Miller 2015
Miller 2015
Cancer Cell
Perspective
Recent advances in cancer immunotherapy have directly built on 50 years of fundamental and technological
advances that made checkpoint blockade and T cell engineering possible. In this review, we intend to show
that research, not specifically designed to bring relief or cure to any particular disease, can, when creatively
exploited, lead to spectacular results in the management of cancer. The discovery of thymus immune func-
tion, T cells, and immune surveillance bore the seeds for today’s targeted immune interventions and chimeric
antigen receptors.
Nanos gigantum humeris insidentes. (Like dwarfs sitting notherapy were recognized by the Science magazine as the
on the shoulders of giants.) ‘‘breakthrough of the year’’ in 2013 (Couzin-Frankel, 2013) and
by the US Food and Drug Administration, which approved the
—Bernard de Chartres (1120)
anti-CTLA-4 monoclonal antibody Ipilimumab in 2011 and
granted breakthrough status to the CD19-specific chimeric anti-
If I have seen further than others, it is by standing on the
gen receptors (CARs) utilized at the University of Pennsylvania
shoulders of giants.
and Memorial Sloan Kettering Cancer Center for the treatment
—Isaac Newton (1676) of pediatric and adult acute lymphoblastic leukemia in 2014.
In this review, we aim to expose how basic discoveries in immu-
The immune system is the guardian of our organismal integrity nology led to these promising advances in cancer therapy.
in protecting us from infectious and other foreign invaders, such
as grafts and certain tumors. Immunology and oncology thus Leukemogenesis, the Thymus and Immunological
have a long relationship, in evolutionary terms as well as within Ontogeny
the biomedical sciences. The two fields have intersected again Prior to 1960, the thymus was thought to be a vestigial organ that
and again over the past half-century. We recount here some of had become redundant during evolution and was just a grave-
these events, with a deliberate focus on T lymphocytes—from yard for dying lymphocytes. Even though recirculating small
their discovery to their genetic engineering and use in cancer lymphocytes had been found by Gowans (Gowans et al., 1962)
immunotherapy. Insightful observations into leukemogenesis to be immunocompetent cells able to initiate either cellular or hu-
in mice were at the root of the discovery of thymopoiesis moral immune responses, thymus lymphocytes were deemed
and lymphocyte subsets. Subsequent observations led to the immunoincompetent since they did not recirculate, nor could
concept of immune surveillance, with eventual controversies they transfer immune responses to appropriate recipients.
on the role of immunity in tumor prevention and protection that Furthermore, thymectomy, which had always been performed
were reconciled in the cancer immunoediting hypothesis. As in adult animals, had no untoward effects on immune capacity.
progress was made in our fundamental understanding of antigen In 1959–1961, however, results obtained in a mouse model of
recognition, T cell activation, and T cell costimulation, transla- lymphocytic leukemia induced by the Gross leukemia virus led
tional researchers began to exploit the accumulating knowledge to experiments using neonatally thymectomized mice.
for cancer therapy. Tumor-infiltrating T cells were harnessed for To obtain a high incidence of leukemia in mouse strains that
adoptive cell therapy in a subset of melanoma patients; cancer were not highly prone to develop this malignancy, the virus had
vaccines were developed in an attempt to amplify endogenous to be given at birth, and not later (Gross, 1951). Thymectomy
tumor-specific T cell responses. Two of the most recent, most of virus-inoculated mice at 1 month of age prevented the disease
exciting therapeutic developments rest on the manipulation of (Miller, 1959a), and grafting a neonatal thymus as late as
costimulatory pathways governing T cell function. One, based 6 months after adult thymectomy restored the potential for
on monoclonal antibody technology, enables the release of tu- leukemia development (Miller, 1959b). Clearly, the virus must
mor-infiltrating T cells from inhibition mediated by costimulatory have remained latent, and indeed, it could be recovered from
receptors such as CTLA-4 and PD-1, through either checkpoint the non-leukemic tissues of thymectomized mice not grafted
blockade or depletion of regulatory T cells. The other, based on with thymus tissue (Miller, 1960).
gene transfer technology, enables to repurpose patient’s T cells, It was surmised that to induce a high percentage of leukemia in
targeting them to tumor antigens and augmenting their func- low-leukemic strains of mice, the virus had to be given neonatally
tional properties to overcome barriers erected by tumor cells because it needed to multiply in some cells present only in a
and their microenvironment. These two forms of cancer immu- newborn thymus. To test this hypothesis, the virus was given
Cancer Cell
Perspective
at birth but immediately after neonatal thymectomy, based on All of these results were initially regarded with some skepti-
the prediction that thymus grafting performed later would not cism. Vague criticisms abounded, such as the one stating that
restore leukemogenesis. The neonatally thymectomized mice mice must be unusual and that the results obtained would never
fared well until some weeks after weaning, when many became be seen in humans. What needed to be checked, however, was
sick, wasted, and died. This had never been seen by anyone who that because the mice used had been raised in converted horse
had thymectomized adult mice (Miller, 1961a). Post-mortem stables, the added trauma of neonatal thymectomy made them
examination revealed lesions in the liver suggestive of mouse highly susceptible to infections. This criticism was soon quashed
hepatitis virus infection and marked diminution of lymphocytes when it was shown that mice reared in a germfree facility, when
in the blood and in the lymphoid tissues (Miller, 1961b, 1962a). thymectomized at birth did not develop wasting disease and yet
As lymphocytes were known to be involved in graft rejection were unable to reject skin grafts, even those that differed at the
and other immune responses, the neonatally thymectomized H-2 locus (McIntire et al., 1964). In 1968, the nude mouse was
mice were tested for immunocompetence by grafting them discovered (Rygaard, 1973), and immunologists no longer had
with foreign skin before they had begun to show signs of wasting doubts about the immunological function of the thymus.
disease. Remarkably, they failed to reject foreign skin grafts, In children with thymus aplasia, as in di George syndrome,
even when donors and recipients differed at the major histo- thymus transplantation has reversed immunodeficiency (August
compatibility locus, H-2 (Miller, 1961b, 1962a). The neonatally et al., 1970). The problem lies in donor availability and tissue
thymectomized mice also lacked the ability to produce a normal compatibility. However, the recent spectacular success in
antibody response to certain antigens, such as Salmonella typhi creating a functional thymus organ by enforcing Foxn1 expres-
H antigen (Miller, 1962a) and sheep erythrocytes (Miller et al., sion to reprogram mouse embryonic fibroblasts into fetal thymic
1965). epithelium (Bredenkamp et al., 2014) constitutes a first prom-
Thymus grafting restored immune potential, and grafting of a ising step in the provision of appropriate thymus tissue.
foreign thymus induced specific tolerance to skin from the donor
of the thymus graft (Miller, 1962a). This suggested that the The Identification of T and B Cells in Mice
thymus may be the seat where tolerance is learned: ‘‘Antigenic As stated before, Gowans had clearly shown that recirculating
material might make contact with certain cell types differenti- small lymphocytes could respond both by a cellular immune
ating in the thymus and in some way prevent these cells from response (as in skin graft rejection) and by producing antibody
maturing to a stage when they would be capable of reacting (Gowans et al., 1962). He considered that the same cell could
immunologically’’ (Miller, 1962a). This clear prediction of nega- take part in either, depending on the antigenic stimulus. As
tive selection was proven some years later by using the so-called neonatally thymectomized mice were deficient in both cellular
super antigens and transgenic mice, and both positive and and at least some humoral responses, it was urgent to show
negative selection of thymocytes were then worked out (Bevan, that they had markedly reduced numbers of recirculating lym-
1977; Kappler et al., 1987). The major events in thymus cell dif- phocytes, not just blood lymphocytes. This was performed after
ferentiation have been summarized in a recent Timeline review cannulating the thoracic duct of neonatally thymectomized and
(Miller, 2011). ATxBM mice and collecting the lymph over a 24- to 48-hr period
It seemed important to determine whether the thymus exerted (Miller et al., 1967; Miller and Mitchell, 1967). The conclusion was
its influence by seeding cells into the rest of the lymphoid sys- made that most thoracic duct lymphocytes (TDLs) in mice were
tem. Since no cell-surface markers had at that time been found thymus derived, and this was actually proven in subsequent
to identify cells from different locations, use was made of the experiments (Miller and Sprent, 1971).
