You are on page 1of 11

The n e w e ng l a n d j o u r na l of m e dic i n e

Review Article

Dan L. Longo, M.D., Editor

Tolerance in the Age of Immunotherapy


Jeffrey A. Bluestone, Ph.D., and Mark Anderson, M.D., Ph.D.​​

T
From the Sean N. Parker Autoimmune he induction and maintenance of robust immune tolerance
Research Laboratory (J.A.B.) and the Dia- has been the holy grail of immunology for decades. In the absence of robust
betes Center (J.A.B., M.A.), University of
California, San Francisco, San Francisco. unresponsiveness of the immune system to self antigens (immune toler-
Address reprint requests to Dr. Bluestone ance), uncontrolled reactivity can lead to disorders like food allergies and autoim-
at the UCSF Diabetes Center, 513 Parnas- mune diseases. Although the first seminal experiments in tolerance were con-
sus Ave., Box 0540, San Francisco, CA
94143, or at ­jeff​.­bluestone@​­ucsf​.­edu. ducted in the 1950s,1 the development of novel tolerance-inducing therapeutic
drugs has been fraught with clinical challenges and few durable successes, in spite
N Engl J Med 2020;383:1156-66.
DOI: 10.1056/NEJMra1911109 of advances in our understanding of the fundamental aspects of the immune sys-
Copyright © 2020 Massachusetts Medical Society. tem. However, in recent years, new breakthroughs in our understanding of these
basic mechanisms and maintenance of immune tolerance have led to clinical suc-
cesses in the fields of organ transplantation and allergic and autoimmune dis-
eases. Moreover, novel peptide therapeutic drugs, anti–T-cell antibodies, and cell
therapies have set the stage for short-term treatments of autoimmune diseases that
have long-term efficacy and eliminate the need for continuous therapy.
During the past few decades, a more detailed understanding of the molecular
events associated with T-cell recognition and activation has advanced various ap-
proaches to tolerance, such as reprogramming, costimulatory blockade, check-
point inhibition, and antigen-specific immune regulation. The term “unresponsive-
ness” that has been associated with immune tolerance refers to a lack of
pathogenic immunity characterized by immune-cell inactivation or deletion or to
the diversion of pathogenic immunity to protective immunity through the engage-
ment of regulatory cells, deviation in cell differentiation, or development of im-
mune barriers. Thus, implicit in current approaches to the development of tolero-
genic drugs is the assumption that successful therapies would treat and prevent
allergic and autoimmune diseases, as well as lead to immunosuppression-free
organ and stem cell–derived tissue transplantation and protein-replacement ap-
proaches in congenital diseases such as hemophilia. Many of the most recent
therapeutic successes involve novel drugs that have been designed to break toler-
ance to cancers that are dependent on tumor-specific and microenvironment-
mediated tolerogenic signals. However, these treatments can lead to autoimmune
syndromes, underscoring the delicate balance between breaking tolerance to treat
tumors and altering immune homeostasis systemically.

Mech a nisms of Im mune T ol er a nce


The Thymus and Central Tolerance
In the early 1960s, Jacques Miller and Max Cooper independently defined two
distinct types of immune cells, T and B cells, that are the hallmark of the antigen-
specific adaptive immune system.2,3 T cells orchestrate immune responses both
indirectly, by providing soluble and membrane-associated signals that promote the
survival, expansion, and differentiation of B cells (which produce antibodies that

1156 n engl j med 383;12  nejm.org  September 17, 2020

The New England Journal of Medicine


Downloaded from nejm.org at WEILL CORNELL MEDICAL COLLEGE LIBRARY on September 16, 2020. For personal use only. No other uses without permission.
Copyright © 2020 Massachusetts Medical Society. All rights reserved.
Toler ance in the Age of Immunother apy

Glossary

Autoimmune regulator (AIRE): A protein that binds to chromatin and regulates the process of gene transcription such
that a plethora of self-proteins are ectopically expressed in medullary thymic epithelial cells (mTECs) involved in
thymic selection.
Bystander suppression: Immunosuppression in a local environment through direct cell-to-cell contacts or short-range
cytokines that are independent of specific antigen reactivity.
Infectious tolerance: Tolerance that results when forkhead box P3 (FOXP3)–positive regulatory T cells (Tregs) convert
conventional T cells into peripherally derived Tregs through secretion of the suppressive cytokines transforming
growth factor β (TGF-β), interleukin-10, or interleukin-35 or indirectly through the alteration of antigen-presenting
cells.
In vitro–induced Tregs (iTregs): Tregs that can clearly be distinguished from Treg populations generated in vivo; iTregs
can be induced by cytokines such as TGF-β, altered antigen-presenting cells, or costimulatory blockade.
Medullary thymic epithelial cells (mTECs): An epithelial-cell population present in the thymic medulla that expresses
AIRE protein and is responsible for negative selection and Treg development.
Peripherally derived Tregs (pTregs): Tregs that develop in tissue sites from conventional CD4+ T cells as a consequence
of exposure to certain cytokines, microbial products, or altered antigen-presenting cells.
T-cell receptor (TCR) repertoire: The diverse use of unique TCR alpha and beta chains to recognize individual antigen–
major histocompatibility complexes.
Thymus-derived Tregs (tTregs): Tregs induced as a consequence of negative selection, in which high-affinity, self-reac-
tive T cells develop into FOXP3-positive Tregs.
Toll-like receptors (TLRs): A class of proteins, usually expressed on macrophages and dendritic cells, that recognize
structurally conserved molecules derived from microbes to activate the innate immune system.

