ArticlePDF Available

CD8 T cell differentiation status correlates with the feasibility of sustained unresponsiveness following oral immunotherapy

Springer Nature
Nature Communications
Authors:

Abstract and Figures

While food allergy oral immunotherapy (OIT) can provide safe and effective desensitization (DS), the immune mechanisms underlying development of sustained unresponsiveness (SU) following a period of avoidance are largely unknown. Here, we compare high dimensional phenotypes of innate and adaptive immune cell subsets of participants in a previously reported, phase 2 randomized, controlled, peanut OIT trial who achieved SU vs. DS (no vs. with allergic reactions upon food challenge after a withdrawal period; n = 21 vs. 30 respectively among total 120 intent-to-treat participants). Lower frequencies of naïve CD8⁺ T cells and terminally differentiated CD57⁺CD8⁺ T cell subsets at baseline (pre-OIT) are associated with SU. Frequency of naïve CD8⁺ T cells shows a significant positive correlation with peanut-specific and Ara h 2-specific IgE levels at baseline. Higher frequencies of IL-4⁺ and IFNγ⁺ CD4⁺ T cells post-OIT are negatively correlated with SU. Our findings provide evidence that an immune signature consisting of certain CD8⁺ T cell subset frequencies is potentially predictive of SU following OIT.
Peanut OIT effects an immune shift away from Th2 phenotype a Top panel: schematic representation of POISED study timeline and sample collection time points. Bottom panel: depiction of approach for mechanistic analyses. Here, “SU” stands for sustained unresponsive and “DS” stands for desensitized alone participants. Illustration created with BioRender.com. b Immune cell subsets among peanut-stimulated week 104 PBMCs from participants on active peanut OIT (n = 80) were compared against those treated with placebo (n = 15). Significant frequency (i.e., percentage) and expression (i.e., MSI) changes among peanut-reactive (pr) CD4⁺ T cells are shown. c, d Significant changes in peanut-stimulated week 104 PBMC culture supernatant of active participants (n = 16) compared to those treated with placebo (n = 7) as a heatmap (c) and as individual boxplots (d). e, f Significant changes in peanut-stimulated week 104 PBMC culture supernatant of active participants (n = 13) compared to BL as a heatmap (e) and individual boxplots (f). In the boxplots, medians are shown, and the “hinges” represent the first and third quartile. The whiskers are the smallest and largest values after exclusion of outliers (greater than the 75th percentile plus 1.5 times the interquartile range (IQR), or less than 25th percentile minus 1.5 times the IQR). q values were computed using the χ² tests in mixed-effects models, adjusted for multiple hypothesis testing using the Benjamini and Hochberg method. Source data are provided as a Source Data file.
… 
CD8⁺ T cell differentiation status distinguishes SU participants from DS Immune cell subsets among peanut-stimulated (a, b, d, f) and unstimulated (c, e, g) PBMCs from SU participants were compared with DS participants at BL, week 104, and week 117. a–c Significant frequency and CD57 expression differences among pr CD8⁺ T cells (a), and peanut non-reactive CD8⁺ T cells (b) among peanut-stimulated (SU, n = 20; DS, n = 29), and unstimulated (c) (SU, n = 20; DS, n = 26), BL PBMCs. d, e Significant differences in the naive CD8⁺ T cell frequency in peanut-stimulated (d) and unstimulated (e) PBMCs at BL (pea stim: SU, n = 20, DS, n = 29; unstim: SU, n = 20, DS, n = 26), week 104 (pea stim: SU, n = 21, DS, n = 30; unstim: SU, n = 20, DS, n = 29), and week 117 (pea stim: SU, n = 17, DS, n = 27; unstim: SU, n = 17, DS, n = 27). f, g Significant differences in the frequency or expression of CD86 in memory B cells among peanut-stimulated PBMCs (f) and APC subsets among unstimulated PBMCs (g) at BL (pea stim: SU, n = 20, DS, n = 29; unstim: SU, n = 20, DS, n = 26). h IL-2 levels in peanut-stimulated BL PBMC culture supernatants from SU (n = 7) vs. DS (n = 7) participants. Boxplots show medians and “hinges” represent the first and third quartile. Whiskers denote the smallest and largest values after outlier exclusion. q values computed using the χ² tests in mixed-effects models, adjusted for multiple hypothesis testing using the Benjamini and Hochberg method. Source data are provided as a Source Data file.
