You are on page 1of 12

Published OnlineFirst July 16, 2018; DOI: 10.1158/2159-8290.

CD-18-0297

REVIEW

CARs versus BiTEs: A Comparison


between T Cell–Redirection Strategies
for Cancer Treatment
Clare Y. Slaney1,2, Pin Wang3, Phillip K. Darcy1,2, and Michael H. Kershaw1,2

ABSTRACT The redirection of T cells against tumors holds much promise for the treatment of
cancer. Two main approaches for T-cell redirection involve their genetic modifica-
tion with chimeric antigen receptors (CAR), or the use of recombinant proteins designated bispecific
T-cell engagers (BiTE). These approaches have demonstrated dramatic effects in patients with hemato-
logic cancers, although limited effect against solid cancers. Here, we review and compare the successes
and challenges of these two types of immunotherapies, with special focus on their mechanisms, and
discuss strategies to improve their efficacy against cancer.

Significance: CAR and BiTE cancer therapies have generated much excitement, but although the thera-
pies are potentially competitive, information directly comparing the two is difficult to obtain. Here, we
present the fundamentals of each approach and compare the range and level of functions they can elicit
from T cells, and their efficacy against cancers. Cancer Discov; 8(8); 924–34. ©2018 AACR.

INTRODUCTION ously, BiTEs mediate T-cell responses and killing of tumor


cells. Importantly, the T-cell/target cell adherence facilitated
Immunotherapy that utilizes the body’s immune system
by BiTE is independent of MHC haplotype (1).
against tumors is a promising field of cancer research. In recent
A variety of formats for bispecific antibodies have been
years, exciting technologies and platforms have been developed
developed in addition to BiTEs, including bispecific IgG, dia-
for the intricate design and production of anticancer immune
bodies, and conjugates, which have been the subject of some
reagents using antibodies in the form of single-chain variable
comprehensive recent reviews (2, 3). BiTEs have the advan-
fragments (scFv; Fig. 1). The scFv technology has been adopted
tages of relatively simple recombinant production and puri-
and used in some promising cancer immunotherapies, includ-
fication, and a molecular weight enabling tissue penetration.
ing bispecific T-cell engager (BiTE) and chimeric antigen
BiTEs have also advanced through the regulatory framework,
receptor (CAR). Both strategies use scFv to direct cytotoxic T
gaining FDA approval for certain indications, and will be the
lymphocytes (CTL) to specific surface antigens on cancer cells
focus of this review.
to facilitate a polyclonal T-cell response to tumor antigens.
The other area that has a high level of excitement for using
A BiTE is a recombinant bispecific protein that has two
scFv technologies is the CAR T-cell approach for adoptive cell
linked scFvs from two different antibodies, one targeting a
transfer. A CAR is a synthetic receptor comprising an extra-
cell-surface molecule on T cells (for example, CD3ε) and the
cellular tumor antigen recognition domain, which is fused
other targeting antigens on the surface of malignant cells.
to the CD3ζ chain for intracellular signaling and contains
The two scFvs are linked together by a short flexible linker
one or more costimulatory domains (Fig. 2). The binding of
(Fig. 1). By binding to tumor antigens and T cells simultane-
scFv to tumor cells triggers the T-cell receptor (TCR) intracel-
lular domain and leads to T-cell responses against antigen-
1
Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, expressing cells.
Victoria, Australia. 2Sir Peter MacCallum Department of Oncology, Univer- Various molecular formats of CAR have been developed,
sity of Melbourne, Parkville, Victoria, Australia. 3National Key Laboratory
differing in their extracellular, transmembrane, and cytoplas-
of Medical Immunology and Institute of Immunology, Second Military
Medical University, Shanghai, China. mic domains, which have been the subject of some previous
Corresponding Authors: Michael H. Kershaw, Peter MacCallum Cancer Cen-
excellent reviews (4). In this review, we focus on CARs in
tre, 305 Grattan Street, Melbourne, Victoria 3000, Australia. Phone: 613- which the predominant cytoplasmic signaling domains are
8559-7092; Fax: 613-8559-5039; E-mail: michael.kershaw@petermac.org; derived from CD3ζ and the costimulatory molecules CD28 or
and Clare Y. Slaney, clare.slaney@petermac.org CD137, which endow T cells with significant antitumor activ-
doi: 10.1158/2159-8290.CD-18-0297 ity, and which have received FDA approval for the treatment
©2018 American Association for Cancer Research. of some hematologic malignancies.

924 | CANCER DISCOVERY August 2018 www.aacrjournals.org

Downloaded from cancerdiscovery.aacrjournals.org on July 12, 2020. © 2018 American Association for Cancer
Research.
Published OnlineFirst July 16, 2018; DOI: 10.1158/2159-8290.CD-18-0297

CAR T-cell and BiTE Therapies for Cancers REVIEW

scFv
T cell
A Antigen C
Antigen
binding site binding site

Variable
Light chain
BiTE CD3
Anti-CD3 scFv
BiTE
Constant
Anti-TAA scFv
Heavy chain Linkers

TAA

IgG1 IgG2

B
VH Linker VL Linker VH Linker VL
Tumor cell
IgG1 scFv IgG2 scFv

Figure 1.  The design of a BiTE. A, Schematic representation of the derivation and structure of a BiTE generated from two antibodies, with specificity
for a T-cell activation molecule and a tumor-associated antigen (TAA). B, Schema of a BiTE gene used to produce the recombinant BiTE protein. Linkers
were constructed between VH and VL domains of the scFv and between the two scFvs. C, A BiTE creates an immunologic synapse by binding simultane-
ously to a tumor cell, via TAA, and a T cell, via CD3.

Recent advances and positive clinical results from BiTE MECHANISM KEY POINTS
and CAR T-cell therapies have generated hope and excite-
T-cell Types
ment. However, both therapies are facing similar dilemmas,
such as toxicity concerns in hematologic cancers and a lack T-cell populations are made up predominantly of two cell
of response in solid cancers. In this article, we review and subsets, expressing either CD4 or CD8. The main function
compare the recent progress in the application of BiTE and of CD8+ T cells is considered to be cytolysis of tumor cells,
CAR T-cell therapies, summarize the current understanding whereas CD4+ T cells secrete cytokines to regulate immune
of their anticancer mechanisms, and discuss strategies to responses. In the CAR T-cell approach, CD4+ T cells and CD8+
improve their efficacy against cancers (Table 1). T cells are usually mixed and genetically modified with CAR
and infused back to the patients in clinical trials. Although
there is no doubt that CD8+ CAR T cells are the major players
in killing cancer cells, CD4+ T cells armed with a CAR can also
Tumor cell be activated via CAR and showed cytolytic activities. Although
CD4+ CAR T cells eradicated leukemia cells more slowly, these
CD4+ CAR T cells persisted longer in vivo (5).
Clinical trials, mixing CAR-equipped CD4+ and CD8+ T cells
at a fixed ratio in treating patients, have generated remarkable
responses. A study using a defined ratio of CD4+:CD8+ CAR T
cells treating refractory B-cell acute lymphoblastic leukemia
(B-ALL) achieved bone marrow disease remission in 27 of 29
evaluable patients (6). In that study, 27 patients received a 1:1
TAA
ratio of CD4+:CD8+ CAR T cells, whereas 2 patients received
alternate ratios. Of the 2 patients receiving alternate ratios,
TCR Extracellular domain (scFv)
one relapsed and one achieved minimal residual disease. The
Hinge and TM domains CAR low patient numbers and intratrial differences in treatment
Signaling domains regimens did not allow a statistically meaningful comparison
between patients receiving a 1:1 and other ratios. Interest-
ingly, a similar approach using a defined mixture of CD4+
and CD8+ CAR T cells in patients with relapsed/refractory
B-cell non-Hodgkin lymphoma (NHL) achieved 64% com-
Figure 2.  The design of a CAR T cell. A CAR comprises an extracellular plete responses (7). This was comparable with other studies
domain, which is an scFv targeting TAA. The scFv is followed by a hinge of using CAR T cells of variable composition (8, 9), although
varying length and flexibility (CD8 as an example) and various combina-
tions of endodomains that provide T-cell activation signals, such as CD3ζ, differences in doses and treatment regimens did not allow a
and a costimulation signal, such as CD28. TM, transmembrane. direct comparison of efficacy.

August 2018 CANCER DISCOVERY | 925

Downloaded from cancerdiscovery.aacrjournals.org on July 12, 2020. © 2018 American Association for Cancer
Research.
Published OnlineFirst July 16, 2018; DOI: 10.1158/2159-8290.CD-18-0297

REVIEW Slaney et al.

