You are on page 1of 7

Received: 15 November 2018 Revised: 17 January 2019 Accepted: 22 January 2019

DOI: 10.1002/ajh.25418

CRITICAL REVIEW

An introduction to chimeric antigen receptor (CAR) T-cell


immunotherapy for human cancer
Steven Feins1,2,3 | Weimin Kong1,2,3 | Erik F. Williams1,2,3 | Michael C. Milone2,3,4,5 |
Joseph A. Fraietta1,2,3,4,5

1
Department of Microbiology, Perelman
School of Medicine, University of Chimeric antigen receptor (CAR) T-cell therapy represents a major advancement in personalized
Pennsylvania, Philadelphia, Pennsylvania cancer treatment. In this strategy, a patient's own T cells are genetically engineered to express a
2
Department of Pathology and Laboratory synthetic receptor that binds a tumor antigen. CAR T cells are then expanded for clinical use
Medicine, Perelman School of Medicine,
and infused back into the patient's body to attack and destroy chemotherapy-resistant cancer.
University of Pennsylvania, Philadelphia,
Pennsylvania Dramatic clinical responses and high rates of complete remission have been observed in the set-
3
Center for Cellular Immunotherapies, ting of CAR T-cell therapy of B-cell malignancies. This resulted in two recent FDA approvals of
University of Pennsylvania, Philadelphia, CAR T cells directed against the CD19 protein for treatment of acute lymphoblastic leukemia
Pennsylvania and diffuse large B-cell lymphoma. Thus, CAR T cells are arguably one of the first successful
4
Abramson Cancer Center, University of examples of synthetic biology and personalized cellular cancer therapy to become commercially
Pennsylvania, Philadelphia, Pennsylvania
5
available. In this review, we introduce the concept of using CAR T cells to break immunological
Parker Institute for Cancer Immunotherapy,
tolerance to tumors, highlight several challenges in the field, discuss the utility of biomarkers in
University of Pennsylvania, Philadelphia,
Pennsylvania the context of predicting clinical responses, and offer prospects for developing next-generation
Correspondence CAR T cell-based approaches that will improve outcome.
Joseph A. Fraietta, South Pavilion Expansion,
Room 9-104, 3400 Civic Center Blvd., Bldg.
421, Philadelphia, PA 19104.
Email: jfrai@upenn.edu
Funding information
Gabrielle's Angel Foundation for Cancer
Research; National Cancer Institute, Grant/
Award Number: P01CA214278-01; Parker
Institute for Cancer Immunotherapy

1 | INTRODUCTION TO ADOPTIVE CELL A major obstacle facing the field of cancer immunotherapy is
THERAPY FOR CANCER breaking tolerance to “self” antigens. Based on the nature of the target,
tumor-specific immune responses are directed against either “self” or
Adoptive transfer is a term coined by Billingham, Brent, and Medawar1,2 “foreign” antigens.5 Foreign antigens can arise in virally-induced cancers
to describe allograft rejection, and the phrase “adoptive immuno- or from genetic aberrations unique to a particular tumor, such as point
therapy” denotes the infusion of immunocompetent cells for the mutations or chromosomal rearrangements that generate epitopes rec-
treatment of cancer or infectious disease.3 Mitchison and colleagues ognized by T cells and B cells. Immune responses against such foreign
first reported specific targeting of tumor grafts through the adoptive epitopes are likely similar to those elicited against pathogens, and
transfer of lymphocytes in murine models over 60 years ago.4 This therefore may not be susceptible to normal pathways that mediate
strategy has the potential to overcome many of the pitfalls associ- self-tolerance. In contrast, immune responses mounted against tumor-
ated with vaccine-based approaches for treating cancer, such as the associated self-antigens are subject to suppression by central or periph-
de novo activation and expansion of tumor-specific T cells in vivo, eral tolerance mechanisms. One major issue is thymic elimination of
even in immune compromised patients. Accordingly, adoptive cell high-affinity T-cell receptors (TCRs), resulting in the predominance of
transfer is a robust form of immunotherapy for treatment of estab- relatively low-affinity TCRs to self-antigens compared to foreign anti-
lished tumors. gens. In humans, TCRs that recognize self-tumor antigens have a 1.5

Am J Hematol. 2019;94:S3–S9. wileyonlinelibrary.com/journal/ajh © 2019 Wiley Periodicals, Inc. S3


S4 FEINS ET AL.

