Download as pdf or txt
Download as pdf or txt
You are on page 1of 20

Please cite this article in press as: Wagner et al.

, CAR T Cell Therapy for Solid Tumors: Bright Future or Dark Reality?, Molecular Therapy (2020), https://
doi.org/10.1016/j.ymthe.2020.09.015

Review

CAR T Cell Therapy for Solid Tumors:


Bright Future or Dark Reality?
Jessica Wagner,1 Elizabeth Wickman,1,2 Christopher DeRenzo,1 and Stephen Gottschalk1
1Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; 2Graduate School of Biomedical
Sciences, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA

Chimeric antigen receptor (CAR) T cell therapy has garnered apeutic failure is most likely multifactorial and includes (1) CAR
significant excitement due to its success for hematological ma- design and CAR T cell generation, (2) a limited array of targetable an-
lignancies in clinical studies leading to the US Food and Drug tigens and heterogeneous antigen expression (aka “antigen
Administration (FDA) approval of three CD19-targeted CAR dilemma”), (3) limited T cell fitness, (4) inefficient homing to and
T cell products. In contrast, the clinical experience with CAR penetration of solid tumors, and (5) the hostile tumor microenviron-
T cell therapy for solid tumors and brain tumors has been ment (TME) (Figure 1).27–30 Herein, we review CAR design, the cur-
less encouraging, with only a few patients achieving complete rent clinical experience with autologous CAR T cells for solid tumors,
responses. Clinical and preclinical studies have identified mul- including brain tumors, and genetic approaches to overcome the
tiple “roadblocks,” including (1) a limited array of targetable aforementioned roadblocks. Since allogeneic CAR T cells are at pre-
antigens and heterogeneous antigen expression, (2) limited sent mainly explored for hematological malignancies in early phase
T cell fitness and survival before reaching tumor sites, (3) an clinical studies, we refer the interested reader to recently published re-
inability of T cells to efficiently traffic to tumor sites and pene- views.31,32 We also do not review CRS and neurotoxicity since several
trate physical barriers, and (4) an immunosuppressive tumor recent reviews have covered this topic.14,33
microenvironment. Herein, we review these challenges and
discuss strategies that investigators have taken to improve the Design of CARs
effector function of CAR T cells for the adoptive immuno- CARs have a modular design that consists of an antigen-binding
therapy of solid tumors. domain, most commonly a single-chain variable fragment (scFv)
derived from a monoclonal antibody (mAb), a hinge and transmem-
Adoptive cell therapies utilizing T cells expressing chimeric antigen brane domain, and an intracellular signaling domain.27,34–38 In addi-
receptors (CARs) have been propelled to the forefront of experi- tion to scFvs, natural ligands of receptors such as cytokines or pep-
mental cell therapies due to their clinical success for hematological tides that bind cell surface molecules are also being explored as
malignancies targeting a range of antigens, including CD19, CD22, antigen-binding domains.22,39,40 While most CARs recognize epi-
CD30, kappa, and B cell maturation antigen (BCMA).1–10 The land- topes of cell surface proteins in a major histocompatibility complex
scape of CAR T cell therapies for hematological malignancies, (MHC)-independent manner, scFvs have also been incorporated
including successes and challenges, has been the subject of multiple into CARs that recognize a peptide in the context of a human leuko-
recent reviews.11–13 We therefore do not discuss these in detail, cyte antigen (HLA) molecule.41–43 While this approach allows CAR
except for highlighting the “lessons learned” as they relate to target- T cells to recognize intracellular molecules, it renders target antigen
ing solid tumors, including brain tumors. First, CAR T cells can recognition dependent on a particular HLA type, restricting its appli-
eradicate chemorefractory cancer cells regardless of their underlying cation to a subset of patients. In addition, CAR T cells become sensi-
oncogenic driver mutations; second, lymphodepleting chemo- tive to decreased HLA expression and defects in the antigen process-
therapy is at present a sine qua non to enable expansion and persis- ing pathway, with both pathways used by tumor cells to actively evade
tence of infused CAR T cells; third, inclusion of at least one co-stim- immune responses.44
ulatory signaling domain in the CAR is critical for its success;
fourth: antigen loss variants have emerged as one mechanism of First-generation CARs contained only a T cell activation domain,
therapeutic failure, even for antigens that are highly and homoge- and the most commonly utilized domain is derived from
neously expressed such as CD19; and fifth, CAR T cells can induce CD3z.37,38 Since optimal T cell activation also relies on co-stimula-
significant side effects, including cytokine release syndrome (CRS) tion, signaling domains from co-stimulatory molecules were
and neurotoxicity.11–15

https://doi.org/10.1016/j.ymthe.2020.09.015.
In contrast to CAR T cell therapy for hematological malignancies,
Correspondence: Stephen Gottschalk, Department of Bone Marrow Trans-
CAR T cell therapies for solid tumors, including brain tumors, have
plantation and Cellular Therapy, St. Jude Children’s Research Hospital, 262 Danny
shown limited antitumor activity in early phase clinical testing despite Thomas Place, MS321, Memphis, TN 38105, USA.
targeting a variety of target antigens and tumor types.16–26 This ther- E-mail: stephen.gottschalk@stjude.org

Molecular Therapy Vol. 28 No 11 November 2020 ª 2020 The American Society of Gene and Cell Therapy. 1
Please cite this article in press as: Wagner et al., CAR T Cell Therapy for Solid Tumors: Bright Future or Dark Reality?, Molecular Therapy (2020), https://
doi.org/10.1016/j.ymthe.2020.09.015

www.moleculartherapy.org

Review

Optimizing CAR function remains a challenge since there is an intri-


cate interplay between the functional (antigen recognition and
signaling domains) and nonfunctional components (hinge and trans-
membrane domain) of CARs.57,58 In addition, the location of the
epitope within the targeted cell surface molecule determines CAR
T cell activity.59 For example, epitopes that are proximal to the plasma
membrane induce greater CAR T cell activation than do distal epi-
topes.57,59,60 Studies have also highlighted that too much CAR
T cell activation is detrimental to T cell function. For example,
CARs with two of three mutated immunoreceptor tyrosine-based
activation motifs (ITAMs) in the CD3z chain endow CAR T cells
with improved effector function.61 In addition, excessive CD28 co-
stimulation has detrimental effects,62 and mutations in the YMXM
motif of CD28 that reduce its signaling activity improve T cell func-
tion.63 In addition to CAR signaling after activation, baseline (aka
tonic) signaling determines CAR T cell activity.64,65 Recently, investi-
gators have also focused on studying the immunological synapse that
is formed between CAR T cells and target cells, and they have corre-
lated synapse formation with CAR T cell effectiveness.66,67

Lastly, several studies have highlighted limitations of CARs with stan-


dard co-stimulatory molecules, which most likely cannot be over-
come by further refining its structure.27,37,38 Foremost, CARs only
provide signal 1 (activation) and signal 2 (co-stimulation) of canon-
ical T cell activation and not signal 3 (inflammatory cytokines). While
CAR-activated T cells initially produce cytokines, they do not secrete
sufficient amounts of cytokines after recursive exposure to antigen-
Figure 1. CAR T Cell Immunotherapy “Roadblocks” for Solid Tumors positive target cells, leading to rapid erosion of their effector func-
Top left: antigen dilemma. Heterogeneous antigen expression results in immune
tion.68–70 This occurs even in the absence of an immunosuppressive
escape variants, and antigen expression in normal tissues can lead to on target/off
TME, highlighting that additional modification of CAR T cells or
cancer toxicity. Top right: CAR T cell fitness. Expansion, contractions, and persis-
tence of CAR T cells is often limited after infusion. Bottom left: homing/penetration. combinatorial therapies are needed to sustain and/or enhance their
CAR T cells have a limited ability to traffic to and penetrate solid tumors. Bottom effector function. These are discussed in detail in the section
right: microenvironment. The solid tumor microenvironment is hostile, limiting CAR Improving CAR T Cell Fitness.
T cell effector function.
CAR T Cells for Solid Tumors and Brain Tumors—Clinical
included into CARs. Initial studies focused on domains derived Experience
from the canonical co-stimulatory molecules CD28 and 41BB.37,38 CAR T cells have been evaluated in early phase clinical studies for
Since then, signaling domains from a broad range of molecules more than a decade, targeting a broad range of tumor antigens,
have been explored, including OX40, CD27, and ICOS.37,45–49 including B7-H3, CAIX, CEACAM5, CD133, CD171, epidermal
CD28 and 41BB co-stimulation in the context of CAR T cells has growth factor receptor (EGFR), EGFRvIII, FRa, GD2, GPC3,
been extensively studied, including detailed phosphoproteomic HER2, interleukin (IL)-13Ra2, mesothelin, MUC1, prostate-specific
and single-cell RNA sequencing (RNA-seq) analyses.50–52 They acti- membrane antigen (PSMA), ROR1, and vascular endothelial growth
vate different pathways within T cells, with CD28 signaling promot- factor (VEGF)-R2.19,20,22–26,71–82 Table 1 lists selected published
ing glycolytic metabolism and an effector memory phenotype, in studies, and Table 2 lists ongoing clinical studies. Early studies
contrast to 41BB signaling, which induces oxidative metabolism focused on first-generation CARs specific for CAIX, CD171, FRa,
and a central memory phenotype.53 Depending on the number of GD2, or IL-13Ra2.16,20,22,83 Two studies utilized polyclonal, activated
co-stimulatory molecules included in the CAR signaling domain, CAR T cells (CAIX, FRa);20,84 two studies focused on CAR T cell
CARs are either designated second (one domain) or third (two clones (CD171, IL-13Ra2);75,83,85 and one study compared GD2-
domains) generation. In preclinical studies, the benefit of including CAR T cells with GD2-CAR/Epstein-Barr virus (EBV)-specific
two co-stimulatory molecules in CARs is model-dependent.54,55 T cells.81,82 First-generation CAR T cells did not expand after infusion
Results of one clinical study, comparing CD28-CAR versus and had limited antitumor activity, except for the study with GD2-
CD28.41BB-CAR T cells targeting CD19, suggest that third-genera- CAR/EBV-specific T cells, in which 3 of 11 patients had a complete
tion CARs endow T cells with a greater ability to expand after infu- response (CR). Despite having no antitumor activity, CAIX-CAR
sion in humans.56 T cells induced “on target/off cancer” toxicity in the form of

2 Molecular Therapy Vol. 28 No 11 November 2020


Please cite this article in press as: Wagner et al., CAR T Cell Therapy for Solid Tumors: Bright Future or Dark Reality?, Molecular Therapy (2020), https://
doi.org/10.1016/j.ymthe.2020.09.015

www.moleculartherapy.org

Review

Table 1. Selected Published CAR T Cell Clinical Studies for Solid Tumors and Brain Tumors

Antigen Diagnosis Signaling Domain Vector T Cell Product IL-2 after T Cells Comment References
Intravenous Administration, No Lymphodepleting Chemotherapy
FRa OVCA z RV ATCsa Y 14/14 NR 21

12/12 NR; on target/off cancer 19,20


CAIX RCC z RV ATCs N
toxicity—bile ducts
83
CD171 NB z Plasmid T cell clone N 1/6 PR
17
EGFRvIII HGG 4-1BB LV ATCs N 1/10 SD
81,82
GD2 NB z RV ATCs, VSTs N 3/11 CR
23
HER2 sarcoma CD28z RV ATCs N 4/17 SD
26
HER2 HGG CD28z RV VSTs N 3/17 SD
80
Mesothelin PCA 41BBz mRNA ATCs N 2/6 SD
Intravenous Administration after Lymphodepleting Chemotherapy
24
CD133 HCC, PCA, CRC 41BBz LV ATCs N 3/23 PR, 14/23 SD
CRC, PDCA, stomach, 7/14 SD; on target/off cancer 25
CEACAM5 z RV ATCs Y
esophagus toxicity—lung
86
EGFR PCA 41BBz LV ATCs N 4/16 PR, 8/16 SD, 2/16 NE
b 16
EGFRvIII HGG CD28.41BBz RV ATCs Y 17/18 NR, 1/18 NE due to TRM
GD2 NB CD28.OX40z RVc ATCs N 5/11 SD 87

88
GPC3 HCC CD28z LV ATCs N 2/13 PR, 1/13 SD, 4/13 NE
b
1/1 TRM ; on target/off cancer 18
HER2 CRC CD28.41BBz LV ATCs Y
toxicity—lung
89,90
HER2 sarcoma CD28z RV ATCs N 1/10 CR; 3/10 SD
Mesothelin MPM, PCA, OVCA 41BBz LV ATCs N 11/15d SD 91

71
PSMA prostate z RV ATCs Y 2/5 PR
4/6 mixed response; 72
ROR1 TNBC, NSCLC 41BBz LV ATCs N
1/6 SD; 1/6 not reported
VEGF-R2 metastatic CA – RV ATCs Y 1/23 PR e

Intravenous Administration after Allotransplant


f
GD2 NB z RV VSTs N 3/3 PD
Locoregional Administration
73
B7-H3 meningioma 41BBz LV ATCs N 1/1 evidence of ALV
74
IL-13Ra2 HGG z plasmid T cell clone N 1/3 tumor necrosis
a 75
IL-13Ra2 HGG z plasmid T cell clone N imaging study; outcome not reported
22
IL-13Ra2 HGG 41BBz LV ATCs N 1/1 CR
Mesothelin MPM, lung CA, breast CA CD28z RVc ATCs N 2/21g CR, 5/21 PR, 4/21 SD 76

h 92
MUC1 SVA 41BBz or CD28z LV ATCs N 1/1 tumor necrosis
ALV, antigen loss variant; ATC, activated T cell; CA, cancer; CR, complete response; CRC, colorectal cancer; HCC, hepatocellular carcinoma; HGG, high-grade glioma; LV, lentivirus;
MPM, malignant pleural mesothelioma; N, no; NB, neuroblastoma; NE, not evaluable; NR, no response; OVCA, ovarian cancer; PCA, pancreatic adenocarcinoma; PR, partial response;
RCC, renal cell carcinoma; RV, retrovirus; SD, stable disease; SVA, seminal vesicle adenocarcinoma; VST, virus-specific T cell; Y, yes.
a
Several patients received allogeneic cell products.
b
Pulmonary complications.
c
CAR vector also encoded inducible caspase-9.
d
6 of 15 patients received lymphodepleting chemotherapy.
e
ClinicalTrials.gov: NCT01218867.
f
ClinicalTrials.gov: NCT01460901.
g
18 of 21 patients received lymphodepleting chemotherapy and 13 of 21 patients received pembrolizumab.
h
41BBz CAR vector also encoded IL-12.

Molecular Therapy Vol. 28 No 11 November 2020 3


4

Review
www.moleculartherapy.org

doi.org/10.1016/j.ymthe.2020.09.015
Please cite this article in press as: Wagner et al., CAR T Cell Therapy for Solid Tumors: Bright Future or Dark Reality?, Molecular Therapy (2020), https://
Molecular Therapy Vol. 28 No 11 November 2020

Table 2. Selected Actively Recruiting CAR T Cell Clinical Studies for Solid Tumors and Brain Tumors

Antigen Diagnosis Signaling Domain Additional Engineering Vector T Cell Product Other Therapy NCT No.: ClinicalTrials.gov
Intravenous Administration, No Lymphodepleting Chemotherapy Listed on ClinicalTrials.gov Site
HCC – TGF-b-CAR, IL-7-CCL19 – ATCs N NCT03198546
GPC3
HCC – N – ATCs N NCT04121273
PSMA solid tumors – N – ATCs N NCT04429451
Intravenous Administration after Lymphodepleting Chemotherapy
B7-H3 solid tumors – iC9 LV ATCs N NCT04432649
CD171 NB 41BBz or CD28.41BBz N LV ATCs N NCT02311621
Claudin 18.2 solid tumors – N – ATCs N NCT03874897
EGFR806 solid tumors 41BBz CD19-CAR LV ATCs N NCT03618381
IL-13Ra2 Melanoma 41BBz N LV ATCs IL-2 NCT04119024
NB – iC9, IL-15 RV ATCs N NCT03721068
GD2 solid tumors – iC9 – ATCs N NCT03373097
solid tumors – C7R RV ATCs N NCT03635632
DIPG, HGG – C7R RV ATCs N NCT04099797
NB – IL-15 RV iNKT N NCT03294954
NB – N – ATCs N NCT02761915
DIPG 41BBz iC9 RV ATCs N NCT04196413
solid tumors 41BBz N RV ATCs N NCT02932956
GPC3 HCC 41BBz N RV ATCs N NCT02905188
HCC – N – ATCs N NCT03884751
solid tumors – PD-1 KO – ATCs N NCT03747965
Mesothelin
solid tumors – N LV ATCs N NCT03054298
esophageal CA – PD-1 KO – ATCs IL-2 NCT03706326
MUC-1 NSCLC – PD-1 KO – ATCs IL-2 NCT03525782
breast Cancer 41BBz N LV ATCs N NCT04020575
prostate CA 41BBz N LV ATCs N NCT03873805
PSCA
solid tumors z iMC RV ATCs N NCT02744287
solid tumors – iC9 gene editing ATCs N NCT04249947
PSMA prostate CA – DNR TGFb LV ATCs N NCT03089203
prostate CA – DNR TGF-b LV ATCs N NCT04227275
Intravenous and Locoregional Administration After Lymphodepleting Chemotherapy
MUC16ecto solid tumors 41BBz IL-12 LV ATCs N NCT02498912
(Continued on next page)
Please cite this article in press as: Wagner et al., CAR T Cell Therapy for Solid Tumors: Bright Future or Dark Reality?, Molecular Therapy (2020), https://
doi.org/10.1016/j.ymthe.2020.09.015

www.moleculartherapy.org

Review

cholangitis.19,20 In hindsight, limited antitumor activity was not sur-

caspase-9; iMC, inducible Myd88.CD40; ipi, ipilimumab; KO, knockout; LV, lentivirus; N, no; NB, neuroblastoma; nivo, nivolumab; OVCA, ovarian cancer; pembro, pembrolizumab; RV, retrovirus; SCCHN, squamous cell
ATC, activated T cell; CA, cancer; CCR, chimeric cytokine receptor; C7R, constitutive active IL-7 receptor a; DNR, dominant negative receptor; HCC, hepatocellular carcinoma; HGG, high-grade glioma; iC9, inducible
prising since CARs without co-stimulatory endodomains were
NCT No.: ClinicalTrials.gov

grafted onto T cells, and patients did not receive lymphodepleting


chemotherapy prior to the adoptive transfer of CAR T cells—two pre-
requisites of successful CAR T cell therapies for hematological malig-
NCT04185038
NCT04385173
NCT04077866
NCT03618381
NCT01818323
NCT03696030
NCT02442297
NCT03500991
NCT04003649
NCT02208362

NCT03585764
NCT02414269
nancies.1–10

In subsequent studies, CARs were evaluated with co-stimulatory


endodomains (CD28, 41BB, CD28/41BB, CD28/OX40) targeting
B7-H3, CEACAM5, CD133, CD171, EGFR, EGFRvIII, FRa, GD2,
GPC3, HER2, IL-13Ra2, mesothelin, MUC1, PSMA, ROR1, and
Other Therapy

VEGF-R2.72,86–89,91,92 Similar to first-generation CAR T cells, adop-


Pembro tive transfer of second- or third-generation CAR T cells or CAR vi-
nivo/ipi
TMZ
TMZ

rus-specific T cells did not result in significant T cell expansion in


N

N
N
N
N
N

two studies, and variable expansion in one study.23,26,87 Thus, inclu-


sion of a co-stimulatory domain into the CAR by itself is not sufficient
to consistently induce systemic T cell expansion. In contrast, lympho-
T Cell Product

depleting chemotherapy prior to CAR T cell infusion resulted in a sig-


nificant expansion of CAR T cells irrespective of the utilized co-stim-
ATCs
ATCs
ATCs
ATCs
ATCs
ATCs
ATCs
ATCs
ATCs
ATCs

ATCs
ATCs

ulatory domain and targeted antigen.9,93,94 Despite significant in vivo


expansion, antitumor activity remained limited (Table 1). However,
single case reports of patients with recurrent refractory solid tumors
or brain tumors, who achieved CRs after CAR T cell therapy, have
been reported (Table 1).22,90 Clinical studies have also highlighted
Vector

safety concerns. Two patients died of acute respiratory failure after


RV
RV

RV

RV
LV

LV

LV

LV
LV
LV

LV

having received lymphodepleting chemotherapy, a high dose (1 to


6  1010) of third-generation CAR T cells, and IL-2.16,18 CAR
T cells either recognized an antigen that is expressed (HER2) or not
expressed (EGFRvIII) on lung endothelial cells, suggesting that on
Additional Engineering

target/off cancer toxicity as well as unspecific CAR T cell activation


can result in fatal respiratory failure. Adoptive transfer of high cell
IL-4/IL-2 CCR

doses (1  109 to 5  1010) of CEACAM5-CAR T cells in conjunction


with IL-2 also resulted in pulmonary toxicities (mild pulmonary
edema), which was attributed to CEACAM5 expression on lung
iC9
N
N
N
N

N
N
N
N
N

epithelium.25

Tumor tissue after CAR T cell infusion has only been studied sys-
Locoregional Administration after Lymphodepleting Chemotherapy
Locoregional Administration, No Lymphodepleting Chemotherapy

tematically in one study in which patients with high-grade glioma


Signaling Domain

(HGG) received EGFRvIII-CAR T cells.17 Collectively, the analysis


showed that CAR T cells were able to migrate to and proliferate
at tumor sites. CAR T cells killed tumor cells as evidenced by
CD28z

CD28z

CD28z
41BBz

41BBz

41BBz
41BBz
41BBz

41BBz

the selection of target antigen-negative tumors, and, strikingly,




induced a reactive immunosuppressive environment as judged


by expression of IDO1 and PD-L1, and an influx of regulatory
carcinoma of the head and neck; Y, yes.

