Vous êtes sur la page 1sur 11

Review

Special Focus Issue: Adoptive cell immunotherapy for cancer


For reprint orders, please contact: reprints@futuremedicine.com

CAR T-cell therapy: toxicity and the


relevance of preclinical models

Chimeric antigen receptor (CAR) T cells form part of a broad wave of immunotherapies Milena Kalaitsidou1, Gray
that are showing promise in early phase cancer clinical trials. This clinical delivery has Kueberuwa1, Antje Schütt1
been based upon preclinical efficacy testing that confirmed the proof of principle of & David Edward Gilham*,1
1
Clinical & Experimental Immunotherapy
the therapy. However, CAR T-cell therapy does not exist alone as T cells are generally
Group, Institute of Cancer Sciences,
given in combination with patient preconditioning, most commonly in the form of Academic Healthcare Science Centre,
chemotherapy, and may also include systemic cytokine support, both of which are University of Manchester, Manchester
associated with toxicity. Consequently, complete CAR T-cell therapy includes elements Paterson Building, Wilmslow Road,
where the toxicity profile is well known, but also includes the CAR T cell itself, for Withington, Manchester, M20 4BX, UK
*Author for correspondence:
which toxicity profiles are largely unknown. With recent reports of adverse events
Tel.: +44 161 446 3236
associated with CAR T-cell therapy, there is now concern that current preclinical Fax: +44 161 446 3236
models may not be fit for purpose with respect to CAR T-cell toxicity profiling. Here, David.Gilham@ ics.manchester.ac.uk
we explore the preclinical models used to validate CAR T-cell function and examine
their potential to predict CAR T-cell driven toxicities for the future.

Keywords:  adoptive cell therapy • CAR T cells • cytokine storm • preclinical models • toxicity

Chimeric antigen receptor (CAR, also known the wide number of excellent recent reviews
as chimeric immune receptor [CIR] and detailing CAR structures and issues relating
T body) T-cell technology has developed rap- to the clinical delivery of this therapy [4–10] . In
idly over the last 20 years. The initial concept brief here, to provide an overview of the diver-
featured the fusion of T-cell signaling proteins, sity of CARs, the modular nature of CARs has
including the T-cell receptor β chain, CD3γ encouraged the incorporation of multiple sig-
and CD3ζ with target-binding domains. naling domains to form hybrids that increase
When expressed in a T cell, these fusion pro- downstream signaling potency (Figure 1).
teins result in T-cell effector functions being Hence, CARs containing two signaling
controlled by the ligand binding specificity of domains fused together are termed ‘second
the target-binding domain [1] . Over time, this generation,’ while those bearing three are listed
linear arrangement of targeting domain fused as ‘third generation.’ The combinations of sig-
to one signaling domain has been termed ‘first- naling domains vary, however, for the majority,
generation’ CARs. Upon this basic structure, the cytosolic sequence from CD3ζ protein is
a diverse range of CARs have been developed included to provide the main T-cell activating
and shown to function. The target-binding signal. Recently developed ‘fourth-generation’
domain most commonly used involves single- CARs (or TRUCKs) involve two separate
chain antibody variable fragments (scFvs) transgenes with a first, second- or third-gener-
derived from monoclonal antibody hybrid- ation CAR expressed alongside a T-cell activa-
omas or by phage display technology. Though tion responsive promoter linked to a cytokine
less common, intact extracellular protein such as IL-12. Consequently, upon T-cell acti-
domains and receptor ligands have also been vation, high levels of IL-12 are produced that
successfully used in the CAR context [2,3] . The can modulate the local microenvironment and
part of
explosion of interest in CARs is highlighted by aid CAR T-cell function [11] .

10.2217/IMT.14.123 © 2015 Future Medicine Ltd Immunotherapy (2015) 7(5), 487–497 ISSN 1750-743X 487
Review  Kalaitsidou, Kueberuwa, Schütt & Gilham

Antibody CARs TRUCKs

Fab 1st 2nd 3rd 4th

ScFv

Spacer

Transmembrane domain
CD3ζ CD28/ CD28
Signaling domain
4-1BB
4-1BB
CD3ζ OX-40 NFAT
CD3ζ IL-12
(IL-2)
Cytotoxicity (IL-8)
IL-12
Proliferation, persistence
NFAT min. prom.
Survival

Figure 1. Chimeric antigen receptors. First-generation CARs contain solely the CD3ζ chain as a single activation
domain, leading to T-cell activation and cytotoxicity based on scFv specificity. Second- and third-generation
CARs consist of one or two additional costimulatory signaling domains, respectively, such as CD28, CD27, OX-40
(CD134) and 4-1BB (CD137). Costimulation enhances the overall survival as well as proliferation and persistence of
activated T cells. TRUCKs represent the fourth generation and are currently the newest approach in adoptive T-cell
therapy. T cells are genetically modified to express CARs along with an inducible cytokine gene cassette driven
by an NFAT sensitive promoter. Consequently, immune stimulatory cytokines such as IL-12 are secreted upon CAR
engagement.
CAR: Chimeric antigen receptor; min. prom.: Minimum promoter. 

Against this background of CAR structural diver- involve direct targeting of the cytokine action by
sity, there is no current consensus on the optimal com- blocking access to their receptors (i.e., tocilizumab)
bination of signaling domains and also no obvious or indirect reduction of cytokines by removing the
rules that define the extracellular domains upon which transduced cells via lymphodepletion using cortico­
target binding by scFv is dependent. Consequently, steroids  [14] . Similar management strategies for cyto­
clinical studies arising from this diversity have pro- kine-release syndrome are reported in acute B cell lym-
ceeded in a largely empirical manner, with individual phoblastic leukemia patients receiving CD28.CD3ζ
research groups using locally developed CARs with CAR T cells [15,16] . In the very worst-case scenario, tar-
differing targeting moieties against varying diseases, geting of normal, essential tissue through the CAR can
associated with different levels of patient precondition- result in rapid autotoxicity and can lead to death. This
ing and using T cells generated using different ex vivo has occurred in a colorectal cancer patient with lung
systems  [12] . Despite this, objective clinical responses and liver metastases who received treatment with a
have been reported in several centers, predominantly Her2/neu-specific ‘third-generation’ CAR T cells [17] .
targeting B-cell leukemia through the CD19 antigen This highlights the fine line between achieving a clini-
(see  [8] for recent review), and this is driving a major cal anticancer response and preventing autotoxicity as
surge of interest and activity in CAR T-cell therapy [13] . a result of CAR T-cell therapy. Antitumor activity of
Few, if any, therapies deliver clinical responses in CAR T cells results in the production of cytokines that
the absence of some form of toxicity and CAR T-cell are advantageous through the direct killing of target
therapy is no exception to this. The clinical success of cells and recruiting other elements of the immune sys-
targeting the CD19 antigen with ‘second-generation’ tem to elicit an enhanced antitumor response. How-
4-1BB.CD3ζ CARs (CART019) is associated with ever, ensuring that the magnitude of response does
cytokine-release syndrome (CRS), in which IL-6, not exceed the threshold for causing systemic toxicity
IL-10 and IFN-γ play a major role, that is potentially and that CAR T cells are only targeted to tumor and
life-threatening if not appropriately managed [14] . not healthy tissue, remain major hurdles for clinicians
Examples of main strategies used to overcome CRS using CAR T cells. Going forward, will it be possible

