You are on page 1of 8

DERENZO, KRENCIUTE, AND GOTTSCHALK

The Landscape of CAR T Cells Beyond Acute Lymphoblas c


Leukemia for Pediatric Solid Tumors
Christopher DeRenzo, MD, MBA, Giedre Krenciute, PhD, and Stephen Go schalk, MD

OVERVIEW
Adop ve cell therapy with gene cally modified T cells holds the promise to improve outcomes for children with recurrent/
refractory solid tumors and has the poten al to reduce treatment complica ons for all pa ents. Although T cells that ex-
press chimeric an gen receptors (CARs) specific for CD19 have had remarkable success for B-cell–derived malignancies,
which has led to their approval by the U.S. Food and Drug Administra on, CAR T cells have been less effec ve for solid
tumors and brain tumors. Lack of efficacy is most likely mul factorial, but heterogeneous an gen expression; limited mi-
gra on of T cells to tumor sites; and the immunosuppressive, hos le tumor microenvironment have emerged as major
roadblocks that must be addressed. In this review, we summarize the clinical experience with CAR T-cell therapy for pe-
diatric solid tumors, including brain tumors. In addi on, we review strategies that have been and are being developed to
enhance their an tumor ac vity.

I mmunotherapy for pediatric malignancies holds the promises


of improving outcomes and reducing treatment-related
complica ons. Among different forms of immunotherapy that
expansion of adop vely transferred CD19–CAR T cells. Sec-
ond, CD19–CAR T cells can eradicate B-cell malignancies re-
gardless of their underlying gene c altera on. Third, CD19–
are ac vely being pursued, the adop ve transfer of T cells CAR T cells, can eradicate B-cell malignancies, which are
that express chimeric an gen receptors (CARs) has garnered refractory to chemotherapy and/or radia on, highligh ng
great excitement because of the success of CAR T-cell ther- that T cells kill their target cells through different cytotoxic
apy for CD19-posi ve malignancies.1-11 mechanisms than conven onal therapies. Fourth, CD19-
CARs are synthe c molecules that combine the specific- CARs have to encode a cos mulatory signaling domain
ity of monoclonal an bodies with the effector func on of to be effec ve. Although CD19–CAR.CD28.ζ and CD19–
T cells.12-14 The prototypic CAR consists of an an gen bind- CAR.41BB.ζ T cells have not been compared directly in a sin-
ing domain encoded by a single-chain variable fragment gle clinical study, results of published studies indicate that
derived from a monoclonal an body, a hinge and trans- CD19–CAR.41BB.ζ T cells persist longer.1-11 However, it re-
membrane domain, and signaling domains derived from mains unclear whether this translates into improved an tu-
the CD3.ζ chain as well as cos mulatory molecules, such mor ac vity.1-11 Fi h, targe ng a single an gen can result in
as CD28 and/or 41BB (Fig. 1A). The majority of CAR T-cell an gen loss variants. Last, CD19–CAR T-cell therapy can be
products are generated by viral transduc on that uses rep- associated with important clinical adverse effects, including
lica on incompetent retroviral or len viral vectors (Fig. 1B). cytokine release syndrome (CRS) and neurotoxicity.1-11
Since the submission of the first inves ga onal new drug In this educa onal review, results of early-phase clinical
applica on for CD19–CAR T cells, the field has moved rapidly studies with CAR T cells for pediatric solid tumors and brain
and culminated in 2017 with approval by the U.S. Food and tumors are summarized (Table 1). In addi on, strategies to
Drug Administra on (FDA) of two CD19–CAR T-cell prod- improve their an tumor ac vity and current challenges are
ucts.15 The interested reader is referred to recent reviews, discussed.
which summarize the clinical results of CD19–CAR T cells
in detail.1,2 Here, we highlight only the key findings, which CLINICAL STUDIES WITH CAR T CELLS FOR
must be considered as we develop and improve current CAR PEDIATRIC BRAIN AND SOLID TUMORS
T-cell therapy approaches for pediatric solid tumors. Neuroblastoma
First, lymphodeple ng chemotherapy with cyclophospha- Two clinical studies have been conducted with first-
mide and fludarabine is cri cal to allow engra ment and genera on CAR T cells in pa ents with neuroblastoma. In

From the Department of Bone Marrow Transplanta on and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN.

Disclosures of poten al conflicts of interest provided by the authors are available with the online ar cle at asco.org/edbook.

Corresponding author: Stephen Go schalk, MD, Department of Bone Marrow Transplanta on and Cellular Therapy, St Jude Children’s Research Hospital, 262 Danny Thomas Pl.,
MS321, Memphis, TN 38105; email: stephen.go schalk@stjude.org.

© 2018 American Society of Clinical Oncology

830 2018 ASCO EDUCATIONAL BOOK | asco.org/edbook


Downloaded from ascopubs.org by 41.68.253.21 on June 26, 2021 from 041.068.253.021
Copyright © 2021 American Society of Clinical Oncology. All rights reserved.
LANDSCAPE OF CAR T CELLS BEYOND ACUTE LYMPHOBLASTIC LEUKEMIA

FIGURE 1. Scheme of CAR T-Cell Genera on


A 1st Gen 2nd Gen B
Activation Viral Transduction Freeze
Cytokines

Cytokines

zeta costim
Day 0 Day 1-2 Day 9-10

zeta

(A) CARs consist of an ectodomain, hinge and transmembrane domain, and endodomain. CARs have been designed with or without cos mulatory (cos m) signaling domains. As examples, first- and second-
genera on (Gen) CARs are shown. (B) CAR T cells are produced by ac va ng T cells in the presence of cytokines. When T cells proliferate, they are transduced with viral vectors that encode CARs. CAR T cells
subsequently are expanded with cytokines, and sufficient CAR T cells for preclinical or clinical studies are generated within 9 to 10 days of culture ini a on.