T cells (Tregs).
CNS metastases

pleural disease
CNS tumors
CNS tumors
CNS tumors
CNS tumors

CNS tumors
CNS tumors
Diagnosis

SCCHN

Thus, CAR T cells at present rarely induce CRs in patients with solid
OVCA
HGG
HGG

tumors or brain tumors. In addition, several studies have highlighted


their potential to induce on target/off cancer toxicity. In the following
Table 2. Continued

sections we review approaches that investigators have taken to


improve their efficacy and safety, with special focus on (1) clinical
EGFR family

Mesothelin

grade CAR T cell production and T cell subsets for CAR T cell ther-
IL-13Ra2
EGFR806
Antigen

B7-H3

HER2

apy, (2) expanding the array of targetable antigens and overcoming


FRa

heterogeneous antigen expression, (3) CAR T cell fitness, (4) CAR

Molecular Therapy Vol. 28 No 11 November 2020 5


Please cite this article in press as: Wagner et al., CAR T Cell Therapy for Solid Tumors: Bright Future or Dark Reality?, Molecular Therapy (2020), https://
doi.org/10.1016/j.ymthe.2020.09.015

www.moleculartherapy.org

Review

Figure 2. Strategies to Overcome CAR T Cell Immunotherapy Roadblocks for Solid Tumors
Summary of strategies that are discussed in detail in this review.

T cell trafficking and tumor penetration, and (5) counteracting the of CAR T cells increases the frequency of CD8+CD45RA+CCR7+
immunosuppressive TME (Figure 2). stem cell-like CAR T cells with superior antitumor functionality.108
Based on these findings, many clinical-grade CAR T cell production
Improving Clinical-Grade CAR T Cell Production protocols are currently using IL-7 and IL-15. In addition, IL-21 has
Preclinical studies have highlighted that the effector function of been shown to prevent differentiation of genetically modified
T cells progressively decreases as T cells differentiate from naive T cells paired with improved effector function in preclinical
(TN) to stem cell memory (TSCM), central memory (TCM), effector studies.109,110 Inhibiting or activating pathways in T cells that pro-
memory (TEM), and terminally differentiated T cells that express mote or prevent T cell differentiation are alternative strategies that
the CD45RA effector (TEMRA).95 Transcription factors govern T cell have been explored. These include activating Wnt/b-catenin signaling
differentiation. For example, TCF7 promotes self-renewal and mem- pathways or inhibiting mTOR or AKT signaling.111,112
ory formation, while TOX, BLIMP1, and TBET promote terminal
T cell differentiation.96–102 More recent studies have also highlighted Notwithstanding these advances, there is at present a lack of generally
the critical role of epigenetic programs enforced by the de novo DNA accepted biomarkers that predict efficacy of the infused CAR T cell
methyltransferase 3A (DNMT3A) in determining T cell fate.103–105 In product. Several studies with CD19-CAR T cells are starting to
addition, a recent case report highlighted that disruption of TET2 in shed light on this issue. In one study in patients with chronic lympho-
CD19-CAR T cells, an enzyme that regulates DNA demethylation, re- cytic leukemia (CLL), the presence of CD27+PD-1CD8+ CD19-CAR
sulted in clonal T cell expansion.106 T cells expressing high levels of the IL-6 receptor in the T cell product
correlated with antitumor activity,113 and in another study in patients
Despite these insights in T cell biology, most initial clinical studies uti- with acute lymphocytic leukemia (ALL), the presence of tumor
lized polyclonal, CD3/CD28-activated T cells that are expanded in the necrosis factor (TNF)-a+CD8+ CD19-CAR T cells, respectively.114
g-cytokine IL-2. These expansion protocols in general result in signif- One recent study, using RNA-seq analysis, demonstrated that
icant T cell differentiation paired with inferior effector function in CD19-CAR T cell clones that expand after infusion are derived
preclinical models. Investigators have explored alternative g-cyto- from clusters of CAR T cells in the product that express higher levels
kines to preserve CAR T cell function.107 For example, replacing of genes associated with T cell proliferation and cytolytic activity.115
IL-2 with a combination of IL-7 and IL-15 for the ex vivo expansion While this study found no correlation with vector integration sites,

6 Molecular Therapy Vol. 28 No 11 November 2020


Please cite this article in press as: Wagner et al., CAR T Cell Therapy for Solid Tumors: Bright Future or Dark Reality?, Molecular Therapy (2020), https://
doi.org/10.1016/j.ymthe.2020.09.015

www.moleculartherapy.org

Review

another recent report demonstrated that vector integration site distri- cacy of GD2-CAR/varicella zoster virus (VZV)-specific T cells (Clin-
bution in CD19-CAR T cells is linked to clinical outcome.116 Clearly, icalTrials.gov: NCT01953900). Advantages of this approach are not
additional studies are needed to identify biomarkers not only for the only that CAR/virus-specific T cells might have improved effector
CAR T cell product, but also for the targeted tumor. function in comparison to their unselected CAR T cell counterparts,
but these CAR/virus-specific T cells can be boosted after infusion
Besides improving culture conditions for CAR T cell generation, in- through their endogenous, virus-specific ab TCR either by vaccina-
vestigators have also focused on the genetic modification step of tion (e.g., VZV vaccine) or reactivation of the latent virus (e.g.,
T cells during CAR T cell production. Most clinical studies so far EBV). Lastly CAR-expressing gd T cells133 and invariant natural killer
have utilized CAR T cells that were generated with retroviral or len- (iNK)T cells have been successfully explored in preclinical studies,134
tiviral vectors with an excellent safety record.117–119 However, clin- and a clinical study with iNK T cells expressing GD2-CARs and IL-15
ical-grade production and release testing of recombinant viral vectors is in progress (ClinicalTrials.gov: NCT03294954).
is expensive, and patients require additional monitoring. Therefore,
alternative approaches have been developed, including the use of Pig- CAR T Cell Therapy Targets—The Antigen Dilemma
gyBac or Sleeping Beauty transposon systems.120,121 More recently, An “ideal” CAR target should be highly and homogeneously ex-
combining gene-editing technologies (e.g., CRISPR-Cas9, TALEN, pressed throughout the tumor, across multiple patients, and have
ZFN, meganucleases) with homology-directed DNA repair (HDR) minimal to no expression in vital normal tissues. Unfortunately, these
has enabled the integration of transgenes into specific loci.122–124 characteristics do not apply to most CAR targets that are currently be-
This approach allows for control of transgene expression with endog- ing explored for solid tumors and brain tumors (Table 2). In general,
enous promoters. For example, CD19-CARs have been knocked in to current targets are differentially expressed antigens, which may be
the T cell receptor (TCR) a constant (TRAC) locus using CRISPR- present at low levels in normal tissues. The only exceptions are EGFR-
Cas9 gene editing to drive CAR expression from the endogenous vIII, a cancer-specific splice variant expressed in HGG, and EGFR806,
TCR a chain promoter. This resulted in consistent CAR expression, a conformational EGFR epitope that is also present in HGG.135,136
reduced baseline signaling, and improved antitumor activity While neoantigens are a potential solution to this “antigen dilemma,”
compared to CD19-CAR T cells generated by viral transduction.125 most of these are patient specific, raising concerns about feasibility. In
The clinical experience with CAR T cells generated with non-viral addition, most neoantigens described so far are derived from intracel-
methods is so far limited,78 and most ongoing studies are focused lular proteins, which are difficult to target with standard CAR
on targeting hematological malignancies. T cells.137 Investigators have therefore embarked on discovering novel
CAR targets using genomic and proteomic approaches.138–140 In
T Cell Subsets for CAR T Cell Therapy addition, there is an intense focus on engineering CARs and CAR
Investigators have explored CAR T cell products with a defined T cells to increase their specificity for antigens expressed on tumor
CD8+/CD4+ T cell subset ratio and products that were derived cells. These include (1) modulating the affinity of the scFv, (2) target-
from TSCM, TCM, or T helper (Th17) cells.48,126–129 These studies ing multiple antigens, and (3) restricting CAR activity to tumor sites.
have highlighted that T cell subset-derived CAR T cell populations In addition, investigators have developed safety switches that can be
have improved effector function in comparison to polyclonal, CD3/ activated in the event of adverse events secondary to CAR T cells,
CD28-activated CAR T cells. Importantly, the co-stimulatory domain including on target/off cancer toxicity.
of the CAR might have to be tailored for a specific T cell subset with
one study demonstrating superior antitumor activity when CD4+ Modulating scFv Affinity
T cells, expressing CARs with an ICOS co-stimulatory endodomain, This approach is only applicable to antigens that are expressed at very
were combined with CD8+ T cells, expressing CARs with a 41BB high levels such as gene-amplified HER2 in HER2-positive breast
co-stimulatory endodomain.130 cancer. For example, investigators have shown that reducing the
scFv affinity of HER2-CARs or EGFR-CARs decreases the risk of
However, the translation of these approaches into the clinic might be on target/off cancer toxicity.141 In addition, in murine models, the
limited due to the frequency of some of these T cell subset popula- incidence of neurotoxicity of GD2-CAR T cells varied according to
tions; for example, TSCM cells are very low in the peripheral circula- the affinity of the expressed CAR.142 In addition to modulating
tion.131 In addition, sequential cycles of chemotherapy have depleted scFv affinity, recent studies have also highlighted that the choice
T cell subsets, including TN cells, in heavily pretreated cancer patients, of hinge, transmembrane, and/or costimulatory domains of
who are the current CAR T cell study population.132 CARs can influence antigen density requirements for CAR T cell
activation.58,143,144
Selecting virus-specific T cells for CAR T cell generation is an alterna-
tive approach to selecting TCM and TEM cells with unknown speci- Targeting Multiple Antigens
ficity. Examples include the aforementioned clinical study with Heterogeneity is a hallmark of cancer and a major roadblock for all
GD2-CAR/EBV-specific T cells in which investigators demonstrated targeted cancer therapies, including immunotherapy.145 Heteroge-
improved persistence in comparison to GD2-CAR T cells.81 In addi- neous antigen expression is one consequence of tumor heterogeneity
tion, a clinical study is ongoing, which evaluates the safety and effi- leading to the outgrowth of tumor cell subpopulations when a single

Molecular Therapy Vol. 28 No 11 November 2020 7


Please cite this article in press as: Wagner et al., CAR T Cell Therapy for Solid Tumors: Bright Future or Dark Reality?, Molecular Therapy (2020), https://
doi.org/10.1016/j.ymthe.2020.09.015

www.moleculartherapy.org

Review

antigen is targeted. Investigators have therefore focused on targeting full CAR T cell activation has been restricted to (1) PSMA- and
cancer stem cells (CSCs) or tumor-initiating cells (TICs),146–148 or PSCA-positive or (2) MUC1- and HER2-positive tumor cells in pre-
multiple antigens to prevent immune escape. In addition, multi-spe- clinical models.173,174 Expressing inhibitory CARs that recognizes an
cific CAR T cells have the potential to reduce the risk of on target off/ antigen expressed on normal tissues in conjunction with a tumor-spe-
cancer toxicity if they target an array of antigens present in a partic- cific CAR is another approach that has been evaluated using CD19 as
ular tumor and not in normal tissues. Several approaches are actively a tumor-specific antigen and PSMA as a model antigen expressed on
being pursued, including expressing CARs with two scFvs. Investiga- non-targeted cancer cells.175 Lastly, inactive CARs have been de-
tors have either assembled two scFvs in a linear (aka tandem CAR) signed (e.g., EGFR)176 that require proteolytic processing into an
fashion or by alternating heavy and light chains of the two scFvs active form by matrix metalloproteinases (MMPs) within the TME.
(aka loop CAR), similar to the dual-affinity retargeting (DART) bis-
pecific antibody platform.149–151 Investigators have also expressed Safety Switches
two or three different CARs in T cells.152 The clinical experience Neither of the aforementioned strategies may end up being suffi-
with T cells expressing bispecific CARs or two CARs is currently cient to prevent on target/off cancer toxicities. Investigators have
limited to CD19/CD22- and CD19/CD20-targeted approaches for therefore developed inducible safety switches to ablate CAR
leukemia and/or lymphoma. An alternate approach is to design T cells when the need arises. These safety switches can be concep-
CARs in which the antigen-binding domain is derived from a ligand tually divided into four approaches. The first approach relies on
that binds multiple antigens, and the most relevant example for solid activating a prodrug that is toxic to T cells. The best studied
tumors is the T1E peptide that binds to the EGFR family of recep- example is the antiviral ganciclovir, which readily kills T cells that
tors.40 So-called “universal CARs” have also been developed to target are genetically modified with the herpes simplex virus thymidine ki-
multiple antigens that contain an ectodomain that can be paired with nase (HSV-tk).177 The second approach takes advantage of dimer-
multiple antigen recognition domains. Examples include (1) avidin- izer drugs to activate a molecule that is transgenically expressed
CARs/biotin-labeled scFvs, (2) CD16-CAR/mAbs, (3) anti-fluores- in CAR T cells. Several dimerizer systems have been developed;178
cein isothiocyanate (FITC)-CARs/FITC-labeled scFvs, (4) coiled- of these, the inducible caspase-9 (iC9) system has been evaluated
coil CARs (SUPRA CARs), (5) anti-PNE-CARs/PNE-scFvs, and (6) in humans with encouraging results.179,180 Other promising strate-
NKG2D-CARs/ULBP2-mAbs.153–159 Lastly, bispecific T cell engagers gies to increase the safety profile of CAR T cells include splitting
(BiTEs) have been expressed in T cells,160,161 and more recently CARs into two domains, which need to be linked with a small mole-
adapted to CAR T cells, opening up the opportunity to target multiple cule to be active,181,182 or taking advantage of the small molecule-as-
antigens and redirecting bystander T cells to tumor cells.162 sisted shut-off (SMAsh) technology.183 The third approach relies on
expressing a molecule on the cell surface of T cells that can be tar-
In addition to designing CAR T cells to target multiple antigens, stra- geted with a clinically approved mAb (e.g., CD20 and truncated
tegies are being pursued to engineer T cells to activate bystander EGFR).184,185 At present the clinical experience with these ap-
T cells to recognize tumor cells (aka induce antigen/epitope proaches is limited. Finally, the last approach relies on exploiting
spreading). These include the transgenic expression of cytokines intrinsic vulnerabilities of T cells with a prime example being the
(e.g., IL-18 or IL-36g) or CD40L.163–165 Likewise, one recent study tyrosine kinase inhibitor (TKI) dasatinib, which inhibits TCR and
has demonstrated that engineering T cells to secrete the FLT3 ligand CAR signaling through inhibition of Lck.186
promotes epitope spreading.166
Improving CAR T Cell Fitness
Restricting CAR Activity to Tumor Sites As mentioned in the Design of CARs section, even outside the
CAR activity can be restricted to tumor sites by several measures. immunosuppressive TME, CAR T cells have a limited ability to pro-
First, CAR expression can be induced once T cells have migrated to duce cytokines, expand, and kill when recursively exposed to tumor
tumor sites with small molecules, such as a doxycycline-inducible cells. T cell exhaustion is a widely used term to describe this T cell
expression system.167 Second, CAR T cells can take clues from the response to chronic antigen exposure, and there is significant debate
TME to express a CAR. These include hypoxia-inducible expression in the field how to best define T cell exhaustion.187 While tradition-
systems,168 as well as synthetic signaling circuits that take advantage ally cell surface markers, such as PD-1, LAG-3, TIM3, and TIGIT,
of Boolean logic.169–171 One of the best examples of Boolean logic- have been used to define T cell exhaustion, more recent studies
gated CARs is the synthetic notch (synNotch) system,169–171 in which have highlighted that expression of transcription factors such as
an antigen-specific synNotch receptor induces the expression of a sec- TOX or epigenetic programs might be more suitable to reliably
ond CAR upon activation. For example, EpCAM- or B7H3-specific define T cell exhaustion.100–103,105 To improve the fitness of CAR
synNotch receptors have been used to restrict ROR1-CAR expression T cells, investigators have focused on (1) providing additional sig-
to tumor sites, reducing on target/off cancer toxicity in preclinical nals to promote T cell activation/co-stimulation, (2) providing
models.172 Other strategies include engineering T cells to express signal 3 by transgenic expression of cytokines or constitutively
two CARs with different specificity in which one CAR provides signal active cytokine receptors, (3) silencing or deleting molecules that
1 and the other signal 2 to limit full CAR T cell activation to tumor restrict T cell activation, and (4) transgenic expression of transcrip-
sites at which both antigens are present.173,174 With this approach tion factors.