… 
Higher frequency and expression of IFNγ as an additional distinctive feature of DS vs. SU participants a Frequency of IL-4⁺ CD4⁺ memory T effectors at week 104, frequencies of IFNγ⁺ CD4⁺ memory T effectors at week 104 and week 117, and expression of IFNγ in CD4⁺ memory T effectors at week 104 in PMAi-stimulated PBMCs from SU vs. DS [week 104 (PMAi stim: SU, n = 21, DS, n = 29); week 117 (PMAi stim: SU, n = 17, DS, n = 27)]. b Expression of IFNγ in memory γδ T cells among PMAi-stimulated week 104 PBMCs from SU vs. DS; expression of CXCR3 in memory and naive γδ T cells among unstimulated PBMCs at week 104 [week 104 (PMAi stim: SU, n = 21, DS, n = 29; unstim: SU, n = 20, DS, n = 29); week 117 (PMAi stim and unstim: SU, n = 17, DS, n = 27)]. c Plasma CXCL9 level compared between SU (n = 8) and DS (n = 13) at week 117. In the boxplots, medians are shown, and the “hinges” represent the first and third quartile. The whiskers are the smallest and largest values after exclusion of outliers (greater than the 75th percentile plus 1.5 times the interquartile range (IQR), or less than 25th percentile minus 1.5 times the IQR). q values were computed using the χ² tests in mixed-effects models, adjusted for multiple hypothesis testing using the Benjamini and Hochberg method. d Age of wheezing onset for SU participants with asthma (n = 11) was compared with that for DS participants (n = 15). p value was computed using unpaired, two-sided, non-parametric Mann–Whitney test. Source data are provided as a Source Data file.
… 
This content is subject to copyright. Terms and conditions apply.
Article https://doi.org/10.1038/s41467-022-34222-8
CD8+T cell differentiation status correlates
with the feasibility of sustained unrespon-
siveness following oral immunotherapy
Abhinav Kaushik
1
, Diane Dunham
1
,XiaoruiHan
1
,EvanDo
1
, Sandra Andorf
1,2,3
,
Sheena Gupta
4
, Andrea Fernandes
1
,LaurieElizabethKost
1
,
Sayantani B. Sindher
1
, Wong Yu
1,5
,MindyTsai
1,6
, Robert Tibshirani
7
,
Scott D. Boyd
1,6
,ManishaDesai
7
,HoldenT.Maecker
4,8
,StephenJ.Galli
1,6,8
,
R. Sharon Chinthrajah
1
,RosemarieH.DeKruyff
1,9
, Monali Manohar
1,9
&
Kari C. Nadeau
1,9
While food allergy oral immunotherapy (OIT) can provide safe and effective
desensitization (DS), the immune mechanisms underlying development of
sustained unresponsiveness (SU) following a period of avoidance are largely
unknown. Here, we compare high dimensional phenotypes of innate and
adaptive immune cell subsets of participants in a previously reported, phase 2
randomized, controlled, peanut OIT trial who achieved SU vs. DS (no vs. with
allergic reactions upon food challenge after a withdrawal period; n=21vs.30
respectively among total 120 intent-to-treat participants). Lower frequencies
of naïve CD8+T cells and terminally differentiated CD57+CD8+Tcellsubsetsat
baseline (pre-OIT) are associated with SU. Frequency of naïve CD8+Tcells
shows a signicant positive correlation with peanut-specicandArah
2-specic IgE levels at baseline. Higher frequencies of IL-4+and IFNγ+CD4+
T cells post-OIT are negatively correlated with SU. Our ndings provide evi-
dence that an immune signature consisting of certain CD8+Tcellsubsetfre-
quencies is potentially predictive of SU following OIT.
Oral immunotherapy (OIT), during which food allergens are gradually
introduced at increasing doses until a maintenance dose is reached, has
been successful in desensitizing patients to offending food antigens
and many trials administering peanut OIT have been performed13.
While these trials have demonstrated the efcacy of peanut OIT to
desensitize participants to peanut, the proportion of such desensitized
participants achieving sustained unresponsiveness (SU), i.e., a lack of
clinical reactivity following a period of OIT discontinuation is highly
variable13. Moreover, while some aspects of the immune mechanisms
resulting in desensitization (i.e., DS, for the purposes of this study,
dened by the recurrence of any allergic reaction upon food challenge
after a period of withdrawal post-successful OIT outcome) have been
largely elucidated, the immune pathways or biomarkers that may dis-
tinguish SU vs. DS are yet to be fully characterized47.
To address this issue and to gain insight into mechanisms
underlying development of SU, we performed a comprehensive post-
Received: 2 November 2021
Accepted: 19 October 2022
Check for updates
1
Sean N. Parker Center for Allergy and Asthma Research, Department of Medicine, Stanford University School ofMedicine, Stanford, CA,USA.
2
Department of
Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
3
Divisions of Biomedical Informatics and Allergy & Immunology, Cincinnati
Childrens Hospital Medical Center,Cincinnati, OH,USA.
4
Human Immune Monitoring Center, Institute for Immunity, Transplantation, and Infection, Stanford
University School of Medicine, Stanford, CA, USA.
5
Division of Allergy, Immunology, and Blood and Marrow Transplantation, Department of Pediatrics,
University of California, San Francisco, CA, USA.
6
Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.