the first study, pa ents received up to 109/m2 CD8 CD171- More recently, ac vated T cells that expressed a third-
specific CAR T-cell clones.16 Adop ve transfer of T cells was genera on GD2–CAR with a CD28.OX40.ζ endodomain were
well tolerated, but T cells persisted for only 6 weeks, and evaluated in pa ents with neuroblastoma.19 Three cohorts
only one of six pa ents had a par al response. One strat- of pa ents were infused. The first cohort (four pa ents) re-
egy to improve the persistence of adop vely transferred T ceived only CAR T cells; the second cohort (four pa ents) re-
cells takes advantage of the specificity of the endogenous ceived lymphodeple ng chemotherapy (cyclophosphamide
αβ T-cell receptor expressed by CAR T cells. For example, and fludarabine) before CAR T-cell infusion; and the third
virus-specific T cells receive appropriate s mula on by viral cohort (three pa ents) received two doses of pembroli-
an gens presented by professional an gen-presen ng cells. zumab in addi on to CAR T-cell infusion: on day −1 and day
This concept was explored in the second clinical study, in 21 rela ve to CAR T-cell infusion. Lymphodeple ng chemo-
which inves gators expressed a first-genera on disialogan- therapy induced expansion of CAR T cells for up to 3 logs,
glioside (GD2)-specific CAR in polyclonal Epstein Barr virus and expansion peaked at 1 to 2 weeks post infusion. Addi-
(EBV)–specific T cells. The in vivo fate of GD2–CAR EBV- on of pembrolizumab did not improve T-cell expansion or
specific T cells was directly compared with ac vated T cells persistence further. There was a large increase in the fre-
that expressed the same CAR in individual patients.17,18 quency of circula ng myeloid cells in the peripheral blood
Although GD2–CAR EBV-specific T cells did not expand, they with an immunosuppressive M2 phenotype a er CAR T-cell
persisted longer than GD2–CAR ac vated T cells. Five of 11 infusion in all three cohorts. Addi onal studies are needed
patients with active disease showed tumor responses or to inves gate the significance of this finding.
necrosis. Three of them had complete responses. In addi on to these published studies, several clinical
trials, including one study to evaluate second-genera on
(41BB.ζ) and third-genera on (CD28.41BB.ζ) CAR T cells that
PRACTICAL APPLICATIONS target CD17124 in pa ents with neuroblastoma and gangli-
oneuroblastoma (NCT02311621), are in progress. In addi on,
• CAR T cells for pediatric solid tumors are safe but have studies con nue to explore GD2–CAR T cells for neuroblas-
limited an tumor ac vity in early-phase clinical studies.
toma; these include NCT02107963, NCT02761915, and
• Carefully designed correla ve studies will be cri cal to
understand current failures of CAR T-cell therapies and
NCT03373097.
to devise strategies to improve them.
• Rou ne, noninvasive, clinical imaging of infused CAR Sarcoma
T cells would provide invaluable insight into their T cells that express second-genera on HER2–CARs (CD28.ζ)
biodistribu on in humans. have been evaluated in 19 pa ents with refractory HER2-
• Addi onal gene c modifica ons have improved the posi ve sarcoma (16 with osteosarcoma, one with Ewing
an tumor ac vity of CAR T cells in preclinical models, sarcoma, one with primi ve neuroectodermal tumor, and
but improved CAR T cells have not been tested in the one with desmoplas c small round cell tumor). HER2–CAR
clinic. T-cell infusions were well tolerated and had no dose-limi ng
• Costs and regulatory requirements associated with toxicity.20 HER2–CAR T cells persisted for at least 6 weeks in
clinical tes ng of CAR T cells have the poten al to
pa ents who received greater than 1 × 106/m2 HER2–CAR
impede progress.
T cells and were detected at tumor sites. Of 17 evaluable

asco.org/edbook | 2018 ASCO EDUCATIONAL BOOK  831


Downloaded from ascopubs.org by 41.68.253.21 on June 26, 2021 from 041.068.253.021
Copyright © 2021 American Society of Clinical Oncology. All rights reserved.
DERENZO, KRENCIUTE, AND GOTTSCHALK

TABLE 1. Selected Completed and Future CAR T-Cell Therapy Studies for Pediatric Solid Tumors or Brain
Tumors

CAR Signaling Cell Type and Lymph Comment, NCT No., or


Disease Target Domain Vector Delivery Deple on Reference (if published)
Neuroblastoma CD171 ζ Pl ATC, IV — Park et al16
41BB.ζ or
CD171 LV ATC, IV + NCT02311621
CD28.41BB.ζ
GD2 ζ RV VST or ATC, IV — Pule et al,17 Louis et al18
GD2 CD28.OX40.ζ RV ATC, IV ± Pembrolizumab, Heczey et al19
GD2 ζ, ND RV ATC, IV + NCT03373097
GD2 ζ, ND RV ATC, IV ± NCT02761915
Second transgene: IL-15,
GD2 ζ, ND RV iNKT, IV +
NCT03294954
Neuroblastoma and
GD2 CD28.OX40.ζ RV ATC, IV + NCT02107963
sarcoma
Sarcoma Ahmed et al (2015),20 Hegde
HER2 CD28.ζ RV ATC, IV ±
et al21
GD2 CD28.OX40.ζ RV VST, IV ± VZV vaccine, NCT02932956
Hepatoblastoma and
GPC3 41BB.ζ RV ATC, IV + NCT02932956
sarcoma
Brain tumors HER2 CD28.ζ RV VST, IV — Ahmed et al (2017)22
IL-13Rα2 41BB.ζ LV ATC, IC — NCT02208362, Brown et al23

Abbrevia ons: ATC, ac vated T cells; CAR, chimeric an gen receptor; IC, intracranial; IL, interleukin; IL-13R, IL-13 receptor; iNKT, invariant NK T cells; IV, intravenous; LV, len virus; ND, not disclosed;
Pl, plasmid; RV, retrovirus; VST, virus-specific T cells.