8 Molecular Therapy Vol. 28 No 11 November 2020


Please cite this article in press as: Wagner et al., CAR T Cell Therapy for Solid Tumors: Bright Future or Dark Reality?, Molecular Therapy (2020), https://
doi.org/10.1016/j.ymthe.2020.09.015

www.moleculartherapy.org

Review

Provision of Additional Signals to Promote T Cell Activation/Co- control of potential side effects. Alternative approaches to activate
stimulation the JAK/STAT pathways are also being pursued, including expression
Standard CARs only provide co-stimulation, and not directly signal 3, of constitutively active cytokine receptors (e.g., constitutively active
a necessary signal for T cell expansion. To overcome this limitation, IL-7 receptor a [C7R]).68
investigators have incorporated the truncated cytoplasmic domain
of IL-2Rb and a STAT3-binding YXXQ motif into CD28.z-CARs tar- Cytokines That Signal through MyD88. IL-1b and IL-18 signal
geting CD19.188 Other groups have shown that T cells expressing through MyD88, and at present only transgenic expression of IL-18
CD28.z-CARs that incorporate the Toll/IL-1 receptor domain of has been explored by several groups of investigators. Consistently,
Toll-like receptor (TLR)2 improve CAR T cell effector function.189 IL-18 improved the effector function of CAR T cells not only in xeno-
In both approaches, signals are provided simultaneously, which is graft models but also in syngeneic murine models.163,203,204 However,
in contrast to physiological T cell activation in which signals are pro- in some models, administration of CAR/IL-18 T cells was associated
vided in a temporospatial fashion. T cells express 41BB upon activa- with significant toxicities, which could be mitigated in one study by
tion, and studies have demonstrated that expression of the 41BB expressing IL-18 under the transcriptional control of the NFAT
ligand (41BBL) on the cell surface of CD28.z-CAR T cells endows promoter.204
CAR T cells with superior effector function in comparison to incor-
porating the 41BB signaling domain directly into the CAR.55,190 Silencing or Deletion of Molecules That Restrict T Cell Activation
Other ligands of the TNF superfamily have also been expressed on T cells have developed an intricate system to limit their activation,
the cell surface of CAR T cells, including CD40L.165 Lastly, investiga- and investigators have conducted unbiased screens to discover key
tors are actively exploring the activation of TLR pathways through molecules within this tightly regulated system. While the first screens
inducible co-stimulatory molecules containing signaling domains of relied on short hairpin RNA (shRNA) approaches identifying the
MyD88,69,191 the central signaling molecule of TLRs, IL-1b, and IL- phosphatase pp2r2d as a negative regulator of ab TCR T cell activa-
18.192 tion in tumor models,205 more recent studies have taken advantage of
CRISPR-Cas9 gene-editing technology.206 One recently published
Transgenic Expression of Cytokines or Constitutively Active in vitro screen identified TCEB2, SOCS1, CBLB, and RASA2 as nega-
Cytokine Receptors tive regulators of ab TCR activation.206 An in vivo screen also has
Cytokines or Cytokine Receptors That Signal through the JAK/STAT highlighted REGNASE1 as a key negative regulator, and REGNASE1
Pathway. Common g-cytokines (IL-2, IL-7, IL-15, IL-21) and IL– knockout CD19-CAR T cells had improved antitumor activity in a
12 and IL23 activate the JAK/STAT signaling pathways in T cells. syngeneic leukemia model.207
While common g-cytokines signal through JAK1/JAK2, and predom-
inantly STAT5, IL-12 and IL-23 signal through JAK2, TYK2, and Modulating Transcription Factors
STAT3 and/or STAT4.193–195 Transgenic expression of these cyto- Transcription factor networks are critical for T cell plasticity, and
kines, demonstrated in numerous preclinical studies, improves the several recent studies have highlighted their importance. For example,
ability of CAR T cells to expand and/or persist, resulting in improved c-Myb (1) is a transcriptional activator of TCF7, a transcription factor
antitumor activity.196–198 Investigators have not only explored secre- critical for memory formation, and (2) represses ZEB2, a transcrip-
tory but also membrane-bound versions of cytokines, which might be tion factor that promotes T cell differentiation.208 In addition, TOX
advantageous since they have the potential to improve cytokine activ- has emerged as a key transcription factor that enforces T cell exhaus-
ity (e.g., IL-15) and also restrict most of the cytokine’s action to the tion, and investigators have shown that TOX- and TOX2-deficient
genetically modified cell.199,200 To mitigate systemic side effects of murine CAR T cells have improved effector function.100–102,209 Other
secreted cytokines, investigators have engineered T cells in which studies have highlighted that the nuclear receptor transcription fac-
cytokine expression is controlled by the nuclear factor of activated tors NR4A1 (NUR77), NR4A2 (NURR1), and NR4A3 (NOR1)
T cells (NFAT) promoter. However, while systemic side effects of orchestrate transcription programs that promote T cell exhaustion
IL-12 could be mitigated in preclinical studies, one clinical study and that deleting all three factors in CAR T cells is necessary for
with NFAT-IL-12-modified tumor-infiltrating lymphocytes (TILs) optimal function.210 Lastly, overexpression of c-Jun (transcription
indicated that the NFAT promoter is insufficient to restrict gene factor AP-1) in CAR T cells increases their functional capacities
expression to activated T cells.201 Another approach to reduce cyto- and reduces terminal T cell differentiation, resulting in improved
kine secretion of gene-modified CAR T cells is to use a construct in antitumor activity.211
which the cytokine gene is placed 30 prime of an internal ribosomal
entry site (IRES).202 A clinical study with MUC16ecto-CAR/IRES- CAR T Cell Trafficking and Tumor Penetration
IL-12 T cells is in progress (ClinicalTrials.gov: NCT02498912). In hematological malignancies, CAR T cells and their corresponding
GD2-CAR T cells (ClinicalTrials.gov: NCT03721068) or GD2-CAR target malignancies share hematopoietic origins, and thus have a
iNK T cells (ClinicalTrials.gov: NCT03294954) expressing IL-15 are higher propensity to migrate to similar areas such bone marrow or
also currently being evaluated in patients with neuroblastoma. In lymph nodes. In contrast, many solid tumors do not attract T cells.
one of these studies (ClinicalTrials.gov: NCT03721068), CAR/IL-15 These tumors are called cold tumors, and they can be further subdi-
T cells are also modified with the iC9 safety switch to allow for the vided into tumors that do not (cold, ignored) or have (cold, excluded)

Molecular Therapy Vol. 28 No 11 November 2020 9


Please cite this article in press as: Wagner et al., CAR T Cell Therapy for Solid Tumors: Bright Future or Dark Reality?, Molecular Therapy (2020), https://
doi.org/10.1016/j.ymthe.2020.09.015

www.moleculartherapy.org

Review

infusion of CAR T cells with the local delivery of an oncolytic adeno-


virus encoding RANTES and IL-15 has improved homing to and
persistence of CAR T cells at tumor sites in preclinical models.218

The tumor stroma itself can be extremely dense with various cell types
protecting the tumor. Targeting the extracellular matrix (ECM) and
degrading these physically protective proteins has also shown
promise preclinically. Heparin sulfate proteoglycans (HSPGs) are
expressed in the ECM as well as neuronal tissue during develop-
ment.219,220 Many cancer cells, especially neuroblastoma, express
high levels of HSPGs. Expression of heparinase on CAR T cells to
degrade HSPGs has demonstrated improved tumor infiltration and
antitumor activity in vivo.221 However, the addition of ECM-degrad-
ing enzymes to standard chemotherapies has provided mixed results,
with some clinical trials showing decreased survival in patients with
the combination therapy.222

Counteracting the Immunosuppressive TME


Solid tumor cells are intermixed with suppressive cell populations
such as tumor-associated macrophages (TAMs), myeloid-derived
suppressor cells (MDSCs), Tregs, and cancer-associated fibroblasts
(CAFs) (Figure 3).223–225 Tumor cells and/or cells of the TME express
a broad range of immune checkpoints, including PD-L1 and ligands
for LAG-3, TIM-3, and TIGIT. MDSCs, Tregs, and/or CAFs also
secrete immunosuppressive cytokines such as transforming growth
factor (TGF)-b and IL-10.226,227 In addition, amino acids that are
critical for T cell function such as arginine and glutamine are often
depleted within the TME, contributing to limited T cell fitness
(Figure 3).228,229 Some of the approaches that counteract the immu-
nosuppressive microenvironment rely on strategies that were
discussed in the section Improving CAR T Cell Fitness. For example,
transgenic expression of IL-15 in cytotoxic T cells counteracts Treg-
Figure 3. Immunosuppressive Solid Tumor Microenvironment
Scheme of the solid tumor microenvironment that consists of (1) tumor vasculature; mediated suppression.230 Herein, we review additional strategies,
(2) stromal cells, including cancer-associated fibroblasts; (3) immunosuppressive including (1) directly counteracting immunosuppressive factors, (2)
cells, including myeloid cells, macrophages, and regulatory T cells (Tregs); (4) hijacking cytokines or growth factors to promote T cell effector func-
suppressive metabolites; (5) inhibitory ligands, including PD-1; and (6) suppressive tion, (3) improving T cell metabolism in the TME, and (4) targeting
cytokines. See text for additional details. non-malignant cells of the tumor stroma.

a T cell infiltrate in their periphery.212 T cell migration into tumors is Directly Counteracting Immunosuppressive Factors
influenced not only by chemokines and adhesion molecules, but also Several preclinical studies have shown that combining PD-1/PD-L1
the tumor vasculature and the presence of other immune cells within blockade with CAR T cell therapy improves antitumor activity.231
the tumor.212–214 Recent studies have highlighted the role of tissue- In an effort to reduce side effects from systemic checkpoint blockade,
resident memory T cells for cancer immunotherapies, which express CAR T cells were genetically modified to express a PD-1/CD28 switch
a unique pattern of adhesion molecules and residency markers.215 or a truncated PD-1 receptor that functions as a dominant negative
Engineering approaches have so far mainly focused on taking receptor (DNR).70,232 In addition, PD-1 was deleted in CAR T cells
advantage of chemokines that are secreted by tumor cells for which by CRISPR-Cas9 gene editing.233–235 All three approaches resulted
T cells do not express the corresponding chemokine receptor (e.g., in improved effector function of CAR T cells in preclinical models.
CXCL8 by melanoma or CCL2 by neuroblastoma).216,217 In preclin- Thus far the clinical experience with these approaches is limited,
ical models, transgenic expression of chemokine receptors on tumor- and only PD-1 knockout abTCR T cells have been evaluated clini-
specific T cells, including CAR T cells, has been shown to enhance cally.236 Other approaches include silencing CTLA-4 or FAS expres-
their ability to home to tumor sites, resulting in improved antitumor sion on the cell surface of tumor-specific or CAR T cells.237,238 More
activity,216,217 and this approach is being evaluated in a clinical recently, investigators demonstrated that expression of a DNR FAS
study for the adoptive immunotherapy of TILs in melanoma receptor also improved the effector function of adoptively transferred
(ClinicalTrials.gov: NCT01740557). Alternatively, combining the tumor-specific T cells.239

10 Molecular Therapy Vol. 28 No 11 November 2020


Please cite this article in press as: Wagner et al., CAR T Cell Therapy for Solid Tumors: Bright Future or Dark Reality?, Molecular Therapy (2020), https://
doi.org/10.1016/j.ymthe.2020.09.015

www.moleculartherapy.org

Review

Hijacking Cytokines or Growth Factors to Promote T Cell antitumor activity in preclinical models, some safety concerns were
Effector Function raised due to FAP expression on stromal cells in healthy tissues;
The TME is rich with cytokines that either inhibit T cell function or nevertheless, this approach has been translated into the clinic.258–261
for which T cells do not express the corresponding receptors. The TAMs have been targeted with CD123- or folate receptor b (FRb)-
inhibitory effects of these cytokines can be negated by expressing CAR T cells in preclinical models,262,263 and CSF1R-CARs have
DNRs or cytokine switch receptors (CSRs) that convert an inhibitory been developed, opening up the possibility to target CSF1R-positive
signal into a growth-promoting signal. Examples of cytokine DNRs TAMs.264 MDSCs express high levels of NKG2D ligands, and in
include DNR-TGF-b receptors.240 DNR-TGF-b-expressing EBV- one preclinical model, combining NKG2D-CAR NK cells with
specific T cells for EBV+ lymphoma have been evaluated in early GD2-CAR T cells resulted in improved antitumor activity for neuro-
phase clinical studies, demonstrating safety and suggesting improved blastoma.265 Lastly, endothelial cells of the tumor vasculature have
efficacy in comparison to their unmodified counterparts.241 CSRs been targeted with CARs specific for VEGF-R2, PSMA, or the
have been developed to take advantage of IL-4 produced within the EDB/EIIIB splice variant of fibronectin in preclinical models.266–268
TME, including IL-4/IL-2, IL-4/IL-7, and IL-4/IL-21 CSRs.242–244 In Of these approaches, only VEGF-R2 CAR T cells have been success-
addition, a TGF-b CSR, consisting of a TGF-b-specific scFv and a fully evaluated in one clinical study. While the results have not been
CD28z signaling domain, has shown promise in preclinical CAR published, they are available (ClinicalTrials.gov: NCT01218867), and
T cell therapy models.245 Of these, the IL-4/IL-2 CSR is currently out of 23 infused patients, only 1 patient achieved a partial response.
being evaluated in one early phase clinical study in the context of
TE1-CAR T cells (ClinicalTrials.gov: NCT01818323). Lastly, col- Combinatorial Therapies
ony-stimulating factor-1 (CSF-1) is an example of a cytokine that is While this review has focused on additional genetic modification to
present in the TME and for which T cells do not express the cognate improve the effector function of CAR T cells, numerous studies are
receptor. One preclinical study has shown that expression of CSF-1R underway to combine CAR T cell therapy with other treatment mo-
in CAR T cells improves their effector function in a CSF-1-dependent dalities to improve outcomes by creating therapeutic synergies.269,270
manner.246 These include efforts, as already mentioned in this review, to combine
CAR T cell therapy with cytokine administration, checkpoint
Improving Metabolic Fitness of CAR T Cells in the TME blockade, oncolytic viruses, radiation, and/or vaccines. Examples
The TME is hypoxic and deprived of critical nutrients that are for oncolytics/CAR T cell therapy combinations include the adminis-
required for T cell proliferation. In addition, the TME (1) is enriched tration of oncolytic viruses encoding BiTEs, cytokines, and/or check-
in metabolic end products that are immunosuppressive, for example, point inhibitors,271–273 or oncolytic viruses that encode CAR tar-
R-2-hydroxyglutarate in tumors with isocitrate dehydrogenase 1/2 gets.274,275 In addition, investigators have explored the use of T cells
mutations;247–249 (2) has increased concentrations of electrolytes, to directly deliver viruses into tumors.276–278 Checkpoint blockade/
such as potassium, that are immunosuppressive;250 and (3) harbors CAR T cell therapy combinations are actively being pursued,279
reactive oxygen species (ROS) that impair T cell function.251,252 including the aforementioned clinical studies combining mesothe-
Several strategies are being actively explored to improve the metabolic lin-CAR T cells with pembrolizumab for the treatment of mesotheli-
function of adoptively transferred T cells. These include ex vivo oma.76 Furthermore, several studies have highlighted the benefit of
loading of tumor-specific T cells with arginine,228 or the genetic combining radiotherapy with the administration of CAR T cells,
modification of CAR T cells with enzymes that are critical for arginine and early clinical studies for patients with B cell lymphoma have pro-
re-synthesis, including argininosuccinate synthase or ornithine trans- duced encouraging results.280 Recent preclinical studies have also
carbamylase.253 Investigators have also focused on manipulating demonstrated the benefit of boosting CAR T cells outside the immu-
glutamine metabolism in the TME, and recent studies have high- nosuppressive TME with amphiphile CAR ligands or RNA-based
lighted that glutamine antagonists and transient inhibition of gluta- vaccines.281,282
minase increase T cell effector function.254,255 To protect CAR
T cells from ROS, investigators have genetically modified T cells to Other approaches include combining CAR T cells with small mole-
secrete catalase. This not only protected T cells from ROS damage cules that target cancer cells that do not affect CAR T cells, with
but also protected unmodified bystander immune cells.256 Lastly, the best example being combining the BTK inhibitor ibrutinib with
small molecules such a metformin are being explored to reduce tumor CD19-CAR T cell therapies for lymphoma.283 In addition, combining
hypoxia.257 CAR T cells with chemotherapeutic agents that upregulate the expres-
sion of the targeted antigens is an attractive option to enhance the
Targeting Non-malignant Cells of the Tumor Stroma antitumor activity of CAR T cells that is actively being pursued.284
CAFs, MDSCs, TAMs, and tumor-associated endothelial cells are Finally, combinational treatment of HER2-CAR T cells with the
present in the tumor stroma, and while not malignant, they promote smac-mimetic birinapant, which promotes apoptosis, significantly
tumor growth/metastasis and immunosuppression and/or secrete enhanced antitumor activity in a TNF-dependent manner.285 Thus,
components to the ECM that hinder immune cell penetration. there is a myriad of combinatorial approaches, and selection of an
CAFs have been targeted by several groups with fibroblast activation optimal combination will most likely depend on the targeted tumor
protein (FAP)-CAR T cells. While FAP-CAR T cells had potent type and disease status. For example, brain tumors, which often

Molecular Therapy Vol. 28 No 11 November 2020 11


Please cite this article in press as: Wagner et al., CAR T Cell Therapy for Solid Tumors: Bright Future or Dark Reality?, Molecular Therapy (2020), https://
doi.org/10.1016/j.ymthe.2020.09.015

www.moleculartherapy.org

Review

present as local disease or with limited sites of recurrences, might be and does not necessarily represent the official views of the NIH. Fig-
ideal tumors to combine local radiation or delivery of an oncolytic vi- ures were created with BioRender.
rus with CAR T cell therapy. In contrast, combining local delivery of
agents with CAR T cell therapy for metastatic cancer should only be REFERENCES
pursued if treating selected metastases induces an abscopal effect,286 1. Porter, D.L., Levine, B.L., Kalos, M., Bagg, A., and June, C.H. (2011). Chimeric an-
leading to the regression of distant, untreated metastases. Feasibility tigen receptor-modified T cells in chronic lymphoid leukemia. N. Engl. J. Med. 365,
725–733.
and costs are also a major consideration; in this regard, repurposing
2. Grupp, S.A., Kalos, M., Barrett, D., Aplenc, R., Porter, D.L., Rheingold, S.R.,
FDA-approved drugs for combinatorial CAR T cell therapy should
Teachey, D.T., Chew, A., Hauck, B., Wright, J.F., et al. (2013). Chimeric antigen
actively be explored as for other cancer therapeutics.287,288 receptor-modified T cells for acute lymphoid leukemia. N. Engl. J. Med. 368,
1509–1518.
Conclusions 3. Maude, S.L., Frey, N., Shaw, P.A., Aplenc, R., Barrett, D.M., Bunin, N.J., Chew, A.,
CAR T cells for solid tumors so far have limited activity in early phase Gonzalez, V.E., Zheng, Z., Lacey, S.F., et al. (2014). Chimeric antigen receptor T cells
clinical studies, raising concerns of whether CAR T cells can be devel- for sustained remissions in leukemia. N. Engl. J. Med. 371, 1507–1517.

