regulatory cells
transfer immunotherapy for B-cell malignancies human disseminated tumor. Cancer Res through selection of CD127-positive cells results
and solid cancers. Mol Ther 2011;19:1928– 2010;70:9053–9061. in a population enriched in memory T cells:
1930. 237. Mitchell DA, Karikari I, Cui X, Xie W, implications for anti-tumor cell therapy.
219. Sadelain M. T-cell engineering for cancer Schmittling R, Sampson JH. Selective Haematologica 2012;97:1678–1685.
immunotherapy. Cancer J 2009;15:451–455. modification of antigen-specific T cells by RNA 253. Terakura S, Yamamoto TN, Gardner RA, Turtle
220. Naldini L, et al. In vivo gene delivery and stable electroporation. Human Gene Ther CJ, Jensen MC, Riddell SR. Generation of
transduction of nondividing cells by a lentiviral 2008;19:511–521. CD19-chimeric antigen receptor modified CD8+
vector. Science 1996;272:263–267. 238. Yoon SH, et al. Adoptive immunotherapy using T cells derived from virus-specific central
221. Cavalieri S, et al. Human T lymphocytes human peripheral blood lymphocytes transferred memory T cells. Blood 2012;119:72–82.
transduced by lentiviral vectors in the absence of with RNA encoding Her-2/neu-specific chimeric 254. Keenan RD, et al. Purification of
TCR activation maintain an intact immune immune receptor in ovarian cancer xenograft cytomegalovirus-specific CD8 T cells from
competence. Blood 2003;102:497–505. model. Cancer Gene Ther 2009;16:489–497. peripheral blood using HLA-peptide tetramers. Br
222. Circosta P, et al. T cell receptor (TCR) gene 239. Chicaybam L, Sodre AL, Curzio BA, Bonamino J Haematol 2001;115:428–434.
transfer with lentiviral vectors allows efficient MH. An efficient low cost method for gene 255. Cobbold M, et al. Adoptive transfer of
redirection of tumor specificity in naive and transfer to T lymphocytes. PLoS ONE 2013;8: cytomegalovirus-specific CTL to stem cell
memory T cells without prior stimulation of e60298. transplant patients after selection by HLA-peptide
endogenous TCR. Hum Gene Ther 240. Riddell SR, Greenberg PD. The use of anti-CD3 tetramers. J Exp Med 2005;202:379–386.
2009;20:1576–1588. and anti-CD28 monoclonal antibodies to clone 256. Maus MV, et al. Ex vivo expansion of polyclonal
223. Hacein-Bey-Abina S, et al. Insertional and expand human antigen-specific T cells. J and antigen-specific cytotoxic T lymphocytes by
oncogenesis in 4 patients after Immunol Methods 1990;128:189–201. artificial APCs expressing ligands for the T-cell
retrovirus-mediated gene therapy of SCID-X1. J 241. Dudley ME, et al. A phase I study of receptor, CD28 and 4-1BB. Nat Biotechnol
Clin Invest 2008;118:3132–3142. nonmyeloablative chemotherapy and adoptive 2002;20:143–148.
224. Hacein-Bey-Abina S, et al. A serious adverse transfer of autologous tumor antigen-specific T 257. Thomas AK, Maus MV, Shalaby WS, June CH,
event after successful gene therapy for X-linked lymphocytes in patients with metastatic Riley JL. A cell-based artificial antigen-presenting
severe combined immunodeficiency. N Engl J melanoma. J Immunother 2002;25:243–251. cell coated with anti-CD3 and CD28 antibodies
Med 2003;348:255–256. 242. Duval L, et al. Adoptive transfer of allogeneic enables rapid expansion and long-term growth of
225. Newrzela S, et al. Resistance of mature T cells to cytotoxic T lymphocytes equipped with a CD4 T lymphocytes. Clin Immunol
oncogene transformation. Blood 2008;112:2278– HLA-A2 restricted MART-1 T-cell receptor: a 2002;105:259–272.