log lower affinity for cognate major histocompatibility complex (MHC): receptor-tyrosine-based-activation-motif [ITAM]-containing protein),
6
peptide complexes, relative to their virus-specific TCR counterparts. and optional tandem co-stimulation domains such as the 4-1BB or
Thus, the endogenous T-cell repertoire is often inadequate for control CD28 signaling modules. First generation CARs contain CD3ζ alone,
of human cancer. while second generation chimeric receptors also incorporate a co-
stimulatory endodomain (e.g., 4-1BB/CD3ζ). Future studies with third
generation CARs containing multiple co-stimulatory signaling modules
2 | BREAKING IMMUNOLOGICAL are also underway (reviewed in Ref. 15; see Figure 1). The main
T O L E R A N C E T O CA N C E R WI T H CA R T C E L L S advantage of using CAR-based approaches for cancer immunotherapy
is that the scFv is derived from an antibody with affinities several
The potential for immunotherapy to treat the most challenging human orders of magnitude higher than TCRs (reviewed in Ref. 16). Because
cancers has been demonstrated in pre-clinical models and clinical tri- scFvs possess the capacity to recognize intact cell surface proteins,
als. A number of approaches have been taken to circumvent the issue CAR T cell-mediated targeting of tumors is neither restricted and nor
of tolerance in the setting of cancer immunotherapy, including dependent on antigen processing and presentation. CAR T cells are
infusion of allogeneic T cells, adoptive transfer of expanded tumor therefore insensitive to tumor escape mechanisms related to MHC
infiltrating lymphocytes (TILs), and inhibition of negative regulators of loss. In addition, CARs can target antigens such as glycolipids, aber-
immune activation (e.g., cytotoxic T-lymphocyte associated protein rantly glycosylated proteins and conformational epitopes that cannot
4 [CTLA-4], programmed cell death-1 [PD-1], transforming growth be easily recognized, if at all, by TCRs. Based on results from clinical
factor beta [TGFβ], reviewed in Ref. 7). Some therapeutic monoclonal trials conducted at our institution and elsewhere, there is an increas-
antibodies (mAbs) are designed to bind a tumor antigen and induce ing consensus that CAR T cells have the potential to deliver powerful
cell death through various mechanisms or to carry tumoricidal sub- anti-tumor effects. These efforts culminated in the recent FDA
stances.8 In addition, a newer class of mAbs known as bispecific T-cell approval of CAR T cells directed against the CD19 protein for the
enhancing antibodies has been designed to simultaneously target a treatment of acute lymphoblastic leukemia (ALL) and diffuse large B-
tumor-associated antigen and a T-cell antigen (e.g., CD19 and CD3, cell lymphoma (DLBCL).
respectively), which results in enhanced co-localization of T cells and Although CARs were first proposed in the late 1980s, these syn-
tumors.9 Recently, the use of mAbs to inhibit the aforementioned thetic antigen receptors were not tested clinically as a cancer therapy
negative immune checkpoints (i.e., CTLA-4 and PD-1) has demon- until 2006. The initial results in adult patients were not promising due
strated remarkable benefit in the treatment of a variety of cancer to poor persistence of genetically-engineered lymphocytes.17,18 One
types (reviewed in Ref. 10). Thus, antibody-based immunotherapies study had shown some evidence of anti-tumor effects in 4 of 8 chil-
for cancer are now well established in the clinician's armamentarium. dren with metastatic neuroblastoma following a single infusion of first
The adoptive transfer of autologous T cells engineered by gene generation CAR T cells containing CD3ζ alone, but the persistence of
transfer to express receptors targeting molecules expressed on malig- these cells was short in most subjects.19 Although CAR T cells were
nant cells may have greater potential to transform cancer therapeutics safe, they continued to show disappointing clinical activity in the
compared to TIL- and antibody-based approaches. Genes encoding majority of early trials.20 The central issue facing the field was that
antigen receptors can be introduced into the patient's T cells, which CAR T cells did not persist and expand in vivo for more than a few
are then rapidly expanded to derive memory and effector lympho- days. Efforts to overcome this challenge with dose escalation were
cytes capable of proliferating robustly in vivo and eliciting potent anti- not successful, which underscored the need to enhance the persis-
tumor activity. One such strategy to overcome tolerance in cancer is tence and sustain the function of CAR T cells in vivo.
to genetically redirect and reprogram T cells with chimeric antigen The incorporation of CD28 and 4-1BB signaling modules into
receptors (CARs). These synthetic receptors combine the effector CARs can recapitulate natural co-stimulation and increases the
functions of T lymphocytes and the ability of antibodies to recognize potency of engineered T cells.21–27 Our pre-clinical work led to the
pre-defined surface antigens with a high degree of specificity in a identification of the 4-1BB (CD137) signaling domain as a critical
non-MHC restricted manner. determinant of survival and proliferation of CAR T cells in mice with
Expression of the first synthetic immunoglobulin/TCR chimeric pre-B ALL and carcinoma.28,29 In these studies, 4-1BB over CD28
molecule with antibody-like specificity was reported over 3 decades co-stimulation or CD3ζ signaling alone in CAR T cells enabled
ago.11,12 Shortly following these initial discoveries, Irving and Weiss enhanced survival of tumor-bearing mice.29 We then conducted the
determined that a CAR composed of CD8 and the CD3ζ chain could first clinical trial of a second generation CAR signaling through
mediate T-cell activation independently of the endogenous TCR.13 4-1BB/CD3ζ delivered by lentiviral vector transduction and found
CARs typically encode an extracellular domain for tumor antigen rec- that these CAR T cells are capable of unprecedented on-target clinical
ognition, linked to one or more intracellular signaling domains that activity in chronic lymphocytic leukemia (CLL).30,31 These findings
mediate T-cell activation. The antigen-binding single chain variable served as the springboard to test this approach in other indications,
fragment (scFv) portion of a CAR traditionally consists of the variable including ALL32–34 and DLBCL35,36 for which autologous CD19 CAR T
heavy (VH) and variable light (VL) chains of an antibody, fused by a cells are now commercially available. Recent trials of CAR T cells for
peptide spacer of ~15 residues in length.14 This molecule is joined to the treatment of multiple myeloma have also exhibited promising
an intracellular signaling molecule comprised of the TCR CD3ζ signal- results.37–39 Clinical outcomes of patients infused with CAR T cells for
ing chain (or the intracellular signaling domain of another immune- therapy of some of the above indications are summarized in the
FEINS ET AL. S5