Table 1. Comparison of CAR T cells and BiTEs

CAR T cell BiTE


Structure A synthetic gene construct encoding an scFv against A recombinant protein composed of two linked
tumor antigen linked to activation and costimulatory scFvs; one binds to CD3 on T cells and the other
motifs. to target a tumor antigen on tumor cells.
Effector cell types Engineered CD8+ and CD4+ T cells (5). Less-differentiated Endogenous CD8+ and CD4+ T cells (13). Antigen-
subsets displaying better antitumor activity experienced TEM but not TN effective (14).
in vivo (TSCM and TCM; ref. 10).
Immune synapse Atypical (15). Typical (17–19).
Serial killing Yes (16). Yes (22).
Killing mechanisms Perforin and granzyme B (16), Fas/Fas-L, or TNF/TNF-R. Perforin and granzyme B (17).
Trafficking Active. Trafficking of CAR T cells involves comprehen- Passive. Biodistribution depends on factors
sive interactions between various molecules and related to rates of diffusion through vascular
cell–cell interactions (57). endothelium, fluid flow rates, and interaction
with target.
Toxicity CRS, neurotoxicity, B-cell aplasia (31, 49). CRS, neurotoxicity, B-cell aplasia (62, 64).
Clinical applications Pretreatment lymphodepleting regime using cyclophos- No lymphodepletion regime required. Premedi-
phamide and fludarabine. cate with dexamethasone. Repeat administra-
Premedicate with acetaminophen and an H1-antihista- tion necessary, including continuous i.v. infusion
mine. One infusion. regimens.
FDA approval Yescarta was approved to treat adult patients with Blinatumomab was approved to treat relapsed/
relapsed/refractory large B-cell lymphoma in 2017. refractory B-ALL in 2014 and 2017.
Kymriah was approved to treat patients up to 25 years of
age with refractory/relapsed B-ALL in 2017.
Other characteristics Individually produced for each patient. “Off the shelf” reagents.

In addition to cell subsets, T cells can exist in several dif- T cells showed better efficacy, in BiTE treatment, antigen-
ferentiation states that have different functional abilities. T experienced T-cell subsets mediate BiTE-induced tumor cell
cells can be classified into naïve T cells (TN) and four main death, whereas naïve T cells are not activated when engaged
antigen-experienced or activated subtypes: stem cell memory by BiTE (1). In an early study, Bargou and colleagues reported
(TSCM), central memory (TCM), effector memory (TEM), and that following injection of the anti-CD19 BiTE blinatu-
effector (TEFF) T cells. It is now evident that less differentiated momab in patients with relapsed and refractory NHL, T cells
CAR T cells have better efficacy in vivo. In CD19-CAR–treated were redistributed and activated. T cells transiently declined
patients with relapsed/refractory B-cell malignancies, the fre- in numbers within 24 to 48 hours, as the T cells adhered to
quency of TSCM cells within the transferred lymphocytes cor- blood vessels endothelium and/or extravasated. T cells then
related with their in vivo expansion and persistence during the returned to baseline numbers with an activated phenotype,
first 6 weeks after infusion (10). Preclinical studies showed driven by an expansion of CD4+ and CD8+ TEM cells. T-cell lev-
that TSCM cells had greater resistance to cell death caused by els eventually exceeded pretreatment levels, and this increase
repetitive encounter with antigen and had better migration was due to the expansion of TEM cells, but not naïve T cells
ability to secondary lymphoid organs (10). It is proposed that (14). Despite some differences in the optimal cell phenotype
TCM are capable of self-renewal and give rise to shorter-lived for each approach, both therapies mediated tumor cell killing
TEM and TEFF cells that are incapable of self-renewal (11). independent of MHC and provide an opportunity to redirect
Therefore, although TEM and TEFF can be present at high fre- T-cell cytotoxicity to tumor cells.
quency at the peak of the anticancer immune response, their
persistence is poor. Accordingly, strategies favoring the gen- T-cell Cytotoxicity
eration and preservation of TSCM and TCM have been applied CAR T cells can induce target tumor cell death through the
in the CAR T-cell therapy field (12). formation of synapses, cytokine secretion, perforin/granzyme
Similar to CAR T-cell therapy, BiTE therapies involve poly- B release and/or death receptor ligation. Interestingly, CAR-
clonal T-cell responses, which are independent of MHC and mediated immune synapse formation has been reported to
TCR recognition and costimulation. It has been demon- differ from the traditional TCR-induced synapse, in that
strated that when incubated with BiTEs, both CD8+ and the CAR-induced synapse has a much smaller actin ring and
CD4+ T cells can be activated and induce target tumor cell diffuse distribution of LCK. In addition, the microtubule-
death, with CD8+ cells killing faster than CD4+ T cells (13). organizing center is distant from the immune synapse in
In contrast to CAR T-cell therapy, where less differentiated comparison with the classic TCR-induced synapse, which is

926 | CANCER DISCOVERY August 2018 www.aacrjournals.org

Downloaded from cancerdiscovery.aacrjournals.org on July 12, 2020. © 2018 American Association for Cancer
Research.
Published OnlineFirst July 16, 2018; DOI: 10.1158/2159-8290.CD-18-0297

CAR T-cell and BiTE Therapies for Cancers REVIEW

proximal to the LCK accumulation. The adhesion molecule malignancies, CAR T cells have been demonstrated to expand
LFA1 does not accumulate at the synapse, which could result up to, or even exceeding, 1,000-fold (31, 32) and to reach
in rapid detachment from the dying tumor cells, thereby greater than 20% of circulating lymphocytes (33). In clini-
enabling CAR T cells to act rapidly (15). In fact, CAR T-cell cal studies targeting CD19 on B-cell malignancies, absolute
serial killing (one T cell kills multiple targets) requires a sig- counts of circulating CAR T cells can vary among patients,
nificantly shorter time than TCR-mediated serial killing of but still achieve levels from several thousand per mL of blood
cancer cells (16). up to several hundred thousand per mL (8, 34, 35).
Similar to CAR T cells, it has been reported that, when In the case of BiTEs, a large pool of antigen-experienced T
incubated with BiTEs, both CD8+ and CD4+ T cells could be cells can be exploited to provide substantial numbers of redi-
activated to kill target tumor cells in vitro, via perforin and rected T cells, with less reliance on expansion. Nevertheless,
granzyme B (17), although roles for TNF and FAS do not increased numbers of circulating T cells have been demon-
appear to have been investigated. However, initial studies strated following BiTE administration, suggesting expansion
suggest that differences exist between the synapse formed of the pool of T cells with the potential to respond against
between T cells and target cells when mediated by BiTEs tumor cells. Increases of the order of 2- to 4-fold in circulat-
compared with CARs. The linkage of tumor cells and T cells ing T cells have been described (14, 36, 37). However, because
by BiTE leads to the formation of cytolytic synapses that are the number of antigen-experienced T cells (able to respond
similar to the ones formed during regular cytotoxic T-cell independently of costimulation) comprises only a propor-
recognition (17–19). Once activated, these T cells start to tion of total T cells, the actual level of expansion of respond-
proliferate and also upregulate the expression of activation ing T cells is likely several folds higher than that reflected in
markers, such as CD69 and CD25 (20). Although the reason increases in total circulating T cells.
for the difference in synapses formed by CARs and BiTEs is The provision of large numbers of tumor-reactive T cells in
not clear, it potentially arises because BiTEs engage the natu- the solid-tumor setting presents different challenges for the
ral CD3 complex, whereas CARs do not. BiTE and CAR T-cell approaches. Whereas extensive expan-
Both the size and the position of the epitope to the cell sion and prolonged persistence has been demonstrated for
surface determine the extent of BiTE-mediated cancer cell CAR T cells in hematologic cancers, little if any expansion of
killing (21). Activated T cells kill the target cancer cells via CAR T cells is usually demonstrated when treating solid can-
triggering apoptosis, as evidenced by caspase 3/7 activation, cers (38–40), except for isolated reports of expansion exceed-
PARP cleavage, DNA fragmentation, and membrane blebbing ing 100-fold (41, 42). CAR T cells directed against blood
(17). Similar to CAR T cells, it has been shown that BiTEs can cancers may receive further costimulatory signals from cancer
induce serial killing by T cells, with one T-cell able to kill sev- cells, or other antigen-positive leukocytes, in a microenviron-
eral target cells (22). However, unlike CAR-redirected T-cell ment that is less immunosuppressive than that found in solid
function, BiTE-redirected function is sensitive to the dose of tumors. This may explain the observed differences in T-cell
BiTE administered. Although this may have advantages in expansion in liquid cancers compared with solid cancers.
tuning the response to avoid an overreaction and potential Approaches combining CAR T-cell transfer with vaccines, to
toxic levels of cytokine, an excess of BiTE may completely provide T-cell support away from the tumor microenviron-
coat CD3 and target antigen separately, thereby reducing ment, are being used to enhance CAR T-cell proliferation and
the opportunity for coengagement of T cells and target cells persistence in the solid-tumor setting (43, 44).
(23–25). Because the BiTE approach is potentially less reliant on
Although direct cancer cell lysis is seen as the most potent T-cell expansion, it may be more successful against solid
means to reduce cancer burden by T cells, large amounts of tumors. However, the success of this approach is dependent
cytokines can be secreted by both CAR- and BiTE-redirected on the sustained loading of BiTEs on T cells, which may
means (26). These effector cytokines have the potential to be compromised during the process of penetration of solid
change the tumor microenvironment and induce endog- tumors.
enous antitumor immunity. This potential was evident in a Differences between the CAR and BiTE approaches also exist
study that demonstrated the ability of CAR T cell–secreted when considering tumor recurrence. CAR T cells are able to
cytokines to change the recruitment and activation of endog- engraft long-term and provide an ongoing source of tumor-
enous macrophages and create a favorable milieu for antican- reactive T cells to respond against recurring tumors, even before
cer immune response (27). Interestingly, studies using both they become evident. This has been demonstrated in mouse
BiTEs and CARs suggest that cytokines secreted upon target tumor models, in which mice that rejected a primary tumor also
cell ligation can contribute to lysis of antigen-negative tumor rejected a secondary challenge with tumor cells (43). In contrast,
cells in close proximity to the antigen-specific engagement, without continuous administration of BiTEs, recurrent tumors
so-called bystander lysis (28, 29), although this is not always have the opportunity to grow large before detection, which may
the case for all target antigens (30). render subsequent treatment with BiTEs less effective.

