Vous êtes sur la page 1sur 15

REVIEWS

Food allergy: immune mechanisms,


diagnosis and immunotherapy
Wong Yu13*, Deborah M.Hussey Freeland1,2* and Kari C.Nadeau13
Abstract | Food allergy is a pathological, potentially deadly, immune reaction triggered by normally
innocuous food protein antigens. The prevalence of food allergies is rising and the standard of care
is not optimal, consisting of food-allergen avoidance and treatment of allergen-induced systemic
reactions with adrenaline. Thus, accurate diagnosis, prevention and treatment are pressing needs,
research into which has been catalysed by technological advances that are enabling a mechanistic
understanding of food allergy at the cellular and molecular levels. We discuss the diagnosis and
treatment of IgE-mediated food allergy in the context of the immune mechanisms associated with
healthy tolerance to common foods, the inflammatory response underlying most food allergies,
and immunotherapy-induced desensitization. We highlight promising research advances,
therapeutic innovations and the challenges that remain.

The atopic diseases tend to occur in a progression termed States increased by 18% to an estimated prevalence of
Atopic diseases
Disorders characterized by the atopic march13, in which the initial manifestation 3.9%8, and prevalence had reached ~5% by 2011 (REF.9).
pathological immune of atopic disease in infancy or early childhood is often Another study of 38,480 children under 18years of age
responses to normally atopic dermatitis, followed by the staggered development in the United States yielded an estimated food allergy
innocuous antigens. of food allergy, allergic rhinitis and allergic asthma. Food prevalence of 8%, as well as the findings that approxi-
Oral food challenge
allergy is a pathological reaction of the immune system mately 40% of patients with food allergy have experi-
(OFC). Used for food allergy triggered by the ingestion of a food protein antigen. enced a life-threatening allergic reaction, and 30% of
diagnosis. Under supervision, Exposure to very small amounts of allergenic foods can children with food allergy have multiple food allergies10.
the patient is asked to consume trigger clinical symptoms such as gastrointestinal dis- Increases in the numbers of food allergy anaphylaxis-
small but increasing amounts of
orders, urticaria and airway inflammation, ranging in related hospital admissions and fatalities may reflect this
the suspected food allergen
until a predetermined maximum severity from mild to life-threatening. Food allergy is apparent increase in food allergy prevalence, although
dose is reached or afood distinct from food intolerance in that intolerance does other factors (such as increased recognition of anaphy-
allergic reaction isobserved. not arise from immune system dysregulation; for exam- laxis, increased severity of food allergy or increased
ple, lactose intolerance arises from non-immune factors, exposure of allergic individuals to food allergens) may
such as lactose malabsorption and lactase deficiency. also contribute. Between 1998 and 2012, the number of
Food allergy is both common and costly. In a recently food allergy anaphylaxis-related hospital admissions
1
Sean N.Parker Center for
published epidemiological study defining a retrospective increased by 137% in children 14years and younger in
Allergy and Asthma Research,
Stanford University, cross-sectional cohort of 333,200 children in the United England and Wales without an increase in the number
2
Division of Pulmonary States, food allergy prevalence was found to be 6.7%4. of fatalities11. Between 1997 and 2013, the number of
andCritical Care Medicine, A global survey of food allergy in children found that food allergy anaphylaxis-related deaths increased by an
Department of Medicine, only 9 of the 89 countries surveyed had accurate preva- average of 9.7% per year in Australia12. The apparent
Stanford University,
3
Division of Allergy,
lence data determined by oral food challenge (OFC), 7 of increase in food allergy prevalence over a short time
Immunology and which reported a prevalence ranging from 0.45% to 10% period suggests that environmental factors have a role
Rheumatology, Department among children less than 5 years old; the remaining 28 in its aetiology. Although the mechanisms by which
of Medicine, Stanford countries that had food allergy prevalence data relied environmental factors could promote food allergy are
University, Stanford,
on methods such as self-reporting, which is known to not well understood, recent findings indicate that envi-
California 94305, USA.
overestimate food allergy prevalence5. Annually, food ronmental exposures may interfere with the normal
*These authors contributed
equally to this work.
allergy results in costs to the US healthcare system of ability of the immune system to promote tolerance to
Correspondence to K.C.N. approximately $24.8billion6. In addition, the global foodallergens1315.
knadeau@stanford.edu prevalence of all food allergy seems to be increasing 7. In light of the prevalence and costs of food allergy,
doi:10.1038/nri.2016.111 Between 1997 and 2007, the self-reported prevalence of effective methods of prevention and treatment would be
Published online 31 Oct 2016 food allergy in children under 18years old in the United of substantial clinical value. The current standard of care

NATURE REVIEWS | IMMUNOLOGY VOLUME 16 | DECEMBER 2016 | 751



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

is food-allergen avoidance and the treatment of food Variants of IgE-mediated food allergy include
allergen-induced systemic reactions with adrenaline16. oralallergy syndrome (OAS), in which individuals
Recent advances in food allergy research have led to the with allergic rhinitis produce IgE molecules specific
development of new therapies, many of which are cur- for pollen-derived epitopes that are crossreactive with
rently in clinical trials. This Review presents a summary fruit- or vegetable-protein epitopes22. Signs and symp-
of food allergy classification and mechanisms of dis- toms of OAS include immediate oral pruritus, mucosal
ease, diagnostic methods, strategies for prevention and angioedema and/or abdominal pain. As the crossreactive
treatment, and future directions in food allergy research. IgE antibodies may be specific for a heat-labile food
epitope, uncooked fruits or vegetables should be used
Classification of food allergies in diagnostic testing for OAS. In the context of peanut
Food allergies are atopic disorders, which can be broadly allergy, testing for IgE antibodies specific for particular
classified into those that are IgE mediated, those that are components of peanut protein (IgE componenttesting) can
mediated by both IgE-dependent and IgE-independent be used to distinguish OAS from standard IgE-mediated
pathways (mixed), and those that are not IgE mediated allergy: in individuals with birch-pollen allergy, IgE
(TABLE1). Food allergies are mechanistically distinct from antibodies specific for the pollen antigen Bet v 1 may
non-atopic disorders, such as coeliac disease. crossreact with the peanut component Ara h 8 to cause
OAS23, but reactivity against Ara h 8 is also associated
IgE-mediated food allergies. This class of food allergy with a decreased risk of clinically relevant IgE-mediated
is associated with the risk of severe or fatal reactions; peanut allergy; by contrast, reactivity against the peanut
accordingly, it is the most fully characterized type of food components Ara h1 or Arah2 is more likely to be
allergy and is the main focus of this Review. Whereas IgE- associated with clinically relevant IgE-mediated peanut
mediated allergies to cows milk, egg, wheat and soy tend allergy 24. IgE- and Tcell-mediated crossreactivities have
to be outgrown, atopic responses to peanuts, tree nuts also been observed between Betv1 and the hazelnut
and shellfish usually persist into adulthood17. The most allergen Cor a 1 (REF.25).
common IgE-mediated food allergies vary somewhat Another variant of IgE-mediated food allergy occurs
by region and with dietary habits; for example, children in individuals who produce IgE antibodies that are
in Ghana between 5 and 16years of age are commonly specific for the red meat carbohydrate galactose-1,3
allergic to pineapple, pawpaw, orange and mango, as well galactose, rather than specific for a protein epitope26. As
as to peanut 18, whereas children in North America are patients with this type of food allergy have usually con-
commonly allergic to peanut, milk, egg, shellfish and sumed red meat uneventfully for many years, it has been
soy 4. The most common food allergen in Asia is shell- proposed that sensitivity is initiated through tick bites,
fish; in most Asian countries wheat allergy is uncommon, via exposure to antigens transmitted in the saliva of ticks
but inJapan and Korea it is the leading cause of anaphy- that have fed on mammals26. Although these reactions
laxis19. In Singapore, allergy to birds nest was the leading can be severe, they are unlike other IgE-mediated allergic
cause of anaphylaxis in children more than 15years ago, reactions to food in that symptom onset is delayed by
whereas peanut allergy is currently the most common 46hours or longer 26,27.
type of foodallergy 19.
In food allergen-sensitized individuals those Mixed food allergies. This class of food allergy is char-
who have been exposed to the allergen and had an ini- acterized by both IgE-dependent and IgE-independent
tial immune response subsequent exposure to the pathways. Atopic manifestations arising from IgE-
food allergen triggers IgE-mediated degranulation of independent factors include delayed food-allergy-
immune effector cells, such as mast cells and basophils, associated atopic dermatitis (648hours post exposure28)
resulting in the rapid manifestation of symptoms. Food caused by the action of Thelper 2 (TH2) cells29, and
allergen-derived epitopes attach to IgE molecules bound eosinophilic gastrointestinal disorders, such as eosino
to FcRI receptors on the surface of these effector cells; philic oesophagitis (EoE), which are often triggered
epitope-specific crosslinking of IgE-bound receptors then by milk allergens and caused by the eosinophilic infil-
occurs, leading to the release of preformed histamine and tration of tissues30,31. Further research is underway to
other inflammatory mediators of the immediate allergic discern the possible role of food allergies in these con-
reaction20. After this immediate phase of the response, the ditions and to assess the relative contributions of IgE-
denovo production of leukotrienes, platelet activating fac- dependent and -independent pathways in individuals
tor and cytokines such as interleukin4 (IL4), IL5 and with thesedisorders.
IL13 maintains allergic inflammation20. Gastrointestinal
manifestations can include oral tingling, pruritus and/or Non-IgE-mediated food allergies. Most of the known
swelling, as well as nausea, abdominal pain and/or vom- non-IgE-mediated food allergies primarily affect the
iting. Respiratory effects include wheezing and/or airway gastrointestinal tract, rather than the skin and respira-
IgE component testing inflammation. Skin manifestations include flushing, urti- tory tracts. For example, allergen-specific Tcells are
Purified allergen extracts or caria, angioedema and/or pruritus. Systemic responses thought to have roles in the largely unknown aetiolo-
recombinant allergens are used may also occur, such as hypotension due to fluid leakage gies of food protein-induced enterocolitis syndrome
to diagnose food allergies in a
more accurate manner than is
from the vasculature and/or hypothermia. Anaphylaxis (FPIES), food protein-induced proctocolitis (FPIP)
possible with crude allergen is a serious allergic reaction that involves multiple organ and food protein enteropathy (FPE)32. FPIES, FPIPand
extracts. systems and can rapidly become life-threatening 21. FPE primarily involve infants and toddlers who are

752 | DECEMBER 2016 | VOLUME 16 www.nature.com/nri



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Table 1 | Classification of food allergies


Subtype Prevalence Age group Common allergens Usual symptoms Diagnosis Treatment
affected
IgE-mediated food allergy
No subtype 0.410% Children Milk, egg, wheat, soy, Pruritus, urticaria, sIgE levels, SPT and OFC Standard:
> adults peanut, tree nuts, angioedema, food-allergen
shellfish and fish abdominal pain, avoidance and
vomiting, wheezing emergency
and hypotension medication
Research: OIT,
SLIT, EPIT and
omalizumab
Mixed IgE- and cell-mediated food allergy
Food-allergy- 2737% of patients Children Milk, egg, wheat, soy, Exacerbation sIgE levels, SPT and OFC Standard:
associated with atopic > adults peanut, tree nuts, of dermatitis food-allergen
atopic dermatitis183; shellfish and fish with allergen avoidance
dermatitis 1427% of patients ingestion (as an Research: OIT,
with self-reported addition to typical SLIT, EPIT and
atopic IgE-mediated food omalizumab
dermatitis184 allergy symptoms)
EoE Up to ~50 patients Children Milk, wheat, egg, beef, Vomiting, failure to Oesophageal biopsies Standard:
per 100,000 and adults soy and chicken186 thrive, dysphagia, showing eosinophil infiltrates topical
(REF.185) (3:1 male food impaction and after a 23month course of steroids (in the
to female heartburn protein pump inhibitors to oesophagus) or
ratio)185 exclude gastro-oesophageal food-allergen
reflux disease as a cause avoidance
Other Rare187 EC: infants EC: milk and soy Manifestations Increased numbers Standard:
eosinophilic EG: adults EG: possibly milk, vary with affected of eosinophils seen in steroids for
gastrointestinal > children wheat, soy, egg, nuts, gastrointestinal gastrointestinal biopsy; EC and EG;
disorders (EC, EGE: adults seafood and red tract region and eosinophils in ascites if avoidance of
EG or EGE) meats layer (mucosal, serosal layer affected; other, specific foods
EGE: may not have muscular or serosal) more common, causes of
food allergy aetiology eosinophilia must be ruled out
Non-IgE-mediated food allergy32
FPIES Few data: one Infants and Milk, soy, rice, oat and Intermittent Food-allergen avoidance and Standard:
study reports children egg allergen exposure: food challenge food-allergen
0.34% of infants severe vomiting avoidance
with FPIES to cows Chronic allergen
milk188 exposure:
diarrhoea and
failure to thrive
FPIP Few data: one Infants Milk, soy, wheat and Rectal bleeding Food-allergen avoidance and Standard:
study reports egg food challenge food-allergen
0.16% of infants avoidance
with potential FPIP
to cows milk189
FPE Few data Infants and Milk, soy, wheat and Steatorrhoea from Food-allergen avoidance and Standard:
toddlers egg malabsorption, food challenge together with food-allergen
diarrhoea and jejunal biopsy showing villous avoidance
failure to thrive atrophy and crypt hyperplasia
EC, eosinophilic colitis; EG, eosinophilic gastritis; EGE, eosinophilic gastroenteritis; EoE, eosinophilic oesophagitis; EPIT, epicutaneous immunotherapy; FPE, food
protein enteropathy; FPIES, food protein-induced enterocolitis syndrome; FPIP, food protein-induced proctocolitis; OFC, oral food challenge; OIT, oral immunotherapy;
sIgE, allergen-specific IgE; SLIT, sublingual immunotherapy; SPT, skin prick test.