support productive humoral immunity), and di- 1010 distinct receptor combinations and is key to
rectly, by killing foreign and infected tissues the ability of the T-cell population as a whole to
through cellular and soluble mediators.3 T cells recognize the vast array of potential targets (see
recognize foreign antigens through a unique, the Glossary). However, in each person, this
highly diverse set of T-cell receptors (TCRs) de- repertoire is restricted and skewed as each T cell
signed to mediate immunity without the collat- is selected for its ability to bind a self-peptide in
eral damage of destroying native tissues. In a the context of a polymorphic and polygenic com-
parallel system, regulatory T cells (Tregs) recog- ponent of its ligand, molecules encoded by the
nize self-peptides and, when activated, control major histocompatibility complex (MHC). This
self-reactive pathogenic T cells. This complex, selection step, termed positive selection, ensures
dynamic process of self–nonself discrimination that T cells are able to recognize a foreign pep-
is the basis of immune tolerance. In this review, tide antigen bound to proteins encoded by spe-
we focus on TCR αβ cells in immune tolerance, cific self MHC alleles, most commonly MHC
given their dual role in pathogenicity and im- class I and class II molecules for CD8+ and
mune suppression, and on the wealth of new CD4+ T cells, respectively.4 (Details are provided
approaches being developed to induce or break in note 1 in the Supplementary Appendix, avail-
tolerance by targeting TCR αβ cells and the anti- able with the full text of this article at NEJM.org.)
gen-presenting cells that drive their function. After positive selection, T cells pass through
The thymus is the birthplace of T cells. Bone an important second selection step, called nega-
marrow–derived CD34+ stem cells migrate to the tive selection. This is a filtering step to remove
thymus, where they differentiate and acquire the T cells that have a strong binding affinity to
expression of the TCR. Each T cell expresses its self-peptides that are bound to the same MHC
own unique receptor, which is composed of a molecules and are consequently autoreactive. Such
heterodimer of two chains (TCRα and TCRβ), T cells arise by chance through the stochastic
each of which is generated through somatic re- gene-rearrangement process that generated the
combination of multiple genetic elements, includ- TCR and, thus, could potentially lead to autoim-
ing the addition of a few nucleotides at the site munity. To accomplish negative selection, a small,
of recombination. This combinatorial diversity resident cell subset, termed medullary thymic epi-
results in a broad TCR repertoire with more than thelial cells (mTECs), and bone marrow–derived

n engl j med 383;12  nejm.org  September 17, 2020 1157


The New England Journal of Medicine
Downloaded from nejm.org at WEILL CORNELL MEDICAL COLLEGE LIBRARY on September 16, 2020. For personal use only. No other uses without permission.
Copyright © 2020 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

A B

Antigen- Antigen-
presenting cell presenting cell

CD86 CD80 CD80 CD86


MHC MHC
Inhibitory
Peptide Peptide
Signal 1 Signal 2 signal
TCR TCR
CD28 CTLA-4

T cell CTLA-4 T cell CD28

Inactivation
Activation Inhibition of proliferation and cytokine
Proliferation production by T cells
Cytokine production T-cell exhaustion and apoptosis
Prevention of anergy Altered antigen-presenting-cell function
Differentiation of helper T cells and suppressive cytokine production

Figure 1. Two-Signal Models of Costimulatory and Inhibitory Pathways.


Initiation of a productive T-cell response involves integration of a primary signal delivered through the T-cell recep-
tor (TCR) and major histocompatibility complex (MHC)–peptide, followed by a second signal delivered through the
CD28–CD80 or CD28–CD86 pathway (Panel A). After initiation of T-cell activation, other inhibitory checkpoint inter-
actions can shut down T-cell activity (Panel B). Pathways that may be affected as a consequence of both positive and
negative second signals are listed at the bottom of the figure. CTLA-4 denotes cytotoxic T-lymphocyte–associated
protein 4.

dendritic cells5 interact with the maturing T cells. Peripheral Tolerance


The mTECs express a transcriptional activator As efficient as the thymus is in eliminating self-
called the autoimmune regulator (AIRE), which reactive cells, many self-reactive T cells escape
enables the expression of thousands of other- thymic negative selection, which leads to the
wise tissue-restricted proteins, so that peptides, need for peripheral mechanisms to ensure that
derived from these proteins, are displayed to self-tolerance is maintained.8 A variety of cell
developing T cells; this process results in robust types and processes control peripheral tolerance,
elimination of self-reactive CD4+ and CD8+ con- including cells of the adaptive and innate im-
ventional T cells.6 The key role of AIRE-positive mune system and signaling components within
mTECs in central tolerance is indicated by the T cells and antigen-presenting cells themselves.9
clinical manifestation of a profound, multiorgan Effective T-cell signaling requires both engage-
autoimmune syndrome, called autoimmune poly- ment of the primary antigen-specific receptor and
glandular syndrome type 1 (APS1), in patients a second, costimulatory signal to induce prolif-
with mutations in AIRE.7 eration, differentiation, and survival.10 A consti-

1158 n engl j med 383;12 nejm.org September 17, 2020

The New England Journal of Medicine


Downloaded from nejm.org at WEILL CORNELL MEDICAL COLLEGE LIBRARY on September 16, 2020. For personal use only. No other uses without permission.
Copyright © 2020 Massachusetts Medical Society. All rights reserved.
Toler ance in the Age of Immunother apy

tutive T-cell–surface molecule, CD28, was the


first T-cell costimulatory receptor to be identi- CD28 CD80
fied (Fig. 1).11 The ligands CD80 and CD86 are +
expressed selectively on antigen-presenting cells, CD86
CTLA-4
especially after activation through innate ago- –
nists of toll-like receptors (TLRs) and soluble
PD-1 PDL1 or L2
factors. Only cells that recognize nominal anti- –
gen — often termed “signal one” to reflect the TIM-3
first signal delivered to T cells during an activa- –
tion event — respond to a second costimulatory BTLA HVEM