pa ents, four had stable disease. Three of these had their However, responses were observed; these included one
tumor removed; one showed 90% or greater necrosis. Sub- par al response22 and one complete response,23 and sev-
sequently, six pa ents received lymphodeple ng chemo- eral pa ents had stable disease for a prolonged period of
therapy before HER2–CAR T-cell infusion. HER2-CAR T-cell me.22
expansion was observed without apparent toxicity, and Detailed correla ve studies performed a er infusion
one pa ent with refractory rhabdomyosarcoma, who had of EGFRvIII–CAR T cells revealed that T cells were able to
persistent bone marrow disease only, achieved a complete migrate to glioma sites a er intravenous infusion.26 Target
response for greater than 12 months.21 In addi on to HER2– an gen expression was reduced in resected gliomas, which
CAR T cells, GD2–CAR T cells are ac vely being explored was indica ve of an on-target CAR T-cell effect. In addi on,
(NCT01953900, NCT02107963) for sarcoma. In one study, a gliomas upregulated the expression of immunosuppressive
third-genera on GD2–CAR with a CD28.OX40.ζ endodomain molecules, including indoleamine 2,3 dioxygenase and IL-
is expressed in varicella zoster virus–specific T cells, and 10, which highlighted the ability of gliomas to counteract
the inves gators are evalua ng whether GD2–CAR virus- infiltra ng pro-inflammatory CAR T cells. Currently, only one
specific T cells can be boosted with an FDA-approved var- clinical study (NCT02208362) is ac vely recrui ng pa ents,
icella zoster virus vaccine a er infusion. Another clinical including children older than age 12 years, with gliomas.
study (NCT02932956) is approved by the FDA to evaluate This study is evalua ng the safety and efficacy of intracranial
the safety and efficacy of GPC3-CAR T cells25 in pa ents with injec ons of IL-13Rα2–CAR.41BB.ζ T cells.
pediatric solid tumors, including rhabdomyosarcoma. How- The ini al foray into the clinic with CAR T cells has demon-
ever, the study currently is not open for accrual. strated their safety for pediatric solid tumors and brain
tumors. However, only subsets of pa ents have benefited
Brain Tumors from this approach so far. Poten al strategies to increase
Clinical studies with CAR T cells that target HER2, EGFRvIII, the efficacy of CAR T cells are reviewed in the next sec on.
and interleukin (IL)-13 receptor α2 (IL-13Rα2) have been
conducted in pa ents with high-grade glioma.22,23,26,27 Two Strategies to Improve CAR T-Cell Therapy for Solid
studies infused only adults, whereas 10 of 17 pa ents in the Tumors
HER2–CAR T-cell therapy study were children. T cells were Lack of CAR T-cell efficacy is most likely mul factorial. Major
given either intravenously (HER2, EGFRvIII) or directly in- roadblocks include the availability of targeted an gens and
jected into the tumor and/or ventricle (IL-13Rα2). Like the their heterogeneous expression, the homing of T cells to
CAR T-cell therapy studies for solid tumors, the majority of tumor cells, and the immunosuppressive tumor microenvi-
pa ents in the brain tumor studies had progressive disease. ronment (Fig. 2).

832 2018 ASCO EDUCATIONAL BOOK | asco.org/edbook


Downloaded from ascopubs.org by 41.68.253.21 on June 26, 2021 from 041.068.253.021
Copyright © 2021 American Society of Clinical Oncology. All rights reserved.
LANDSCAPE OF CAR T CELLS BEYOND ACUTE LYMPHOBLASTIC LEUKEMIA