T6 strain of mice, the cells of which could be identified at meta- But were the same cells involved in antibody production and
phase by the presence of two minute chromosomes. Neonatally skin graft rejection? Two experimental systems suggested that
thymectomized F1 hybrid mice, in which one parent was T6, this might not be the case. Claman (Claman et al., 1966) showed
were grafted with thymus from the other parental strain and that irradiated mice given syngeneic bone marrow cells and
immunized with skin from various donors. An analysis of the syngeneic thymus cells could produce more antibody than
chromosome constitution of the cells in metaphase in the when given either cell source alone. As no antibody markers
spleen showed that 15% to 20% had originated from the thymus were available at the time, the origin of the antibody-forming cells
graft (Miller, 1962a). This suggested that the thymus did produce could not be easily identified. The situation was different for
cells capable of migrating to the periphery and that presumably chickens in Burnet’s laboratory, where impairment of bursa func-
such recent thymus emigrants would have just matured before tion by testosterone injection caused the birds not to produce
leaving or would mature in the peripheral lymphocyte pool to antibody, whereas thymus atrophy in sick birds prevented graft
become fully competent lymphocytes. rejection (Warner et al., 1962). Because mammals do not have
Although adult thymectomy had no effect on immunological a bursa, Burnet (Burnet, 1962) surmised, ‘‘In mammals it is highly
capacity, it could conceivably have caused a problem in mice probable that the thymus also carries out the function of the
whose lymphoid system had been destroyed in some way, as bursa of Fabricius in the chicken, which is to feed into the
for example by total-body irradiation. This proved to be the body the cells whose descendants will produce antibody.’’
case in partially irradiated mice (Miller, 1962c) and in lethally In 1967 and 1968, Miller and Mitchell (Miller and Mitchell, 1967,
irradiated mice protected with bone marrow (Cross et al., 1968, 1969), reconstituted neonatally thymectomized and adult
1964). The use of adult thymectomized, heavily irradiated mice thymectomized, irradiated CBA strain mice with CBA bone
protected with bone marrow (ATxBM mice) proved invaluable marrow and (CBAXC57BL)F1 TDLs and challenged them with
for further elucidation of immune functions. sheep erythrocytes. The ATxBM mice in those experiments
Cancer Cell
Perspective
Cancer Cell
Perspective
surface and that the specificity of the TCR was directed to both of antimicrobial bodies but is determined purely by cellular fac-
MHC molecules and virus-encoded products. The phenomenon tors. These may be weakened in older age groups where cancer
became known as MHC restriction and the MHC molecules is more prevalent.’’ Burnet echoed this view in his paper on tu-
involved as restriction elements. T cells in fact recognize mor immunosurveillance (Burnet, 1971).
relatively short peptide fragments (Townsend et al., 1986) that A large amount of work has been done by many to determine
become wedged in the jaws of the MHC molecules. How the importance of immunocompetent lymphocytes in guarding
T and B cells perceive antigen is highly relevant for tumor against the emergence of neoplastic clones of cells. There is
immunity and the escape of immunogenic tumor cells from any no doubt that a deficiency of such cells is associated with an
host immune response. Loss of cell-surface antigen or MHC increased incidence of those neoplasms that happen to be
molecules from genetically unstable tumor variants would of strongly immunogenic and to be produced by strong carcino-
course prejudice or prevent the response of any specific B and gens as cited above (Miller et al., 1963). However, extensive
T cells. CARs would later capitalize on the respective advan- studies performed using athymic nude mice failed to provide
tages of both targeting modalities—and Ig VH genes would any evidence of an increased susceptibility to the development
make their way into T cells, after all (see below). of spontaneous tumors (Rygaard and Povlsen, 1976). On the
Further T cell subsets and their function were soon identified: other hand, RAG-deficient mice (lacking T, B, and NKT cells)
CD4 and CD8 T cells (Kisielow et al., 1975; Cantor and Boyse, do spontaneously develop gastrointestinal epithelial malig-
1975), natural killer T (NKT) cells (Makino et al., 1995; Godfrey nancies by the age of 18 months (Shankaran et al., 2001);
et al., 2004), and CD4+CD25+FoxP3+ regulatory T (Treg) cells immunocompromised pathogen-free severe combined immu-
(Sakaguchi et al., 1985). Self-tolerance, as mentioned above, nodeficiency (SCID) mice have a high incidence of spontaneous
is mostly achieved intra-thymically by negative selection of thymic lymphomas, and about 2% of retired breeders developed
self-reactive thymocytes. Since, however, some self-reactive a variety of non-thymic tumors (Huang et al., 2011).
T cells (e.g., those with low affinity to self-antigens) escape to It is well established that immunosuppressed humans, e.g.,
the periphery, a peripheral tolerance mechanism exists, enabling following therapy for transplantation, have an increased inci-
Treg cells to prevent autoimmunity (reviewed in Josefowicz et al., dence of tumors such as skin or cervical carcinomas and lym-
2012). Treg cells are either produced in the thymus during T cell phomas (Penn and Starzl, 1973). A key question is whether this
development (natural Treg [nTreg] cells) or induced from naive is the result of failed immunosurveillance or of increased suscep-
CD4+ T cells in the periphery (so-called induced Treg [iTreg] tibility to infection by potentially oncogenic viruses (human papil-
cells) and are marked by expression of the transcription factor lomavirus, hepatitis C virus, human immunodeficiency virus, and
Foxp3 (Sakaguchi and Powrie, 2007). These cells soak up inter- Epstein-Barr virus [EBV]).
leukin-2 (IL-2) (Smith, 1989) through their high-affinity trimeric Many of the antigens expressed by spontaneous tumors are
IL-2 receptor, thus depriving other T cells of this growth factor, expected to be self-antigens, and hence thymus-derived lym-
but the exact mechanism by which, when recruited into the phocytes responding to these antigens would have been deleted
tumor environment, they prevent the activity of other immune in the thymus. Low-affinity cells specific for these self-antigens
cells capable of destroying the tumor is not clear. It may involve may, however, have escaped deletion and migrated to the pe-
the production of inhibitory cytokines such as transforming riphery, where they may be held in check by the growing tumors
growth factor (TGFb) (Powrie et al., 1996), IL-10 (Asseman as described below. Some spontaneous tumors have a high fre-
et al., 1999), and IL-35 (Collison et al., 2007). quency of mutations that may therefore generate de novo anti-
It is not the aim of this review to describe in further detail each gens (e.g., BCR-Abl, mutated forms of p53) that will not have
of the key advances that have led to our current knowledge of been present in the thymus and may thus appear foreign to the
the function of T and B cells, since this has been extensively immune system (Coulie et al., 2014; Gubin et al., 2014). In these
covered recently in a Timeline review (Miller, 2011). It is note- cases, therefore, both T cells and antibodies directed to tumor
worthy that several genes that govern T cell development or antigens should be able to eliminate the tumor. It is, however,
function, e.g., those encoding the IL receptor common g chain, not clear how likely it is that a point mutation in a single oncogene
the Wiskott-Aldrich syndrome protein, and others, are now being would give rise to an antigenic peptide. In some instances,
developed as genetic therapies for the treatment of severe pathogen-elicited thymus-derived cells that cross-react with
immunodeficiencies (Fischer et al., 2013) neoantigens acquired through somatic mutation may contribute
to anti-tumor responses (Snyder et al., 2014).