2286. phase I trial in metastatic melanoma. Clin Cancer 258. Ye Q, et al. Engineered artificial antigen
226. Scholler J, et al. Decade-long safety and function Res 2006;12:1229–1236. presenting cells facilitate direct and efficient
of retroviral-modified chimeric antigen receptor 243. Jensen MC, et al. Antitransgene rejection expansion of tumor infiltrating lymphocytes. J
T cells. Sci Transl Med 2012;4:132ra153. responses contribute to attenuated persistence of Transl Med 2011;9:131.

© 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
36 Immunological Reviews 257/2014
Ruella & Kalos  Adoptive immunotherapy for cancer

259. Numbenjapon T, Serrano LM, Chang WC, 275. Berger C, Flowers ME, Warren EH, Riddell SR. 291. Fu X, Rivera A, Tao L, Zhang X. Genetically
Forman SJ, Jensen MC, Cooper LJ. Analysis of transgene-specific immune responses modified T cells targeting neovasculature
Antigen-independent and antigen-dependent that limit the in vivo persistence of adoptively efficiently destroy tumor blood vessels, shrink
methods to numerically expand CD19-specific transferred HSV-TK-modified donor T cells after established solid tumors and increase
CD8+ T cells. Exp Hematol 2007;35:1083–1090. allogeneic hematopoietic cell transplantation. nanoparticle delivery. Int J Cancer
260. Hasan AN, Selvakumar A, Doubrovina E, Riviere Blood 2006;107:2294–2302. 2013;133:2483–2492.
I, Sadelain MW, O’Reilly RJ. Artificial antigen 276. Straathof KC, et al. An inducible caspase 9 safety 292. Marx J. Cancer biology. All in the stroma:
presenting cells that express prevalent HLA switch for T-cell therapy. Blood 2005;105:4247– cancer’s Cosa Nostra. Science 2008;320:38–41.
alleles: a step towards the broad application of 4254. 293. Polyak K, Haviv I, Campbell IG. Co-evolution of
antigen-specific adoptive cell therapies. Discov 277. Di Stasi A, et al. Inducible apoptosis as a safety tumor cells and their microenvironment. Trends
Med 2009;8:210–218. switch for adoptive cell therapy. N Engl J Med Genet 2009;25:30–38.
261. Li Y, Zhi W, Wareski P, Weng NP. IL-15 2011;365:1673–1683. 294. Chometon G, Jendrossek V. Targeting the tumour
activates telomerase and minimizes telomere loss 278. Wang X, et al. A transgene-encoded cell surface stroma to increase efficacy of chemo- and
and may preserve the replicative life span of polypeptide for selection, in vivo tracking, and radiotherapy. Clin Transl Oncol 2009;11:75–81.
memory CD8+ T cells in vitro. J Immunol ablation of engineered cells. Blood 295. Dvorak HF. Tumors: wounds that do not heal.
2005;174:4019–4024. 2011;118:1255–1263. Similarities between tumor stroma generation
262. Klebanoff CA, et al. IL-15 enhances the in vivo 279. Griffioen M, van Egmond EH, Kester MG, and wound healing. N Engl J Med
antitumor activity of tumor-reactive CD8+ T Willemze R, Falkenburg JH, Heemskerk MH. 1986;315:1650–1659.
cells. Proc Natl Acad Sci USA 2004;101:1969– Retroviral transfer of human CD20 as a suicide 296. Tanaka T, Bai Z, Srinoulprasert Y, Yang BG,
1974. gene for adoptive T-cell therapy. Haematologica Hayasaka H, Miyasaka M. Chemokines in tumor
263. Cieri N, et al. IL-7 and IL-15 instruct the 2009;94:1316–1320. progression and metastasis. Cancer Sci
generation of human memory stem T cells from 280. van der Bruggen P, et al. A gene encoding an 2005;96:317–322.
naive precursors. Blood 2013;121:573–584. antigen recognized by cytolytic T lymphocytes 297. Alexander S, Friedl P. Cancer invasion and
264. Hinrichs CS, et al. IL-2 and IL-21 confer on a human melanoma. Science 1991;254:1643– resistance: interconnected processes of disease
opposing differentiation programs to CD8+ T 1647. progression and therapy failure. Trends Mol Med
cells for adoptive immunotherapy. Blood 281. Kalos M. Tumor antigen-specific T cells and 2012;18:13–26.