Expanding the Repertoire of Targetable An gens enables the targe ng of an gens that are expressed in nor-
The majority of CARs developed so far recognize cell sur- mal ssues.37,38
face proteins (GD2, HER2, Glypican (GPC)3, and IL-13Rα2),
which originally were discovered as targets for monoclonal Enhancing Migra on of CAR T-Cells to Tumor Sites
an bodies. Efforts are underway with gene expression ar- and Within Tumors
ray data and proteomics to iden fy new targetable cell sur- Several preclinical studies have highlighted the mismatch
face an gens.28 The recent iden fica on of GPC2, which is between chemokines secreted by solid tumors and chemo-
expressed at high levels on neuroblastoma, highlights the kine receptors expressed by CAR T cells. Transgenic expres-
feasibility of this approach.29 However, it might be difficult sion of chemokine receptors has been shown to overcome
to discover an gens that are not expressed at low levels in this roadblock.39 For example, neuroblastoma secretes high
normal ssues. Although private neoan gens are present, levels of CCL2; however, CAR T cells lack expression of the
albeit at low frequency, in pediatric tumors,30 directly tar- corresponding chemokine receptor (i.e., CCR2).40 Trans-
ge ng these with CAR T cells is not feasible with current genic expression of CCR2b on GD2–CAR T cells resulted
technology that uses viral vectors to generate CAR T cells. in enhanced homing and an tumor ac vity in preclinical
In addi on to the large regulatory burden and cost, me is neuroblastoma models.40 Besides migra on to tumor sites,
a barrier: it takes more than 6 months to generate a clinical- limited migra on within tumors might contribute to the
grade viral vector. However, op miza on of CAR T cells to reduced an tumor ac vity observed in clinical studies. For
efficiently induce immune responses against nontargeted example, CAR T cells are limited in their ability to degrade
an gen (i.e., an gen spreading) would be one approach to the extracellular matrix, and this limita on results in poor
target private neoan gens and could increase the an tumor tumor penetra on. However, penetra on can be improved
ac vity of CAR T cells in a manner similar to that of cancer by expressing heparanase.41 Another approach consists of
vaccines.31 directly targe ng cancer-associated fibroblasts, the main
CAR T cells are being developed to recognize an gen producer of collagen within tumors, with CAR T cells.42
pa erns. Examples include designing CARs that recognize
two an gens or engineering T cells that express mul ple Engineering CAR T Cells to Resist the
CARs.32,33 T cells that express these CARs become fully ac - Immunosuppressive Tumor Environment
vated only in the presence of all targeted an gens. Target- Brain and solid tumors create a hos le tumor environment
ing mul ple an gens also should offset the risk of selec ng that favors T-cell exhaus on and/or dysfunc on induced by
an gen loss variants. In addi on, so-called inhibitory CARs immunosuppressive cytokines (e.g., IL-4, IL-10, transforming
that block poten al off-target T-cell responses have been growth factor β [TGFβ]); expression of inhibitory molecules
developed.34 Development of CARs that recognize pep des (e.g., first apoptosis signal receptor [FAS] ligand, PD-L1); the
in the context of major histocompa bility complex class I metabolic environment; and/or recruitment of immunosup-
molecules also might increase the poten al repertoire of pressive cells, including myeloid-derived suppressor cells,
targetable an gens, because two-thirds of all expressed cancer-associated fibroblasts, and/or regulatory T cells.43-45
proteins reside within the cell. This can be achieved with a Although this sec on is focused on engineering CAR T
single-chain variable fragment derived from a T-cell receptor cells to improve their an tumor ac vity in the tumor mi-
to mimic a monoclonal an body as a CAR an gen-binding croenvironment, combina on therapies also are a rac ve
domain. Examples include CARs specific for an HLA-A2– approaches. For example, combina on of CAR T-cell therapy
restricted pep de derived from intracellular proteins, such with checkpoint blockade, oncoly c viruses, chemotherapy,
as Wilms tumor 1 protein or proteinase 3.35,36 radia on, and/or small molecules is being explored in pre-
Inducible expression systems may provide a poten al clinical studies with encouraging results.46-48 Gene c engi-
solu on to the an gen dilemma. So-called synthe c notch neering approaches to enhance the an tumor ac vity of
signaling receptors allow the expression of CARs only af- CAR T cells can be divided into two broad categories: trans-
ter a T cell has migrated to tumor sites, which poten ally genic expression of immune s mulatory molecules and si-
lencing nega ve regulators.
Several preclinical studies have shown that transgenic ex-
FIGURE 2. Roadblocks of CAR T Cells for Solid pression of cytokines (e.g., IL-12, IL-15, IL-18),49-51 cons tu-
Tumors ve cytokine receptors,52 41BBL,53 or CD40L54 enhance the
Antigen &
an tumor ac vity of CAR T cells. A recent study also demon-
Heterogeneity strated that transgenic expression of IL-7 in combina on
with the chemokine CCL19 not only enhances the effector
func on of CAR T cells but also enables the cells to induce
CART endogenous T-cell responses against the targeted tumor,
Roadblocks
which is indica ve of an gen spreading.55
Direct blockade of inhibitory cytokines or conversion of
Tumor
environment Homing their signal into a T-cell s mulatory signal are other ap-
proaches that are being pursued. For example, expression

asco.org/edbook | 2018 ASCO EDUCATIONAL BOOK  833


Downloaded from ascopubs.org by 41.68.253.21 on June 26, 2021 from 041.068.253.021
Copyright © 2021 American Society of Clinical Oncology. All rights reserved.
DERENZO, KRENCIUTE, AND GOTTSCHALK