oped into a curative therapeutic approach in the future. While 4. Fry, T.J., Shah, N.N., Orentas, R.J., Stetler-Stevenson, M., Yuan, C.M., Ramakrishna,
S., Wolters, P., Martin, S., Delbrook, C., Yates, B., et al. (2018). CD22-targeted CAR
currently ineffective, clinical and preclinical studies have identified
T cells induce remission in B-ALL that is naive or resistant to CD19-targeted CAR
roadblocks, including (1) an antigen dilemma, (2) T cell fitness and immunotherapy. Nat. Med. 24, 20–28.
survival before reaching the tumor site, (3) inability of T cells to effi- 5. Gardner, R.A., Finney, O., Annesley, C., Brakke, H., Summers, C., Leger, K.,
ciently traffic to tumor sites and penetrate physical barriers, and (4) Bleakley, M., Brown, C., Mgebroff, S., Kelly-Spratt, K.S., et al. (2017). Intent-to-treat
an immunosuppressive TME. In the last decade, investigators have leukemia remission by CD19 CAR T cells of defined formulation and dose in chil-
dren and young adults. Blood 129, 3322–3331.
developed elegant solutions and/or countermeasures in preclinical
studies, engineering CAR T cells with significantly improved effector 6. Ramos, C.A., Grover, N.S., Beaven, A.W., Lulla, P.D., Wu, M.F., Ivanova, A., Wang,
T., Shea, T.C., Rooney, C.M., Dittus, C., et al. (2020). Anti-CD30 CAR-T cell therapy
function and/or combining them with other treatment modalities. in relapsed and refractory Hodgkin lymphoma. J. Clin. Oncol. Published online July
Thus, we strongly believe that the future of CAR T cells for solid tu- 23, 2020. 10.1200/JCO.20.01342.
mors is bright, and they will not become a mere footnote in the history 7. Ramos, C.A., Savoldo, B., Torrano, V., Ballard, B., Zhang, H., Dakhova, O., Liu, E.,
of cancer immunotherapy. A key challenge will be how to prioritize Carrum, G., Kamble, R.T., Gee, A.P., et al. (2016). Clinical responses with T lympho-
and streamline the clinical evaluation of the developed approaches. cytes targeting malignancy-associated k light chains. J. Clin. Invest. 126, 2588–2596.
In addition, there is a continued need to advance our understanding 8. Raje, N., Berdeja, J., Lin, Y., Siegel, D., Jagannath, S., Madduri, D., Liedtke, M.,
in basic CAR T cell biology and how cancer cells and resident immune Rosenblatt, J., Maus, M.V., Turka, A., et al. (2019). Anti-BCMA CAR T-cell therapy
bb2121 in relapsed or refractory multiple myeloma. N. Engl. J. Med. 380, 1726–1737.
cells interact with adoptively transferred CAR T cells. Lastly, future
9. Turtle, C.J., Hanafi, L.A., Berger, C., Hudecek, M., Pender, B., Robinson, E.,
CAR T cell design might benefit from considering the “ten principles
Hawkins, R., Chaney, C., Cherian, S., Chen, X., et al. (2016). Immunotherapy of
for good design” that were put forward by the eminent industrial non-Hodgkin’s lymphoma with a defined ratio of CD8+ and CD4+ CD19-specific
designer Dieter Rams:289 good design (1) is innovative, (2) makes a chimeric antigen receptor-modified T cells. Sci. Transl. Med. 8, 355ra116.
product useful, (3) is aesthetic, (4) makes a product understandable, 10. Kochenderfer, J.N., Somerville, R.P.T., Lu, T., Yang, J.C., Sherry, R.M., Feldman,
(5) is unobtrusive, (6) is honest, (7) is long-lasting, (8) is thorough S.A., McIntyre, L., Bot, A., Rossi, J., Lam, N., and Rosenberg, S.A. (2017). Long-dura-
down to the last detail, (9) is environmentally friendly, and (10) in- tion complete remissions of diffuse large B cell lymphoma after anti-CD19 chimeric
antigen receptor T cell therapy. Mol. Ther. 25, 2245–2253.
volves as little design as possible.
11. Holstein, S.A., and Lunning, M.A. (2020). CAR T-cell therapy in hematologic ma-
lignancies: a voyage in progress. Clin. Pharmacol. Ther. 107, 112–122.
AUTHOR CONTRIBUTIONS
12. Boyiadzis, M.M., Dhodapkar, M.V., Brentjens, R.J., Kochenderfer, J.N., Neelapu,
J.W., E.W., and S.G. wrote the manuscript. All authors (J.W., E.W., S.S., Maus, M.V., Porter, D.L., Maloney, D.G., Grupp, S.A., Mackall, C.L., et al.
C.D., and S.G.) reviewed and edited the manuscript. (2018). Chimeric antigen receptor (CAR) T therapies for the treatment of
hematologic malignancies: clinical perspective and significance. J. Immunother.
Cancer 6, 137.
CONFLICTS OF INTEREST
J.W., C.D., and S.G. have patent applications in the fields of T cell and/ 13. Majzner, R.G., and Mackall, C.L. (2019). Clinical lessons learned from the first leg of
the CAR T cell journey. Nat. Med. 25, 1341–1355.
or gene therapy for cancer. S.G. has a research collaboration with
TESSA Therapeutics, is a DSMB member of Immatics, and is on 14. Neelapu, S.S., Tummala, S., Kebriaei, P., Wierda, W., Locke, F.L., Lin, Y., Jain, N.,
Daver, N., Gulbis, A.M., Adkins, S., et al. (2018). Toxicity management after
the Scientific Advisory Board of Tidal. chimeric antigen receptor T cell therapy: one size does not fit “ALL”. Nat. Rev.
Clin. Oncol. 15, 218.
ACKNOWLEDGMENTS 15. Lee, D.W., Santomasso, B.D., Locke, F.L., Ghobadi, A., Turtle, C.J., Brudno, J.N.,
The work was supported by National Institutes of Health grant Maus, M.V., Park, J.H., Mead, E., Pavletic, S., et al. (2019). ASTCT consensus
R01NS106379 (to S.G.); the Alex's Lemonade Stand Foundation grading for cytokine release syndrome and neurologic toxicity associated with im-
mune effector cells. Biol. Blood Marrow Transplant. 25, 625–638.
and the Cure4Cam Foundation (ALSF; Young Investigator Grant;
16. Goff, S.L., Morgan, R.A., Yang, J.C., Sherry, R.M., Robbins, P.F., Restifo, N.P.,
to J.W.); the Alliance for Cancer Gene Therapy (ACGT; to S.G.);
Feldman, S.A., Lu, Y.C., Lu, L., Zheng, Z., et al. (2019). Pilot trial of adoptive transfer
and by the American Lebanese Syrian Associated Charities (to C.D. of chimeric antigen receptor-transduced T cells targeting EGFRvIII in patients with
and S.G.). The content is solely the responsibility of the authors glioblastoma. J. Immunother. 42, 126–135.

12 Molecular Therapy Vol. 28 No 11 November 2020


Please cite this article in press as: Wagner et al., CAR T Cell Therapy for Solid Tumors: Bright Future or Dark Reality?, Molecular Therapy (2020), https://
doi.org/10.1016/j.ymthe.2020.09.015

www.moleculartherapy.org

Review

17. O’Rourke, D.M., Nasrallah, M.P., Desai, A., Melenhorst, J.J., Mansfield, K., noglobulin-derived V regions and T-cell receptor-derived C regions. Biochem.
Morrissette, J.J.D., Martinez-Lage, M., Brem, S., Maloney, E., Shen, A., et al. Biophys. Res. Commun. 149, 960–968.
(2017). A single dose of peripherally infused EGFRvIII-directed CAR T cells medi-
35. Gross, G., Waks, T., and Eshhar, Z. (1989). Expression of immunoglobulin-T-cell
ates antigen loss and induces adaptive resistance in patients with recurrent glioblas-
receptor chimeric molecules as functional receptors with antibody-type specificity.
toma. Sci. Transl. Med. 9, eaaa0984.
Proc. Natl. Acad. Sci. USA 86, 10024–10028.
18. Morgan, R.A., Yang, J.C., Kitano, M., Dudley, M.E., Laurencot, C.M., and
36. Eshhar, Z., Waks, T., Gross, G., and Schindler, D.G. (1993). Specific activation and
Rosenberg, S.A. (2010). Case report of a serious adverse event following the admin-
targeting of cytotoxic lymphocytes through chimeric single chains consisting of
istration of T cells transduced with a chimeric antigen receptor recognizing ERBB2.
antibody-binding domains and the gamma or zeta subunits of the immunoglobulin
Mol. Ther. 18, 843–851.
and T-cell receptors. Proc. Natl. Acad. Sci. USA 90, 720–724.
19. Lamers, C.H., Sleijfer, S., Vulto, A.G., Kruit, W.H., Kliffen, M., Debets, R., Gratama,
37. June, C.H., and Sadelain, M. (2018). Chimeric antigen receptor therapy. N. Engl. J.
J.W., Stoter, G., and Oosterwijk, E. (2006). Treatment of metastatic renal cell carci-
Med. 379, 64–73.
noma with autologous T-lymphocytes genetically retargeted against carbonic anhy-
drase IX: first clinical experience. J. Clin. Oncol. 24, e20–e22. 38. Dotti, G., Gottschalk, S., Savoldo, B., and Brenner, M.K. (2014). Design and devel-
opment of therapies using chimeric antigen receptor-expressing T cells. Immunol.
20. Lamers, C.H., Sleijfer, S., van Steenbergen, S., van Elzakker, P., van Krimpen, B.,
Rev. 257, 107–126.
Groot, C., Vulto, A., den Bakker, M., Oosterwijk, E., Debets, R., and Gratama,
J.W. (2013). Treatment of metastatic renal cell carcinoma with CAIX CAR-engi- 39. Shaffer, D.R., Savoldo, B., Yi, Z., Chow, K.K., Kakarla, S., Spencer, D.M., Dotti, G.,
neered T cells: clinical evaluation and management of on-target toxicity. Mol. Wu, M.F., Liu, H., Kenney, S., and Gottschalk, S. (2011). T cells redirected
Ther. 21, 904–912. against CD70 for the immunotherapy of CD70-positive malignancies. Blood 117,
4304–4314.
21. Kershaw, M.H., Westwood, J.A., Parker, L.L., Wang, G., Eshhar, Z., Mavroukakis,
S.A., White, D.E., Wunderlich, J.R., Canevari, S., Rogers-Freezer, L., et al. (2006). 40. Davies, D.M., Foster, J., Van Der Stegen, S.J., Parente-Pereira, A.C., Chiapero-
A phase I study on adoptive immunotherapy using gene-modified T cells for ovarian Stanke, L., Delinassios, G.J., Burbridge, S.E., Kao, V., Liu, Z., Bosshard-Carter, L.,
cancer. Clin. Cancer Res. 12, 6106–6115. et al. (2012). Flexible targeting of ErbB dimers that drive tumorigenesis by using
genetically engineered T cells. Mol. Med. 18, 565–576.
22. Brown, C.E., Alizadeh, D., Starr, R., Weng, L., Wagner, J.R., Naranjo, A., Ostberg,
J.R., Blanchard, M.S., Kilpatrick, J., Simpson, J., et al. (2016). Regression of glioblas- 41. Ma, Q., Garber, H.R., Lu, S., He, H., Tallis, E., Ding, X., Sergeeva, A., Wood, M.S.,
toma after chimeric antigen receptor T-cell therapy. N. Engl. J. Med. 375, 2561– Dotti, G., Salvado, B., et al. (2016). A novel TCR-like CAR with specificity for
2569. PR1/HLA-A2 effectively targets myeloid leukemia in vitro when expressed in hu-
man adult peripheral blood and cord blood T cells. Cytotherapy 18, 985–994.
23. Ahmed, N., Brawley, V.S., Hegde, M., Robertson, C., Ghazi, A., Gerken, C., Liu, E.,
Dakhova, O., Ashoori, A., Corder, A., et al. (2015). Human epidermal growth factor 42. Rafiq, S., Purdon, T.J., Daniyan, A.F., Koneru, M., Dao, T., Liu, C., Scheinberg, D.A.,
receptor 2 (HER2)-specific chimeric antigen receptor-modified T cells for the and Brentjens, R.J. (2017). Optimized T-cell receptor-mimic chimeric antigen re-
immunotherapy of HER2-positive sarcoma. J. Clin. Oncol. 33, 1688–1696. ceptor T cells directed toward the intracellular Wilms tumor 1 antigen. Leukemia
31, 1788–1797.
24. Wang, Y., Chen, M., Wu, Z., Tong, C., Dai, H., Guo, Y., Liu, Y., Huang, J., Lv, H.,
Luo, C., et al. (2018). CD133-directed CAR T cells for advanced metastasis malig- 43. Maus, M.V., Plotkin, J., Jakka, G., Stewart-Jones, G., Rivière, I., Merghoub, T.,
nancies: a phase I trial. OncoImmunology 7, e1440169. Wolchok, J., Renner, C., and Sadelain, M. (2017). An MHC-restricted antibody-
based chimeric antigen receptor requires TCR-like affinity to maintain antigen spec-
25. Thistlethwaite, F.C., Gilham, D.E., Guest, R.D., Rothwell, D.G., Pillai, M., Burt, D.J.,
ificity. Mol. Ther. Oncolytics 3, 1–9.
Byatte, A.J., Kirillova, N., Valle, J.W., Sharma, S.K., et al. (2017). The clinical efficacy
of first-generation carcinoembryonic antigen (CEACAM5)-specific CAR T cells is 44. Töpfer, K., Kempe, S., Müller, N., Schmitz, M., Bachmann, M., Cartellieri, M.,
limited by poor persistence and transient pre-conditioning-dependent respiratory Schackert, G., and Temme, A. (2011). Tumor evasion from T cell surveillance.
toxicity. Cancer Immunol. Immunother. 66, 1425–1436. J. Biomed. Biotechnol. 2011, 918471.

26. Ahmed, N., Brawley, V., Hegde, M., Bielamowicz, K., Kalra, M., Landi, D., 45. Rafiq, S., Yeku, O.O., Jackson, H.J., Purdon, T.J., van Leeuwen, D.G., Drakes, D.J.,
Robertson, C., Gray, T.L., Diouf, O., Wakefield, A., et al. (2017). HER2-specific Song, M., Miele, M.M., Li, Z., Wang, P., et al. (2018). Targeted delivery of a PD-
chimeric antigen receptor-modified virus-specific T cells for progressive glioblas- 1-blocking scFv by CAR-T cells enhances anti-tumor efficacy in vivo. Nat.
toma: a phase 1 dose-escalation trial. JAMA Oncol. 3, 1094–1101. Biotechnol. 36, 847–856.

27. Rafiq, S., Hackett, C.S., and Brentjens, R.J. (2020). Engineering strategies to over- 46. Hombach, A.A., and Abken, H. (2011). Costimulation by chimeric antigen receptors
come the current roadblocks in CAR T cell therapy. Nat. Rev. Clin. Oncol. 17, revisited the T cell antitumor response benefits from combined CD28-OX40 signal-
147–167. ling. Int. J. Cancer 129, 2935–2944.

28. Rodriguez-Garcia, A., Palazon, A., Noguera-Ortega, E., Powell, D.J., Jr., and Guedan, 47. Collinson-Pautz, M.R., Chang, W.C., Lu, A., Khalil, M., Crisostomo, J.W., Lin, P.Y.,
S. (2020). CAR-T cells hit the tumor microenvironment: strategies to overcome tu- Mahendravada, A., Shinners, N.P., Brandt, M.E., Zhang, M., et al. (2019).
mor escape. Front. Immunol. 11, 1109. Constitutively active MyD88/CD40 costimulation enhances expansion and efficacy
of chimeric antigen receptor T cells targeting hematological malignancies. Leukemia
29. Schmidts, A., and Maus, M.V. (2018). Making CAR T cells a solid option for solid
33, 2195–2207.
tumors. Front. Immunol. 9, 2593.
48. Guedan, S., Chen, X., Madar, A., Carpenito, C., McGettigan, S.E., Frigault, M.J., Lee,
30. Knochelmann, H.M., Smith, A.S., Dwyer, C.J., Wyatt, M.M., Mehrotra, S., and
J., Posey, A.D., Jr., Scholler, J., Scholler, N., et al. (2014). ICOS-based chimeric anti-
Paulos, C.M. (2018). CAR T cells in solid tumors: blueprints for building effective
gen receptors program bipolar TH17/TH1 cells. Blood 124, 1070–1080.
therapies. Front. Immunol. 9, 1740.
49. Nair, S., Wang, J.B., Tsao, S.T., Liu, Y., Zhu, W., Slayton, W.B., Moreb, J.S., Dong, L.,
31. Depil, S., Duchateau, P., Grupp, S.A., Mufti, G., and Poirot, L. (2020). “Off-the-shelf”
and Chang, L.J. (2019). Functional improvement of chimeric antigen receptor
allogeneic CAR T cells: development and challenges. Nat. Rev. Drug Discov. 19,
through intrinsic interleukin-15Ra signaling. Curr. Gene Ther. 19, 40–53.
185–199.
50. Salter, A.I., Ivey, R.G., Kennedy, J.J., Voillet, V., Rajan, A., Alderman, E.J.,
32. Qasim, W. (2019). Allogeneic CAR T cell therapies for leukemia. Am. J. Hematol. 94
Voytovich, U.J., Lin, C., Sommermeyer, D., Liu, L., et al. (2018).
(S1), S50–S54.
Phosphoproteomic analysis of chimeric antigen receptor signaling reveals kinetic
33. Frey, N., and Porter, D. (2019). Cytokine release syndrome with chimeric antigen and quantitative differences that affect cell function. Sci. Signal. 11, eaat6753.
receptor T cell therapy. Biol. Blood Marrow Transplant. 25, e123–e127.
51. Boroughs, A.C., Larson, R.C., Marjanovic, N.D., Gosik, K., Castano, A.P., Porter,
34. Kuwana, Y., Asakura, Y., Utsunomiya, N., Nakanishi, M., Arata, Y., Itoh, S., Nagase, C.B.M., Lorrey, S.J., Ashenberg, O., Jerby, L., Hofree, M., et al. (2020). A distinct
F., and Kurosawa, Y. (1987). Expression of chimeric receptor composed of immu- transcriptional program in human CAR T cells bearing the 4-1BB signaling domain