7
Department of
Biomedical Data Science, Stanford University, Stanford, CA, USA.
8
Department of Microbiology and Immunology, Stanford University School of Medicine,
Stanford, CA, USA.
9
These authors contributed equally: Rosemarie H. DeKruyff, Monali Manohar, Kari C. Nadeau. e-mail: knadeau@stanford.edu
Nature Communications | (2022) 13:6646 1
1234567890():,;
1234567890():,;
Content courtesy of Springer Nature, terms of use apply. Rights reserved
hoc mechanistic analysis of peripheral blood mononuclear cells
(PBMCs) and plasma from participants in our phase 2 OIT POISED
study (Peanut Oral Immunotherapy: Safety, Efcacy, Discovery) (NCT
02103270). The design, objectives, and primary outcomes of this
randomized, double-blind, placebo-controlled (DBPC) clinical trial
have been previously published8. Briey, in this study, after dosage
build-up over ~52 weeks, peanut-allergic patients were maintained on
4000 mg peanut protein daily for another 52 weeks, while a blinded
placebo group received oat our. At week 104, 80 (98.77%) of per-
protocol active participants passed the DBPC food challenge
(DBPCFC) and peanut ingestionwas withdrawnin a group of 51 blinded
participants (i.e., peanut avoidance group) for 12 weeks. At week 117, 21
(41.2%) of per-protocol participants in the peanut avoidance group
passed the 4000mg DBPCFC without any allergic reaction, thus
demonstrating SU. Those 21 participants continued to have DBPCFCs
every 3 months and were allowed to continue peanut OIT dis-
continuation if they passed a DBPCFC. At week 156, 12 months after
peanut discontinuation, only 8 participants passed the DBPCFCs (i.e.,
had achieved long-term SU with no allergic reaction upon food
challenge).
We recently reported higher frequencies of peanut-reactive (pr)
CD8+T cells in peanut-allergic individuals and demonstrated interac-
tions between pr CD8+and Th2 cells leading to enhancement of
allergic pathways9,10. Based on these ndings, we postulated a pr CD8+
T cell-based mechanism in distinguishing SU vs. DS outcomes. Fur-
thermore, our group and others have demonstrated distinct functions
of innate immune cells in food allergy11,12, and so we further hypothe-
sized that pr CD8+T cells along with innate immune cell subsets likely
impact the possibility achieving SU.
To test thesehypotheses,we perform high dimensional functional
immunophenotyping of peanut-stimulated, PMA/Ionomycin-stimu-
lated, and unstimulated PBMCs with mass cytometry, and plasma and
peanut-stimulated PBMC supernatants with Luminex technology from
all per-protocol trialparticipants. We quantitate immune changes from
SU (i.e., SU dened as those who had no allergic reaction at the
DBPCFC at week 117 or later) vs. DS (i.e., those who had allergic reac-
tions to the DBPCFC at week 117) at week 117. To identify potential
biomarkers predictive of SU, we examine differences in immune pro-
les of SU vs. DS participants at baseline (BL, i.e., pre-OIT) as well as at
the end of OIT at week 104 (i.e., after successful desensitization). One
of the primary baseline inclusion criterion for study participation was
tolerance to 500 mg of cumulative peanut protein dose during the
DBPCFC at the screening stage. The participants who successfully
passed 4000 mg peanut OFC at week 104, but failed at a peanut dose
of 500 mg at week 117 represented a subset having relapsed to the
baseline threshold following mere 12 weeks of avoidance. Thus, as a
nested analysis of participants at the ends of SU vs. DS outcome
spectrum, we compare a subset of DS participants, who failed week 117
DBPCFC at a peanut protein dose 500 mg (n= 7) with the 8 SU par-
ticipants,whopassedallDBPCFCsuptoweek156.Wealsoexamine
peanut OIT-induced immune changes by comparing active vs. placebo
groups at week 104.
Our results show that lower frequencies of naive CD8+T cells and
terminally differentiated, CD57+pr CD8+T cells at BL are strongly
associated with SU. On the other hand, higher frequencies of IL-4+and
IFNγ+CD4+T cells post-OIT are negatively correlated with SU. In
summary, we discover a distinct CD8+T cell pattern in vivo that is
associated with the impairment of achieving SU during food allergy
OIT. In addition, our data conrm the attenuation of Th2 phenotype
post-OIT.
Results
Peanut OIT leads to attenuation of Type 2 phenotype
Figure 1a depicts the POISED study design (top panel) and the work-
ow for mechanistic assays (bottom panel). High dimensional
intracellular and extracellular proteins in peanut-stimulated PBMCs
from participants on active peanut OIT vs. placebo were quantied by
mass cytometry at week 104.