From Immune Surveillance to Cancer Vaccines The bulk of the observations on immune surveillance have in
and Adoptive T Cell Therapy fact shown that T cells and antibodies could recognize and elim-
The finding that neonatally thymectomized mice were more sus- inate tumors (Boon et al., 1994). How tumors spontaneously
ceptible than normal mice to the cancer-producing activities of generate tumor immunity has led to the cancer immunoediting
strong carcinogens (e.g., 3,4-benzopyrene) (Miller et al., 1963) hypothesis (Schreiber et al., 2011). This consists of three
and polyoma virus (Miller et al., 1964), (Miller, 1967) provided sequential phases: elimination, equilibrium, and escape. In the
some experimental support for Paul Ehrlich’s concept of immu- first phase, tumors developing before being clinically apparent
nological surveillance (Ehrlich and Himmelweit, 1957): ‘‘I am are destroyed and eliminated by both innate and adaptive im-
convinced that during development and growth malignant cells mune mechanisms (e.g., Shankaran et al., 2001). If variants arise
arise extremely frequently, but in the majority of people they and fail to be eliminated, cells of the adaptive immune system
remain latent due to the protective action of the host. I am also may restrain their growth during the lifetime of their host and
convinced that this natural immunity is not due to the presence a state of equilibrium is reached. Constant immune selective
Cancer Cell
Perspective
pressure of genetically unstable tumors may, however, lead to ligands CD80 (B7-1) and CD86 (B7-2) on the APC (Lafferty and
variants that can no longer be recognized by effector T cells Cunningham, 1975; Harding et al., 1992). Additional costimula-
because they have downregulated antigen or MHC, secreted tory receptors support T cell expansion and the maintenance
immunosuppressive cytokines such as TGFb or IL-10, or of T cell memory (Croft, 2003). Both activating and costimulatory
recruited Treg cells in their environment. They then enter the molecules would eventually be turned into powerful anti-cancer
escape phase and become clinically apparent. agents, via very different therapeutic strategies, one based
The administration of a cancer vaccine, such as GVAX (Dranoff on monoclonal antibody technology and the other on genetic
et al., 1993), has been used in the management of non- or weakly engineering.
immunogenic cancers. GVAX expresses tumor antigens in the
context of two important adjuvant activators, granulocyte- Reversing Tumor-Protective Immune Inhibition
macrophage colony-stimulating factor (GM-CSF) and pathogen Under normal circumstances, T cell responses are regulated
associated molecular patterns (PAMPs), that promote the acti- by inhibitory checkpoints, mediated in part by CTLA-4 (CD152),
vation of T cells with low affinity for self-antigens expressed by PD-1 (CD279), and BTLA (CD272), to prevent unrestrained multi-
tumors. This vaccine, together with the DNA-damage-inducing plication, collateral damage by cytotoxic effector molecules, and
chemotherapeutic drug cyclophosphamide that preferentially even autoimmunity.
targets Treg cells in a mixed cell population (Ghiringhelli et al., CTLA-4 competes with CD28 for the ligands CD80 and CD86
2007), with further assistance from the dual cocktail of anti- and acts as a signal dampener during the early stages of activa-
CTLA-4 plus anti-PD-1 (or anti-PD-L1) antibodies, discussed tion of naive and memory T cells, in fact 24–48 hr after antigen
below, may be successful in the treatment of certain tumors— presentation (Krummel and Allison, 2011). It has been shown
if potent, truly tumor-specific T cell responses are indeed eli- to act as an effector molecule that inhibits CD28 costimulation
cited. This goal may be harder to reach in tumors with a low by the cell-extrinsic depletion of its ligands (Qureshi et al.,
rate of mutations (Alexandrov et al., 2013). 2011). CTLA-4 is expressed on T cells, and its main physiological
The focus on T cell responses inspired yet another approach in function is to down-modulate CD4 T helper cell activity and
cancer immunotherapy, pioneered by Rosenberg and col- increase CD4+CD25+FoxP3+ Treg-cell-mediated immunosup-
leagues, focusing directly on the patient’s own tumor-infiltrating pression (reviewed by Pardoll, 2012). A deficiency or mutation
lymphocytes (TILs) when those exist and are accessible. In TIL in CTLA-4 in humans has contributed to the development of
therapy, T cells retrieved from surgical melanoma specimens autoimmune diseases including autoimmune hypothyroidism,
are cultured under activating conditions and reinfused with IL-2 type I diabetes, systemic lupus erythematosus, and celiac dis-
(Rosenberg et al., 1986) after host preconditioning (Gattinoni ease (reviewed by Watanabe and Nakajima, 2012). Recently, a
et al., 2006). This approach is, however, not applicable to all heterozygous nonsense mutation in exon 1 of CTLA4 has been
melanoma patients nor many other cancers. In EBV-associated identified in one of the commoner subgroups of common
lymphoproliferative disease, for which EBV-transformed B cells variable immunodeficiency syndromes (CVIDs) characterized
uniquely serve as effective APCs for tumor antigens of viral by hypogammaglobulinemia, recurrent infections, and multiple
origin, tumor-specific T cell lines have been successfully gener- autoimmune manifestations (Schubert et al., 2014).
ated from peripheral blood of healthy donors and used to treat PD-1 is expressed following T cell activation and during
immunocompromised recipients (Papadopoulos et al., 1994; chronic antigen-induced T cell stimulation, and it limits the activ-
Heslop et al., 1996). Artificial APC systems substituting for ity of T cells that have already been activated (Keir et al., 2008).
EBV-transformed B cells or dendritic cells (Kim et al., 2004) Unlike CTLA-4, PD-1 is expressed not only on all T cells subsets
have since been developed to facilitate the selection and/or but also on activated B cells and NK cells. It exerts its main func-
expansion of therapeutic T cells. tion during the inflammatory response associated with infection.
Chronic antigen exposure, as occurs in certain viral infections,
From T Cell Activation Biology to Checkpoint Blockade can induce exhaustion of cognate antigen-specific activated
and T Cell Engineering T cells. Furthermore, signals from inflammatory tissues induce
Most spontaneous tumors express self-antigens and present the expression of the PD-1 ligands, PD-L1 (B7-H1) or PD-L2
peptides derived from them embedded in MHC proteins on their (B7-DC), that downregulate T cell activity, thereby limiting collat-
cell surface. T cells with high-affinity TCRs for these would have eral damage and even autoimmunity. As with CTLA-4, mutations
been deleted by negative selection in the thymus. In melanomas, in PD-1 have also been associated with autoimmunity in humans
which are known to undergo a high mutation rate (Berger et al., (Watanabe and Nakajima, 2012).