FIGURE 1 Schematic of chimeric antigen receptor (CAR) structure. CARs are composed of a single-chain variable fragment (scFv) consisting of
antibody variable heavy (VH) and variable light (VL) chains, fused by a peptide linker. This antigen-binding scFv of a CAR is joined to an
intracellular CD3ζ signaling domain. First generation CARs contain CD3ζ alone, while second generation synthetic antigen receptors incorporate
an addition co-stimulatory endodomain. Third generation CARs include multiple co-stimulatory signaling modules

proceeding reviews found in this special issue of the American Journal CAR T cells.41 Thus, new manufacturing technologies focused on
of Hematology. achieving efficient T-cell purification and tumor cell purging will improve
the safety and potency of cellular products for adoptive transfer
therapies.
3 | GENERATION OF S A FE AND To achieve durable clinical responses to cell-based gene therapies,
C L I N I C A L L Y E F F E C T I V E C A R T CE L L S permanent transgene expression is often required. Murine gammare-
troviruses and lentiviruses are two available clinical gene therapy
The production of CAR T cells typically involves cell collection from a vector systems that afford long-term CAR transgene expression. Con-
patient by leukapheresis, followed by elutriation to remove myeloid siderable clinical evidence shows that retroviral vectors are safe when
cells, T lymphocyte enrichment, transgene delivery, and ex vivo expan- expressed in human T cells. Longitudinal analysis of gammaretroviral
sion (see Figure 2). One of the critical cellular manufacturing constraints vector-engineered T cells for treatment of human immunodeficiency
is efficient isolation of T cells from leukapheresis samples. Leukapheresis virus (HIV) infection revealed the persistence of CAR T cells for over a
products from cancer patients consist of a heterogeneous population of decade in patients, without evidence of vector-induced immortaliza-
cells, including T cells, myeloid cells (e.g., monocytes, neutrophils, and tion of the engineered lymphocytes.42 However, retroviral vectors
dendritic cells), natural killer cells, erythroid cells, and malignant cells. appear to be less safe when used in human stem cells.43,44 In contrast,
With certain hematological malignancies such as leukemia, tumor cells lentiviral vectors, especially third generation self-inactivating vectors,
may comprise the bulk of the leukapheresis material. Despite the appli- have a lower risk of insertional mutagenesis, which may be attributed
cation of positive and negative selection methods, enriched T lympho- to the absence of strong enhancer elements present in oncogenic
cytes may remain contaminated by inhibitory cell types that can hamper murine gammaretroviruses.45,46 Lentiviral vectors also have substan-
efficient CAR T cell expansion in culture and subsequently, in vivo. Poor tially higher efficiency for genetically engineering human T cells.47
CAR T cell manufacturing outcome has been attributed to contamina- Nevertheless, we recently reported a case of a CLL patient who had a
tion of autologous peripheral blood mononuclear cells with mono- clonal expansion of his autologous CAR T cells due in part to lentiviral
cytes.40 Furthermore, we recently reported a rare case in which a single disruption of the Tet2 gene.41 Long-term monitoring of patients
leukemia cell was inadvertently transduced with the CAR transgene dur- receiving CAR T cells engineered with integrating viruses should
ing T-cell manufacturing. This resulted in a CAR binding in cis to the therefore continue to be carried out in pharmacovigilance studies. As
CD19 epitope on the surface of a leukemia clone that had expanded “safe harbor” sites in the genome are identified, it may be possible to
massively in an ALL patient, masking it from recognition by anti-CD19 develop clinically feasible cellular manufacturing processes based on
S6 FEINS ET AL.

FIGURE 2 Depiction of a standard chimeric antigen receptor (CAR) T-cell manufacturing process. Cells are collected from a patient by
leukapheresis, followed by monocyte elutriation and T-cell selection. The selected product is then activated (e.g., using paramagnetic beads
coated with anti-CD3 and anti-CD28 agonistic antibodies) and transduced with a viral vector encoding the CAR transgene. Following gene
delivery, T cells are expanded in culture and subsequently harvested to formulate the final CAR T-cell infusion product

guided integration of antigen receptor transgenes into specific loci 4 | P R E D I C T I N G R E S P O N S E T O C A R T- C E L L


using viral and non-viral methods.48,49 THERAPY
Compositions of CD4+ and CD8+ T cell subsets differ in cancer
patients relative to healthy individuals, and this variability may con- Despite a number of clinical successes, CAR T cells do not expand or
tribute to the efficacy of CAR T cell infusion products. Stem cell-like durably persist in some patients, while in others robust proliferation
memory T cells (Tscm) were identified by Gattinoni and colleagues to and clonal outgrowth of transferred T lymphocytes occur. A certain
be a self-renewing pool of lymphocytes with multipotent capacity to level of CAR T-cell expansion and persistence is necessary to induce
differentiate into central memory (TCM), effector memory (TEM) and meaningful tumor regressions, but the predictive indicators and
50
effector (TEFF) T cells. When these cells were enriched during CAR mechanism(s) associated with remarkable proliferation, persistence and