The frequencies of IL-4+,IL-9
+and IL-10+peanut-reactive (pr) CD4+
T cells (i.e., CD69+CD40L+CD4+T cells) among total CD4+Tcellswere
signicantly reduced in participants on active OIT (Fig. 1b; q= 0.002,
0.09 and 0.04, respectively). Peanut-reactive CD4+cells also demon-
strated a reduced frequency and expression of Th2-polarization sur-
rogate marker OX40 in active vs. placebo groups (Fig. 1b; q=0.01and
0.03, respectively), indicating downregulation of the Type 2 pheno-
type in the active peanut OIT-treated, desensitized participants.
Peanut-reactive CD4+T cells also demonstrated increased frequencies
of subpopulations expressing the costimulatory molecule CD27 and
the activation marker CD38 in active vs. placebo (Fig. 1b; q=0.046and
0.092, respectively). Similar differences in OX40 and CD27 expression
were observed inactive vs. placebo comparison of pr CD4+cell clusters
identied through FlowSOM-based unsupervised clustering (Supple-
mentary Fig. 1a, b; q= 0.01 and 0.006, respectively). Expression of
other functional markers evaluated in pr CD4+T cells including IL-17,
LAP (TGF-β)andIFN-γand frequencies of pr CD4+and pr CD8+Tcells
among peanut-stimulated PBMCs were similar in active vs. placebo
comparison at week 104 (Supplementary Fig. 1c; q> 0.1). Also, fre-
quencies of all the manually gated immune subpopulations examined
within the scope of our mass cytometry panel including Treg
(CD25hiCD127-CD4+), NKT (CD56+CD3+αβ T), naive γδ (CD45RA+
CCR7+γδTCR+CD3+) T cells, and plasmablasts (CD27hiCD38hiHLA-
DR+CD19+CD3-) among peanut-stimulated and unstimulated PBMCs
were comparable across active and placebo participants at week 104
(Supplementary Fig. 1d; q>0.1).
Analysis of supernatants of peanut-stimulated PBMCs by Luminex
assay showed that expression of the Type 2 cytokines IL-4, IL-5, IL-9
and IL-13 was signicantly decreased at week 104 in active vs. placebo
(q= 0.09, 0.006, 0.005 and 0.006, respectively) (Fig. 1c, d). This
marked decrease in IL-4, IL-5, IL-9 and IL-13 was also evident when
comparing their levels in peanut-stimulated culture supernatants of
active participantsPBMCs at week 104 with those at BL (q= 0.05, 0.09,
0.09 and 0.09, respectively) (Fig. 1e, f), further supporting a shift away
from a Type 2 prole. Apart from Type 2 cytokine changes, peanut-
stimulated PBMC culture supernatant from active participants showed
an increase in Monocyte Chemoattractant Proteins MCP1 and MCP3
(active vs. placebo, q= 0.02 and 0.01, respectively), as well asIL-12 p70
and FLT3 Ligand (q= 0.03 and 0.07, respectively) compared with
placebo (Supplementary Fig. 1e). In contrast, peanut-stimulated PBMC
culture supernatants from active participants showed a marked
increase in the eosinophil chemotactic protein Eotaxin at week 104 vs.
BL (q= 0.05), and a concomitant decrease in proinammatory cyto-
kines including RANTES, TNF, and IL-22 (Supplementary Fig. 1f;
q= 0.098, 0.065 and 0.059, respectively).
CD8+T cell subset distinctions as a hallmark of SU vs. DS
outcome
While all 51 per-protocol active participants randomized into the
avoidance group were successfully desensitized at week 104, 12 weeks
of peanut avoidance thereafter led to two distinct clinical outcomes,
viz., SU (n=21) vs. DS (n= 30) (no vs. with allergic reactions on the
4000 mg DBPCFC at week 117, respectively). To identify changes in
immune markers or cell type frequencies that might identify SU par-
ticipants, we compared immune changes in peanut-stimulated and
unstimulated PBMCs from SU participants with those from DS at BL,
week 104, and week 117. Interestingly, in both peanut-stimulated and
unstimulated PBMCs, differences distinguishing SU vs. DS were iden-
tied within CD8+T cell subsets but not in CD4+T cells. Peanut-rea ctive
(pr) CD8+T cells (i.e., CD69+CD8+) among peanut-stimulated PBMCs of
SU participants were primarily of effector memory (EM; CD45RA-CCR7-
) phenotype while those from DS participants exhibited a naive
Article https://doi.org/10.1038/s41467-022-34222-8
Nature Communications | (2022) 13:6646 2
Content courtesy of Springer Nature, terms of use apply. Rights reserved
(CD45RA+CCR7+) phenotype (Fig. 2a). Also, pr CD8+TcellsfromDS
participants had a signicantly higher frequency and expression of
CD57 (q=0.079 and 0.079, respectively), a marker of terminal differ-
entiation (Fig. 2a). Similarly, EM and TEMRA (CD45RA+CCR7) CD8+T
cell subsets of peanut non-reactive (i.e., CD69-)CD8
+T cells among
peanut-stimulated PBMCs from DS participants showed a signicantly
higher frequency and expression of CD57 (Fig. 2b; q= 0.019, 0.05, 0.02
and 0.02, respectively). This CD57 expression pattern among EM and
TEMRA CD8+cells was evident in unstimulated PBMCs as well (Fig. 2c;
SU vs. DS q= 0.05, 0.009, 0.03 and 0.01, respectively). Interestingly,
CD56dimCD16+NK cells, a subset involved in host protective immunity
throughcytolysisalongwithCD8
+T cells, from DS participants also
Article https://doi.org/10.1038/s41467-022-34222-8
Nature Communications | (2022) 13:6646 3
Content courtesy of Springer Nature, terms of use apply. Rights reserved
showed an elevated frequency and expression of CD57 at BL and at
week 117 in peanut-stimulated (Supplementary Fig. 2a; q= 0.059 and
0.068, respectively) and unstimulated PBMCs (Supplementary Fig. 2a;
q= 0.02 and 0.01, respectively).