2008;111:5326–5333. cancer immunotherapy: current issues and future 298. Zhang B, et al. Induced sensitization of tumor
265. Li Y, Bleakley M, Yee C. IL-21 influences the prospects. Vaccine 2003;21:781–786. stroma leads to eradication of established cancer
frequency, phenotype, and affinity of the 282. Li Y, et al. Directed evolution of human T-cell by T cells. J Exp Med 2007;204:49–55.
antigen-specific CD8 T cell response. J Immunol receptors with picomolar affinities by phage 299. Condeelis J, Pollard JW. Macrophages: obligate
2005;175:2261–2269. display. Nat Biotechnol 2005;23:349–354. partners for tumor cell migration, invasion, and
266. Singh H, et al. Reprogramming CD19-specific T 283. Riddell SR, Berger C, Murata M, Randolph S, metastasis. Cell 2006;124:263–266.
cells with IL-21 signaling can improve adoptive Warren EH. The graft versus leukemia response 300. Kuraishy A, Karin M, Grivennikov SI. Tumor
immunotherapy of B-lineage malignancies. after allogeneic hematopoietic stem cell promotion via injury- and death-induced
Cancer Res 2011;71:3516–3527. transplantation. Blood Rev 2003;17:153–162. inflammation. Immunity 2011;35:467–477.
267. Wilkie S, et al. Selective expansion of chimeric 284. Robbins PF, et al. A mutated beta-catenin gene 301. Friedl P, Alexander S. Cancer invasion and the
antigen receptor-targeted T-cells with potent encodes a melanoma-specific antigen recognized microenvironment: plasticity and reciprocity. Cell
effector function using interleukin-4. J Biol by tumor infiltrating lymphocytes. J Exp Med 2011;147:992–1009.
Chem 2010;285:25538–25544. 1996;183:1185–1192. 302. Ladoire S, et al. In situ immune response after
268. Rao RR, Li Q, Odunsi K, Shrikant PA. The mTOR 285. Terakura S, et al. A single minor neoadjuvant chemotherapy for breast cancer
kinase determines effector versus memory CD8+ histocompatibility antigen encoded by UGT2B17 predicts survival. J Pathol 2011;224:389–400.
T cell fate by regulating the expression of and presented by human leukocyte 303. Pages F, et al. Effector memory T cells, early
transcription factors T-bet and Eomesodermin. antigen-A*2902 and -B*4403. Transplantation metastasis, and survival in colorectal cancer. N
Immunity 2010;32:67–78. 2007;83:1242–1248. Engl J Med 2005;353:2654–2666.
269. O’Connor RS, et al. Substrate rigidity regulates 286. Wilkie S, et al. Dual targeting of ErbB2 and 304. Beyer M, Schultze JL. Regulatory T cells in
human T cell activation and proliferation. J MUC1 in breast cancer using chimeric antigen cancer. Blood 2006;108:804–811.
Immunol 2012;189:1330–1339. receptors engineered to provide complementary 305. Galon J, et al. Type, density, and location of
270. Rufer N, Migliaccio M, Antonchuk J, Humphries signaling. J Clin Immunol 2012;32:1059–1070. immune cells within human colorectal tumors
RK, Roosnek E, Lansdorp PM. Transfer of the 287. Mihara K, et al. Synergistic and persistent effect predict clinical outcome. Science
human telomerase reverse transcriptase (TERT) of T-cell immunotherapy with anti-CD19 or 2006;313:1960–1964.