signal essential for complete T-cell activation. Antigen-
T cell presenting cell
Blockade of costimulatory pathways leads to an TCR–CD3 MHC-pep
antigen-specific apoptotic cell death, clonal in- +
activation, and tolerance induction.12 Studies in
CD70 CD27
animal models have shown that costimulatory +
blockade with monoclonal antibodies and soluble CD154 CD40
+ +
forms of the CD80 and CD86 high-affinity re-
ceptor, cytotoxic T-lymphocyte–associated pro- + OX40L OX40
tein 4 (CTLA-4), induces tolerance in the context ICOS B7RP-1
of autoimmune disorders or organ transplanta- +
tion.12 Identification of additional costimulatory CD137L CD137
+
pathways, such as CD154–CD40, CD11A–CD54,
CD18–CD54, and CD2–CD58, has validated and
extended the two-signal model of T-cell activa- Cytokines
(TGFβ, interleukin-1, interleukin-6, interleukin-10, interleukin-12, interleukin-18)
tion. These findings have provided new thera-
peutic opportunities for blocking autoreactive
T-cell activity and inducing long-term tolerance Figure 2. Additional Costimulatory and Checkpoint Pathways.
without the need for continuous therapy.13 In addition to the two-signal models of costimulatory and checkpoint path-
Additional controls on the surface of activated ways, additional stimulatory and inhibitory pathways (indicated by plus and
T cells — so-called negative regulators, or check- minus signs, respectively) influence the immune response, including mole-
cules of the tumor necrosis factor (TNF)–related family, other members of
points — are as important as costimulatory
the CD28 family, adhesion molecules, and T-cell immunoglobulin and mu-
pathways in controlling T-cell activation. How- cin (TIM) molecules. The various stimulatory and inhibitory pathways can
ever, unlike costimulatory pathways, checkpoints, influence and be influenced by cytokines. Pep denotes peptide, and TGF-β
including CTLA-4 and programmed death 1 transforming growth factor β.
(PD-1),14 shut down immune activation when
engaged by their ligands, which leads to active
tolerance induction (Fig. 1). Direct evidence of a ance is the presence of specialized cell popula-
role for checkpoints in tolerance has been shown tions designed to suppress pathogenic immune
in cases in which checkpoint pathways have responses that inadvertently target self-tissue.
been inhibited. For instance, gene ablation or These include naturally occurring Treg cells and
treatment with checkpoint inhibitors can exacer- in vitro–induced Treg (iTreg) cells, as well as
bate autoimmunity and, in some cases, break interleukin-10–producing type 1 regulatory T (Tr1)
tolerance induced by other therapies.15 Thus, cells and transforming growth factor β (TGF-β)–
targeting of checkpoint pathways has resulted in producing type 3 helper T (Th3) cells.16 Of these
a new generation of tolerance-manipulating drugs subsets, the most extensively studied are Tregs,
(Fig. 2). Inhibition of checkpoint pathways has a subset of the self-reactive CD4+ T cells that
revolutionized cancer immunotherapy by turn- develop as a consequence of the expression of a
ing once deadly cancers such as melanoma and master transcriptional repressor, forkhead box
non–small-cell lung cancer into treatable dis- P3 (FOXP3). FOXP3 alters the differentiation of
eases.14 Checkpoint agonists are being used in the mature CD4+ T-cell population into this
the treatment of autoimmune disease and organ- regulatory T-cell subset, which plays a funda-
transplant rejection, as described below. mental role in immune homeostasis.17 Func-
Another key mechanism of peripheral toler- tional disruption of FOXP3, either genetically (in

n engl j med 383;12 nejm.org September 17, 2020 1159


The New England Journal of Medicine
Downloaded from nejm.org at WEILL CORNELL MEDICAL COLLEGE LIBRARY on September 16, 2020. For personal use only. No other uses without permission.
Copyright © 2020 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

Scurfy mice and patients with the IPEX [immune producing Th3 cells, may provide the broadest
dysregulation, polyendocrinopathy, enteropathy, antigen-recognition repertoire for controlling
X-linked] syndrome) or pharmacologically, leads pathogenic self-reactivity. Moreover, because of
to severe autoimmune disorders that cause death thymic involution in adulthood, the development
at a very young age unless the patient is given a and maintenance of tolerance may differ accord-
bone marrow transplant.18 ing to age, with peripheral pathways having a
The primary source of Tregs is the thymus, more important regulatory role in autoimmunity
where the cells are generated by an alternative in adults than in children25 (note 2 in the Supple-
developmental pathway. During negative selec- mentary Appendix).
tion, a subset of developing T cells expressing Antigen-presenting cells, including tolerogenic
TCRs with high affinity develop into Treg pre- dendritic cells, immature macrophages, suppres-
cursors that up-regulate FOXP3, leading to a sor antigen-presenting cells of the myeloid lin-
stable epigenetic state and resulting in a mature, eage, and even certain B-cell subsets, function
self-reactive population of thymus-derived Tregs in conjunction with Tregs and other suppressor
(tTregs) that populate lymphoid and nonlym- T cells to control immunity. These cells develop
phoid tissues in the periphery.19,20 The role of in response to a variety of cell-surface and sol-
Tregs in the periphery is to halt self-reactivity uble factors, including those controlled by
and promote tissue repair and regeneration Tregs.26-28 For instance, Tregs express high levels
(Fig. 3).21,22 AIRE-expressing mTECs play a role of the checkpoint CTLA-4, which can block
in the generation of tTregs through presentation CD28-mediated costimulation and deliver in-
of tissue-specific antigens. Moreover, AIRE is hibitory signals that change antigen-presenting
expressed in a subset of bone marrow–derived cells into tolerogenic cells.29,30 Tregs, as well as
cells in peripheral lymphoid organs and, thus, other regulatory cells, produce cytokines, such
may influence tolerance in peripheral tissues.23 as interleukin-10, interleukin-35, TGF-β, and
This finding highlights the connection between other soluble factors involved in metabolism,
peripheral tolerance and T-cell development in such as indoleamine 2,3-dioxygenase. The Tregs
the thymus. can induce myeloid-derived suppressor cells and
Tregs are generated in the immune periphery alter antigen presentation.27,31 Tregs express CD39
as well, under conditions in which naive CD4+ and CD73, which affect the duration, magni-
T cells encounter antigen in the context of sup- tude, and chemical nature of purinergic signals
pressive factors such as TGF-β, interleukin-10, delivered to immune cells through the conver-
bacterially derived metabolic products, or altered sion of ADP or ATP to adenosine.32 Finally,
stimulatory pathways. Under certain conditions, metabolic products of the gut microbiome, in-
antigen presentation leads to the induction of cluding the production of short-chain fatty acids
stable FOXP3 and transformation of conven- such as butyrate, can have a profound effect on
tional T cells into peripherally derived Tregs immune function. These and other regulatory
(pTregs).24 Thus, unlike tTregs, which develop in factors can act on antigen-presenting cells to
the thymus from T cells undergoing negative promote the generation of Tr1 cells and Tregs or
selection based on high self-reactivity, pTregs directly affect pathogenic T cells, altering dif-
develop from a conventional peripheral T-cell ferentiation, trafficking, and function. Oral tol-
repertoire selected for low self-reactivity. A grow- erance studies with short-chain fatty acids sup-
ing number of examples show that Tregs recog- port a strong interface between the immune
nize commensal bacteria, which have many system and resident microbiota33 (Fig. 4).
characteristics normally attributed to self-anti- Thus, it is clear that a dynamic, homeostatic
gens. This process diversifies the Treg repertoire, immune system has evolved to deal with endog-
which may be most effective in shutting down enous and exogenous insults. There is a balanc-
inflammatory responses. Moreover, pTregs may ing act between the need to develop potent effec-
recognize modified proteins, such as citrulli- tor cells in order to combat foreign pathogens
nated peptides, hybrid peptides, and phosphory- and the need for homeostatic control of the
lated proteins, which are often present in auto- immune system to shut down unwanted autoin-
immune states but not in the thymus. Together, flammation, as has been reported in some pa-
the combination of tTregs and pTregs, as well as tients with coronavirus disease 2019 (Covid-19)34
interluekin-10–producing Tr1 cells and TGF-β– and autoimmunity.