T-cell Numbers and Persistence


The resolution of malignant disease requires large num- CLINICAL APPLICATIONS
bers of responding T cells and their continued action over CAR T-cell therapies have generated fantastic responses
prolonged time periods. In the case of CAR T cells, these in B-cell malignancies and revolutionized the field of cancer
requirements are best achieved by expansion in vivo and long- immunotherapy. CD19 CAR T-cell therapy has been tested in
term engraftment following transfer. In some hematologic treating different B-cell malignancies, including NHL (7, 45,

August 2018 CANCER DISCOVERY | 927

Downloaded from cancerdiscovery.aacrjournals.org on July 12, 2020. © 2018 American Association for Cancer
Research.
Published OnlineFirst July 16, 2018; DOI: 10.1158/2159-8290.CD-18-0297

REVIEW Slaney et al.

46), chronic lymphocytic leukemia (CLL; refs. 47, 48), and ALL are less satisfactory than the ones used in blood cancers.
(6, 31, 34, 49, 50). In these trials, the relapsed/refractory patients There is great enthusiasm to improve CAR T-cell treatment
who had limited treatment options were treated with CD19 efficacy against solid cancers in preclinical research, and this
CAR T cells, and the treatment generated remarkably high is discussed in the next section.
rates of complete responses, over 50% in NHL (7, 8, 46), 90% in Similar to CAR T cells, BiTEs have been used in clinical
ALL (6, 49, 50), and over 70% complete response and/or partial trials targeting a variety of antigens. Blinatumomab was
response in CLL (48). In 2017, two CD19 CAR T-cell adoptive the first clinically tested and FDA-approved BiTE, receiving
transfer treatments were approved by the FDA. Yescarta (axi- accelerated approval in 2014 for the treatment of CD19+
cabtagene ciloleucel; Kite Pharma) was approved to treat adult Philadelphia chromosome–negative (Ph−) relapsed and refrac-
patients with relapsed or refractory large B-cell lymphoma. tory B-ALL. In July 2017, the FDA further approved its use to
Kymriah (tisagenlecleucel; Novartis) was approved for the treat- include the treatment of Philadelphia chromosome–positive
ment of patients up to 25 years of age with refractory/relapsed (Ph+) relapsed or refractory B-ALL.
B-ALL. Preconditioning that depletes endogenous lymphocytes A phase II trial (ALCANTARA, NCT02000427) enrolled
is important for CAR T-cell therapy; without this, significant 45 patients with Ph+ relapsed or refractory B-ALL for
clinical benefit is rarely observed (51). the blinatumomab treatment. In this trial, 16 patients
Given the remarkable initial response rate observed using (36%) achieved complete remission (CR) or CR with partial
CAR T cells in some blood cancers, but a significant relapse hematologic recovery, and the median duration of remis-
frequency at present, the question remains as to whether sion was 6.7 months (62). Thus, blinatumomab has led to
this form of therapy would be best applied as a stand-alone promising results for patients with Ph+ relapsed/refractory
treatment or as a bridge to stem-cell transplantation. The use ALL. A recent phase II trial (TOWER, NCT02013167) com-
of CAR T cells as a bridge to transplant has some attractive pared blinatumomab with standard-care chemotherapy
features including dramatic reduction in disease burden often in 405 patients with Ph− relapsed and refractory B-ALL.
to undetectable levels, perhaps enabling a subsequent less- Blinatumomab demonstrated a significant improvement
intensive conditioning regimen prior to transplant. In the CAR in overall survival (OS) and higher rates of hematologic
T-cell setting, transplantation following conditioning would remission than chemotherapy. The median OS was 7.7
have the added benefit of eliminating CAR T cells to allow months with blinatumomab compared with 4 months
recovery of normal B cells, because B-cell aplasia can persist with chemotherapy (63).
long-term in a substantial proportion of patients with leuke- Apart from ALL, blinatumomab has also shown promise
mia/lymphoma achieving complete responses. This is likely to in treating NHL. The initial clinical studies used short infu-
continue to be an important option for clinicians in the treat- sion of blinatumomab and did not show efficacy but did
ment of hematologic cancers. However, the increased cost of show toxicities such as cytokine release syndrome (CRS) and
an additional transplant procedure, together with transplant- neurotoxicity (14). Subsequently, lessons were learned from
related toxicities if using allogeneic donors, indicates that with the ALL studies for continuous infusion of blinatumomab.
improvements to cell production and efficacy, and controlled A clinical trial using continuous infusion of blinatumomab
gene expression or inclusion of suicide genes for CAR T cells, a 60 μg/m2/day in NHL (4-fold higher than ALL) has proven to
stand-alone approach would be an ultimate goal. be feasible. In this phase I trial, single-agent blinatumomab
Although highly effective against CD19 B-cell malignan- showed promising activity with an objective response rate of
cies, CAR T-cell therapy did not generate the same potent 69% (24/35 patients; ref. 64).
response against other lymphomas/leukemias. In addition, As with CAR T cells, treatment with BiTEs can lead to
the loss of CD19 expression in patients can limit the CD19- high initial response rates but a significant relapse rate, and
CAR therapeutic benefit. Recently, a few additional CAR the use of BiTEs in the hematologic setting offers an option
targets have been tested. CD22 in treating patients with ALL as a bridge to transplantation. As with CAR T-cell therapy,
who are refractory to CD19 CAR T-cell therapy (52), CD20 in dramatic reductions in disease and complete responses are
indolent B-cell and mantle cell lymphomas (53), B-cell matu- observed in many patients but, in contrast to CAR T cells
ration antigen (BCMA) in multiple myeloma (54), and Lewis where T cells persist long-term, BiTEs require continuous
Y in acute myeloid leukemia (AML; ref. 55) have been tested administration to provide protection against disease recur-
in the clinic and generated encouraging results. rence. Therefore, hematopoietic stem-cell transplant follow-
In solid-cancer clinical trials, CAR T cells have not dem- ing BiTE therapy may be beneficial. Longer-term follow-up of
onstrated the same dramatic effect as seen in hematologic complete responders receiving BiTEs (or CAR T cells) will be
cancers. The reasons are believed to be the solid-tumor immu- informative to determine the feasibility of using T-cell redi-
nosuppressive microenvironment, impaired T-cell trafficking, rection as a stand-alone therapy.
and the suboptimal phenotype of the infused CAR T cells (11, Other tumor antigens have also been explored for BiTE
56, 57). A number of tumor antigens have been targeted in therapy. For example, CD3–CD33 (AMG330) BiTE has been
solid cancers, such as GD2 in neuroblastoma (58), HER2 in constructed to treat patients with relapsed/refractory AML
glioblastoma and other solid cancers (40, 59), and EGFRvIII (NCT02715011).
in glioblastoma (60). Interestingly, a recent inspiring study For treating solid tumors, CD3-coupled BiTEs specific for
reported that CAR T-cell treatment targeting IL13Rα-2 has EPCAM (solitomab), CEA, and prostate-specific membrane
successfully eradicated a case of glioblastoma, and the com- antigen (PSMA) have all been tested. Limited responses were
plete response lasted several months (61). Although some of observed in treating patients with solid tumors, and toxicity
the trials have observed some response, the overall outcomes was observed. In a trial using solitomab treating different

928 | CANCER DISCOVERY August 2018 www.aacrjournals.org

Downloaded from cancerdiscovery.aacrjournals.org on July 12, 2020. © 2018 American Association for Cancer
Research.
Published OnlineFirst July 16, 2018; DOI: 10.1158/2159-8290.CD-18-0297