In addition, among peanut non-reactive CD8+T cells in peanut-
stimulated PBMCs, the frequency of naive CD8+cells was substantially
lower in SU vs. DS participants at BL, week 104 and week 117 (Fig. 2d;
q= 0.02, 0.01 and 0.04, respectively). Unstimulated SU vs. DS BL, week
104 and week 117 PBMCs also reected these differences in naive CD8+
T cell frequency (Fig. 2e; q= 0.02, 0.008 and 0.03, respectively). A
signicantly reduced frequency of CD8+naive T cells among total live
cells was also detected in SU vs. DS participants at BL, week 104 and
week 117 using unsupervised clustering of peanut-stimulated PBMCs
(q= 0.09, 0.009 and 0.09, respectively), and unstimulated PBMCs
(q= 0.09, 0.01 and 0.06, respectively) (Supplementary Fig. 2bd). On
the contrary, SU participants showed a markedly higher frequency of
peanut non-reactive EM CD8+cells among peanut-stimulated and
ustimulated total CD8+cells at week 104, and week 117 PBMCs com-
pared to DS participants (Supplementary Fig. 2e; q= 0.02, 0.03
respectively, and Supplementary Fig. 2f q= 0.025 each). This trend of
higher EM CD8+T cell frequency could be observed at BL as well.
Frequencies of pr CD4+and pr CD8+T cells among peanut-
stimulated PBMCs and expression of the evaluated functional markers
within these subsets were similar between SU and DS participants at
BL, week 104, and week 117 (Supplementary Fig. 3ah; q>0.1). Parti-
cularly, while there were no signicant differences in the respective
frequencies and no signicant differences in the expression of IL-4, IL-
9, and IL-10 between SU vs. DS participants at week 117, SU participants
had lower frequencies of IL-4+and IL-9+pr CD4+T cells and lower
expression of IL-4 compared to DS participants at BL, week 104,as well
as week 117 (Supplementary Fig. 3bd). The frequencies of all the
manually gated immune subpopulations examined within the scope of
our mass cytometry panel including Treg, NKT, naive γδ T cells and
plasmablasts among peanut-stimulated and unstimulated PBMCs were
comparable across SU and DS participants at BL, week 104 and week
117 (Supplementary Fig. 3il; q>0.1).
Eligible participants further avoided peanut consumption past
week 117 in a blinded protocol, and eight participants in this group
successfully tolerated a 4000 mg peanut DBPCFC at week 156, thereby
having achieved long-termSU (henceforth termed, SU week 156)
following 1 year of peanut avoidance (Supplementary Fig. 4a). Immune
changes in these SU week 156 participants were compared with those
of participants who failed week 117 DBPCFC at a dose lower than
500 mg peanut (termed DS 500 mg; n= 7). Such subset selection
afforded immune evaluation of participants representing each end of
the treatment outcome spectrum. The frequency of IL-9+pr CD4+
T cells at week 104 and expression of IL-9 among pr CD4+T cells at
week 117 were signicantly lower in the SU week 156 vs. DS 500 mg
(Supplementary Fig. 4b; q= 0.03 and 0.05, respectively). While the
CD57 expression and frequency among CD8 subsets did not sig-
nicantly differ in SU week 156 vs . DS 500mg, peanut-stimulated and
unstimulated week 117 PBMCs from SU week 156 exhibited a lower
frequency of CD57+CD56dimCD16+NK cells than DS500 mg
(Supplementary Fig. 4c; q= 0.05 and 0.06, respectively). The fre-
quency of naive CD8+T cells in total live peanut-stimulated and
unstimulated PBMCs from SU week 156 participants was substantially
lower at week 104 (Supplementary Fig. 4d, e; q= 0.002 and 0.003,
respectively) and week 117 (Supplementary Fig. 4d, e; q= 0.002 and
0.006, respectively) compared with those in the DS 500 mg group.