of a dominant nega ve TGFβ receptor renders T cells re- Need for Preclinical Tes ng in Range of Animal
sistant to TGFβ in preclinical as well as in clinical studies.56 Models
In addi on, dominant nega ve receptors have been de- The majority of preclinical studies have relied on xenogra
veloped to provide intrinsic protec on from PD-1/PD-L1 models, which do not reliably recapitulate the complex tu-
checkpoint blockade.57 Chimeric cytokine or switch recep- mor microenvironment. Immunocompetent animal mod-
tors not only block an inhibitory signal but also convert it els have been adapted for CAR T-cell therapies, and these
into a T-cell s mulatory signal. Examples include receptors models will be invaluable as combina on therapies are
that consist of the ectodomain of the IL-4 receptor and developed.79-81 In addi on, pa ent-derived xenogra mod-
the transmembrane and intracellular signaling domain of els should enable preclinical tes ng of CAR T cells against
the IL-2 or IL-7 receptor.58,59 Although siRNA approaches a panel of human tumors that more closely mimic pa ent
were used ini ally to silence nega ve regulators, more re- tumors than tumor cell lines that have been propagated in
cent studies have focused on gene edi ng technologies, vitro. Also, large animal models hold promise to evaluate
such as TALENs and CRIPSR/Cas9. Per nent examples in- CAR T cells in spontaneous tumor models.82
clude the silencing of FAS ligand or the knockout of PD-1 in
T cells.60,61 Correla ve Studies, In Vivo Tracking of Infused T
As we enhance the effector func on of CAR T cells, it is Cells, and Clinical Response Criteria
advisable to insert safety switches that can be ac vated if There currently is only one published CAR T-cell therapy
adverse effects develop. Safety switches that have been (noted in the Brain Tumor sec on) that has systema cally
tested clinically include HSV thymidine kinase (HSV-tk), studied tumor biopsies a er infusion.26 These types of stud-
which enables cell killing in the presence of ganciclovir,62 or ies will be cri cal to understand current therapeu c failures
an inducible caspase 9,63 which can be ac vated by a chem- and to devise evidence-based approaches to overcome
ical inducer of dimeriza on. In addi on, expression of cell them. In addi on, our ability to track infused CAR T cells
surface molecules (e.g., EGFR, CD20),64,65 which can be tar- in pa ents is limited unless the cells are gene cally modi-
geted with FDA-approved monoclonal an bodies, is another fied with a reporter gene, such HSV thymidine kinase.83 The
suicide gene op on that has been evaluated successfully in ability to rou nely track CAR T cells for 48 to 72 hours a er
preclinical models. infusion would be a major advance that could provide in-
valuable insight into ini al biodistribu on of CAR T cells and
CURRENT CHALLENGES their ability to migrate to tumor sites. Although diagnos c
CAR T-Cell Genera on imaging immune response criteria have been implemented
The majority of clinical studies have used retroviral or len - for the assessment of immunotherapies for solid tumors
viral vectors to generate clinical-grade CAR T-cell products. and brain tumors,84,85 these were developed in the cancer
Genera on of clinical-grade viral vectors is me consuming vaccine era and might require addi onal fine tuning for cell-
and costly, and their use is associated with a large regulatory based immunotherapies, including CAR T cells.
burden. In this regard, reducing some of the required test-
ing of CAR T-cell products, as recently advocated,66,67 would CONCLUSIONS
be a step in the right direc on. Required tes ng could also The ini al foray of CAR T cells into the clinic for pediatric
be reduced with the use of nonviral DNA delivery systems solid tumors and brain tumors demonstrated their safety but
that have been successfully used to generate CAR T cells.68-70 also highlighted their limited an tumor ac vity. Addi onal
Closed-cell manufacturing systems71,72 and the use of off- gene c modifica on of CAR T cells has greatly enhanced the
the-shelf CAR T-cell products73 also hold the promise to an tumor ac vity of these cells in preclinical studies, and
streamline CAR T-cell produc on and/or distribu on. we hope that some of the devised strategies will translate
into improved an tumor ac vity in humans. To advance
What Is the Op mal T-Cell Subset to Generate CAR T the field, there is an urgent need to discover novel an gens
Cells? that can be targeted with CAR T cells and to improve our
Several studies have highlighted that CAR T cells genera on ability to evaluate CAR T-cell therapies in preclinical mod-
from central memory T cells with a defined CD4:CD8 ra o els. The abili es to track CAR T cells noninvasively and to
have superior effector func on compared with CAR T cells perform detailed studies with pa ents enrolled in CAR T-cell
generated from bulk T cells in preclinical models.74,75 Epigen- therapy should enable us to advance the field. We remain
e c profiling of T cells has provided novel insight76 and holds hopeful that, within the next 5 to 10 years, pa ents with
the promise to advance our ability to select the most potent solid tumors will benefit from CAR T-cell therapies to the
T-cell subset for CAR T-cell genera on. Also, γδ T cells and same degree that pa ents with B-cell–derived malignancies
invariant natural killer T cells are being explored as T-cell do today.
pla orms for CAR T-cell therapy.77,78 In this regard, a clinical
study with invariant natural killer T cells that express GD2– ACKNOWLEDGMENT
CARs and IL-15 for pa ents with neuroblastoma has been This work was supported by Na onal Ins tutes of Health
approved by the FDA (NCT03294954) but is not ac vely ac- Grants No. 5T32HL092332 and 1R01CA173750 and CRPIT
cruing pa ents. Grant No. RP101335.

834 2018 ASCO EDUCATIONAL BOOK | asco.org/edbook


Downloaded from ascopubs.org by 41.68.253.21 on June 26, 2021 from 041.068.253.021
Copyright © 2021 American Society of Clinical Oncology. All rights reserved.
LANDSCAPE OF CAR T CELLS BEYOND ACUTE LYMPHOBLASTIC LEUKEMIA