CAR T-cell and BiTE Therapies for Cancers REVIEW

solid tumors, transient elevation of liver enzymes and diarrhea plasma precursor cells, resulting in substantial hypogamma-
as dose-limiting toxicities were observed. This may be due to globulinemia. This presents risk of infections, which can be
the targeting of EPCAM to normal tissue in bowel epithelial reduced by administration of intravenous immunoglobulin.
and bile ducts. The toxicity precluded further dose escalation
(65). Although some tumor response was observed, higher
doses were limited by toxicity. New targets, such as gpc3 to RESEARCH IN ENHANCING TREATMENT
treat hepatocellular carcinoma, have also been tested (66). EFFICACY AGAINST SOLID CANCERS
Both BiTE and CAR T-cell treatment strategies have pro-
duced remarkable effects in hematologic malignancies; how-
TOXICITIES ever, the efficacy for both was generally poor against solid
CAR T-cell therapy can be associated with some toxicities. cancers. Considerable effort is being invested in enhancing
The most commonly observed toxicity is CRS, a systemic the treatment efficacy against solid cancers.
response due to elevated levels of cytokines. Studies have dem- For CAR T-cell therapy, the hurdles for treating solid can-
onstrated that high levels of 24 serum cytokines, such as IL6 cers lie in the combination of the immunosuppressive effect
and IFNγ in the first months after CAR T-cell infusion, were of the tumor microenvironment and inefficient penetration
associated with severe CRS (67). CRS occurs several hours of CAR T cells into tumors. The tumor microenvironment
to 14 days after CAR T-cell infusion, and symptoms include is a complex assortment of cell types including malignant
fever, fatigue, anorexia, hypotension, tachycardia, and capil- cells, fibroblasts, endothelium, and immunoregulatory cells.
lary leak, which can be life-threatening. It was demonstrated Regulatory cell types include myeloid-derived suppressor cells
that the patients with highly elevated levels of IL6 are the ones (MDSC), M2 macrophages, and regulatory T cells (Treg; ref.
with maximum CAR T-cell activation and proliferation (49). 73). Using a preclinical model, Moon and colleagues demon-
Anti-IL6 receptor antibody (tocilizumab) that blocks IL6 from strated that CAR T cells were able to slow down tumor growth,
binding to its receptor can reverse the life-threatening CRS but did not cause regression. The reason was the rapid loss of
without inhibiting CAR T-cell treatment efficacy (31). functions of CAR T cells when exposed to the tumor microen-
Neurotoxicity, also termed CAR T cell–related encephalopa- vironment in vivo. Diminished functions included reduction
thy syndrome (CRES), has also been reported (31, 48, 49). in the ability to secrete cytokines, killing ability, and phospho-
CRES is typically manifested by symptoms of confusion and ERK expression. When CAR T cells were isolated and rested
delirium and occasionally by seizures and cerebral edema (6, away from the tumor, their hypofunction was reversed (74).
68). The pathogenesis of neurologic toxicity remains unclear, For targeting the suppressive tumor microenvironment,
although it has been proposed that CRES could be due to a number of strategies have been tested. For example, using
cytokine-mediated endothelial activation and passive diffusion all-trans retinoic acid (75) or GM-CSF neutralization to dis-
of cytokines into the brain (69) and/or trafficking of T cells rupt MDSC function (76), targeting CD123 on macrophages
into the central nervous system (CNS; ref. 68). Although CD19 (77), or fibroblast activation protein on cancer-associated
CAR T cells were identified in the cerebrospinal fluid (CSF), fibroblasts (78) inhibiting adenosine 2A receptors (79) and
levels of the transgene in the brain did not correlate with rates altering metabolic components such as inhibiting protein
of high-grade CRES events (32). CRES is generally reversible kinase A activation (80) all demonstrated enhancement of
using anti-IL6 receptor and/or corticosteroids (68, 69). CAR T-cell treatment efficacy in preclinical models of solid
B-cell aplasia is also associated with CD19 CAR T-cell tumors. In addition, there has been great interest in using
therapy, because CD19 is expressed on normal B cells, but checkpoint inhibitors as adjuvant therapy for CAR T-cell
injections of immunoglobulin can be used to maintain levels treatment. Blocking a number of checkpoints, such as PD-1
of circulating antibodies (49). Infections with microorganisms (81) and LAG3 (82), has been shown to enhance CAR T-cell
can also occur following CAR T-cell transfer, which was largely treatment in solid tumors.
due to depletion of endogenous leukocytes by precondition- Other strategies that have shown promising results include
ing regimens (70). Bacterial infections predominated, which supplying CAR T cells with cytokine support to enhance
included bacteremia and more localized infections to sites their activity within the tumor microenvironment (83). Such
including gastrointestinal tissue. Viral and fungal infections approaches include inducing the local release of stimulatory
were also experienced. These infections are largely manageable cytokines, including IL12 (84), and transducing the IL7 recep-
using antibiotics, although some fatalities have resulted. tor into CAR T cells to provide signal 3 for T-cell support
Although leukodepleting preconditioning is not required (85). Inhibitory cytokines present in the tumor microenviron-
for BiTE therapy, toxicities after blinatumomab treatment are ment can also be harnessed to provide support for T cells. In
common, but mostly manageable. The most frequent adverse this approach, T cells are genetically modified with chimeric
effects were CRS and neurologic adverse effects including cytokine receptors. For example, chimeric cytokine receptors
headache, tremor, aphasia, and seizure (62, 64). These effects composed of the extracellular domain of the IL4 receptor
might be associated with T-cell migration to the CNS or a fused to the intracellular domain of the IL7 receptor led to
transient cytokine storm in serum. Currently, the blinatu- activation and proliferation of T cells in the presence of the
momab treatment dosage and administration schedule is normally inhibitory cytokine IL4 (86, 87).
based on body weight, and patients are required to be pre- To enhance CAR T-cell infiltration to the tumor bed, effort
medicated with dexamethasone, to reduce cytokine produc- has been made to modify chemokine receptors on CAR T cells
tion while retaining cytolytic activity (71, 72). In addition, the to enhance their migration toward tumors. For example, CAR
treatment of blinatumomab also depleted normal B cells and T cells have been modified to express CCR2b, the chemokine

August 2018 CANCER DISCOVERY | 929

Downloaded from cancerdiscovery.aacrjournals.org on July 12, 2020. © 2018 American Association for Cancer
Research.
Published OnlineFirst July 16, 2018; DOI: 10.1158/2159-8290.CD-18-0297

REVIEW Slaney et al.

receptor for CCL2 that is highly expressed by the target Strategies for changing the tumor microenvironment may
tumors (88). To facilitate the penetration into tumors, con- greatly enhance BiTE treatment efficacy in solid cancers. It
struction of CARs targeting VEGFR2 or αvβ3 integrin over- was reported that tumors upregulated PD-L1 post–BiTE
expressed on tumor vasculature has been reported (89, 90). administration (102) and adding anti–PD-L1 to AMG 330
Similar to CAR T-cell therapy, the efficacy of BiTEs in treat- treatment enhanced killing in vitro (103). Other inhibitor
ing solid tumors in preclinical models is modest. One of the molecules to target include galectin 1, which has been docu-
major hurdles for BiTEs is their short half-life in serum. After mented to be linked with BiTE resistance (104). Enhancing
injection, BiTEs are rapidly catabolized and cleared from the tumor antigen presentation has also been shown to be a
circulation, leading to a short serum half-life of around 2 good strategy for enhancing BiTE treatment efficacy. Adding
hours (91). To overcome the limitation, current approaches the epigenetic modifier drugs panobinostat and azacitidine
include attaching BiTEs to heavy-chain fragments or other increased CD33 expression in some cell lines and enhanced
novel designs to increase the serum life. A number of these AMG 330–induced cytotoxicity (105).
methods significantly increased the BiTE circulation time A few studies directly compared the functional efficacy of
without hampering its binding to the target cells (92). the two strategies in their antitumor effect (106). In an early
The other major hurdle for treating solid tumors using study, Stone and colleagues compared the CAR and BiTE
BiTE is the density and types of T cells already in the tumor treatment antitumor response in vitro using the same antican-
bed. Small-animal PET and fluorescent imaging studies have cer scFv against a murine fibrosarcoma epitope, 237. Although
demonstrated the penetration of BiTE into solid tumors both strategies enhanced T cell–mediated antitumor effect,
(93). However, the profiles of the T cells already existing in CAR T cells were more potent than the BiTE approach against
the tumor are difficult to predict, as the intensity, location, cancers expressing lower levels of antigens (107). In a recent
phenotype, and activation phenotypes of these T cells vary study, Hoseini and colleagues compared a bispecific antibody
dramatically among tumors (94). In addition, preexisting T (specific for both CD3 and GD2), with a CAR that contained
cells in the tumor may already have an exhausted phenotype the same anti-GD2 scFv (106). Incubating GD2+ target cancer
or may be Tregs. Approaches in increasing T-cell infiltration cells with anti-GD2 CAR T cells for 24 hours led to the deple-
into tumor tissue and combinational approaches to reverse tion of the CARhi T cells, and, in contrast, in the presence of
T-cell exhaustion and targeting the tumor immunosuppres- BC119, the exposure of untransduced T cells to GD2+ target
sive microenvironment warrant further investigation to boost cancer cells did not deplete the T cells. This GD2-specific CAR
the efficacy of BiTE against solid cancers. T-cell depletion severely compromised CAR T-cell cytotoxic-
Recent enthusiasm surrounding BiTEs extends toward ity in vitro and impaired their in vivo antitumor efficacy. For
their production in vivo. One strategy is to produce geneti- treating the GD2+ tumors in immunocompromised mice,
cally modified T cells for making BiTE to overcome the BC119 with untransduced T cells conferred rapid shrinkage
limitations of BiTEs’ short half-life and passive biodistribu- of the established tumors, whereas CAR T cell–treated tumors
tion. Technologies were developed to generate T cells that showed a delayed regression for 2 weeks. The percentage of
secrete BiTEs (ENG-T cells) targeting tumor antigens such tumor-infiltrating lymphocytes (TIL) in the BC119 treatment
as EPHA2 (95), CD123 (96), and CD19 (97). These ENG-T was higher than that in the CAR T-cell treatment. Interest-
cells can recognize tumor cells in an antigen-dependent man- ingly, the ratio of CD4+ and CD8+ TILs in BC119-treated mice
ner in vitro and in vivo, redirecting T cells to the target cancer was nearly equal, whereas the TILs in the CAR T cell–treated
cells, and have potent antitumor activity in cancer models mice were nearly all CD8+. The number and composition of
(95, 96). Importantly, these ENG-T cells expanded in vivo and TILs could partially explain the superior antitumor effect
increased the production of BiTEs after activation, obviating mediated by BiTE over CAR T-cell treatment. It is note­worthy
the need for continuous infusion of engager molecules (95). that the above study used immunocompromised mice with
Interestingly, a recent report compared the efficacy of RNA- adoptive transferred T cells, whereas in the clinic, BiTEs are
electroporated ENG-T cells to the RNA-electroporated CAR usually administered without adoptive transfer. The TIL pro-
T cells and concluded that the ENG-T cells showed more files in immunocompetent mice are poorly studied, albeit a
efficacy in the NALM6 leukemia model (98). few studies demonstrated that BiTE could suppress tumor
Another recent promising strategy is to use an oncolytic virus progression in these mice (108–110).
engineered to express a BiTE. Oncolytic viruses have been tested In the clinic, it is possible that patients refractory to one
in the clinic and have demonstrated safety and some anticancer therapy respond to the other. For example, in a trial run by
effects. Two recent studies engineered an oncolytic adenovi- Maude and colleagues in 2014, 2 patients refractory to bli-
rus to secrete an EGFR-targeting BiTE. The BiTE-expressing natumomab achieved CR after CD19-CAR treatment (49).
adenovirus induced a strong T-cell response against cancer cells This suggests that a lack of response to one immunotherapy
both in vitro and in vivo (99, 100). The use of oncolytic virus in against the same target may not preclude a successful therapy
combination with BiTE or the oncolytic virus secreting BiTE is outcome with another, even with the same target. In addition,
another strategy for changing the tumor microenvironment. there could conceivably be some benefit from using CAR T
Several oncolytic viruses are currently under development, and cells and BiTEs simultaneously against tumors. A preclinical
whether different strains of these viruses are better than others study targeting the alpha folate receptor using a CAR, while
to be engineered to express and secrete BiTE is yet to be inves- also targeting EGFR using an oncolytic virus encoding a
tigated. Post–oncolytic virus administration, large numbers of BiTE, led to increased inhibition of tumors (111). This study
immune cells infiltrated to the tumors, and the addition of suggested that combining these approaches, and oncolytic
BiTE redirects infiltrating T cells against tumors (101). viruses, could address the issues of antigen heterogeneity and

