ISSN 0006-2979, Biochemistry (Moscow), 2024, Vol. 89, No. 5, pp. 765-783 © Pleiades Publishing, Ltd., 2024.
765
REVIEW
CAR Cells beyond Classical CAR T Cells:
Functional Properties and Prospects of Application
Elizaveta P. Minina
1
, Dmitry V. Dianov
1
, Saveliy A. Sheetikov
1
,
and Apollinariya V. Bogolyubova
1,a
*
1
National Medical Research Centre for Hematology, Ministry of Health of the Russian Federation,
125167 Moscow, Russia
a
e-mail: apollinariya.bogolyubova@gmail.com
Received August 14, 2023
Revised November 23, 2023
Accepted December 2, 2023
AbstractChimeric antigen receptors (CARs) are genetically engineered receptors that recognize antigens and
activate signaling cascades in a cell. Signal recognition and transmission are mediated by the CAR domains derived
from different proteins. T cells carrying CARs against tumor-associated antigens have been used in the develop-
ment of the CAR T cell therapy, a new approach to fighting malignant neoplasms. Despite its high efficacy in the
treatment of oncohematological diseases, CAR T cell therapy has a number of disadvantages that could be avoided
by using other types of leukocytes as effector cells. CARs can be expressed in a wide range of cells of adaptive and
innate immunity with the emergence or improvement of cytotoxic properties. This review discusses the features
of CAR function in different types of immune cells, with a particular focus on the results of preclinical and clinical
efficacy studies and the safety of potential CAR cell products.
DOI: 10.1134/S0006297924050018
Keywords: chimeric antigen receptor, immunotherapy, cell therapy, CAR, CAR T cell, CAR NK cell
Abbreviations: CAR,chimeric antigen receptor; CARM,macrophage expressing CAR; CAR T,T cell expressing CAR; CBCR,chi-
meric B cell receptor; CIK,cytokine-induced killers; CRS,cytokine release syndrome; DC,dendritic cell; DN T cell,double-neg-
ative T cell; GD2,disialoganglioside GD2; GVHD,graft-versus-host disease; iNKT cell,invariant NKT cell; MAIT cell,mucosal-
associated invariant T cell; MHC,major histocompatibility complex; NK,natural killer cell; NKT cells,natural killer T cell;
NSCAR,non-signaling CAR; TCR,T cell receptor; Treg,regulatory T cell.
* To whom correspondence should be addressed.
INTRODUCTION
A chimeric receptor consisting of the variable
immunoglobulin domain and constant regions of the
T cell receptor (TCR) was first created in 1987 [1].
It recognized the bacterial antigen phosphorylcholine
and was expressed in EL4 lymphoblastic T cells. The
first T lymphocytes with a chimeric antigen receptor
(CAR) were obtained in 1993 [2]. This receptor recog-
nized 2,4,6-trinitrophenol and was one of the first gen-
eration of CARs. It consisted of the extracellular anti-
gen-recognizing single-chain variable fragment (scFv)
linked by a transmembrane region to the intracellular
CD3ζ signaling domain (a fragment of the endogenous
TCR) [3]. CAR T cells expressing the first-generation re-
ceptors were poorly effective against malignant cells
because, despite of their strong cytotoxic properties,
they were easily exhausted [4]. In 2002, second-gen-
eration CARs were obtained. They contained the CD28
costimulatory domain between the signaling and trans-
membrane domains [5]. Second-generation CAR T cells
targeting the CD19 antigen were effective in recogniz-
ing and eliminating B cell tumors in a mouse model
[6]. Since then, CAR T lymphocytes targeting various tu-
mor-associated antigens have been actively developed.
CAR T therapy has been particularly effective in
the treatment of hematological oncological diseases.
To date, the Food and Drug Administration (FDA) has
approved six CAR-T therapies for the treatment of
B-cell neoplasms [7]. So far, CAR-T products that have
MININA et al.766
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
successfully passed clinical trials and have been ap-
proved by FDA are based on the second-generation
CARs containing CD28 or 4-1BB signaling domains as
costimulatory domains. Other variants of CAR T cells
containing receptors with different immunomodulato-
ry domains are being researched and developed. For
example, third-generation CARs containing two costim-
ulatory domains have been developed, although their
clinical use has been limited due to strong severe side
effects. CAR T cells expressing fourth-generation CARs
also secrete cytokines that increase the persistence of
these cells in the tumor microenvironment [8].
Despite significant success in the treatment of
oncohematological diseases, CAR T cell therapy has a
number of drawbacks. For example, it becomes inef-
fective when malignant cells lose the tumor antigens
targeted by the CARs [8]. CAR T cells are often unable
to infiltrate solid tumors and recognize the antigen.
In addition, the cytotoxic function of CAR T cells is
significantly influenced by the immunosuppressive
tumor microenvironment. Currently, progress in the
treatment of solid tumors with CAR T cells has reached
its limits [9]. Finally, the secretion of pro-inflammatory
cytokines by activated CAR T cells can lead to severe
side effects, such as cytokine release syndrome (CRS)
and neurotoxicity [10], which are due to the rapid ac-
tivation and proliferation of T cells producing pro-in-
flammatory cytokines and typically develop within
one week of CAR T cell administration. It has been sug-
gested that excessive release of pro-inflammatory cy-
tokines increases capillary permeability in the brain,
leading to severe neurological symptoms and even
death, although the molecular mechanisms of neuro-
toxicity in the case of CAR T cell therapy are not ful-
ly understood [10]. The adverse effects of this therapy
are often due to the poor regulation of the activity of
CAR T cells, which can sometimes be activated even
inthe absence of an antigenic stimulus [11].
Fig. 1. Advantages and disadvantages of different types of immune cells in the development of CAR-bearing cell products.
Nomenclature: TCR, T cell receptor; CAR, chimeric antigen receptor; NKG2D, activating receptor; MR1, non-canonical major
histocompatibility complex close to classI; CD1,non-canonical major histocompatibility complex close to classI; CRS,cytokine
release syndrome; MHC,major histocompatibility complex; GVHD,graft-versus-host disease.
CAR CELLS BEYOND CLASSICAL CAR T CELLS 767
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
To overcome these drawbacks, new strategies for
the CAR T cell generation are being actively being de-
veloped [12, 13], including the use of different (non-
T cells) leukocytes as CAR-bearing cells (Fig. 1). CAR-
expressing cells have been derived from γδ T cells,
regulatory T cells (Tregs), mucosal-associated invariant
T cells (MAIT cells), double-negative T cells (DNTcells),
natural killer (NK) cells, natural killer T cells (NKT
cells), cytokine-induced killers (CIKs), macrophages,
dendritic cells (DCs), and even B lymphocytes.
Preclinical studies have shown that many alterna-
tive immune CAR cells with a high antitumor activity
have fewer or none of the typical drawbacks of CAR
T cells. However, such cells are poorly studied. Only a
few cell products have entered phase I/II clinical tri-
als; some have preclinical data, while others are still
under development. This review discusses the features
of CAR function in different types of immune cells and
the results of available preclinical and clinical studies
of CAR-bearing immune cells.
CARs IN DIFFERENT TYPES
OF IMMUNE CELLS
Below we discuss the properties of different im-
mune cells that could be used in the development of
novel CAR-mediated immunotherapies, focusing on
the results of preclinical and, if available, clinical stud-
ies of CAR expression and efficacy in these cells.
Natural killer (NK) cells are innate immune cells
of the lymphoid lineage. NK cells account approx-
imately 10% of peripheral blood lymphocytes [14].
Unlike T lymphocytes, they recognize malignant and
infected cells with a variety of non-polymorphic acti-
vating and inhibitory receptors. The directionality of
the NK cell response in each case is the result of a bal-
ance of various signals. When the signals from the ac-
tivating receptors outweigh those from the inhibitory
receptors, an NK cell causes a lysis of the target cell.
NK cells express a variety of activating and inhibiting
receptors on their surface, the most important among
them are receptors that recognize the major histocom-
patibility complex MHC I and MHC I-like molecules.
Cells that express these molecules are recognized as
healthy by NK cells [15]. It is important to note that red
blood cells do not carry the ligands (either activating
or inhibiting) for the NK cell receptors on their surface
and therefore are not recognized as targets by these
cells. In addition to MHC, NK cells can recognize other
cell surface ligands, in particular, the stress markers
MICA, MICB, and UL16BP1, due to the presence of spe-
cific receptors, including the NKG2D receptor, which is
also present on the surface of γδ T cells [16]. In addi-
tion, the expression of FcγRIIIa allows NK cells to rec-
ognize and kill cells opsonized by antibodies [17].
The cytotoxic function of NK cells is manifested by
the formation of an immunological synapse between
the NK cell and the target cell and the subsequent se-
cretion of granzyme-containing lytic granules. NK cells
can also kill target cells by inducing programmed cell
death through FasL and TRAIL. Finally, activated NK
cells release a number of pro-inflammatory cytokines,
in particular, interferon γ (IFNγ) and tumor necrosis
factor (TNF) [14].
The cytotoxic properties of NK cells and the lack
of need for the antigen presentation in the MHC con-
text required for the recognition of target cells (unlike
in T cells), have significantly contributed to the devel-
opment of anti-cancer therapies based on genetically
modified NK cells, including those expressing CARs.
In most studies, NK cells have been transduced with
the CAR constructs originally developed for the CAR
T cells therapy. In addition to the CD28 and 4-1BB co-
stimulatory domains, some of the CARs expressed
in NK cells contained the 2B4 costimulatory domain
(Fig. 2). The 2B4 receptor is one of the activating re-
ceptors of the SLAM (signaling lymphocytic activation
molecule) family on NK cells. It is important to note
that CAR constructs originally designed for expression
in T cells can also function in NK cells due to the pres-
ence of common signaling pathways that control cell
activation in both cell types. In particular, the signal-
ing from some activating receptors in NK cells involves
the CD3ζ signaling domain which is intended for use
in T cells [18]. NK cells transduced with the second-
generation 2B4-containing CAR targeting CD5 had a
higher cytotoxic activity against malignant cells and
showed more rapid proliferation and enhanced cyto-
kine production compared to NK cells expressing CARs
with the CD28 domain [19]. In addition to the CD3ζ do-
main, CARs developed for the expression in NK cells
can contain the DAP10 and DAP12 domains, which are
involved in the signaling from a number of activating
NK cell receptors. It has been shown that the antitu-
mor activity observed for the constructs with the CD3ζ
domain was higher than that of those with the DAP10
domain, but lower than the activity of CARs with the
DAP12 domain [20,21]. Cifaldi etal. [22] proposed the
use of DNAM-1 as a part of a CAR adapted to NK cells.
DNAM-1 recognizes the poliovirus receptor (PVR) and
nectin-2, which are expressed on the virus-infected
cells and many malignant cells. Receptors contain-
ing the 2B4 and CD3ζ domains in addition to DNAM-1
cause further activation of NK cells.
It should be emphasized that CAR-expressing NK
cells can potentially exhibit cytotoxic activity against
malignant cells in a CAR-independent manner due to
their own activating receptors, as well as their abili-
ty to recognize cells opsonized by antibodies [20,23].
The presence of a CAR-independent antitumor activ-
ity enhances the efficacy of the CAR NK cell therapy.
MININA et al.768
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
Fig. 2. Structure of CARs used in the development of products based on different cell types. Nomenclature: NK cells,natural killer
cells; NKT cells,natural killer T cells.
Inaddition, the diversity of activating receptors allows
CAR NK cells to recognize and destroy tumor cells with
an altered phenotype that have survived a long-term
treatment [24]. NK cells treated with exogenous IL-12
and IL-18 acquire a phenotype similar to that of mem-
ory cells. The efficacy of such cells has already been
demonstrated in phase I trials in patients with re-
lapsed or resistant acute myeloid leukemia. Additional
expression of a CAR directed against nucleophosmin1
(NPM1) in these cells increased the efficacy of the
therapy and reduced the incidence of side effects [25].
Anincreased sensitivity of NK cells to reactive oxygen
species (compared to T and B cells) can be reduced by
expression of peroxiredoxin 1, which would promote
the persistence of CAR NK cells in the acidified envi-
ronment of solid tumors [26].
Preclinical and clinical studies have shown that
CAR NK cells lack many of the disadvantages of CAR T
cells. Since NK cells do not carry variable TCRs, their
adoptive transfer does not lead to the development
of the graft-versus-host disease (GVHD) [27, 28]. This
opens up the possibility for the producing off-the-shelf
allogeneic cell preparations suitable for many patients
at the same time. In addition, CAR NK cell transfer
carries almost no risk of CRS or neurotoxicity [29,30].
Finally, CAR NK cells have been shown in preclinical
studies to be effective not only against oncohematolog-
ical diseases, but also in the treatment of solid tumors
[31]. Currently, many developed CAR NK cell prepa-
rations, including those against solid tumors, have
moved from preclinical studies to clinical trials.
Several dozen clinical trials have been registered
to investigate the use of CAR NK cells directed against
various tumor antigens, both as a monotherapy and
in combination with other therapeutic approaches.
For example, in one trial (NCT04847466), the partici-
pants treated with CAR NK cells against PD-L1 also
received the immunostimulant N-803, which induces
proliferation and activation of NK and CD8
+
T cells, and
pembrolizumab (monoclonal antibody against PD-1).