allergic to cows milk, and they commonly resolve after unresponsiveness to common food antigens occurs
1 to 5years32. Their prevalence is uncertain owing to in non-food-allergic (in other words, healthy or
a lack of diagnostic tests, and treatment consists of immune-tolerant) individuals. In individuals with food
food-allergen avoidance. allergy, sensitization to food allergens results in inappro
priate inflammatory immune responses to common
Immune mechanisms foods. Many clinical trials in food allergy aim to establish
For overall health, it is essential that the immune functional immune tolerance in place of this sensitiza-
system distinguishes pathogenic antigens from innoc- tion. This is defined by clinical endpoints in a clinical
uous environmental antigens. Accordingly, a state of trial after a period of withdrawal from therapy, otherwise

NATURE REVIEWS | IMMUNOLOGY VOLUME 16 | DECEMBER 2016 | 753



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

known as sustained unresponsiveness. These studies under some circumstances, CD103+ DCs may capture
involve food allergen immunotherapy, in which an indi- antigens directly from the lumen via periscoping behav-
vidual with food allergy is exposed to initially small but iour (extending a process through a tight junction) or by
gradually increasing amounts of allergenic food proteins extending a process through a transcellular pore in an
over many months, so that the immune system slowly Mcell38. An estimated 2% of proteins pass through the
adjusts to the exposure, and the probability of an aller- gut epithelium intact and could be transported directly
gic reaction decreases. Immunotherapy often results in or indirectly to the liver or to secondary lymphoid
desensitization a temporary increase in the threshold tissues for antigen presentation by other cell types41,42.
for allergen reactivity instead of durable sustained CX3C-chemokine receptor 1 (CX3CR1)+ cells (proba-
unresponsiveness. The mechanisms of desensitization bly macrophages) extend dendrites between the epithelial
are not well understood and may differ from those of cells, sample antigens in the gut lumen and may promote
the healthy state of immune tolerance. Because it is tolerance through IL10 secretion43. These CX3CR1+ cells
unknown whether a patient with food allergy who has also seem to transfer captured antigens via gap junctions
been desensitized through immunotherapy will remain to CD103+ DCs, a subset of which migrates from the
permanently unresponsive to that food allergen, we lamina propria to the draining lymph nodes44. Here,
refer to sustained unresponsiveness as distinct from the the CD103+ DCs produce transforming growth factor
healthy, pre-allergic state of immune tolerance. (TGF) and retinoic acid, thereby inducing naive Tcells
The deduction of disease mechanisms lies at the fore- to differentiate into antigen-specific forkhead box P3
front of food allergy research. Technological advances, (FOXP3)+ regulatory T (Treg) cells35,4448. CD103+ DCs
such as high-throughput instruments for CyTOF may also induce the differentiation of naive CD4+ Tcells
(cytometry by time of flight mass spectrometry) and into FOXP3, IL10secreting T cells, possibly type1
single-cell quantitative PCR, that are capable of quantify- regulatory Tcells (Tr1 cells)35,49,50.
ing multiple parameters simultaneously have converged The cell-mediated trafficking of antigen to the sec-
with rapid advances in big data analytics to enable ondary lymphoid tissue promotes the establishment of
unprecedented discoveries in the signalling pathways tolerance: mice deficient in CCR7, which is required
underlying immune tolerance, sensitization and desensi for DC trafficking to secondary lymphoid tissue, have
tization to food allergens. This section examines our cur- impaired oral tolerance51. The relative importance of
rent understanding of the mechanisms involved in the the mesenteric lymph nodes, Peyers patches and other
progression of food allergy from health (tolerance), to types of secondary lymphoid tissue remains unclear5254,
disease (sensitization), to treatment (desensitization), although many reports have suggested that the mesen-
focusing mainly on research in humans but drawing teric lymph nodes are a major destination of migrating
from research in mouse models where relevant. DCs52,53. This hypothesis is supported by a mouse model
showing that the absence of mesenteric lymph nodes
Tolerance. The establishment of immune tolerance to impairs oral tolerance53. CD103+ DCs are thought to be
orally administered antigens is a subject of intensive one of the major DC subsets responsible for the devel-
study 33 (FIG.1). The mechanisms have been covered in opment of mucosal tolerance35. In addition, CCR9+ DCs
detail elsewhere34, and only the main points are high- have been identified in lymphoid tissues and have been
lighted here. The healthy immune system is tolerant to shown to induce Treg cell function, thereby suppressing
foods, characterized by a sustained (that is, memory) antigen-specific immune responses55.
unresponsiveness towards potential food allergens even Retinoic acid, produced by CD103+ DCs, also induces
Sustained unresponsiveness
The state in which an individual
in the absence of regular allergen exposure. Immune tol- Treg cell expression of integrin 47, which results in the
with food allergy no longer erance is an active process that begins with the uptake of homing of Treg cells to the gut where they may dampen
requires regular ingestion of potential food antigens in the small intestine, which con- the immune response49,50,56. In mouse models, the reti-
an allergen to maintain tains much of the gut-associated lymphoid tissue (GALT)35. noic acid-induced expression of integrin 47 by Tcells is
desensitization to it;
Diverse cell types collaborate in the series of events that required for the development of Tcell-mediated oral tol-
phenotypically, it resembles
the normal immune tolerance maintains immune tolerance, involving transmission of erance57. Peripherally induced regulatory Tcells return to
of a healthy individual but may antigen from the gut lumen to the lamina propria, trans- the lamina propria, where they maintain their regulatory
be mechanistically distinct. fer of antigen to the lymphoid tissue, antigen presenta- phenotype in response to IL10 secretion by CX3CR1+
tion and induction of a Tcell response in the lymphoid cells58,59. The presence of a large number of regulatory
Gut-associated lymphoid
tissue
tissue, and subsequent return of immune effector cells T cells in the gut (including FOXP3+ Treg cells and
(GALT). The immune system of to thegut. Tr1cells) depends upon the availability of food antigens60.
the gastrointestinal tract is Potential food antigens can be captured from the Further evidence supporting the role of Treg cells in
collectively called the GALT; gut lumen by passing between (paracytosis) or through oral tolerance is provided by human clinical studies
examples include the tonsils,
(transcytosis) epithelial cells3537. Specialized cells, such reporting hypomethylation (and thus increased accessi-
Peyers patches and lymphoid
cells in the lamina propria. as the M (microfold) cells that overlie the GALT, can bility for transcription) of the FOXP3 locus of Treg cells in
endocytose antigen38, and myeloid cells such as dendritic those patients who develop and maintain functional tol-
Type1 regulatory Tcells cells (DCs) and/or macrophages may capture potential erance in response to oral immunotherapy (OIT)61. Also,
(Tr1 cells). A subset of food antigens from the gut lumen39,40. For example, the IL4 receptor (IL4R) has been implicated in food
regulatory Tcells that are
FOXP3. Tr1 cells mediate
CD103+ DCs sample antigens that pass through the allergy pathogenesis, and IL4R inhibition may augment
suppressive effects through epithelial barrier via M cell-mediated transcytosis or Treg cell induction and the enforcement of tolerance, as
IL10 secretion. through translocation by mucin-secreting goblet cells; shown in mouse models62.

754 | DECEMBER 2016 | VOLUME 16 www.nature.com/nri



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Normal immune tolerance is not well understood. to the allergen was associated with the expansion of
A study comparing the responses of CD4+ Tcells to these protective Tcells63. Although food antigen-specific
the birch pollen allergen Bet v 1 in healthy and allergic lymphocytes are very rare in the peripheral blood, a
individuals found that allergen-specific CD4+ Tcells flow cytometry study compared peanut-specific cells
from healthy individuals secrete IFN and IL10, in among children who are allergic to peanuts, children
contrast to those from patients with allergy: tolerance with no peanut allergy, and those who have naturally

Lumen Filamentous
High microbial bacteria
Dimeric diversity
IgA Food
Intestinal Antigen allergen
epithelial Tuft cell sampling
cell M cell

Transcytosis
of antigen

CX3CR1+
macrophage Mast CD103+
cell
Antigen
transfer
B cell
IL-10
CD103+ production Peyers patch
DC
Treg cell Treg cell
proliferation

IL-10,
DC TGF
migration
CTLA4
Gut
homing
TH2 cell
47
Treg cell
Gut lamina
propria
Anergy Apoptosis
TGF and retinoic
acid production
IgA
Peptide IL-10,
MHC TGF
Draining
lymph node
Naive
CD4+ T cell TFH cell B cell

Figure 1 | Immune tolerance to oral antigens in the gut. CX3CR1 cells (most likely to be macrophages) extend dendrites
+

between the intestinal epithelial cells, sample antigens in the gut lumen59,162 and transfer captured Nature Reviews
antigens | Immunology
via gap junctions
to CD103+ dendritic cells (DCs). A subset of these DCs migrates from the lamina propria to the draining lymph nodes where the
DCs express transforming growth factor- (TGF) and retinoic acid, thereby inducing naive Tcells to differentiate into
regulatory T (Treg) cells44,46. Macrophages also seem to secrete interleukin10 (IL10), leading to Treg cell proliferation43,59,162;
however, this is debated163. Several types of regulatory T cell (resting, effector, and memory164) have been reported to be
associated with mucosal tolerance, including induced forkhead box P3 (FOXP3)+ Treg cells, IL10secreting Tr1 cells and
TGF-secreting T helper 3 (TH3) cells165. Retinoic acid also induces Treg cell expression of integrin 47, which results in homing
to the gut where Treg cells may dampen the immune response49,50,56. CD103+ DCs also sample antigens that pass through the
epithelial barrier via M cell-mediated transcytosis or through translocation by mucin-secreting goblet cells; under some
circumstances, CD103+ DCs may capture antigens from the lumen directly, via periscoping behaviour (extending a process
through a tight junction) or by extending a process through a transcellular pore in an M cell38. Bcell clones expressing antibody
specific for food allergen may undergo isotype switching in the secondary lymphoid organs with the aid of follicular Thelper
(TFH) cells. Food tolerance and allergen desensitization are associated with IgA (FIG.1) and IgG4 (FIG.3), respectively86,166.
Bycontrast, food allergen-specific IgE (FIG.2) will be bound by FcRI on mast cells (which are normally found in tissues forming
environmental barriers167) and basophils, thus leading to immediate hypersensitivity reactions to food. High-dose exposure to
oral antigens has been reported to lead to the anergy or deletion of antigen-specific Tcells, possibly after DC interaction168.
TFHcells secreting different cytokine combinations favour Bcell switch recombination to produce particular antibody isotypes,
whereas follicular Treg cells suppress the germinal centre reaction169,170. The roles of tissue-resident Tcells, CD8+ Tcells and
Tcells remain to be determined. The relationship between TFH cells and the conventional TH cell subsets is not clear,
andconversion between the two has been reported171,172. CTLA4, cytotoxic T lymphocyte antigen 4.