930 | CANCER DISCOVERY August 2018 www.aacrjournals.org

Downloaded from cancerdiscovery.aacrjournals.org on July 12, 2020. © 2018 American Association for Cancer
Research.
Published OnlineFirst July 16, 2018; DOI: 10.1158/2159-8290.CD-18-0297

CAR T-cell and BiTE Therapies for Cancers REVIEW

immunosuppressive microenvironment of solid tumors to 5. Yang Y, Lin T, Jacoby E, Qin H, Gardner EG, Chien CD, et  al.
enhance antitumor responses. CD4 CAR T Cells Mediate CD8-like cytotoxic anti-leukemic effects
resulting in leukemic clearance and are less susceptible to attenua-
tion by endogenous TCR activation than CD8 CAR T cells. Blood
CONCLUSIONS 2015;126:100.
6. Turtle CJ, Hanafi LA, Berger C, Gooley TA, Cherian S, Hudecek M,
We have summarized the current clinical status of the et  al. CD19 CAR-T cells of defined CD4+:CD8+ composition in
two immunotherapies BiTE and CAR T-cell treatment, their adult B cell ALL patients. J Clin Invest 2016;126:2123–38.
modes of action, toxicities, and preclinical development. Both 7. Turtle CJ, Hanafi LA, Berger C, Hudecek M, Pender B, Robinson E,
platforms have demonstrated excellent efficacy in clinical et al. Immunotherapy of non-Hodgkin’s lymphoma with a defined
ratio of CD8+ and CD4+ CD19-specific chimeric antigen receptor-
trials against hematologic malignancies and were recently modified T cells. Sci Transl Med 2016;8:355ra116.
approved by the FDA for treating B-ALL. With their applica- 8. Kochenderfer JN, Somerville RPT, Lu T, Shi V, Bot A, Rossi J, et al.
tions in the clinic, we will understand more about the anti- Lymphoma remissions caused by anti-CD19 chimeric antigen recep-
cancer mechanisms, investigate these treatments as single tor T cells are associated with high serum interleukin-15 levels.
or combined methods in different situations, and eventually J Clin Oncol 2017;35:1803–13.
expand their success to other cancers. 9. Locke FL, Neelapu SS, Bartlett NL, Siddiqi T, Chavez JC, Hos-
ing CM, et  al. Phase 1 results of ZUMA-1: a multicenter study of
Despite the excitement, a few issues still remain to be
KTE-C19 Anti-CD19 CAR T cell therapy in refractory aggressive
overcome. Interestingly, both platforms are facing the same lymphoma. Mol Ther 2017;25:285–95.
dilemma: CRS and neurologic adverse effects and moderate 10. Xu Y, Zhang M, Ramos CA, Durett A, Liu E, Dakhova O, et  al.
anticancer effects in solid-tumor settings. Substantial effort Closely related T-memory stem cells correlate with in vivo expansion
has been invested in finding strategies for reducing toxicities of CAR.CD19-T cells and are preserved by IL-7 and IL-15. Blood
and enhancing treatment efficacy against solid tumors, such 2014;123:3750–9.
11. Sadelain M, Riviere I, Riddell S. Therapeutic T cell engineering.
as targeting the immunosuppressive microenvironment and
Nature 2017;545:423–31.
enhancing T-cell infiltration to the tumors. Both treatments 12. Hurton LV, Singh H, Najjar AM, Switzer KC, Mi T, Maiti S, et  al.
warrant further exploration in a wide variety of tumor set- Tethered IL-15 augments antitumor activity and promotes a stem-
tings. cell memory subset in tumor-specific T cells. Proc Natl Acad Sci U S A
2016;113:E7788–E97.
Disclosure of Potential Conflicts of Interest 13. Mack M, Gruber R, Schmidt S, Riethmüller G, Kufer P. Biologic
No potential conflicts of interest were disclosed. properties of a bispecific single-chain antibody directed against
17-1A (EpCAM) and CD3: tumor cell-dependent T cell stimulation
Acknowledgments and cytotoxic activity. J Immunol 1997;158:3965.
14. Bargou R, Leo E, Zugmaier G, Klinger M, Goebeler M, Knop S, et al.
This work was supported by grants from Cure Cancer Australia Tumor regression in cancer patients by very low doses of a T cell-
(1100199), the Peter MacCallum Cancer Centre Foundation, the engaging antibody. Science 2008;321:974–7.
National Health and Medical Research Council (NHMRC) of Aus- 15. Davenport AJ, Cross RS, Watson KA, Liao Y, Shi W, Prince HM,
tralia (1103352 and 1132373), the National Breast Cancer Foun- et al. Chimeric antigen receptor T cells form nonclassical and potent
dation (NBCF) of Australia (IIRS-18-064), and Susan G. Komen immune synapses driving rapid cytotoxicity. Proc Natl Acad Sci U S A
(16376637). P.K. Darcy and M.H. Kershaw were supported by 2018;115:E2068–E76.
NHMRC Senior Research Fellowships. C.Y. Slaney was supported by 16. Davenport AJ, Jenkins MR, Cross RS, Yong CS, Prince HM, Ritchie
a postdoctoral fellowship from the NBCF. P. Wang was supported DS, et  al. CAR-T cells inflict sequential killing of multiple tumor
by grants from the National Natural Science Foundation of China target cells. Cancer Immunol Res 2015;3:483–94.
(31470871, 81671566, and 31722019). 17. Haas C, Krinner E, Brischwein K, Hoffmann P, Lutterbuse R,
Schlereth B, et al. Mode of cytotoxic action of T cell-engaging BiTE
The costs of publication of this article were defrayed in part by antibody MT110. Immunobiology 2009;214:441–53.
the payment of page charges. This article must therefore be hereby 18. Loffler A, Kufer P, Lutterbuse R, Zettl F, Daniel PT, Schwenkenbe-
marked advertisement in accordance with 18 U.S.C. Section 1734 cher JM, et al. A recombinant bispecific single-chain antibody, CD19 ×
solely to indicate this fact. CD3, induces rapid and high lymphoma-directed cytotoxicity by
unstimulated T lymphocytes. Blood 2000;95:2098–103.
19. Offner S, Hofmeister R, Romaniuk A, Kufer P, Baeuerle PA. Induction of
Received March 29, 2018; revised May 20, 2018; accepted June 1,
­regular cytolytic T cell synapses by bispecific single-chain antibody con-
2018; published first July 16, 2018.
structs on MHC class I-negative tumor cells. Mol Immunol 2006;43:763–71.
20. Brischwein K, Parr L, Pflanz S, Volkland J, Lumsden J, Klinger M,
et al. Strictly target cell-dependent activation of T cells by bispecific
REFERENCES single-chain antibody constructs of the BiTE class. J Immunother
1. Klinger M, Benjamin J, Kischel R, Stienen S, Zugmaier G. Harness- 2007;30:798–807.
ing T cells to fight cancer with BiTE(R) antibody constructs— 21. Bluemel C, Hausmann S, Fluhr P, Sriskandarajah M, Stallcup WB,
past developments and future directions. Immunol Rev 2016;270: Baeuerle PA, et al. Epitope distance to the target cell membrane and
193–208. antigen size determine the potency of T cell-mediated lysis by BiTE
2. Spiess C, Zhai Q, Carter PJ. Alternative molecular formats and thera- antibodies specific for a large melanoma surface antigen. Cancer
peutic applications for bispecific antibodies. Mol Immunol 2015; Immunol Immunother 2010;59:1197–209.
67:95–106. 22. Hoffmann P, Hofmeister R, Brischwein K, Brandl C, Crommer S,
3. Velasquez MP, Bonifant CL, Gottschalk S. Redirecting T cells to Bargou R, et  al. Serial killing of tumor cells by cytotoxic T cells
hematological malignancies with bispecific antibodies. Blood 2018; redirected with a CD19-/CD3-bispecific single-chain antibody con-
131:30–8. struct. Int J Cancer 2005;115:98–104.
4. Sadelain M, Brentjens R, Riviere I. The basic principles of chimeric 23. Amann M, D’Argouges S, Lorenczewski G, Brischwein K, Kischel R,
antigen receptor design. Cancer Discov 2013;3:388–98. Lutterbuese R, et al. Antitumor activity of an EpCAM/CD3-bispecific