NK cells sequentially transfected with the chemokine
receptor CXCR4 and a CAR directed against B cell mat-
uration antigen (BCMA) efficiently destroyed multiple
myeloma cells [32]. Currently, all clinical trials of the
CAR NK cell therapy are in phase I/II. Several regis-
tered trials are testing CAR NK cells against oncohema-
tological diseases, such as multiple myeloma (target,
BCMA; NCT05008536), B cell lymphomas (target, CD19;
NCT05379647) and acute lymphoblastic leukemia (tar-
get, CD19; NCT05563545). Clinical trials investigating
the efficacy and safety of CAR NK cells in the treat-
ment of various solid tumors are ongoing: ovarian
cancer (target, claudin 6; NCT05410717); colon cancer
(target, NKGD2L; NCT05213195); pancreatic cancer
(target, ROBO1; NCT03941457); prostate cancer (target,
PSMA; NCT03692663); and others (table). The success-
ful completion of phase I in several clinical trials of
CAR NK cells has demonstrated the high safety of this
type of therapy and the almost complete absence of
side effects [33].
Despite many advantages, CAR NK cell thera-
py has some limitations. CAR NK cells are character-
ized by a low persistence after adoptive transfer: the
lifespan of CAR T cells in a patient’s body can be up
to 10 years [34], while the lifespan of CAR NK cells
does not exceed several weeks [35]. CAR NK cells are
CAR CELLS BEYOND CLASSICAL CAR T CELLS 769
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
Some registered CAR NK cell clinical trials
Clinical trial Started in Phase Disease Target
NCT02892695 2016 I/II lymphomas and leukemias CD19
NCT03940833 2019 I/II multiple myeloma BCMA
NCT02742727 2016 I/II lymphomas and leukemias CD7
NCT02944162 2016 I/II acute myeloid leukemia CD33
NCT02839954 2016 I/II solid tumors MUCL
NCT03383978 2017 I glioblastoma HER2
NCT03415100 2018 I metastatic solid tumors NKG2DL
NCT03941457 2019 I/II pancreatic cancer ROBO1
NCT03692663 2018 I prostate cancer PSMA
NCT05410717 2022 I/II ovarian cancer claudin6 (CLDN6)
NCT05194709 2021 I developed solid tumors 5T4
NCT05507593 2022 I non-small cell lung cancer DLL3
difficult to expand ex vivo and do not tolerate freez-
ing and storage well [36]. However, there is hope that
these technical difficulties will be soon be overcome.
In particular, it has been shown that the persistence
of CAR NK cells can be doubled by knocking out the
CISH gene, which encodes the CIS protein, a nega-
tive regulator of the IL-15 signaling pathway. The ab-
sence of CIS in CAR NK cells activates IL-15 secretion
and, since CAR NK cells carry IL-15 receptors on their
surface, causes autocrine activation of the IL-15 sig-
naling pathway. Activation of IL-15-mediated signal-
ing, in turn, promotes the expansion of CAR NK cells
ex vivo, increases their persistence and enhances the
antitumor properties of these cells [37]. Incorporation
of IL-15 into a CAR construct can also improve the cell
metabolic status of the cells and activate the effector
functions of CAR NK cells that are attenuated by the
interaction with metabolically active tumors [38].
Another factor that can reduce the efficacy of CAR
NK therapy is the capture of tumor antigens by the
therapeutic CAR NK cells via the trogocytosis mecha-
nism. After capturing the tumor antigen, CAR NK cells
become the target of the therapy and are destroyed
by other NK cells via the fratricide pathway, while
the amount of tumor antigen on the targeted malig-
nant cells decreases. The risk of fratricide among CAR
NK cells can be reduced by co-expression of activating
CARs directed against the tumor antigen and inhibitory
CARs that recognize antigens specific for NK cells [39].
γδ T cells. Approximately 3.7% of T cells circulat-
ing in the bloodstream have TCRs formed by the γ and
δ chains on their surface [40,41]. Such “non-classical”
(as opposed to “classical” αβ T cells) γδ T cells differ
significantly from αβ T cells, which carry TCRs formed
by the α and β chains and constitute a predominant
population of circulating T cells. Recognition of anti-
gens by αβ T cells is only possible when the antigen
is presented in the context of the MHC, whereas γδ T
cells do not require the involvement of classical MHC
molecules in the antigen recognition, greatly expand-
ing the possibilities for their transfer between differ-
ent organisms and significantly reducing the likelihood
of complications such as GVHD. Relatively safe adop-
tive transfer of γδ T cells is also possible because the
repertoire of γ and δ chains in a population is much
less diverse than the repertoire of α and β chains, so
that γδ T cells tend to recognize molecular signatures
common to different individuals, indicating the devel-
opment of an infectious process or the emergence of
malignant cells. For this reason, they are often thought
to be similar to the cells of the innate immunity. γδ T
cells also resemble innate immune cells in the expres-
sion of Toll-like receptors and receptors similar to the
activating receptors of NK cells, in particular, NKG2D
[42]. However, γδ T cells are also capable of forming
immunological memory and are therefore classified as
components of the adaptive immunity [43]. γδ T cells
can differentiate into Th-like cells and produce a wide
range of cytokines [44].
Most circulating γδ T cells are Vγ9Vδ2 T cells. Their
TCRs consist of the Vγ9 and Vδ2 segments of the γ and
δ chains, respectively. Since Vγ9Vδ2 T cells recognize
MININA et al.770
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
phosphoantigens, synthetic analogs such as zoledron-
ic acid are used to expand Vγ9Vδ2 T cells exvivo [45].
γδ T cells, which have the Vδ1 segment in the TCRs,
localize mainly to the mucosa [23].
The role of γδ T cells in providing antitumor pro-
tection was first demonstrated in mice lacking these
cells. In these mice, chemical mutagens caused early
development of squamous cell carcinoma of the skin
[46]. Later, an important role of γδ T cells in antitumor
immunity has been demonstrated in various tumor
models. In particular, the extent of malignant tumor
infiltration by γδ T cells has been shown to correlate
with a favorable prognosis in many types of cancer,
such as melanoma [47] and gastric cancer [48]. It is be-
lieved that the activity of γδ T cells against malignant
cells is ensured by their cytotoxic properties, as well as
the production of IFNγ and TNF [33]. The cytotoxicity
of γδ T cells is provided by the action of perforin and
granzymes; the cells are also capable of antibody-de-
pendent cellular cytotoxicity. Antitumor activity is in-
herent to both Vγ9Vδ2 and Vδ1 T cells [45, 48, 49].
γδ T cells have both the antitumor and pro-tumor
properties. The pro-tumor effect of γδ T cells is usual-
ly due to the production of interleukin-17 (IL-17) and
some other cytokines. For example, in pancreatic can-
cer, γδ T cells suppress the activity of αβ T cells by se-
creting IL-10 and IL-17 and promoting PD-L1 expres-
sion in tumor cells [50].
The strong intrinsic antitumor activity of γδ T cells,
together with a highly safe adoptive transfer, makes
these cells a promising tool in the immunotherapy of
oncological diseases. Since the transfer of unmodified
γδ T cells to a patient after their exvivo expansion ap-
peared to be safe but ineffective [45], the enhancement
of the antitumor activity of γδ T cells via CAR expres-
sion has been actively investigated. It was shown that
CD19-directed CAR γδ T cells effectively recognized and
destroyed malignant cells, including those that had
lost CD19. The cytotoxic properties of CAR γδ T cells
were enhanced by their treatment with zoledronic
acid [51, 52]. The ability of CAR γδ T lymphocytes to
kill malignant blood cells that have lost antigen is due
to the high non-specific cytotoxicity of γδ T cells to-
wards leukemia cells, which is further enhanced by
zoledronic acid during exvivo expansion of CAR γδ
T cells. Such retention of cytotoxic properties against
malignant blood cells that have lost the antigen is an
important advantage of CAR γδ T cells [52].
CAR γδ T cells can also be effective against solid
tumors. It has been shown that CAR γδ T cells direct-
ed against the neuroblastoma antigen (GD2) not only
destroy malignant cells, but also present antigens to
and activate αβ T cells invitro. It is possible that CAR
γδ T cells also present antigens and activate αβ T cells
invivo, but further experiments are needed to clarify
this issue [23].
Most CARs that have been successfully expressed
in γδ T cells are the second-generation receptors and
contain CD28 and CD3ζ as costimulatory and signaling
domains, respectively [23] (Fig. 2). In [53], the first-
generation GD-recognizing CAR with a single intracel-
lular domain DAP10 was introduced into γδ T cells.
Itshould be noted that γδ T cells express of costimula-
tory molecules, such as CD28, CD27, and 4-1BB, which
contribute to the activation of these cells during sig-
naling by CARs [54].
Some studies have used γδ T cells to create a com-
binatorial antigen recognition system acting as logical
operators. For example, Fisher et al. [53] obtained γδ
CAR T cells expressing GD2-recognizing DAP10-CAR and
γδ TCR that bound to phosphoantigens expressed by
malignant (but not normal) cells. These γδ CAR Tcells
were activated only by interaction with the target cells
exposing both antigens recognized by the CAR and
TCR, thus significantly reducing the likelihood of kill-
ing off-target cells.
Fleischer et al. [55] obtained γδ T cells expressing
non- signaling CAR (NSCAR), which lack intracellular ac-
tivation domains and are therefore unable to trigger the
cytotoxic response upon antigen recognition. NSCAR-
expressing γδ T cells can recognize the tumor antigens
CD19 and CD5 and, through this interaction, approach
malignant cells. However, direct antitumor activity re-
quires endogenous MHC-independent cytotoxicity of
γδ T cells. In acute lymphoblastic leukemia, γδ T cells
expressing NSCARs were found to be more cytotoxic
against B and T cells than γδ T cells expressing CARs
against the same antigens (CD19 and CD5). Expression of
NSCARs instead of CARs in αβ T cells did not significant-
ly increase their cytotoxic properties, suggesting that
the molecular mechanisms underlying the enhanced
antitumor activity of γδ T cells expressing NSCARs are
fundamentally different from those of αβ T cells [55].
Several ongoing clinical trials are focusing on the
use of γδ CAR T cells in the immunotherapy of different
types of cancer, such as acute myeloid leukemia (tar-
gets, CD33 and CD123; NCT03885076, NCT04796441),
relapsed or resistant CD7
+
T cell leukemia (target, CD7;
NCT04702841), various B cell oncohematological
diseases (targets, CD20 and CD19; NCT04735471,
NCT02656147, NCT04911478) and relapsed or resistant
solid tumors of various origins (target, NKG2DL,
NCT04107142, NCT05302037). To date, all these clinical
trials are in phaseI, so it is too early to assess the clin-
ical efficacy of γδ CAR T cells. It is believed that the
γδ CAR T cell-based therapy will have an advantage
over conventional CAR T cell therapy by reducing the
risk of developing side effects, such as CRS and neuro-
toxicity [49]. Studies on the function of γδ CAR T cells
invivo have shown that these cells only persist in the
body for a short time [52], which may be a significant
limitation for their clinical application.
CAR CELLS BEYOND CLASSICAL CAR T CELLS 771
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
NKT cells are similar to γδ T cells and combine
the properties of adaptive and innate immune cells.
NKT cells represent less than 1% of T cells in the blood
[56] and express αβ TCRs, as well as molecules charac-
teristic of NK cells, such as CD16 (FcγR) and CD56 [57].
Unlike T lymphocytes, NKT cells recognize antigens
only in the context of CD1d, a non-classical MHC mol-
ecule close to MHC classI. NKT cells are usually clas-
sified into invariant NKT (iNKT) cells, or type I NKT
cells, and type II NKT cells. iNKT cells are character-
ized by a restricted TCR repertoire and recognize α-ga-
lactosylceramide (α-GalCer), whereas typeII NKT cells
are characterized by a broader TCR repertoire and do
not recognize α-GalCer [58].
The number of NKT cells in the blood is often re-
duced in patients with various malignancies, partic-
ularly oncohematological ones, and the functions of
these cells (e.g., IFNγ production) are also impaired
[60]. An increased number of NKT cells in the periph-
eral blood, as well as NKT cell infiltration into the tu-
mor tend to correlate with a favorable prognosis for
patients [61, 62]. Only iNKT cells have a pronounced
antitumor effect, whereas type II NKT cells can even
suppress the immune response against malignant
cells. The cytotoxicity of iNKT cells, which is mediat-
ed by granzymes or FasL, is promoted by the recog-
nition of the lipid antigen associated with CD1d [63].
Inaddition to direct lysis of tumor cells, iNKT cells can
modulate the activity of other immune cells (in par-
ticular, DCs, NK cells, B and T lymphocytes) through
the secretion of pro- and anti-inflammatory cytokines,
such as IFNγ, TNF, IL-4, IL-10, and IL-13 [64,  65]. Cyto-
kines released by iNKT cells can also affect the tumor
microenvironment and thus indirectly influence the
immune response against tumors [64]. NKT cells with
a transient CAR expression produced less IL-6 than
transduced CD8
+
CAR T lymphocytes, while maintain-
ing their cytotoxic activity. For this reason, it has been
suggested that the risk of CRS is lower with CAR NKT
therapy than with CAR-T therapy, but the issue needs
further clarification [66].
Some properties of NKT cells make them partic-
ularly attractive as effectors for CAR therapy. Their
intrinsic antitumor activity (not only the cytotoxic
properties, but also the ability to rearrange the tumor
environment) can complement the CAR-mediated ef-
fect against malignant cells. Since NKT cells recognize
antigens in the context of non-polymorphic CD1d, their
allogeneic adoptive transfer does not cause GVHD, al-
lowing the generation of allogeneic CAR iNKT cells
from the cells of healthy donors [23]. In addition, NKT
cells are able to proliferate rapidly exvivo and are rel-
atively easy to obtain in the quantities required to pro-
duce a clinical product [56].