488 Immunotherapy (2015) 7(5) future science group


CAR T-cell therapy: toxicity & the relevance of preclinical models  Review

to identify such potential toxicities in the preclinical Moreover, the majority of these studies involve CAR
setting that will then allow further refinement of the T-cells targeting human antigens whose expression is
CAR strategy to enhance efficacy in the absence of restricted to the human tumor cells used to challenge
autotoxicity? mice in the model. Thus, targeting of the antigen which
may be expressed on normal, healthy tissue cannot be
Getting CAR T cells to the clinic: preclinical evaluated. To examine this, immune-competent mouse
models to deliver the proof of principle models have been used where the target antigen is the
The majority of preclinical studies investigating CAR mouse homologue of the human target antigen [25] .
T-cell function to date have focused upon verifying While these models provide an array of additional infor-
specificity and potency of antitumor activity. Speci- mation about CAR T-cell function with background
ficity has been generally confirmed by in vitro assays antigen expression and a functional immune system,
establishing that the T cells endowed with a specific ‘mouse CAR-mouse homologue’ interactions may dif-
CAR show a differential functional response between fer from the equivalent human scenario. In addition,
the target antigen (either in purified format or on rel- this type of model is dependent upon minimal disparity
evant target cells) and nonspecific antigens. The key between expression profiles of mouse and human pro-
advantage of the CAR is the ability to redirect T-cell tein homologues. Another approach is to utilize trans-
effector function in the absence of HLA-restriction. genic animals expressing the human antigen under the
Consequently, allogeneic T-cells can be readily used to control of a physiologically relevant promoter where the
demonstrate this in vitro specificity without the need CAR specific for the human antigen can be tested [26] .
for matched targets. Background activity, which is These combined approaches have been used to suc-
the result of allo-recognition, is generally equivalent cessfully demonstrate CAR T-cell function against
between cells transduced with irrelevant CARs and a wide diversity of target antigens, with the growing
nontransduced T cells. This is typically measurable interest in CAR T-cell approaches confirmed by the
in vitro and the benefit of CARs is seen as fold increase rapidly rising number of publications specifically citing
above this background. Such assays prove CAR func- CARs (Figure 2) .
tion and support translation to the animal model sys-
tem. It should be noted that background allo-recogni- Potential mechanisms of
tion has not been associated with antitumor activity CAR T-cell-mediated toxicity
in vivo in a large number of xenograft tumor studies. While the complete CAR T-cell therapy involves
For clinical translation, regulatory authorities administration of agents that have well described tox-
generally require information relating to the specific icity profiles, toxicities arising from CAR T cells them-
agent to be used in the clinical trial. In the case of selves are not as well defined. There are three poten-
CAR T cells, this means that testing in immune-com- tially key routes by which CAR T cells may contribute
promised mouse models represent the only possible to toxicity and must be considered (Figure 3) .
route to assess the actual clinical agent. However, the
success of human T-cell engraftment in the majority On-target, on-tumor toxicity
of immune-compromised mice is limited since the On target, on tumor toxicities are generally acute and
residual elements of the mouse immune system still relate directly to the effects of binding of the CAR to
challenge the engrafting human T cells. Nonethe- cognate antigen resident on the target tumor cell. The
less, several studies have shown efficacy of CAR T-cell underlying premise of the therapy is the activation of
function using Nude, NOD/SCID or SCID/Beige T-cell effector functions as a result of successful CAR
animals (for examples see [18–21]). More recently, engagement, including cytokine release and cyto­
the availability of the highly immune-compromised toxicity. However, excessive cytokine production may
NOG/NSG mouse (NOD/SCID IL-2Rγ-/-) has result in cytokine release syndrome typified by chills
allowed efficient human T-cell engraftment. However, and fevers but can also result in much more severe,
engrafted T cells can drive a xeno-graft versus host potentially life threatening reactions [27] . Moreover,
disease (xGVHD) which occurs around 50 days after the rapid destruction of large quantities of tumor can
adoptive T-cell transfer at doses above 109 cells/kg [22] . result in tumor lysis syndrome due to the abrupt release
The occurrence of xGVHD is associated with trans- of tumor cellular contents causing an array of systemic
duced cell persistence and this is affected by the cyto- metabolic disturbances [28] .
kine conditions used to culture the cells prior to injec-
tion into mice. Consequently, this limits the ability On-target, off-tumor toxicity
to investigate the long-term effects of CAR T cells in In this situation, the CAR T-cell engages the target
this model [22–24] . antigen that is expressed upon healthy, nontumor cells.

future science group www.futuremedicine.com 489


Review  Kalaitsidou, Kueberuwa, Schütt & Gilham

B cells. Successful targeting of CD19 by CAR T cells


100
leads to ‘on-target, off-tumor’ aplasia of developing
Number of Publications 80
B cells but spares CD19-negative plasma cells and,
crucially, hematopoietic stem cells, allowing repletion
60 of the B-cell compartment post-CAR T-cell therapy.
This B-cell aplasia may be an acceptable side effect
40 due to clinical management strategies that are avail-
able. Indeed, B-cell aplasia has been used as surrogate
20 readout for CD19 CAR T-cell activity in preclinical
models  [29] . Moreover, the use of lymphotoxic steroids
0 can eradicate CAR T cells and allow B-cell repletion
1980