References
1. Ruella M, June CH. Chimeric an gen receptor T cells for B cell 18. Louis CU, Savoldo B, Do G, et al. An tumor ac vity and long-term
neoplasms: choose the right CAR for you. Curr Hematol Malig Rep. fate of chimeric an gen receptor-posi ve T cells in pa ents with
2016;11:368-384. neuroblastoma. Blood. 2011;118:6050-6056.
2. Park JH, Geyer MB, Brentjens RJ. CD19-targeted CAR T-cell therapeu cs 19. Heczey A, Louis CU, Savoldo B, et al. CAR T cells administered in
for hematologic malignancies: interpre ng clinical outcomes to date. combina on with lymphodeple on and PD-1 inhibi on to pa ents
Blood. 2016;127:3312-3320. with neuroblastoma. Mol Ther. 2017;25:2214-2224.
3. Kochenderfer JN, Dudley ME, Feldman SA, et al. B-cell deple on and 20. Ahmed N, Brawley VS, Hegde M, et al. Human epidermal growth
remissions of malignancy along with cytokine-associated toxicity in factor receptor 2 (HER2) -specific chimeric an gen receptor-modified
a clinical trial of an -CD19 chimeric-an gen-receptor-transduced T T cells for the immunotherapy of HER2-posi ve sarcoma. J Clin Oncol.
cells. Blood. 2012;119:2709-2720. 2015;33:1688-1696.
4. Kochenderfer JN, Dudley ME, Kassim SH, et al. Chemotherapy- 21. Hegde M, DeRenzo CC, Zhang H, et al. Expansion of HER2-CAR T
refractory diffuse large B-cell lymphoma and indolent B-cell cells a er lymphodeple on and clinical responses in pa ents with
malignancies can be effec vely treated with autologous T cells advanced sarcoma. J Clin Oncol. 2017;35 (suppl; abstr 10508).
expressing an an -CD19 chimeric an gen receptor. J Clin Oncol. 22. Ahmed N, Brawley V, Hegde M, et al. HER2-specific chimeric an gen
2015;33:540-549. receptor-modified virus-specific T cells for progressive glioblastoma: a
5. Maude SL, Frey N, Shaw PA, et al. Chimeric an gen receptor T cells for phase I dose-escala on trial. JAMA Oncol. 2017;3:1094-1101.
sustained remissions in leukemia. N Engl J Med. 2014;371:1507-1517. 23. Brown CE, Alizadeh D, Starr R, et al. Regression of glioblastoma a er
6. Lee DW, Kochenderfer JN, Stetler-Stevenson M, et al. T cells expressing chimeric an gen receptor T-cell therapy. N Engl J Med. 2016;375:2561-
CD19 chimeric an gen receptors for acute lymphoblas c leukemia 2569.
in children and young adults: a phase I dose-escala on trial. Lancet. 24. Künkele A, Taraseviciute A, Finn LS, et al. Preclinical assessment
2015;385:517-528. of CD171-directed CAR T-cell adop ve therapy for childhood
7. Turtle CJ, Hanafi LA, Berger C, et al. Immunotherapy of non-Hodgkin’s neuroblastoma: CE7 epitope target safety and product manufacturing
lymphoma with a defined ra o of CD8+ and CD4+ CD19-specific chimeric feasibility. Clin Cancer Res. 2017;23:466-477.
an gen receptor-modified T cells. Sci Transl Med. 2016;8:355ra116.
25. Li W, Guo L, Rathi P, et al. Redirec ng T cells to glypican-3 with 4-1BB
8. Turtle CJ, Riddell SR, Maloney DG. CD19-targeted chimeric an gen zeta chimeric an gen receptors results in Th1 polariza on and potent
receptor-modified T-cell immunotherapy for B-cell malignancies. Clin an tumor ac vity. Hum Gene Ther. 2017;28:437-448.
Pharmacol Ther. 2016;100:252-258.
26. O’Rourke DM, Nasrallah MP, Desai A, et al. A single dose of peripherally
9. Gardner RA, Finney O, Annesley C, et al. Intent-to-treat leukemia infused EGFRvIII-directed CAR T cells mediates an gen loss and
remission by CD19 CAR T cells of defined formula on and dose in induces adap ve resistance in pa ents with recurrent glioblastoma.
children and young adults. Blood. 2017;129:3322-3331. Sci Transl Med. 2017;9:9.
10. Brentjens RJ, Davila ML, Riviere I, et al. CD19-targeted T cells rapidly 27. Brown CE, Badie B, Barish ME, et al. Bioac vity and safety of IL13Rα2-
induce molecular remissions in adults with chemotherapy-refractory redirected chimeric an gen receptor CD8+ T cells in pa ents with
acute lymphoblas c leukemia. Sci Transl Med. 2013;5:177ra38. recurrent glioblastoma. Clin Cancer Res. 2015;21:4062-4072.
11. Park JH, Rivière I, Gonen M, et al. Long-term follow-up of CD19 CAR 28. Orentas RJ, Yang JJ, Wen X, et al. Iden fica on of cell surface proteins
therapy in acute lymphoblas c leukemia. N Engl J Med. 2018;378:449- as poten al immunotherapy targets in 12 pediatric cancers. Front
459. Oncol. 2012;2:194.
12. Eshhar Z, Waks T, Gross G, et al. Specific ac va on and targe ng of 29. Bosse KR, Raman P, Zhu Z, et al. Iden fica on of GPC2 as an
cytotoxic lymphocytes through chimeric single chains consis ng oncoprotein and candidate immunotherapeu c target in high-risk
of an body-binding domains and the gamma or zeta subunits of neuroblastoma. Cancer Cell. 2017;32:295-309.e12
the immunoglobulin and T-cell receptors. Proc Natl Acad Sci USA. 30. Gröbner SN, Worst BC, Weischenfeldt J, et al; ICGC MMML-Seq
1993;90:720-724. Project. The landscape of genomic altera ons across childhood
13. Do G, Go schalk S, Savoldo B, et al. Design and development of cancers. Nature. 2018;555:321-327.
therapies using chimeric an gen receptor-expressing T cells. Immunol 31. Bu erfield LH, Ribas A, Disse e VB, et al. Determinant spreading
Rev. 2014;257:107-126. associated with clinical response in dendri c cell-based immunotherapy
14. Sadelain M. Chimeric an gen receptors: driving immunology towards for malignant melanoma. Clin Cancer Res. 2003;9:998-1008.
synthe c biology. Curr Opin Immunol. 2016;41:68-76. 32. Hegde M, Mukherjee M, Grada Z, et al. Tandem CAR T cells targe ng
15. Sadelain M. CD19 CAR T cells. Cell. 2017;171:1471. HER2 and IL13Rα2 mi gate tumor an gen escape. J Clin Invest.
2016;126:3036-3052.
16. Park JR, Digiusto DL, Slovak M, et al. Adop ve transfer of chimeric
an gen receptor re-directed cytoly c T lymphocyte clones in pa ents 33. Bielamowicz K, Fousek K, Byrd TT, et al. Trivalent CAR T-cells
with neuroblastoma. Mol Ther. 2007;15:825-833. overcome interpa ent an genic variability in glioblastoma. Neuro-
oncol. 2018;20:506-518.
17. Pule MA, Savoldo B, Myers GD, et al. Virus-specific T cells engineered
to coexpress tumor-specific receptors: persistence and an tumor 34. Fedorov VD, Themeli M, Sadelain M. PD-1- and CTLA-4-based inhibitory
ac vity in individuals with neuroblastoma. Nat Med. 2008;14:1264- chimeric an gen receptors (iCARs) divert off-target immunotherapy
1270. responses. Sci Transl Med. 2013;5:215ra172.

asco.org/edbook | 2018 ASCO EDUCATIONAL BOOK  835


Downloaded from ascopubs.org by 41.68.253.21 on June 26, 2021 from 041.068.253.021
Copyright © 2021 American Society of Clinical Oncology. All rights reserved.
DERENZO, KRENCIUTE, AND GOTTSCHALK