2012), some mutated peptides are displayed as altered self- Theoretically, therefore, antagonists directed to these immune
antigens on the cell surface, where they can be recognized by inhibitory checkpoints should enable T cells to proliferate and
cytotoxic CD8 T cells. So why are these immunogenic tumors respond more vigorously and would be beneficial in controlling
not spontaneously killed by activated CD8 cells? Extensive virus infections and suppressing tumor growth (Figure 2). Ago-
studies of the molecules and processes involved in T cell acti- nists of CTLA-4 or PD-1, on the other hand, may prevent unde-
vation have shed light on this conundrum and yielded levers to sirable immune responses as occur in autoimmunity, allergy,
enable or restore T cell function. and transplant rejection (Okazaki and Honjo, 2007). In many
T cell activation initially requires two signals, one from the cases, cancer cells express the ligand PD-L1 on their surface,
recognition of antigen-derived peptides displayed in the context which may restrict tumor-reactive T cell responses by engaging
of MHC molecules on the surface of APCs and one from the the inhibitory receptor PD-1. Furthermore, PD-L1 on tumor cells
interaction of the T cell costimulatory molecule CD28 with its and the surrounding stromal cells is upregulated by g-interferon
Cancer Cell
Perspective
Cancer Cell
Perspective
Figure 3. Principles of T Cell Engineering
and CAR Design
(A) Integration of B cell and T cell antigen recog-
nition principles in the design of CARs. The heavy
and light chain chains, which are components
of the B cell receptor and Igs, are fused to the
T-cell-activating z chain of the TCR-associated
CD3 complex to generate non-MHC restricted,
activating receptors capable of redirecting T cell
antigen recognition and cytotoxicity.
(B and C) Integration of T cell activation and
costimulation principles in dual signaling CARs
designed to enhance T cell function and persis-
tence in addition to retargeting T cell specificity.
In (B), the physiological abTCR associated with
the CD3 signaling complex is flanked by the CD28
costimulatory receptor. (C) shows a prototypic
second-generation CAR, which comprises three
canonical components: an scFv for antigen
recognition, the cytoplasmic domain of the CD3z
chain for T cell activation, and a costimulatory
domain to enhance T cell function and persistence
(here CD28, as described in Maher et al. [2002],
and currently utilized in ALL patients as reported in
Brentjens et al. [2013] and Davila et al. [2014]).
Unlike the abTCR/CD3 complex, which comprises
g, d, ε, and z signaling chains and is modulated
by a multitude of costimulatory receptors, CARs
possess in a single molecule the ability to trigger
and modulate antigen-specific T cell functions.
Cancer Cell
Perspective
2012) to further enhance T cell potency, specificity, or safety. (2012). Melanoma genome sequencing reveals frequent PREX2 mutations.
Nature 485, 502–506.
Unlike CARs, CCRs do not trigger T cell activation, but rather
offset apoptosis and extend T cell persistence in antigen-spe- Bevan, M.J. (1977). In a radiation chimaera, host H-2 antigens determine
cific fashion (Krause et al., 1998). Antigen-specific inhibitory immune responsiveness of donor cytotoxic cells. Nature 269, 417–418.
receptors, such as iCARs modeled on CTLA-4 and PD-1 (Fe- Boon, T., Van den Eynde, B., Hirsch, H., Moroni, C., De Plaen, E., van der Brug-
dorov et al., 2013), further extend this growing panoply of recom- gen, P., De Smet, C., Lurquin, C., Szikora, J.P., and De Backer, O. (1994).
binant receptors. Genes coding for tumor-specific rejection antigens. Cold Spring Harb.
Symp. Quant. Biol. 59, 617–622.
In a futuristic embodiment, bringing together thymopoiesis and
cell engineering (Zhao et al., 2007; Themeli et al., 2013), the ability Bredenkamp, N., Ulyanchenko, S., O’Neill, K.E., Manley, N.R., Vaidya, H.J.,
and Blackburn, C.C. (2014). An organized and functional thymus generated
to recapitulate thymic maturation in vitro and generate T cells from FOXN1-reprogrammed fibroblasts. Nat. Cell Biol. 16, 902–908.
from pluripotent stem cells (Nishimura et al., 2013; Vizcardo
et al., 2013; Themeli et al., 2013) may open a path to design Brentjens, R.J., Latouche, J.B., Santos, E., Marti, F., Gong, M.C., Lyddane, C.,
King, P.D., Larson, S., Weiss, M., Rivière, I., and Sadelain, M. (2003). Eradica-
and manufacture therapeutic T cells with broad histocompatibil- tion of systemic B-cell tumors by genetically targeted human T lymphocytes
ity and optimized functional features (Themeli et al., 2015). co-stimulated by CD80 and interleukin-15. Nat. Med. 9, 279–286.
Brentjens, R.J., Davila, M.L., Riviere, I., Park, J., Wang, X., Cowell, L.G.,
Perspective Bartido, S., Stefanski, J., Taylor, C., Olszewska, M., et al. (2013). CD19-
targeted T cells rapidly induce molecular remissions in adults with chemo-
The spectacular successes that have been achieved in the im- therapy-refractory acute lymphoblastic leukemia. Sci. Transl. Med. 5, 77ra38.
mune management of various clinical conditions and especially
cancer were borne out of basic research that was creatively Brocker, T. (2000). Chimeric Fv-zeta or Fv-epsilon receptors are not sufficient
to induce activation or cytokine production in peripheral T cells. Blood 96,
exploited by translational researchers. No one could have pre- 1999–2001.
dicted that investigating how or why virus-induced lymphocytic
leukemia needs to develop in the neonatal mouse thymus would Brocker, T., Peter, A., Traunecker, A., and Karjalainen, K. (1993). New simpli-
fied molecular design for functional T cell receptor. Eur. J. Immunol. 23, 1435–
reveal the latter’s immunological function. The extensive world- 1439.
wide research that followed was crucial to our understanding
Bunnell, B.A., Muul, L.M., Donahue, R.E., Blaese, R.M., and Morgan, R.A.
of what cells and molecules regulate T cell activation and how (1995). High-efficiency retroviral-mediated gene transfer into human and
this may be used to our benefit in the clinic. The advent of nonhuman primate peripheral blood lymphocytes. Proc. Natl. Acad. Sci.
monoclonal antibody and gene transfer technologies later USA 92, 7739–7743.
enabled creative exploitation of fundamental knowledge on Burnet, F.M. (1962). The thymus gland. Sci. Am. 207, 50–57.
T and B cell antigen recognition, T cell activation, and T cell
costimulation, leading to the invention of checkpoint blockade Burnet, F.M. (1971). Immunological surveillance in neoplasia. Transplant. Rev.
7, 3–25.
and CAR T cell therapy. While some key molecular pathways
and cellular interactions have been decoded, much more is yet Cantor, H., and Boyse, E.A. (1975). Functional subclasses of T-lymphocytes
bearing different Ly antigens. I. The generation of functionally distinct T-cell
to be discovered. More immunology research is warranted. In subclasses is a differentiative process independent of antigen. J. Exp. Med.
the meantime, immunology has spawned immunotherapy, which 141, 1376–1389.
is about to claim a seat in the therapeutic pantheon of oncology,
Chen, Y.T., Scanlan, M.J., Sahin, U., Türeci, O., Gure, A.O., Tsang, S., William-
next to surgery, radiation therapy, and chemotherapy. son, B., Stockert, E., Pfreundschuh, M., and Old, L.J. (1997). A testicular anti-
gen aberrantly expressed in human cancers detected by autologous antibody
screening. Proc. Natl. Acad. Sci. USA 94, 1914–1918.
ACKNOWLEDGMENTS
Chinnasamy, D., Yu, Z., Kerkar, S.P., Zhang, L., Morgan, R.A., Restifo, N.P.,
We thank Drs. Tony Basten, Marc Feldmann, Dale Godfrey, Robyn Slattery, and Rosenberg, S.A. (2012). Local delivery of interleukin-12 using T cells
and Andreas Strasser for reading the manuscript and for helpful suggestions. targeting VEGF receptor-2 eradicates multiple vascularized tumors in mice.
Clin. Cancer Res. 18, 1672–1683.