August 2018 CANCER DISCOVERY | 931

Downloaded from cancerdiscovery.aacrjournals.org on July 12, 2020. © 2018 American Association for Cancer
Research.
Published OnlineFirst July 16, 2018; DOI: 10.1158/2159-8290.CD-18-0297

REVIEW Slaney et al.

BiTE antibody during long-term treatment of mice in the absence chimeric antigen receptor-modified T Cells for the immunotherapy
of T-cell anergy and sustained cytokine release. J Immunother of HER2-positive sarcoma. J Clin Oncol 2015;33:1688–96.
2009;32:452–64. 41. Gargett T, Yu W, Dotti G, Yvon ES, Christo SN, Hayball JD, et  al.
24. Dreier T, Baeuerle PA, Fichtner I, Grun M, Schlereth B, Lorencze- GD2-specific CAR T cells undergo potent activation and deletion
wski G, et  al. T cell costimulus-independent and very efficacious following antigen encounter but can be protected from activation-
inhibition of tumor growth in mice bearing subcutaneous or leuke- induced cell death by PD-1 blockade. Mol Ther 2016;24:1135–49.
mic human B cell lymphoma xenografts by a CD19-/CD3-bispecific 42. Junghans RP, Ma Q, Rathore R, Gomes EM, Bais AJ, Lo AS, et  al.
single-chain antibody construct. J Immunol 2003;170:4397–402. Phase I Trial of Anti-PSMA designer CAR-T cells in prostate cancer:
25. Amann M, Friedrich M, Lutterbuese P, Vieser E, Lorenczewski possible role for interacting interleukin 2-T cell pharmacodynamics
G, Petersen L, et  al. Therapeutic window of an EpCAM/CD3- as a determinant of clinical response. Prostate 2016;76:1257–70.
specific BiTE antibody in mice is determined by a subpopulation of 43. Slaney CY, von Scheidt B, Davenport AJ, Beavis PA, Westwood JA,
EpCAM-expressing lymphocytes that is absent in humans. Cancer Mardiana S, et  al. Dual-specific chimeric antigen receptor T cells
Immunol Immunother 2009;58:95–109. and an indirect vaccine eradicate a variety of large solid tumors in an
26. Brentjens RJ, Santos E, Nikhamin Y, Yeh R, Matsushita M, La Perle immunocompetent, self-antigen setting. Clin Cancer Res 2017;23:
K, et al. Genetically targeted T cells eradicate systemic acute lympho- 2478–90.
blastic leukemia xenografts. Clin Cancer Res 2007;13:5426–35. 44. Rossig C, Pule M, Altvater B, Saiagh S, Wright G, Ghorashian S,
27. Spear P, Barber A, Rynda-Apple A, Sentman CL. Chimeric antigen et  al. Vaccination to improve the persistence of CD19CAR gene-
receptor T cells shape myeloid cell function within the tumor modified T cells in relapsed pediatric acute lymphoblastic leukemia.
microenvironment through IFN-gamma and GM-CSF. J Immunol Leukemia 2017;31:1087–95.
2012;188:6389–98. 45. Schuster SJ, Svoboda J, Chong EA, Nasta SD, Mato AR, Anak O,
28. Ross SL, Sherman M, McElroy PL, Lofgren JA, Moody G, Baeuerle et al. Chimeric antigen receptor T cells in refractory B-cell lympho-
PA, et al. Bispecific T cell engager (BiTE(R)) antibody constructs can mas. N Engl J Med 2017;377:2545–54.
mediate bystander tumor cell killing. PLoS One 2017;12:e0183390. 46. Neelapu SS, Locke FL, Bartlett NL, Lekakis LJ, Miklos DB, Jacobson
29. Lanitis E, Poussin M, Hagemann IS, Coukos G, Sandaltzopoulos R, CA, et  al. Axicabtagene ciloleucel cAR T-cell therapy in refractory
Scholler N, et al. Redirected antitumor activity of primary human large B-cell lymphoma. N Engl J Med 2017;377:2531–44.
lymphocytes transduced with a fully human anti-mesothelin chi- 47. Kalos M, Levine BL, Porter DL, Katz S, Grupp SA, Bagg A, et al. T
meric receptor. Mol Ther 2012;20:633–43. cells with chimeric antigen receptors have potent antitumor effects
30. Henderson MA, Yong CS, Duong CP, Davenport AJ, John LB, and can establish memory in patients with advanced leukemia. Sci
Devaud C, et al. Chimeric antigen receptor-redirected T cells display Transl Med 2011;3:95ra73.
multifunctional capacity and enhanced tumor-specific cytokine 48. Turtle CJ, Hay KA, Hanafi LA, Li D, Cherian S, Chen X, et al. Dura-
secretion upon secondary ligation of chimeric receptor. Immuno- ble molecular remissions in chronic lymphocytic leukemia treated
therapy 2013;5:577–90. with CD19-specific chimeric antigen receptor-modified T cells after
31. Grupp SA, Kalos M, Barrett D, Aplenc R, Porter DL, Rheingold SR, failure of ibrutinib. J Clin Oncol 2017;35:3010–20.
et al. Chimeric antigen receptor-modified T cells for acute lymphoid 49. Maude SL, Frey N, Shaw PA, Aplenc R, Barrett DM, Bunin NJ, et al.
leukemia. N Engl J Med 2013;368:1509–18. Chimeric antigen receptor T cells for sustained remissions in leuke-
32. Mueller KT, Maude SL, Porter DL, Frey N, Wood P, Han X, et  al. mia. N Engl J Med 2014;371:1507–17.
Cellular kinetics of CTL019 in relapsed/refractory B-cell acute 50. Maude SL, Laetsch TW, Buechner J, Rives S, Boyer M, Bittencourt
lymphoblastic leukemia and chronic lymphocytic leukemia. Blood H, et  al. Tisagenlecleucel in children and young adults with B-cell
2017;130:2317–25. lymphoblastic leukemia. N Engl J Med 2018;378:439–48.
33. Porter DL, Levine BL, Kalos M, Bagg A, June CH. Chimeric antigen 51. Brentjens RJ, Riviere I, Park JH, Davila ML, Wang X, Stefanski J,
receptor-modified T cells in chronic lymphoid leukemia. N Engl J et  al. Safety and persistence of adoptively transferred autologous
Med 2011;365:725–33. CD19-targeted T cells in patients with relapsed or chemotherapy
34. Brentjens RJ, Davila ML, Riviere I, Park J, Wang X, Cowell LG, et al. refractory B-cell leukemias. Blood 2011;118:4817–28.
CD19-targeted T cells rapidly induce molecular remissions in adults 52. Fry TJ, Shah NN, Orentas RJ, Stetler-Stevenson M, Yuan CM,
with chemotherapy-refractory acute lymphoblastic leukemia. Sci Ramakrishna S, et al. CD22-targeted CAR T cells induce remission
Transl Med 2013;5:177ra38. in B-ALL that is naive or resistant to CD19-targeted CAR immuno-
35. Kochenderfer JN, Dudley ME, Carpenter RO, Kassim SH, Rose therapy. Nat Med 2018;24:20–8.
JJ, Telford WG, et  al. Donor-derived CD19-targeted T cells cause 53. Till BG, Jensen MC, Wang J, Qian X, Gopal AK, Maloney DG, et al.
regression of malignancy persisting after allogeneic hematopoietic CD20-specific adoptive immunotherapy for lymphoma using a chi-
stem cell transplantation. Blood 2013;122:4129–39. meric antigen receptor with both CD28 and 4-1BB domains: pilot
36. Topp MS, Kufer P, Gokbuget N, Goebeler M, Klinger M, Neumann clinical trial results. Blood 2012;119:3940–50.
S, et  al. Targeted therapy with the T-cell-engaging antibody blina- 54. Ali SA, Shi V, Maric I, Wang M, Stroncek DF, Rose JJ, et  al. T
tumomab of chemotherapy-refractory minimal residual disease in cells expressing an anti-B-cell maturation antigen chimeric antigen
B-lineage acute lymphoblastic leukemia patients results in high receptor cause remissions of multiple myeloma. Blood 2016;128:
response rate and prolonged leukemia-free survival. J Clin Oncol 1688–700.
2011;29:2493–8. 55. Ritchie DS, Neeson PJ, Khot A, Peinert S, Tai T, Tainton K, et  al.
37. Nagele V, Kratzer A, Zugmaier G, Holland C, Hijazi Y, Topp MS, Persistence and efficacy of second generation CAR T cell against the
et  al. Changes in clinical laboratory parameters and pharmacody- LeY antigen in acute myeloid leukemia. Mol Ther 2013;21:2122–9.
namic markers in response to blinatumomab treatment of patients 56. Kershaw MH, Westwood JA, Darcy PK. Gene-engineered T cells for
with relapsed/refractory ALL. Exp Hematol Oncol 2017;6:14. cancer therapy. Nat Rev Cancer 2013;13:525–41.
38. Kershaw MH, Westwood JA, Parker LL, Wang G, Eshhar Z, Mavrouka- 57. Slaney CY, Kershaw MH, Darcy PK. Trafficking of T cells into
kis SA, et al. A phase I study on adoptive immunotherapy using gene- tumors. Cancer Res 2014;74:7168–74.
modified T cells for ovarian cancer. Clin Cancer Res 2006;12:6106–15. 58. Louis CU, Savoldo B, Dotti G, Pule M, Yvon E, Myers GD, et al. Anti-
39. Feng K, Liu Y, Guo Y, Qiu J, Wu Z, Dai H, et al. Phase I study of chi- tumor activity and long-term fate of chimeric antigen receptor-posi-
meric antigen receptor modified T cells in treating HER2-positive tive T cells in patients with neuroblastoma. Blood 2011;118:6050–6.
advanced biliary tract cancers and pancreatic cancers. Protein Cell 59. Ahmed N, Brawley V, Hegde M, Bielamowicz K, Kalra M, Landi D,
2017 [Epub ahead of print]. et al. HER2-specific chimeric antigen receptor-modified virus-specific
40. Ahmed N, Brawley VS, Hegde M, Robertson C, Ghazi A, Gerken C, T cells for progressive glioblastoma: a phase 1 dose-escalation trial.
et  al. Human epidermal growth factor receptor 2 (HER2) -specific JAMA Oncol 2017;3:1094–101.