NATURE REVIEWS | IMMUNOLOGY VOLUME 16 | DECEMBER 2016 | 755



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

of peanut protein extract to undamaged skin in mice


Box 1 | Additional cell types involved in food allergy pathogenesis
leads to a TH2 type immune response through IL33
Tuft cells induction70, and mechanical injury to the skin results
These are component cells of the small intestinal epithelium that secrete interleukin25 in cutaneous TSLP expression and the elicitation of an
(IL25) to maintain the homeostasis of type2 innate lymphoid cells (ILC2s)153. Under inflammatory TH2 cell-mediated response71. The mech-
certain conditions, tuft cells can induce ILC2s to secrete IL13, thereby initiating a
anisms by which a TH2 cell-mediated response of cutan
Thelper 2 (TH2) cell-mediated response153.
eous origin can reach the gut are unclear. One possibility
Tcells is that DCs in the skin may secrete retinoic acid, which
Found in the intestinal epithelium and the lamina propria, these cells can modulate induces allergen-specific Tcells to express gut-homing
andcontribute to the breakdown of oral tolerance in mice, and may have a similar role
markers as they d
ifferentiate down the TH2cell pathway72.
in humans154.
CD8+ Tcells Desensitization. The immune changes that accompany
These cells may influence gastrointestinal manifestations of food allergy: increased food allergen immunotherapy are thought to occur as a
numbers of CD8+ Tcells have been observed in a mouse model of eosinophilic progression of several elements7375 (FIG.3). The mechan
oesophagitis155, and CD8+ Tcells are reported to suppress food allergen-triggered
isms by which mast cell and basophil responsiveness to
diarrhoea in another mouse model156.
degranulation are decreased early during immunotherapy
Epithelial cells are under investigation76. Other changes to the immune
Cells of the gut and respiratory mucosae and skin that secrete thymic stromal system observed after immunotherapy include increased
lymphopoietin (TSLP), IL25 and IL33; these cytokines tend to elicit a TH2 cell-mediated
numbers of anergic antigen-specific Tcells77, apoptotic
response through their actions on dendritic cells (DCs) and ILC2s157. Mutations in
filaggrin impair epithelial barrier function and can be associated with an increased risk
antigen-specific Tcells7881 and FOXP3+ Tregcells80,82,
of atopic dermatitis and food allergy119,120,158,159. aswell as increased levels ofTGF1 (REFS77,83). For
example, over the course of OIT, antigen-specific CD4+
Type2 innate lymphoid cells
Tcells were found to transition from allergic and regu
ILC2s can be influenced by TSLP, IL25 or IL33 to secrete IL5 and IL13, which promote
TH2 cell-mediated immune responses157,160.
latory phenotypes to anergic and non-allergic pheno-
types, in association with decreased allergic symptoms
Mast cells and the development of sustained unresponsiveness77.
Mast cells provide both immediate and delayed release of inflammatory mediators,
Allergen-specific immunotherapy has also been associ
and their activation is a hallmark of atopic disease20,66.
ated with the preferential deletion of allergen-specific
CD14+ monocytes TH2 cells, without significant concomitant changes in
These cells occur at an increased ratio to CD4+ Tcells in the cord blood of infants who the frequencies of TH1 and Tr1 cells78.
develop food allergies161. CD14+ monocytes are hyperresponsive to lipopolysaccharide In summary, Treg cells are likely to be a part of, but
(LPS), secreting increased levels of the inflammatory cytokines IL1, IL6 and tumour
not unique to, the mechanisms involved in immune
necrosis factor in response to LPS stimulation161.
tolerance. These observations are consistent with a
model involving a shift from allergen-specific TH2cells
outgrown a peanut allergy. Cells from peanut-allergic to another cell subset as the key event in sustained
individuals have a TH2-polarized cytokine production, unresponsiveness. This shift reduces the inflammatory
whereas those from the non-allergic and nolonger response of tissues that encounter food allergen, thereby
allergic groups have a TH1type cytokine response to mitigating the recruitment and/or activation of innate
peanutantigens64. immune cells such as mast cells and basophils84. Recently
published data in mouse models show some similarities
Sensitization. DCmediated tolerance to food anti- and differences among the mechanisms of desensitiza-
gens can be reprogrammed by inflammatory stimuli tion induced by OIT, sublingual immunotherapy (SLIT)
to give rise to food-allergic immune responses medi- and epicutaneous immunotherapy (EPIT), which seem
ated by TH2cells, as well as by additional cell types to induce different subsets of regulatory Tcells85.
(BOX1). Because several excellent reviews6567 address Other changes observed over the course of OIT
the immune mechanisms of sensitization in detail, we include those in allergen-specific IgE levels, which
touch only upon research concerning a pivotal step in decrease, whereas allergen-specific IgG4 levels increase86.
the postulated atopic march from cutaneous sensitiza- Although levels of all IgG subclasses rise during OIT
tion to food allergy. A current model of the induction of and may contribute differentially to the suppression of
Type2 innate lymphoid cells
a TH2cellmediated inflammatory immune response in IgE-mediated responses through binding FcRIIb, the
(ILC2s). Cells that produce the gut is given in FIG.2. increase in the ratio of IgG4 to IgE is particularly nota-
Thelper 2 cell cytokines, such Cutaneous exposure to food allergens may promote ble, and IgG4 has become an important research focus87.
as IL4, IL5, IL9 and IL13, sensitization. Inflammatory cytokines released by the Intests of basophil activation, sera from peanut-sensitive
and are implicated in the
skin epithelium, including IL25, IL33 and thymic stro- patients drawn at week 52 of OIT were found to sup-
development of allergic
inflammation. mal lymphopoietin (TSLP), act on DCs and other cells press the responsiveness to peanut antigens of basophils
to deviate the immune response towards TH2 cell-related from normal, non-peanut-allergic donors that had been
Sublingual immunotherapy allergic responses, rather than tolerogenic responses68. activated with allergic sera; when IgG was removed
(SLIT). An immunotherapeutic For example, the cutaneous sensitization of mice with from these sera, their suppressive effect was mitigated,
method in which the food
allergen (in the form of a tablet
ovalbumin (OVA) and TSLP, followed by repeated oral and this suppressive effect was restored when the IgG
or drops) is administered under challenge with OVA, results in IL25dependent diar- fractions were restored87. Intests of mast cell activation,
the tongue. rhoea and anaphylaxis69. Also, repeated application plasma from peanut-sensitive patients with detectable

756 | DECEMBER 2016 | VOLUME 16 www.nature.com/nri



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Lumen Food
Mucosal Increased Intestinal
damage allergen epithelial
antigen uptake Tuft cell
M cell cell

IgE IL-25,
Mast cell IL-33, IL-25
FcRI accumulation TSLP Cytokine
secretion
Mast Class-switching
cell B cell to IgE
IL-4
DC ILC2 Peyers
IL-9 patch
IL-5,
IL-4 IL-13

Basophil
Type 2
? cytokines
DC maturation
and migration Treg cell Eosinophil Eosinophil and
basophil
? TH2 cell accumulation

TH2 cell Circulation


Gut lamina OX40L dierentiation
propria
OX40

Peptide Naive
TH9 cell TH2 cell- CD4+ T cell
promoting MHC
Draining phenotype
lymph node

Figure 2 | TH2 cell-mediated inflammatory response to oral antigen in the gut. EpithelialNature damage or inflammation
Reviews | Immunology
(forexample, due to toxin exposure or trauma) in the gut, skin (not shown) or airways (not shown) allows increased antigen
entry and promotes the secretion of the epithelium-derived cytokines interleukin25 (IL25), IL33 and thymic stromal
lymphopoietin (TSLP)157. These mediators set the immune system towards a T helper 2 (TH2) cell response, and it is
thought that the initial sensitization to food allergens often takes place at the skin70,120. In particular, TSLP may promote
dendritic cell (DC) differentiation into a TH2 cell-promoting phenotype157,173. For example, OX40L may be upregulated in
DCs that promote TH2 cell differentiation of naive CD4+ Tcells173. IL25 secretion by epithelial tuft cells also may aid the
expansion of type2 innate lymphoid cell (ILC2) populations174, which together with TH2 cells secrete cytokines that
promote the TH2 cell-mediated immune response175, which includes tissue eosinophil accumulation and IgE class-
switching by Bcells20. TH9cells also contribute to the allergic immune response by increasing tissue mast cell
accumulation176, and IL4mediated signalling may convert regulatory Tcells into TH2 cells62. The roles of follicular Tcells,
tissue-resident Tcells, CD8+ Tcells and Tcells remain to be determined. Treg cell, regulatory T cell.

peanut-specific IgG4 was found to inhibit mast cell Diagnostic tests for food allergies
activation, which was restored when the plasma was Clinical history and physical examination are the first-
depleted of IgG4 (REF.88). Other findings suggest that line approach to diagnosing a food allergy, which should
food allergen OIT can stimulate the somatic mutation address specific risk factors such as atopic dermatitis and
of allergen-specific IgG4(REF.89). a family history of atopic disease16. When IgE-mediated
Furthermore, an increase in the number of regulatory food allergy is suspected, measurements of total and
Bcells (Breg cells) that secrete IL10 and IgG4 specific for allergen-specific serum IgE (sIgE) levels and skin prick
bee venom phospholipase A2 has been observed in indi- tests (SPTs) are recommended to identify the causative
viduals undergoing treatment for bee-sting allergy, which food, although they are not in themselves diagnostic16.
indicates that Breg cells may have an important role in Increased sIgE levels may suggest food allergy, and sIgE
immunotherapy 90. In addition, immunotherapy-related thresholds with a 95% positive predictive value have
increases in IgA2 levels have been observed in a study been determined for some food allergens; for example,
Regulatory Bcells of children with egg allergy, who had lower IgA2 levels in peanut-allergic patients, the threshold sIgE level for
(Breg cells). These cells support than did control individuals at baseline91. Successful OIT peanut antigen is 14kUA/L (UA=allergen-specific units;
immune tolerance through
secretion of the
has resulted in desensitization over a period of months to 1 unit2.4ng IgE)17,92,93. SPTs provoke allergen-mediated
anti-inflammatory cytokines a year, and in some cases has succeeded in establishing mast cell degranulation in the skin, leading to wheal-
IL10, IL35 and TGF. sustained unresponsiveness to food allergens77. and-flare reactions. A lancet coated with allergen extract

NATURE REVIEWS | IMMUNOLOGY VOLUME 16 | DECEMBER 2016 | 757



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

is used to puncture the epithelial barrier to elicit a skin and intestinal epithelia encounter food proteins: the
reaction; if a wheal forms, it is measured after 15min- skin epithelium encounters intact protein molecules in
utes. Wheal size thresholds indicating a high probabil- a SPT, whereas the intestinal epithelium also encounters
ity of food allergy have been defined for some common digested proteins, which may have altered capacities to
foods; for example, a peanut SPT resulting in a wheal of elicit immune responses.
8mm diameter has a 95% positive predictivevalue94. The only definitive diagnostic method for food allergy
However, sIgE levels and SPTs indicate allergen sensi is the OFC, in which increasing doses of a potential food
tization only (which is necessary, but not sufficient, for allergen are ingested over fixed time intervals, until either
food allergy), rather than clinically significant food an allergic reaction is observed or a maximum dose is
allergy, and both tests are associated with a high rate reached17. However, this diagnostic method is infre-
of false positives, due in part to crossreactivity between quently used outside properly equipped and staffed spe-
homologous proteins95. False-positive SPTs may also cialist centres because it is resource-intensive and carries
arise from differences in the manner in which the skin the risk of inducing a severe anaphylactic reaction. Ideally,

Lumen
Increased Food Intestinal
Dimeric levels of IgA
IgA Tuft cell allergen epithelial
cell
M cell

Competition for B cell


food allergens
IgE
IgG4
IL-4,
IL-13
Class-switching
from IgE to IgG
IL-10,
FcRIIb TGF
OX40
Peyers patch
Mast cell
Treg cells TH2
Decreased cell
mast cell IL-10,
Basophil TGF
and basophil
activation CTLA4, Increased ratio
LAG3 of Treg cells to
DC
TH2 cells

Treg Treg cell TH2 cell


cell dierentiation

Gut lamina
propria
Anergy Apoptosis
Peptide IgA
Tolerogenic Naive IL-10,
phenotype MHC CD4+ T cell
TGF

Draining
lymph node TFH cell B cell

Figure 3 | Desensitization to oral antigens in the gut. Differences between the immune responses associated with
Nature Reviews | Immunology
desensitization and tolerance are under active investigation. Initially during desensitization, mast cell and basophil
activation are decreased through an unclear mechanism, and a shift occurs from the predominance of T helper 2 (TH2)
cells177 to that of allergen-specific Treg cells77, which in turn may lead to the observed shift in allergen-specific antibodies
from the IgE to the IgG4 isotype86,178. As has been proposed in immunotherapy for venom allergies, IgG4 antibodies may
compete with IgE antibodies for food allergens, further dampening the TH2 cell-mediated immune response179,180. The
source of these IgG4 antibodies may be regulatory Bcells, which also promote immune tolerance through the secretion
ofIL10 (REF.90). The proposed mechanism of dendritic cell (DC) inhibition by regulatory T (Treg) cells is possibly mediated
via cytotoxic T lymphocyte antigen 4 (CTLA4) and lymphocyte activation gene 3 (LAG3)181. Treg cells inhibit mast cells via
contact involving OX40OX40L interaction182. The roles of follicular Tcells, tissue-resident Tcells, CD8+ Tcells and
Tcells remain to be determined.