1990

2000

2010
if required [30] .
However, in the case of many solid tumors, the CAR
Year
target may not be tumor specific but a tumor-associ-
Figure 2. In vivo application of chimeric antigen
ated antigen (TAA) where expression of the target anti-
receptors. Timeline showing the number of publications gen is high on tumors but may be present at low levels
citing chimeric antigen receptors utilized between 1980 on healthy tissue. In this case, CAR T cells may target
and 2014, highlighting an intensive increase in interest and respond to such healthy tissue, and where this tis-
in the past 5 years (search terms: ‘chimeric antigen sue resides in an organ essential for life, catastrophic
receptor’ or ‘chimeric immunoreceptor’ or ‘chimeric
T-cell receptor’ or ‘chimeric immune receptor’).
consequences may occur [31] . With these toxicities in
mind, target antigen selection is probably the most crit-
Such targeting can result in the destruction of healthy ical determinant to successful CAR therapy. Antigens
cells expressing the specific target antigen. In the case with strict tumor specificity are ideal (e.g., EphA2 [32] ,
of B-cell malignancies, the CD19 antigen is expressed mutated EGFRvIII [33,34] and ‘unmasked’ Mucin-1
on cells of B-cell lineage from pro-B cells to mature with truncated glycosylations [35]), or at least where

T cell 2 On-target off-tumor


T cell
T cell T cell Healthy tissue
T cell
T cellT cell T cell
Nontumor
T cellT cell tissue
T cell

Excessive CAR-T-cell activation


• Cytokine storm

T cell 3 Off-target off-tumor


1 On-target on-tumor

Tumor lysis syndrome Nonspecific CAR Activation of


• Tumor-derived toxicity affinity endogenous MDSCs
TCRs

Nontumor
tissue
MDSC
Tumor

Figure 3. Categorization of chimeric antigen receptors toxicities. Once T cells are activated their cytotoxic potential can lead to
undesired side effects. (1) Even in the optimal scenario of specific activation of CAR T cells via their target expressed on tumor tissue
can lead to toxicity. This is termed on-target on-tumor toxicity and may be due to excessive T-cell activation with (A) a Th1 dominated
cytokine storm or (B) due to a very high tumor load leading to massive destruction of tumor tissue, resulting in tumor lysis syndrome.
(2) In cases where the target is also expressed on normal tissue it is likely that on-target off-tumor toxicity will occur, subsequently
damaging healthy tissue. (3) Additionally, there is a possibility of off-target off-tumor toxicity. This can occur through, (C) CAR T-cell
activation by similar but distinct epitopes or the endogenous T-cell receptor engagement, or (D) Th2 cytokine secreting MDSCs
associated with long-term engraftment of CAR T cells.
CAR: Chimeric antigen receptor; MDSC: Myeloid-derived suppressor cell; TCR: T-cell receptor.

490 Immunotherapy (2015) 7(5) future science group


CAR T-cell therapy: toxicity & the relevance of preclinical models  Review

background antigen expression is on nonessential tis- but are likely to include factors such as differences in
sues (e.g., prostate-specific antigen, NY-ESO [36] , CAR structures, T-cell biology between the mouse
MAGE  [37,38] , CD19 [39] and CD20 [40]). In practice, and human and differential drug metabolism capacity
however, such antigens have been difficult to identify, between species [46] . However, while these observations
particularly in the setting of solid malignancies. Given raise questions concerning the ability of current model
a known background expression of a target antigen it systems to predict toxicities, there have been recent
becomes paramount to determine whether levels are reports of toxicity in preclinical models systems in
over a threshold that can cause ‘on-target, off-tumor’ three specific CAR T-cell experimental investigations
toxicity, and to determine the potential severity thereof that illustrate the mechanisms of CAR T-cell toxicity.
in humans.
On target off-tumor toxicity resulting from
Off target, off tumor toxicity ‘atypical’ CAR targeting domains driving
This third potential mechanism of CAR T-cell toxicity toxicity through the recognition of multiple
has arisen in experimental models of CAR T-cell ther- target antigens present on healthy tissue
apy recently and relates to the response of non-CAR In contrast to the absence of toxicity observed using
T cells to the therapy [41] . Prolonged, potentially low CAR’s armed with scFv targeting Her2/neu in pre-
level, CAR T-cell activity may indirectly impact upon clinical model systems, human T cells armed with the
the local environment resulting in skewing of normal T1E28z second-generation CAR, which binds ErbB-1
homeostatic cellular responses, that in turn develop homodimers and also ErbB2/3 heterodimers, dis-
into toxicities. This mechanism of toxicity is discussed played toxicity in mice [47] . This CAR transgene was
in further detail below. co-expressed with a chimeric protein consisting of the
IL-4α chain fused to the IL-2/IL-15 β chain permit-
Genotoxicity ting IL-4-driven selection of gene-modified T cells [47] .
Integrating vectors based upon retroviral and lentiviral Interestingly, the route of administration of T1E28z
backbones have been most commonly used to facili- CAR T cells strongly influenced toxicity and antitumor
tate the stable expression in primary T cells. However, efficacy in the SCID/Beige mouse. CAR T cells admin-
such vectors may pose a potential risk of oncogenic istered by the intravenous route demonstrated reduced
events including disruption of normal gene expression tumor growth with no outward symptoms of adverse
as observed in the hematopoietic stem cell situation effects and no evidence of pathological abnormality at
with the loss of control of the LMO2 gene in X-linked necropsy 7 days post-CAR T-cell infusion [48] . How-
SCID [42] . However, thus far, no such toxicity has been ever, intraperitoneal administration of T1E28z CAR
reported in the context of CAR therapy [43] , but this is T cells to challenge local tumor xenografts resulted in
clearly an important consideration for the future where toxicities associated with a major cytokine release syn-
CAR T cells may prevail for life time of the treated drome mediated largely by human IFNγ, human IL-2
patient. and mouse IL-6 that were readily detectable within the
circulation of mice receiving a lethal dose of T1E28z
CAR T-cell toxicity in preclinical models CAR T cells [48] . Clodronate depletion of macrophages
Across the breadth of preclinical publications focusing prior to CAR T-cell transfer confirmed that these cells
upon CAR T cells, there are very few that document are the likely source of mouse IL-6 and, interestingly,
toxicity, even where toxicity has been observed in the this also reduced human IFN-γ levels. The dose and
clinic. One example involves CAR T cells targeting the volume of administered CAR T cells along with tumor
Her2/neu (ErbB2) antigen which is overexpressed on a all appeared to correlate with toxicity. The expression of
range of tumors. There are a wealth of studies docu- ErbB proteins on healthy tissue and, as the authors sug-
menting the antitumor potency of Her2/neu-specific gest, specifically upon normal mesodermal cells within
CAR T cells, including studies targeting this antigen the peritoneal cavity may also have resulted in the pro-
in Her2 transgenic animals combined with lympho­ found on-target off-tumor toxicity associated with the
depletion  [44] or anti-PD-1 antibody therapy [45] . In T1E28z CAR T cells. Nonetheless, targeting of multiple
each case, there has been no evidence of any pathol- ErbB proteins was associated with substantial antitumor
ogy associated with the Her2/neu-directed therapy, activity underscoring the potential of the approach [48] .
yet a patient receiving Her2/neu-specific CAR T cells Taken together, controlling the route of administra-
died, potentially as a result of cytokines released due tion and reducing tumor burden prior to adoptive CAR
to targeting of Her2/neu present on lung epithelial T-cell transfer would be important factors to potentially
cells [17] . The reasons for this dichotomy between pre- optimize the therapy while reducing adverse side effects.
clinical results and the patient situation are not clear Indeed, the authors of this study have designed a Phase I