REFERENCES Chmielewski, M., and Abken, H. (2012). CAR T cells transform to trucks:
chimeric antigen receptor-redirected T cells engineered to deliver inducible
Abu-Eid, R., Samara, R.N., Ozbun, L., Abdalla, M.Y., Berzofsky, J.A., Fried- IL-12 modulate the tumour stroma to combat cancer. Cancer Immunol.
man, K.M., Mkrtichyan, M., and Khleif, S.N. (2014). Selective inhibition of reg- Immunother. 61, 1269–1277.
ulatory T cells by targeting the PI3K-Akt pathway. Cancer Immunol Res 2,
1080–1089. Claman, H.N., Chaperon, E.A., and Triplett, R.F. (1966). Thymus-marrow cell
combinations. Synergism in antibody production. Proceedings of the Society
Alexandrov, L.B., Nik-Zainal, S., Wedge, D.C., Aparicio, S.A., Behjati, S., for Experimental Biology and Medicine Society for Experimental Biology and
Biankin, A.V., Bignell, G.R., Bolli, N., Borg, A., Børresen-Dale, A.L., et al.; Medicine 122, 1167–1171.
Australian Pancreatic Cancer Genome Initiative; ICGC Breast Cancer Con-
sortium; ICGC MMML-Seq Consortium; ICGC PedBrain (2013). Signatures Collison, L.W., Workman, C.J., Kuo, T.T., Boyd, K., Wang, Y., Vignali, K.M.,
of mutational processes in human cancer. Nature 500, 415–421. Cross, R., Sehy, D., Blumberg, R.S., and Vignali, D.A.A. (2007). The inhibitory
cytokine IL-35 contributes to regulatory T-cell function. Nature 450, 566–569.
Asseman, C., Mauze, S., Leach, M.W., Coffman, R.L., and Powrie, F. (1999).
An essential role for interleukin 10 in the function of regulatory T cells that Coulie, P.G., Van den Eynde, B.J., van der Bruggen, P., and Boon, T. (2014).
inhibit intestinal inflammation. J. Exp. Med. 190, 995–1004. Tumour antigens recognized by T lymphocytes: at the core of cancer immuno-
therapy. Nat. Rev. Cancer 14, 135–146.
August, C.S., Berkel, A.I., Levey, R.H., Rosen, F.S., and Kay, H.E. (1970).
Establishment of immunological competence in a child with congenital thymic Couzin-Frankel, J. (2013). Breakthrough of the year 2013. Cancer immuno-
aplasia by a graft of fetal thymus. Lancet 1, 1080–1083. therapy. Science 342, 1432–1433.
Berger, M.F., Hodis, E., Heffernan, T.P., Deribe, Y.L., Lawrence, M.S., Croft, M. (2003). Co-stimulatory members of the TNFR family: keys to effective
Protopopov, A., Ivanova, E., Watson, I.R., Nickerson, E., Ghosh, P., et al. T-cell immunity? Nat. Rev. Immunol. 3, 609–620.
Cancer Cell
Perspective
Cross, A.M., Leuchars, E., and Miller, J.F. (1964). Studies on the recovery of lated polypeptide chain of the human T-cell receptor/T3 complex. Nature
the immune response in irradiated mice thymectomized in adult life. J. Exp. 321, 431–434.
Med. 119, 837–850.
Gong, M.C., Latouche, J.B., Krause, A., Heston, W.D., Bander, N.H., and
Curiel, T.J. (2007). Tregs and rethinking cancer immunotherapy. J. Clin. Invest. Sadelain, M. (1999). Cancer patient T cells genetically targeted to prostate-
117, 1167–1174. specific membrane antigen specifically lyse prostate cancer cells and release
cytokines in response to prostate-specific membrane antigen. Neoplasia 1,
Curran, M.A., Montalvo, W., Yagita, H., and Allison, J.P. (2010). PD-1 and 123–127.
CTLA-4 combination blockade expands infiltrating T cells and reduces regula-
tory T and myeloid cells within B16 melanoma tumors. Proc. Natl. Acad. Sci. Good, R.A. (1969). Discussion after Miller, J.F.A.P. In Immunological Toler-
USA 107, 4275–4280. ance: A Reassessment of Mechanisms of the Immune Response, M. Landy
and W. Braun, eds. (New York: Academic Press), p. 136.
Davila, M.L., Riviere, I., Wang, X., Bartido, S., Park, J., Curran, K., Chung, S.S.,
Stefanski, J., Borquez-Ojeda, O., Olszewska, M., et al. (2014). Efficacy and Gowans, J.L. (1969). Discussion after Miller, J.F.A.P. In Immunological Toler-
toxicity management of 19-28z CAR T cell therapy in B cell acute lympho- ance: A Reassessment of Mechanisms of the Immune Response, M. Landy
blastic leukemia. Sci. Transl. Med. 6, 24ra25. and W. Braun, eds. (New York: Academic Press), pp. 164–169.
Delgoffe, G.M., Woo, S.-R., Turnis, M.E., Gravano, D.M., Guy, C., Overacre, Gowans, J.L., McGREGOR, D.D., and Cowen, D.M. (1962). Initiation of
A.E., Bettini, M.L., Vogel, P., Finkelstein, D., Bonnevier, J., et al. (2013). immune responses by small lymphocytes. Nature 196, 651–655.
Stability and function of regulatory T cells is maintained by a neuropilin-1-
semaphorin-4a axis. Nature 501, 252–256. Gross, L. (1951). Pathogenic properties, and ‘‘vertical’’ transmission of the
mouse leukemia agent. Proceedings of the Society for Experimental Biology
Dembic , Z., Haas, W., Weiss, S., McCubrey, J., Kiefer, H., von Boehmer, H., and Medicine Society for Experimental Biology and Medicine 78, 342–348.
and Steinmetz, M. (1986). Transfer of specificity by murine alpha and beta
T-cell receptor genes. Nature 320, 232–238. Grupp, S.A., Kalos, M., Barrett, D., Aplenc, R., Porter, D.L., Rheingold, S.R.,
Teachey, D.T., Chew, A., Hauck, B., Wright, J.F., et al. (2013). Chimeric antigen
Dranoff, G., Jaffee, E., Lazenby, A., Golumbek, P., Levitsky, H., Brose, K., receptor-modified T cells for acute lymphoid leukemia. N. Engl. J. Med. 368,
Jackson, V., Hamada, H., Pardoll, D., and Mulligan, R.C. (1993). Vaccination 1509–1518.
with irradiated tumor cells engineered to secrete murine granulocyte-macro-
phage colony-stimulating factor stimulates potent, specific, and long-lasting Gubin, M.M., Zhang, X., Schuster, H., Caron, E., Ward, J.P., Noguchi, T., Iva-
anti-tumor immunity. Proc. Natl. Acad. Sci. USA 90, 3539–3543. nova, Y., Hundal, J., Arthur, C.D., Krebber, W.-J., et al. (2014). Checkpoint
blockade cancer immunotherapy targets tumour-specific mutant antigens.
Duong, C.P., Westwood, J.A., Berry, L.J., Darcy, P.K., and Kershaw, M.H. Nature 515, 577–581.
(2011). Enhancing the specificity of T-cell cultures for adoptive immunotherapy
of cancer. Immunotherapy 3, 33–48. Harding, F.A., McArthur, J.G., Gross, J.A., Raulet, D.H., and Allison, J.P.
(1992). CD28-mediated signalling co-stimulates murine T cells and prevents
Ehrlich, P., and Himmelweit, F. (1957). The Collected Papers of Paul Ehrlich. induction of anergy in T-cell clones. Nature 356, 607–609.
(Oxford: Pergamon Press).