In the first work on the generation of CAR iNKT
cells, the expressed CAR was directed against the neu-
roblastoma antigen GD2 and contained the 4-1BB and
CD3ζ signaling domains (Fig. 2). 4-1BB was shown to
polarize CAR iNKT cells towards the Th1 phenotype,
enabling these cells to efficiently destroy neuroblasto-
ma cells invivo and persist in the body for a long time
[67]. CD62L expression has been shown to be a mark-
er of increased persistence and anti-tumor activity of
NKT cells, including those expressing CARs. Adminis-
tration of anti-CD19 CAR NKT cells expressing CD62L
to mice with the B cell lymphoma resulted in a signif-
icant disease regression [68]. The study of the efficacy
of CAR NKT cells recognizing chondroitin sulfate pro-
teoglycan 4 (CSPG4; melanoma cell antigen) showed
that CAR NKT cells destroyed melanoma cells invitro
no less efficiently than CAR T cells [69]. The generation
of NKT cells expressing the third-generation CARs di-
rected against glypican-3 (hepatocellular carcinoma
cell antigen) were obtained and containing CD28 and
4-1BB costimulatory domains has recently been an-
nounced [70].
Advances in the preclinical application of CAR
NKT cells have led to the initiation of clinical tri-
als for the treatment of neuroblastoma (target, GD2;
NCT03294954, NCT02439788), relapsed or resistant
B cell lymphoma (target, CD19; NCT03774654) and
other B cell neoplasms (target, CD19; NCT05487651).
The small number of CAR NKT clinical trials is proba-
bly due to the insufficient preclinical research. Anoth-
er major limitation for the production of clinical prod-
ucts from CAR NKT cells is the low number of NKT
cells in the peripheral blood.
Regulatory T cells (Tregs) are a specialized subset
of CD4
+
T lymphocytes with the immunosuppressive
function. Tregs account for 5 to 10% of CD4
+
Tcells in
the circulation [71]. The inhibitory influence of Tregs
on the effector T cells or antigen-presenting cells can
be direct or indirect. In the first case, Tregs act on
target cells by secreting anti-inflammatory cytokines
(IL-35, IL-10, or TGF-β) or by releasing granzymes from
the lytic granules, resulting in the target cell death.
Indirect mechanisms include targeting of other cells
exposed to Tregs [72].
The ability of Tregs to inhibit effector immune
cells makes them potentially useful for therapeutic
applications and in various conditions associated with
an exaggerated immune response, such as transplanta-
tion. However, the first studies of the therapeutic prop-
erties of polyclonal Tregs showed that their adoptive
transfer induces non-specific tolerance, reduces the
body’s resistance to infection and increases the risk of
developing malignant neoplasms [73]. CAR expression
allows Tregs to selectively attack target tissues while
maintaining the original inhibitory activity of these
cells and avoiding induction of unwanted immunolog-
ical tolerance. The first-generated CAR Tregs were in-
tended for the therapy of colitis in a mouse model [74];
MININA et al.772
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
soon after, the development of the first human CAR
Tregs was reported [75].
In most studies of CAR Tregs, these cells expressed
second-generation CARs with the CD28 or 4-1BB as
costimulatory domains. Systematic analysis of the ac-
tivity of second-generation CARs directed against the
human leukocyte antigen HLA-A2 with different co-
stimulatory domains in Tregs showed that CARs with
CD28 had the most pronounced activity. The suppres-
sive properties of these cells were further increased by
the expression of IL-10 [76]. CAR Tregs with the CD28
domain showed better persistence than CAR Tregs
with the 4-1BB domain (unlike classic CAR T) [77,78].
Tregs expressing third-generation CARs with both CD28
and 4-1BB costimulatory domains are currently being
studied [79-81].
In the decade since the first CAR Treg cells were
generated, several CAR Tregs with different specifici-
ties have been obtained. These cells are characterized
by a high efficacy, stability, and increased persistence
in mouse models [82]. CAR Tregs are also character-
ized by a reduced requirement for IL-2 compared to
CAR T [83]. CAR Treg prevent or reduce the symptoms
of GVHD, disorders associated with the excessive activ-
ity of the immune system, hemophilia, and other dis-
eases in mouse models [73]. Currently, only one CAR
Treg study has entered clinical trial (NCT04817774),
which aims to assess the efficacy and safety of CAR
Treg in kidney transplantation. In this research, CARs
recognize HLA-A2 in the recipient organ, so that CAR
Tregs are attracted to the antigen and suppress the
immune response.
Mucosal-associated T cells. Mucosal-associated in-
variant T cells (MAIT) recognize metabolites of vita-
mins B2 and B9 of bacterial origin in a complex with
the non-classical MHC molecule MR1 [84]. MAIT cells
account for 1 to 8% of peripheral blood T cells [85] and
are localized to mucous membranes and lymphoid tis-
sues. MAIT cell TCRs consist of non-polymorphic α and
β chains; most MAIT cells also express CD8. They also
carry granzyme-containing lytic granules and have
cytotoxic properties [86]. When activated, MAIT cells
secrete the pro-inflammatory cytokines IFNγ, TNF,
and IL-17 [87].
MAIT cells are involved in protection against most-
ly bacterial infections and in the pathogenesis of many
non-communicable diseases, such as autoimmune dis-
orders, inflammatory bowel disease, celiac disease, and
cancer [84]. The level of MAIT cells in the blood decreas-
es in colorectal cancer. MAIT cells migrate into the tu-
mors, where they exert their antitumor cytotoxic effect
by releasing granzymes [88]. Areduction in the number
of MAIT cells is also observed in patients with multiple
myeloma [89]. In some cases, actively migrating MAIT
cells become immunosuppressed by the tumor micro-
environment and lose the ability to secrete IFNγ [90].
The fact that MAIT cells actively infiltrate tumor
microenvironment and have the cytotoxic properties
suggests that they can be used in the development
of novel anti-tumor immunotherapeutic approaches.
Since MAIT cells only recognize antigens associat-
ed with the non-classical MHC class I molecule MR1,
the likelihood of GVHD development when using CAR
MAIT cells would be lower than in the case of cytotox-
ic CAR T cells [23]. Dogan et al. [86] expressed CARs
against CD19 or HER2 antigens in primary human
MAIT cells and showed that the resulting cells were
cytotoxic effects against B cell lymphoma and breast
cancer cells, respectively. The activated CAR MAIT cells
were not inferior to CD8
+
CAR T lymphocytes in their
cytotoxicity against target cells, but produced signifi-
cantly lower levels of IFNγ and other pro-inflammato-
ry cytokines. This latter observation suggests that the
likelihood of CRS and neurotoxicity following injection
of CAR MAIT cells into the patient’s body may be low-
er than with CAR T cells [87]. Further evaluation of
the safety and antitumor properties of CAR MAIT cells
should be performed in in vivo in laboratory animals
and in clinical trials.
Double negative (DN) T cells. DN T cells are char-
acterized by CD3 expression in the absence of CD4 and
CD8 expression. DN T cells also express CD25 [91], a
molecule that is highly expressed by Tregs. TCRs of DN
T cells can contain both αβ and γδ chains. DN Tcells
make up 3 to 5% of T cells in the blood [92]. They ex-
ert a strong suppressive effect on several groups of
immune cells (CD4
+
and CD8
+
T cells, B cells, and NK
cells) both in vitro and in vivo. Thus, DN T cells play
an important role in the prevention of GVHD and the
maintenance of tolerance to allografts and xenografts
[93-95]. Thus, DN T cells can be considered as a non-
canonical subset of Tregs.
Despite their immunosuppressive effect on many
immune cells, DN T cells possess their own MHC-in-
dependent cytotoxic properties. DN T cells can induce
malignant cell death via FasL [96], TRAIL, and other
cytotoxicity-associated surface proteins [97]. They also
express perforin and granzymes and secrete TNF and
IFNγ [97]. In addition to MHC-independent cytotoxici-
ty, DN T cells have other properties, such as the possi-
bility of easy expansion ex vivo [98], lack of extra-tu-
mor cytotoxic activity, and reduced risk of rejection
after allogeneic transfer [99], which allow their use in
immunotherapy of malignant diseases.
The possibility of CAR expression in DN T cells
has been poorly investigated. In 2022, Vasic etal. [99]
reported the generation of DN T cells with the CD19-
directed CAR and compared their antitumor proper-
ties with traditional CAR T cells directed against the
same antigen. The authors found that the CAR DN
Tcells were not inferior to conventional CAR T cells in
their in vitro and in vivo anti-tumor properties, but did
CAR CELLS BEYOND CLASSICAL CAR T CELLS 773
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
not cause GVHD [99]. However, these results require
independent confirmation by other investigators.
Cytokine-induced killer (CIK) cells are a hetero-
geneous group of CD8
+
T cells that are phenotypically
and functionally close to NK cells [100, 101]. CIKs are
derived from CD3
+
T lymphocytes that begin to express
CD56 during expansion. Initially, CIKs were derived
from the lymphokine-activated killer cells (LAKs, lym-
phocytes with the ability to lyse tumor cells after in-
cubation with IL-2 [102]) during the optimization of
the expansion protocol. LAKs had already been ob-
tained in the 1980s [103], but despite their strong cy-
totoxic properties against tumor cells, the difficulties
in expanding these cells exvivo have limited their use.
The timed addition of IFNγ, IL-2, and anti-CD3 mono-
clonal antibody (which had the mitogenic effect) to
LAKs led to the development of CIKs [104]. Currently,
these cells can be produced relatively cheaply from
peripheral or umbilical cord blood T cells.
The cytotoxic effect of CIKs requires the involve-
ment of the NKG2D receptor and is manifested by the
release of the contents of granzyme-containing lytic
granules. NKG2D recognizes stress-induced molecules,
such as UL16BP and MICA/MICB, on the surface of tar-
get cells [105]. As TCR signaling is also possible, so the
use of CIKs has for a long time been limited to autol-
ogous therapy due to the fear of acute GVHD. How-
ever, it has been recently been shown that the use of
donor-based CIKs after hematopoietic stem cells al-
lotransplantation is relatively safe, which may be due
to the short persistence of terminally differentiated
CD3
+
CD56
+
cells and the reduced expression of some
homing receptors [106].
Due to their strong cytotoxic properties, rapid
ex vivo expansion, tumor cell recognition via NKG2D,
and safety of allogeneic adoptive transfer, CIKs are
considered as promising effectors for CAR cell thera-
py [107]. Several preclinical studies have demonstrat-
ed the efficacy of CAR CIKs against hematological and
solid tumors. For example, CIKs expressing a CAR di-
rected against CD19 and containing CD28/4-1BB and
CD3ζ domains efficiently destroyed B cell acute lym-
phoblastic leukemia cells [108,109]. CIKs bearing CARs
to relevant tumor antigens effectively destroyed colon
cancer cells, acute myeloid leukemia cells, soft tissue
sarcoma cells, and cells of other tumor [107]. Expres-
sion of CARs in CIKs leads to increased production of
IFNγ and TNF compared to CIKs that do not express
CARs [110,111].
As of 2022, there is only one clinical trial inves-
tigating the efficacy and safety of CD19-directed CAR
CIKs in acute lymphoblastic leukemia (NCT03389035).
Further studies are therefore needed to provide infor-
mation on the therapeutic efficacy of CAR CIKs.
Macrophages as specialized phagocytic cells that
heavily infiltrate the stroma of solid tumors and are
an important component of the tumor microenviron-
ment. Following recognition of foreign agents by in-
nate immune receptors (mainly Toll-like and NOD-like
receptors), macrophages become activated and start to
produce pro-inflammatory cytokines (TNF, IL-1β, IL-6,
IL-12, and IL-23) that affect many cells in the tumor mi-
croenvironment and in particular promote the antitu-
mor activity of T cells and NK cells. Macrophages with
the pro-inflammatory properties have the so-called
M1 phenotype, whereas macrophages with M2 phe-
notype inhibit inflammation and the development of
antitumor T cell immunity and promote tumor growth
by producing IL-4, IL-5, and IL-13 cytokines. The ma-
jor physiological role of M2 macrophages is wound
healing [112]. Macrophages in the tumor microenvi-
ronment are known as tumor-associated macrophages
(TAMs). The TAMs with M2 phenotype promote tumor
growth and metastasis and has the immunosuppres-
sive properties [66]. Therefore, polarization of TAMs
towards the M1 phenotype, which contributes to the
destruction of neoplasia, is an important component
of the anti-tumor immunotherapy using these cells.
The phagocytic capacity of macrophages can be
enhanced by the expression of CARs bearing intracel-
lular domains that trigger phagocytosis-activating sig-
naling. In the one of the first studies focusing on CAR
macrophages (CAR Ms), primary human monocytes
were transduced with a CAR directed against carci-
noembryonic antigen (CEA) and bearing CD64 (FcγRI)
as a signaling domain. The resulting CAR Ms showed
antitumor activity in vitro and in vivo, although the
molecular mechanism of this activity remained un-
clear [113]. Further studies showed that expression
of CARs containing the phagocytic receptor Megf10 or
FcγR as signaling domains promoted the phagocytic
activity of macrophages [114, 115]. Phagocytosis was
also activated by the expression of CARs with the CD3ζ
signaling domain, which is homologous to FcγRI.