Notably, on comparing the frequencies of naive CD8+TcellsinSU
week 156 (n= 8), DS week 130 to 156 (i.e., per-protocol participants,
who passed at week 117, but failed at week 130/143/156; n=10), DS
week 117 > 500 mg (n= 23), and DS week 117 500 mg (n=7) at BL,
week 104, and week 117, a trend of gradual increase in the frequency of
naive CD8+cells going from SU week 156 to DS week 117 500 mg was
observed (Supplementary Fig. 4f, g).
DifferencesinCD86expressiononAPCsubsetsfromSUvs.DS
participants
Besides CD8+T cell subsets, we found signicant differences in CD86
expression on APC subsets in SU vs. DS. The frequencies of CD86
expressing memory B (CD27+CD38+HLA-DR+CD19+CD3-) cells among
peanut-stimulated PBMCs (Fig. 2f; q= 0.039) and that among classical
monocytes (CD14+CD16-HLA-DR+) and myeloid dendritic cells 2
(mDC2; CD11c+CD123-CD14-HLA-DR+) among unstimulated PBMCs
from SU were markedly lower than DS at BL (Fig. 2g; q=0.05and0.03
respectively). Intermediate monocytes (CD14+CD16+HLA-DR+)and
myeloid dendritic cells 1 (mDC1; CD11c+CD123+CD14-HLA-DR+)among
unstimulated PBMCs from SU participants also showed a lower
expression of CD86 (Fig. 2g; q= 0.03 and 0.01, respectively). In addi-
tion, a singular Luminex readout distinctive of SU vs. DS was lower
levels of IL-2 in peanut-stimulated PBMC culture supernatants at BL
(Fig. 2h; q=0.056).
Similarly, reduced CD86+frequency or CD86 expression was seen
in APC subsets among PBMCs from SU week 156 group compared to
the DS 500mg group of participants. The frequency of CD86+clas-
sical monocytes and memory B cells among peanut-stimulated week
104 PBMCs, and frequency of CD86+classicalmonocytes, intermediate
monocytes and mDC1s among unstimulated BL PBMCs, was sig-
nicantly lower in SU week 156 vs. DS 500 mg (Supplementary
Fig. 4h, i; q= 0.067, 0.080, and 0.007, 0.06 and 0.02, respectively).
Classical monocytes, mDC1, and mDC2 subsets at BL from SU week 156
participants also showed lower expression of CD86 than DS 500 mg
participants (Supplementary Fig. 4i; q= 0.03, 0.02 and 0.02,
respectively).
Correlations between distinctive immune subsets and specic
immunoglobulin readouts
We and others have shown that an increased peanut-specicand
peanut component-specic IgG4/IgE ratio driven by increased
allergen-specic IgG4 is associated with induction of SU8,13,14. Analyses
were thus performed to probe correlations of peanut- or peanut
component-specic IgE, IgG4 and IgG4/IgE antibody levels at BL with
immune cell subset frequencies, which we had observed to be asso-
ciated with SU in this study. We observed a positive correlation of the
frequency of naive CD8+T cells in peanut-stimulated PBMCs with
Fig. 1 | Peanut OIT effects an immune shift away from Th2 phenotype. a Top
panel: schematic representation of POISED study timeline and sample collection
time points. Bottom panel: depiction of approach for mechanistic analyses. Here,
SUstands for sustained unresponsive and DSstands for desensitized alone
participants. Illustration created with BioRender.com. bImmune cell subsets
among peanut-stimulated week104 PBMCs from participants on active peanut OIT
(n= 80) were compared against those treated with placebo (n=15).Signicant
frequency (i.e., percentage) and expression (i.e., MSI) changes among peanut-
reactive (pr) CD4+T cells are shown. c,dSignicant changes in peanut-stimulated
week 104 PBMC culture supernatant of active participants (n= 16) compared to
those treated with placebo (n=7)as a heatmap (c) and as individual boxplots (d).
e,fSignicant changes in peanut-stimulatedweek 104 PBMC culture supernatantof
activeparticipants(n= 13) comparedto BL as a heatmap (e)and individualboxplots
(f). In the boxplots, medians are shown, and the hingesrepresent the rst and
third quartile. The whiskers are the smallest and largest values after exclusion of
outliers (greater than the 75th percentile plus 1.5 times the interquartile range
(IQR),or less than 25th percentileminus 1.5 times theIQR). qvalues were comp uted
using the χ2tests in mixed-effects models, adjusted for multiple hypothesistesting
using the Benjamini and Hochberg method. Source data are provided as a Source
Data le.