gene into T lymphocytes results in extension of anti-CD38 chimeric receptor in conjunction with 306. Shimizu K, Nakata M, Hirami Y, Yukawa T,
replicative potential. Blood 2001;98:597–603. rituximab on B-cell non-Hodgkin lymphoma. Br Maeda A, Tanemoto K. Tumor-infiltrating
271. Eaton D, Gilham DE, O’Neill A, Hawkins RE. J Haematol 2010;151:37–46. Foxp3+ regulatory T cells are correlated with
Retroviral transduction of human peripheral 288. Cameron BJ, et al. Identification of a titin-derived cyclooxygenase-2 expression and are associated
blood lymphocytes with Bcl-X(L) promotes in HLA-A1-presented peptide as a cross-reactive with recurrence in resected non-small cell lung
vitro lymphocyte survival in pro-apoptotic target for engineered MAGE A3-directed T cells. cancer. J Thorac Oncol 2010;5:585–590.
conditions. Gene Ther 2002;9:527–535. Sci Transl Med 2013;5:197ra103. 307. Eljaszewicz A, et al. Collaborating with the
272. Dotti G, et al. Human cytotoxic T lymphocytes 289. Parente-Pereira AC, et al. Trafficking of enemy: function of macrophages in the
with reduced sensitivity to Fas-induced apoptosis. CAR-engineered human T cells following development of neoplastic disease. Mediators
Blood 2005;105:4677–4684. regional or systemic adoptive transfer in Inflamm 2013;2013:831387.
273. Bollard CM, et al. Adapting a transforming SCID beige mice. J Clin Immunol 308. Steidl C, et al. Tumor-associated macrophages
growth factor beta-related tumor protection 2011;31:710–718. and survival in classic Hodgkin’s lymphoma. N
strategy to enhance antitumor immunity. Blood 290. Canevari S, et al. Regression of advanced ovarian Engl J Med 2010;362:875–885.
2002;99:3179–3187. carcinoma by intraperitoneal treatment with 309. Dave SS, et al. Prediction of survival in follicular
274. Bonini C, et al. The suicide gene therapy autologous T lymphocytes retargeted by a lymphoma based on molecular features of
challenge: how to improve a successful gene bispecific monoclonal antibody. J Natl Cancer tumor-infiltrating immune cells. N Engl J Med
therapy approach. Mol Ther 2007;15:1248–1252. Inst 1995;87:1463–1469. 2004;351:2159–2169.

© 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
Immunological Reviews 257/2014 37
Ruella & Kalos  Adoptive immunotherapy for cancer

310. Ahmadzadeh M, et al. Tumor antigen-specific 327. Gattinoni L, et al. Removal of homeostatic cancer cells. Proc Natl Acad Sci USA
CD8 T cells infiltrating the tumor express high cytokine sinks by lymphodepletion enhances 2012;109:15877–15881.
levels of PD-1 and are functionally impaired. the efficacy of adoptively transferred 343. Miyazaki Y, et al. Development of a novel
Blood 2009;114:1537–1544. tumor-specific CD8+ T cells. J Exp Med redirected T-cell-based adoptive immunotherapy
311. Fridlender ZG, et al. Polarization of 2005;202:907–912. targeting human telomerase reverse transcriptase
tumor-associated neutrophil phenotype by 328. Paulos CM, et al. Microbial translocation for adult T-cell leukemia. Blood 2013;121:4894–
TGF-beta: “N1” versus “N2” TAN. Cancer Cell augments the function of adoptively transferred 4901.
2009;16:183–194. self/tumor-specific CD8+ T cells via TLR4 344. Tsuji T, et al. Generation of tumor-specific,
312. Biswas SK, Mantovani A. Macrophage plasticity signaling. J Clin Invest 2007;117:2197–2204. HLA class I-restricted human Th1 and Tc1 cells
and interaction with lymphocyte subsets: cancer 329. Brentjens RJ, et al. Safety and persistence of by cell engineering with tumor peptide-specific
as a paradigm. Nat Immunol 2010;11:889–896. adoptively transferred autologous CD19-targeted T-cell receptor genes. Blood 2005;106:470–