Molecular Therapy Vol. 28 No 11 November 2020 13


Please cite this article in press as: Wagner et al., CAR T Cell Therapy for Solid Tumors: Bright Future or Dark Reality?, Molecular Therapy (2020), https://
doi.org/10.1016/j.ymthe.2020.09.015

www.moleculartherapy.org

Review

revealed by scRNA-seq. Mol. Ther. , Published online July 25, 2020. https://doi.org/ 68. Shum, T., Omer, B., Tashiro, H., Kruse, R.L., Wagner, D.L., Parikh, K., Yi, Z., Sauer,
10.1016/j.ymthe.2020.07.023. T., Liu, D., Parihar, R., et al. (2017). Constitutive signaling from an engineered IL7
receptor promotes durable tumor elimination by tumor-redirected T cells. Cancer
52. Xhangolli, I., Dura, B., Lee, G., Kim, D., Xiao, Y., and Fan, R. (2019). Single-cell anal-
Discov. 7, 1238–1247.
ysis of CAR-T cell activation reveals a mixed TH1/TH2 response independent of dif-
ferentiation. Genomics Proteomics Bioinformatics 17, 129–139. 69. Mata, M., Gerken, C., Nguyen, P., Krenciute, G., Spencer, D.M., and Gottschalk, S.
(2017). Inducible activation of MyD88 and CD40 in CAR T cells results in control-
53. Kawalekar, O.U., O’ Connor, R.S., Fraietta, J.A., Guo, L., McGettigan, S.E., Posey,
lable and potent antitumor activity in preclinical solid tumor models. Cancer
A.D., Jr., Patel, P.R., Guedan, S., Scholler, J., Keith, B., et al. (2016). Distinct signaling
Discov. 7, 1306–1319.
of coreceptors regulates specific metabolism pathways and impacts memory devel-
opment in CAR T cells. Immunity 44, 712. 70. Cherkassky, L., Morello, A., Villena-Vargas, J., Feng, Y., Dimitrov, D.S., Jones, D.R.,
Sadelain, M., and Adusumilli, P.S. (2016). Human CAR T cells with cell-intrinsic
54. Quintarelli, C., Orlando, D., Boffa, I., Guercio, M., Polito, V.A., Petretto, A.,
PD-1 checkpoint blockade resist tumor-mediated inhibition. J. Clin. Invest. 126,
Lavarello, C., Sinibaldi, M., Weber, G., Del Bufalo, F., et al. (2018). Choice of costi-
3130–3144.
mulatory domains and of cytokines determines CAR T-cell activity in neuroblas-
toma. OncoImmunology 7, e1433518. 71. Junghans, R.P., Ma, Q., Rathore, R., Gomes, E.M., Bais, A.J., Lo, A.S., Abedi, M.,
Davies, R.A., Cabral, H.J., Al-Homsi, A.S., and Cohen, S.I. (2016). Phase I trial of
55. Zhao, Z., Condomines, M., van der Stegen, S.J.C., Perna, F., Kloss, C.C., Gunset, G.,
anti-PSMA designer CAR-T cells in prostate cancer: possible role for interacting
Plotkin, J., and Sadelain, M. (2015). Structural design of engineered costimulation
interleukin 2-T cell pharmacodynamics as a determinant of clinical response.
determines tumor rejection kinetics and persistence of CAR T cells. Cancer Cell
Prostate 76, 1257–1270.
28, 415–428.
72. Specht, J.M., Lee, S., Turtle, C., Berger, C., Veatch, J., Gooley, T., Mullane, E.,
56. Ramos, C.A., Rouce, R., Robertson, C.S., Reyna, A., Narala, N., Vyas, G., Mehta, B.,
Chaney, C., Riddell, S., and Maloney, D.G. (2018). Phase I study of immunotherapy
Zhang, H., Dakhova, O., Carrum, G., et al. (2018). In vivo fate and activity of second-
for advanced ROR1+ malignancies with autologous ROR1-specific chimeric antigen
versus third-generation CD19-specific CAR-T cells in B cell non-Hodgkin’s lym-
receptor-modified (CAR)-T cells. J. Clin. Oncol. 36 (Suppl ), TPS79.
phomas. Mol. Ther. 26, 2727–2737.
73. Tang, X., Liu, F., Liu, Z., Cao, Y., Zhang, Z., Wang, Y., Huang, J., Fan, S., Zhao, S.,
57. Guest, R.D., Hawkins, R.E., Kirillova, N., Cheadle, E.J., Arnold, J., O’Neill, A., Irlam, Chen, Y., et al. (2020). Bioactivity and safety of B7-H3-targeted chimeric antigen re-
J., Chester, K.A., Kemshead, J.T., Shaw, D.M., et al. (2005). The role of extracellular ceptor T cells against anaplastic meningioma. Clin. Transl. Immunology 9, e1137.
spacer regions in the optimal design of chimeric immune receptors: evaluation of
four different scFvs and antigens. J. Immunother. 28, 203–211. 74. Brown, C.E., Badie, B., Barish, M.E., Weng, L., Ostberg, J.R., Chang, W.C., Naranjo,
A., Starr, R., Wagner, J., Wright, C., et al. (2015). Bioactivity and safety of IL13Ra2-
58. Majzner, R.G., Rietberg, S.P., Sotillo, E., Dong, R., Vachharajani, V.T., Labanieh, L., redirected chimeric antigen receptor CD8+ T cells in patients with recurrent glio-
Myklebust, J.H., Kadapakkam, M., Weber, E.W., Tousley, A.M., et al. (2020). Tuning blastoma. Clin. Cancer Res. 21, 4062–4072.
the antigen density requirement for CAR T-cell activity. Cancer Discov. 10,
702–723. 75. Keu, K.V., Witney, T.H., Yaghoubi, S., Rosenberg, J., Kurien, A., Magnusson, R.,
Williams, J., Habte, F., Wagner, J.R., Forman, S., et al. (2017). Reporter gene imaging
59. Hudecek, M., Lupo-Stanghellini, M.T., Kosasih, P.L., Sommermeyer, D., Jensen, of targeted T cell immunotherapy in recurrent glioma. Sci. Transl. Med. 9, eaag2196.
M.C., Rader, C., and Riddell, S.R. (2013). Receptor affinity and extracellular domain
modifications affect tumor recognition by ROR1-specific chimeric antigen receptor 76. Adusumilli, P.S., Zauderer, M.G., Rusch, V.W., O’Cearbhaill, R.E., Zhu, A., Ngai,
T cells. Clin. Cancer Res. 19, 3153–3164. D.A., McGee, E., Chintala, N.K., Messinger, J.C., Vincent, A., et al. (2019). A phase
I clinical trial of malignant pleural disease treated with regionally delivered autolo-
60. James, S.E., Greenberg, P.D., Jensen, M.C., Lin, Y., Wang, J., Till, B.G., Raubitschek, gous mesothelin-targeted CAR T cells: safety and efficacy. Cancer Res. 79 (Suppl ),
A.A., Forman, S.J., and Press, O.W. (2008). Antigen sensitivity of CD22-specific CT026.
chimeric TCR is modulated by target epitope distance from the cell membrane.
J. Immunol. 180, 7028–7038. 77. Hegde, M.,D.C., Zhang, H., Mata, M., Gerken, C., Shree, A., Yi, Z., Brawley, V.,
Dakhova, O., Wu, M.-F., et al. (2017). Expansion of HER2-CAR T cells after lym-
61. Feucht, J., Sun, J., Eyquem, J., Ho, Y.J., Zhao, Z., Leibold, J., Dobrin, A., Cabriolu, A., phodepletion and clinical responses in patients with advanced sarcoma. J. Clin.
Hamieh, M., and Sadelain, M. (2019). Calibration of CAR activation potential di- Oncol 35 (15 Suppl), 10508–10508.
rects alternative T cell fates and therapeutic potency. Nat. Med. 25, 82–88.
78. Kebriaei, P., Singh, H., Huls, M.H., Figliola, M.J., Bassett, R., Olivares, S., Jena, B.,
62. Wijewarnasuriya, D., Bebernitz, C., Lopez, A.V., Rafiq, S., and Brentjens, R.J. (2020). Dawson, M.J., Kumaresan, P.R., Su, S., et al. (2016). Phase I trials using Sleeping
Excessive costimulation leads to dysfunction of adoptively transferred T cells. Beauty to generate CD19-specific CAR T cells. J. Clin. Invest. 126, 3363–3376.
Cancer Immunol. Res. 8, 732–742.
79. Zhu, A.X., Gold, P.J., El-Khoueiry, A.B., Abrams, T.A., Morikawa, H., Ohishi, N.,
63. Guedan, S., Madar, A., Casado-Medrano, V., Shaw, C., Wing, A., Liu, F., Young, Ohtomo, T., and Philip, P.A. (2013). First-in-man phase I study of GC33, a novel
R.M., June, C.H., and Posey, A.D., Jr. (2020). Single residue in CD28-costimulated recombinant humanized antibody against glypican-3, in patients with advanced he-
CAR-T cells limits long-term persistence and antitumor durability. J. Clin. Invest. patocellular carcinoma. Clin. Cancer Res. 19, 920–928.
130, 3087–3097.
80. Beatty, G.L., O’Hara, M.H., Lacey, S.F., Torigian, D.A., Nazimuddin, F., Chen, F.,
64. Long, A.H., Haso, W.M., Shern, J.F., Wanhainen, K.M., Murgai, M., Ingaramo, M., Kulikovskaya, I.M., Soulen, M.C., McGarvey, M., Nelson, A.M., et al. (2018).
Smith, J.P., Walker, A.J., Kohler, M.E., Venkateshwara, V.R., et al. (2015). 4-1BB Activity of mesothelin-specific chimeric antigen receptor T cells against pancreatic
costimulation ameliorates T cell exhaustion induced by tonic signaling of chimeric carcinoma metastases in a phase 1 trial. Gastroenterology 155, 29–32.
antigen receptors. Nat. Med. 21, 581–590.
81. Pule, M.A., Savoldo, B., Myers, G.D., Rossig, C., Russell, H.V., Dotti, G., Huls, M.H.,
65. Gomes-Silva, D., Mukherjee, M., Srinivasan, M., Krenciute, G., Dakhova, O., Zheng, Liu, E., Gee, A.P., Mei, Z., et al. (2008). Virus-specific T cells engineered to coexpress
Y., Cabral, J.M.S., Rooney, C.M., Orange, J.S., Brenner, M.K., and Mamonkin, M. tumor-specific receptors: persistence and antitumor activity in individuals with neu-
(2017). Tonic 4-1BB costimulation in chimeric antigen receptors impedes T cell sur- roblastoma. Nat. Med. 14, 1264–1270.
vival and is vector-dependent. Cell Rep. 21, 17–26.
82. Louis, C.U., Savoldo, B., Dotti, G., Pule, M., Yvon, E., Myers, G.D., Rossig, C.,
66. Xiong, W., Chen, Y., Kang, X., Chen, Z., Zheng, P., Hsu, Y.H., Jang, J.H., Qin, L., Liu, Russell, H.V., Diouf, O., Liu, E., et al. (2011). Antitumor activity and long-term
H., Dotti, G., and Liu, D. (2018). Immunological synapse predicts effectiveness of fate of chimeric antigen receptor-positive T cells in patients with neuroblastoma.
chimeric antigen receptor cells. Mol. Ther. 26, 963–975. Blood 118, 6050–6056.
67. Davenport, A.J., Cross, R.S., Watson, K.A., Liao, Y., Shi, W., Prince, H.M., Beavis, 83. Park, J.R., Digiusto, D.L., Slovak, M., Wright, C., Naranjo, A., Wagner, J.,
P.A., Trapani, J.A., Kershaw, M.H., Ritchie, D.S., et al. (2018). Chimeric antigen re- Meechoovet, H.B., Bautista, C., Chang, W.C., Ostberg, J.R., and Jensen, M.C.
ceptor T cells form nonclassical and potent immune synapses driving rapid cytotox- (2007). Adoptive transfer of chimeric antigen receptor re-directed cytolytic
icity. Proc. Natl. Acad. Sci. USA 115, E2068–E2076. T lymphocyte clones in patients with neuroblastoma. Mol. Ther. 15, 825–833.

14 Molecular Therapy Vol. 28 No 11 November 2020


Please cite this article in press as: Wagner et al., CAR T Cell Therapy for Solid Tumors: Bright Future or Dark Reality?, Molecular Therapy (2020), https://
doi.org/10.1016/j.ymthe.2020.09.015

www.moleculartherapy.org

Review

84. Ao, X., Yang, Y., Li, W., Tan, Y., Guo, W., Ao, L., He, X., Wu, X., Xia, J., Xu, X., and 101. Scott, A.C., Dündar, F., Zumbo, P., Chandran, S.S., Klebanoff, C.A., Shakiba, M.,
Guo, J. (2019). Anti-aFR CAR-engineered NK-92 cells display potent cytotoxicity Trivedi, P., Menocal, L., Appleby, H., Camara, S., et al. (2019). TOX is a critical regu-
against aFR-positive ovarian cancer. J. Immunother. 42, 284–296. lator of tumour-specific T cell differentiation. Nature 571, 270–274.
85. Brown, C.E., Aguilar, B., Starr, R., Yang, X., Chang, W.C., Weng, L., Chang, B., 102. Khan, O., Giles, J.R., McDonald, S., Manne, S., Ngiow, S.F., Patel, K.P., Werner,
Sarkissian, A., Brito, A., Sanchez, J.F., et al. (2018). Optimization of IL13Ra2-tar- M.T., Huang, A.C., Alexander, K.A., Wu, J.E., et al. (2019). TOX transcriptionally
geted chimeric antigen receptor T cells for improved anti-tumor efficacy against and epigenetically programs CD8+ T cell exhaustion. Nature 571, 211–218.
glioblastoma. Mol. Ther. 26, 31–44. 103. Ghoneim, H.E., Fan, Y., Moustaki, A., Abdelsamed, H.A., Dash, P., Dogra, P.,
86. Liu, Y., Guo, Y., Wu, Z., Feng, K., Tong, C., Wang, Y., Dai, H., Shi, F., Yang, Q., and Carter, R., Awad, W., Neale, G., Thomas, P.G., and Youngblood, B. (2017). De
Han, W. (2020). Anti-EGFR chimeric antigen receptor-modified T cells in metasta- novo epigenetic programs inhibit PD-1 blockade-mediated T cell rejuvenation.
tic pancreatic carcinoma: a phase I clinical trial. Cytotherapy 22, P573–P580. Cell 170, 142–157.e19.

87. Heczey, A., Louis, C.U., Savoldo, B., Dakhova, O., Durett, A., Grilley, B., Liu, H., Wu, 104. Youngblood, B., Hale, J.S., Kissick, H.T., Ahn, E., Xu, X., Wieland, A., Araki, K.,
M.F., Mei, Z., Gee, A., et al. (2017). CAR T cells administered in combination with West, E.E., Ghoneim, H.E., Fan, Y., et al. (2017). Effector CD8 T cells dedifferentiate
lymphodepletion and PD-1 inhibition to patients with neuroblastoma. Mol. Ther. into long-lived memory cells. Nature 552, 404–409.
25, 2214–2224. 105. Zebley, C.C., Gottschalk, S., and Youngblood, B. (2020). Rewriting history: epige-
88. Shi, D., Shi, Y., Kaseb, A.O., Qi, X., Zhang, Y., Chi, J., Lu, Q., Gao, H., Jiang, H., netic reprogramming of CD8+ T cell differentiation to enhance immunotherapy.
Wang, H., et al. (2020). Chimeric antigen receptor-glypican-3 T-cell therapy for Trends Immunol. 41, 665–675.
advanced hepatocellular carcinoma: results of phase I trials. Clin. Cancer Res 26, 106. Fraietta, J.A., Nobles, C.L., Sammons, M.A., Lundh, S., Carty, S.A., Reich, T.J.,
3979–3989. Cogdill, A.P., Morrissette, J.J.D., DeNizio, J.E., Reddy, S., et al. (2018). Disruption
of TET2 promotes the therapeutic efficacy of CD19-targeted T cells. Nature 558,
89. Navai, S.A., Derenzo, C., Joseph, S., Sanber, K., Byrd, T., Zhang, H., Mata, M.,
307–312.
Gerken, C., Shree, A., Mathew, P.R., et al. (2019). Administration of HER2-CAR
T cells after lymphodepletion safely improves T cell expansion and induces clinical 107. Dwyer, C.J., Knochelmann, H.M., Smith, A.S., Wyatt, M.M., Rangel Rivera, G.O.,
responses in patients with advanced sarcomas. Cancer Res. 79 (Suppl ), LB-147. Arhontoulis, D.C., Bartee, E., Li, Z., Rubinstein, M.P., and Paulos, C.M. (2019).
Fueling cancer immunotherapy with common gamma chain cytokines. Front.
90. Hegde, M., Joseph, S.K., Pashankar, F., DeRenzo, C., Sanber, K., Navai, S., Byrd, T.T.,
Immunol. 10, 263.
Hicks, J., Xu, M.L., Gerken, C., et al. (2020). Tumor response and endogenous im-
mune reactivity after administration of HER2 CAR T cells in a child with metastatic 108. Xu, Y., Zhang, M., Ramos, C.A., Durett, A., Liu, E., Dakhova, O., Liu, H., Creighton,
rhabdomyosarcoma. Nat. Commun. 11, 3549. C.J., Gee, A.P., Heslop, H.E., et al. (2014). Closely related T-memory stem cells
correlate with in vivo expansion of CAR.CD19-T cells and are preserved by IL-7
91. Haas, A.R., Tanyi, J.L., O’Hara, M.H., Gladney, W.L., Lacey, S.F., Torigian, D.A.,
and IL-15. Blood 123, 3750–3759.
Soulen, M.C., Tian, L., McGarvey, M., Nelson, A.M., et al. (2019). Phase I study
of lentiviral-transduced chimeric antigen receptor-modified T cells recognizing 109. Hinrichs, C.S., Spolski, R., Paulos, C.M., Gattinoni, L., Kerstann, K.W., Palmer, D.C.,
mesothelin in advanced solid cancers. Mol. Ther. 27, 1919–1929. Klebanoff, C.A., Rosenberg, S.A., Leonard, W.J., and Restifo, N.P. (2008). IL-2 and
IL-21 confer opposing differentiation programs to CD8+ T cells for adoptive immu-
92. You, F., Jiang, L., Zhang, B., Lu, Q., Zhou, Q., Liao, X., Wu, H., Du, K., Zhu, Y., notherapy. Blood 111, 5326–5333.
Meng, H., et al. (2016). Phase 1 clinical trial demonstrated that MUC1 positive met-
astatic seminal vesicle cancer can be effectively eradicated by modified Anti-MUC1 110. Li, Y., Bleakley, M., and Yee, C. (2005). IL-21 influences the frequency, phenotype,
chimeric antigen receptor transduced T cells. Sci. China Life Sci. 59, 386–397. and affinity of the antigen-specific CD8 T cell response. J. Immunol. 175, 2261–2269.
111. Muralidharan, S., Hanley, P.J., Liu, E., Chakraborty, R., Bollard, C., Shpall, E.,
93. Brentjens, R.J., Rivière, I., Park, J.H., Davila, M.L., Wang, X., Stefanski, J., Taylor, C.,
Rooney, C., Savoldo, B., Rodgers, J., and Dotti, G. (2011). Activation of Wnt
Yeh, R., Bartido, S., Borquez-Ojeda, O., et al. (2011). Safety and persistence of adop-
signaling arrests effector differentiation in human peripheral and cord blood-
tively transferred autologous CD19-targeted T cells in patients with relapsed or
derived T lymphocytes. J. Immunol. 187, 5221–5232.
chemotherapy refractory B-cell leukemias. Blood 118, 4817–4828.
112. Zheng, W., O’Hear, C.E., Alli, R., Basham, J.H., Abdelsamed, H.A., Palmer, L.E.,
94. Andersch, L., Radke, J., Klaus, A., Schwiebert, S., Winkler, A., Schumann, E.,
Jones, L.L., Youngblood, B., and Geiger, T.L. (2018). PI3K orchestration of the
Grunewald, L., Zirngibl, F., Flemmig, C., Jensen, M.C., et al. (2019). CD171- and
in vivo persistence of chimeric antigen receptor-modified T cells. Leukemia 32,
GD2-specific CAR-T cells potently target retinoblastoma cells in preclinical
1157–1167.
in vitro testing. BMC Cancer 19, 895.
113. Fraietta, J.A., Lacey, S.F., Orlando, E.J., Pruteanu-Malinici, I., Gohil, M., Lundh, S.,
95. Kaech, S.M., Wherry, E.J., and Ahmed, R. (2002). Effector and memory T-cell dif-
Boesteanu, A.C., Wang, Y., O’Connor, R.S., Hwang, W.T., et al. (2018).
ferentiation: implications for vaccine development. Nat. Rev. Immunol. 2, 251–262.
Determinants of response and resistance to CD19 chimeric antigen receptor
96. Danilo, M., Chennupati, V., Silva, J.G., Siegert, S., and Held, W. (2018). Suppression (CAR) T cell therapy of chronic lymphocytic leukemia. Nat. Med. 24, 563–571.
of Tcf1 by inflammatory cytokines facilitates effector CD8 T cell differentiation. Cell 114. Finney, O.C., Brakke, H.M., Rawlings-Rhea, S., Hicks, R., Doolittle, D., Lopez, M.,
Rep. 22, 2107–2117. Futrell, R.B., Orentas, R.J., Li, D., Gardner, R.A., and Jensen, M.C. (2019). CD19
97. Kim, E.H., Sullivan, J.A., Plisch, E.H., Tejera, M.M., Jatzek, A., Choi, K.Y., and CAR T cell product and disease attributes predict leukemia remission durability.
Suresh, M. (2012). Signal integration by Akt regulates CD8 T cell effector and mem- J. Clin. Invest. 129, 2123–2132.
ory differentiation. J. Immunol. 188, 4305–4314. 115. Sheih, A., Voillet, V., Hanafi, L.A., DeBerg, H.A., Yajima, M., Hawkins, R., Gersuk,
98. Rutishauser, R.L., Martins, G.A., Kalachikov, S., Chandele, A., Parish, I.A., Meffre, V., Riddell, S.R., Maloney, D.G., Wohlfahrt, M.E., et al. (2020). Clonal kinetics and
E., Jacob, J., Calame, K., and Kaech, S.M. (2009). Transcriptional repressor single-cell transcriptional profiling of CAR-T cells in patients undergoing CD19
Blimp-1 promotes CD8+ T cell terminal differentiation and represses the acquisition CAR-T immunotherapy. Nat. Commun. 11, 219.
of central memory T cell properties. Immunity 31, 296–308. 116. Nobles, C.L., Sherrill-Mix, S., Everett, J.K., Reddy, S., Fraietta, J.A., Porter, D.L., Frey,
99. Joshi, N.S., Cui, W., Chandele, A., Lee, H.K., Urso, D.R., Hagman, J., Gapin, L., and N., Gill, S.I., Grupp, S.A., Maude, S.L., et al. (2020). CD19-targeting CAR T cell
Kaech, S.M. (2007). Inflammation directs memory precursor and short-lived immunotherapy outcomes correlate with genomic modification by vector integra-
effector CD8+ T cell fates via the graded expression of T-bet transcription factor. tion. J. Clin. Invest. 130, 673–685.
Immunity 27, 281–295. 117. Cornetta, K., Duffy, L., Turtle, C.J., Jensen, M., Forman, S., Binder-Scholl, G., Fry, T.,
100. Alfei, F., Kanev, K., Hofmann, M., Wu, M., Ghoneim, H.E., Roelli, P., Utzschneider, Chew, A., Maloney, D.G., and June, C.H. (2018). Absence of replication-competent
D.T., von Hoesslin, M., Cullen, J.G., Fan, Y., et al. (2019). TOX reinforces the pheno- lentivirus in the clinic: analysis of infused T cell products. Mol. Ther. 26, 280–288.
type and longevity of exhausted T cells in chronic viral infection. Nature 571, 118. Marcucci, K.T., Jadlowsky, J.K., Hwang, W.T., Suhoski-Davis, M., Gonzalez, V.E.,
265–269. Kulikovskaya, I., Gupta, M., Lacey, S.F., Plesa, G., Chew, A., et al. (2018).