Article https://doi.org/10.1038/s41467-022-34222-8
Nature Communications | (2022) 13:6646 4
Content courtesy of Springer Nature, terms of use apply. Rights reserved
peanut-specic IgE (Fig. 3a; p=0.43, p=0.002)aswellasArah
2-specic IgE (Fig. 3a; p=0.47, p< 0.001) levels at BL. Such positive
correlation with peanut- and Ara h 2-specic IgE was also observed
with naive CD8+T cells among unstimulated PBMCs (Fig. 3a; p=0.48
and 0.53 respectively; p< 0.001 for both). Notably, participants who
demonstrated SU at week 117 had both signicantly lower levels of
peanut-specic IgE at BL and signicantly lower numbers of naive CD8+
T cells at BL (Fig. 2a) than participants that were DS (Fig. 3a). On the
other hand, the BL frequency of EM CD8+cells among peanut-
stimulated total CD8+cells exhibited a signicant negative correlation
with BL peanut-specic IgE (Supplementary Fig. 5a; p=0.43,
p= 0.0016). Similarly, a signicant negative correlation was observed
Fig. 2 | CD8+T celldifferentiation statusdistinguishes SU participants from DS.
Immune cellsubsets among peanut-stimulated (a,b,d,f) and unstimulated (c,e,g)
PBMCs from SU participants were compared with DS participants at BL, week 104,
and week 117. acSignicantfrequency and CD57 expressiondifferences amongpr
CD8+Tcells(a), and peanut non-reactive CD8+Tcells(b) among peanut-stimulated
(SU, n=20;DS,n= 29), and unstimulated (c)(SU,n= 20; DS, n= 26), BL PBMCs.
d,eSignicant differences in the naive CD8+T cell frequency in peanut-stimulated
(d) and unstimulated (e) PBMCs at BL (pea stim: SU, n=20,DS,n= 29; unstim: SU,
n=20,DS,n=26), week 104 (pea stim:SU, n= 21, DS, n= 30; unstim: SU,n= 20, DS,
n= 29), and week 117 (pea stim:SU, n= 17, DS, n= 27; unstim: SU, n=17,DS,n=27).
f,gSignicant differences in the frequency or expression of CD86 in memory B
cells among peanut-stimulated PBMCs (f) and APC subsets among unstimulated
PBMCs (g) at BL (pea stim: SU, n=20,DS,n=29;unstim:SU,n=20,DS,n=26).hIL-
2 levels in peanut-stimulated BL PBMC culture supernatants from SU (n=7)vs.DS
(n= 7) participants. Boxplots show medians and hingesrepresent the rst and
third quartile. Whiskers denote the smallest and largest values after outlier exclu-
sion. qvalues computed using the χ2tests in mixed-effects models, adjusted for
multiple hypothesis testing using the Benjamini and Hochberg method. Source
data are provided as a Source Data le.
Article https://doi.org/10.1038/s41467-022-34222-8
Nature Communications | (2022) 13:6646 5
Content courtesy of Springer Nature, terms of use apply. Rights reserved
between the frequency of EM CD8+cells among peanut-stimulated
total CD8+cells and Ara h 2-specic IgE at BL (Supplementary Fig. 5b;
p=0.46, p= 0.0008). To further dissect this intriguing link between
relative frequencies of naive vs. EM CD8+T cell subsets with serological
readouts and hence SU as an outcome, we probed a convenience
sample set of week 52 PBMCs from 5 SU vs. DS participants each.
Interestingly, unstimulated and PMA/Ionomycin (PMAi)-stimulated
CD8+cells from SU participants exhibited a signicantly higher fre-
quency of the key cytolytic effector molecule, Granzyme B (GzB)-
expressing subset compared to DS participants. A signicantly higher
GzB expression was observed in PMAi-stimulated CD8+cells from SU
participants as well compared to DS participants (Supplementary
Fig. 5c; p< 0.01). Overall, EM CD8+followed by TEMRA CD8+Tcells
primarily contributed to GzB production. Nevertheless, DS partici-
pants had a lower mean proportion of GzB+EM CD8+cells (43.28% vs.
54.14%) and a higher mean proportion of GzB+naive CD8+cells (18.46
vs. 9.44%) compared to SU participants (Supplementary Fig. 5d).
As expected, we found that the frequency of IL-4+peanut-
reactive CD4+T cells was positively correlated with the level of Ara h
2-specic IgE (Fig. 3b; p= 0.44, p= 0.002) and negatively correlated
with the ratio of Ara h 2-specic IgG4/IgE at BL (Fig. 3b; p=0.45,
p= 0.002). Those participants who developed SU displayed lower
values of Ara h 2-specic IgE and higher ratios of Ara h 2-specic
IgG4/IgE1. Also, the frequency of CD57+pr CD8+T cells at BL nega-
tively correlated with peanut and Ara h 2-specic IgG4/IgE levels
(Fig. 3c; p=0.41, p= 0.005).
Supervised learning-based identication of features predictive
of SU vs. DS outcome
As an independent approach to validate the above-mentioned obser-
vations, we applied supervised learning on the BL CyTOF readouts to
identify features that could potentially predict the outcome as SU vs.