T-cell culture, they demonstrated superior anti-tumor activity com- favorable clinical responses are largely unknown. Central questions

pared to other early memory and effector subsets. 50


Clinical studies currently facing the field are how to enhance the potency and sustain

are currently being conducted to evaluate the safety and efficacy of the function of CAR T cells in vivo and how to develop mechanism-
based approaches to increase the resistance of CAR T cells to immuno-
TSCM CAR T cells directed against the B-cell maturation antigen for
senescence and exhaustion. In consideration of these challenges, there
the treatment of relapsed/refractory multiple myeloma.51 Similarly,
+ − + is an urgent need for identification of biomarkers to enable clinicians
we recently identified a population of CD27 PD-1 CD8 T cells that
to determine which patients will respond to CAR T-cell treatment a
was significantly enriched in CD19 CAR T-cell infusion products from
priori and so that treatments can be adjusted for maximization of ther-
CLL patients who had complete and durable responses to therapy.41
apeutic effects.
Low frequencies of this lymphocyte population in CAR T-cell infusion
We now have 8 years of experience with using CD19-directed
products are associated with reduced T-cell engraftment and poor
CAR T cells to treat B-cell malignancies, and our longest involvement
tumor control.41 Other groups have focused on formulating CAR
is in treating CLL. In the context of treating CLL with anti-CD19 CAR
T cells with defined CD4+:CD8+ compositions and these cellular prod-
T cells, we have not identified patient- or disease-specific features
ucts have demonstrated robust clinical potency in adult B-cell ALL
that forecast which subjects respond best to this treatment or why.
patients.52 Synergistic or additive augmentation of CAR T-cell efficacy
Response (or lack thereof) is not related to patient age, the number of
could be achieved by infusion of a defined ratio of CAR T cells derived
prior therapies received, p53 mutational status, peripheral or nodal
from early memory (e.g., TSCM and TCM) CD8+ and CD4+ T cells.
disease burden, CAR T-cell dose, or other usual risk factors.41,60 How-
In addition, because the presence of antigen-experienced T cells ever, in our series of CLL patients, we determined that the majority of
is known to impair the anti-tumor function of less differentiated subjects responding to CAR T-cell therapy displayed dramatic in vivo
lymphocytes,53 strategies to enrich early memory subsets at the expansion of CAR T cells in the first 2 weeks following infusion, which
beginning of CAR T-cell manufacturing or during culture may be was accompanied by the complete and persistent eradication of
beneficial for increasing the efficacy of the engineered cellular leukemia cells.30,31,60 The degree of CAR T-cell expansion in vivo
product. In addition to the aforementioned upfront T-cell purification strongly and directly correlated with the proliferative capacity of these
approaches, potency enhancement may be achieved by shortening the cells during ex vivo culture.41,61 In addition, the infused T cells per-
54
duration of CAR T-cell culture, limiting replicative lifespan through sisted long-term in the responding patients and were polyfunctional
prevention of telomere loss,55 metabolic programming,56 use of homeo- upon CAR-specific stimulation ex vivo. In line with these findings, it
57
static cytokines during lymphocyte expansion and/or epigenetic T-cell was recently reported that the anti-tumor activity and toxicity of
modulation.41,58,59 CD19 CAR T cells in non-Hodgkin's lymphoma (NHL) patients as well
FEINS ET AL. S7

as overall clinical outcome were associated with the polyfunctional vivo transfer.69 Successful development of “universal” CAR T cells
62
nature of the preinfusion cellular product. Additional predictive derived from healthy, allogeneic donors using precision genome editing
indicators of response to anti-CD19 CAR T-cell therapy include a high technologies (e.g., transcription activator-like effector nucleases, zinc-
frequency of CD3+CD8+CD27+CD45RO− lymphocytes at the time of finger nucleases, clustered regularly interspaced short palindromic
collection, cellular infusion products enriched in transcriptomic signa- repeats [CRISPR]/CRISPR associated protein 9) has the potential to cir-
tures of early memory T-cell differentiation (i.e., STAT3-related genes), cumvent issues associated with autologous T-cell defects such as termi-
and elevated pretreatment serum IL-15 levels.41,62 Given the intensive nal differentiation.
manufacturing process, the potential toxicity and possible new alter- In CLL, we found that CD19-targeted CAR T-cell infusion products
native treatments for some patients, the ability to use biomarkers to from non-responding patients when compared with those of complete
identify which subjects are most likely to respond to CAR T-cell ther- responders contained significantly higher frequencies of CD4+ and
apy would have a tremendous clinical benefit. Predictive biomarkers CD8+ T cells expressing multiple inhibitory receptors (e.g., PD-1, TIM-3,
can be utilized by physicians to prescribe CAR T-cell therapy only
and LAG-3).41 This finding suggests that in vivo blockade of negative
to the responding patient population, thus eliminating the unneces-
immune checkpoint molecules with concurrent CAR T-cell therapy rep-
sary expense and risk of immune-related adverse events for non-
resents a novel treatment strategy for overcoming resistance. Accord-
responders. Finally, systematic investigations of the mechanisms that
ingly, administration of a PD-1 blocking antibody therapy to a patient
potentiate the anti-tumor activity of CAR T cells at the cellular and
with refractory diffuse large B-cell lymphoma (DLBCL) that had
molecular level may reveal ways to generate potent cell infusion prod-
relapsed after receiving CAR-modified cells induced re-expansion of
ucts for many future patients.
CAR T cells, which was accompanied by a clinically significant anti-
tumor response.70 Future studies are needed to define whether com-
5 | OVERCOMING RESISTANCE TO CAR bining in vivo blockade with PD-1 or multiple other inhibitory receptors
T - C E L L T H E R A P Y OF H E M A T O P O I E T I C after CAR T-cell transfer is beneficial.
MALIGNANCIES