Hedrick, S.M., Cohen, D.I., Nielsen, E.A., and Davis, M.M. (1984). Isolation of
Eshhar, Z., Waks, T., Gross, G., and Schindler, D.G. (1993). Specific activation cDNA clones encoding T cell-specific membrane-associated proteins. Nature
and targeting of cytotoxic lymphocytes through chimeric single chains con- 308, 149–153.
sisting of antibody-binding domains and the gamma or zeta subunits of the
immunoglobulin and T-cell receptors. Proc. Natl. Acad. Sci. USA 90, 720–724. Heslop, H.E., Ng, C.Y., Li, C., Smith, C.A., Loftin, S.K., Krance, R.A., Brenner,
M.K., and Rooney, C.M. (1996). Long-term restoration of immunity against
Eshhar, Z., Bach, N., Fitzer-Attas, C.J., Gross, G., Lustgarten, J., Waks, T., and Epstein-Barr virus infection by adoptive transfer of gene-modified virus-spe-
Schindler, D.G. (1996). The T-body approach: potential for cancer immuno- cific T lymphocytes. Nat. Med. 2, 551–555.
therapy. Springer Semin. Immunopathol. 18, 199–209.
Ho, W.Y., Blattman, J.N., Dossett, M.L., Yee, C., and Greenberg, P.D. (2003).
European Society for Medical Oncology (2014). Immune checkpoint inhibitors Adoptive immunotherapy: engineering T cell responses as biologic weapons
provide antitumour activity across malignant diseases. http://www.esmo.org/ for tumor mass destruction. Cancer Cell 3, 431–437.
Conferences/Past-Conferences/ESMO-2014-Congress.
Hombach, A., Wieczarkowiecz, A., Marquardt, T., Heuser, C., Usai, L., Pohl,
Fedorov, V.D., Themeli, M., and Sadelain, M. (2013). PD-1- and CTLA-4-based C., Seliger, B., and Abken, H. (2001). Tumor-specific T cell activation by re-
inhibitory chimeric antigen receptors (iCARs) divert off-target immunotherapy combinant immunoreceptors: CD3 zeta signaling and CD28 costimulation
responses. Sci. Transl. Med. 5, ra172. are simultaneously required for efficient IL-2 secretion and can be integrated
into one combined CD28/CD3 zeta signaling receptor molecule. J. Immunol.
Finney, H.M., Lawson, A.D., Bebbington, C.R., and Weir, A.N. (1998). Chimeric 167, 6123–6131.
receptors providing both primary and costimulatory signaling in T cells from a
single gene product. J. Immunol. 161, 2791–2797. Hoyos, V., Savoldo, B., Quintarelli, C., Mahendravada, A., Zhang, M., Vera, J.,
Heslop, H.E., Rooney, C.M., Brenner, M.K., and Dotti, G. (2010). Engineering
Fischer, A., Hacein-Bey-Abina, S., and Cavazzana-Calvo, M. (2013). Gene CD19-specific T lymphocytes with interleukin-15 and a suicide gene to
therapy of primary T cell immunodeficiencies. Gene 525, 170–173. enhance their anti-lymphoma/leukemia effects and safety. Leukemia 24,
1160–1170.
Gallardo, H.F., Tan, C., Ory, D., and Sadelain, M. (1997). Recombinant retrovi-
ruses pseudotyped with the vesicular stomatitis virus G glycoprotein mediate Huang, P., Westmoreland, S.V., Jain, R.K., and Fukumura, D. (2011). Sponta-
both stable gene transfer and pseudotransduction in human peripheral blood neous nonthymic tumors in SCID mice. Comp. Med. 61, 227–234.
lymphocytes. Blood 90, 952–957.
Hudson, L., Sprent, J., Miller, J.F., and Playfair, J.H.L. (1974). B cell-derived
Gattinoni, L., Powell, D.J., Jr., Rosenberg, S.A., and Restifo, N.P. (2006). immunoglobulin on activated mouse T lymphocytes. Nature 251, 60–62.
Adoptive immunotherapy for cancer: building on success. Nat. Rev. Immunol.
6, 383–393. Imai, C., Mihara, K., Andreansky, M., Nicholson, I.C., Pui, C.H., Geiger, T.L.,
and Campana, D. (2004). Chimeric receptors with 4-1BB signaling capacity
Ghiringhelli, F., Menard, C., Puig, P.E., Ladoire, S., Roux, S., Martin, F., Solary, provoke potent cytotoxicity against acute lymphoblastic leukemia. Leukemia
E., Le Cesne, A., Zitvogel, L., and Chauffert, B. (2007). Metronomic cyclophos- 18, 676–684.
phamide regimen selectively depletes CD4+CD25+ regulatory T cells and
restores T and NK effector functions in end stage cancer patients. Cancer Irving, B.A., and Weiss, A. (1991). The cytoplasmic domain of the T cell
Immunol. Immunother. 56, 641–648. receptor zeta chain is sufficient to couple to receptor-associated signal
transduction pathways. Cell 64, 891–901.
Godfrey, D.I., MacDonald, R., Kronenberg, M., Smyth, M.J., and van Kaer, L.
(2004). NKT cells: what’s in a name? Nat. Rev. Immunol. 4, 231–237. Iwai, Y., Ishida, M., Tanaka, Y., Okazaki, T., Honjo, T., and Minato, N. (2002).
Involvement of PD-L1 on tumor cells in the escape from host immune system
Gold, D.P., Puck, J.M., Pettey, C.L., Cho, M., Coligan, J., Woody, J.N., and and tumor immunotherapy by PD-L1 blockade. Proc. Natl. Acad. Sci. USA 99,
Terhorst, C. (1986). Isolation of cDNA clones encoding the 20K non-glycosy- 12293–12297.
Cancer Cell
Perspective
Jensen, M.C., and Riddell, S.R. (2015). Designing chimeric antigen receptors Mavilio, F., Ferrari, G., Rossini, S., Nobili, N., Bonini, C., Casorati, G., Traver-
to effectively and safely target tumors. Curr. Opin. Immunol. 33C, 9–15. sari, C., and Bordignon, C. (1994). Peripheral blood lymphocytes as target cells
of retroviral vector-mediated gene transfer. Blood 83, 1988–1997.
Josefowicz, S.Z., Lu, L.-F., and Rudensky, A.Y. (2012). Regulatory T cells:
mechanisms of differentiation and function. Annu. Rev. Immunol. 30, 531–564. McIntire, K.R., Sell, S., and Miller, J.F. (1964). Pathogenesis of the post-
neonatal thymectomy wasting syndrome. Nature 204, 151–155.
Kappler, J.W., Roehm, N., and Marrack, P. (1987). T cell tolerance by clonal
elimination in the thymus. Cell 49, 273–280. Miller, J.F. (1959a). Role of the thymus in murine leukaemia. Nature 183, 1069.
Keir, M.E., Butte, M.J., Freeman, G.J., and Sharpe, A.H. (2008). PD-1 and its Miller, J.F. (1959b). Fate of subcutaneous thymus grafts in thymectomized
ligands in tolerance and immunity. Annu. Rev. Immunol. 26, 677–704. mice inoculated with leukaemic filtrate. Nature 184 (23), 1809–1810.
Köhler, G., and Milstein, C. (1975). Continuous cultures of fused cells secreting Miller, J.F., and Mitchell, G.F. (1967). The thymus and the precursors of anti-
antibody of predefined specificity. Nature 256, 495–497. gen-reactive cells. Nature 216, 659–663.
Miller, J.F., and Mitchell, G.F. (1968). Cell to cell interaction in the immune
Krause, A., Guo, H.F., Latouche, J.B., Tan, C., Cheung, N.K., and Sadelain, M.
response. I. Hemolysin-forming cells in neonatally thymectomized mice recon-
(1998). Antigen-dependent CD28 signaling selectively enhances survival and
stituted with thymus or thoracic duct lymphocytes. J. Exp. Med. 128, 801–820.
proliferation in genetically modified activated human primary T lymphocytes.