1160 n engl j med 383;12  nejm.org  September 17, 2020

The New England Journal of Medicine


Downloaded from nejm.org at WEILL CORNELL MEDICAL COLLEGE LIBRARY on September 16, 2020. For personal use only. No other uses without permission.
Copyright © 2020 Massachusetts Medical Society. All rights reserved.
Toler ance in the Age of Immunother apy

A Pathways in Thymus

AIRE-expressing
mTEC T cell

T cell
FOXP3
tTreg

T cell Naive
T cells

Apoptosis of
T cells

B Pathways in Immune Periphery

Naive
CD4+ T cell
CD4+
effector T cell AIRE-expressing
eTAC

Antigen-
presenting cell
CD4+
Antigen- effector T cell
Conventional macrophages presenting cell
and dendritic cells
CD8+
effector T cell

Tolerogenic macrophages
and dendritic cells

CD8+
effector T cell
Apoptosis
of effector FOXP3
T cells pTreg

Pathogenic Protective

Figure 3. Tolerance-Inducing Pathways in the Thymus and Periphery.


The majority of cells interacting with autoimmune regulator (AIRE)–expressing medullary thymic epithelial cells (mTECs) during thymic
development undergo negative selection and die (Panel A). A subset of high-affinity, self-reactive CD4+ T cells interact with the mTECs,
leading to the development of regulatory T cells (Tregs). The remaining mature naive T cells migrate into the immune periphery, where
they have either a pathogenic role in mediating immunity (Panel B, left) or a protective role as peripherally derived Tregs (through inter-
action with tolerogenic antigen-presenting cells and cytokines) that control potential autoreactive responses (Panel B, right). Additional
cell types, such as extrathymic AIRE–expressing cells (eTACs), can also modify potentially autoreactive T cells.

Inducing T ol er a nce tion has been the use of broadly immunosup-


pressive drugs, which carry substantial risks of
Historically, the only option for patients with serious side effects. Approved therapies, such as
autoimmune diseases or organ-transplant rejec- calcineurin inhibitors, glucocorticoids, and tumor

n engl j med 383;12 nejm.org September 17, 2020 1161


The New England Journal of Medicine
Downloaded from nejm.org at WEILL CORNELL MEDICAL COLLEGE LIBRARY on September 16, 2020. For personal use only. No other uses without permission.
Copyright © 2020 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

to the tolerogenic antigen during regeneration


Microbiome
and recovery of the immune system. Autologous
CD4+ HST has been reported to halt the progression
Suppressive factors effector T cell
of certain forms of autoimmunity, such as multi-
SCFAs ple sclerosis.35 Combining autologous and donor-
derived HST can result in lasting tolerance to the
donor MHC while allowing effective immune
Antigen- reconstitution (i.e., mixed chimerism).36,37 The
Interleukin-2
presenting cell Bystander immune-cell depletion approach to rebooting
suppression
the immune system has been enabled by the use
of broad immune-depleting agents such as alem-
tuzumab (anti-CD52 antibody), B-cell–depleting
Treg agents (rituximab, ocrelizumab, and obinutuzu-
Th3 mab), and antilymphocyte antibody therapies.38-41
Tr1
These drugs have been successful in slowing the
progression of disease, in some cases through
the elimination of antigen presentation by the
CD4+ Treg
autoreactive B cells, which are very efficient in
Infectious
effector T cell tolerance Th3 capturing and presenting autoantigens.38,42 How-
Tr1 ever, both autologous HST and the broad cell-
depleting strategies lead to the elimination of
immune-cell subsets, including those involved in
Figure 4. Activation and Functional Consequences of Suppressive Cells. protection from infection and, potentially, cancer.
Tregs and other suppressive cells circulate and reside in lymphoid and so-
matic tissues to control unwanted autoimmune and inflammatory respons- Targeting Pathogenic T Cells
es. Multiple cell–cell contacts, as well as soluble molecules (including the
production of metabolites by microbiota), are generated by Tregs or anti-
In the past several years, treatments have been
gen-presenting cells after Treg interactions to control immunity. Tregs can developed to selectively target the highly acti-
also act through bystander suppression, leading to dominant local immu- vated, pathogenic T cells involved in mediating
nosuppression and tolerance induction. IDO denotes indoleamine 2,3-di- tissue-reactive immunity. These treatments in-
oxygenase, SCFA short-chain fatty acid, Th3 type 3 helper T cell, and Tr1 clude an anti-CD3 monoclonal antibody (teplizu-
type 1 regulatory T cell.
mab), a soluble form of the CD2 costimulatory
receptor LFA-3 (alefacept), and low-dose antithy-
mocyte globulin.40,43,44 These biologics have been
necrosis factor α and interleukin-1 antagonists, shown to induce apoptosis and functional inac-
require continuous treatment and target the end tivation of highly activated effector cells, leaving
stage of the immune dysfunction rather than the naive T cells and Tregs intact or even expanding
induction of tolerance. An improved understand- the regulatory pathways. This class of therapeu-
ing of the molecular and functional basis for tic drugs has been shown to be efficacious in a
immune tolerance has driven the development variety of autoimmune diseases, even when pa-
of tolerogenic drugs that may fundamentally tients were treated for only a short period of
change the therapeutic landscape. time. In the case of teplizumab, Herold and col-
leagues reported that the median time until dis-
Resetting the Immune System ease onset was prolonged by more than 3 years
The most effective way to generate immune tol- and the median disease-free survival was 5 or
erance would be to reset and rebalance the im- more years among patients who were at risk for
mune system in order to prevent the develop- type 1 diabetes and who were treated for only
ment and progression of the autoimmune 2 weeks.45,46 The therapy not only delays the de-
response. In this regard, two approaches — autol- velopment of disease but also may induce more
ogous hematopoietic stem-cell transplantation sustained tolerance in some patients. In trials
(HST) and massive immune-cell depletion — conducted by the Immune Tolerance Network,
have been pioneered. Both of these methods are one or two courses of teplizumab given over a
intended to induce tolerance through exposure 3-month period to patients with newly diagnosed