Macrophages expressing CARs directed against
CD19 with Megf10 or FcγR domains efficiently de-
stroyed CD19-positive tumor cells. Phagocytosis was
only observed for a small fraction of malignant cells,
while the majority were destroyed by trogocytosis
[116]. Zhang etal. [116] derived macrophages express-
ing second-generation anti-CD19 CAR and possessing
CD3ζ signaling and 4-1BB costimulatory domains from
the induced pluripotent stem cells (iPSCs). These mac-
rophages phagocytized tumor cells in culture and pro-
duced pro-inflammatory cytokines, but had little effect
on the tumor growth invivo [117].
It has been shown that the process of CAR M gen-
eration itself induces their polarization into pro-in-
flammatory cells. In particular, it was shown that trans-
duction with a CAR-encoding adenoviral vector and
subsequent CAR expression polarized macrophages
towards the M1 phenotype, which was maintained
MININA et al.774
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
in the tumor microenvironment [118]. In addition,
CAR Ms are capable of cross-presenting tumor anti-
gens after phagocytosis. Macrophages expressing CARs
directed against HER2 (breast cancer antigen) phago-
cytized tumor cells and presented the processed an-
tigens. CAR Ms have been shown to activate systemic
antitumor immunity [119]. A single administration of
CAR Ms in a mouse model of ovarian cancer signifi-
cantly prolonged animal survival and slowed (but did
not completely suppress) tumor development [115].
Nevertheless, the ability of CAR Ms to transform the
tumor microenvironment into a pro-inflammatory one
makes them an additional tool in the CAR T cell thera-
py or other immunotherapies.
Macrophages can produce matrix metallopro-
teinases that cause significant changes in the tumor
extracellular matrix and affect the architecture of the
neoplasm, which can be exploited in the CAR therapy.
Expression of CARs directed against HER2 and contain-
ing CD147 as a transmembrane/intracellular domain in
macrophages stimulated the secretion of matrix metal-
loproteinases by these cells. Using a mouse model of
breast cancer, it was shown that administration of these
CAR Ms led to the tumor matrix rearrangement, which
promoted T cell infiltration into the tumor. Levels of
CRS-associated pro-inflammatory cytokines (IFNγ, TNF,
and IL-6) in the blood of mice with breast cancer were
reduced, while the levels of IL-12 and IFNγ in the tumor
were increased [120]. It is reasonable to assume that
CAR M therapy is associated with a reduced risk of CRS.
At present, there is not enough information to
draw conclusions about the efficacy and safety of CAR
M therapy in humans. The fact that macrophages are
one of the main cell types of that trigger the CRS re-
quires a detailed review of the safety of CAR M thera-
py. In addition, peripheral blood monocytes are hetero-
geneous, and there is a possibility that CAR Ms derived
from these cells may migrate to healthy tissues more
often than to the tumor when used systemically [23],
which may lead to significant side effects in different
organ systems. To date, only one HER2-overexpressing
CAR M against solid tumors (NCT04660929) has en-
tered the clinical trial.
Dendritic cells. As “professional” antigen-present-
ing cells, DCs are involved in the maturation of naive
T cells and the reactivation of memory T cells. During
antigen presentation to T cells, DCs produce cytokines
that modulate T cell activity. When present in the tu-
mor microenvironment, DCs can both induce immune
tolerance and contribute to the development of the an-
titumor immune response [121,122]. In addition, DCs
are able to cross-present tumor antigens to the cyto-
toxic CD8
+
T cells, thereby promoting the antitumor
response [123].
The potential of CAR-expressing DCs in controlling
of the antitumor activity of CAR T cells has been poor-
ly investigated. Suh et al. [123] investigated the pos-
sibility of using CAR-expressing DCs (CAR DCs) to at-
tract to the tumor microenvironment and to activate
CAR T cells through the cytokine secretion. Expression
of CARs against CD33 on DCs promoted the homing
of these cells to the bone marrow in mice with acute
myeloid leukemia. Mice receiving CAR DCs and CAR
T cells targeting CD33 had significantly elevated lev-
els of IL-12, IFNγ, and TNF, and the overall survival of
these animals was longer than that of mice receiving
CAR T cells alone. Thus, co-administration of CAR DCs
and CAR T cells significantly increased the efficiency of
malignant cell destruction [124]. The safety of clinical
application of CAR DCs should be investigated, as IL-12
secreted in high concentrations may have a systemic
toxic effect [125]. To date, there are no registered clini-
cal trials focusing on CAR DCs.
B cells. In addition to T cells and innate immune
cells, B cells are also of interest for CAR technology.
Because of their ability to differentiate into long-lived
antibody-secreting plasma cells, B lymphocytes could
become a safe and controlled source of therapeutic
monoclonal antibodies. By expressing CARs against tu-
mor antigens in these cells, targeted antibody delivery
can avoid the side effects of systemic antibody admin-
istration.
Despite these prospects, the studies on the CAR-ex-
pressing B cells and their therapeutic use are current-
ly limited. The possibility of lentiviral transduction of
B cells and subsequent expression of CARs in them
was described in 2018, when a patient experienced a
relapse of B cell lymphoma caused by a single B cell
clone accidentally transduced with a CAR [126]. Soon
after, Pesch etal. [126] demonstrated that genes encod-
ing chimeric B cell receptors (CBCRs) could be insert-
ed into B cells using the CRISPR/Cas9 genome editing
technology. The CBCR used in this study contained the
transmembrane region CD28 and the BCR signaling do-
main CD79β instead of the classical CD3ζ. Introducing
of the gene encoding this CBCR into primary mouse
B cells resulted in the abundant presence of CBCR on
their surface, allowing the cells to recognize antigens
without the involvement of their own B cell receptors
[127]. The antitumor activity and the therapeutic effi-
cacy of CAR-expressing B cells, as well as their safety
in humans, are currently unknown.
ONE TARGET – MANY TYPES OF CAR CELLS
Early studies focused on the antitumor effects of
CAR-expressing leukocytes of different types have used
a small set of tumor-associated antigens, including
CD19, GD2 and others as CAR targets. At present, CAR
cells that effectively recognize tumor antigens have
been derived from different types of white blood cells.
CAR CELLS BEYOND CLASSICAL CAR T CELLS 775
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
The generation of CAR cells that recognize the same
tumor antigen from different cell sources could pro-
vide scope for selecting the optimal therapy for each
patient, as the effector properties of different groups
of white blood cells are different.
Four out of the six FDA-approved CAR T cell ther-
apies target the CD19 antigen and are designed to treat
B cell neoplasias. Despite proven high efficacy, the
anti- CD19 CAR T cell therapy is associated with serious
side effects, primarily CRS and neurotoxicity, in a sig-
nificant proportion of patients. For example, the use
of axicabtagene ciloleucel (CAR T cells with the CD28
costimulatory domain and CD3ζ signaling domain)
causes CRS in 93% of patients and neurotoxicity in
67% [128,129]. To date, CAR cells that efficiently rec-
ognize and destroy CD19-positive malignant cells have
been derived from γδ T cells [52], MAIT cells [87], NKT
cells [68], NK cells [29], DN T cells [100], CIKs [108]
and macrophages [114, 115]. Tregs expressing CD19
have been engineered to suppress the antibody pro-
duction and prevent GVHD [130].
Preclinical studies and studies in small groups of
patients, have shown that anti-CD19 CAR cells, which
are different from the cytotoxic T cells, have a strong
antitumor effect with fewer side effects than the CAR
T cell therapy. However, many of the cell products
mentioned above have not yet reached full-scale clin-
ical trials, while those that have are still in the early
stages of clinical testing, so it is too early to draw con-
clusions about their safety and efficacy.
Two other FDA-approved CAR T cell therapies,
idecabtagene vicleucel and ciltacabtagene autoleucel,
which target the BCMA antigen, have been developed
for the treatment of multiple myeloma. As with the an-
ti-CD19 CAR T therapy, the high efficacy of these drugs
is associated with a significant risk of CRS or neuro-
toxicity: 76 and 42% for idecabtagene vicleucel and
92 and 20% for ciltacabtagene autoleucel, respectively
[131,132]. It has been reported that CAR NK cells tar-
geting BCMA have been obtained [133], but have not
yet entered clinical trials.
There are also CAR cells targeting the same tu-
mor-associated antigen, derived from different types
of white blood cells, intended for the therapy of solid
tumors. For example, GD2-recognizing CAR cells have
been derived from γδ T cells and NKT cells. Currently,
GD2-recognizing CAR NKT cells are in phaseI clinical
trials, so the safety and efficacy of both cell products
remain to be investigated.
CONCLUSION
Over the past 20 years, the CAR T cell approach
has been marked by a recognized success in the treat-
ment of hematologic cancers, and six CAR T cell ther-
apies have been approved for clinical use by the FDA.
Despite the drawbacks of “traditional” CAR T cell ther-
apy, the possibility of generating CAR cells from other
types of white blood cells has long been overlooked.
The results of preclinical studies of the efficacy and
safety of CAR cells in cell culture and mouse models
suggest a wide range of applications for CAR cells in
the treatment of human cancer, including solid tu-
mors. Data on the expression of CARs against different
targets and in different leukocyte subsets are summa-
rized in Table 1 in the article by Qin et al. [33]. Cur-
rently, existing cellular products are only in phaseI/II
of clinical trials, while the vast majority of therapeutic
drugs based on CAR-expressing immune cells have not
yet progressed beyond the preclinical studies.
Although alternative CAR cell products have sig-
nificant advantages over traditional CAR T cells (e.g.,
reduced risk of CRS and neurotoxicity), the latter are
still superior in a number of parameters. Since the sig-
naling pathways underlying the activity of CARs are
different in different groups of adaptive and innate
immune cells, the choice of costimulatory and signal-
ing domains is crucial for the development of CARs in-
tended for the expression in different types of immune
cells. Inclusion of a domain that is not sufficiently func-
tional in a particular cell type can lead to a reduction
in the CAR activity and therapeutic efficacy. For ex-
ample, in macrophages, the Megf10 signaling domain
in the CAR construct is more efficient than the CD3ζ
domain, which has shown optimal activity in T cells.
Therefore, each new CAR-expressing cell may require
“customization” of the receptors and identification of
domains that provide the required level of signaling
from the activated receptor. In the case of traditional
CAR T cells, the choice of domains for CARs has been
much better studied. In addition, some immune cells
are difficult to expand ex vivo, and obtaining sufficient
numbers of these cells can be expensive and time-con-
suming, significantly limiting the ability to produce
clinically relevant products. However, in some cases, it
is possible to find alternative sources of white blood
cells that can at least partially overcome the problem
of cell expansion. For example, CAR NK cells may be
derived from cell lines and iPSCs [20]; CAR Ms may
be derived from monocytes and iPSCs [116]. However,
this method of generating CAR cells may be associat-
ed with a potential risk of tumor formation due to the
high oncogenic potential of iPSCs, and therefore its
safety needs to be further investigated.
Despite significant limitations associated with the
development of CAR-mediated therapies based on dif-
ferent types of white blood cells, these cells have good
prospects in the immunotherapy of solid tumors. The
use of CAR T cells in the treatment of solid tumors has
been largely ineffective due to the insufficient infiltra-
tion of CAR T cells into the tumor, suppressive effects
MININA et al.776
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
of the tumor microenvironment, and heterogeneity
and frequent loss of tumor antigens. Preclinical stud-
ies have shown that many alternative CAR-expressing
immune cells successfully destroy solid tumors. Some
cells, in particular CAR-Ms, can transform tumor mi-
croenvironment into an inflammatory environment
that promotes tumor elimination. The issue of infil-
tration of CAR-expressing immune cells into solid tu-
mors has also not been resolved. Efficient tumor in-
filtration of CAR cells has only been demonstrated for
NK cells and macrophages [117]. Nevertheless, it can
be assumed that CAR leukocytes will become effective
agents for the treatment of solid tumors when used in
combination with other therapeutic approaches.
In conclusion, CAR cell therapy based on different
types of immune cells is moving towards its clinical
application. However, it may be years before alterna-
tive CAR cells become effective, safe, and routinely used
tools in the treatment of cancer and other diseases.
Contributions. E.P.M. and A.V.B. developed the
concept of the review; E.P.M. prepared the manuscript;
S.A.Sh. created illustrations; D.V.D. and A.V.B. edited the
manuscript.
Ethics declarations. This work does not contain
any studies involving human and animal subjects.
Theauthors of this work declare that they have nocon-
flicts of interest.
REFERENCES
1. Kuwana,Y., Asakura,Y., Utsunomiya,N., Nakanishi,M.,
Arata,Y., Itoh, S., Nagase,F., and Kurosawa,Y. (1987)
Expression of chimeric receptor composed of immuno-
globulin-derived Vregions and T-cell receptor- derived
C regions, Biochem. Biophys. Res. Commun., 149, 960-
968, doi:10.1016/0006-291X(87)90502-X.
2. Eshhar, Z., Waks, T., Gross, G., and Schindler, D. G.
(1993) Specific activation and targeting of cytotoxic
lymphocytes through chimeric single chains consist-
ing of antibody-binding domains and the γ or ζ sub-
units of the immunoglobulin and T-cell receptors,
Proc. Natl. Acad. Sci. USA, 90, 720-724, doi: 10.1073/
pnas.90.2.720.
3. Sadelain, M., Brentjens, R., and Riviere, I. (2013)
The basic principles of chimeric antigen recep-
tor design, Cancer Discov., 3, 388-398, doi: 10.1158/
2159-8290.CD-12-0548.