313. Gabrilovich DI, Nagaraj S. Myeloid-derived T cells in patients with relapsed or chemotherapy 476.
suppressor cells as regulators of the immune refractory B-cell leukemias. Blood 345. Chapuis AG, et al. Transferred WT1-reactive
system. Nat Rev Immunol 2009;9:162–174. 2011;118:4817–4828. CD8+ T cells can mediate antileukemic activity
314. Gabrilovich DI, Ostrand-Rosenberg S, Bronte V. 330. Grupp SA, et al. Adoptive transfer of autologous and persist in post-transplant patients. Sci Transl
Coordinated regulation of myeloid cells by T cells improves T cell repertoire diversity and Med 2013;5:174ra127.
tumours. Nat Rev Immunol 2012;12:253–268. long term B cell function in pediatric patients 346. Meij P, et al. Generation and administration
315. Allavena P, Sica A, Solinas G, Porta C, Mantovani with neuroblastoma. Clin Cancer Res of HA-1-specific T-cell lines for the treatment
A. The inflammatory micro-environment in 2012;18:6732–6741. of patients with relapsed leukemia after
tumor progression: the role of tumor-associated 331. Wang W, et al. Specificity redirection by CAR allogeneic stem cell transplantation: a pilot
macrophages. Crit Rev Oncol Hematol with human VEGFR-1 affinity endows T study. Haematologica 2012;97:1205–1208.
2008;66:1–9. lymphocytes with tumor-killing ability and 347. Kochenderfer JN, et al. B-cell depletion and
316. Solinas G, Germano G, Mantovani A, Allavena P. anti-angiogenic potency. Gene Ther remissions of malignancy along with
Tumor-associated macrophages (TAM) as major 2013;20:970–978. cytokine-associated toxicity in a clinical trial of
players of the cancer-related inflammation. J 332. Chinnasamy D, Tran E, Yu Z, Morgan RA, Restifo anti-CD19 chimeric-antigen-receptor-transduced
Leukoc Biol 2009;86:1065–1073. NP, Rosenberg SA. Simultaneous targeting of T cells. Blood 2012;119:2709–2720.
317. Hodi FS, et al. Improved survival with tumor antigens and the tumor vasculature using 348. Teachey DT, et al. Cytokine release syndrome
ipilimumab in patients with metastatic T lymphocyte transfer synergize to induce after blinatumomab treatment related to
melanoma. N Engl J Med 2010;363:711–723. regression of established tumors in mice. Cancer abnormal macrophage activation and ameliorated
318. Brahmer JR, et al. Safety and activity of Res 2013;73:3371–3380. with cytokine-directed therapy. Blood
anti-PD-L1 antibody in patients with advanced 333. Feig C, Gopinathan A, Neesse A, Chan DS, Cook 2013;121:5154–5157.
cancer. N Engl J Med 2012;366:2455–2465. N, Tuveson DA. The pancreas cancer 349. Walter EA, et al. Reconstitution of cellular
319. Kang MJ, et al. Tumor-infiltrating PD1-positive microenvironment. Clin Cancer Res immunity against cytomegalovirus in recipients
lymphocytes and FoxP3-positive regulatory T 2012;18:4266–4276. of allogeneic bone marrow by transfer of T-cell
cells predict distant metastatic relapse and 334. Tripathi M, Billet S, Bhowmick NA. clones from the donor. N Engl J Med
survival of clear cell renal cell carcinoma. Transl Understanding the role of stromal fibroblasts in 1995;333:1038–1044.