1162 n engl j med 383;12 nejm.org September 17, 2020

The New England Journal of Medicine


Downloaded from nejm.org at WEILL CORNELL MEDICAL COLLEGE LIBRARY on September 16, 2020. For personal use only. No other uses without permission.
Copyright © 2020 Massachusetts Medical Society. All rights reserved.
Toler ance in the Age of Immunother apy

type 1 diabetes led to prolonged preservation of than block) the key checkpoints and inactivate
c-peptide (insulin) production, with no increase pathogenic T cells.
in rates of infection or cancer. A significant per- The third approach, the induction of antigen-
centage of residual CD8+ effector T cells ex- specific tolerance with the use of autoantigen
pressed an exhausted or anergic T-cell pheno- therapeutic drugs, has been the most challeng-
type that has been shown to be the major ing.55-58 These therapies are potentially the most
marker of tumor tolerance.47 These results high- selective in eliminating autoreactive pathogenic
light the importance of targeting pathogenic T cells while reducing the risks of infections and
T cells to induce tolerance while maintaining cancer, which make broadly immunosuppressive
naive T cells and regulatory pathways to preserve agents problematic. However, the requirement for
immune competence and homeostasis. Moreover, knowledge of the autoantigen, the large number
a combination of these therapies, as well as the of pathogenic epitopes, and the consequence of
pro-Treg drugs described below, provides an op- epitope spreading (which expands the number of
portunity to rebalance the immune system, since pathogenic epitopes) complicate these deletional
it is likely that control of autoimmunity requires therapies. Such therapies have nevertheless been
both a decreased number of pathogenic T cells successful in the treatment of immune respons-
and increased regulatory activity. es to allergens, such as peanuts, dust mites, and
The second approach to tolerance induction certain grasses, and metabolic deficiencies, such
with more selective tolerogenic therapeutic drugs as hemophilia,59,60 in which the uncontrolled
has been the development of inhibitors of the inflammatory responses to the proteins after
costimulatory pathway, as noted above and exposure can prevent the use of these lifesaving
shown in Figures 1 and 2. Two soluble CD28 treatments. In a seminal proof-of-principle study,
antagonists, abatacept and belatacept (CTLA-4– newborn children at high risk for atopic aller-
IgG1 and mutant CTLA-4–IgG1, respectively), gies were randomly assigned to be exposed to
have been approved by the Food and Drug Ad- peanuts or to have no exposure over a 5-year
ministration for the treatment of rheumatoid period. A majority of the children who were ex-
arthritis and kidney-transplant rejection, respec- posed to peanuts had a higher sustained reduc-
tively.48,49 Unfortunately, the ability of these tion in the incidence of peanut allergy than
drugs to induce protracted tolerance in humans those who had not been exposed.61 At a mecha-
is unclear. In a clinical trial involving patients nistic level, tolerance was due in part to immune
with newly diagnosed type 1 diabetes, treatment deviation from a pathogenic IgE response to a
with abatacept for 6 months resulted in signifi- nonpathogenic IgG response.
cant prolongation of insulin production, as com- In an equally dramatic demonstration of the
pared with insulin production in the control power of antigens to induce tolerance under the
group, but this protection diminished over time.50 right conditions, allograft tolerance has been
One reason for the decline in the efficacy of the maintained after withdrawal of immunosuppres-
drug may have been its depletion of Tregs, sive drugs in a subset of liver-transplant recipi-
which may be essential for the development of ents, presumably because of the constant expo-
tolerance.12,51 Treg development and survival are sure of alloantigen in a protolerogenic hepatic
dependent on CD28 signaling in vivo.52 A sec- environment.62 These successes have expanded
ond generation of costimulatory antagonists to the use of peptides, either administered alone
(including those that block CD154–CD40 inter- intravenously or coupled to cells, nanoparticles,
actions) that may synergize with CD28 costim- or other multimeric scaffolds; DNA vaccines,
ulatory blockade are currently being evaluated which incorporate both antigenic peptides and
in a number of clinical settings.53 The substan- tolerogenic therapeutics to modify TCR recogni-
tial effect of antibodies directed at negative tion; immobilized HLA–peptide complexes; and
regulators in cancer immune therapy, including tolerogenic dendritic cells pulsed with multiple
multiple antibodies against PD-1 and its ligand, peptides.63 Peptide-induced tolerance has often
PD-L1, and antibodies against CTLA-4 to pro- been associated with the induction of Tregs or
mote activation signals to T-cells, opens up a tolerogenic dendritic cells. Uptake of antigen
new area for tolerance induction.54 New treat- through scavenger receptors, such as DEC-205,
ments are being developed to activate (rather SR-A, or MARCO, may alter the cell phenotype

n engl j med 383;12  nejm.org  September 17, 2020 1163


The New England Journal of Medicine
Downloaded from nejm.org at WEILL CORNELL MEDICAL COLLEGE LIBRARY on September 16, 2020. For personal use only. No other uses without permission.
Copyright © 2020 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