A mechanism of CAR T-cell failure in pediatric ALL is loss of the CD19


6 | CONC LU SION
antigen or epitope, despite otherwise adequate persistence of trans-
ferred cells. Antigen-negative tumor escape variants likely emerge
CAR T cells are changing the paradigm for cancer therapy. This new
through selection of sub-clonal leukemia cells that possess a survival
era of synthetic biology and medicine has been heralded by the recent
advantage in the face of a powerful antigen-specific assault mediated by
FDA approval of CD19 CAR T cells for the treatment of ALL and
CAR T cells. Unlike other CAR targets, CD19 escape was unexpected
DLBCL. CAR T-cell therapy for multiple other cancers will likely enter
due to its essential role in B-lineage development.63,64 Nevertheless, the
mainstream oncology. The capacity of CAR T cells to overcome toler-
first major mechanism of CD19 escape identified in anti-CD19 CAR
ance, expand as well as traffic to tumor sites, synergize with the
T-cell therapy was alternative splicing, leading to the production of
endogenous immune response, and persist long-term in vivo may
either a non-membrane bound form of CD19 (Δexon 5-6) or a poorly
allow this living, replicating drug to induce permanent anti-tumor
expressed variant that lacks an epitope recognized by the CAR scFv
(Δexon 2) on the cell surface. 65
Jacoby and colleagues subsequently effects in patients. Engineered T cells could therefore provide signifi-

reported that the immune pressure exerted on CD19 by CAR T cells cant therapeutic and economic advantages over antibody-based treat-

gives rise to either a rapidly relapsing leukemia lacking the epitope rec- ment regimens and other targeted inhibitors, as these modalities
ognized by CAR, or lineage-switched disease resulting from hematopoi- require prolonged administration and are not generally curative.
etic reprogramming. 66
More recently, Orlando et al. reported that Furthermore, administration of CAR T cells could obviate the need for
homozygous or biallelic frameshift mutations in CD19, rather than alter- allogeneic stem cell salvage procedures in the setting of particular hema-
native splicing, are the main cause of CD19 loss and acquired resistance topoietic cancers. Larger studies are currently being conducted to evalu-
67
to CAR T-cell therapy. Regardless of the escape mechanism, trials ate the safety and reliability of CAR T cells for diseases other than
based on administering combinations of CARs against multiple targets CD19-positive B-cell malignancies. With the emergence of improved cel-
will likely eliminate the emergence of antigen-negative leukemia that lular manufacturing techniques, novel cellular engineering approaches,
leads to a high degree of relapse in patients. precision genome editing technologies, and combination therapy strate-
Resistance to CAR therapy has also been attributed to failure of gies, we believe that CAR T cells will soon become an off-the-shelf, cost-
engraftment and/or expansion and persistence of engineered T cells. effective, and potentially curative treatment of human cancer.
As previously discussed, we and others have demonstrated that T cells
with an early memory phenotype are endowed with superior expansion
potential.41,68,69 In one study, T cells from patients with ALL had higher
absolute numbers of early-lineage lymphocytes than individuals with B ACKNOWLEDGMENTS

cell Non-Hodgkin lymphoma (NHL), and there was heterogeneity even This work was supported by P01CA214278 (M.C.M. and J.A.F.), the
among ALL patients, leading us to postulate that in some patients pre- Parker Institute for Cancer Immunotherapy (M.C.M. and J.A.F.) and
existing T cell quality may influence the degree of expansion upon in the Gabrielle's Angel Foundation (J.A.F.).
S8 FEINS ET AL.