Molecular Therapy Vol. 28 No 11 November 2020 15


Please cite this article in press as: Wagner et al., CAR T Cell Therapy for Solid Tumors: Bright Future or Dark Reality?, Molecular Therapy (2020), https://
doi.org/10.1016/j.ymthe.2020.09.015

www.moleculartherapy.org

Review

Retroviral and lentiviral safety analysis of gene-modified T cell products and infused 135. Reilly, E.B., Phillips, A.C., Buchanan, F.G., Kingsbury, G., Zhang, Y., Meulbroek,
HIV and oncology patients. Mol. Ther. 26, 269–279. J.A., Cole, T.B., DeVries, P.J., Falls, H.D., Beam, C., et al. (2015). Characterization
of ABT-806, a humanized tumor-specific anti-EGFR monoclonal antibody. Mol.
119. Bear, A.S., Morgan, R.A., Cornetta, K., June, C.H., Binder-Scholl, G., Dudley, M.E.,
Cancer Ther. 14, 1141–1151.
Feldman, S.A., Rosenberg, S.A., Shurtleff, S.A., Rooney, C.M., et al. (2012).
Replication-competent retroviruses in gene-modified T cells used in clinical trials: 136. Panousis, C., Rayzman, V.M., Johns, T.G., Renner, C., Liu, Z., Cartwright, G., Lee,
is it time to revise the testing requirements? Mol. Ther. 20, 246–249. F.T., Wang, D., Gan, H., Cao, D., et al. (2005). Engineering and characterisation
of chimeric monoclonal antibody 806 (ch806) for targeted immunotherapy of tu-
120. Singh, H., Huls, H., Kebriaei, P., and Cooper, L.J. (2014). A new approach to gene
mours expressing de2-7 EGFR or amplified EGFR. Br. J. Cancer 92, 1069–1077.
therapy using Sleeping Beauty to genetically modify clinical-grade T cells to target
CD19. Immunol. Rev. 257, 181–190. 137. Wang, Z., and Cao, Y.J. (2020). Adoptive cell therapy targeting neoantigens: a fron-
tier for cancer research. Front. Immunol. 11, 176.
121. Morita, D., Nishio, N., Saito, S., Tanaka, M., Kawashima, N., Okuno, Y., Suzuki, S.,
Matsuda, K., Maeda, Y., Wilson, M.H., et al. (2017). Enhanced expression of anti- 138. Abbott, R.C., Cross, R.S., and Jenkins, M.R. (2020). Finding the keys to the CAR:
CD19 chimeric antigen receptor in piggyBac transposon-engineered T cells. Mol. identifying novel target antigens for T cell redirection immunotherapies. Int. J.
Ther. Methods Clin. Dev. 8, 131–140. Mol. Sci. 21, 515.

122. Bailey, S.R., and Maus, M.V. (2019). Gene editing for immune cell therapies. Nat. 139. Orentas, R.J., Yang, J.J., Wen, X., Wei, J.S., Mackall, C.L., and Khan, J. (2012).
Biotechnol. 37, 1425–1434. Identification of cell surface proteins as potential immunotherapy targets in 12 pe-
diatric cancers. Front. Oncol. 2, 194.
123. Roth, T.L., Puig-Saus, C., Yu, R., Shifrut, E., Carnevale, J., Li, P.J., Hiatt, J., Saco, J.,
Krystofinski, P., Li, H., et al. (2018). Reprogramming human T cell function and 140. Bosse, K.R., Raman, P., Zhu, Z., Lane, M., Martinez, D., Heitzeneder, S., Rathi, K.S.,
specificity with non-viral genome targeting. Nature 559, 405–409. Kendsersky, N.M., Randall, M., Donovan, L., et al. (2017). Identification of GPC2 as
an oncoprotein and candidate immunotherapeutic target in high-risk neuroblas-
124. Hale, M., Lee, B., Honaker, Y., Leung, W.H., Grier, A.E., Jacobs, H.M., Sommer, K., toma. Cancer Cell 32, 295–309.e12.
Sahni, J., Jackson, S.W., Scharenberg, A.M., et al. (2017). Homology-directed recom-
141. Liu, X., Jiang, S., Fang, C., Yang, S., Olalere, D., Pequignot, E.C., Cogdill, A.P., Li, N.,
bination for enhanced engineering of chimeric antigen receptor T cells. Mol. Ther.
Ramones, M., Granda, B., et al. (2015). Affinity-tuned ErbB2 or EGFR chimeric an-
Methods Clin. Dev. 4, 192–203.
tigen receptor T cells exhibit an increased therapeutic index against tumors in mice.
125. Eyquem, J., Mansilla-Soto, J., Giavridis, T., van der Stegen, S.J., Hamieh, M., Cancer Res. 75, 3596–3607.
Cunanan, K.M., Odak, A., Gönen, M., and Sadelain, M. (2017). Targeting a CAR
142. Richman, S.A., and Milone, M.C. (2018). Neurotoxicity associated with a high-affin-
to the TRAC locus with CRISPR/Cas9 enhances tumour rejection. Nature 543,
ity GD2 CAR-response. Cancer Immunol. Res. 6, 496–497.
113–117.
143. Priceman, S.J., Gerdts, E.A., Tilakawardane, D., Kennewick, K.T., Murad, J.P., Park,
126. Bowers, J.S., Nelson, M.H., Majchrzak, K., Bailey, S.R., Rohrer, B., Kaiser, A.D.,
A.K., Jeang, B., Yamaguchi, Y., Yang, X., Urak, R., et al. (2017). Co-stimulatory
Atkinson, C., Gattinoni, L., and Paulos, C.M. (2017). Th17 cells are refractory to
signaling determines tumor antigen sensitivity and persistence of CAR T cells target-
senescence and retain robust antitumor activity after long-term ex vivo expansion.
ing PSCA+ metastatic prostate cancer. OncoImmunology 7, e1380764.
JCI Insight 2, e90772.
144. Watanabe, N., Bajgain, P., Sukumaran, S., Ansari, S., Heslop, H.E., Rooney, C.M.,
127. Klebanoff, C.A., Gattinoni, L., and Restifo, N.P. (2012). Sorting through subsets:
Brenner, M.K., Leen, A.M., and Vera, J.F. (2016). Fine-tuning the CAR spacer im-
which T-cell populations mediate highly effective adoptive immunotherapy?
proves T-cell potency. OncoImmunology 5, e1253656.
J. Immunother. 35, 651–660.
145. Dagogo-Jack, I., and Shaw, A.T. (2018). Tumour heterogeneity and resistance to
128. Blaeschke, F., Stenger, D., Kaeuferle, T., Willier, S., Lotfi, R., Kaiser, A.D., cancer therapies. Nat. Rev. Clin. Oncol. 15, 81–94.
Assenmacher, M., Döring, M., Feucht, J., and Feuchtinger, T. (2018). Induction of
a central memory and stem cell memory phenotype in functionally active CD4+ 146. Ahmed, N., Salsman, V.S., Kew, Y., Shaffer, D., Powell, S., Zhang, Y.J., Grossman,
and CD8+ CAR T cells produced in an automated good manufacturing practice sys- R.G., Heslop, H.E., and Gottschalk, S. (2010). HER2-specific T cells target primary
tem for the treatment of CD19+ acute lymphoblastic leukemia. Cancer Immunol. glioblastoma stem cells and induce regression of autologous experimental tumors.
Immunother. 67, 1053–1066. Clin. Cancer Res. 16, 474–485.

129. Schmueck-Henneresse, M., Omer, B., Shum, T., Tashiro, H., Mamonkin, M., 147. Vora, P., Venugopal, C., Salim, S.K., Tatari, N., Bakhshinyan, D., Singh, M., Seyfrid,
Lapteva, N., Sharma, S., Rollins, L., Dotti, G., Reinke, P., et al. (2017). M., Upreti, D., Rentas, S., Wong, N., et al. (2020). The rational development of
Comprehensive approach for identifying the T cell subset origin of CD3 and CD133-targeting immunotherapies for glioblastoma. Cell Stem Cell 26, 832–844.e6.
CD28 antibody-activated chimeric antigen receptor-modified T cells. J. Immunol. 148. Alhabbab, R.Y. (2020). Targeting cancer stem cells by genetically engineered
199, 348–362. chimeric antigen receptor T cells. Front. Genet. 11, 312.
130. Guedan, S., Posey, A.D., Jr., Shaw, C., Wing, A., Da, T., Patel, P.R., McGettigan, S.E., 149. Velasquez, M.P., Bonifant, C.L., and Gottschalk, S. (2018). Redirecting T cells to he-
Casado-Medrano, V., Kawalekar, O.U., Uribe-Herranz, M., et al. (2018). Enhancing matological malignancies with bispecific antibodies. Blood 131, 30–38.
CAR T cell persistence through ICOS and 4-1BB costimulation. JCI Insight 3, 150. Qin, H., Ramakrishna, S., Nguyen, S., Fountaine, T.J., Ponduri, A., Stetler-
e96976. Stevenson, M., Yuan, C.M., Haso, W., Shern, J.F., Shah, N.N., and Fry, T.J.
131. Costa Del Amo, P., Lahoz-Beneytez, J., Boelen, L., Ahmed, R., Miners, K.L., Zhang, (2018). Preclinical development of bivalent chimeric antigen receptors targeting
Y., Roger, L., Jones, R.E., Marraco, S.A.F., Speiser, D.E., et al. (2018). Human TSCM both CD19 and CD22. Mol. Ther. Oncolytics 11, 127–137.
cell dynamics in vivo are compatible with long-lived immunological memory and 151. Hegde, M., Mukherjee, M., Grada, Z., Pignata, A., Landi, D., Navai, S.A., Wakefield,
stemness. PLoS Biol. 16, e2005523. A., Fousek, K., Bielamowicz, K., Chow, K.K., et al. (2019). Tandem CAR T cells tar-
132. Das, R.K., Vernau, L., Grupp, S.A., and Barrett, D.M. (2019). Naïve T-cell deficits at geting HER2 and IL13Ra2 mitigate tumor antigen escape. J. Clin. Invest. 129, 3464.
diagnosis and after chemotherapy impair cell therapy potential in pediatric cancers. 152. Bielamowicz, K., Fousek, K., Byrd, T.T., Samaha, H., Mukherjee, M., Aware, N., Wu,
Cancer Discov. 9, 492–499. M.F., Orange, J.S., Sumazin, P., Man, T.K., et al. (2018). Trivalent CAR T cells over-
133. Rozenbaum, M., Meir, A., Aharony, Y., Itzhaki, O., Schachter, J., Bank, I., Jacoby, E., come interpatient antigenic variability in glioblastoma. Neuro-oncol. 20, 506–518.
and Besser, M.J. (2020). Gamma-delta CAR-T cells show CAR-directed and inde- 153. Urbanska, K., Lanitis, E., Poussin, M., Lynn, R.C., Gavin, B.P., Kelderman, S., Yu, J.,
pendent activity against leukemia. Front. Immunol. 11, 1347. Scholler, N., and Powell, D.J., Jr. (2012). A universal strategy for adoptive immuno-
134. Heczey, A., Liu, D., Tian, G., Courtney, A.N., Wei, J., Marinova, E., Gao, X., Guo, L., therapy of cancer through use of a novel T-cell antigen receptor. Cancer Res. 72,
Yvon, E., Hicks, J., et al. (2014). Invariant NKT cells with chimeric antigen receptor 1844–1852.
provide a novel platform for safe and effective cancer immunotherapy. Blood 124, 154. Liu, K., Liu, X., Peng, Z., Sun, H., Zhang, M., Zhang, J., Liu, S., Hao, L., Lu, G., Zheng,
2824–2833. K., et al. (2015). Retargeted human avidin-CAR T cells for adoptive immunotherapy

16 Molecular Therapy Vol. 28 No 11 November 2020


Please cite this article in press as: Wagner et al., CAR T Cell Therapy for Solid Tumors: Bright Future or Dark Reality?, Molecular Therapy (2020), https://
doi.org/10.1016/j.ymthe.2020.09.015