758 | DECEMBER 2016 | VOLUME 16 www.nature.com/nri



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

a clinical setting in which food allergy SPT and sIgE meas- These studies indicate that the early introduction of
urements are carried out should have a referral system in allergenic foods may be a promising prevention strategy
place for obtaining definitive diagnoses usingOFC. for food allergy. However, many questions regarding
Of substantial clinical value would be simple and the effects of allergen type, dose and frequency require
reliable invitro assays for the accurate diagnosis of food further consideration before recommendations can be
allergy, as well as tests capable of predicting the severity of made105, and a systematic review has concluded that
an allergic reaction upon OFC. These assays could draw there is insufficient evidence to support a role for mater-
from our mechanistic understanding of the submolecular nal allergen consumption in the risk of food allergy 107.
specificity of the inflammatory T H2 cell-mediated Although a definitive, protective value of early exposure
response, as well as our understanding of the roles and to potential food allergens has not been established, cur-
behaviours of the immune cells that are involved. rent guidelines no longer recommend allergen avoidance;
Towards this end, diagnostic tests that are in devel- instead, it is recommended that mothers follow a normal
opment include component-resolved diagnosis (CRD)96 diet during pregnancy and breastfeeding, and they avoid
and the basophil activation test (BAT)97. CRD uses puri- delaying the introduction of solid foods including
fied or recombinant allergens (such as rAra h 2)98 to iden- potentially allergenic foods beyond 46months of age,
tify allergen-specific IgE and IgG4 antibodies99, which even in infants at high risk of developing allergic disease.
may distinguish various allergenic, non-allergenic and The role of the microbiota in the induction of
crossreactive molecules in standardized amounts. BATs immune tolerance is a very active area of research.
are carried out using allergenic proteins as stimulants. Microorganisms that normally colonize the skin and
Upon stimulation, basophils induce surface expression or the gut influence immune system development and can
upregulate the surface level of proteins such as CD63 and promote or restrain the development of immune tol-
CD203c97. Although the BAT provides highly accurate erance to foods108,109. Although causality has not been
diagnoses of peanut 97 and other food allergies, it is cur- established, one epidemiological study found an associ-
rently used primarily in research settings. With further ation between the use of antibiotics and an increased risk
standardization, these invitro tests for food allergy may of cows milk allergy 110. Variations in the composition of
become more widely accepted in clinical settings. the human skin and gut microbiota have been correlated
with food allergy, but the observed changes in bacterial
Prevention groups are inconsistent across several studies111113. Many
Increasing attention is being devoted to research on risk studies have investigated whether caesarean birth (which
factors and preventive measures to mitigate the rising is thought to alter the microbiota of the infant compared
prevalence of food allergies. Whereas the avoidance of with the maternal vaginal microbiota acquired during
common food allergens during pregnancy and breast- vaginal delivery) is associated with an increased risk of
feeding has been previously advocated100, some evidence food allergy; a systematic review of 13 studies reports
now suggests that early exposure to potential food aller- that 6 observed significant associations, although only
gens may decrease the risk of developing the respective 2 of those relied on clinical food allergy diagnosis114.
allergy. For example, maternal consumption of common Similarly, many studies have examined the ingestion of
food allergens, such as peanuts, tree nuts, milk and wheat, probiotics during pregnancy or breastfeeding, or by indi-
during pregnancy may reduce the risk of food allergy in viduals at risk of or suffering from food allergy; however,
infants101. Similarly, the introduction of an increased uncertainty persists as to whether the incidence of food
diversity of foods during infancy may be associated allergy is altered115.
with a reduced risk of allergic disease102. A landmark Recent studies suggest that higher serum levels of
study and its follow-up indicate that the early initiation vitaminD are inversely associated with risk of food
of peanut consumption (at 411months) may prevent allergy. VitaminD is an immunomodulator, the defi-
peanut allergy; increased levels of peanut-specific IgG4 ciency of which not only interferes with its general
characterized the peanut-consuming group, whereas the tolerogenic effects but also may increase an individu-
avoidance group had increased levels of peanut-specific als susceptibility to gastrointestinal infection, thereby
IgE103,104. On the basis of this Learning Early About compromising the integrity of the gastrointestinal epi-
Peanut Allergy (LEAP) trial, global consensus guidance thelial barrier and undermining tolerogenic encoun-
has been published recommending the early introduction ters of immune cells with food protein antigens116. The
of peanuts to high-risk infants because of the potential incidence of peanut or egg allergy was 3 times higher in
association between delaying peanut introduction and vitaminDdeficient infants, and it was 6 times higher
an increased risk of peanut allergy 105. Another large, among food-sensitized vitaminDdeficient infants;
prospective, randomized trial from the Enquiring About those infants with lower levels of vitaminDbinding
Tolerance (EAT) study group, which enrolled more than protein, and thus higher levels of biologically available
1,300 infants, found that the introduction of six com- serum vitaminD, experienced some compensation for
monly allergenic foods (peanut, cows milk, egg, sesame, the unfavourable effects of low serum levels of vita-
whitefish and wheat) to breastfeeding infants at 3months minD117. The immunological mechanisms underlying
of age may be feasible without interfering with breast- these observations are ripe for further investigation.
feeding, although difficulty with compliance precluded Other factors associated with increased risk of food
further definitive findings regarding the prevention of allergy include atopic dermatitis and family history of
food allergy in this feasibility study 106. atopic disease118. Predisposition to the development

NATURE REVIEWS | IMMUNOLOGY VOLUME 16 | DECEMBER 2016 | 759



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

of TH2cell-mediated diseases, including food allergy, range and increase gradually over a period of days or
has been associated with mutations in filaggrin, which weeks. A large, multicentre, randomized, placebo-
is a protein important for skin barrier integrity 119,120. controlled, double-blind, crossover study evaluated SLIT
Additional study of the mechanisms that mediate this for peanut allergy in 40 subjects, with a response defined
association, which may involve an improper or alarm- as tolerating a dose of 5g peanut powder or a 10fold
ing introduction of food proteins to the immune system increase in the tolerated dose: after 44weeks, 70% of
through a weakened skin barrier, are warranted. Studies the 20 subjects in the treatment group had responded,
of the application of skin emollients to maintain skin compared with 15% in the control group; the median
barrier integrity have shown beneficial effects in terms tolerated dose of peanut powder in the treatment group
of atopic dermatitis, but they have not yet focused on the rose from an initial 3.5mg to 496mg (the equivalent of
development of food allergies121,122. Genetic studies have about twopeanuts)127.
identified seven susceptibility loci that are associated OIT is a promising approach that enables the majority
with the atopic march from atopic dermatitis to asthma, of children with a food allergy to become desensitized to
but their role in susceptibility to food allergy was not substantial amounts of the allergenic food. In OIT, a low
studied123. Further investigation of the extent to which dose of allergen (in the milligram range) is ingested daily
the prevention and treatment of atopic dermatitis may and the dose is gradually increased (for example, every
also function in food allergy prevention and treatment two weeks) over a period of several months. Because
may be very valuable. of the larger allergen doses that are used in OIT com-
pared with other forms of immunotherapy that patients
Therapy may choose, patients can often be desensitized not only
Currently, there is no definitive treatment for food allergy: to amounts of the allergen sufficient to avoid a life-
the avoidance of food allergens and treatment of food threatening reaction due to accidental exposure, but also
allergen-induced systemic reactions with adrenaline to the extent that they are able to consume gram amounts
remain the standard of care16. Adrenaline can reverse of allergenicfoods.
oedema, urticaria, bronchospasm, hypotension and The question of sustained unresponsiveness to higher
gastrointestinal symptoms within minutes. Early treat- doses of allergen, distinct from desensitization requiring
ment with adrenaline after allergen exposure (such as continued, regular allergen exposure, was addressed in
within the first 6minutes after exposure) is more effec- a double-blind, randomized, placebo-controlled trial for
tive than later treatment (such as more than 20minutes egg OIT involving 55 children128. After an initial buildup
after the onset of reaction), and early response is a crucial and maintenance phase, 55% and 75% of the 40 children
factor in preventing death from anaphylaxis124. Between in the treatment arm passed an OFC at 10 and 22months,
1% and 20% of people who experience an allergic food respectively, whereas none in the placebo group did,
reaction may have a biphasic reaction that involves a showing again that OIT can effectively induce allergen
recurrence of their symptoms within hours after initial desensitization. Those children who passed the OFC at
adrenaline treatment; a late or insufficient first dose of 22months avoided egg for 2months and were retested
adrenaline has been postulated to increase the risk of a by OFC at 24months. Only 11 children passed the OFC
biphasicreaction125. at 24months (28% of the 40 participants in the treatment
Other pharmaceuticals, such as antihistamines, arm), but all who passed were able to pass a subsequent
including diphenhydramine, or the more specific OFC one year later (after 36months on an adlib diet).
H1 receptorblockers such as cetirizine, are used to treat In conclusion, although desensitization does not neces
localized food allergy symptoms. Gastrointestinal symp- sarily lead to tolerance, sustained unresponsiveness is
toms can be treated with H2 receptor blockers such achievable in a subset of patients.
as famotidine. However, currently available pharma EPIT uses an adhesive containing microgram amounts
ceuticals only control the symptoms of food allergy, and of allergen to deliver antigen to the skin surface. This
they do not address the underlying immunedisorder. routeof delivery seems to have fewer and less intense side
Immunotherapy to desensitize individuals to potential effects than OIT, and some subjects may prefer wearing
food allergens represents tremendous progress in treat- a skin patch to orally consuming the same food allergen
ing food allergy. In this section, we discuss the different each day 129. A phaseII study evaluating a milk patch and
types of immunotherapy, ongoing clinical trials (TABLE2) a phaseIII study evaluating a peanut patch are currently
and other innovative treatments in clinical development. underway.

Antigen desensitization. New immunotherapies for food Monoclonal antibodies. Our mechanistic understand-
allergy are being designed based on our increasing mech- ing of the molecular processes involved in sensitization
anistic understanding of how desensitization to food has inspired the development of monoclonal antibodies
protein antigens occurs. Desensitizing immunotherapy as therapeutic agents to help block these processes. For
is generally delivered sublingually, orally or through the example, because allergic reactions are common dur-
skin. The first double-blind, placebo-controlled trial ing OIT and can be severe, the use of monoclonal anti
ofSLIT for food allergy was published in 2005 (REF.126). bodies specific for IgE in conjunction with OIT has been
H1 receptor blockers
H1 receptor antagonists that
SLIT involves administering a liquid extract of the explored to increase safety. The monoclonal antibody
block histamine action, allergen under the tongue, where it is held for several omalizumab binds to the Fc region of IgE antibodies,
dampening allergic reactions. minutes. Daily allergen doses begin in the submilligram blocking IgE binding to FcRI and thus preventing the

760 | DECEMBER 2016 | VOLUME 16 www.nature.com/nri



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Table 2 | Immunotherapy for food allergy


Treatment Type of drug Mechanism Trial examples (Preliminary) results
ANB020 IL33specific Blocks IL33induced signalling, PhaseI NA
antibody which normally favours TH2 cell-
mediated allergic responses
Reslizumab Humanized mouse Binds free IL5, preventing IL5 Off label137 No clinical effect in EoE, though decrease in
monoclonal antibody receptor activation eosinophilic infiltration of oesophagus
Mepolizumab Humanized mouse Binds free IL5, preventing IL5 Off label136 Decrease in eosinophilic infiltration of
monoclonal antibody receptor activation oesophagus in EoE
Omalizumab Humanized mouse Binds Fc region of IgE, blocking PhaseI and II Reduces allergen-mediated mast cell
monoclonal antibody FcRI binding (seemain text degranulation; FcRI signal blockade blunts TH2
andREFS131134) cell-mediated response; mitigates allergen side
effects and allows rapid escalation of OIT dose
OIT for Allergen taken orally Potentially functions via Egg (CoFAR 75% desensitization and 28% sustained
treatment expansion of allergen-specific study)128 unresponsiveness
CD4+FOXP3+ Treg cell population
OIT for Allergen taken orally Potentially functions via Peanut (LEAP 13.7% of SPT-negative infants developed
prevention expansion of allergen-specific study)104 peanut allergy in the avoidance group versus
CD4+FOXP3+ Treg cell population 1.9% in the consumption group
SLIT Allergen Potentially functions via Hazelnut126 45% of the treatment group reached the
administered expansion of allergen-specific andpeanut127 highest dose of hazelnut (20g), versus 9% of
sublingually CD4+FOXP3+ Treg cell population the placebo group; modest desensitization to
peanut in most test subjects versus placebo
EPIT Allergen absorbed Potentially functions via PhaseI (for milk)190 Abstracts report promising increases in dose
through skin expansion of allergen-specific threshold for food allergic symptoms
CD4+FOXP3+ Treg cell population
EMP123 Recombinant Arah Rectal administration to induce PhaseI (REF.191) Increased food allergic reactions with drug
1, Ara h 2 and Ara h 3 mucosal tolerance
Peanut vaccine Ara h 1, Arah2 and Presentation of peanut protein PhaseI and NA
Arah3 without inflammation induces preclinical
tolerance
CoFAR, Consortium for Food Allergy Research; EoE, eosinophilic oesophagitis; EPIT, epicutaneous immunotherapy; IL, interleukin; LEAP, Learning Early About
Peanut Allergy; NA, not applicable; OIT, oral immunotherapy; SLIT, sublingual immunotherapy; SPT, skin prick test; TH2, T helper 2; Treg cell, regulatory T cell.