CONF LICT OF IN TE RE ST 20. Till BG, Jensen MC, Wang J, et al. Adoptive immunotherapy for indo-
lent non-Hodgkin lymphoma and mantle cell lymphoma using geneti-
M.C.M. and J.A.F. hold patents related to CAR T cell therapy. M.C.M. is cally modified autologous CD20-specific T cells. Blood. 2008;112(6):
a co-founder and equity holder in Cabaletta Bio. J.A.F. receives research 2261-2271.
funding from Novartis Pharmaceuticals Corporation and Tmunity 21. Finney HM, Lawson AD, Bebbington CR, Weir AN. Chimeric receptors
providing both primary and costimulatory signaling in T cells from a
Therapeutics. The remaining authors declare no competing interests.
single gene product. J Immunol. 1998;161(6):2791-2797.
22. Eshhar Z, Waks T, Bendavid A, Schindler DG. Functional expression of
ORCID chimeric receptor genes in human T cells. J Immunol Methods. 2001;
248(1–2):67-76.
Joseph A. Fraietta https://orcid.org/0000-0001-7900-8993 23. Krause A, Guo HF, Latouche JB, Tan C, Cheung NK, Sadelain M.
Antigen-dependent CD28 signaling selectively enhances survival and
proliferation in genetically modified activated human primary T lym-
RE FE R ENC E S phocytes. J Exp Med. 1998;188(4):619-626.
24. Imai C, Mihara K, Andreansky M, et al. Chimeric receptors with 4-1BB
1. Billingham RE, Brent L, Medawar PB. Quantitative studies on tissue
signaling capacity provoke potent cytotoxicity against acute lympho-
transplantation immunity. II. The origin, strength and duration of
blastic leukemia. Leukemia. 2004;18(4):676-684.
actively and adoptively acquired immunity. Proc R Soc Lond B Biol Sci.
25. Friedmann-Morvinski D, Bendavid A, Waks T, Schindler D, Eshhar Z.
1954;143(910):58-80.
Redirected primary T cells harboring a chimeric receptor require costi-
2. Medawar PB. The behaviour and fate of skin autografts and skin
mulation for their antigen-specific activation. Blood. 2005;105(8):
homografts in rabbits: A report to the War Wounds Committee of the
3087-3093.
Medical Research Council. J Anat. 1944;78(Pt 5):176-199.
26. Maher J, Brentjens RJ, Gunset G, Riviere I, Sadelain M. Human
3. Rosenberg SA. Adoptive immunotherapy for cancer. Sci Am. 1990;262(5):
T-lymphocyte cytotoxicity and proliferation directed by a single chimeric
62-69.
TCRzeta /CD28 receptor. Nat Biotechnol. 2002;20(1):70-75.
4. Mitchison NA. Studies on the immunological response to foreign 27. Finney HM, Akbar AN, Lawson AD. Activation of resting human pri-
tumor transplants in the mouse. I. The role of lymph node cells in con- mary T cells with chimeric receptors: costimulation from CD28, induc-
ferring immunity by adoptive transfer. J Exp Med. 1955;102(2): ible costimulator, CD134, and CD137 in series with signals from the
157-177. TCR zeta chain. J Immunol. 2004;172(1):104-113.
5. Pardoll D. Does the immune system see tumors as foreign or self? 28. Milone MC, Fish JD, Carpenito C, et al. Chimeric receptors containing
Annu Rev Immunol. 2003;21:807-839. CD137 signal transduction domains mediate enhanced survival of
6. Aleksic M, Liddy N, Molloy PE, et al. Different affinity windows for T cells and increased antileukemic efficacy in vivo. Mol Ther. 2009;17
virus and cancer-specific T-cell receptors: implications for therapeutic (8):1453-1464.
strategies. Eur J Immunol. 2012;42(12):3174-3179. 29. Carpenito C, Milone MC, Hassan R, et al. Control of large, established
7. Sharma P, Hu-Lieskovan S, Wargo JA, Ribas A. Primary, adaptive, and tumor xenografts with genetically retargeted human T cells containing
acquired resistance to cancer immunotherapy. Cell. 2017;168(4): CD28 and CD137 domains. Proc Natl Acad Sci USA. 2009;106(9):
707-723. 3360-3365.
8. Hafeez U, Gan HK, Scott AM. Monoclonal antibodies as immunomod- 30. Porter DL, Levine BL, Kalos M, Bagg A, June CH. Chimeric antigen