932 | CANCER DISCOVERY August 2018 www.aacrjournals.org

Downloaded from cancerdiscovery.aacrjournals.org on July 12, 2020. © 2018 American Association for Cancer
Research.
Published OnlineFirst July 16, 2018; DOI: 10.1158/2159-8290.CD-18-0297

CAR T-cell and BiTE Therapies for Cancers REVIEW

60. O’Rourke DM, Nasrallah MP, Desai A, Melenhorst JJ, Mansfield ronment of hodgkin lymphoma using chimeric antigen receptor T
K, Morrissette JJD, et  al. A single dose of peripherally infused cells. Cancer Discov 2017;7:1154–67.
EGFRvIII-directed CAR T cells mediates antigen loss and induces 78. Kakarla S, Chow KK, Mata M, Shaffer DR, Song XT, Wu MF, et al.
adaptive resistance in patients with recurrent glioblastoma. Sci Antitumor effects of chimeric receptor engineered human T cells
Transl Med 2017;9:eaaa0984. directed to tumor stroma. Mol Ther 2013;21:1611–20.
61. Brown CE, Alizadeh D, Starr R, Weng L, Wagner JR, Naranjo A, et al. 79. Beavis PA, Henderson MA, Giuffrida L, Mills JK, Sek K, Cross RS,
Regression of glioblastoma after chimeric antigen receptor T-cell et al. Targeting the adenosine 2A receptor enhances chimeric anti-
therapy. N Engl J Med 2016;375:2561–9. gen receptor T cell efficacy. J Clin Invest 2017;127:929–41.
62. Martinelli G, Boissel N, Chevallier P, Ottmann O, Gokbuget N, 80. Newick K, O’Brien S, Sun J, Kapoor V, Maceyko S, Lo A, et al. Augmen-
Topp MS, et  al. Complete hematologic and molecular response in tation of CAR T-cell trafficking and antitumor efficacy by blocking
adult patients with relapsed/refractory Philadelphia chromosome- protein kinase a localization. Cancer Immunol Res 2016;4:541–51.
positive B-precursor acute lymphoblastic leukemia following treat- 81. John LB, Devaud C, Duong CP, Yong CS, Beavis PA, Haynes NM,
ment with blinatumomab: results from a phase II, single-arm, et al. Anti-PD-1 antibody therapy potently enhances the eradication
multicenter study. J Clin Oncol 2017;35:1795–802. of established tumors by gene-modified T Cells. Clin Cancer Res
63. Kantarjian H, Stein A, Gokbuget N, Fielding AK, Schuh AC, Ribera 2013;19:5636–46.
JM, et  al. Blinatumomab versus chemotherapy for advanced acute 82. Zhang Y, Zhang X, Cheng C, Mu W, Liu X, Li N, et al. CRISPR-Cas9
lymphoblastic leukemia. N Engl J Med 2017;376:836–47. mediated LAG-3 disruption in CAR-T cells. Front Med 2017;11:554–62.
64. Goebeler ME, Knop S, Viardot A, Kufer P, Topp MS, Einsele H, 83. Shum T, Kruse RL, Rooney CM. Strategies for enhancing adop-
et  al. Bispecific T-Cell Engager (BiTE) antibody construct blinatu- tive T-cell immunotherapy against solid tumors using engineered
momab for the treatment of patients with relapsed/refractory non- cytokine signaling and other modalities. Expert Opin Biol Ther
Hodgkin lymphoma: final results from a phase I study. J Clin Oncol 2018;18:653–64.
2016;34:1104–11. 84. Koneru M, Purdon TJ, Spriggs D, Koneru S, Brentjens RJ. IL-12
65. Fiedler WM, Wolf M, Kebenko M, Goebeler M-E, Ritter B, Quaas A, secreting tumor-targeted chimeric antigen receptor T cells eradicate
et al. A phase I study of EpCAM/CD3-bispecific antibody (MT110) ovarian tumors in vivo. Oncoimmunology 2015;4:e994446.
in patients with advanced solid tumors. J Clin Oncol 30:15, 2012 85. Shum T, Omer B, Tashiro H, Kruse RL, Wagner DL, Parikh K, et al. constitu-
(suppl; abstr 2504). tive signaling from an engineered IL7 receptor promotes durable tumor
66. Bi Y, Jiang H, Wang P, Song B, Wang H, Kong X, et  al. Treatment elimination by tumor-redirected T cells. Cancer Discov 2017;7:1238–47.
of hepatocellular carcinoma with a GPC3-targeted bispecific T cell 86. Leen AM, Sukumaran S, Watanabe N, Mohammed S, Keirnan J,
engager. Oncotarget 2017;8:52866–76. Yanagisawa R, et al. Reversal of tumor immune inhibition using a
67. Teachey DT, Lacey SF, Shaw PA, Melenhorst JJ, Maude SL, Frey N, chimeric cytokine receptor. Mol Ther 2014;22:1211–20.
et  al. Identification of predictive biomarkers for cytokine release 87. Mohammed S, Sukumaran S, Bajgain P, Watanabe N, Heslop HE,
syndrome after chimeric antigen receptor T-cell therapy for acute Rooney CM, et al. Improving chimeric antigen receptor-modified T
lymphoblastic leukemia. Cancer Discov 2016;6:664–79. cell function by reversing the immunosuppressive tumor microen-
68. Neelapu SS, Tummala S, Kebriaei P, Wierda W, Gutierrez C, Locke vironment of pancreatic cancer. Mol Ther 2017;25:249–58.
FL, et al. Chimeric antigen receptor T-cell therapy - assessment and 88. Moon EK, Carpenito C, Sun J, Wang LC, Kapoor V, Predina J, et al.
management of toxicities. Nat Rev Clin Oncol 2018;15:47–62. Expression of a functional CCR2 receptor enhances tumor localiza-
69. Gust J, Hay KA, Hanafi LA, Li D, Myerson D, Gonzalez-Cuyar LF, tion and tumor eradication by retargeted human T cells expressing
et  al. Endothelial activation and blood-brain barrier disruption in a mesothelin-specific chimeric antibody receptor. Clin Cancer Res
neurotoxicity after adoptive immunotherapy with CD19 CAR-T 2011;17:4719–30.
Cells. Cancer Discov 2017;7:1404–19. 89. Chinnasamy D, Yu Z, Kerkar SP, Zhang L, Morgan RA, Restifo NP,
70. Hill JA, Li D, Hay KA, Green ML, Cherian S, Chen X, et  al. Infec- et al. Local delivery of interleukin-12 using T cells targeting VEGF
tious complications of CD19-targeted chimeric antigen receptor- receptor-2 eradicates multiple vascularized tumors in mice. Clin
modified T-cell immunotherapy. Blood 2018;131:121–30. Cancer Res 2012;18:1672–83.
71. Topp MS, Gokbuget N, Stein AS, Zugmaier G, O’Brien S, Bargou RC, 90. Fu X, Rivera A, Tao L, Zhang X. Genetically modified T cells target-
et  al. Safety and activity of blinatumomab for adult patients with ing neovasculature efficiently destroy tumor blood vessels, shrink
relapsed or refractory B-precursor acute lymphoblastic leukaemia: a established solid tumors and increase nanoparticle delivery. Int J
multicentre, single-arm, phase 2 study. Lancet Oncol 2015;16:57–66. Cancer 2013;133:2483–92.
72. Brandl C, Haas C, d’Argouges S, Fisch T, Kufer P, Brischwein K, 91. Zhu M, Wu B, Brandl C, Johnson J, Wolf A, Chow A, et al. Blinatu-
et  al. The effect of dexamethasone on polyclonal T cell activation momab, a bispecific T-cell engager (BiTE(®)) for CD-19 targeted
and redirected target cell lysis as induced by a CD19/CD3-bispecific cancer immunotherapy: clinical pharmacology and its implications.
single-chain antibody construct. Cancer Immunol Immunother Clin Pharmacokinet 2016;55:1271–88.
2007;56:1551–63. 92. Liu H, Saxena A, Sidhu SS, Wu D. Fc engineering for developing
73. Binnewies M, Roberts EW, Kersten K, Chan V, Fearon DF, Merad M, therapeutic bispecific antibodies and novel scaffolds. Front Immunol
et al. Understanding the tumor immune microenvironment (TIME) 2017;8:38.
for effective therapy. Nat Med 2018;24:541–50. 93. Warnders FJ, Waaijer SJ, Pool M, Lub-de Hooge MN, Friedrich M,
74. Moon EK, Wang LC, Dolfi DV, Wilson CB, Ranganathan R, Sun J, Terwisscha van Scheltinga AG, et al. Biodistribution and PET imag-
et al. Multifactorial T-cell hypofunction that is reversible can limit ing of labeled bispecific T cell-engaging antibody targeting EpCAM.
the efficacy of chimeric antigen receptor-transduced human T cells J Nucl Med 2016;57:812–7.
in solid tumors. Clin Cancer Res 2014;20:4262–73. 94. Whiteside TL. The tumor microenvironment and its role in promot-
75. Highfill SL, Long AH, Orentas RJ, Mackall CL. Neutralization of ing tumor growth. Oncogene 2008;27:5904.
murine myeloid-derived suppressor cells enhances the efficacy of 95. Iwahori K, Kakarla S, Velasquez MP, Yu F, Yi Z, Gerken C, et  al.
a chimeric antigen receptor T-cells directed against pediatric solid Engager T cells: a new class of antigen-specific T cells that redirect
tumors. J Immunother Cancer 2013;1:P265. bystander T cells. Mol Ther 2015;23:171–8.
76. Burga RA, Thorn M, Point GR, Guha P, Nguyen CT, Licata LA, et al. 96. Bonifant CL, Szoor A, Torres D, Joseph N, Velasquez MP, Iwahori
Liver myeloid-derived suppressor cells expand in response to liver K, et al. CD123-Engager T cells as a novel immunotherapeutic for
metastases in mice and inhibit the anti-tumor efficacy of anti-CEA acute myeloid leukemia. Mol Ther 2016;24:1615–26.
CAR-T. Cancer Immunol Immunother 2015;64:817–29. 97. Velasquez MP, Torres D, Iwahori K, Kakarla S, Arber C, Rodriguez-Cruz
77. Ruella M, Klichinsky M, Kenderian SS, Shestova O, Ziober A, Kraft T, et  al. T cells expressing CD19-specific engager molecules for the
DO, et  al. Overcoming the immunosuppressive tumor microenvi- immunotherapy of CD19-positive malignancies. Sci Rep 2016;6:27130.