www.moleculartherapy.org

Review

of EGFRvIII expressing gliomas and their evaluation via optical imaging. Oncotarget 172. Srivastava, S., Salter, A.I., Liggitt, D., Yechan-Gunja, S., Sarvothama, M., Cooper, K.,
6, 23735–23747. Smythe, K.S., Dudakov, J.A., Pierce, R.H., Rader, C., and Riddell, S.R. (2019). Logic-
gated ROR1 chimeric antigen receptor expression rescues T cell-mediated toxicity to
155. Lohmueller, J.J., Ham, J.D., Kvorjak, M., and Finn, O.J. (2017). mSA2 affinity-
normal tissues and enables selective tumor targeting. Cancer Cell 35, 489–503.e8.
enhanced biotin-binding CAR T cells for universal tumor targeting.
OncoImmunology 7, e1368604. 173. Kloss, C.C., Condomines, M., Cartellieri, M., Bachmann, M., and Sadelain, M.
(2013). Combinatorial antigen recognition with balanced signaling promotes selec-
156. Rataj, F., Jacobi, S.J., Stoiber, S., Asang, F., Ogonek, J., Tokarew, N., Cadilha, B.L., van
tive tumor eradication by engineered T cells. Nat. Biotechnol. 31, 71–75.
Puijenbroek, E., Heise, C., Duewell, P., et al. (2019). High-affinity CD16-polymor-
phism and Fc-engineered antibodies enable activity of CD16-chimeric antigen re- 174. Wilkie, S., van Schalkwyk, M.C., Hobbs, S., Davies, D.M., van der Stegen, S.J.,
ceptor-modified T cells for cancer therapy. Br. J. Cancer 120, 79–87. Pereira, A.C., Burbridge, S.E., Box, C., Eccles, S.A., and Maher, J. (2012). Dual tar-
geting of ErbB2 and MUC1 in breast cancer using chimeric antigen receptors engi-
157. Liu, D., Zhao, J., and Song, Y. (2019). Engineering switchable and programmable
neered to provide complementary signaling. J. Clin. Immunol. 32, 1059–1070.
universal CARs for CAR T therapy. J. Hematol. Oncol. 12, 69.
175. Fedorov, V.D., Themeli, M., and Sadelain, M. (2013). PD-1- and CTLA-4-based
158. Rodgers, D.T., Mazagova, M., Hampton, E.N., Cao, Y., Ramadoss, N.S., Hardy, I.R.,
inhibitory chimeric antigen receptors (iCARs) divert off-target immunotherapy re-
Schulman, A., Du, J., Wang, F., Singer, O., et al. (2016). Switch-mediated activation
sponses. Sci. Transl. Med. 5, 215ra172.
and retargeting of CAR-T cells for B-cell malignancies. Proc. Natl. Acad. Sci. USA
113, E459–E468. 176. Han, X., Bryson, P.D., Zhao, Y., Cinay, G.E., Li, S., Guo, Y., Siriwon, N., and Wang,
P. (2017). Masked chimeric antigen receptor for tumor-specific activation. Mol.
159. Landgraf, K.E., Williams, S.R., Steiger, D., Gebhart, D., Lok, S., Martin, D.W.,
Ther. 25, 274–284.
Roybal, K.T., and Kim, K.C. (2020). convertibleCARs: a chimeric antigen receptor
system for flexible control of activity and antigen targeting. Commun. Biol. 3, 296. 177. Bonini, C., Ferrari, G., Verzeletti, S., Servida, P., Zappone, E., Ruggieri, L., Ponzoni,
M., Rossini, S., Mavilio, F., Traversari, C., and Bordignon, C. (1997). HSV-TK gene
160. Iwahori, K., Kakarla, S., Velasquez, M.P., Yu, F., Yi, Z., Gerken, C., Song, X.T., and
transfer into donor lymphocytes for control of allogeneic graft-versus-leukemia.
Gottschalk, S. (2015). Engager T cells: a new class of antigen-specific T cells that
Science 276, 1719–1724.
redirect bystander T cells. Mol. Ther. 23, 171–178.
178. Jones, B.S., Lamb, L.S., Goldman, F., and Di Stasi, A. (2014). Improving the safety of
161. Liu, X., Barrett, D.M., Jiang, S., Fang, C., Kalos, M., Grupp, S.A., June, C.H., and
cell therapy products by suicide gene transfer. Front. Pharmacol. 5, 254.
Zhao, Y. (2016). Improved anti-leukemia activities of adoptively transferred
T cells expressing bispecific T-cell engager in mice. Blood Cancer J. 6, e430. 179. Di Stasi, A., Tey, S.K., Dotti, G., Fujita, Y., Kennedy-Nasser, A., Martinez, C.,
Straathof, K., Liu, E., Durett, A.G., Grilley, B., et al. (2011). Inducible apoptosis as
162. Choi, B.D., Yu, X., Castano, A.P., Bouffard, A.A., Schmidts, A., Larson, R.C., Bailey,
a safety switch for adoptive cell therapy. N. Engl. J. Med. 365, 1673–1683.
S.R., Boroughs, A.C., Frigault, M.J., Leick, M.B., et al. (2019). CAR-T cells secreting
BiTEs circumvent antigen escape without detectable toxicity. Nat. Biotechnol. 37, 180. Straathof, K.C., Pulè, M.A., Yotnda, P., Dotti, G., Vanin, E.F., Brenner, M.K., Heslop,
1049–1058. H.E., Spencer, D.M., and Rooney, C.M. (2005). An inducible caspase 9 safety switch
for T-cell therapy. Blood 105, 4247–4254.
163. Avanzi, M.P., Yeku, O., Li, X., Wijewarnasuriya, D.P., van Leeuwen, D.G., Cheung,
K., Park, H., Purdon, T.J., Daniyan, A.F., Spitzer, M.H., and Brentjens, R.J. (2018). 181. Giordano-Attianese, G., Gainza, P., Gray-Gaillard, E., Cribioli, E., Shui, S., Kim, S.,
Engineered tumor-targeted T cells mediate enhanced anti-tumor efficacy both Kwak, M.J., Vollers, S., Corria Osorio, A.J., Reichenbach, P., et al. (2020). A compu-
directly and through activation of the endogenous immune system. Cell Rep. 23, tationally designed chimeric antigen receptor provides a small-molecule safety
2130–2141. switch for T-cell therapy. Nat. Biotechnol. 38, 426–432.
164. Li, X., Daniyan, A.F., Lopez, A.V., Purdon, T.J., and Brentjens, R.J. (2020). Cytokine 182. Zajc, C.U., Dobersberger, M., Schaffner, I., Mlynek, G., Pühringer, D., Salzer, B.,
IL-36g improves CAR T-cell functionality and induces endogenous antitumor Djinovic-Carugo, K., Steinberger, P., De Sousa Linhares, A., Yang, N.J., et al.
response. Leukemia. , Published online May 23, 2020. https://doi.org/10.1038/ (2020). A conformation-specific ON-switch for controlling CAR T cells with an
s41375-020-0874-1. orally available drug. Proc. Natl. Acad. Sci. USA 117, 14926–14935.
165. Kuhn, N.F., Purdon, T.J., van Leeuwen, D.G., Lopez, A.V., Curran, K.J., Daniyan, 183. Juillerat, A., Tkach, D., Busser, B.W., Temburni, S., Valton, J., Duclert, A., Poirot, L.,
A.F., and Brentjens, R.J. (2019). CD40 ligand-modified chimeric antigen receptor Depil, S., and Duchateau, P. (2019). Modulation of chimeric antigen receptor surface
T cells enhance antitumor function by eliciting an endogenous antitumor response. expression by a small molecule switch. BMC Biotechnol. 19, 44.
Cancer Cell 35, 473–488.e6.
184. Griffioen, M., van Egmond, E.H., Kester, M.G., Willemze, R., Falkenburg, J.H., and
166. Lai, J., Mardiana, S., House, I.G., Sek, K., Henderson, M.A., Giuffrida, L., Chen, Heemskerk, M.H. (2009). Retroviral transfer of human CD20 as a suicide gene for
A.X.Y., Todd, K.L., Petley, E.V., Chan, J.D., et al. (2020). Adoptive cellular therapy adoptive T-cell therapy. Haematologica 94, 1316–1320.
with T cells expressing the dendritic cell growth factor Flt3L drives epitope
185. Wang, X., Chang, W.C., Wong, C.W., Colcher, D., Sherman, M., Ostberg, J.R.,
spreading and antitumor immunity. Nat. Immunol. 21, 914–926.
Forman, S.J., Riddell, S.R., and Jensen, M.C. (2011). A transgene-encoded cell sur-
167. Sakemura, R., Terakura, S., Watanabe, K., Julamanee, J., Takagi, E., Miyao, K., face polypeptide for selection, in vivo tracking, and ablation of engineered cells.
Koyama, D., Goto, T., Hanajiri, R., Nishida, T., et al. (2016). A Tet-On inducible sys- Blood 118, 1255–1263.
tem for controlling CD19-chimeric antigen receptor expression upon drug admin-
186. Mestermann, K., Giavridis, T., Weber, J., Rydzek, J., Frenz, S., Nerreter, T., Mades,
istration. Cancer Immunol. Res. 4, 658–668.
A., Sadelain, M., Einsele, H., and Hudecek, M. (2019). The tyrosine kinase inhibitor
168. Juillerat, A., Marechal, A., Filhol, J.M., Valogne, Y., Valton, J., Duclert, A., dasatinib acts as a pharmacologic on/off switch for CAR T cells. Sci. Transl. Med. 11,
Duchateau, P., and Poirot, L. (2017). An oxygen sensitive self-decision making en- eaau5907.
gineered CAR T-cell. Sci. Rep. 7, 39833.
187. Blank, C.U., Haining, W.N., Held, W., Hogan, P.G., Kallies, A., Lugli, E., Lynn, R.C.,
169. Morsut, L., Roybal, K.T., Xiong, X., Gordley, R.M., Coyle, S.M., Thomson, M., and Philip, M., Rao, A., Restifo, N.P., et al. (2019). Defining “T cell exhaustion”. Nat. Rev.
Lim, W.A. (2016). Engineering customized cell sensing and response behaviors us- Immunol. 19, 665–674.
ing synthetic notch receptors. Cell 164, 780–791.
188. Kagoya, Y., Tanaka, S., Guo, T., Anczurowski, M., Wang, C.H., Saso, K., Butler,
170. Roybal, K.T., Rupp, L.J., Morsut, L., Walker, W.J., McNally, K.A., Park, J.S., and Lim, M.O., Minden, M.D., and Hirano, N. (2018). A novel chimeric antigen receptor con-
W.A. (2016). Precision tumor recognition by T cells with combinatorial antigen- taining a JAK-STAT signaling domain mediates superior antitumor effects. Nat.
sensing circuits. Cell 164, 770–779. Med. 24, 352–359.
171. Yang, Z.J., Yu, Z.Y., Cai, Y.M., Du, R.R., and Cai, L. (2020). Engineering of an 189. Lai, Y., Weng, J., Wei, X., Qin, L., Lai, P., Zhao, R., Jiang, Z., Li, B., Lin, S., Wang, S.,
enhanced synthetic Notch receptor by reducing ligand-independent activation. et al. (2018). Toll-like receptor 2 costimulation potentiates the antitumor efficacy of
Commun. Biol. 3, 116. CAR T cells. Leukemia 32, 801–808.

Molecular Therapy Vol. 28 No 11 November 2020 17


Please cite this article in press as: Wagner et al., CAR T Cell Therapy for Solid Tumors: Bright Future or Dark Reality?, Molecular Therapy (2020), https://
doi.org/10.1016/j.ymthe.2020.09.015

www.moleculartherapy.org

Review

190. Nguyen, P., Okeke, E., Clay, M., Haydar, D., Justice, J., O’Reilly, C., Pruett-Miller, S., Myb regulates CD8+ T cell stemness and antitumor immunity. Nat. Immunol. 20,
Papizan, J., Moore, J., Zhou, S., et al. (2020). Route of 41BB/41BBL costimulation 337–349.
determines effector function of B7-H3-CAR.CD28z T cells. Mol. Ther. Oncolytics
209. Seo, H., Chen, J., González-Avalos, E., Samaniego-Castruita, D., Das, A., Wang,
18, 202–214.
Y.H., López-Moyado, I.F., Georges, R.O., Zhang, W., Onodera, A., et al. (2019).
191. Foster, A.E., Mahendravada, A., Shinners, N.P., Chang, W.C., Crisostomo, J., Lu, A., TOX and TOX2 transcription factors cooperate with NR4A transcription factors
Khalil, M., Morschl, E., Shaw, J.L., Saha, S., et al. (2017). Regulated expansion and to impose CD8+ T cell exhaustion. Proc. Natl. Acad. Sci. USA 116, 12410–12415.
survival of chimeric antigen receptor-modified T cells using small molecule-depen-
210. Chen, J., López-Moyado, I.F., Seo, H., Lio, C.J., Hempleman, L.J., Sekiya, T.,
dent inducible MyD88/CD40. Mol. Ther. 25, 2176–2188.
Yoshimura, A., Scott-Browne, J.P., and Rao, A. (2019). NR4A transcription factors
192. Deguine, J., and Barton, G.M. (2014). MyD88: a central player in innate immune limit CAR T cell function in solid tumours. Nature 567, 530–534.
signaling. F1000Prime Rep. 6, 97.
211. Lynn, R.C., Weber, E.W., Sotillo, E., Gennert, D., Xu, P., Good, Z., Anbunathan, H.,
193. Horvath, C.M. (2004). The Jak-STAT pathway stimulated by interferon a or inter- Lattin, J., Jones, R., Tieu, V., et al. (2019). c-Jun overexpression in CAR T cells in-
feron b. Sci. STKE 2004, tr10. duces exhaustion resistance. Nature 576, 293–300.
194. Majoros, A., Platanitis, E., Kernbauer-Hölzl, E., Rosebrock, F., Müller, M., and 212. van der Woude, L.L., Gorris, M.A.J., Halilovic, A., Figdor, C.G., and de Vries, I.J.M.
Decker, T. (2017). Canonical and non-canonical aspects of JAK-STAT signaling: les- (2017). Migrating into the tumor: a roadmap for T cells. Trends Cancer 3, 797–808.
sons from interferons for cytokine responses. Front. Immunol. 8, 29.
213. Lanitis, E., Dangaj, D., Irving, M., and Coukos, G. (2017). Mechanisms regulating T-
195. Liau, N.P.D., Laktyushin, A., Lucet, I.S., Murphy, J.M., Yao, S., Whitlock, E., cell infiltration and activity in solid tumors. Ann. Oncol. 28 (suppl 12), xii18–xii32.
Callaghan, K., Nicola, N.A., Kershaw, N.J., and Babon, J.J. (2018). The molecular ba-
sis of JAK/STAT inhibition by SOCS1. Nat. Commun. 9, 1558. 214. Vilgelm, A.E., and Richmond, A. (2019). Chemokines modulate immune surveil-
lance in tumorigenesis, metastasis, and response to immunotherapy. Front.
196. Ma, X., Shou, P., Smith, C., Chen, Y., Du, H., Sun, C., Porterfield Kren, N., Michaud, Immunol. 10, 333.
D., Ahn, S., Vincent, B., et al. (2020). Interleukin-23 engineering improves CAR
T cell function in solid tumors. Nat. Biotechnol. 38, 448–459. 215. Mami-Chouaib, F., Blanc, C., Corgnac, S., Hans, S., Malenica, I., Granier, C., Tihy, I.,
and Tartour, E. (2018). Resident memory T cells, critical components in tumor
197. Liu, Y., Di, S., Shi, B., Zhang, H., Wang, Y., Wu, X., Luo, H., Wang, H., Li, Z., and immunology. J. Immunother. Cancer 6, 87.
Jiang, H. (2019). Armored inducible expression of IL-12 enhances antitumor activity
of glypican-3-targeted chimeric antigen receptor-engineered T cells in hepatocellu- 216. Peng, W., Ye, Y., Rabinovich, B.A., Liu, C., Lou, Y., Zhang, M., Whittington, M.,
lar carcinoma. J. Immunol. 203, 198–207. Yang, Y., Overwijk, W.W., Lizée, G., and Hwu, P. (2010). Transduction of tumor-
specific T cells with CXCR2 chemokine receptor improves migration to tumor
198. Pegram, H.J., Lee, J.C., Hayman, E.G., Imperato, G.H., Tedder, T.F., Sadelain, M., and antitumor immune responses. Clin. Cancer Res. 16, 5458–5468.
and Brentjens, R.J. (2012). Tumor-targeted T cells modified to secrete IL-12 eradi-
cate systemic tumors without need for prior conditioning. Blood 119, 4133–4141. 217. Craddock, J.A., Lu, A., Bear, A., Pule, M., Brenner, M.K., Rooney, C.M., and Foster,
A.E. (2010). Enhanced tumor trafficking of GD2 chimeric antigen receptor T cells by
199. Hurton, L.V., Singh, H., Najjar, A.M., Switzer, K.C., Mi, T., Maiti, S., Olivares, S., expression of the chemokine receptor CCR2b. J. Immunother. 33, 780–788.
Rabinovich, B., Huls, H., Forget, M.A., et al. (2016). Tethered IL-15 augments anti-
tumor activity and promotes a stem-cell memory subset in tumor-specific T cells. 218. Nishio, N., Diaconu, I., Liu, H., Cerullo, V., Caruana, I., Hoyos, V., Bouchier-Hayes,
Proc. Natl. Acad. Sci. USA 113, E7788–E7797. L., Savoldo, B., and Dotti, G. (2014). Armed oncolytic virus enhances immune func-
tions of chimeric antigen receptor-modified T cells in solid tumors. Cancer Res. 74,
200. Krenciute, G., Prinzing, B.L., Yi, Z., Wu, M.F., Liu, H., Dotti, G., Balyasnikova, I.V., 5195–5205.
and Gottschalk, S. (2017). Transgenic expression of IL15 improves antiglioma activ-
ity of IL13Ra2-CAR T cells but results in antigen loss variants. Cancer Immunol. 219. Iozzo, R.V. (2005). Basement membrane proteoglycans: from cellar to ceiling. Nat.
Res. 5, 571–581. Rev. Mol. Cell Biol. 6, 646–656.

201. Zhang, L., Morgan, R.A., Beane, J.D., Zheng, Z., Dudley, M.E., Kassim, S.H., Nahvi, 220. Iozzo, R.V., and Sanderson, R.D. (2011). Proteoglycans in cancer biology, tumour
A.V., Ngo, L.T., Sherry, R.M., Phan, G.Q., et al. (2015). Tumor-infiltrating lympho- microenvironment and angiogenesis. J. Cell. Mol. Med. 15, 1013–1031.
cytes genetically engineered with an inducible gene encoding interleukin-12 for the 221. Caruana, I., Savoldo, B., Hoyos, V., Weber, G., Liu, H., Kim, E.S., Ittmann, M.M.,
immunotherapy of metastatic melanoma. Clin. Cancer Res. 21, 2278–2288. Marchetti, D., and Dotti, G. (2015). Heparanase promotes tumor infiltration and
202. Renaud-Gabardos, E., Hantelys, F., Morfoisse, F., Chaufour, X., Garmy-Susini, B., antitumor activity of CAR-redirected T lymphocytes. Nat. Med. 21, 524–529.
and Prats, A.C. (2015). Internal ribosome entry site-based vectors for combined 222. Ramanathan, R.K., McDonough, S.L., Philip, P.A., Hingorani, S.R., Lacy, J.,
gene therapy. World J. Exp. Med. 5, 11–20. Kortmansky, J.S., Thumar, J., Chiorean, E.G., Shields, A.F., Behl, D., et al. (2019).
203. Hu, B., Ren, J., Luo, Y., Keith, B., Young, R.M., Scholler, J., Zhao, Y., and June, C.H. Phase IB/II randomized study of FOLFIRINOX plus pegylated recombinant human
(2017). Augmentation of antitumor immunity by human and mouse CAR T cells hyaluronidase versus FOLFIRINOX alone in patients with metastatic pancreatic
secreting IL-18. Cell Rep. 20, 3025–3033. adenocarcinoma: SWOG S1313. J. Clin. Oncol. 37, 1062–1069.

204. Chmielewski, M., and Abken, H. (2017). CAR T cells releasing IL-18 convert to T- 223. Fane, M., and Weeraratna, A.T. (2020). How the ageing microenvironment influ-
Bethigh FoxO1low effectors that exhibit augmented activity against advanced solid tu- ences tumour progression. Nat. Rev. Cancer 20, 89–106.
mors. Cell Rep. 21, 3205–3219. 224. Binnewies, M., Roberts, E.W., Kersten, K., Chan, V., Fearon, D.F., Merad, M.,
205. Zhou, P., Shaffer, D.R., Alvarez Arias, D.A., Nakazaki, Y., Pos, W., Torres, A.J., Coussens, L.M., Gabrilovich, D.I., Ostrand-Rosenberg, S., Hedrick, C.C., et al.
Cremasco, V., Dougan, S.K., Cowley, G.S., Elpek, K., et al. (2014). In vivo discovery (2018). Understanding the tumor immune microenvironment (TIME) for effective
of immunotherapy targets in the tumour microenvironment. Nature 506, 52–57. therapy. Nat. Med. 24, 541–550.
206. Shifrut, E., Carnevale, J., Tobin, V., Roth, T.L., Woo, J.M., Bui, C.T., Li, P.J., Diolaiti, 225. Duan, Q., Zhang, H., Zheng, J., and Zhang, L. (2020). Turning cold into hot: firing
M.E., Ashworth, A., and Marson, A. (2018). Genome-wide CRISPR screens in pri- up the tumor microenvironment. Trends Cancer 6, 605–618.
mary human T cells reveal key regulators of immune function. Cell 175, 1958–
226. Jarnicki, A.G., Lysaght, J., Todryk, S., and Mills, K.H. (2006). Suppression of anti-
1971.e15.
tumor immunity by IL-10 and TGF-b-producing T cells infiltrating the growing tu-
207. Wei, J., Long, L., Zheng, W., Dhungana, Y., Lim, S.A., Guy, C., Wang, Y., Wang, mor: influence of tumor environment on the induction of CD4+ and CD8+ regula-
Y.D., Qian, C., Xu, B., et al. (2019). Targeting REGNASE-1 programs long-lived tory T cells. J. Immunol. 177, 896–904.
effector T cells for cancer therapy. Nature 576, 471–476.
227. Jiang, X., Wang, J., Deng, X., Xiong, F., Ge, J., Xiang, B., Wu, X., Ma, J., Zhou, M., Li,
208. Gautam, S., Fioravanti, J., Zhu, W., Le Gall, J.B., Brohawn, P., Lacey, N.E., Hu, J., X., et al. (2019). Role of the tumor microenvironment in PD-L1/PD-1-mediated tu-
Hocker, J.D., Hawk, N.V., Kapoor, V., et al. (2019). The transcription factor c- mor immune escape. Mol. Cancer 18, 10.

18 Molecular Therapy Vol. 28 No 11 November 2020


Please cite this article in press as: Wagner et al., CAR T Cell Therapy for Solid Tumors: Bright Future or Dark Reality?, Molecular Therapy (2020), https://
doi.org/10.1016/j.ymthe.2020.09.015

www.moleculartherapy.org

Review

228. Geiger, R., Rieckmann, J.C., Wolf, T., Basso, C., Feng, Y., Fuhrer, T., Kogadeeva, M., 245. Chang, Z.L., Lorenzini, M.H., Chen, X., Tran, U., Bangayan, N.J., and Chen, Y.Y.
Picotti, P., Meissner, F., Mann, M., et al. (2016). L-arginine modulates T cell meta- (2018). Rewiring T-cell responses to soluble factors with chimeric antigen receptors.
bolism and enhances survival and anti-tumor activity. Cell 167, 829–842.e13. Nat. Chem. Biol. 14, 317–324.
229. Rivadeneira, D.B., and Delgoffe, G.M. (2018). Antitumor T-cell reconditioning: 246. Lo, A.S., Taylor, J.R., Farzaneh, F., Kemeny, D.M., Dibb, N.J., and Maher, J. (2008).
improving metabolic fitness for optimal cancer immunotherapy. Clin. Cancer Harnessing the tumour-derived cytokine, CSF-1, to co-stimulate T-cell growth and
Res. 24, 2473–2481. activation. Mol. Immunol. 45, 1276–1287.