35. Rafiq S, Purdon TJ, Daniyan AF, et al. Op mized T-cell receptor-mimic 53. Zhao Z, Condomines M, van der Stegen SJC, et al. Structural design
chimeric an gen receptor T cells directed toward the intracellular of engineered cos mula on determines tumor rejec on kine cs and
Wilms Tumor 1 an gen. Leukemia. 2017;31:1788-1797. persistence of CAR T cells. Cancer Cell. 2015;28:415-428.
36. Ma Q, Garber HR, Lu S, et al. A novel TCR-like CAR with specificity 54. Curran KJ, Seinstra BA, Nikhamin Y, et al. Enhancing an tumor efficacy
for PR1/HLA-A2 effec vely targets myeloid leukemia in vitro when of chimeric an gen receptor T cells through cons tu ve CD40L
expressed in human adult peripheral blood and cord blood T cells. expression. Mol Ther. 2015;23:769-778.
Cytotherapy. 2016;18:985-994. 55. Adachi K, Kano Y, Nagai T, et al. IL-7 and CCL19 expression in CAR-T
37. Roybal KT, Lim WA. Synthe c immunology: hacking immune cells cells improves immune cell infiltra on and CAR-T cell survival in the
to expand their therapeu c capabili es. Annu Rev Immunol. tumor. Nat Biotechnol. 2018;36:346-351.
2017;35:229-253. 56. Bollard CM, Tripic T, Cruz CR, et al. Tumor-specific T-cells engineered to
38. Morsut L, Roybal KT, Xiong X, et al. Engineering customized cell overcome tumor immune evasion induce clinical responses in pa ents
sensing and response behaviors using synthe c notch receptors. Cell. with relapsed Hodgkin lymphoma. J Clin Oncol. Epub 2018 Jan 9.
2016;164:780-791. 57. Cherkassky L, Morello A, Villena-Vargas J, et al. Human CAR T cells
39. Kershaw MH, Wang G, Westwood JA, et al. Redirec ng migra on of T with cell-intrinsic PD-1 checkpoint blockade resist tumor-mediated
cells to chemokine secreted from tumors by gene c modifica on with inhibi on. J Clin Invest. 2016;126:3130-3144.
CXCR2. Hum Gene Ther. 2002;13:1971-1980. 58. Levy R. A perspec ve on monoclonal an body therapy: where we
40. Craddock JA, Lu A, Bear A, et al. Enhanced tumor trafficking of GD2 have been and where we are going. Semin.Hematol. 2000;37:43-46
chimeric an gen receptor T cells by expression of the chemokine 59. Leen AM, Sukumaran S, Watanabe N, et al. Reversal of tumor immune
receptor CCR2b. J Immunother. 2010;33:780-788. inhibi on using a chimeric cytokine receptor. Mol Ther. 2014;22:1211-
41. Caruana I, Savoldo B, Hoyos V, et al. Heparanase promotes tumor 1220.
infiltra on and an tumor ac vity of CAR-redirected T lymphocytes. 60. Do G, Savoldo B, Pule M, et al. Human cytotoxic T lymphocytes with
Nat Med. 2015;21:524-529. reduced sensi vity to Fas-induced apoptosis. Blood. 2005;105:4677-
42. Kakarla S, Chow KK, Mata M, et al. An tumor effects of chimeric 4684.
receptor engineered human T cells directed to tumor stroma. Mol 61. Rupp LJ, Schumann K, Roybal KT, et al. CRISPR/Cas9-mediated PD-1
Ther. 2013;21:1611-1620. disrup on enhances an -tumor efficacy of human chimeric an gen
43. Kesarwani P, Kant S, Prabhu A, et al. The interplay between metabolic receptor T cells. Sci Rep. 2017;7:737.
remodeling and immune regula on in glioblastoma. Neuro-oncol. 62. Lupo-Stanghellini MT, Provasi E, Bondanza A, et al. Clinical impact
2017;19:1308-1315. of suicide gene therapy in allogeneic hematopoie c stem cell
44. Kamran N, Chandran M, Lowenstein PR, et al. Immature myeloid cells transplanta on. Hum Gene Ther. 2010;21:241-250.
in the tumor microenvironment: implica ons for immunotherapy. Clin 63. Di Stasi A, Tey SK, Do G, et al. Inducible apoptosis as a safety switch
Immunol. 2016;S1521-6616(16)30518-6. for adop ve cell therapy. N Engl J Med. 2011;365:1673-1683.
45. Zou W. Immunosuppressive networks in the tumour environment and 64. Vogler I, Newrzela S, Hartmann S, et al. An improved bicistronic
their therapeu c relevance. Nat Rev Cancer. 2005;5:263-274. CD20/tCD34 vector for efficient purifica on and in vivo deple on
46. Nishio N, Diaconu I, Liu H, et al. Armed oncoly c virus enhances of gene-modified T cells for adop ve immunotherapy. Mol Ther.
immune func ons of chimeric an gen receptor-modified T cells in 2010;18:1330-1338.
solid tumors. Cancer Res. 2014;74:5195-5205. 65. Wang X, Chang WC, Wong CW, et al. A transgene-encoded cell surface
47. Tanoue K, Rosewell Shaw A, Watanabe N, et al. Armed oncoly c polypep de for selec on, in vivo tracking, and abla on of engineered
adenovirus-expressing PD-L1 mini-body enhances an tumor effects cells. Blood. 2011;118:1255-1263.
of chimeric an gen receptor T cells in solid tumors. Cancer Res. 66. Corne a K, Duffy L, Turtle CJ, et al. Absence of replica on-competent
2017;77:2040-2051. len virus in the clinic: analysis of infused T cell products. Mol Ther.
48. John LB, Devaud C, Duong CP, et al. An -PD-1 an body therapy 2018;26:280-288.
potently enhances the eradica on of established tumors by gene- 67. Heslop HE, Brenner MK. Seek and you will not find: ending the hunt
modified T cells. Clin Cancer Res. 2013;19:5636-5646. for replica on-competent retroviruses during human gene therapy.
49. Krenciute G, Prinzing BL, Yi Z, et al. Transgenic expression of IL15 Mol Ther. 2018;26:1-2.
improves an glioma ac vity of IL13Rα2-CAR T cells but results in 68. Kebriaei P, Singh H, Huls MH, et al. Phase I trials using Sleeping Beauty
an gen loss variants. Cancer Immunol Res. 2017;5:571-581. to generate CD19-specific CAR T cells. J Clin Invest. 2016;126:3363-
50. Koneru M, Purdon TJ, Spriggs D, et al. IL-12 secre ng tumor-targeted 3376.
chimeric an gen receptor T cells eradicate ovarian tumors in vivo. 69. Manuri PV, Wilson MH, Mai SN, et al. PiggyBac transposon/
OncoImmunology. 2015;4:e994446. transposase system to generate CD19-specific T cells for the treatment
51. Hu B, Ren J, Luo Y, et al. Augmenta on of an tumor immunity of B-lineage malignancies. Hum Gene Ther. 2010;21:427-437.
by human and mouse CAR T cells secre ng IL-18. Cell Reports. 70. Nakazawa Y, Huye LE, Salsman VS, et al. PiggyBac-mediated cancer
2017;20:3025-3033. immunotherapy using EBV-specific cytotoxic T-cells expressing HER2-
specific chimeric an gen receptor. Mol Ther. 2011;19:2133-2143.
52. Shum T, Omer B, Tashiro H, et al. Cons tu ve signaling from an
engineered IL7 receptor promotes durable tumor elimina on by 71. Zhu F, Shah N, Xu H, et al. Closed-system manufacturing of CD19 and
tumor-redirected T cells. Cancer Discov. 2017;7:1238-1247. dual-targeted CD20/19 chimeric an gen receptor T cells using the