J. Exp. Med. 188, 619–626. Miller, J.F., and Mitchell, G.F. (1969). Thymus and antigen-reactive cells.
Transplant. Rev. 1, 3–42.
Krummel, M.F., and Allison, J.P. (2011). Pillars article: CD28 and CTLA-4 have
opposing effects on the response of T cells to stimulation. The journal of Miller, J.F., and Sprent, J. (1971). Thymus-derived cells in mouse thoracic duct
experimental medicine. 1995. 182: 459-465. J. Immunol. 187, 3459–3465. lymph. Nat. New Biol. 230, 267–270.
Lafferty, K.J., and Cunningham, A.J. (1975). A new analysis of allogeneic Miller, J.F., Grant, G.A., and Roe, F.J. (1963). Effect of thymectomy on the
interactions. Aust. J. Exp. Biol. Med. Sci. 53, 27–42. induction of skin tumours by 3,4-benzopyrene. Nature 199, 920–922.
Leach, D.R., Krummel, M.F., and Allison, J.P. (1996). Enhancement of anti- Miller, J.F., Law, L.W., and Ting, R.C. (1964). Influence of thymectomy on tu-
tumor immunity by CTLA-4 blockade. Science 271, 1734–1736. mor induction by polyoma virus in C57BL mice. Proceedings of the Society
for Experimental Biology and Medicine Society for Experimental Biology and
Lee, D.W., Kochenderfer, J.N., Stetler-Stevenson, M., Cui, Y.K., Delbrook, C., Medicine 116, 323–327.
Feldman, S.A., Fry, T.J., Orentas, R., Sabatino, M., Shah, N.N., et al. (2015).
T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic Miller, J.F., De Burgh, P.M., and Grant, G.A. (1965). Thymus and the produc-
leukaemia in children and young adults: a phase 1 dose-escalation trial. Lancet tion of antibody-plaque-forming cells. Nature 208, 1332–1334.
385, 517–528.
Miller, J.F., Mitchell, G.F., and Weiss, N.S. (1967). Cellular basis of the immu-
Letourneur, F., and Klausner, R.D. (1991). T-cell and basophil activation nological defects in thymectomized mice. Nature 214, 992–997.
through the cytoplasmic tail of T-cell-receptor zeta family proteins. Proc.
Natl. Acad. Sci. USA 88, 8905–8909. Mitchell, G.F., and Miller, J.F. (1968). Cell to cell interaction in the immune
response. II. The source of hemolysin-forming cells in irradiated mice given
Maher, J., Brentjens, R.J., Gunset, G., Rivière, I., and Sadelain, M. (2002). bone marrow and thymus or thoracic duct lymphocytes. J. Exp. Med. 128,
Human T-lymphocyte cytotoxicity and proliferation directed by a single 821–837.
chimeric TCRzeta /CD28 receptor. Nat. Biotechnol. 20, 70–75.
Nishimura, T., Kaneko, S., Kawana-Tachikawa, A., Tajima, Y., Goto, H., Zhu,
D., Nakayama-Hosoya, K., Iriguchi, S., Uemura, Y., Shimizu, T., et al. (2013).
Makino, Y., Kanno, R., Ito, T., Higashino, K., and Taniguchi, M. (1995).
Generation of rejuvenated antigen-specific T cells by reprogramming to plurip-
Predominant expression of invariant V a 14+ TCR a chain in NK1.1+ T cell
otency and redifferentiation. Cell Stem Cell 12, 114–126.
populations. Int. Immunol. 7, 1157–1161.
Oettgen, H.C., Terhorst, C., Cantley, L.C., and Rosoff, P.M. (1985). Stimulation
Marchalonis, J.J., Cone, R.E., and Atwell, J.L. (1972). Isolation and partial of the T3-T cell receptor complex induces a membrane-potential-sensitive cal-
characterization of lymphocyte surface immunoglobulins. J. Exp. Med. 135, cium influx. Cell 40, 583–590.
956–971.
Okazaki, T., and Honjo, T. (2007). PD-1 and PD-1 ligands: from discovery to
Markley, J.C., and Sadelain, M. (2010). IL-7 and IL-21 are superior to IL-2 and clinical application. Int. Immunol. 19, 813–824.
IL-15 in promoting human T cell-mediated rejection of systemic lymphoma in
immunodeficient mice. Blood 115, 3508–3519. Ostrand-Rosenberg, S. (2010). Myeloid-derived suppressor cells: more mech-
anisms for inhibiting antitumor immunity. Cancer Immunol. Immunother. 59,
Maude, S.L., Frey, N., Shaw, P.A., Aplenc, R., Barrett, D.M., Bunin, N.J., Chew, 1593–1600.
A., Gonzalez, V.E., Zheng, Z., Lacey, S.F., et al. (2014). Chimeric antigen
receptor T cells for sustained remissions in leukemia. N. Engl. J. Med. 371, Papadopoulos, E.B., Ladanyi, M., Emanuel, D., Mackinnon, S., Boulad, F.,
1507–1517. Carabasi, M.H., Castro-Malaspina, H., Childs, B.H., Gillio, A.P., Small, T.N.,
Cancer Cell
Perspective
et al. (1994). Infusions of donor leukocytes to treat Epstein-Barr virus-associ- Shankaran, V., Ikeda, H., Bruce, A.T., White, J.M., Swanson, P.E., Old, L.J.,
ated lymphoproliferative disorders after allogeneic bone marrow transplanta- and Schreiber, R.D. (2001). IFNgamma and lymphocytes prevent primary
tion. N. Engl. J. Med. 330, 1185–1191. tumour development and shape tumour immunogenicity. Nature 410, 1107–
1111.
Pardoll, D.M. (2012). The blockade of immune checkpoints in cancer immuno-
therapy. Nat. Rev. Cancer 12, 252–264. Simpson, T.R., Li, F., Montalvo-Ortiz, W., Sepulveda, M.A., Bergerhoff, K.,
Arce, F., Roddie, C., Henry, J.Y., Yagita, H., Wolchok, J.D., et al. (2013).
Pegram, H.J., Lee, J.C., Hayman, E.G., Imperato, G.H., Tedder, T.F., Sadelain, Fc-dependent depletion of tumor-infiltrating regulatory T cells co-defines
M., and Brentjens, R.J. (2012). Tumor-targeted T cells modified to secrete the efficacy of anti-CTLA-4 therapy against melanoma. J. Exp. Med. 210,
IL-12 eradicate systemic tumors without need for prior conditioning. Blood 1695–1710.
119, 4133–4141.
Smith, K.A. (1989). The interleukin 2 receptor. Annu. Rev. Cell Biol. 5, 397–425.
Penn, I., and Starzl, T.E. (1973). Immunosuppression and cancer. Transplant.
Proc. 5, 943–947. Snyder, A., Makarov, V., Merghoub, T., Yuan, J., Zaretsky, J.M., Desrichard,
A., Walsh, L.A., Postow, M.A., Wong, P., Ho, T.S., et al. (2014). Genetic basis
Powrie, F., Carlino, L., Leach, M.W., Mauze, S., and Coffman, R.L. (1996). for clinical response to CTLA-4 blockade in melanoma. N. Engl. J. Med. 371,
A critical role for transforming growth factor-b but not intreleukin-4 in the 2189–2199.
suppression of T helper type 1-mediated colitis by CD45RB(low)CD4+ T cells.
J. Exp. Med. 183, 2669–2674. Steinman, R.M., and Cohn, Z.A. (1973). Identification of a novel cell type in
peripheral lymphoid organs of mice. I. Morphology, quantitation, tissue
Prosser, M.E., Brown, C.E., Shami, A.F., Forman, S.J., and Jensen, M.C. distribution. J. Exp. Med. 137, 1142–1162.