4. Gomes-Silva, D., Mukherjee, M., Srinivasan, M.,
Krenciute,G., Dakhova,O., Zheng, Y., Cabral, J.M.S.,
Rooney, C. M., Orange, J. S., Brenner, M. K., and Ma-
monkin, M. (2017) Tonic 4-1BB costimulation in chi-
meric antigen receptors impedes T cell survival and
is vector-dependent, Cell Rep., 21, 17-26, doi:10.1016/
j.celrep.2017.09.015.
5. Maher, J., Brentjens, R. J., Gunset, G., Rivière, I., and
Sadelain,M. (2002) Human T-lymphocyte cytotoxicity
and proliferation directed by a single chimeric TCRζ/
CD28 receptor, Nat. Biotechnol., 20, 70-75, doi:10.1038/
nbt0102-70.
6. Brentjens, R. J., Latouche, J. B., Santos, E., Marti, F.,
Gong, M. C., Lyddane, C., King, P. D., Larson, S.,
Weiss,M., Riviere,I., and Sadelain,M. (2003) Eradica-
tion of systemic B-cell tumors by genetically targeted
human T lymphocytes co-stimulated by CD80 and in-
terleukin-15, Nat. Med., 9, 279-286, doi:10.1038/nm827.
7. List of FDA approved cell and gene therapies, URL:
https://www.fda.gov/vaccines-blood-biologics/cellular-
gene-therapy-products/approved-cellular-and-gene-
therapy-products.
8. Morgan, R. A., Yang, J. C., Kitano, M., Dudley, M. E.,
Laurencot, C.M., and Rosenberg, S.A. (2010) Case re-
port of a serious adverse event following the adminis-
tration of Tcells transduced with a chimeric antigen
receptor recognizing ERBB2, Mol. Ther., 18, 843-851,
doi:10.1038/mt.2010.24.
9. D’Aloia, M. M., Zizzari, I. G., Sacchetti, B., Pierelli, L.,
and Alimandi, M. (2018) CAR-T cells: the long and
winding road to solid tumors, Cell Death Dis., 9, 282,
doi:10.1038/s41419-018-0278-6.
10. Larson, R.C., and Maus, M.V. (2021) Recent advances
and discoveries in the mechanisms and functions of
CAR Tcells, Nat. Rev. Cancer, 21, 145-161, doi:10.1038/
s41568-020-00323-z.
11. Long, A.H., Haso, W.M., Shern, J.F., Wanhainen, K.M.,
Murgai, M., Ingaramo, M., Smith, J. P., Walker, A. J.,
Kohler, M.E., Venkateshwara, V.R., Kaplan, R.N., Pat-
terson, G.H., Fry, T.J., Orentas, R.J., and Mackall, C.L.
(2015) 4-1BB costimulation ameliorates Tcell exhaus-
tion induced by tonic signaling of chimeric antigen re-
ceptors, Nat. Med., 21, 581-590, doi:10.1038/nm.3838.
12. Hong, M., Clubb, J. D., and Chen, Y. Y. (2020) Engi-
neering CAR-T cells for next-generation cancer ther-
apy, Cancer Cell, 38, 473-488, doi: 10.1016/j.ccell.
2020.07.005.
13. Rafiq,S., Hackett, C.S., and Brentjens, R.J. (2020) En-
gineering strategies to overcome the current road-
blocks in CAR Tcell therapy, Nat. Rev. Clin. Oncol., 17,
147-167, doi:10.1038/s41571-019-0297-y.
14. Meza Guzman, L. G., Keating, N., and Nicholson,
S. E. (2020) Natural killer cells: tumor surveillance
and signaling, Cancers (Basel), 12, 952, doi: 10.3390/
cancers12040952.
15. Kumar,S. (2018) Natural killer cell cytotoxicity and its
regulation by inhibitory receptors, Immunology, 154,
383-393, doi:10.1111/imm.12921.
16. Fauriat, C., Long, E. O., Ljunggren, H. G., and Bry-
ceson, Y. T. (2010) Regulation of human NK-cell cy-
tokine and chemokine production by target cell
recognition, Blood, 115, 2167-2176, doi: 10.1182/
blood-2009-08-238469.
CAR CELLS BEYOND CLASSICAL CAR T CELLS 777
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
17. Lo Nigro, C., Macagno, M., Sangiolo, D., Bertolacci-
ni,L., Aglietta, M., and Merlano, M. C. (2019) NK-me-
diated antibody-dependent cell-mediated cytotoxicity
in solid tumors: biological evidence and clinical per-
spectives, Ann. Transl. Med., 7, 105, doi:10.21037/atm.
2019.01.42.
18. Tyshchuk, E. V., Mikhailova, V. A., Selkov, S. A., and
Sokolov, D. I. (2021) Natural killer cells: origin, phe-
notype, function, Medical Immunology (Russia), 23,
1207-1228, doi:10.15789/1563-0625-NKC-2330.
19. Xu,Y., Liu,Q., Zhong,M., Wang,Z., Chen,Z., Zhang,Y.,
Xing,H., Tian,Z., Tang,K., Liao,X., Rao,Q., Wang,M.,
and Wang, J. (2019) 2B4 costimulatory domain en-
hancing cytotoxic ability of anti-CD5 chimeric antigen
receptor engineered natural killer cells against Tcell
malignancies, J.Hematol. Oncol., 12, 49, doi: 10.1186/
s13045-019-0732-7.
20. Xie, G., Dong, H., Liang, Y., Ham, J. D., Rizwan, R.,
and Chen, J. (2020) CAR-NK cells: a promising cellu-
lar immunotherapy for cancer, EBioMed., 59, 102975,
doi:10.1016/j.ebiom.2020.102975.
21. Töpfer, K., Cartellieri, M., Michen, S., Wiedemuth, R.,
Muller, N., Lindemann, D., Bachmann, M., Fussel,M.,
Schackert, G., and Temme, A. (2015) DAP12-based
activating chimeric antigen receptor for NK cell tu-
mor immunotherapy, J. Immunol., 194, 3201-3212,
doi:10.4049/jimmunol.1400330.
22. Cifaldi, L., Melaiu, O., Giovannoni, R., Benvenu-
to, M., Focaccetti, C., Nardozi, D., Barillari, G., and
Bei, R. (2023) DNAM-1 chimeric receptor-engineered
NKcells: a new frontier for CAR-NK cell-based immu-
notherapy, Front. Immunol., 14, 1197053, doi:10.3389/
fimmu.2023.1197053.
23. Oei, V.Y.S., Siernicka,M., Graczyk-Jarzynka,A., Hoel,
H. J., Yang, W., Palacios, D., Almasbak, H., Bajor, M.,
Clement,D., Brandt,L., Onfelt, B., Goodridge,J., Win-
iarska, M., Zagozdzon, R., Olweus, J., Kyte, J. A., and
Malmberg, K. J. (2018) Intrinsic functional potential
of NK-cell subsets constrains retargeting driven by
chimeric antigen receptors, Cancer Immunol. Res., 6,
467-480, doi:10.1158/2326-6066.CIR-17-0207.
24. Xia,J., Minamino,S., and Kuwabara,K. (2020) CAR-ex-
pressing NKcells for cancer therapy: a new hope, Bios-
ci. Trends, 14, 354-359, doi:10.5582/bst.2020.03308.
25. Dong, H., Ham, J. D., Hu, G., Xie, G., Vergara, J., Li-
ang,Y., Ali,A., Tarannum,M., Donner,H., Baginska,J.,
Abdulhamid,Y., Dinh,K., Soiffer, R.J., Ritz,J., Glimch-
er, L. H., Chen, J., and Romee, R. (2022) Memory-like
NKcells armed with a neoepitope-specific CAR exhibit
potent activity against NPM1 mutated acute myeloid
leukemia, Proc. Natl. Acad. Sci. USA, 119, e2122379119,
doi:10.1073/pnas.2122379119.
26. Klopotowska, M., Bajor, M., Graczyk-Jarzynka, A.,
Kraft,A., Pilch,Z., Zhylko,A., Firczuk,M., Baranows-
ka, I., Lazniewski, M., Plewczynski, D., Goral, A.,
Soroczynska, K., Domagala, J., Marhelava, K., Slu-
sarczyk, A., Retecki, K., Ramji, K., Krawczyk, M.,
Temples, M. N., Sharma, B., Lachota, M., Netskar, H.,
Malmberg, K.-J., Zagozdzon, R., and Winiarska, M.
(2022) PRDX-1 supports the survival and antitumor
activity of primary and CAR-modified NK cells under
oxidative stress, Cancer Immunol. Res., 10, 228-244,
doi:10.1158/2326-6066.CIR-20-1023.
27. Ruggeri, L., Capanni, M., Urbani, E., Perruccio, K.,
Shlomchik, W.D., Tosti,A., Posati,S., Rogaia,D., Fras-
soni,F., Aversa,F., Martelli, M.F., and Velardi,A. (2002)
Effectiveness of donor natural killer cell aloreactivi-
ty in mismatched hematopoietic transplants, Science,
295, 2097-2100, doi:10.1126/science.1068440.
28. Miller, J. S., Soignier, Y., Panoskaltsis-Mortari, A.,
McNearney, S. A., Yun, G. H., Fautsch, S. K., McKen-
na, D., Le, C., Defor, T. E., Burns, L. J., Orchard, P. J.,
Blazar, B. R., Wagner, J. E., Slungaard, A., Weisdorf,
D.J., Okazaki, I.J., and McGlave, P.B. (2005) Success-
ful adoptive transfer and invivo expansion of human
haploidentical NKcells in patients with cancer, Blood,
105, 3051-3057, doi:10.1182/blood-2004-07-2974.
29. Liu, E., Marin, D., Banerjee, P., Macapinlac, H. A.,
Thompson, P., Basar, R., Nassif Kerbauy, L., Over-
man, B., Thall, P., Kaplan, M., Nandivada, V., Kaur, I.,
Nunez Cortes,A., Cao,K., Daher,M., Hosing,C., Cohen,
E. N., Kebriaei, P., Mehta, R., Neelapu, S., Nieto, Y.,
Wang, M., Wierda, W., Keating, M., Champlin, R.,
Shpall, E., and Rezvani, K. (2020) Use of CAR-trans-
duced natural killer cells in CD19-positive lymphoid
tumors, N. Engl. J. Med., 382, 545-553, doi: 10.1056/
NEJMoa1910607.
30. Xiao,L., Cen,D., Gan,H., Sun,Y., Huang,N., Xiong,H.,
Jin, Q., Su, L., Liu, X., Wang, K., Yan, G., Dong, T.,
Wu,S., Zhou,P., Zhang, J., Liang,W., Ren,J., Teng, Y.,
Chen, C., and Xu, X. H. (2019) Adoptive transfer of
NKG2D CAR mRNA-engineered natural killer cells in
colorectal cancer patients, Mol. Ther., 27, 1114-1125,
doi:10.1016/j.ymthe.2019.03.011.
31. Wrona,E., Borowiec,M., and Potemski,P. (2021) CAR-
NK cells in the treatment of solid tumors, Int.J. Mol.
Sci., 22, 5899, doi:10.3390/ijms22115899.
32. Ng, Y. Y., Du, Z., Zhang, X., Chng, W. J., and Wang, S.
(2022) CXCR4 and anti-BCMA CAR co-modified natural
killer cells suppress multiple myeloma progression in
a xenograft mouse model, Cancer Gene Ther., 29, 475-
483, doi:10.1038/s41417-021-00365-x.
33. Qin, V.M., D’Souza,C., Neeson, P.J., and Zhu, J.J. (2021)
Chimeric antigen receptor beyond CAR-T cells, Can-
cers (Basel), 13, 404, doi:10.3390/cancers13030404.
34. Melenhorst, J. J., Chen, G. M., Wang, M., Porter, D.L.,
Chen, C., Collins, M. A., Gao, P., Bandyopadhyay, S.,
Sun,H., Zhao,Z., Lundh,S., Pruteanu-Malinici,I., No-
bles, C.L., Maji,S., Frey, N.V., Gill, S.I., Loren, A.W.,
Tian, L., Kulikovskaya, I., Gupta, M., Ambrose, D. E.,
Davis, M. M., Fraietta, J. A., Brogdon, J. L., Young,
R. M., Chew, A., Levine, B. L., Siegel, D.L., Alanio,C.,
MININA et al.778
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
Wherry,E.J., Bushman, F.D., Lacey, S.F., Tan,K., and
June, C. H. (2022) Decade-long leukaemia remissions
with persistence of CD4
+
CAR T cells, Nature, 602,
503-509, doi:10.1038/s41586-021-04390-6.
35. Sabbah, M., Jondreville, L., Lacan, C., Norol, F., Vieil-
lard, V., Roos-Weil, D., and Nguyen, S. (2022) CAR-NK
cells: a chimeric hope or a promising therapy? Cancers
(Basel), 14, 3839, doi:10.3390/cancers14153839.
36. Ebrahimiyan, H., Tamimi, A., Shokoohian, B., Mi-
naei, N., Memarnejadian, A., Hossein-Khannazer, N.,
Hassan,M., and Vosough,M. (2022) Novel insights in
CAR-NK cells beyond CAR-T cell technology; promis-
ing advantages, Int. Immunopharmacol., 106, 108587,
doi:10.1016/j.intimp.2022.108587.