Fc receptor-mediated activation and degranulation 122.5 to 40.2 eosinophils per high-powered field; mean
of mast cells and basophils130. Omalizumab was origi decreased from 39.1 to 9.3)136. In a study of reslizumab,
nally approved for the treatment of allergic asthma, 226 patients received four, monthly antibody infusions137,
but has now been tested in combination with OIT for which resulted in an approximately 60% reduction in the
the treatment of food allergies in a series of smaller median oesophageal eosinophil count, compared with a
studies131134. Promising results indicate that OIT plus 24% reduction for the placebo group. However, physi
omalizumab may allow the immune system to be cian assessment of EoE severity improved in both pla-
desensitized to food allergens more quickly and safely cebo and treatment groups, with no statistically significant
than OIT alone. Patients initially receive injections of difference between the groups.
omalizumab on a biweekly to monthly basis for at least
8weeks to deplete free IgE and downregulate surface Future treatments. Our increasing understanding of
expression of FcRI on mast cells135. Then, OIT begins: the biology of allergic responses gives rise to new tar-
after an initial overlap period of several weeks in which gets for the experimental treatment of food allergy 138
both omalizumab and food allergen are given, the (TABLE2). The IL33mediated signalling pathway is one
monoclonal antibody is discontinued134. Additional such target: knocking out the IL33 receptor, ST2, in
clinical trials are being carried out to confirm the safety a mouse model of house dust mite and peanut allergy
and efficacy of omalizumab, to optimize maintenance showed that this pathway is necessary for driving the
doses and to evaluate the sustainability of desensitization TH2 cell-mediated allergic response139.
or the establishment of tolerance. Mutations in the -subunit of the IL4 receptor
Additional examples include monoclonal anti- (IL4R)140 and in IL13 (REF.141) have been found to
bodies that target upstream mediators of food allergy. be associated with an increased risk of food allergy.
For example, pilot studies have been carried out using Accordingly, another strategy for treating food allergy
IL5specific monoclonal antibodies (such as mepoli- is directly to block signalling by the canonical TH2 cell
zumab and reslizumab) to treat EoE. In a study of cytokines, IL4 and IL13. Although this approach has
mepolizumab, 59 patients received three, monthly anti- yet to be tested in food allergy, there is reason to believe
body infusions, which resulted in a decrease in oesopha that it may provide an effective treatment. Dupilumab
geal eosinophil counts (peak count decreased from is a fully human monoclonal antibody directed against

NATURE REVIEWS | IMMUNOLOGY VOLUME 16 | DECEMBER 2016 | 761



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Box 2 | Tolerance versus desensitization versus sustained unresponsiveness outer membrane protein 16 (OMP16), have been found
in mouse models to reduce atopic responses to peanut
Tolerance is the state of healthy immunological nonresponsiveness to common food and cows milk, respectively 144,145.
antigens. Desensitization is a temporary increase in the threshold for allergen reactivity, Toll-like receptor (TLR) agonists also have been
the mechanisms of which are not well understood and may differ from those of the used to treat atopic conditions other than food allergy
permanently allergen-unresponsive state of immune tolerance.
by immune deviation, and are therefore of interest as
An individual with food allergy who has been desensitized to a food allergen must
continue to consume that allergen regularly, or risk losing his or her desensitization. potential therapies for food allergy. R848, a TLR7 agonist,
Immunotherapy often results in desensitization rather than a durable state of sustained was found to decrease airway inflammation in a mouse
unresponsiveness. Because it is unknown whether a food-allergic patient who has been model of asthma through its action on natural killer T
desensitized through immunotherapy will remain permanently unresponsive to that (NKT) cells146; also in mice, CpG-oligodeoxynucleotides
food allergen, we refer to sustained unresponsiveness as distinct from the healthy, (TLR9 agonists), when combined with protamine to
pre-allergic state of immune tolerance. form nanoparticles and then complexed with the peanut
The immune changes that are observed in oral desensitization to food allergens protein Arah2, were found to blunt the peanut-allergic
include a decrease in the number of allergen-specific T helper 2 (TH2) cells and in IgE TH2cell-mediated response147. In humans, a TLR9 ago-
levels, as well as an increase in the number of allergen-specific regulatory Tcells and in nist has been shown to be effective in the treatment of
IgG4 levels. Other cells are probably eliminated through depletion, exhaustion or
allergic rhinitis148, and a modified allergoid preparation
anergic mechanisms. One study found preferential depletion of allergen-specific
TH2cells after repeated stimulation with high-dose antigen78. Another recent study including a TLR4 agonist has been effective in treating
demonstrated that peanut-specific CD4+ Tcells from subjects who developed sustained ragweed pollen allergy149. Taken together, these studies
unresponsiveness tended to transition more strongly towards the gene-expression suggest that TLR modulation in humans may be a fruitful
clusters defined as non allergic or anergic memory (REF.77). approach to food allergy treatment.
Although preliminary research has identified certain immunological changes that CRTH2, a G proteincoupled receptor expressed by
occur with immunotherapy, further research is necessary to identify appropriate TH2 cells, eosinophils and basophils, provides another
biomarkers of successful immunotherapy. There are no reliable markers to distinguish therapeutic target. Activated mast cells release the
desensitization from durable, sustained unresponsiveness. CRTH2 ligand, prostaglandin D2, which is thought to
attract and activate CRTH2+ cells and thereby to augment
the TH2 cell-mediated immune response. CRTH2 antag-
IL4R, which is a component of typeI and typeII IL4 onists have been tested in humans for the treatment of
receptors as well as of the IL13 receptor (which is a asthma and allergic rhinitis, with promising results150,151.
heterodimer composed of IL4R and IL13RA1). This It would be informative to test the effectiveness of
monoclonal antibody has been found to be effective for CRTH2 antagonists in food allergy, which also involves
the treatment of asthma and atopic dermatitis142,143. These many nonrespiratory symptoms.
findings support the potential clinical benefit in allergic Another potential approach to tolerizing the immune
diseases of blockade of the IL4- and IL13induced sig- system uses a DNA vaccine that expresses peanut protein.
nalling pathways, and dupilumab is currently being tested In 1999, this approach was demonstrated in mice using
in patients withEoE. oral delivery of a DNA plasmid encoding theArah2pro-
In a mouse model of peanut allergy featuring a muta- tein on a nanoparticle carrier 152. Subsequent Arah2
tion in IL4R that increases ligand-activated signalling, expression in the gut epithelium resulted in partial pro-
TH2 cell responses and IgE production are enhanced; the tection from anaphylaxis. A clinical trial is currently
additional IgE then binds FcRI on mast cells, promot- underway to test a DNA vaccine for peanut allergy.
ing atopic TH2 cell-mediated responses to food allergens As our understanding of the biology of food allergy
and leading to Treg cell suppression140. Tofocus specifi- develops through the immune monitoring of individu-
cally on the IgE-mediated activation of mast cells in this als enrolled in both preventive and therapeutic clinical
model, a mast-cell-specific deletion of spleen tyrosine studies, we anticipate that cellular and molecular mark-
kinase (Syk), which is essential for FcRI signalling, was ers will identify subgroups of patients who differ in their
engineered; this deletion restored the induction of tol- disease behaviour and in their response to targeted ther-
erance-promoting Treg cells140. Furthermore, the trans- apies. This classification of endotypes and phenotypes
fer of Treg cells from mice treated with both peanut and of food allergy will enable the design of safer and more
Syk inhibitor blocked allergic sensitization to peanut effective treatments. In addition, markers may be devel-
in recipient mice, which is consistent with the hypoth- oped to aid in monitoring the course of food allergy
esis that Treg cells have an important role in tolerizing treatment. For example, an invitro diagnostic test on a
the immune system to allergens and could be use- blood sample could be used to identify when a patient
ful as a means of monitoring patients or as a target of has been tolerized to an allergen, rather than simply
OIT(BOX2). desensitized, so that the need for maintenance ingestion
As FIG.3 suggests, the balance between atopy and of allergen in immunotherapy can bedetermined.
tolerance can be understood in mechanistic terms
as a balance between the predominance of TH2 cells Conclusions
(atopy)and that of allergen-specific Treg cells (desensi- Food allergy is a common and potentially fatal condi-
tization and tolerance). Adjuvants that can deviate the tion that affects the daily lives of a growing number of
immune response from a TH2 cell-mediated state when individuals and their families. Although the traditional
administered with food allergen thus may offer a poten- standard of care has been food-allergen avoidance, there
tial treatment forfood allergy. Chitin plus chitosan, and is recent progress in using allergen exposure both in the

762 | DECEMBER 2016 | VOLUME 16 www.nature.com/nri



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

prevention of food allergies and as a method of desensi sustained unresponsiveness, a possibly permanent state of
tization. Although desensitization therapies for food nonreactivity to a food allergen, without requiring regu
allergy have been shown to be effective, many practical lar, continued exposure to the food allergen. Gaps persist
barriers restrict their general use. Diagnostic invitro tests in our understanding of the immune mechanisms of food
that can be carried out reliably and inexpensively without allergy and of how desensitization and conversion to sus-
the risk of an anaphylactic reaction are required to enable tained unresponsiveness take place. New technologies for
immunotherapy to become the standard of care and pre- monitoring and analysing the human immune system in
vention for food allergy. The frequent, extended clinical individuals undergoing immunotherapy may help to elu-
visits that are required for immunotherapy also pose a cidate these mechanisms and to reveal new cell types that
barrier to its general adoption. Furthermore, desensitiza- are important in this process. In particular, understand-
tion resulting from immunotherapy is often temporary, ing the markers and mechanisms that differentiate toler-
and recurrence of the food allergy is often observed after ance, desensitization and sustained unresponsiveness will
regular intake of the maintenance dose of the food aller- be useful for monitoring immunotherapy and providing
gen is stopped. The goal and the challenge are to achieve the next generation of treatment targets.