future science group www.futuremedicine.com 491


Review  Kalaitsidou, Kueberuwa, Schütt & Gilham

trial protocol using intratumoral infusion of T1E28z different scFvs used between the studies may them-
CAR T cells in head and neck cancer as a practical selves endow the specific CARs with differential activi-
means to avoid such undesired toxicity [49] . ties. Consequently, there are many potential individual
and combinatorial factors that could underlie these
On-target off-tumor toxicity driven by differential observations and further study is clearly
‘standard’ CAR’s targeting rare, specialized cell required. However, the initial suggestion is that FAP
types targeted toxicity resulted from the combination of
Whilst the direct targeting of tumor cells by CAR third-generation CAR and optimal conditions to drive
T cells is the natural mechanistic choice for such a CAR T-cell engraftment. In summary, studies exam-
therapeutic approach, the inherent genetic instability ining FAP targeting highlight the potential impact of
of tumors raises the possibility that antigen-negative the full therapy and the potency of third-generation
variants may arise and allow the tumor to effectively CARs in targeting non-neoplastic, tumor associated
circumvent targeting by the CAR. Targeting of non- stromal cells [52] .
neoplastic tumor associated stromal cells potentially
avoids this issue and can be achieved through targeting Chronic off-target off-tumor toxicity resulting
antigens such as fibroblast activation protein α (FAP), from CD19 targeted CAR T cells
which is expressed on cancer associated fibroblasts. The cytokine release syndromes observed in certain
T cells armed with a ‘second-generation’ CAR with patients receiving CD19 CAR T cells [14] have not been
an FAP-specific scFv CAR showed effective antitumor exactly reproduced in mouse model systems. However,
activity against established A549 lung cancer cells in a recent study from our group has reported short-term
SCID mice [50] and FAP+HT1080 cells in the NSG Th1-biased cytokine driven toxicity in mice receiving
mouse model [51] . In the lung cancer model, on-target mouse CD19 targeted, second-generation CAR T cells
off-tumor toxicity was considered but no evidence of and also reported a chronic toxicity resulting from the
adverse pathology was observed in mice sacrificed at specific expansion of myeloid cells in response to Th2-
short time points after CAR T-cell infusion [50] . The biased cytokine production arising from chronic CD4 +
human antigen restriction of the CAR used in the CAR T-cell activity [41] . In this system, acute toxicity
mesothelioma study prevented any in depth study of manifested as rapid weight loss and cachexia with death
toxicity due to lack of background somatic expres- occurring 1–5 days after adoptive T-cell transfer. How-
sion  [51] . By contrast, mouse T cells engrafted with ever, this acute toxicity primarily occurred after the
a FAP-specific third-generation CAR (consisting of adoptive transfer of T-cells engrafted with a CAR con-
CD28, 4-1BB and CD3ζ signaling domains) drove taining the full length CD28 sequence fused to CD3ζ.
significant morbidity and mortality in recipient mice, T cells expressing CARs bearing a truncated CD28
including a drastically diminished frequency of live extracellular domain fused to CD3ζ induced a reduced
bone marrow and osteogenic cells resident within severity of weight loss and the animals survived. How-
femurs and tibia [52] . Further analysis identified a pre- ever, a significant number of animals engrafted with
viously unknown FAP+ multipotent bone marrow stem either CD28-based CAR developed symptoms includ-
cell population, which was depleted in FAP-specific ing weight loss and cachexia resulting in death from
CAR immunotherapy resulting in the lethal bone approximately 2 months after adoptive transfer. Tumor
toxicity and cachexia [52] . Subsequent to this, stud- burden was not implicated as causative as animals lack-
ies employing a second-generation FAP-specific CAR ing tumor also suffered the same fate. Upon necropsy,
reported encouraging antitumor activity against a affected animals presented with enlarged spleen and
number of tumor targets with no evidence of on-target lymph nodes filled with granuloma like cells, shown
off-tumor toxicity [53] . to consist of large numbers of CD11b + Gr-1+ myeloid
The question is ‘how such a major discrepancy is cells reminiscent of myeloid-derived suppressor cells
possible between effectively similar studies targeting (MDSC). Cytokine analysis showed increased levels of
the same antigen?’ The answer may lie in the total ther- Th2 cytokines, specifically IL-10 and IL-13 that were
apy. In the report describing toxicity, third-generation postulated to be derived from the large numbers of
CAR T cells were employed along with a total body CD4 + CAR T cells identified within secondary lym-
irradiation preconditioning regimen (5Gy) and six phoid structures. Interestingly, affected animals were
doses of systemic IL-2 support (2.2 × 105 IU/dose) [52] . essentially devoid of CD8 + CAR T cells and B cells.
By contrast, the three other publications used second- Together, this suggests that chronic stimulation of
generation CAR’s in the absence of preconditioning resident CD4 + CAR T cells, most likely due to B cells
and only one gave supportive IL-2 albeit at a reduced emerging from hematopoietic progenitor cells, caused
dose (1500 IU, single dose) [50,51,53] . Additionally, the production of Th2 cytokines that mediated the expan-