37. Daher,M., Basar,R., Gokdemir,E., Baran,N., Uprety,N.,
Nunez Cortes, A.K., Mendt,M., Kerbauy, L.N., Baner-
jee, P.P., Shanley,M., Imahashi,N., Li,L., Lim, F.L.W.I.,
Fathi,M., Rezvan,A., Mohanty,V., Shen,Y., Shaim,H.,
Lu,J., Ozcan,G., Ensley,E., Kaplan,M., Nandivada,V.,
Bdiwi,M., Acharya,S., Xi,Y., Wan,X., Mak,D., Liu,E.,
Jiang, X.R., Ang,S., Muniz-Feliciano,L., Li,Y., Wang,J.,
Kordasti, S., Petrov, N., Varadarajan, N., Marin, D.,
Brunetti, L., Skinner, R. J., Lyu, S., Silva, L., Turk, R.,
Schubert, M.S., Rettig, G.R., McNeill, M.S., Kurgan,G.,
Behlke, M. A., Li, H., Fowlkes, N. W., Chen, K., Kono-
pleva,M., Champlin, R.E., Shpall, E.J., and Rezvani,K.
(2021) Targeting a cytokine checkpoint enhances the
fitness of armored cord blood CAR-NK cells, Blood,
137, 624-636, doi:10.1182/blood.2020007748.
38. Pulè, M. A., Straathof, K. C., Dotti, G., Heslop, H. E.,
Rooney, C. M., and Brenner, M. K. (2005) A chime-
ric T cell antigen receptor that augments cytokine
release and supports clonal expansion of primary
human T cells, Mol. Ther., 12, 933-941, doi: 10.1016/
j.ymthe.2005.04.016.
39. Li,Y., Basar,R., Wang,G., Liu,E., Moyes, J.S., Li,L., Ker-
bauy, L.N., Uprety,N., Fathi,M., Rezvan,A., Banerjee,
P. P., Muniz-Feliciano, L., Laskowski, T. J., Ensley, E.,
Daher,M., Shanley,M., Mendt,M., Acharya,S., Liu,B.,
Biederstadt,A., Rafei,H., Guo,X., Garcia, L.M., Lin,P.,
Ang,S., Marin,D., Chen,K., Bover,L., Champlin, R.E.,
Varadarajan, N., Shpall, E. J., and Rezvani, K. (2022)
KIR-based inhibitory CARs overcome CAR-NK cell tro-
gocytosis-mediated fratricide and tumor escape, Nat.
Med., 28, 2133-2144, doi:10.1038/s41591-022-02003-x.
40. Li, L., Mohanty, V., Dou, J., Huang, Y., Banerjee, P. P.,
Miao,Q., Lohr, J.G., Vijaykumar,T., Frede,J., Knoech-
el, B., Muniz-Feliciano, L., Laskowski, T. J., Liang, S.,
Moyes, J.S., Nandivada, V., Basar, R., Kaplan, M., Da-
her,M., Liu,E., Li,Y., Acharya,S., Lin,P., Shanley,M.,
Rafei,H., Marin,D., Mielke,S., Champlin, R.E., Shpall,
E. J., Chen, K., and Rezvani, K. (2023) Loss of meta-
bolic fitness drives tumor resistance after CAR-NK
cell therapy and can be overcome by cytokine engi-
neering, Sci. Adv., 9, eadd6997, doi: 10.1126/sciadv.
add6997.
41. Papadopoulou, M., Sanchez Sanchez, G., and Ver-
mijlen, D. (2020) Innate and adaptive γδ T cells:
how, when, and why, Immunol. Rev., 298, 99-116,
doi:10.1111/imr.12926.
42. Simões, A.E., Di Lorenzo,B., and Silva-Santos,B. (2018)
Molecular determinants of Target cell recognition by
human γδTcells, Front. Immunol., 9, 929, doi:10.3389/
fimmu.2018.00929.
43. Chien, Y. H., Meyer, C., and Bonneville, M. (2014)
γδTcells: first line of defense and beyond, Annu. Rev.
Immunol., 32, 121-155, doi:10.1146/annurev-immunol-
032713-120216.
44. Deng,J., and Yin,H. (2022) Gamma delta (γδ) Tcells in
cancer immunotherapy; where it comes from, where
it will go? Eur.J. Pharmacol., 919, 174803, doi:10.1016/
j.ejphar.2022.174803.
45. Sebestyen,Z., Prinz,I., Dechanet-Merville,J., Silva-San-
tos, B., and Kuball, J. (2020) Translating gammadel-
ta(γδ) Tcells and their receptors into cancer cell ther-
apies, Nat. Rev. Drug Discov., 19, 169-184, doi:10.1038/
s41573-019-0038-z.
46. Girardi,M., Oppenheim, D.E., Steele, C.R., Lewis, J.M.,
Glusac,E., Filler,R., Hobby,P., Sutton,B., Tigelaar, R.E.,
and Hayday, A.C. (2001) Regulation of cutaneous ma-
lignancy by gammadelta Tcells, Science, 294, 605-609,
doi:10.1126/science.1063916.
47. Donia,M., Ellebaek,E., Andersen, M.H., Straten, P.T.,
and Svane, I. M. (2012) Analysis of Vdelta1 T cells
in clinical grade melanoma-infiltrating lympho-
cytes, Oncoimmunology, 1, 1297-1304, doi: 10.4161/
onci.21659.
48. Wang,J., Lin,C., Li,H., Li,R., Wu,Y., Liu,H., Zhang,H.,
He,H., Zhang,W., and Xu,J. (2017) Tumor-infiltrating
γδTcells predict prognosis and adjuvant chemother-
apeutic benefit in patients with gastric cancer, On-
coimmunology, 6, e1353858, doi: 10.1080/2162402X.
2017.1353858.
49. Silva-Santos, B., Mensurado, S., and Coffelt, S. B.
(2019) γδ T cells: pleiotropic immune effectors with
therapeutic potential in cancer, Nat. Rev. Cancer, 19,
392-404, doi:10.1038/s41568-019-0153-5.
50. Gunderson, A.J., Kaneda, M.M., Tsujikawa,T., Nguyen,
A. V., Affara, N. I., Ruffell, B., Gorjestani, S., Liudahl,
S.M., Truitt,M., Olson,P., Kim,G., Hanahan,D., Tem-
pero, M.A., Sheppard,B., Irving,B., Chang, B.Y., Var-
ner, J. A., and Coussens, L. M. (2016) Bruton tyrosine
kinase-dependent immune cell cross-talk drives pan-
creas cancer, Cancer Discov., 6, 270-285, doi: 10.1158/
2159-8290.CD-15-0827.
51. Deniger, D.C., Switzer,K., Mi,T., Maiti,S., Hurton,L.,
Singh, H., Huls, H., Olivares, S., Lee, D.A., Champlin,
R.E., and Cooper, L.J. (2013) Bispecific T-cells express-
ing polyclonal repertoire of endogenous γδT-cell re-
ceptors and introduced CD19-specific chimeric an-
tigen receptor, Mol. Ther., 21, 638-647, doi: 10.1038/
mt.2012.267.
CAR CELLS BEYOND CLASSICAL CAR T CELLS 779
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
52. Rozenbaum, M., Meir, A., Aharony, Y., Itzhaki, O.,
Schachter,J., Bank,I., Jacoby,E., and Besser, M.J. (2020)
Gamma-delta CAR-T cells show CAR-directed and in-
dependent activity against leukemia, Front. Immunol.,
11, 1347, doi:10.3389/fimmu.2020.01347.
53. Fisher, J., Abramowski, P., Wisidagamage Don, N. D.,
Flutter, B., Capsomidis, A., Cheung, G. W., Gustafs-
son, K., and Anderson, J. (2017) Avoidance of on-tar-
get off-tumor activation using a co-stimulation-only
chimeric antigen receptor, Mol. Ther., 25, 1234-1247,
doi:10.1016/j.ymthe.2017.03.002.
54. Morandi, F., Yazdanifar, M., Cocco, C., Bertaina, A.,
and Airoldi,I. (2020) Engineering the bridge between
innate and adaptive immunity for cancer immuno-
therapy: focus on γδ T and NK cells, Cells, 9, 1757,
doi:10.3390/cells9081757.
55. Fleischer, L. C., Becker, S.A., Ryan, R.E., Fedanov,A.,
Doering, C. B., and Spencer, H. T. (2020) Non-signal-
ing chimeric antigen receptors enhance antigen-di-
rected killing by γδ T cells in contrast to αβ T cells,
Mol. Ther. Oncolytics, 18, 149-160, doi: 10.1016/j.omto.
2020.06.003.
56. Ali, A.K., Tarannum,M., and Romee,R. (2021) Is adop-
tive cellular therapy with non-T-cell immune effec-
tors the future? Cancer J., 27, 168-175, doi: 10.1097/
PPO.0000000000000517.
57. Vivier, E., and Anfossi, N. (2004) Inhibitory NK-cell
receptors on Tcells: witness of the past, actors of the
future, Nat. Rev. Immunol., 4, 190-198, doi: 10.1038/
nri1306.
58. Kriegsmann, K., Kriegsmann, M., von Bergwelt-Bail-
don,M., Cremer,M., and Witzens-Harig,M. (2018) NKT
cells– new players in CAR cell immunotherapy? Eur.J.
Haematol., 101, 750-757, doi:10.1111/ejh.13170.
59. Yoneda,K., Morii,T., Nieda,M., Tsukaguchi,N., Ama-
no,I., Tanaka,H., Yagi,H., Narita,N., and Kimura,H.
(2005) The peripheral blood Valpha24
+
NKT cell
numbers decrease in patients with haematopoiet-
ic malignancy, Leuk. Res., 29, 147-152, doi: 10.1016/
j.leukres.2004.06.005.
60. Tahir, S.M., Cheng,O., Shaulov,A., Koezuka,Y., Bubley,
G.J., Wilson, S.B., Balk, S.P., and Exley, M. A. (2001)
Loss of IFN-gamma production by invariant NKTcells
in advanced cancer, J. Immunol., 167, 4046-4050,
doi:10.4049/jimmunol.167.7.4046.
61. Tachibana, T., Onodera, H., Tsuruyama, T., Mori, A.,
Nagayama, S., Hiai, H., and Imamura, M. (2005) In-
creased intratumor Valpha24-positive natural kill-
er T cells: a prognostic factor for primary colorec-
tal carcinomas, Clin. Cancer Res., 11, 7322-7327,
doi:10.1158/1078-0432.CCR-05-0877.
62. Schneiders, F. L., de Bruin, R. C., van den Eertwegh,
A.J., Scheper, R. J., Leemans, C. R., Brakenhoff, R. H.,
Langendijk, J. A., Verheul, H. M., de Gruijl, T. D.,
Molling, J.W., and van der Vliet, H.J. (2012) Circulat-
ing invariant natural killer T-cell numbers predict
outcome in head and neck squamous cell carcinoma:
updated analysis with 10-year follow-up, J. Clin. On-
col., 30, 565-567, doi:10.1200/jco.2011.39.3975.
63. Bassiri, H., Das, R., Guan, P., Barrett, D.M., Brennan,
P. J., Banerjee, P. P., Wiener, S. J., Orange, J. S., Bren-
ner, M.B., Grupp, S.A., and Nichols, K.E. (2014) iNKT
cell cytotoxic responses control T-lymphoma growth
in vitro and in vivo, Cancer Immunol. Res., 2, 59-69,
doi:10.1158/2326-6066.CIR-13-0104.
64. Nair, S., and Dhodapkar, M. V. (2017) Natural killer
Tcells in cancer immunotherapy, Front. Immunol., 8,
1178, doi:10.3389/fimmu.2017.01178.
65. McEwen-Smith, R. M., Salio, M., and Cerundolo, V.
(2015) The regulatory role of invariant NKT cells in
tumor immunity, Cancer Immunol. Res., 3, 425-435,
doi:10.1158/2326-6066.CIR-15-0062.
66. Cortés-Selva, D., Dasgupta, B., Singh, S., and Grew-
al, I. S. (2021) Innate and innate-like cells: the fu-
ture of chimeric antigen receptor (CAR) cell thera-
py, Trends Pharmacol. Sci., 42, 45-59, doi: 10.1016/j.
tips.2020.11.004.
67. Heczey, A., Liu, D., Tian, G., Courtney, A. N., Wei, J.,
Marinova, E., Gao, X., Guo, L., Yvon, E., Hicks, J.,
Liu,H., Dotti,G., and Metelitsa, L.S. (2014) Invariant
NKT cells with chimeric antigen receptor provide
a novel platform for safe and effective cancer im-
munotherapy, Blood, 124, 2824-2833, doi: 10.1182/
blood-2013-11-541235.
68. Tian, G., Courtney, A. N., Jena, B., Heczey, A., Liu, D.,
Marinova,E., Guo,L., Xu,X., Torikai,H., Mo, Q., Dot-
ti, G., Cooper, L. J., and Metelitsa, L. S. (2016) CD62L
+
NKT cells have prolonged persistence and antitu-
mor activity in vivo, J. Clin. Invest., 126, 2341-2355,
doi:10.1172/JCI83476.
69. Simon, B., Wiesinger, M., Marz, J., Wistuba-Ham-
precht, K., Weide, B., Schuler-Thurner, B., Schul-
er, G., Dorrie, J., and Uslu, U. (2018) The generation
of CAR-transfected natural killer T cells for the im-
munotherapy of melanoma, Int. J. Mol. Sci., 19, 2365,
doi:10.3390/ijms19082365.
70. Shaik, R. S., Rathi, P., Courtney, A., Schneller, N.,
Guo, L., Barragan, G., Zhang, C., Xu, X., Sumazin, P.,
Metelitsa, L., and Heczey, A. (2022) Glypican-3-spe-
cific CAR-NKT cells overexpressing BATF3 mediate
potent antitumor activity against hepatocellular car-
cinoma, J. Clin. Oncol., 40, e14521, doi: 10.1200/jco.