1. Thomsen,S.F. Epidemiology and natural history of 19. Lee,A.J., Thalayasingam,M. & Lee,B.W. Food allergy 39. Rescigno,M. etal. Dendritic cells express tight
atopic diseases. Eur. Clin. Respir. J. 2, 24642 (2015). in Asia: how does it compare? Asia Pac. Allergy junction proteins and penetrate gut epithelial
2. Anto,J.M. etal. Understanding the complexity of IgE- 3, 314 (2013). monolayers to sample bacteria. Nat. Immunol. 2,
related phenotypes from childhood to young adulthood: 20. Stone,K.D., Prussin,C. & Metcalfe,D.D. 361367 (2001).
a Mechanisms of the Development of Allergy (MeDALL) IgE, mast cells, basophils, and eosinophils. 40. Niess,J.H. etal. CX3CR1mediated dendritic cell
seminar. J.Allergy Clin. Immunol. 129, 943954 J.Allergy Clin. Immunol. 125, S73S80 (2010). access to the intestinal lumen and bacterial clearance.
(2012). 21. Sampson,H.A. etal. Second symposium on the Science 307, 254258 (2005).
3. Alduraywish,S.A. etal. The march from early life food definition and management of anaphylaxis: summary 41. Chinthrajah,R.S., Hernandez,J.D., Boyd,S.D.,
sensitization to allergic disease: a systematic review and report Second National Institute of Allergy and Galli,S.J. & Nadeau,K.C. Molecular and cellular
meta-analyses of birth cohort studies. Allergy 71, Infectious Disease/Food Allergy and Anaphylaxis mechanisms of food allergy and food tolerance.
7789 (2016). Network symposium. J.Allergy Clin. Immunol. 117, J.Allergy Clin. Immunol. 137, 984997 (2016).
4. Hill,D.A., Grundmeier,R.W., Ram,G. & Spergel,J.M. 391397 (2006). 42. Warshaw,A.L., Walker,W.A. & Isselbacher,K.J.
The epidemiologic characteristics of healthcare 22. Ivkovic-Jurekovic,I. Oral allergy syndrome in children. Protein uptake by the intestine: evidence for absorption
provider-diagnosed eczema, asthma, allergic rhinitis, Int. Dent. J. 65, 164168 (2015). of intact macromolecules. Gastroenterology 66,
and food allergy in children: a retrospective cohort 23. Hurlburt,B.K. etal. Structure and function of the 987992 (1974).
study. BMC Pediatr. 16, 133 (2016). peanut panallergen Ara h 8. J.Biol. Chem. 288, 43. Steinbach,E.C. & Plevy,S.E. The role of macrophages
5. Prescott,S.L. etal. A global survey of changing 3689036901 (2013). and dendritic cells in the initiation of inflammation in
patterns of food allergy burden in children. 24. Asarnoj,A. etal. Peanut component Ara h 8 IBD. Inflamm. Bowel Dis. 20, 166175 (2014).
World Allergy Organ. J. 6, 21 (2013). sensitization and tolerance to peanut. J.Allergy Clin. 44. Mazzini,E., Massimiliano,L., Penna,G. & Rescigno,M.
6. Gupta,R. etal. The economic impact of childhood food Immunol. 130, 468472 (2012). Oral tolerance can be established via gap junction
allergy in the United States. JAMA Pediatr. 167, 25. Hofmann,C. etal. Cor a 1reactive Tcells and IgE are transfer of fed antigens from CX3CR1+ macrophages
10261031 (2013). predominantly cross-reactive to Bet v 1 in patients with to CD103+ dendritic cells. Immunity 40, 248261
7. Koplin,J.J., Mills,E.N. & Allen,K.J. Epidemiology of birch pollen-associated food allergy to hazelnut. (2014).
food allergy and food-induced anaphylaxis: is there J.Allergy Clin. Immunol. 131, 13841392 (2013). CX3CR1+ macrophages capture food antigens and
really a Western world epidemic? Curr. Opin. Allergy 26. Commins,S.P., Jerath,M.R., Cox,K., Erickson,L.D. transfer them to CD103+ DCs in a connexin43
Clin. Immunol. 15, 409416 (2015). &Platts-Mills,T. Delayed anaphylaxis to -gal, an dependent manner, cooperating in the process of
8. Branum, A. M. & Lukacs, S. L. Food allergy among U. S. oligosaccharide in mammalian meat. Allergol. Int. 65, antigen capture and presentation that contributes
children: trends in prevalence and hospitalizations. 1620 (2016). to Treg cell differentiation involved in tolerance.
NCHS data brief, no 10. Hyattsville, MD: National 27. Steinke,J.W., Platts-Mills,T.A. & Commins,S.P. 45. Schulz,O. etal. Intestinal CD103+, but not CX3CR1+,
Center for Health Statistics (2008). The alpha-gal story: lessons learned from connecting antigen sampling cells migrate in lymph and serve
9. Jackson, K. D., Howie, L. D. & Akinbami, L. J. Trends in the dots. J.Allergy Clin. Immunol. 135, 589596 classical dendritic cell functions. J.Exp. Med. 206,
allergic conditions among children: United States, (2015). 31013114 (2009).
19972011.CDC NCHS data brief, no 121. Hyattsville, 28. Sidbury,R. etal. Guidelines of care for the 46. Coombes,J.L. etal. A functionally specialized
MD: National Center for Health Statistics (2013). management of atopic dermatitis: section 4. population of mucosal CD103+ DCs induces Foxp3+
10. Gupta,R.S. etal. The prevalence, severity, and Prevention of disease flares and use of adjunctive regulatory Tcells via a TGF-- and retinoic acid-
distribution of childhood food allergy in the United therapies and approaches. J.Am. Acad. Dermatol. 71, dependent mechanism. J.Exp. Med. 204, 17571764
States. Pediatrics 128, e9e17 (2011). 12181233 (2014). (2007).
11. Turner,P.J. etal. Increase in anaphylaxis-related 29. Spergel, J. M. Nonimmunoglobulin e-mediated 47. Persson,E.K., Scott,C.L., Mowat,A.M. &
hospitalizations but no increase in fatalities: an immune reactions to foods.. AAllergy Asthma Clin. Agace,W.W. Dendritic cell subsets in the intestinal
analysisof United Kingdom national anaphylaxis data, Immunol. 2, 7885 (2006). lamina propria: ontogeny and function. Eur.J.Immunol.
19922012. J.Allergy Clin. Immunol. 135, 956963 30. Zuo,L. & Rothenberg,M.E. Gastrointestinal 43, 30983107 (2013).
(2015). eosinophilia. Immunol. Allergy Clin. North Am. 27, 48. Sun,C.M. etal. Small intestine lamina propria
12. Mullins,R.J., Wainstein,B.K., Barnes,E.H., Liew,W.K. 443455 (2007). dendritic cells promote denovo generation of
& Campbell,D.E. Increases in anaphylaxis fatalities 31. Simon,D. etal. Eosinophilic esophagitis is Foxp3 T reg cells via retinoic acid. J.Exp. Med. 204,
in Australia 1997 to 2013. Clin. Exp. Allergy 46, characterized by a non-IgE-mediated food 17751785 (2007).
10991110 (2016). hypersensitivity. Allergy 71, 611620 (2016). 49. Bakdash,G., Vogelpoel,L.T., van Capel,T.M.,
13. Fox,A.T., Sasieni,P., du Toit,G., Syed,H. & Lack,G. 32. Nowak-Wegrzyn,A., Katz,Y., Mehr,S.S. & Koletzko,S. Kapsenberg,M.L. & de Jong,E.C. Retinoic acid
Household peanut consumption as a risk factor for the Non-IgE-mediated gastrointestinal food allergy. primes human dendritic cells to induce gut-homing,
development of peanut allergy. J.Allergy Clin. Immunol. J.Allergy Clin. Immunol. 135, 11141124 (2015). IL10producing regulatory Tcells. Mucosal Immunol.
123, 417423 (2009). 33. Berin,M.C. & Sampson,H.A. Mucosal immunology of 8, 265278 (2015).
14. Brough,H.A. etal. Peanut protein in household dust food allergy. Curr. Biol. 23, R389R400 (2013). 50. Mucida,D. etal. Reciprocal TH17 and regulatory Tcell
is related to household peanut consumption and is 34. du Toit,G., Tsakok,T., Lack,S. & Lack,G. Prevention of differentiation mediated by retinoic acid. Science 317,
biologically active. J.Allergy Clin. Immunol. 132, food allergy. J.Allergy Clin. Immunol. 137, 9981010 256260 (2007).
630638 (2013). (2016). 51. Worbs,T. etal. Oral tolerance originates in the
15. Bened,S., Blzquez,A.B., Chiang,D., Tordesillas,L. 35. Pabst,O. & Mowat,A.M. Oral tolerance to food intestinal immune system and relies on antigen
&Berin,M.C. The rise of food allergy: environmental protein. Mucosal Immunol. 5, 232239 (2012). carriage by dendritic cells. J.Exp. Med. 203, 519527
factors and emerging treatments. EBioMedicine 36. Menard,S., Cerf-Bensussan,N. & Heyman,M. (2006).
7, 2734 (2016). Multiple facets of intestinal permeability and epithelial 52. Kraus,T.A. etal. Induction of mucosal tolerance in
16. Boyce, J. A. et al. Guidelines for the diagnosis and handling of dietary antigens. Mucosal Immunol. Peyers patch-deficient, ligated small bowel loops.
management of food allergy in the United States: 3, 247259 (2010). J.Clin. Invest. 115, 22342243 (2005).
report of the NIAID-sponsored expert panel. J. Allergy 37. McDole,J.R. etal. Goblet cells deliver luminal antigen 53. Spahn,T.W. etal. Mesenteric lymph nodes are critical
Clin. Immunol. 126, S1-58 (2010). to CD103+ dendritic cells in the small intestine. for the induction of high-dose oral tolerance in the
17. Chinthrajah,R.S. etal. Diagnosis of food allergy. Nature 483, 345349 (2012). absence of Peyers patches. Eur. J.Immunol. 32,
Pediatr. Clin. North Am. 62, 13931408 (2015). 38. Mabbott,N.A., Donaldson,D.S., Ohno,H., 11091113 (2002).
18. Obeng,B.B. etal. Food allergy in Ghanaian Williams,I.R. & Mahajan,A. Microfold (M) cells: 54. Fujihashi,K. etal. Peyers patches are required for oral
schoolchildren: data on sensitization and reported food important immunosurveillance posts in the intestinal tolerance to proteins. Proc. Natl Acad. Sci. USA 98,
allergy. Int. Arch. Allergy Immunol. 155, 6373 (2011). epithelium. Mucosal Immunol. 6, 666677 (2013). 33103315 (2001).