492 Immunotherapy (2015) 7(5) future science group


CAR T-cell therapy: toxicity & the relevance of preclinical models  Review

sion of the myeloid cell compartment resulting in a cytokine secretion may allow for dynamic control of
lethal granulomatous reaction [41] . CAR T cells to avoid unwanted on-target, off-tumor
The acute and chronic toxicities observed with the toxicities [60] .
mouse CD19-specific scFv appears to be directly related Transient expression of CARs may provide the
to the mouse strain used with toxicity apparent in the short-duration of redirected T-cell activity that is
BALB/c strain but largely absent in C57BL/6 or C3H required to eliminate tumor. T-cells transfected with
strains. This suggests that the Th2 bias of the mouse mRNA encoding the CAR receptor are functional
strain is a critical element in driving this observed toxic- in mouse models [61] although there is some question
ity. Nonetheless, this study demonstrates the auto-toxic of the ability of mRNA transiently expressing CAR
potential that exists with chronic long-term stimulation T cells to deliver durable antitumor responses in solid
of CAR T cells in vivo and poses the question whether tumor models [62] . The use of nonintegrating, transient
the benefits of long-lived immunotherapeutic cells that expression vectors also avoids the potential of any pos-
could offer prolonged immunological memory and pro- sible long-term genotoxicities. Finally, the type of T cell
tection against subsequent tumor growth may be coun- used for adoptive transfer may be critical, with T cells
ter-balanced by the possible toxicity induced through displaying a less differentiated phenotype potentially
extended CAR T-cell stimulation. delivering improved therapy in vivo [63] . This has been
shown to be applicable to CARs in a recently study
Overcoming CAR T-cell-related issues by Xu and colleagues showing that CD19 transduced
Each of these ‘issues’ lies at the core of the successful T memory stem cells cultured in IL-7 and IL-15 cyto-
antitumor activity of the CAR T cell itself. Thus, there kines expanded more efficiently and had more potent
is a fine balancing act to be achieved between tumor survival and antitumor effect in preclinical models [64] .
elimination and unwanted treatment-related toxicity. These examples of technological developments will
To control this, many groups have developed suicide impact upon the CAR T-cell approach and assessing
gene systems, including HSV-TK, iCasp9 and CD20, the impact of each upon toxicity will be the challenge
that once activated can selectively eliminate the CAR to deliver practical information that can aid clinical
T-cell population and, thereby, eliminate the symp- trial design.
toms of CAR T-cell-driven toxicity [54,55] . However,
this represents the least preferred route to manage this Conclusion
issue since depletion of the CAR T cell will also mean CAR T-cell therapy has rapidly risen to prominence
abrogating the therapeutic potential of the CAR T cell. largely as a result of highly impressive clinical results
In addition, the efficacy of these approaches in manag- emanating from several clinical centers employing
ing potentially life threatening acute toxicities remains CD19-specific CAR technology and the subsequent
to be evaluated. rapid influx of enthusiasm from Industry eager to
Clearly, the targeting of truly tumor-specific targets exploit this potentially powerful therapy. However,
is the main goal, and selection systems based upon the the therapeutic approach is still very much in devel-
CAR T cell may facilitate identification of such targets opment with the ability to predict toxicity resulting
rather than adapting existing CARs [56,57] . However, from the activity of CAR T cells still in its infancy.
in the absence of truly tumor-specific target antigens, Consequently, clinical trials are being initiated without
recently developed systems employing dual CAR tar- significant input from models that may have predic-
geting with antigen specificity for separate tumor anti- tive value to aid their design. At present, preclinical
gens; one CAR containing a ‘signal 1’ and the other model systems are being used to determine whether the
with a ‘signal 2’ activating domain, both signals being adverse events observed in trials can be recapitulated in
required for full T-cell activation. The reduced likeli- mice to permit an examination of underlying mecha-
hood of two separate tumor antigens being simultane- nisms driving toxicity. At the moment, for the large
ously expressed on healthy tissue providing increased part, such recapitulation of the clinical situation has
tumor specificity, thus avoiding on-target off-tumor not been achieved. Toxicity induced by CD19 CAR
toxicity  [58] . Likewise, this has been proven in prin- T cells in the BALB/c autologous model system does
ciple for TanCARs; bispecific CARs that bind two tar- not completely reflect that observed in patients, espe-
get antigens for full downstream signaling. This strat- cially considering that IL-6 does not appear to be a
egy also provides a means to improve tumor specificity major driver of acute toxicity unlike that observed in
of CAR T cells with potential for avoiding antigen patients [41] . Moreover, Her2/neu targeted CAR T cells
escape  [59] . Alternatively, inhibitory CARs (iCARs) have failed to show any evidence of autotoxicity in pre-
where the CAR is engineered to deliver control sig- clinical models yet the death of a patient receiving such
nals upon interaction with healthy tissues to regulate CAR T cells [31] testifies the potency of the therapy and