2022.40.16_suppl.e14521.
71. Itoh,M., Takahashi,T., Sakaguchi,N., Kuniyasu,Y., Shi-
mizu, J., Otsuka, F., and Sakaguchi, S. (1999) Thymus
and autoimmunity: production of CD25
+
CD4
+
natural-
ly anergic and suppressive T cells as a key function
of the thymus in maintaining immunologic self-tol-
erance, J. Immunol., 162, 5317-5326, doi: 10.4049/
jimmunol.162.9.5317.
72. Romano, M., Fanelli,G., Albany, C. J., Giganti, G., and
Lombardi, G. (2019) Past, present, and future of reg-
MININA et al.780
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
ulatory T cell therapy in transplantation and au-
toimmunity, Front. Immunol., 10, 43, doi: 10.3389/
fimmu.2019.00043.
73. Arjomandnejad, M., Kopec, A. L., and Keeler, A. M.
(2022) CAR-T regulatory (CAR-Treg) cells: engineering
and applications, Biomedicines, 10, 287, doi: 10.3390/
biomedicines10020287.
74. Elinav, E., Waks, T., and Eshhar, Z. (2008) Redirec-
tion of regulatory T cells with predetermined spec-
ificity for the treatment of experimental colitis in
mice, Gastroenterology, 134, 2014-2024, doi: 10.1053/
j.gastro.2008.02.060.
75. Hombach, A. A., Kofler, D., Rappl, G., and Abken, H.
(2009) Redirecting human CD4
+
CD25
+
regulatory
Tcells from the peripheral blood with pre-defined tar-
get specificity, Gene Ther., 16, 1088-1096, doi:10.1038/
gt.2009.75.
76. Dawson, N.A.J., Rosado-Sánchez,I., Novakovsky, G.E.,
Fung, V.C.W., Huang,Q., McIver,E., Sun,G., Gillies,J.,
Speck,M., Orban, P.C., Mojibian,M., and Levings, M.K.
(2020) Functional effects of chimeric antigen receptor
co-receptor signaling domains in human regulatory
T cells, Sci. Transl. Med., 12, eaaz3866, doi: 10.1126/
scitranslmed.aaz3866.
77. Mohseni, Y. R., Saleem, A., Tung, S. L., Dudreuilh, C.,
Lang,C., Peng,Q., Volpe,A., Adigbli,G., Cross,A., Hes-
ter, J., Farzaneh, F., Scotta, C., Lechler, R. I., Issa, F.,
Fruhwirth, G. O., and Lombardi, G. (2021) Chimeric
antigen receptor-modified human regulatory T cells
that constitutively express IL-10 maintain their phe-
notype and are potently suppressive, Eur.J. Immunol.,
51, 2522-2530, doi:10.1002/eji.202048934.
78. Boroughs, A. C., Larson, R. C., Choi, B. D., Bouffard,
A. A., Riley, L. S., Schiferle, E., Kulkarni, A. S., Cetru-
lo, C.L., Ting,D., Blazar, B.R., Demehri,S., and Maus,
M. V. (2019) Chimeric antigen receptor costimulation
domains modulate human regulatory Tcell function,
JCI Insight, 5, e126194, doi:10.1172/jci.insight.126194.
79. Fu, R.Y., Chen, A.C., Lyle, M.J., Chen, C.Y., Liu, C.L.,
and Miao, C. H. (2020) CD4
+
T cells engineered with
FVIII-CAR and murine Foxp3 suppress anti-fac-
tor VIII immune responses in hemophilia a mice,
Cell Immunol., 358, 104216, doi: 10.1016/j.cellimm.
2020.104216.
80. Arjomandnejad, M., Sylvia, K., Blackwood, M., Nix-
on,T., Tang,Q., Muhuri,M., Gruntman, A.M., Gao,G.,
Flotte, T. R., and Keeler, A. M. (2021) Modulating im-
mune responses to AAV by expanded polyclonal T-regs
and capsid specific chimeric antigen receptor T-regu-
latory cells, Mol. Ther. Methods Clin. Dev., 23, 490-506,
doi:10.1016/j.omtm.2021.10.010.
81. Rana,J., and Biswas,M. (2020) Regulatory Tcell thera-
py: current and future design perspectives, Cell Immu-
nol., 356, 104193, doi:10.1016/j.cellimm.2020.104193.
82. MacDonald, K.G., Hoeppli, R.E., Huang,Q., Gillies,J.,
Luciani, D. S., Orban, P. C., Broady, R., and Levings,
M. K. (2016) Alloantigen-specific regulatory T cells
generated with a chimeric antigen receptor, J.Clin. In-
vest., 126, 1413-1424, doi:10.1172/JCI82771.
83. D’Souza, C., Chen, Z., and Corbett, A. J. (2018) Re-
vealing the protective and pathogenic potential of
MAIT cells, Mol. Immunol., 103, 46-54, doi: 10.1016/
j.molimm.2018.08.022.
84. Howson, L. J., Salio, M., and Cerundolo, V. (2015)
MR1-restricted mucosal-associated invariant T cells
and their activation during infectious diseases, Front.
Immunol., 6, 303, doi:10.3389/fimmu.2015.00303.
85. Le Bourhis, L., Dusseaux, M., Bohineust, A., Bes-
soles, S., Martin, E., Premel, V., Core, M., Sleurs, D.,
Serriari, N. E., Treiner, E., Hivroz, C., Sansonetti, P.,
Gougeon, M.L., Soudais,C., and Lantz,O. (2013) MAIT
cells detect and efficiently lyse bacterially-infected ep-
ithelial cells, PLoS Pathog., 9, e1003681, doi: 10.1371/
journal.ppat.1003681.
86. Dogan, M., Karhan, E., Kozhaya, L., Placek, L.,
Chen, X., Yigit, M., and Unutmaz, D. (2022) Engi-
neering human mucosal associated invariant T
(MAIT) cells with chimeric antigen receptors for
cancer immunotherapy, J. Immunol., 209, 1523-1531,
doi:10.1101/2022.07.28.501764.
87. Won, E. J., Ju, J. K., Cho, Y. N., Jin, H. M., Park, K. J.,
Kim, T. J., Kwon, Y.-S., Kee, H. J., Kim, J. C., Kee, S.-J.,
and Park, Y.-W. (2016) Clinical relevance of circulating
mucosal-associated invariant T cell levels and their
anti-cancer activity in patients with mucosal-associ-
ated cancer, Oncotarget, 7, 76274-76290, doi:10.18632/
oncotarget.11187.
88. Gherardin, N.A., Loh,L., Admojo,L., Davenport, A.J.,
Richardson,K., Rogers,A., Darcy, P.K., Jenkins, M.R.,
Prince, H. M., Harrison, S.J., Quach,H., Fairlie, D.P.,
Kedzierska, K., McCluskey, J., Uldrich, A. P., Neeson,
P. J., Ritchie, D. S., and Godfrey, D. I. (2018) Enumer-
ation, functional responses and cytotoxic capacity
of MAIT cells in newly diagnosed and relapsed mul-
tiple myeloma, Sci. Rep., 8, 4159, doi: 10.1038/s41598-
018-22130-1.
89. Reantragoon, R., Boonpattanaporn, N., Corbett, A. J.,
and McCluskey,J. (2016) Mucosal-associated invariant
Tcells in clinical diseases, As. Pac.J. Allergy Immunol.,
34, 3-10.
90. Zhang, Z.-X., Yang, L., Young, K. J., Dutemple, B., and
Zhang, L. I. (2000) Identification of a previously un-
known antigen-specific regulatory T cell and its
mechanism of suppression, Nat. Med., 6, 782-789,
doi:10.1038/77513.
91. Wu, Z., Zheng, Y., Sheng, J., Han, Y., Yang, Y., Pan, H.,
and Yao, J. (2022) CD3
+
CD4
CD8
(Double-Negative)
T cells in inflammation, immune disorders and can-
cer, Front. Immunol., 13, 816005, doi: 10.3389/fimmu.
2022.816005.
92. Zhang, Z.-X., Yang,L., Young, K.J., and Zhang,L. (2001)
Suppression of alloimmune responses in vitro and
CAR CELLS BEYOND CLASSICAL CAR T CELLS 781
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
invivo by CD3CD8CD4 regulatory Tcells, Transplant.
Proc., 33, 84-85, doi:10.1016/s0041-1345(00)01915-1.
93. Young, K.J., DuTemple,B., Phillips, M.J., and Zhang,L.
(2003) Inhibition of graft-versus-host disease by dou-
ble-negative regulatory T cells, J.Immunol., 171, 134-
141, doi:10.4049/jimmunol.171.1.134.
94. Chen, W., Ford, M. S., Young, K. J., Cybulsky, M. I.,
and Zhang, L. (2003) Role of double-negative reg-
ulatory T cells in long-term cardiac xenograft sur-
vival, J. Immunol., 170, 1846-1853, doi: 10.4049/
jimmunol.170.4.1846.
95. Chen, J., Hu,P., Wu,G., and Zhou,H. (2019) Antipan-
creatic cancer effect of DNT cells and the underlying
mechanism, Pancreatology, 19, 105-113, doi: 10.1016/
j.pan.2018.12.006.
96. Chen, X., Wang, D., and Zhu,X. (2022) Application of
double-negative T cells in haematological malignan-
cies: recent progress and future directions, Biomark.
Res., 10, 11, doi:10.1186/s40364-022-00360-w.
97. Merims, S., Li, X., Joe, B., Dokouhaki, P., Han, M.,
Childs, R. W., Wang, Z. Y., Gupta, V., Minden, M. D.,
and Zhang, L. (2011) Anti-leukemia effect of ex vivo
expanded DNT cells from AML patients: a potential
novel autologous T-cell adoptive immunotherapy,
Leukemia, 25, 1415-1422, doi:10.1038/leu.2011.99.
98. Lee, J.B., Kang,H., Fang,L., D’Souza,C., Adeyi,O., and
Zhang, L. (2019) Developing allogeneic double-neg-
ative Tcells as a novel off-the-shelf adoptive cellular
therapy for cancer, Clin. Cancer Res., 25, 2241-2253,
doi:10.1158/1078-0432.CCR-18-2291.
99. Vasic,D., Lee, J.B., Leung,Y., Khatri,I., Na,Y., Abate-Da-
ga,D., and Zhang,L. (2022) Allogeneic double-negative
CAR-T cells inhibit tumor growth without off-tumor
toxicities, Sci. Immunol., 7, eabl3642, doi: 10.1126/
sciimmunol.abl3642.
100. Gao, X., Mi, Y., Guo, N., Xu, H., Xu, L., Gou, X., and
Jin,W. (2017) Cytokine-induced killer cells as pharma-
cological tools for cancer immunotherapy, Front. Im-
munol., 8, 774, doi:10.3389/fimmu.2017.00774.
101. Grimm, E.A., Mazumder,A., Zhang, H.Z., and Rosen-
berg, S.A. (1982) Lymphokine-activated killer cell phe-
nomenon. Lysis of natural killer-resistant fresh solid
tumor cells by interleukin 2-activated autologous hu-
man peripheral blood lymphocytes, J.Exp. Med., 155,
1823-1841, doi:10.1084/jem.155.6.1823.
102. Rosenberg, S. A., Spiess, P., and Lafreniere, R. (1986)
A new approach to the adoptive immunotherapy of
cancer with tumor-infiltrating lymphocytes, Science,
233, 1318-1321, doi:10.1126/science.3489291.
103. Cappuzzello,E., Sommaggio,R., Zanovello,P., and Ro-
sato, A. (2017) Cytokines for the induction of antitu-
mor effectors: the paradigm of cytokine-induced kill-
er (CIK) cells, Cytokine Growth Factor Rev., 36, 99-105,
doi:10.1016/j.cytogfr.2017.06.003.
104. Diefenbach,A., Jamieson, A.M., Liu, S.D., Shastri,N.,
and Raulet, D.H. (2000) Ligands for the murine NKG2D
receptor: expression by tumor cells and activation of
NKcells and macrophages, Nat. Immunol., 1, 119-126,
doi:10.1038/77793.
105. Merker,M., Wagner,J., Kreyenberg,H., Heim,C., Mos-
er, L. M., Wels, W. S., Bonig, H., Ivics, Z., Ullrich, E.,
Klingebiel, T., Bader, P., and Rettinger, E. (2020)
ERBB2-CAR-engineered cytokine-induced killer cells
exhibit both CAR-mediated and innate immunity
against high-risk rhabdomyosarcoma, Front. Immu-
nol., 11, 581468, doi:10.3389/fimmu.2020.581468.
106. Rotolo, R., Leuci, V., Donini, C., Cykowska, A., Gam-
maitoni, L., Medico, G., Valabrega, G., Aglietta, M.,
and Sangiolo, D. (2019) CAR-based strategies beyond
T lymphocytes: integrative opportunities for cancer
adoptive immunotherapy, Int. J. Mol. Sci., 20, 2839,
doi:10.3390/ijms20112839.
107. Marin,V., Dander, E., Biagi, E., Introna, M., Fazio, G.,
Biondi,A., and D’Amico,G. (2006) Characterization of
in vitro migratory properties of anti-CD19 chimeric
receptor-redirected CIK cells for their potential use in
B-ALL immunotherapy, Exp. Hematol., 34, 1219-1229,
doi:10.1016/j.exphem.2006.05.004.