ulatory therapy against cancer and autoimmune diseases. Curr Opin receptor-modified T cells in chronic lymphoid leukemia. N Engl J Med.
Pharmacol. 2018;41:114-121. 2011;365(8):725-733.
9. Dahlen E, Veitonmaki N, Norlen P. Bispecific antibodies in cancer 31. Kalos M, Levine BL, Porter DL, et al. T cells with chimeric antigen
immunotherapy. Ther Adv Vaccines Immunother. 2018;6(1):3-17. receptors have potent antitumor effects and can establish memory in
10. Ribas A, Wolchok JD. Cancer immunotherapy using checkpoint block- patients with advanced leukemia. Sci Transl Med. 2011;3(95):95ra73.
ade. Science. 2018;359(6382):1350-1355. 32. Maude SL, Frey N, Shaw PA, et al. Chimeric antigen receptor T cells
11. Kuwana Y, Asakura Y, Utsunomiya N, et al. Expression of chimeric for sustained remissions in leukemia. N Engl J Med. 2014;371(16):
receptor composed of immunoglobulin-derived V regions and T-cell 1507-1517.
receptor-derived C regions. Biochem Biophys Res Commun. 1987;149(3): 33. Maude SL, Laetsch TW, Buechner J, et al. Tisagenlecleucel in children
960-968. and young adults with B-cell lymphoblastic leukemia. N Engl J Med.
2018;378(5):439-448.
12. Gross G, Waks T, Eshhar Z. Expression of immunoglobulin-T-cell
34. Park JH, Riviere I, Gonen M, et al. Long-term follow-up of CD19 CAR
receptor chimeric molecules as functional receptors with antibody-
therapy in acute lymphoblastic leukemia. N Engl J Med. 2018;378(5):
type specificity. Proc Natl Acad Sci USA. 1989;86(24):10024-10028.
449-459.
13. Irving BA, Weiss A. The cytoplasmic domain of the T cell receptor zeta
35. Schuster SJ, Svoboda J, Chong EA, et al. Chimeric antigen receptor
chain is sufficient to couple to receptor-associated signal transduction
T cells in refractory B-cell lymphomas. N Engl J Med. 2017;377(26):
pathways. Cell. 1991;64(5):891-901.
2545-2554.
14. Mullaney BP, Pallavicini MG. Protein–protein interactions in hematol-
36. Neelapu SS, Locke FL, Bartlett NL, et al. Axicabtagene ciloleucel CAR
ogy and phage display. Exp Hematol. 2001;29(10):1136-1146.
T-cell therapy in refractory large B-cell lymphoma. N Engl J Med. 2017;
15. Abate-Daga D, Davila ML. CAR models: next-generation CAR modifi- 377(26):2531-2544.
cations for enhanced T-cell function. Mol Ther Oncolyt. 2016;3:16014. 37. Garfall AL, Maus MV, Hwang WT, et al. Chimeric antigen receptor
16. Stone JD, Kranz DM. Role of T cell receptor affinity in the efficacy T cells against CD19 for multiple myeloma. N Engl J Med. 2015;373
and specificity of adoptive T cell therapies. Front Immunol. 2013; (11):1040-1047.
4:244. 38. Ramos CA, Savoldo B, Torrano V, et al. Clinical responses with T lym-
17. Kershaw MH, Westwood JA, Parker LL, et al. A phase I study on adop- phocytes targeting malignancy-associated kappa light chains. J Clin
tive immunotherapy using gene-modified T cells for ovarian cancer. Invest. 2016;126(7):2588-2596.
Clin Cancer Res. 2006;12(20 Pt 1):6106-6115. 39. Ormhoj M, Bedoya F, Frigault MJ, Maus MV. CARs in the lead against
18. Lamers CH, Sleijfer S, Vulto AG, et al. Treatment of metastatic renal multiple myeloma. Curr Hematol Malig Rep. 2017;12(2):119-125.
cell carcinoma with autologous T-lymphocytes genetically retargeted 40. Stroncek DF, Ren J, Lee DW, et al. Myeloid cells in peripheral blood
against carbonic anhydrase IX: first clinical experience. J Clin Oncol. mononuclear cell concentrates inhibit the expansion of chimeric anti-
2006;24(13):e20-e22. gen receptor T cells. Cytotherapy. 2016;18(7):893-901.
19. Park JR, Digiusto DL, Slovak M, et al. Adoptive transfer of chimeric 41. Ruella M, Xu J, Barrett DM, et al. Induction of resistance to chimeric
antigen receptor re-directed cytolytic T lymphocyte clones in patients antigen receptor T cell therapy by transduction of a single leukemic B
with neuroblastoma. Mol Ther. 2007;15(4):825-833. cell. Nat Med. 2018;24(10):1499-1503.
FEINS ET AL. S9