future science group www.futuremedicine.com 493


Review  Kalaitsidou, Kueberuwa, Schütt & Gilham

highlights the poor predictive nature of current model co-operative CARs, TanCARs or iCARs, transient
systems. expression of the CARs via mRNA electroporation or
Whilst the poor predictive nature of current models using less differentiated populations of T cells grown
may relate to biological differences between rodents in variable cytokine conditions. These approaches are
and humans, there is also the case that therapeutic effi- likely to provide means by which to improve clinical
ciency has been the underlying driver to develop CAR outcomes of CAR therapy while subverting toxicities
T-cell therapy with toxicity taking the back seat to this observed thus far.
central aim. With the observations of toxicity in the
clinic, there is now a stronger drive to develop a better Future perspective
understanding of the potential mechanisms of toxicity. Over the last decade CAR T-cell therapy has pro-
From the current literature examining CAR T-cell gressed from a relatively unknown field to a highly
toxicity in preclinical models, there are some general studied one. Successful treatments of both hemato­
issues that appear to correlate with toxicity: logical and solid malignancies have increased interest
in developing this therapy. Over the next 5–10 years,
• Where toxicity occurs, this is generally exacerbated
this treatment method will likely progress to a first line
by higher cell doses. In most cases to date, reduc-
treatment for certain hematological cancers and in the
ing CAR T cell dose reduces the severity of the
more distant future, will likely be applied to a much
observed toxicity;
broader range of neoplasms. Several novel strategies
• The relative signaling potency of CARs appears to are emerging to overcome existing hurdles to this end,
correlate with toxicity. First-generation CARs appear which include more consistent achievement of com-
to drive minimal levels of toxicity, while second and plete remissions as well as the reduction of treatment
third-generation CARs tend to be associated with related toxicities. The most important probably being
acute and chronic toxicity; identification of tumor-specific antigens, appropriate
signaling domains and optimization of each CAR on
• Enhanced CAR T-cell engraftment resulting from a case by case basis.
patient preconditioning and potentially, through
the use of systemic cytokine support can correlate Financial & competing interests disclosure
with increased toxicity levels; DE Gilham is a cofounder of Cellular Therapeutics Ltd. This
work was supported by Leukaemia and Lymphoma Research,
• High levels of target antigens present on tumor the Kay Kendall Leukaemia Fund, Children with Cancer and
or nontumor tissue can drive cytokine release the Medical Research Council. The authors have no other rele-
­syndromes underpinned by CAR T-cell activity. vant affiliations or financial involvement with any organization
As described, various strategies are currently in or entity with a financial interest in or financial conflict with
development to overcome toxicities observed so far the subject matter or materials discussed in the manuscript
with CAR T-cell therapy. These include identification apart from those disclosed.
of targets with high tumor specificity, incorporation No writing assistance was utilized in the production of this
of suicide genes, targeting multiple antigens utilizing manuscript.

Executive summary
• Types of toxicity mediated by chimeric antigen receptor (CAR) therapy include: ‘on-target, on-tumor’, ‘on-
target, off-tumor’ and ‘off-target, off-tumor’.
• The most serious side effects seen so far result from on-target off- tumor toxicity, therefore one of the main
hurdles of CAR therapy is the identification of antigens with high tumor specificity.
• Evidence thus far indicates the increased efficacy of second- and third-generation CARs as compared with
first-generation CARs is also associated with an increased toxicity.
• Increased input CAR T-cell dose and increased tumor burden correlate with more severe toxicity. Route of
administration and T-cell culturing conditions may also play a role.
• Current preclinical models have proven inadequate for predicting CAR toxicity in patients. However, adverse
events in clinical trials have raised the awareness of the dangers of CAR toxicity, therefore more recently there
has been a marked effort to develop improved animal models, with some success.

1 Gross G, Waks T, Eshhar Z. Expression of immunoglobulin-


References T-cell receptor chimeric molecules as functional receptors
Papers of special note have been highlighted as: with antibody-type specificity. Proc. Natl Acad. Sci.
 • of interest; •• of considerable interest USA 86(24), 10024–10028 (1989).

494 Immunotherapy (2015) 7(5) future science group


CAR T-cell therapy: toxicity & the relevance of preclinical models  Review

2 Kahlon KS, Brown C, Cooper LJN, Raubitschek A, treated with autologous T cells genetically engineered to
Forman SJ, Jensen MC. Specific recognition and killing recognize CD19. Blood 116(20), 4099–4102 (2010).
of glioblastoma multiforme by interleukin 13-zetakine 18 Cheadle EJ, Gilham DE, Hawkins RE. The combination of
redirected cytolytic T cells. Cancer Res. 64(24), 9160–9166 cyclophosphamide and human T cells genetically engineered
(2004). to target CD19 can eradicate established B-cell lymphoma.
3 Ramos CA, Dotti G. Chimeric antigen receptor (CAR)- Br. J. Haematol. 142(1), 65–68 (2008).
engineered lymphocytes for cancer therapy. Expert Opin. 19 Hillerdal V, Ramachandran M, Leja J, Essand M. Systemic
Biol. Ther. 11(7), 855–873 (2011). treatment with CAR-engineered T cells against PSCA delays
4 Bridgeman JS, Hawkins RE, Hombach AA, Abken H, subcutaneous tumor growth and prolongs survival of mice.
Gilham DE. Building better chimeric antigen receptors BMC Cancer 14, 30 (2014).
for adoptive T cell therapy. Curr. Gene Ther. 10(2), 77–90 20 Parente-Pereira AC, Burnet J, Ellison D et al. Trafficking
(2010). of CAR-engineered human T cells following regional or
5 Dotti G, Gottschalk S, Savoldo B, Brenner MK. Design and systemic adoptive transfer in SCID beige mice. J. Clin.
development of therapies using chimeric antigen receptor- Immunol. 31(4), 710–718 (2011).
expressing T cells. Immunol. Rev. 257(1), 107–126 (2014). 21 Zhao Y, Moon E, Carpenito C et al. Multiple injections
6 Jensen MC, Riddell SR. Design and implementation of of electroporated autologous T cells expressing a chimeric
adoptive therapy with chimeric antigen receptor-modified antigen receptor mediate regression of human disseminated
T cells. Immunol. Rev. 257(1), 127–144 (2014). tumor. Cancer Res. 70(22), 9053–9061 (2010).
7 June CH, Maus MV, Plesa G et al. Engineered T cells 22 Alcantar-Orozco EM, Gornall H, Baldan V, Hawkins RE,
for cancer therapy. Cancer Immunol. Immunother. 63(9), Gilham DE. Potential limitations of the NSG humanized
969–975 (2014). mouse as a model system to optimize engineered human
8 Maus MV, Grupp SA, Porter DL, June CH. Antibody- T cell therapy for cancer. Hum. Gene Ther. Methods 24(5),
modified T cells: CARs take the front seat for hematologic 310–320 (2013).
malignancies. Blood 123(17), 2625–2635 (2014). 23 Hannon M, Lechanteur C, Lucas S et al. Infusion of
9 Park TS, Rosenberg SA, Morgan RA. Treating cancer with clinical-grade enriched regulatory T cells delays experimental
genetically engineered T cells. Trends Biotechnol. 29(11), xenogeneic graft-versus-host disease. Transfusion 54(2),
550–557 (2011). 353–363 (2014).