108. Marin, V., Kakuda, H., Dander, E., Imai, C., Campa-
na,D., Biondi,A., and D’Amico,G. (2007) Enhancement
of the anti-leukemic activity of cytokine induced killer
cells with an anti-CD19 chimeric receptor delivering a
4-1BB-zeta activating signal, Exp. Hematol., 35, 1388-
1397, doi:10.1016/j.exphem.2007.05.018.
109. Marin, V., Pizzitola, I., Agostoni, V., Attianese, G. M.,
Finney, H., Lawson, A., Pule, M., Rousseau, R., Bion-
di, A., and Biagi, E. (2010) Cytokine-induced killer
cells for cell therapy of acute myeloid leukemia: im-
provement of their immune activity by expression of
CD33-specific chimeric receptors, Haematologica, 95,
2144-2152, doi:10.3324/haematol.2010.026310.
110. Magnani, C. F., Mezzanotte, C., Cappuzzello, C., Bar-
dini,M., Tettamanti, S., Fazio,G., Cooper, L. J. N., Da-
stoli,G., Cazzaniga,G., Biondi,A., and Biagi,E. (2018)
Preclinical efficacy and safety of CD19CAR cytokine-
induced killer cells transfected with sleeping beauty
transposon for the treatment of acute lymphoblastic
leukemia, Hum. Gene Ther., 29, 602-613, doi: 10.1089/
hum.2017.207.
111. Jarosz-Biej, M., Kaminska, N., Matuszczak, S., Ci-
chon, T., Pamula-Pilat, J., Czapla, J., Smolarczyk, R.,
Skwarzynska,D., Kulik,K., and Szala,S. (2018) M1-like
macrophages change tumor blood vessels and mi-
croenvironment in murine melanoma, PLoS One, 13,
e0191012, doi:10.1371/journal.pone.0191012.
112. Biglari, A., Southgate, T. D., Fairbairn, L. J., and Gil-
ham, D. E. (2006) Human monocytes expressing a
CEA-specific chimeric CD64 receptor specifically tar-
get CEA-expressing tumour cells in vitro and invivo,
Gene Ther., 13, 602-610, doi:10.1038/sj.gt.3302706.
113. Morrissey, M. A., Williamson, A. P., Steinbach, A. M.,
Roberts, E.W., Kern,N., Headley, M.B., and Vale, R.D.
MININA et al.782
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
(2018) Chimeric antigen receptors that trigger phago-
cytosis, Elife, 7, e36688, doi:10.7554/eLife.36688.
114. Klichinsky, M., Ruella, M., Shestova, O., Lu, X. M.,
Best,A., Zeeman,M., Schmierer,M., Gabrusiewicz,K.,
Anderson, N.R., Petty, N.E., Cummins, K.D., Shen,F.,
Shan,X., Veliz,K., Blouch,K., Yashiro-Ohtani,Y., Ken-
derian, S.S., Kim, M.Y., O’Connor, R.S., Wallace, S.R.,
Kozlowski, M.S., Marchione, D.M., Shestov,M., Garcia,
B.A., June, C.H., and Gill,S. (2020) Human chimeric
antigen receptor macrophages for cancer immuno-
therapy, Nat. Biotechnol., 38, 947-953, doi: 10.1038/
s41587-020-0462-y.
115. Cannac,M., Nikolic,J., and Benaroch,P. (2022) Cancer
immunotherapies based on genetically engineered
macrophages, Cancer Immunol. Res., 10, 1156-1166,
doi:10.1158/2326-6066.CIR-22-0030.
116. Zhang, L., Tian, L., Dai, X., Yu, H., Wang, J., Lei, A.,
Zhu, M., Xu, J., Zhao, W., Zhu, Y., Sun, Z., Zhang, H.,
Hu, Y., Wang, Y., Xu, Y., Church, G. M., Huang, H.,
Weng, Q., and Zhang, J. (2020) Pluripotent stem cell-
derived CAR-macrophage cells with antigen-depen-
dent anti-cancer cell functions, J.Hematol. Oncol., 13,
153, doi:10.1186/s13045-020-00983-2.
117. Hagemann,T., Lawrence,T., McNeish,I., Charles, K.A.,
Kulbe,H., Thompson, R.G., Robinson, S.C., and Balk-
will, F.R. (2008) “Re-educating” tumor-associated mac-
rophages by targeting NF-kappaB, J. Exp. Med., 205,
1261-1268, doi:10.1084/jem.20080108.
118. Niu,Z., Chen,G., Chang,W., Sun,P., Luo,Z., Zhang,H.,
Zhi,L., Guo,C., Chen,H., Yin,M., and Zhu, W. (2021)
Chimeric antigen receptor-modified macrophages
trigger systemic anti-tumour immunity, J.Pathol., 253,
247-257, doi:10.1002/path.5585.
119. Zhang, W., Liu, L., Su, H., Liu, Q., Shen, J., Dai, H.,
Zheng, W., Lu, Y., Zhang, W., Bei, Y., and Shen, P.
(2019) Chimeric antigen receptor macrophage ther-
apy for breast tumours mediated by targeting the tu-
mour extracellular matrix, Br.J. Cancer, 121, 837-845,
doi:10.1038/s41416-019-0578-3.
120. Williford, J.M., Ishihara,J., Ishihara,A., Mansurov,A.,
Hosseinchi,P., Marchell, T.M., Potin,L., Swartz, M.A.,
and Hubbell, J.A. (2019) Recruitment of CD103
+
den-
dritic cells via tumor-targeted chemokine delivery en-
hances efficacy of checkpoint inhibitor immunothera-
py, Sci. Adv., 5, eaay1357, doi:10.1126/sciadv.aay1357.
121. Maier,B., Leader, A.M., Chen, S.T., Tung,N., Chang,C.,
LeBerichel,J., Chudnovskiy,A., Maskey,S., Walker,L.,
Finnigan, J. P., Kirkling, M. E., Reizis, B., Ghosh, S.,
D’Amore, N. R., Bhardwaj, N., Rothlin, C. V., Wolf, A.,
Flores, R., Marron, T., Rahman, A. H., Kenigsberg, E.,
Brown, B.D., and Merad,M. (2020) Aconserved den-
dritic-cell regulatory program limits antitumour
immunity, Nature, 580, 257-262, doi: 10.1038/s41586-
020-2134-y.
122. Hildner,K., Edelson, B.T., Purtha, W.E., Diamond,M.,
Matsushita, H., Kohyama, M., Calderon, B., Schraml,
B. U., Unanue, E.R., Diamond, M.S., Schreiber, R. D.,
Murphy, T. L., and Murphy, K. M. (2008) Batf3 defi-
ciency reveals a critical role for CD8α
+
dendritic cells
in cytotoxic T cell immunity, Science, 322, 1097-1100,
doi:10.1126/science.1164206.
123. Suh, H. C., Pohl, K. A., Termini, C., Kan, J., Timmer-
man, J. M., Slamon, D. J., and Chute, J. P. (2018) Bio-
engineered autologous dendritic cells enhance car
t cell cytotoxicity by providing cytokine stimulation
and intratumoral dendritic cells, Blood, 132, 3693,
doi:10.1182/blood-2018-99-115296.
124. Zhang,L., Morgan, R.A., Beane, J. D., Zheng,Z., Dud-
ley, M. E., Kassim, S.H., Nahvi, A.V., Ngo, L. T., Sher-
ry, R. M., Phan, G. Q., Hughes, M.S., Kammula, U.S.,
Feldman, S. A., Toomey, M. A., Kerkar, S. P., Restifo,
N.P., Yang, J.C., and Rosenberg, S.A. (2015) Tumor-in-
filtrating lymphocytes genetically engineered with an
inducible gene encoding interleukin-12 for the immu-
notherapy of metastatic melanoma, Clin. Cancer Res.,
21, 2278-2288, doi:10.1158/1078-0432.CCR-14-2085.
125. Ruella,M., Xu,J., Barrett, D.M., Fraietta, J. A., Reich,
T. J., Ambrose, D. E., Klichinsky, M., Shestova, O., Pa-
tel, P.R., Kulikovskaya,I., Nazimuddin,F., Bhoj, V.G.,
Orlando, E.J., Fry, T.J., Bitter,H., Maude, S.L., Levine,
B.L., Nobles, C.L., Bushman, F.D., Young, R.M., Schol-
ler,J., Gill, S.I., June, C.H., Grupp, S.A., Lacey, S.F., and
Melenhorst, J.J. (2018) Induction of resistance to chi-
meric antigen receptor T cell therapy by transduction
of a single leukemic B cell, Nat. Med., 24, 1499-1503,
doi:10.1038/s41591-018-0201-9.
126. Pesch,T., Bonati,L., Kelton,W., Parola,C., Ehling, R.A.,
Csepregi,L., Kitamura,D., and Reddy, S.T. (2019) Mo-
lecular design, optimization, and genomic integration
of chimeric Bcell receptors in murine B cells, Front.
Immunol., 10, 2630, doi:10.3389/fimmu.2019.02630.
127. Neelapu, S.S., Locke, F.L., Bartlett, N.L., Lekakis, L.J.,
Miklos, D. B., Jacobson, C. A., Braunschweig, I., Olu-
wole, O.O., Siddiqi,T., Lin,Y., Timmerman, J.M., Stiff,
P. J., Friedberg, J. W., Flinn, I. W., Goy, A., Hill, B. T.,
Smith, M.R., Deol,A., Farooq,U., McSweeney,P., Mu-
noz,J., Avivi,I., Castro, J.E., Westin, J.R., Chavez, J.C.,
Ghobadi,A., Komanduri, K.V., Levy,R., Jacobsen, E.D.,
Witzig, T. E., Reagan, P., Bot, A., Rossi, J., Navale, L.,
Jiang,Y., Aycock, J., Elias, M., Chang, D., Wiezorek,J.,
and Go, W.Y. (2017) Axicabtagene ciloleucel CAR T-cell
therapy in refractory large B-cell lymphoma, N.Engl. J.
Med., 377, 2531-2544, doi:10.1056/NEJMoa1707447.
128. Schuster, S. J., Svoboda, J., Chong, E. A., Nasta, S. D.,
Mato, A.R., Anak,O., Brogdon, J.L., Pruteanu-Malini-
ci,I., Bhoj, V., Landsburg,D., Wasik,M., Levine, B.L.,
Lacey, S. F., Melenhorst, J. J., Porter, D. L., and June,
C. H. (2017) Chimeric antigen receptor T cells in re-
fractory B-cell lymphomas, N.Engl. J.Med., 377, 2545-
2554, doi:10.1056/NEJMoa1708566.
129. Imura, Y., Ando, M., Kondo, T., Ito, M., and Yoshimu-
ra,A. (2020) CD19-targeted CAR regulatory Tcells sup-
CAR CELLS BEYOND CLASSICAL CAR T CELLS 783
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
press B cell pathology without GvHD, JCI Insight, 5,
e136185, doi:10.1172/jci.insight.136185.
130. Raje, N., Berdeja, J., Lin, Y., Siegel, D., Jagannath, S.,
Madduri, D., Liedtke, M., Rosenblatt, J., Maus, M. V.,
Turka,A., Lam, L.P., Morgan, R.A., Friedman,K., Mas-
saro, M., Wang, J., Russotti, G., Yang, Z., Campbell, T.,
Hege, K., Petrocca, F., Quigley, M. T., Munshi, N., and
Kochenderfer, J.N. (2019) Anti-BCMA CAR T-cell ther-
apy bb2121 in relapsed or refractory multiple my-
eloma, N. Engl. J. Med., 380, 1726-1737, doi: 10.1056/
NEJMoa1817226.
131. Berdeja, J. G., Madduri, D., Usmani, S. Z., Jakubowi-
ak,A., Agha, M., Cohen, A.D., Stewart, A. K., Hari,P.,
Htut,M., Lesokhin, A., Deol,A., Munshi, N.C., O’Don-
nell, E., Avigan, D., Singh, I., Zudaire, E., Yeh, T. M.,
Allred, A. J., Olyslager, Y., Banerjee, A., Jackson, C. C.,
Goldberg, J. D., Schecter, J. M., Deraed, W., Zhuang,
S. H., Infante, J., Geng, D., Wu, X., Carrasco-Alfonso,
M.J., Akram,M., Hossain, F., Rizvi,S., Fan,F., Lin, Y.,
Martin,T., and Jagannath,S. (2021) Ciltacabtagene au-
toleucel, a B-cell maturation antigen-directed chimeric
antigen receptor T-cell therapy in patients with re-
lapsed or refractory multiple myeloma (CARTITUDE-1):
a phase 1b/2 open-label study, Lancet, 398, 314-324,
doi:10.1016/S0140-6736(21)00933-8.
132. Wang, X., Jasinski, D.L., Medina, J.L., Spencer, D.M.,
Foster, A.E., and Bayle, J.H. (2020) Inducible MyD88/
CD40 synergizes with IL-15 to enhance antitumor
efficacy of CAR-NK cells, Blood Adv., 4, 1950-1964,
doi:10.1182/bloodadvances.2020001510.
133. Shin, M.H., Oh,E., Kim,Y., Nam, D.H., Jeon, S.Y., Yu,
J. H., and Minn, D. (2023) Recent advances in CAR-
based solid tumor immunotherapy, Cells, 12, 1606,
doi:10.3390/cells12121606.
Publishers Note. Pleiades Publishing remains
neutral with regard to jurisdictional claims in pub-
lished maps and institutional affiliations.