Oncol 2013;6:282–289. cancer progression. Cell Adh Migr 2012;6:231– 350. Ramsay AG, et al. Chronic lymphocytic leukemia
320. Muenst S, Soysal SD, Gao F, Obermann EC, Oertli 235. T cells show impaired immunological synapse
D, Gillanders WE. The presence of programmed 335. Kakarla S, et al. Antitumor effects of chimeric formation that can be reversed with an
death 1 (PD-1)-positive tumor-infiltrating receptor engineered human T cells directed to immunomodulating drug. J Clin Invest
lymphocytes is associated with poor prognosis in tumor stroma. Mol Ther 2013;21:1611–1620. 2008;118:2427–2437.
human breast cancer. Breast Cancer Res Treat 336. Spear P, Barber A, Sentman CL. Collaboration of 351. Nummer D, et al. Role of tumor endothelium in
2013;139:667–676. chimeric antigen receptor (CAR)-expressing T CD4+ CD25+ regulatory T cell infiltration of
321. June CH. Adoptive T cell therapy for cancer in cells and host T cells for optimal elimination of human pancreatic carcinoma. J Natl Cancer Inst
the clinic. J Clin Invest 2007;117:1466–1476. established ovarian tumors. Oncoimmunology 2007;99:1188–1199.
322. Curtin JF, et al. Treg depletion inhibits efficacy 2013;2:e23564. 352. Tang ZY, et al. Cytoreduction and sequential
of cancer immunotherapy: implications for 337. Morgan RA, et al. Cancer regression in patients resection: a hope for unresectable primary liver
clinical trials. PLoS ONE 2008;3:e1983. after transfer of genetically engineered cancer. J Surg Oncol 1991;47:27–31.
323. Muranski P, et al. Increased intensity lymphocytes. Science 2006;314:126–129. 353. Li KK, Li F, Li QS, Yang K, Jin B. DNA
lymphodepletion and adoptive immunotherapy– 338. Smith HA, McNeel DG. The SSX family of methylation as a target of epigenetic therapeutics
how far can we go? Nat Clin Pract Oncol cancer-testis antigens as target proteins for tumor in cancer. Anticancer Agents Med Chem
2006;3:668–681. therapy. Clin Dev Immunol 2010;2010:150591. 2013;13:242–247.
324. Lee JC, et al. In vivo inhibition of human 339. Imai N, Ikeda H, Shiku H. [Targeting cancer 354. Karlsson SC, et al. Combining CAR T cells and
CD19-targeted effector T cells by natural T antigen (MAGE-A4, NY-ESO -1) for the Bcl-2 family apoptosis inhibitor ABT-737 for
regulatory cells in a xenotransplant murine immunotherapy]. Nihon Rinsho 2012;70:2125– treating B-cell malignancy. Cancer Gene Ther
model of B cell malignancy. Cancer Res 2129. 2013;20:386–393.
2011;71:2871–2881. 340. Caballero OL, Chen YT. Cancer/testis (CT) 355. Sanchez C, et al. Combining T-cell
325. Klebanoff CA, Khong HT, Antony PA, Palmer antigens: potential targets for immunotherapy. immunotherapy and anti-androgen therapy for
DC, Restifo NP. Sinks, suppressors and antigen Cancer Sci 2009;100:2014–2021. prostate cancer. Prostate Cancer Prostatic Dis
presenters: how lymphodepletion enhances T 341. Morgan RA, et al. Cancer regression and 2013;16(123–131):S121.
cell-mediated tumor immunotherapy. Trends neurological toxicity following anti-MAGE-A3 356. Davids MS, Letai A. Targeting the B-cell
Immunol 2005;26:111–117. TCR gene therapy. J Immunother 2013;36:133– lymphoma/leukemia 2 family in cancer. J Clin
326. Wrzesinski C, Restifo NP. Less is more: 151. Oncol 2012;30:3127–3135.
lymphodepletion followed by hematopoietic 342. Hillerdal V, Nilsson B, Carlsson B, Eriksson F, 357. McGarrity GJ, et al. Patient monitoring and
stem cell transplant augments adoptive Essand M. T cells engineered with a T cell follow-up in lentiviral clinical trials. J Gene Med
T-cell-based anti-tumor immunotherapy. Curr receptor against the prostate antigen TARP 2013;15:78–82.
Opin Immunol 2005;17:195–201. specifically kill HLA-A2+ prostate and breast

© 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
38 Immunological Reviews 257/2014

You might also like