and function and, in some cases, down-regulate and life span of these regulatory cells by insert-
costimulatory ligands. Thus, peptide therapies tap ing specific TCRs or chimeric antigen receptors
into some of the most basic aspects of T-cell and using gene editing to modify durability and
activation. The safety of antigen-specific thera- stability, alter trafficking, and enhance tissue
peutic approaches has been shown in multiple repair of the adoptively transferred cells.
phase 1 clinical trials using several autoantigenic Additional potentially tolerogenic approaches
peptides from multiple proteins, and controlled using antigen-presenting cells are also being
phase 2 trials of efficacy are now under way.64,65 tested in the clinic, including the use of mesen-
chymal stromal cells73 and dendritic cells.74,75
Targeting Regulatory Pathways Each approach has advantages and disadvantages
Defects in or defective regulation of key immune with respect to the potential for “off-the-shelf”
cells such as tolerogenic FOXP3-positive Treg cells treatments, the ability to modify the cell therapy
has been documented in several types of human with current genetic approaches, and the mecha-
autoimmunity, which suggests that enhanced nism (direct vs. bystander) of suppression. Clin-
Treg numbers, increased Treg functioning, or ical-grade tolerogenic dendritic cells, generated
both might stop autoimmunity. Several approach- in vitro, are being tested in autoimmune and
es have been undertaken to increase or enhance organ-transplant settings to induce clonal dele-
Treg numbers and activity in patients with auto- tion of pathogenic T cells in vivo or to induce the
immune disorders. These approaches include generation of antigen-specific regulatory T and
treatment with low-dose interleukin-2 and inter- B cells.76-78 All these approaches are focused on the
leukin-2 mutants to selectively increase Treg induction of active, dominant, antigen-specific
numbers and functioning, short-term combina- tolerance and thus avoid the need to delete all
tion therapies that promote the development of T cells against potential pathogenic specificities.
regulatory cells, such as rapamycin (sirolimus), Finally, other cell therapies are being tested
and the coadministration of tolerogenic peptides, to treat autoantibody-mediated autoimmunity, in-
as described above.66-69 One of the more active cluding the use of genetically engineered effec-
research areas has been the administration of ex tor T cells transduced with a chimeric antigen
vivo–generated, tolerogenic cellular therapies, receptor that is directed at the cell-surface CD19
which can result in a shutdown of autoimmu- molecule. On adoptive transfer, the T cells are
nity, potentially lasting for years.15,70 Treg ther- triggered by CD19 on B cells, which induces
apy takes advantage of two distinct factors that cytolytic activity and B-cell destruction.79,80 An-
enhance tolerance: bystander suppression and other approach is the expression of autoantigen
infectious tolerance. These mechanisms enable (target protein) chimerized to the signal domains
Tregs to suppress the immune response widely of a typical chimeric antigen receptor (a so-called
within a local environment and create a tolero- chimeric autoantigen receptor), which when put
genic environment in which other local cells into CD8+ T cells, leads to the destruction of
take on a tolerogenic phenotype, which leads to B cells and plasma cells expressing a receptor
broadening of the number and specificities of that recognizes the autoantigen.79 These cells are
the cells involved in the control of unwanted im- being introduced into clinical practice and will
munity. More than 50 active and completed soon be assessed for their safety and efficacy in
clinical trials are testing the safety and efficacy a variety of autoantibody-mediated diseases.
of Treg cell therapy for indications such as kid-
ney or liver transplantation, pemphigus vulgaris, C onclusions
systemic lupus erythematosus, inflammatory
bowel disease, autoimmune hepatitis, allergy, Despite the genetic predisposition observed in
and asthma. In patients with chronic graft-versus- the majority of autoimmune diseases, the inabil-
host disease, Treg-cell therapy alleviated symp- ity to predict the development of autoimmunity,
toms, and pharmacologic immunosuppression coupled with the likelihood that disease has al-
could be reduced.71 In addition to Tregs, a sec- ready developed by the time patients come to
ond suppressive T-cell population, Tr1 cells, is clinical attention, means that treatment is usu-
being used therapeutically to induce tolerance.72 ally initiated after the onset of disease. Efforts
Efforts are under way to increase the potency are under way to predict the onset of diseases

1164 n engl j med 383;12  nejm.org  September 17, 2020

The New England Journal of Medicine


Downloaded from nejm.org at WEILL CORNELL MEDICAL COLLEGE LIBRARY on September 16, 2020. For personal use only. No other uses without permission.
Copyright © 2020 Massachusetts Medical Society. All rights reserved.
Toler ance in the Age of Immunother apy

such as rheumatoid arthritis and type 1 diabetes. combination therapies will be required to estab-
Individual tolerogenic therapies may be more lish long-term tolerance.
effective if they can be used before sufficient Disclosure forms provided by the authors are available with
the full text of this article at NEJM.org.
tissue has been damaged to develop a disease We thank Dr. Gerald Nepom at the Benaroya Institute for
phenotype.45 Achieving durable tolerance is like- kindly reading and advising on an earlier version of the manu-
ly to require eliminating or regulating disease script and the members of the Bluestone and Anderson labora-
tory for their work, which has resulted in many of the ideas
effectors and repairing damaged tissue. The com- highlighted in this review, and for their ongoing advice and
plexity of the process suggests that the use of commitment to the discovery effort.