NATURE REVIEWS | IMMUNOLOGY VOLUME 16 | DECEMBER 2016 | 763



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

55. Hadeiba,H. etal. CCR9 expression defines tolerogenic 77. Ryan,J.F. etal. Successful immunotherapy induces Component-resolved diagnostics for the evaluation
plasmacytoid dendritic cells able to suppress acute previously unidentified allergen-specific CD4+ Tcell ofpeanut allergy in a low-prevalence area.
graft-versus-host disease. Nat. Immunol. subsets. Proc. Natl Acad. Sci. USA 113, E1286E1295 Pediatr.Allergy Immunol. 24, 665670 (2013).
9, 12531260 (2008). (2016). 99. Wright,B.L. etal. Component-resolved analysis of
56. Evans,T.I. & Reeves,R.K. All-trans-retinoic acid Antigen-specific CD4+ Tcells transition from allergic IgA, IgE, and IgG4 during egg OIT identifies markers
imprints expression of the gut-homing marker 47 and regulatory phenotypes to an anergic TH2cell associated with sustained unresponsiveness.
while suppressing lymph node homing of dendritic cells. phenotype that is largely absent in both Allergy 71, 15521560 (2016).
Clin. Vaccine Immunol. 20, 16421646 (2013). pretreatment participants and healthy controls, as 100. Lack,G. Update on risk factors for food allergy.
57. Cassani,B. etal. Gut-tropic Tcells that express integrin patients develop sustained unresponsiveness via OIT. J.Allergy Clin. Immunol. 129, 11871197 (2012).
47 and CCR9 are required for induction of oral 78. Wambre,E. etal. Specific immunotherapy modifies 101. Bunyavanich,S. etal. Peanut, milk, and wheat intake
immune tolerance in mice. Gastroenterology 141, allergen-specific CD4+ Tcell responses in an epitope- during pregnancy is associated with reduced allergy
21092118 (2011). dependent manner. J.Allergy Clin. Immunol. 133, and asthma in children. J.Allergy Clin. Immunol. 133,
58. Murai, M. et al. Interleukin 10 acts on regulatory 872879 (2014). 13731382 (2014).
Tcells to maintain expression of the transcription In allergen-specific immunotherapy, allergen-specific 102. Roduit,C. etal. Increased food diversity in the first year
factor Foxp3 and suppressive function in mice with TH2 cells are preferentially deleted whereas the of life is inversely associated with allergic diseases.
colitis. Nat. Immunol. 10, 11781184 (2009). frequencies of TH1 and Tr1 cells are not significantly J.Allergy Clin. Immunol. 133, 10561064 (2014).
59. Hadis,U. etal. Intestinal tolerance requires gut homing changed. 103. Du Toit,G. etal. Effect of avoidance on peanut allergy
and expansion of FoxP3+ regulatory Tcells in the 79. Wambre,E. Effect of allergen-specific immunotherapy after early peanut consumption. N.Engl. J.Med. 374,
lamina propria. Immunity 34, 237246 (2011). on CD4+ Tcells. Curr. Opin. Allergy Clin. Immunol. 15, 14351443 (2016).
60. Kim,K.S. etal. Dietary antigens limit mucosal 581587 (2015). 104. Du Toit,G. etal. Randomized trial of peanut
immunity by inducing regulatory Tcells in the small 80. Kwok,W.W. Modulation of Peanut-specific humoral and consumption in infants at risk for peanut allergy.
intestine. Science 351, 858863 (2016). cellular responses pre- and post-oral immunotherapy. N.Engl. J.Med. 372, 803813 (2015).
61. Syed,A. etal. Peanut oral immunotherapy results in Clin. Exp. Allergy 45, 11461149 (2015). 105. Fleischer,D.M. etal. Consensus communication
increased antigen-induced regulatory Tcell function 81. Gutgemann,I., Fahrer,A.M., Altman,J.D., Davis,M.M. onearly peanut introduction and the prevention
and hypomethylation of forkhead box protein 3 & Chien,Y.H. Induction of rapid Tcell activation and ofpeanut allergy in high-risk infants. J.Allergy
(FOXP3). J.Allergy Clin. Immunol. 133, 500510 tolerance by systemic presentation of an orally Clin.Immunol. 136, 258261 (2015).
(2014). administered antigen. Immunity 8, 667673 (1998). 106. Perkin,M.R. etal. Enquiring About Tolerance (EAT)
Hypomethylation of the FOXP3 locus of 82. Wisniewski,J.A. etal. Analysis of cytokine production study: feasibility of an early allergenic food
antigen-induced Treg cells may predict the achievement by peanut-reactive Tcells identifies residual Th2 introduction regimen. J.Allergy Clin. Immunol. 137,
of operationally defined clinical tolerance during effectors in highly allergic children who received peanut 14771486 (2016).
peanut OIT. oral immunotherapy. Clin. Exp. Allergy 45, 12011213 107. Kramer,M.S. & Kakuma,R. Maternal dietary antigen
62. Noval Rivas,M. etal. Regulatory Tcell reprogramming (2015). avoidance during pregnancy or lactation, or both, for
toward a Th2cell-like lineage impairs oral tolerance and 83. Itoh,N., Itagaki,Y. & Kurihara,K. Rush specific oral preventing or treating atopic disease in the child.
promotes food allergy. Immunity 42, 512523 (2015). tolerance induction in school-age children with severe Cochrane Database Syst. Rev. 9, CD000133 (2012).
IL4R signalling drives Treg cell reprogramming into egg allergy: one year follow up. Allergol. Int. 59, 4351 108. Gensollen,T., Iyer,S.S., Kasper,D.L. &
TH2like cells and interferes with Treg cell-mediated (2010). Blumberg,R.S. How colonization by microbiota in
suppression of mast cell activation and expansion; 84. Berin,M.C. & Shreffler,W.G. Mechanisms underlying early life shapes the immune system. Science 352,
IL4R inhibition has a role in Treg cell induction and induction of tolerance to foods. Immunol. Allergy Clin. 539544 (2016).
the enforcement of tolerance, as shown in mouse North Am. 36, 87102 (2016). 109. Shen,N. & Clemente,J.C. Engineering the
models. 85. Dioszeghy,V. etal. Differences in phenotype, homing microbiome: a novel approach to immunotherapy
63. Van Overtvelt,L. etal. Assessment of Bet v 1specific properties and suppressive activities of regulatory forallergic and immune diseases. Curr. Allergy
CD4+ Tcell responses in allergic and nonallergic Tcells induced by epicutaneous, oral or sublingual Asthma Rep. 15, 39 (2015).
individuals using MHC classII peptide tetramers. immunotherapy in mice sensitized to peanut. 110. Metsala,J. etal. Mothers and offsprings use of
J.Immunol. 180, 45144522 (2008). Cell.Mol. Immunol. 13, 113 (2016). antibiotics and infant allergy to cows milk.
64. Turcanu,V., Maleki,S.J. & Lack,G. Characterization of 86. Sugimoto,M. etal. Differential response in allergen- Epidemiology 24, 303309 (2013).
lymphocyte responses to peanuts in normal children, specific IgE, IgGs, and IgA levels for predicting outcome 111. Berni Canani,R. etal. Lactobacillus rhamnosus
peanut-allergic children, and allergic children who of oral immunotherapy. Pediatr. Allergy Immunol. 27, GGsupplemented formula expands butyrate-
acquired tolerance to peanuts. J.Clin. Invest. 111, 276282 (2016). producing bacterial strains in food allergic infants.
10651072 (2003). 87. Burton,O.T. etal. Oral immunotherapy induces IgG ISME J. 10, 742750 (2016).
65. Pulendran,B. & Artis,D. New paradigms in type2 antibodies that act through FcRIIb to suppress IgE- 112. Ling,Z. etal. Altered fecal microbiota composition
immunity. Science 337, 431435 (2012). mediated hypersensitivity. J.Allergy Clin. Immunol. associated with food allergy in infants. Appl. Environ.
66. Galli,S.J. & Tsai,M. IgE and mast cells in allergic 134, 13101317 (2014). Microbiol. 80, 25462554 (2014).
disease. Nat. Med. 18, 693704 (2012). IgG in sera from patients with peanut allergies 113. Azad,M.B. etal. Infant gut microbiota and food
67. Oyoshi,M.K., Oettgen,H.C., Chatila,T.A., Geha,R.S. undergoing OIT suppresses peanut-induced sensitization: associations in the first year of life.
& Bryce,P.J. Food allergy: insights into etiology, basophil activation. Clin.Exp. Allergy 45, 632643 (2015).
prevention, and treatment provided by murine models. 88. Santos,A.F. etal. IgG4 inhibits peanut-induced 114. Marrs,T. etal. Is there an association between
J.Allergy Clin. Immunol. 133, 309317 (2014). basophil and mast cell activation in peanut-tolerant microbial exposure and food allergy? A systematic
68. Paul,W.E. & Zhu,J. How are TH2type immune children sensitized to peanut major allergens. review. Pediatr. Allergy Immunol. 24, 311320
responses initiated and amplified? Nat. Rev. Immunol. J.Allergy Clin. Immunol. 135, 12491256 (2015). (2013).
10, 225235 (2010). 89. Hoh,R.A. etal. Single Bcell deconvolution of peanut- 115. Fiocchi,A. etal. World Allergy Organization-McMaster
69. Han,H., Thelen,T.D., Comeau,M.R. & Ziegler,S.F. specific antibody responses in allergic patients. University Guidelines for Allergic Disease Prevention
Thymic stromal lymphopoietin-mediated epicutaneous J.Allergy Clin. Immunol. 137, 157167 (2016). (GLADP): probiotics. World Allergy Organ. J. 8, 4
inflammation promotes acute diarrhea and 90. van de Veen,W. etal. IgG4 production is confined to (2015).
anaphylaxis. J.Clin. Invest. 124, 54425452 (2014). human IL10producing regulatory Bcells that suppress 116. Vassallo,M.F. & Camargo,C.A.Jr. Potential
70. Tordesillas,L. etal. Skin exposure promotes a antigen-specific immune responses. J.Allergy Clin. mechanisms for the hypothesized link between
Th2dependent sensitization to peanut allergens. Immunol. 131, 12041212 (2013). sunshine, vitaminD, and food allergy in children.
J.Clin. Invest. 124, 49654975 (2014). 91. Konstantinou,G.N. etal. Egg-white-specific IgA and J.Allergy Clin. Immunol. 126, 217222 (2010).
71. Oyoshi,M.K., Larson,R.P., Ziegler,S.F. & Geha,R.S. IgA2 antibodies in egg-allergic children: is there a role 117. Allen,K.J. & Koplin,J.J. Prospects for prevention of
Mechanical injury polarizes skin dendritic cells to elicit in tolerance induction? Pediatr. Allergy Immunol. 25, food allergy. J.Allergy Clin. Immunol. Pract.
a TH2 response by inducing cutaneous thymic stromal 6470 (2014). 4, 215220 (2016).
lymphopoietin expression. J.Allergy Clin. Immunol. 92. Hamilton,R., G. & Adkinson,N. F.Jr. In vitro assays for 118. Koplin,J.J. etal. The impact of family history of allergy
126, 976984 (2010). the diagnosis of IgE-mediated disorders. J.Allergy Clin. on risk of food allergy: a population-based study of
72. Hammerschmidt,S.I. etal. Retinoic acid induces Immunol. 114, 213225 (2004). infants. Int. J.Environ. Res. Public Health 10,
homing of protective T and Bcells to the gut after 93. Spergel,J.M. etal. Food allergy in infants with atopic 53645377 (2013).
subcutaneous immunization in mice. J.Clin. Invest. dermatitis: limitations of food-specific IgE 119. Marenholz,I. etal. Filaggrin lossoffunction mutations
121, 30513061 (2011). measurements. Pediatrics 136, e1530e1538 (2015). predispose to phenotypes involved in the atopic march.
73. Burks,A.W. etal. Update on allergy immunotherapy: 94. Roberts,G. & Lack,G. Diagnosing peanut allergy with J.Allergy Clin. Immunol. 118, 866871 (2006).
American Academy of Allergy, Asthma and skin prick and specific IgE testing. J.Allergy Clin. 120. Venkataraman,D. etal. Filaggrin lossoffunction
Immunology/European Academy of Allergy and Immunol. 115, 12911296 (2005). mutations are associated with food allergy in childhood
Clinical Immunology/PRACTALL consensus report. 95. Kattan,J.D. & Wang,J. Allergen component testing for and adolescence. J.Allergy Clin. Immunol. 134,
J.Allergy Clin. Immunol. 131, 12881296 (2013). food allergy: ready for prime time? Curr. Allergy 876882 (2014).
74. Jones,S.M. etal. Clinical efficacy and immune Asthma Rep. 13, 5863 (2013). 121. Horimukai,K. etal. Application of moisturizer to
regulation with peanut oral immunotherapy. 96. Tuano,K.S. & Davis,C.M. Utility of component- neonates prevents development of atopic dermatitis.
J.Allergy Clin. Immunol. 124, 292300 (2009). resolved diagnostics in food allergy. Curr. Allergy J.Allergy Clin. Immunol. 134, 824830 (2014).
75. Akdis,M. & Akdis,C.A. Mechanisms of allergen- Asthma Rep. 15, 32 (2015). 122. Leung,D.Y. & Guttman-Yassky,E. Deciphering the
specific immunotherapy: multiple suppressor factors at 97. Santos,A.F. etal. Basophil activation test discriminates complexities of atopic dermatitis: shifting paradigms in
work in immune tolerance to allergens. J.Allergy Clin. between allergy and tolerance in peanut-sensitized treatment approaches. J.Allergy Clin. Immunol. 134,
Immunol. 133, 621631 (2014). children. J.Allergy Clin. Immunol. 134, 645652 769779 (2014).
76. Akdis,C.A. & Akdis,M. Mechanisms of allergen- (2014). 123. Marenholz,I. etal. Meta-analysis identifies seven
specific immunotherapy and immune tolerance to 98. Suratannon,N., Ngamphaiboon,J., susceptibility loci involved in the atopic march.
allergens. World Allergy Organ. J. 8, 17 (2015). Wongpiyabovorn,J., Puripokai,P. & Chatchatee,P. Nat.Commun. 6, 8804 (2015).