230. Perna, S.K., De Angelis, B., Pagliara, D., Hasan, S.T., Zhang, L., Mahendravada, A., 247. DeBerardinis, R.J. (2020). Tumor microenvironment, metabolism, and immuno-
Heslop, H.E., Brenner, M.K., Rooney, C.M., Dotti, G., and Savoldo, B. (2013). therapy. N. Engl. J. Med. 382, 869–871.
Interleukin 15 provides relief to CTLs from regulatory T cell-mediated inhibition: 248. Huang, J., Yu, J., Tu, L., Huang, N., Li, H., and Luo, Y. (2019). Isocitrate dehydro-
implications for adoptive T cell-based therapies for lymphoma. Clin. Cancer Res. genase mutations in glioma: from basic discovery to therapeutics development.
19, 106–117. Front. Oncol. 9, 506.
231. John, L.B., Devaud, C., Duong, C.P., Yong, C.S., Beavis, P.A., Haynes, N.M., Chow, 249. Reiter-Brennan, C., Semmler, L., and Klein, A. (2018). The effects of 2-hydroxy-
M.T., Smyth, M.J., Kershaw, M.H., and Darcy, P.K. (2013). Anti-PD-1 antibody glutarate on the tumorigenesis of gliomas. Contemp. Oncol. (Pozn.) 22, 215–222.
therapy potently enhances the eradication of established tumors by gene-modified 250. Vodnala, S.K., Eil, R., Kishton, R.J., Sukumar, M., Yamamoto, T.N., Ha, N.H., Lee,
T cells. Clin. Cancer Res. 19, 5636–5646. P.H., Shin, M., Patel, S.J., Yu, Z., et al. (2019). T cell stemness and dysfunction in
232. Liu, X., Ranganathan, R., Jiang, S., Fang, C., Sun, J., Kim, S., Newick, K., Lo, A., June, tumors are triggered by a common mechanism. Science 363, eaau0135.
C.H., Zhao, Y., and Moon, E.K. (2016). A chimeric switch-receptor targeting PD1 251. Gelderman, K.A., Hultqvist, M., Holmberg, J., Olofsson, P., and Holmdahl, R.
augments the efficacy of second-generation CAR T cells in advanced solid tumors. (2006). T cell surface redox levels determine T cell reactivity and arthritis suscepti-
Cancer Res. 76, 1578–1590. bility. Proc. Natl. Acad. Sci. USA 103, 12831–12836.
233. Rupp, L.J., Schumann, K., Roybal, K.T., Gate, R.E., Ye, C.J., Lim, W.A., and Marson, 252. Cemerski, S., Cantagrel, A., Van Meerwijk, J.P., and Romagnoli, P. (2002). Reactive
A. (2017). CRISPR/Cas9-mediated PD-1 disruption enhances anti-tumor efficacy of oxygen species differentially affect T cell receptor-signaling pathways. J. Biol. Chem.
human chimeric antigen receptor T cells. Sci. Rep. 7, 737. 277, 19585–19593.
234. Hu, W., Zi, Z., Jin, Y., Li, G., Shao, K., Cai, Q., Ma, X., and Wei, F. (2019). CRISPR/ 253. Fultang, L., Booth, S., Yogev, O., Martins da Costa, B., Tubb, V., Panetti, S., Stavrou,
Cas9-mediated PD-1 disruption enhances human mesothelin-targeted CAR T cell V., Scarpa, U., Jankevics, A., Lloyd, G., et al. (2020). Metabolic engineering against
effector functions. Cancer Immunol. Immunother. 68, 365–377. the arginine microenvironment enhances CAR-T cell proliferation and therapeutic
activity. Blood 136, 1155–1160.
235. Nakazawa, T., Natsume, A., Nishimura, F., Morimoto, T., Matsuda, R., Nakamura,
M., Yamada, S., Nakagawa, I., Motoyama, Y., Park, Y.S., et al. (2020). Effect of 254. Oh, M.H., Sun, I.H., Zhao, L., Leone, R.D., Sun, I.M., Xu, W., Collins, S.L., Tam, A.J.,
CRISPR/Cas9-mediated PD-1-disrupted primary human third-generation CAR-T Blosser, R.L., Patel, C.H., et al. (2020). Targeting glutamine metabolism enhances tu-
cells targeting EGFRvIII on in vitro human glioblastoma cell growth. Cells 9, 998. mor-specific immunity by modulating suppressive myeloid cells. J. Clin. Invest. 130,
3865–3884.
236. Stadtmauer, E.A., Fraietta, J.A., Davis, M.M., Cohen, A.D., Weber, K.L., Lancaster,
255. Leone, R.D., Zhao, L., Englert, J.M., Sun, I.M., Oh, M.H., Sun, I.H., Arwood, M.L.,
E., Mangan, P.A., Kulikovskaya, I., Gupta, M., Chen, F., et al. (2020). CRISPR-engi-
Bettencourt, I.A., Patel, C.H., Wen, J., et al. (2019). Glutamine blockade induces
neered T cells in patients with refractory cancer. Science 367, eaba7365.
divergent metabolic programs to overcome tumor immune evasion. Science 366,
237. Condomines, M., Arnason, J., Benjamin, R., Gunset, G., Plotkin, J., and Sadelain, M. 1013–1021.
(2015). Tumor-targeted human t cells expressing CD28-based chimeric antigen re- 256. Ligtenberg, M.A., Mougiakakos, D., Mukhopadhyay, M., Witt, K., Lladser, A.,
ceptors circumvent CTLA-4 inhibition. PLoS ONE 10, e0130518. Chmielewski, M., Riet, T., Abken, H., and Kiessling, R. (2016). Coexpressed catalase
238. Dotti, G., Savoldo, B., Pule, M., Straathof, K.C., Biagi, E., Yvon, E., Vigouroux, S., protects chimeric antigen receptor-redirected T cells as well as bystander cells from
Brenner, M.K., and Rooney, C.M. (2005). Human cytotoxic T lymphocytes with oxidative stress-induced loss of antitumor activity. J. Immunol. 196, 759–766.
reduced sensitivity to Fas-induced apoptosis. Blood 105, 4677–4684. 257. Zannella, V.E., Dal Pra, A., Muaddi, H., McKee, T.D., Stapleton, S., Sykes, J.,
239. Yamamoto, T.N., Lee, P.H., Vodnala, S.K., Gurusamy, D., Kishton, R.J., Yu, Z., Glicksman, R., Chaib, S., Zamiara, P., Milosevic, M., et al. (2013).
Eidizadeh, A., Eil, R., Fioravanti, J., Gattinoni, L., et al. (2019). T cells genetically en- Reprogramming metabolism with metformin improves tumor oxygenation and
gineered to overcome death signaling enhance adoptive cancer immunotherapy. radiotherapy response. Clin. Cancer Res. 19, 6741–6750.
J. Clin. Invest. 129, 1551–1565. 258. Wang, L.C., Lo, A., Scholler, J., Sun, J., Majumdar, R.S., Kapoor, V., Antzis, M.,
Cotner, C.E., Johnson, L.A., Durham, A.C., et al. (2014). Targeting fibroblast activa-
240. Bollard, C.M., Rössig, C., Calonge, M.J., Huls, M.H., Wagner, H.J., Massague, J.,
tion protein in tumor stroma with chimeric antigen receptor T cells can inhibit tu-
Brenner, M.K., Heslop, H.E., and Rooney, C.M. (2002). Adapting a transforming
mor growth and augment host immunity without severe toxicity. Cancer Immunol.
growth factor beta-related tumor protection strategy to enhance antitumor immu-
Res. 2, 154–166.
nity. Blood 99, 3179–3187.
259. Kakarla, S., Chow, K.K., Mata, M., Shaffer, D.R., Song, X.T., Wu, M.F., Liu, H.,
241. Bollard, C.M., Tripic, T., Cruz, C.R., Dotti, G., Gottschalk, S., Torrano, V., Dakhova,
Wang, L.L., Rowley, D.R., Pfizenmaier, K., and Gottschalk, S. (2013). Antitumor ef-
O., Carrum, G., Ramos, C.A., Liu, H., et al. (2018). Tumor-specific T-cells engi-
fects of chimeric receptor engineered human T cells directed to tumor stroma. Mol.
neered to overcome tumor immune evasion induce clinical responses in patients
Ther. 21, 1611–1620.
with relapsed hodgkin lymphoma. J. Clin. Oncol. 36, 1128–1139.
260. Tran, E., Chinnasamy, D., Yu, Z., Morgan, R.A., Lee, C.C., Restifo, N.P., and
242. Wilkie, S., Burbridge, S.E., Chiapero-Stanke, L., Pereira, A.C., Cleary, S., van der Rosenberg, S.A. (2013). Immune targeting of fibroblast activation protein triggers
Stegen, S.J., Spicer, J.F., Davies, D.M., and Maher, J. (2010). Selective expansion of recognition of multipotent bone marrow stromal cells and cachexia. J. Exp. Med.
chimeric antigen receptor-targeted T-cells with potent effector function using inter- 210, 1125–1135.
leukin-4. J. Biol. Chem. 285, 25538–25544.
261. Curioni, A.B.C., Hiltbrunner, S., Bankel, L., Gulati, P., Weder, W., Opitz, I., Lauk, O.,
243. Wang, Y., Jiang, H., Luo, H., Sun, Y., Shi, B., Sun, R., and Li, Z. (2019). An IL-4/21 Caviezel, C., Knuth, A., Munz, C., et al. (2019). A phase I clinical trial of malignant
Inverted cytokine receptor improving CAR-T cell potency in immunosuppressive pleural mesothelioma treated with locally delivered autologous anti-FAP-targeted
solid-tumor microenvironment. Front. Immunol. 10, 1691. CAR T-cells. Ann. Oncol. 30 (Suppl 5 ), v501.
244. Leen, A.M., Sukumaran, S., Watanabe, N., Mohammed, S., Keirnan, J., Yanagisawa, 262. Tie, Y., Zheng, H., He, Z., Yang, J., Shao, B., Liu, L., Luo, M., Yuan, X., Liu, Y., Zhang,
R., Anurathapan, U., Rendon, D., Heslop, H.E., Rooney, C.M., et al. (2014). Reversal X., et al. (2020). Targeting folate receptor b positive tumor-associated macrophages
of tumor immune inhibition using a chimeric cytokine receptor. Mol. Ther. 22, in lung cancer with a folate-modified liposomal complex. Signal Transduct. Target.
1211–1220. Ther. 5, 6.

Molecular Therapy Vol. 28 No 11 November 2020 19


Please cite this article in press as: Wagner et al., CAR T Cell Therapy for Solid Tumors: Bright Future or Dark Reality?, Molecular Therapy (2020), https://
doi.org/10.1016/j.ymthe.2020.09.015

www.moleculartherapy.org

Review

263. Ruella, M., Klichinsky, M., Kenderian, S.S., Shestova, O., Ziober, A., Kraft, D.O., 275. Aalipour, A., Le Boeuf, F., Tang, M., Murty, S., Simonetta, F., Lozano, A.X., Shaffer,
Feldman, M., Wasik, M.A., June, C.H., and Gill, S. (2017). Overcoming the immu- T.M., Bell, J.C., and Gambhir, S.S. (2020). Viral delivery of CAR targets to solid tu-
nosuppressive tumor microenvironment of Hodgkin lymphoma using chimeric an- mors enables effective cell therapy. Mol. Ther. Oncolytics 17, 232–240.
tigen receptor T cells. Cancer Discov. 7, 1154–1167. 276. Power, A.T., and Bell, J.C. (2007). Cell-based delivery of oncolytic viruses: a new
264. Zhang, P., Zhao, S., Wu, C., Li, J., Li, Z., Wen, C., Hu, S., An, G., Meng, H., Zhang, X., strategic alliance for a biological strike against cancer. Mol. Ther. 15, 660–665.
and Yang, L. (2018). Effects of CSF1R-targeted chimeric antigen receptor-modified 277. Cole, C., Qiao, J., Kottke, T., Diaz, R.M., Ahmed, A., Sanchez-Perez, L., Brunn, G.,
NK92MI & T cells on tumor-associated macrophages. Immunotherapy 10, 935–949. Thompson, J., Chester, J., and Vile, R.G. (2005). Tumor-targeted, systemic delivery
265. Parihar, R., Rivas, C., Huynh, M., Omer, B., Lapteva, N., Metelitsa, L.S., Gottschalk, of therapeutic viral vectors using hitchhiking on antigen-specific T cells. Nat. Med.
S.M., and Rooney, C.M. (2019). NK cells expressing a chimeric activating receptor 11, 1073–1081.
eliminate MDSCs and rescue impaired CAR-T cell activity against solid tumors. 278. Donnelly, O., Vile, R., Pandha, H., Harrington, K., and Melcher, A. (2012). The
Cancer Immunol. Res. 7, 363–375. hitchhiker’s guide to virotherapy. Oncotarget 3, 735–736.
266. Chinnasamy, D., Yu, Z., Kerkar, S.P., Zhang, L., Morgan, R.A., Restifo, N.P., and 279. Grosser, R., Cherkassky, L., Chintala, N., and Adusumilli, P.S. (2019). Combination
Rosenberg, S.A. (2012). Local delivery of interleukin-12 using T cells targeting immunotherapy with CAR T cells and checkpoint blockade for the treatment of
VEGF receptor-2 eradicates multiple vascularized tumors in mice. Clin. Cancer solid tumors. Cancer Cell 36, 471–482.
Res. 18, 1672–1683.
280. Minn, I., Rowe, S.P., and Pomper, M.G. (2019). Enhancing CAR T-cell therapy
267. Xie, Y.J., Dougan, M., Jailkhani, N., Ingram, J., Fang, T., Kummer, L., Momin, N., through cellular imaging and radiotherapy. Lancet Oncol. 20, e443–e451.
Pishesha, N., Rickelt, S., Hynes, R.O., and Ploegh, H. (2019). Nanobody-based 281. Ma, L., Dichwalkar, T., Chang, J.Y.H., Cossette, B., Garafola, D., Zhang, A.Q.,
CAR T cells that target the tumor microenvironment inhibit the growth of solid tu- Fichter, M., Wang, C., Liang, S., Silva, M., et al. (2019). Enhanced CAR-T cell activity
mors in immunocompetent mice. Proc. Natl. Acad. Sci. USA 116, 7624–7631. against solid tumors by vaccine boosting through the chimeric receptor. Science 365,
268. Santoro, S.P., Kim, S., Motz, G.T., Alatzoglou, D., Li, C., Irving, M., Powell, D.J., Jr., 162–168.
and Coukos, G. (2015). T cells bearing a chimeric antigen receptor against prostate- 282. Reinhard, K., Rengstl, B., Oehm, P., Michel, K., Billmeier, A., Hayduk, N., Klein, O.,
specific membrane antigen mediate vascular disruption and result in tumor regres- Kuna, K., Ouchan, Y., Wöll, S., et al. (2020). An RNA vaccine drives expansion and
sion. Cancer Immunol. Res. 3, 68–84. efficacy of claudin-CAR-T cells against solid tumors. Science 367, 446–453.
269. Xu, J., Wang, Y., Shi, J., Liu, J., Li, Q., and Chen, L. (2018). Combination therapy: a 283. Ruella, M., Kenderian, S.S., Shestova, O., Fraietta, J.A., Qayyum, S., Zhang, Q., Maus,
feasibility strategy for CAR-T cell therapy in the treatment of solid tumors. Oncol. M.V., Liu, X., Nunez-Cruz, S., Klichinsky, M., et al. (2016). The addition of the BTK
Lett. 16, 2063–2070. inhibitor ibrutinib to anti-CD19 chimeric antigen receptor T cells (CART19) im-
270. Anderson, K.G., Stromnes, I.M., and Greenberg, P.D. (2017). Obstacles posed by the proves responses against mantle cell lymphoma. Clin. Cancer Res. 22, 2684–2696.
tumor microenvironment to t cell activity: a case for synergistic therapies. Cancer 284. Ramakrishna, S., Highfill, S.L., Walsh, Z., Nguyen, S.M., Lei, H., Shern, J.F., Qin, H.,
Cell 31, 311–325. Kraft, I.L., Stetler-Stevenson, M., Yuan, C.M., et al. (2019). Modulation of target an-
tigen density improves CAR T-cell functionality and persistence. Clin. Cancer Res.
271. Porter, C.E., Rosewell Shaw, A., Jung, Y., Yip, T., Castro, P.D., Sandulache, V.C.,
25, 5329–5341.
Sikora, A., Gottschalk, S., Ittman, M.M., Brenner, M.K., and Suzuki, M. (2020).
Oncolytic adenovirus armed with BiTE, cytokine, and checkpoint inhibitor enables 285. Michie, J., Beavis, P.A., Freeman, A.J., Vervoort, S.J., Ramsbottom, K.M.,
CAR T cells to control the growth of heterogeneous tumors. Mol. Ther. 28, 1251– Narasimhan, V., Lelliott, E.J., Lalaoui, N., Ramsay, R.G., Johnstone, R.W., et al.
1262. (2019). Antagonism of IAPs enhances CAR T-cell efficacy. Cancer Immunol. Res.
7, 183–192.
272. Wing, A., Fajardo, C.A., Posey, A.D., Jr., Shaw, C., Da, T., Young, R.M., Alemany, R.,
June, C.H., and Guedan, S. (2018). Improving CART-cell therapy of solid tumors 286. Ng, J., and Dai, T. (2016). Radiation therapy and the abscopal effect: a concept comes
with oncolytic virus-driven production of a bispecific T-cell engager. Cancer of age. Ann. Transl. Med. 4, 118.
Immunol. Res. 6, 605–616. 287. Zhang, Z., Zhou, L., Xie, N., Nice, E.C., Zhang, T., Cui, Y., and Huang, C. (2020).
273. Rosewell Shaw, A., Porter, C.E., Watanabe, N., Tanoue, K., Sikora, A., Gottschalk, S., Overcoming cancer therapeutic bottleneck by drug repurposing. Signal
Brenner, M.K., and Suzuki, M. (2017). Adenovirotherapy delivering cytokine and Transduct. Target. Ther. 5, 113.
checkpoint inhibitor augments CAR T cells against metastatic head and neck cancer. 288. Mottini, C., Napolitano, F., Li, Z., Gao, X., and Cardone, L. (2019). Computer-aided
Mol. Ther. 25, 2440–2451. drug repurposing for cancer therapy: approaches and opportunities to challenge
274. Park, A.K., Fong, Y., Kim, S.I., Yang, J., Murad, J.P., Lu, J., Jeang, B., Chang, W.C., anticancer targets. Semin. Cancer Biol. , Published online September 25, 2019.
Chen, N.G., Thomas, S.H., et al. (2020). Effective combination immunotherapy us- https://doi.org/10.1016/j.semcancer.2019.09.023.
ing oncolytic viruses to deliver CAR targets to solid tumors. Sci. Transl. Med. 12, 289. Rams, D. (2017). Ten principles for good design. In Ten Principles for Good Design:
eaaz1863. Dieter Rams, C.W. De Jong, ed. (Prestel), pp. 92–133.

20 Molecular Therapy Vol. 28 No 11 November 2020

You might also like