August 2018 CANCER DISCOVERY | 933

Downloaded from cancerdiscovery.aacrjournals.org on July 12, 2020. © 2018 American Association for Cancer
Research.
Published OnlineFirst July 16, 2018; DOI: 10.1158/2159-8290.CD-18-0297

REVIEW Slaney et al.

98. Liu X, Barrett DM, Jiang S, Fang C, Kalos M, Grupp SA, et al. Improved novel CD33/CD3 bispecific T-cell engager (BiTE) antibody, AMG
anti-leukemia activities of adoptively transferred T cells expressing 330, against human AML. Blood 2014;123:554–61.
bispecific T-cell engager in mice. Blood Cancer J 2016;6:e430. 106. Hoseini SS, Dobrenkov K, Pankov D, Xu XL, Cheung NK. Bispecific
99. Fajardo CA, Guedan S, Rojas LA, Moreno R, Arias-Badia M, de Sos- antibody does not induce T-cell death mediated by chimeric anti-
toa J, et al. Oncolytic adenoviral delivery of an EGFR-Targeting T-cell gen receptor against disialoganglioside GD2. Oncoimmunology
engager improves antitumor efficacy. Cancer Res 2017;77:2052–63. 2017;6:e1320625.
100. Freedman JD, Hagel J, Scott EM, Psallidas I, Gupta A, Spiers L, et al. 107. Stone JD, Aggen DH, Schietinger A, Schreiber H, Kranz DM. A
Oncolytic adenovirus expressing bispecific antibody targets T-cell sensitivity scale for targeting T cells with chimeric antigen receptors
cytotoxicity in cancer biopsies. EMBO Mol Med 2017;9:1067–87. (CARs) and bispecific T-cell Engagers (BiTEs). Oncoimmunology
101. Yu F, Wang X, Guo ZS, Bartlett DL, Gottschalk SM, Song XT. T-cell 2012;1:863–73.
engager-armed oncolytic vaccinia virus significantly enhances anti- 108. Amann M, Brischwein K, Lutterbuese P, Parr L, Petersen L, Lor-
tumor therapy. Mol Ther 2014;22:102–11. enczewski G, et al. Therapeutic window of MuS110, a single-chain
102. Kohnke T, Krupka C, Tischer J, Knosel T, Subklewe M. Increase of antibody construct bispecific for murine EpCAM and murine CD3.
PD-L1 expressing B-precursor ALL cells in a patient resistant to Cancer Res 2008;68:143–51.
the CD19/CD3-bispecific T cell engager antibody blinatumomab. 109. Schlereth B, Kleindienst P, Fichtner I, Lorenczewski G, Brischwein
J Hematol Oncol 2015;8:111. K, Lippold S, et  al. Potent inhibition of local and disseminated
103. Krupka C, Kufer P, Kischel R, Zugmaier G, Lichtenegger FS, Kohnke T, tumor growth in immunocompetent mouse models by a bispecific
et al. Blockade of the PD-1/PD-L1 axis augments lysis of AML cells by antibody construct specific for Murine CD3. Cancer Immunol
the CD33/CD3 BiTE antibody construct AMG 330: reversing a T-cell- Immunother 2006;55:785–96.
induced immune escape mechanism. Leukemia 2016;30:484–91. 110. Wu MR, Zhang T, Gacerez AT, Coupet TA, DeMars LR, Sentman CL.
104. Zhang P, Shi B, Gao H, Jiang H, Kong J, Yan J, et al. An EpCAM/CD3 B7H6-Specific bispecific T cell engagers lead to tumor elimination
bispecific antibody efficiently eliminates hepatocellular carcinoma and host antitumor immunity. J Immunol 2015;194:5305–11.
cells with limited galectin-1 expression. Cancer Immunol Immu- 111. Wing A, Fajardo CA, Posey AD Jr., Shaw C, Da T, Young RM, et al.
nother 2014;63:121–32. Improving CART-cell therapy of solid tumors with oncolytic virus-
105. Laszlo GS, Gudgeon CJ, Harrington KH, Dell’Aringa J, Newhall KJ, driven production of a bispecific T-cell engager. Cancer Immunol
Means GD, et al. Cellular determinants for preclinical activity of a Res 2018;6:605–16.

934 | CANCER DISCOVERY August 2018 www.aacrjournals.org

Downloaded from cancerdiscovery.aacrjournals.org on July 12, 2020. © 2018 American Association for Cancer
Research.
Published OnlineFirst July 16, 2018; DOI: 10.1158/2159-8290.CD-18-0297

CARs versus BiTEs: A Comparison between T Cell−Redirection


Strategies for Cancer Treatment
Clare Y. Slaney, Pin Wang, Phillip K. Darcy, et al.

Cancer Discov 2018;8:924-934. Published OnlineFirst July 16, 2018.

Updated version Access the most recent version of this article at:
doi:10.1158/2159-8290.CD-18-0297

Cited articles This article cites 108 articles, 43 of which you can access for free at:
http://cancerdiscovery.aacrjournals.org/content/8/8/924.full#ref-list-1

Citing articles This article has been cited by 5 HighWire-hosted articles. Access the articles at:
http://cancerdiscovery.aacrjournals.org/content/8/8/924.full#related-urls

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications
Subscriptions Department at pubs@aacr.org.

Permissions To request permission to re-use all or part of this article, use this link
http://cancerdiscovery.aacrjournals.org/content/8/8/924.
Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)
Rightslink site.

Downloaded from cancerdiscovery.aacrjournals.org on July 12, 2020. © 2018 American Association for Cancer
Research.

You might also like