References
1. Billingham RE, Brent L, Medawar PB. PD-1–PD-L1 pathway. J Exp Med 2006;​ 30. Walker LSK, Sansom DM. Confusing
Actively acquired tolerance of foreign cells. 203:​2737-47. signals: recent progress in CTLA-4 biol-
Nature 1953;​172:​603-6. 16. Ferreira LMR, Muller YD, Bluestone ogy. Trends Immunol 2015;​36:​63-70.
2. Cooper MD, Peterson RD, Good RA. JA, Tang Q. Next-generation regulatory 31. Josefowicz SZ, Lu L-F, Rudensky AY.
Delineation of the thymic and bursal lym- T cell therapy. Nat Rev Drug Discov 2019;​ Regulatory T cells: mechanisms of differ-
phoid systems in the chicken. Nature 1965;​ 18:​749-69. entiation and function. Annu Rev Immu-
205:​143-6. 17. Rudensky AY. Regulatory T cells and nol 2012;​30:​531-64.
3. Miller JFAP. The golden anniversary Foxp3. Immunol Rev 2011;​241:​260-8. 32. Antonioli L, Pacher P, Vizi ES, Haskó
of the thymus. Nat Rev Immunol 2011;​11:​ 18. Ochs HD, Gambineri E, Torgerson G. CD39 and CD73 in immunity and in-
489-95. TR. IPEX, FOXP3 and regulatory T-cells: flammation. Trends Mol Med 2013;​ 19:​
4. Klein L, Kyewski B, Allen PM, Hogquist a model for autoimmunity. Immunol Res 355-67.
KA. Positive and negative selection of the 2007;​38:​112-21. 33. Pandiyan P, Bhaskaran N, Zou M,
T cell repertoire: what thymocytes see (and 19. Aschenbrenner K, D’Cruz LM, Voll- Schneider E, Jayaraman S, Huehn J. Micro-
don’t see). Nat Rev Immunol 2014;​14:​377- mann EH, et al. Selection of Foxp3+ regu- biome dependent regulation of Tregs and
91. latory T cells specific for self antigen ex- Th17 cells in mucosa. Front Immunol
5. van Meerwijk JPM, Marguerat S, Lees pressed and presented by Aire+ medullary 2019;​10:​426.
RK, Germain RN, Fowlkes BJ, MacDonald thymic epithelial cells. Nat Immunol 2007;​ 34. Mehta P, McAuley DF, Brown M,
HR. Quantitative impact of thymic clonal 8:​351-8. Sanchez E, Tattersall RS, Manson JJ.
­
deletion on the T cell repertoire. J Exp 20. Malchow S, Leventhal DS, Lee V, Nishi ­COVID-19: consider cytokine storm syn-
Med 1997;​185:​377-83. S, Socci ND, Savage PA. Aire enforces im- dromes and immunosuppression. Lancet
6. Anderson MS, Venanzi ES, Klein L, mune tolerance by directing autoreactive 2020;​395:​1033-4.
et al. Projection of an immunological self T cells into the regulatory T cell lineage. 35. Nash RA, Hutton GJ, Racke MK, et al.
shadow within the thymus by the aire Immunity 2016;​44:​1102-13. High-dose immunosuppressive therapy
protein. Science 2002;​298:​1395-401. 21. Josefowicz SZ, Niec RE, Kim HY, et al. and autologous HCT for relapsing-remit-
7. Husebye ES, Anderson MS, Kämpe O. Extrathymically generated regulatory ting MS. Neurology 2017;​88:​842-52.
Autoimmune polyendocrine syndromes. T cells control mucosal TH2 inflamma- 36. Busque S, Scandling JD, Lowsky R,
N Engl J Med 2018;​378:​1132-41. tion. Nature 2012;​482:​395-9. et al. Mixed chimerism and acceptance of
8. Theofilopoulos AN, Kono DH, Baccala 22. Arpaia N, Green JA, Moltedo B, et al. kidney transplants after immunosuppres-
R. The multiple pathways to autoimmu- A distinct function of regulatory T cells sive drug withdrawal. Sci Transl Med
nity. Nat Immunol 2017;​18:​716-24. in tissue protection. Cell 2015;​162:​1078- 2020;​12(528):​eaax8863.
9. Bluestone JA, Bour-Jordan H, Cheng 89. 37. Spitzer TR, Sykes M, Tolkoff-Rubin N,
M, Anderson M. T cells in the control of 23. Gardner JM, Devoss JJ, Friedman RS, et al. Long-term follow-up of recipients
organ-specific autoimmunity. J Clin Invest et al. Deletional tolerance mediated by ex- of combined human leukocyte antigen-
2015;​125:​2250-60. trathymic Aire-expressing cells. Science matched bone marrow and kidney trans-
10. Salomon B, Bluestone JA. Complexi- 2008;​321:​843-7. plantation for multiple myeloma with
ties of CD28/B7: CTLA-4 costimulatory 24. Abbas AK, Benoist C, Bluestone JA, end-stage renal disease. Transplantation
pathways in autoimmunity and transplan- et al. Regulatory T cells: recommendations 2011;​91:​672-6.
tation. Annu Rev Immunol 2001;​19:​225- to simplify the nomenclature. Nat Immu- 38. Sabatino JJ Jr, Zamvil SS, Hauser SL.
52. nol 2013;​14:​307-8. B-cell therapies in multiple sclerosis. Cold
11. Weiss A, Manger B, Imboden J. Syn- 25. Ander SE, Diamond MS, Coyne CB. Spring Harb Perspect Med 2019;​ 9(2):​
ergy between the T3/antigen receptor com- Immune responses at the maternal-fetal a032037.
plex and Tp44 in the activation of human interface. Sci Immunol 2019;​4(31):​eaat6114. 39. Hersh CM, Cohen JA. Alemtuzumab
T cells. J Immunol 1986;​137:​819-25. 26. Rosser EC, Mauri C. Regulatory B cells: for the treatment of relapsing-remitting
12. Esensten JH, Helou YA, Chopra G, origin, phenotype, and function. Immu- multiple sclerosis. Immunotherapy 2014;​
Weiss A, Bluestone JA. CD28 costimula- nity 2015;​42:​607-12. 6:​249-59.
tion: from mechanism to therapy. Immu- 27. Talmadge JE, Gabrilovich DI. History 40. Haller MJ, Long SA, Blanchfield JL,
nity 2016;​44:​973-88. of myeloid-derived suppressor cells. Nat et al. Low-dose anti-thymocyte globulin
13. Sharpe AH. Mechanisms of costimu- Rev Cancer 2013;​13:​739-52. preserves C-peptide, reduces HbA1c, and
lation. Immunol Rev 2009;​229:​5-11. 28. Belkaid Y, Hand TW. Role of the mi- increases regulatory to conventional T-cell
14. Chen DS, Mellman I. Elements of can- crobiota in immunity and inflammation. ratios in new-onset type 1 diabetes: two-
cer immunity and the cancer-immune set Cell 2014;​157:​121-41. year clinical trial data. Diabetes 2019;​68:​
point. Nature 2017;​541:​321-30. 29. Wing K, Onishi Y, Prieto-Martin P, 1267-76.
15. Fife BT, Guleria I, Gubbels Bupp M, et al. CTLA-4 control over Foxp3+ regula- 41. Gitelman SE, Gottlieb PA, Felner EI,
et al. Insulin-induced remission in new- tory T cell function. Science 2008;​ 322:​ et al. Antithymocyte globulin therapy for
onset NOD mice is maintained by the 271-5. patients with recent-onset type 1 diabetes:

n engl j med 383;12  nejm.org  September 17, 2020 1165


The New England Journal of Medicine
Downloaded from nejm.org at WEILL CORNELL MEDICAL COLLEGE LIBRARY on September 16, 2020. For personal use only. No other uses without permission.
Copyright © 2020 Massachusetts Medical Society. All rights reserved.
Toler ance in the Age of Immunother apy