42. Scholler J, Brady TL, Binder-Scholl G, et al. Decade-long safety and donors and untreated chronic lymphocytic leukemia patients. Front
function of retroviral-modified chimeric antigen receptor T cells. Sci Immunol. 2017;8:1956.
Transl Med. 2012;4(132):132ra153. 58. Kagoya Y, Nakatsugawa M, Yamashita Y, et al. BET bromodomain
43. Hacein-Bey-Abina S, Garrigue A, Wang GP, et al. Insertional oncogenesis inhibition enhances T cell persistence and function in adoptive immu-
in 4 patients after retrovirus-mediated gene therapy of SCID-X1. J Clin notherapy models. J Clin Invest. 2016;126(9):3479-3494.
Invest. 2008;118(9):3132-3142. 59. Tyrakis PA, Palazon A, Macias D, et al. S-2-hydroxyglutarate regulates
44. Howe SJ, Mansour MR, Schwarzwaelder K, et al. Insertional mutagen- CD8(+) T-lymphocyte fate. Nature. 2016;540(7632):236-241.
esis combined with acquired somatic mutations causes leukemogene- 60. Porter DL, Hwang WT, Frey NV, et al. Chimeric antigen receptor T
sis following gene therapy of SCID-X1 patients. J Clin Invest. 2008; cells persist and induce sustained remissions in relapsed refractory
118(9):3143-3150. chronic lymphocytic leukemia. Sci Transl Med. 2015;7(303):303ra139.
45. Modlich U, Navarro S, Zychlinski D, et al. Insertional transformation of 61. Fraietta JA, Beckwith KA, Patel PR, et al. Ibrutinib enhances chimeric
hematopoietic cells by self-inactivating lentiviral and gammaretroviral antigen receptor T-cell engraftment and efficacy in leukemia. Blood.
vectors. Mol Ther. 2009;17(11):1919-1928. 2016;127(9):1117-1127.
46. Montini E, Cesana D, Schmidt M, et al. The genotoxic potential of ret- 62. Rossi J, Paczkowski P, Shen YW, et al. Preinfusion polyfunctional anti-
roviral vectors is strongly modulated by vector design and integration CD19 chimeric antigen receptor T cells are associated with clinical
site selection in a mouse model of HSC gene therapy. J Clin Invest. outcomes in NHL. Blood. 2018;132(8):804-814.
2009;119(4):964-975. 63. Del Nagro CJ, Otero DC, Anzelon AN, Omori SA, Kolla RV, Rickert RC.
47. Naldini L, Blomer U, Gallay P, et al. In vivo gene delivery and stable CD19 function in central and peripheral B-cell development. Immunol
transduction of nondividing cells by a lentiviral vector. Science. 1996; Res. 2005;31(2):119-131.
272(5259):263-267. 64. Chung EY, Psathas JN, Yu D, Li Y, Weiss MJ, Thomas-Tikhonenko A.
48. Eyquem J, Mansilla-Soto J, Giavridis T, et al. Targeting a CAR to the CD19 is a major B cell receptor-independent activator of MYC-driven
TRAC locus with CRISPR/Cas9 enhances tumour rejection. Nature. B-lymphomagenesis. J Clin Invest. 2012;122(6):2257-2266.
2017;543(7643):113-117. 65. Sotillo E, Barrett DM, Black KL, et al. Convergence of acquired muta-
49. Roth TL, Puig-Saus C, Yu R, et al. Reprogramming human T cell func- tions and alternative splicing of CD19 enables resistance to CART-19
tion and specificity with non-viral genome targeting. Nature. 2018; immunotherapy. Cancer Discov. 2015;5(12):1282-1295.
559(7714):405-409. 66. Jacoby E, Nguyen SM, Fountaine TJ, et al. CD19 CAR immune pressure
50. Gattinoni L, Lugli E, Ji Y, et al. A human memory T cell subset with induces B-precursor acute lymphoblastic leukaemia lineage switch
stem cell-like properties. Nat Med. 2011;17(10):1290-1297. exposing inherent leukaemic plasticity. Nat Commun. 2016;7:12320.
51. Gregory TK, Berdeja JG, Patel KK, et al. Clinical trial of P- 67. Orlando EJ, Han X, Tribouley C, et al. Genetic mechanisms of target
BCMA-101 T stem cell memory (Tscm) CAR-T cells in relapsed/- antigen loss in CAR19 therapy of acute lymphoblastic leukemia. Nat
refractory (r/r) multiple myeloma (MM). Cancer Res. 2018;78(13): Med. 2018;24(10):1504-1506.
CT130. 68. Wang X, Popplewell LL, Wagner JR, et al. Phase 1 studies of central
52. Turtle CJ, Hanafi LA, Berger C, et al. CD19 CAR-T cells of defined memory-derived CD19 CAR T-cell therapy following autologous
CD4+:CD8+ composition in adult B cell ALL patients. J Clin Invest. HSCT in patients with B-cell NHL. Blood. 2016;127(24):2980-2990.
2016;126(6):2123-2138. 69. Singh N, Perazzelli J, Grupp SA, Barrett DM. Early memory pheno-
53. Klebanoff CA, Scott CD, Leonardi AJ, et al. Memory T cell-driven types drive T cell proliferation in patients with pediatric malignancies.
differentiation of naive cells impairs adoptive immunotherapy. J Clin Sci Transl Med. 2016;8(320):320ra323.
Invest. 2016;126(1):318-334. 70. Chong EA, Melenhorst JJ, Lacey SF, et al. PD-1 blockade modulates
54. Ghassemi S, Nunez-Cruz S, O'Connor RS, et al. Reducing ex vivo cul- chimeric antigen receptor (CAR)-modified T cells: refueling the CAR.
ture improves the antileukemic activity of chimeric antigen receptor Blood. 2017;129(8):1039-1041.
(CAR) T cells. Cancer Immunol Res. 2018;6(9):1100-1109.
55. Bai Y, Kan S, Zhou S, et al. Enhancement of the in vivo persistence
and antitumor efficacy of CD19 chimeric antigen receptor T cells
through the delivery of modified TERT mRNA. Cell Discov. 2015;1:
How to cite this article: Feins S, Kong W, Williams EF,
15040.
56. Kawalekar OU, O'Connor RS, Fraietta JA, et al. Distinct signaling Milone MC, Fraietta JA. An introduction to chimeric antigen
of coreceptors regulates specific metabolism pathways and receptor (CAR) T-cell immunotherapy for human cancer. Am
impacts memory development in CAR T cells. Immunity. 2016;44
J Hematol. 2019;94:S3–S9. https://doi.org/10.1002/ajh.
(2):380-390.
57. Hoffmann JM, Schubert ML, Wang L, et al. Differences in expansion 25418
potential of naive chimeric antigen receptor T cells from healthy

You might also like