10 Sadelain M, Brentjens R, Riviere I. The basic principles 24 Zhou Q, Facciponte J, Jin M, Shen Q, Lin Q. Humanized
of chimeric antigen receptor design. Cancer Discov. 3(4), NOD-SCID IL2rg-/- mice as a preclinical model for cancer
388–398 (2013). research and its potential use for individualized cancer
therapies. Cancer Lett. 344(1), 13–19 (2014).
11 Chmielewski M, Hombach AA, Abken H. Of CARs
and TRUCKs: chimeric antigen receptor (CAR) T cells 25 Davila ML, Riviere I, Wang X et al. Efficacy and toxicity
engineered with an inducible cytokine to modulate the tumor management of 19–28z CAR T cell therapy in B cell acute
stroma. Immunol. Rev. 257(1), 83–90 (2014). lymphoblastic leukemia. Sci. Transl. Med. 6(224), 224ra225
(2014).
12 Lipowska-Bhalla G, Gilham DE, Hawkins RE, Rothwell
DG. Targeted immunotherapy of cancer with CAR 26 Wilkinson RW, Ross EL, Ellison D, Zimmermann W, Snary
T cells: achievements and challenges. Cancer Immunol. D, Mather SJ. Evaluation of a Transgenic Mouse Model
Immunother. 61(7), 953–962 (2012). for Anti-Human CEA Radioimmunotherapeutics. J. Nucl.
Med. 43(10), 1368–1376 (2002).
13 Vonderheide RH, June CH. Engineering T cells for cancer:
our synthetic future. Immunol. Rev. 257(1), 7–13 (2014). 27 Bugelski PJ, Achuthanandam R, Capocasale RJ, Treacy G,
Bouman-Thio E. Monoclonal antibody-induced cytokine-
14 Maude SL, Barrett D, Teachey DT, Grupp SA. Managing
release syndrome. Expert Rev. Clin. Immunol. 5(5), 499–521
cytokine release syndrome associated with novel T cell-
(2009).
engaging therapies. Cancer J. 20(2), 119–122 (2014).
28 Howard SC, Jones DP, Pui CH. The tumor lysis syndrome.
• Highlights the key role of IL-6 in chimeric antigen receptor N. Engl. J. Med. 364(19), 1844–1854 (2011).
(CAR)-mediated toxicities and suggests that it may be a
29 Cheadle EJ, Hawkins RE, Batha H, O’Neill AL, Dovedi SJ,
good target to overcome cytokine release syndrome.
Gilham DE. Natural expression of the CD19 antigen impacts
15 Davila ML, Riviere I, Wang X et al. Efficacy and toxicity the long-term engraftment but not antitumor activity of
management of 19–28z CAR T cell therapy in B cell acute CD19-specific engineered T cells. J. Immunol. 184(4),
lymphoblastic leukemia. Sci. Transl. Med. 6(224), 224ra225 1885–1896 (2010).
(2014).
30 Brentjens RJ, Davila ML, Riviere I et al. CD19-targeted
16 Kochenderfer JN, Dudley ME, Feldman SA et al. B-cell T cells rapidly induce molecular remissions in adults with
depletion and remissions of malignancy along with chemotherapy-refractory acute lymphoblastic leukemia. Sci.
cytokine-associated toxicity in a clinical trial of anti-CD19 Transl. Med. 5(177), 177ra38 (2013).
chimeric-antigen-receptor-transduced T cells. Blood 119(12),
31 Morgan RA, Yang JC, Kitano M, Dudley ME, Laurencot
2709–2720 (2012).
CM, Rosenberg SA. Case report of a serious adverse event
17 Kochenderfer JN, Wilson WH, Janik JE et al. Eradication following the administration of T cells transduced with
of B-lineage cells and regression of lymphoma in a patient