2 year results of a randomised trial. Dia- into anti-CD40/CD154 immunotherapy in 67. Klatzmann D, Abbas AK. The prom-
betologia 2016;​59:​1153-61. transplant tolerance. Immunotherapy 2015;​ ise of low-dose interleukin-2 therapy for
42. Arneth BM. Impact of B cells to the 7:​399-410. autoimmune and inflammatory diseases.
pathophysiology of multiple sclerosis. 54. Paluch C, Santos AM, Anzilotti C, Nat Rev Immunol 2015;​15:​283-94.
J Neuroinflammation 2019;​16:​128. Cornall RJ, Davis SJ. Immune checkpoints 68. Saadoun D, Rosenzwajg M, Joly F, et al.
43. Rigby MR, Harris KM, Pinckney A, as therapeutic targets in autoimmunity. Regulatory T-cell responses to low-dose
et al. Alefacept provides sustained clinical Front Immunol 2018;​9:​2306. interleukin-2 in HCV-induced vasculitis.
and immunological effects in new-onset 55. Miller SD, Turley DM, Podojil JR. Anti- N Engl J Med 2011;​365:​2067-77.
type 1 diabetes patients. J Clin Invest 2015;​ gen-specific tolerance strategies for the 69. Koreth J, Matsuoka K, Kim HT, et al.
125:​3285-96. prevention and treatment of autoimmune Interleukin-2 and regulatory T cells in
44. Herold KC, Gitelman SE, Ehlers MR, disease. Nat Rev Immunol 2007;​7:​665-77. graft-versus-host disease. N Engl J Med
et al. Teplizumab (anti-CD3 mAb) treat- 56. Serra P, Santamaria P. Antigen-specif- 2011;​365:​2055-66.
ment preserves C-peptide responses in pa- ic therapeutic approaches for autoimmu- 70. Bluestone JA, Tang Q. Treg cells — the
tients with new-onset type 1 diabetes in nity. Nat Biotechnol 2019;​37:​238-51. next frontier of cell therapy. Science 2018;​
a randomized controlled trial: metabolic 57. Roep BO, Wheeler DCS, Peakman M. 362:​154-5.
and immunologic features at baseline Antigen-based immune modulation ther- 71. Koreth J, Kim HT, Jones KT, et al. Ef-
identify a subgroup of responders. Diabe- apy for type 1 diabetes: the era of preci- ficacy, durability, and response predictors
tes 2013;​62:​3766-74. sion medicine. Lancet Diabetes Endocri- of low-dose interleukin-2 therapy for
45. Herold KC, Bundy BN, Long SA, et al. nol 2019;​7:​65-74. chronic graft-versus-host disease. Blood
An anti-CD3 antibody, teplizumab, in 58. Wraith DC. The future of immuno- 2016;​128:​130-7.
relatives at risk for type 1 diabetes. N Engl therapy: a 20-year perspective. Front Im- 72. Gregori S, Roncarolo MG. Engineered
J Med 2019;​381:​603-13. munol 2017;​8:​1668. T tegulatory type 1 cells for clinical ap-
46. Sims EK, Bundy BN, Stier KD, et al. 59. Maldonado RA, LaMothe RA, Ferrari plication. Front Immunol 2018;​9:​233.
277-OR: Teplizumab reverses the loss of JD, et al. Polymeric synthetic nanoparti- 73. Rasmusson I. Immune modulation by
C-peptide in relatives at risk for type 1 cles for the induction of antigen-specific mesenchymal stem cells. Exp Cell Res
diabetes (T1D). Diabetes 2020;​69:​Suppl 1 immunological tolerance. Proc Natl Acad 2006;​312:​2169-79.
(https://doi​.­org/​­10​.­2337/​­db20​-­277​-­OR). Sci U S A 2015;​112(2):​E156-E165. 74. Takenaka MC, Quintana FJ. Tolero-
47. Wherry EJ, Kurachi M. Molecular and 60. Adair P, Su Y, Scott DW. Tolerance in- genic dendritic cells. Semin Immuno-
cellular insights into T cell exhaustion. duction in hemophilia A animal models: pathol 2017;​39:​113-20.
Nat Rev Immunol 2015;​15:​486-99. battling inhibitors with antigen-specific 75. Maldonado RA, von Andrian UH.
48. Vincenti F, Rostaing L, Grinyo J, et al. immunotherapies. Discov Med 2013;​15:​ How tolerogenic dendritic cells induce
Belatacept and long-term outcomes in kid- 275-82. regulatory T cells. Adv Immunol 2010;​
ney transplantation. N Engl J Med 2016;​ 61. Du Toit G, Roberts G, Sayre PH, et al. 108:​111-65.
374:​333-43. Randomized trial of peanut consumption 76. Naranjo-Gómez M, Raïch-Regué D,
49. Bathon J, Robles M, Ximenes AC, et al. in infants at risk for peanut allergy. N Engl Oñate C, et al. Comparative study of clini-
Sustained disease remission and inhibi- J Med 2015;​372:​803-13. cal grade human tolerogenic dendritic
tion of radiographic progression in meth- 62. Feng S, Bucuvalas J. Tolerance after cells. J Transl Med 2011;​9:​89.
otrexate-naive patients with rheumatoid liver transplantation: where are we? Liver 77. Beriou G, Moreau A, Cuturi MC.
arthritis and poor prognostic factors Transpl 2017;​23:​1601-14. Tolerogenic dendritic cells: applications
treated with abatacept: 2-year outcomes. 63. Luo X, Miller SD, Shea LD. Immune for solid organ transplantation. Curr Opin
Ann Rheum Dis 2011;​70:​1949-56. tolerance for autoimmune disease and Organ Transplant 2012;​17:​42-7.
50. Orban T, Bundy B, Becker DJ, et al. cell transplantation. Annu Rev Biomed 78. Flórez-Grau G, Zubizarreta I, Cabezón
Costimulation modulation with abatacept Eng 2016;​18:​181-205. R, Villoslada P, Benitez-Ribas D. Tolero-
in patients with recent-onset type 1 diabe- 64. Steinman L, Ho PP, Robinson WH, genic dendritic cells as a promising anti-
tes: follow-up 1 year after cessation of Utz PJ, Villoslada P. Antigen-specific tol- gen-specific therapy in the treatment of
treatment. Diabetes Care 2014;​37:​1069- erance to self-antigens in protein replace- multiple sclerosis and neuromyelitis op-
75. ment therapy, gene therapy and autoim- tica from preclinical to clinical trials.
51. Glatigny S, Höllbacher B, Motley SJ, munity. Curr Opin Immunol 2019;​ 61:​ Front Immunol 2018;​9:​1169.
et al. Abatacept targets T follicular helper 46-53. 79. Ellebrecht CT, Bhoj VG, Nace A, et al.
and regulatory T cells, disrupting molec- 65. Wraith DC. Designing antigens for Reengineering chimeric antigen receptor
ular pathways that regulate their prolifera- the prevention and treatment of autoim- T cells for targeted therapy of autoim-
tion and maintenance. J Immunol 2019;​ mune diseases. Curr Opin Chem Eng 2018;​ mune disease. Science 2016;​353:​179-84.
202:​1373-82. 19:​35-42. 80. Kansal R, Richardson N, Neeli I, et al.
52. Tang Q, Henriksen KJ, Boden EK, et al. 66. Peterson LB, Bell CJM, Howlett SK, et al. Sustained B cell depletion by CD19-tar-
Cutting edge: CD28 controls peripheral A long-lived IL-2 mutein that selectively geted CAR T cells is a highly effective
homeostasis of CD4+CD25+ regulatory activates and expands regulatory T cells treatment for murine lupus. Sci Transl
T cells. J Immunol 2003;​171:​3348-52. as a therapy for autoimmune disease. Med 2019;​11(482):​eaav1648.
53. Pinelli DF, Ford ML. Novel insights J Autoimmun 2018;​95:​1-14. Copyright © 2020 Massachusetts Medical Society.

1166 n engl j med 383;12  nejm.org  September 17, 2020

The New England Journal of Medicine


Downloaded from nejm.org at WEILL CORNELL MEDICAL COLLEGE LIBRARY on September 16, 2020. For personal use only. No other uses without permission.
Copyright © 2020 Massachusetts Medical Society. All rights reserved.

You might also like