836 2018 ASCO EDUCATIONAL BOOK | asco.org/edbook


Downloaded from ascopubs.org by 41.68.253.21 on June 26, 2021 from 041.068.253.021
Copyright © 2021 American Society of Clinical Oncology. All rights reserved.
LANDSCAPE OF CAR T CELLS BEYOND ACUTE LYMPHOBLASTIC LEUKEMIA

CliniMACS Prodigy device at an academic medical center. Cytotherapy. 79. Johnson LA, Sanchez-Perez L, Suryadevara CM, et al. Chimeric
2017;20:394-406 an gen receptor engineered T cells can eliminate brain tumors and
ini ate long-term protec on against recurrence. OncoImmunology.
72. Lock D, Mockel-Tenbrinck N, Drechsel K, et al. Automated
2014;3:e944059.
manufacturing of potent CD20-directed chimeric an gen receptor T
cells for clinical use. Hum Gene Ther. 2017;28:914-925. 80. Barber A, Rynda A, Sentman CL. Chimeric NKG2D-expressing T cells
73. Poirot L, Philip B, Schiffer-Mannioui C, et al. Mul plex genome edi ng eliminate immunosuppression and ac vate immunity within the
of TCRa/CD52 genes as a pla orm for “off the shelf” adop ve T-cell ovarian tumor microenvironment. J Immunol. 2009;183:6939-6947.
immunotherapies. Mol Ther. 2014;22:S201-S202. 81. Pituch KC, Miska J, Krenciute G, et al. Adop ve transfer of IL13Rα2-
74. Greenberg P, Klarnet J, Kern D, et al. Requirements for T cell recogni on specific chimeric an gen receptor T cells creates a pro-inflammatory
and elimina on of retrovirally transformed cells. Princess Takamatsu environment in glioblastoma. Mol Ther. Epub 2018 Feb 8.
Symp. 1988;19:287-301. 82. Panjwani MK, Smith JB, Schutsky K, et al. Feasibility and safety of
75. Sommermeyer D, Hudecek M, Kosasih PL, et al. Chimeric an gen RNA-transfected CD20-specific chimeric an gen receptor T cells in
receptor-modified T cells derived from defined CD8+ and CD4+ subsets dogs with spontaneous B cell lymphoma. Mol Ther. 2016;24:1602-
confer superior an tumor reac vity in vivo. Leukemia. 2016;30:492-500. 1614.

76. Abdelsamed HA, Moustaki A, Fan Y, et al. Human memory CD8 T 83. Keu KV, Witney TH, Yaghoubi S, et al. Reporter gene imaging of
cell effector poten al is epigene cally preserved during in vivo targeted T cell immunotherapy in recurrent glioma. Sci Transl Med.
homeostasis. J Exp Med. 2017;214:1593-1606. 2017;9:9.

77. Heczey A, Liu D, Tian G, et al. Invariant NKT cells with chimeric an gen 84. Okada H, Weller M, Huang R, et al. Immunotherapy response
receptor provide a novel pla orm for safe and effec ve cancer assessment in neuro-oncology: a report of the RANO working group.
immunotherapy. Blood. 2014;124:2824-2833. Lancet Oncol. 2015;16:e534-e542.
78. Capsomidis A, Benthall G, Van Acker HH, et al. Chimeric an gen 85. Wolchok JD, Hoos A, O’Day S, et al. Guidelines for the evalua on of
receptor-engineered human gamma delta T cells: enhanced cytotoxicity immune therapy ac vity in solid tumors: immune-related response
with reten on of cross presenta on. Mol Ther. 2018;26:354-365. criteria. Clin Cancer Res. 2009;15:7412-7420.

asco.org/edbook | 2018 ASCO EDUCATIONAL BOOK  837


Downloaded from ascopubs.org by 41.68.253.21 on June 26, 2021 from 041.068.253.021
Copyright © 2021 American Society of Clinical Oncology. All rights reserved.

You might also like