(2012). Tumor PD-L1 co-stimulates primary human CD8(+) cytotoxic T cells
modified to express a PD1:CD28 chimeric receptor. Mol. Immunol. 51, Stephan, M.T., Ponomarev, V., Brentjens, R.J., Chang, A.H., Dobrenkov, K.V.,
263–272. Heller, G., and Sadelain, M. (2007). T cell-encoded CD80 and 4-1BBL induce
auto- and transcostimulation, resulting in potent tumor rejection. Nat. Med. 13,
Qureshi, O.S., Zheng, Y., Nakamura, K., Attridge, K., Manzotti, C., Schmidt, 1440–1449.
E.M., Baker, J., Jeffery, L.E., Kaur, S., Briggs, Z., et al. (2011). Trans-endocy-
tosis of CD80 and CD86: a molecular basis for the cell-extrinsic function of Stone, J.D., and Kranz, D.M. (2013). Role of T cell receptor affinity in the
CTLA-4. Science 332, 600–603. efficacy and specificity of adoptive T cell therapies. Front. Immunol. 4, 244.
Ramos, C.A., Savoldo, B., and Dotti, G. (2014). CD19-CAR trials. Cancer J. 20, Sugiyama, D., Nishikawa, H., Maeda, Y., Nishioka, M., Tanemura, A.,
112–118. Katayama, I., Ezoe, S., Kanakura, Y., Sato, E., Fukumori, Y., et al. (2013).
Anti-CCR4 mAb selectively depletes effector-type FoxP3+CD4+ regulatory
Robbins, P.F., Morgan, R.A., Feldman, S.A., Yang, J.C., Sherry, R.M., Dudley, T cells, evoking antitumor immune responses in humans. Proc. Natl. Acad.
M.E., Wunderlich, J.R., Nahvi, A.V., Helman, L.J., Mackall, C.L., et al. (2011). Sci. USA 110, 17945–17950.
Tumor regression in patients with metastatic synovial cell sarcoma and mela-
Themeli, M., Kloss, C.C., Ciriello, G., Fedorov, V.D., Perna, F., Gonen, M., and
noma using genetically engineered lymphocytes reactive with NY-ESO-1.
Sadelain, M. (2013). Generation of tumor-targeted human T lymphocytes
J. Clin. Oncol. 29, 917–924.
from induced pluripotent stem cells for cancer therapy. Nat. Biotechnol. 31,
928–933.
Romeo, C., and Seed, B. (1991). Cellular immunity to HIV activated by CD4
fused to T cell or Fc receptor polypeptides. Cell 64, 1037–1046.
Themeli, M., Riviere, I., and Sadelain, M. (2015). New cell sources for T cell
engineering and adoptive immunotherapy. Cell Stem Cell Published online
Rosenberg, S.A., Spiess, P., and Lafreniere, R. (1986). A new approach to
April 2, 2015. http://dx.doi.org/10.1016/j.stem.2015.03.011.
the adoptive immunotherapy of cancer with tumor-infiltrating lymphocytes.
Science 233, 1318–1321. Townsend, A.R.M., Rothbard, J., Gotch, F.M., Bahadur, G., Wraith, D., and
McMichael, A.J. (1986). The epitopes of influenza nucleoprotein recognized
Rygaard, J. (1973). Thymus and Self: Immunobiology of the Mouse Mutant
by cytotoxic T lymphocytes can be defined with short synthetic peptides.
Nude. (København: F.A.D.L.).
Cell 44, 959–968.
Rygaard, J., and Povlsen, C.O. (1976). The nude mouse vs. the hypothesis of Vizcardo, R., Masuda, K., Yamada, D., Ikawa, T., Shimizu, K., Fujii, S., Koseki,
immunological surveillance. Transplant. Rev. 28, 43–61. H., and Kawamoto, H. (2013). Regeneration of human tumor antigen-specific
T cells from iPSCs derived from mature CD8(+) T cells. Cell Stem Cell 12, 31–36.
Sadelain, M., Rivière, I., and Brentjens, R. (2003). Targeting tumours with
genetically enhanced T lymphocytes. Nat. Rev. Cancer 3, 35–45. Warner, N.L., Szenberg, A., and Burnet, F.M. (1962). The immunological role of
different lymphoid organs in the chicken. I. Dissociation of immunological
Sadelain, M., Brentjens, R., and Rivière, I. (2009). The promise and potential responsiveness. Aust. J. Exp. Biol. Med. Sci. 40, 373–387.
pitfalls of chimeric antigen receptors. Curr. Opin. Immunol. 21, 215–223.
Watanabe, N., and Nakajima, H. (2012). Coinhibitory molecules in autoimmune
Sadelain, M., Brentjens, R., and Rivière, I. (2013). The basic principles of diseases. Clin. Dev. Immunol. 2012, 269756.
chimeric antigen receptor design. Cancer Discov 3, 388–398.
Wilkie, S., van Schalkwyk, M.C., Hobbs, S., Davies, D.M., van der Stegen, S.J.,
Sakaguchi, S., and Powrie, F. (2007). Emerging challenges in regulatory T cell Pereira, A.C., Burbridge, S.E., Box, C., Eccles, S.A., and Maher, J. (2012). Dual
function and biology. Science 317, 627–629. targeting of ErbB2 and MUC1 in breast cancer using chimeric antigen recep-
tors engineered to provide complementary signaling. J. Clin. Immunol. 32,
Sakaguchi, S., Fukuma, K., Kuribayashi, K., and Masuda, T. (1985). Organ- 1059–1070.
specific autoimmune diseases induced in mice by elimination of T cell subset.
I. Evidence for the active participation of T cells in natural self-tolerance; deficit Wing, K., Onishi, Y., Prieto-Martin, P., Yamaguchi, T., Miyara, M., Fehervari, Z.,
of a T cell subset as a possible cause of autoimmune disease. J. Exp. Med. Nomura, T., and Sakaguchi, S. (2008). CTLA-4 control over Foxp3+ regulatory
161, 72–87. T cell function. Science 322, 271–275.
Schreiber, R.D., Old, L.J., and Smyth, M.J. (2011). Cancer immunoediting: Yoshikai, Y., Anatoniou, D., Clark, S.P., Yanagi, Y., Sangster, R., Van den
integrating immunity’s roles in cancer suppression and promotion. Science Elsen, P., Terhorst, C., and Mak, T.W. (1984). Sequence and expression of
331, 1565–1570. transcripts of the human T-cell receptor b-chain genes. Nature 312, 521–524.
Schubert, D., Bode, C., Kenefeck, R., Hou, T.Z., Wing, J.B., Kennedy, A., Zhao, Y., Parkhurst, M.R., Zheng, Z., Cohen, C.J., Riley, J.P., Gattinoni, L.,
Bulashevska, A., Petersen, B.S., Schäffer, A.A., Grüning, B.A., et al. (2014). Restifo, N.P., Rosenberg, S.A., and Morgan, R.A. (2007). Extrathymic genera-
Autosomal dominant immune dysregulation syndrome in humans with tion of tumor-specific T cells from genetically engineered human hematopoiet-
CTLA4 mutations. Nat. Med. 20, 1410–1416. ic stem cells via Notch signaling. Cancer Res. 67, 2425–2429.
Seliger, B. (2008). Molecular mechanisms of MHC class I abnormalities Zinkernagel, R.M., and Doherty, P.C. (1974). Immunological surveillance
and APM components in human tumors. Cancer Immunol. Immunother. 57, against altered self components by sensitised T lymphocytes in lymphocytic
1719–1726. choriomeningitis. Nature 251, 547–548.