future science group www.futuremedicine.com 495


Review  Kalaitsidou, Kueberuwa, Schütt & Gilham

a chimeric antigen receptor recognizing ERBB2. Mol. 46 Muruganandan S, Sinal CJ. Mice as clinically relevant
Ther. 18(4), 843–851 (2010). models for the study of cytochrome P450-dependent
• Illustrates the catastrophic effects associated with CAR metabolism. Clin. Pharmacol. Ther. 83(6), 818–828 (2008).
recognizing target on nontumor cells. 47 Davies DM, Foster J, Van Der Stegen SJ et al. Flexible
targeting of ErbB dimers that drive tumorigenesis by using
32 Coffman KT, Hu M, Carles-Kinch K et al. Differential
genetically engineered T cells. Mol. Med. 18, 565–576
EphA2 epitope display on normal versus malignant cells.
(2012).
Cancer Res. 63(22), 7907–7912 (2003).
48 Van Der Stegen SJC, Davies DM, Wilkie S et al. Preclinical
33 Sampson JH, Heimberger AB, Archer GE et al. Immunologic
In vivo Modeling of cytokine release syndrome induced by
escape after prolonged progression-free survival with
ErbB-retargeted human T cells: identifying a window of
epidermal growth factor receptor variant III peptide
therapeutic opportunity? J. Immunol. 191(9), 4589–4598
vaccination in patients with newly diagnosed glioblastoma.
(2013).
J. Clin. Oncol. 28(31), 4722–4729 (2010).
34 Gan HK, Burgess AW, Clayton AHA, Scott AM. Targeting • One of the first studies to document the impact of route
of a conformationally exposed, tumor-specific epitope of of administration, tumor load and on-target off-tumor
EGFR as a strategy for cancer therapy. Cancer Res. 72(12), toxicity driven by ligand-specific CAR.
2924–2930 (2012). 49 Van Schalkwyk MC, Papa SE, Jeannon JP, Guerrero Urbano
35 Maher J, Wilkie S. CAR mechanics: driving T cells into the T, Spicer JF, Maher J. Design of a phase I clinical trial to
MUC of cancer. Cancer Res. 69(11), 4559–4562 (2009). evaluate intratumoral delivery of ErbB-targeted chimeric
antigen receptor T cells in locally advanced or recurrent
36 Gnjatic S, Nishikawa H, Jungbluth AA et al. NY-ESO-1:
head and neck cancer. Hum. Gene Ther. Clin. Dev. 24(3),
Review of an immunogenic tumor antigen. Adv. Cancer
134–142 (2013).
Res.95,1–30 (2006).
50 Kakarla S, Chow KKH, Mata M et al. Antitumor effects
37 Sang M, Lian Y, Zhou X, Shan B. MAGE-A family:
of chimeric receptor engineered human T cells directed to
attractive targets for cancer immunotherapy. Vaccine 29(47),
tumor stroma. Mol. Ther. 21(8), 1611–1620 (2013).
8496–8500 (2011).
51 Schuberth PC, Hagedorn C, Jensen SM et al. Treatment
38 Tsai J-R, Chong I-W, Chen Y-H et al. Differential expression
of malignant pleural mesothelioma by fibroblast activation
profile of MAGE family in non-small-cell lung cancer. Lung
protein-specific re-directed T cells. J. Transl. Med. 11, 187
Cancer 56(2), 185–192 (2007).
(2013).
39 Kochenderfer JN, Rosenberg SA. Treating B-cell cancer with
52 Tran E, Chinnasamy D, Yu Z et al. Immune targeting
T cells expressing anti-CD19 chimeric antigen receptors.
of fibroblast activation protein triggers recognition of
Nat. Rev. Clin. Oncol. 10(5), 267–276 (2013).
multipotent bone marrow stromal cells and cachexia. J. Exp.
40 Till BG, Jensen MC, Wang J et al. CD20-specific adoptive Med. 210(6), 1125–1135 (2013).
immunotherapy for lymphoma using a chimeric antigen
•• Report documenting toxicity resulting from the depletion
receptor with both CD28 and 4–1BB domains. pilot clinical
of a previously unknown population of osteogenic cells
trial results. Blood 119(17), 3940–3950 (2012).
directed by a third-generation FAP-specific CAR combined
41 Cheadle EJ, Sheard V, Rothwell DG et al. Differential role of
with preconditioning and systemic IL-2.
Th1 and Th2 cytokines in autotoxicity driven by CD19-
specific second-generation chimeric antigen receptor T cells 53 Wang LC, Lo A, Scholler J et al. Targeting fibroblast
in a mouse model. J. Immunol. 192(8), 3654–3665 (2014). activation protein in tumor stroma with chimeric antigen
receptor T cells can inhibit tumor growth and augment host
•• First report of acute and chronic toxicity mediated by immunity without severe toxicity. Cancer Immunol. Res. 2(2),
CD19 second-generation CAR involving the dysregulated 154–166 (2014).
response to Th1 and Th2 biased cytokines in a specific
•• A study showing no evidence of toxicity with a FAP-specific
mouse model.
second-generation CAR suggesting that factors enhancing
42 Hacein-Bey-Abina S, Von Kalle C, Schmidt M et al. LMO2- CAR T-cell engraftment and functional activity may
associated clonal T cell proliferation in two patients after
underlie potential on-target off-tumor toxicities.
gene therapy for SCID-X1. Science 302(5644), 415–419
(2003). 54 Marin V, Cribioli E, Philip B et al. Comparison of
different suicide-gene strategies for the safety improvement
43 Scholler J, Brady TL, Binder-Scholl G et al. Decade-long
of genetically manipulated T cells. Hum. Gene Ther.
safety and function of retroviral-modified chimeric antigen
Methods 23(6), 376–386 (2012).
receptor T cells. Sci. Transl. Med. 4(132), 132ra153 (2012).
55 Di Stasi A, Tey S-K, Dotti G et al. Inducible apoptosis
44 Wang LX, Westwood JA, Moeller M et al. Tumor ablation
as a safety switch for adoptive cell therapy. N. Engl.
by gene-modified T cells in the absence of autoimmunity.
J. Med. 365(18), 1673–1683 (2011).
Cancer Res 70(23), 9591–9598 (2010).
56 Alonso-Camino V, Sanchez-Martin D, Compte M et al.
45 John LB, Devaud C, Duong CP et al. Anti-PD-1 antibody
CARbodies: human antibodies against cell surface tumor
therapy potently enhances the eradication of established
antigens selected from repertoires displayed on T cell
tumors by gene-modified T cells. Clin. Cancer Res. 19(20),
chimeric antigen receptors. Mol. Ther. Nucleic Acids 2, e93
5636–5646 (2013).
(2013).

496 Immunotherapy (2015) 7(5) future science group


CAR T-cell therapy: toxicity & the relevance of preclinical models  Review

57 Lipowska-Bhalla G, Gilham DE, Hawkins RE, Rothwell 61 Barrett DM, Zhao Y, Liu X et al. Treatment of Advanced
DG. Isolation of tumor antigen-specific single-chain variable leukemia in mice with mRNA engineered T cells. Hum.
fragments using a chimeric antigen receptor bicistronic Gene Ther. 22(12), 1575–1586 (2011).
retroviral vector in a Mammalian screening protocol. Hum. 62 Singh N, Liu X, Hulitt J et al. Nature of tumor control by
Gene Ther. Methods 24(6), 381–391 (2013). permanently and transiently-modified GD2 chimeric antigen
58 Kloss CC, Condomines M, Cartellieri M, Bachmann receptor T cells in xenograft models of neuroblastoma.
M, Sadelain M. Combinatorial antigen recognition with Cancer Immunol. Res. 2(11), 1059–1070 (2014).
balanced signaling promotes selective tumor eradication by 63 Gattinoni L, Klebanoff CA, Restifo NP. Paths to stemness:
engineered T cells. Nat. Biotechnol. 31(1), 71–75 (2013). building the ultimate antitumour T cell. Nat. Rev.
59 Grada Z, Hegde M, Byrd T et al. TanCAR: a novel bispecific Cancer 12(10), 671–684 (2012).
chimeric antigen receptor for cancer immunotherapy. Mol. 64 Xu Y, Zhang M, Ramos CA et al. Closely related
Ther. Nucleic Acids 2, e105 (2013). T-memory stem cells correlate with in vivo expansion of
60 Fedorov VD, Themeli M, Sadelain M. PD-1-and CTLA- CAR. CD19-T cells and are preserved by IL-7 and IL-15.
4-based inhibitory chimeric antigen receptors (iCARs) Blood 123(24), 3750–3759 (2014).
divert off-target immunotherapy Responses. Sci. Transl.
Med. 5(215), 215ra172 (2013).

future science group www.futuremedicine.com 497

Vous aimerez peut-être aussi