764 | DECEMBER 2016 | VOLUME 16 www.nature.com/nri



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

124. Ho,M.H., Wong,W.H. & Chang,C. Clinical spectrum 145. Ibaez,A.E. etal. Unlipidated outer membrane 169. Crotty,S. T follicular helper cell differentiation,
of food allergies: a comprehensive review. Clin. Rev. protein Omp16 (U-Omp16) from Brucella spp. as function, and roles in disease. Immunity 41, 529542
Allergy Immunol. 46, 225240 (2014). nasal adjuvant induces a Th1 immune response and (2014).
125. Lieberman,P. Biphasic anaphylactic reactions. modulates the Th2 allergic response to cows milk 170. Chung,Y. etal. Follicular regulatory Tcells expressing
Ann.Allergy Asthma Immunol. 95, 217226 (2005). proteins. PLoS ONE 8, e69438 (2013). Foxp3 and Bcl6 suppress germinal center reactions.
126. Enrique,E. etal. Sublingual immunotherapy for 146. Grela,F. etal. The TLR7 agonist R848 alleviates Nat. Med. 17, 983988 (2011).
hazelnut food allergy: a randomized, double-blind, allergic inflammation by targeting invariant NKT cells 171. Ballesteros-Tato,A. etal. T follicular helper cell
placebo-controlled study with a standardized hazelnut to produce IFN-. J.Immunol. 186, 284290 (2011). plasticity shapes pathogenic Thelper 2 cell-mediated
extract. J.Allergy Clin. Immunol. 116, 10731079 147. Pali-Scholl,I. etal. Protamine nanoparticles with CpG- immunity to inhaled house dust mite. Immunity 44,
(2005). oligodeoxynucleotide prevent an allergen-induced 259273 (2016).
127. Fleischer,D.M. etal. Sublingual immunotherapy for Th2response in BALB/c mice. Eur. J.Pharm. Biopharm. 172. Hirota,K. etal. Plasticity of TH17 cells in Peyers
peanut allergy: a randomized, double-blind, placebo- 85, 656664 (2013). patches is responsible for the induction of Tcell-
controlled multicenter trial. J.Allergy Clin. Immunol. 148. Creticos,P.S. etal. Immunotherapy with a ragweed- dependent IgA responses. Nat. Immunol. 14,
131, 119127 (2013). toll-like receptor 9 agonist vaccine for allergic rhinitis. 372379 (2013).
128. Burks,A.W. etal. Oral immunotherapy for treatment of N.Engl. J.Med. 355, 14451455 (2006). 173. Ito,T. etal. TSLP-activated dendritic cells induce an
egg allergy in children. N.Engl. J.Med. 367, 233243 149. Patel,P., Holdich,T., Fischer von Weikersthal- inflammatory Thelper type2 cell response through
(2012). Drachenberg,K.J. & Huber,B. Efficacy of a short OX40 ligand. J.Exp. Med. 202, 12131223
129. Sindher,S., Fleischer,D.M. & Spergel,J.M. course of specific immunotherapy in patients with (2005).
Advances in the treatment of food allergy: sublingual allergic rhinoconjunctivitis to ragweed pollen. 174. Klose,C.S. & Artis,D. Innate lymphoid cells as
and epicutaneous immunotherapy. Immunol. Allergy J.Allergy Clin. Immunol. 133, 121129 (2014). regulators of immunity, inflammation and tissue
Clin. North Am. 36, 3954 (2016). 150. Hall,I.P. etal. Efficacy of BI 671800, an oral CRTH2 homeostasis. Nat. Immunol. 17, 765774 (2016).
130. Pennington,L.F. etal. Structural basis of omalizumab antagonist, in poorly controlled asthma as sole 175. Mirchandani,A.S. etal. Type2 innate lymphoid cells
therapy and omalizumab-mediated IgE exchange. controller and in the presence of inhaled corticosteroid drive CD4+ Th2 cell responses. J.Immunol. 192,
Nat.Commun. 7, 11610 (2016). treatment. Pulm. Pharmacol. Ther. 32, 3744 24422448 (2014).
The crystal structure of the complex of IgE with (2015). 176. Sehra,S. etal. TH9 cells are required for tissue
omalizumab is presented and used to generate a 151. Krug,N. etal. Efficacy of the oral chemoattractant mast cell accumulation during allergic inflammation.
mutant omalizumab-resistant IgEFc fragment receptor homologous molecule on TH2 cells antagonist J.Allergy Clin. Immunol. 136, 433440 (2015).
which, in combination with omalizumab, more BI 671800 in patients with seasonal allergic rhinitis. 177. Blumchen,K. etal. Oral peanut immunotherapy
efficiently blocks basophil activation than does J.Allergy Clin. Immunol. 133, 414419 (2014). inchildren with peanut anaphylaxis. J.Allergy
either agent alone. 152. Roy,K., Mao,H.Q., Huang,S.K. & Leong,K.W. Clin.Immunol. 126, 8391 (2010).178.
131. Nadeau,K.C., Schneider,L.C., Hoyte,L., Borras,I. Oral gene delivery with chitosan DNA nanoparticles Meiler,F., Klunker,S., Zimmermann,M., Akdis,C.A.
&Umetsu,D.T. Rapid oral desensitisation in generates immunologic protection in a murine model &Akdis,M. Distinct regulation of IgE, IgG4 and IgA by
combination with omalizumab therapy in patients of peanut allergy. Nat. Med. 5, 387391 (1999). T regulatory cells and toll-like receptors. Allergy 63,
with cows milk allergy. J.Allergy Clin. Immunol. 127, 153. von Moltke,J., Ji,M., Liang,H.E. & Locksley,R.M. 14551463 (2008).
16221624 (2011). Tuft-cell-derived IL25 regulates an intestinal ILC2 179. James,L.K. etal. Long-term tolerance after allergen
132. Schneider,L.C. etal. A pilot study of omalizumab to epithelial response circuit. Nature 529, 221225 immunotherapy is accompanied by selective
facilitate rapid oral desensitisation in high-risk peanut- (2016). persistence of blocking antibodies. J.Allergy
allergic patients. J.Allergy Clin. Immunol. 132, 154. Frossard,C.P., Asigbetse,K.E., Burger,D. & Clin.Immunol. 127, 509516 (2011).
13681374 (2013). Eigenmann,P.A. Gut Tcell receptor-+ intraepithelial 180. Nouri-Aria,K.T. etal. Grass pollen immunotherapy
133. Wood,R.A. etal. A randomized, double-blind, placebo- lymphocytes are activated selectively by cholera toxin induces mucosal and peripheral IL10 responses and
controlled study of omalizumab combined with oral to break oral tolerance in mice. Clin. Exp. Immunol. blocking IgG activity. J.Immunol. 172, 32523259
immunotherapy for the treatment of cows milk allergy. 180, 118130 (2015). (2004).
J.Allergy Clin. Immunol. 137, 11031110 (2016). 155. Mishra,A., Schlotman,J., Wang,M. & 181. Vignali,D.A., Collison,L.W. & Workman,C.J.
134. Bgin,P. etal. Phase1 results of safety and Rothenberg,M.E. Critical role for adaptive Tcell How regulatory Tcells work. Nat. Rev. Immunol.
tolerabilityin a rush oral immunotherapy protocol to immunity in experimental eosinophilic esophagitis 8, 523532 (2008).
multiple foods using omalizumab. Allergy Asthma inmice. J.Leukoc. Biol. 81, 916924 (2007). 182. Gri,G. etal. CD4+CD25+ regulatory Tcells suppress
Clin.Immunol. 10, 7 (2014). 156. Yamada,A. etal. Antigen-primed splenic CD8+ Tcells mast cell degranulation and allergic responses through
The first study to use omalizumab with OIT to impede the development of oral antigen-induced OX40OX40L interaction. Immunity 29, 771781
multiple allergens simultaneously demonstrates the allergic diarrhea. J.Allergy Clin. Immunol. 123, (2008).
safe and rapid desensitization of patients with 889894 (2009). 183. Eigenmann,P.A. & Calza,A.M. Diagnosis of IgE-
multiple food allergies. 157. Divekar,R. & Kita,H. Recent advances in epithelium- mediated food allergy among Swiss children with
135. Logsdon,S.L. & Oettgen,H.C. Anti-IgE therapy: derived cytokines (IL33, IL25, and thymic stromal atopic dermatitis. Pediatr. Allergy Immunol. 11,
clinical utility and mechanistic insights. Curr. Top. lymphopoietin) and allergic inflammation. Curr. Opin. 95100 (2000).
Microbiol. Immunol. 388, 3961 (2015). Allergy Clin. Immunol. 15, 98103 (2015). 184. Silverberg,J.I. & Simpson,E.L. Association between
136. Assaad,A.H. etal. An antibody against IL5 reduces 158. Palmer,C.N. etal. Common lossoffunction variants severe eczema in children and multiple comorbid
numbers of esophageal intraepithelial eosinophils in of the epidermal barrier protein filaggrin are a conditions and increased healthcare utilization.
children with eosinophilic esophagitis. Gastroenterology major predisposing factor for atopic dermatitis. Pediatr. Allergy Immunol. 24, 476486 (2013).
141, 15931604 (2011). Nat. Genet. 38, 441446 (2006). 185. Hruz,P. Epidemiology of eosinophilic esophagitis.
137. Spergel,J.M. etal. Reslizumab in children and 159. Irvine,A.D., McLean,W.H. & Leung,D.Y. Filaggrin Dig.Dis. 32, 4047 (2014).
adolescents with eosinophilic esophagitis: results of a mutations associated with skin and allergic diseases. 186. Cianferoni,A. & Spergel,J. Eosinophilic esophagitis:
double-blind, randomized, placebo-controlled trial. N.Engl. J.Med. 365, 13151327 (2011). a comprehensive review. Clin. Rev. Allergy Immunol.
J.Allergy Clin. Immunol. 129, 456463 (2012). 160. Lund,S. Walford, H. H. & Doherty, T. A. Type 2 innate 50, 159174 (2016).
138. Cox,L., Compalati,E., Kundig,T. & Larche,M. lymphoid cells in allergic disease. Curr. Immunol. Rev. 187. Cianferoni,A. & Spergel,J.M. Eosinophilic esophagitis
New directions in immunotherapy. Curr. Allergy Asthma 9, 214221 (2013). and gastroenteritis. Curr. Allergy AsthmaRep. 15, 58
Rep. 13, 178195 (2013). 161. Zhang,Y. etal. Cord blood monocyte-derived (2015).
139. Chu,D.K. etal. IL33, but not thymic stromal inflammatory cytokines suppress IL2 and induce 188. Katz,Y., Goldberg,M.R., Rajuan,N., Cohen,A.
lymphopoietin or IL25, is central to mite and peanut nonclassic TH2type immunity associated with &Leshno,M. The prevalence and natural course of
allergic sensitization. J.Allergy Clin. Immunol. 131, development of food allergy. Sci. Transl Med. food protein-induced enterocolitis syndrome to cows
187200 (2013). 8, 321ra8 (2016). milk: a large-scale, prospective population-based
140. Burton,O.T. etal. Immunoglobulin E signal inhibition 162. Mortha,A. etal. Microbiota-dependent crosstalk study. J.Allergy Clin. Immunol. 127, 647653
during allergen ingestion leads to reversal of between macrophages and ILC3 promotes intestinal (2011).
established food allergy and induction of regulatory homeostasis. Science 343, 1249288 (2014). 189. Elizur,A. etal. Cows milk associated rectal bleeding:
Tcells. Immunity 41, 141151 (2014). 163. Zigmond,E. etal. Macrophage-restricted apopulation based prospective study. Pediatr. Allergy
In a mouse model, mutations in IL4R impaired interleukin10 receptor deficiency, but not IL10 Immunol. 23, 766770 (2012).
Treg cell induction; a mast-cell-specific deletion of deficiency, causes severe spontaneous colitis. 190. Dupont,C. etal. Cows milk epicutaneous
Syk, which is essential for FcRI signalling, restored Immunity 40, 720733 (2014). immunotherapy in children: a pilot trial of safety,
the induction of tolerance-promoting Treg cells. 164. Rosenblum,M.D., Way,S.S. & Abbas,A.K. acceptability, and impact on allergic reactivity.
141. Zitnik,S.E. etal. IL13 variants are associated with Regulatory Tcell memory. Nat. Rev. Immunol. 16, J.Allergy Clin. Immunol. 125, 11651167 (2010).
total serum IgE and early sensitization to food 90101 (2016). 191. Wood,R.A. etal. A phase1 study of heat/phenol-
allergens in children with atopic dermatitis. 165. Sakaguchi,S., Yamaguchi,T., Nomura,T. & Ono,M. killed, E.coli-encapsulated, recombinant modified
Pediatr.Allergy Immunol. 20, 551555 (2009). Regulatory Tcells and immune tolerance. Cell 133, peanut proteins Ara h 1, Ara h 2, and Ara h 3
142. Wenzel,S. etal. Dupilumab in persistent asthma with 775787 (2008). (EMP123) for the treatment of peanut allergy.
elevated eosinophil levels. N.Engl. J.Med. 368, 166. Frossard,C.P., Hauser,C. & Eigenmann,P.A. Allergy 68, 803808 (2013).
24552466 (2013). Antigen-specific secretory IgA antibodies in the gut
143. Beck,L.A. etal. Dupilumab treatment in adults with are decreased in a mouse model of food allergy. Acknowledgements
moderate-tosevere atopic dermatitis. N.Engl. J.Med. J.Allergy Clin. Immunol. 114, 377382 (2004). The authors would like to acknowledge the assistance
371, 130139 (2014). 167. Urb,M. & Sheppard,D.C. The role of mast cells in of T. A. Chatila, S. J. Galli, M. T. Graham, V. Sampath and
144. Bae,M.J., Shin,H.S., Kim,E.K., Kim,J. & Shon,D.H. the defence against pathogens. PLoS Pathog. J. M. Spergel.
Oral administration of chitin and chitosan prevents 8, e1002619 (2012).
peanut-induced anaphylaxis in a murine food allergy 168. Weiner,H.L., da Cunha,A.P., Quintana,F. & Wu,H. Competing interests statement
model. Int. J.Biol. Macromol. 61, 164168 (2013). Oral tolerance. Immunol. Rev. 241, 241259 (2011). The authors declare no competing interests.

NATURE REVIEWS | IMMUNOLOGY VOLUME 16 | DECEMBER 2016 | 765



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.

Vous aimerez peut-être aussi