DS (Supplementary Fig. 6a, b). We thus simulated the frequencies and
marker expressions of manually gated immune cell subsets in all the
participantsstudied (300 times data simulation). Each of thesimulated
dataset was then used to build an elastic net machine learning (ML)
model, from which the most important features were elucidated (see
Methods). Here, the data simulation not only enabled probing of the
smallsamplesize(n= 46 at BL), but also facilitated capturing the
important features conserved in the respective group even after con-
stricted random sampling15. The elastic net model could correctly
predict the SU participants with anaverage AUC of 0.94. In agreement
with CyTOF analyses, the most informative features included fre-
quencies of naive CD8+T cells and CD57+TEMRA CD8+Tcellsthus
endorsing the current ndings.
Lower frequencies of certain IFNγ+T cell subsets post-OIT in SU
participants
We further probed immune differences in SU vs. DS participants by
non-specically stimulating PBMCs with PMAi. Lower frequencies of
IL-4+CD4+memory T effector (CD45RA-CD127+CD25lo)cellswere
observed in SU compared with DS participants at week 104, which was
consistent with a reduced Type 2 response (Fig. 4a; q= 0.02). Inter-
estingly, lower frequencies of IFNγ+CD4+memory T effector cells at
weeks 104 and 117 (Fig. 4a; q=0.07 and 0.04 respectively), and lower
expression levels of IFNγby CD4+memory T effector cells (Fig. 4a;
q=0.06)andmemoryγδ Tcells(Fig.4b; q= 0.044) were observed in
SU vs. DS participants at week 104. Unsupervised clustering analysis
also corroborated lower expression levels of IFNγby CD4+memory T
effector cells of SU participants compared to DS at week 117 (Supple-
mentary Fig. 7a; q= 0.003). Moreover, memory and naive γδ T cells in
unstimulated PBMCs displayed lower expression levels of IFNγ-
inducible chemokine receptor CXCR3 at week 104 from SU vs. DS
participants (Fig. 4b). Concomitantly, signicantly lower levels of MIG
(CXCL9), an IFN-γ-induced ligand for CXCR3, were also observed in SU
compared with DS participantsplasma at week 117 (Fig. 4c). Thus,
lower expression of IFN-γin memory γδ T cells was consistent with the
lower expression of CXCR3 and reduced levels of CXCL9.
Since CD8+T cells and IFN-γhave been implicated as contributing
to the development of asthma16, we asked if the age ofwheezing onset,
a prelude to clinical asthma diagnosis by spirometry at the age of 7
years or older, differed in the participants that developed SU com-
pared with DS participants. Notably, most DS participants reported
wheezing onset at a signicantly lower age than SU (Fig. 4d; p=0.04).
Age at onset of other atopic comorbidities, such as atopic dermatitis
and allergic rhinitis, if present, as well as age at diagnosis of peanut
allergy were comparable among SU vs. DS participants (Supplemen-
tary Fig. 7b; p= 0.51, 1 and 0.80, respectively).
Fig. 3 | Naive CD8+,IL-4
+pr CD4+,andCD57
+pr CD8+T cell subsets at baseline
signicantly correlate with peanut and Ara h 2-specic immunoglobulin
readouts. a Spearman correlations of frequency of naive CD8+T cells among total
live cells from peanut-stimulated(top panels) andunstimulated(bottom panels)BL
PBMCs with peanut-specic, and Ara h 2-specicIgE.bSpearman correlations of
frequency of peanut-reactive IL-4+CD4+cellsamong total pr CD4+T cells in peanut-
stimulated PBMCs with Ara h 2-specic IgE and Ara h 2-specic IgG4/IgEratio at BL.
cSpearman correlation of frequency of peanut-reactive CD57+CD8+cells within
total pr CD8+T cells in peanut-stimulatedPBMCs with Ara h 2-specicIgG4/IgE ratio
at BL (pea stim: SU, n=20, DS, n=29; unstim:SU,n=20,DS, n= 26). Data are
presented as the mean ± SE. pvalues in the gure were determined by an unpaired
two-tailed Studentsttest. Plots display the correlation coefcient (ρ)andlinear
regression line with a 95% condence interval shading. Sourcedata are provided as
a Source Data le.
Article https://doi.org/10.1038/s41467-022-34222-8
Nature Communications | (2022) 13:6646 6
Content courtesy of Springer Nature, terms of use apply. Rights reserved
Discussion
Our overarching aim for this study was to understand those T-cell
changes that might elucidate why SU develops only in a subset of
desensitized participants following a period of allergen withdrawal.
Using agnostic computer-based learning algorithms as well as
hypothesis-based analyses from high dimensionalimmune studies, our
ndings suggest a pivotal function of CD8+T cell differentiation status
in dening the likelihood of SU. Our ndings suggest that SU is favored
by: (i) a lower frequency of terminally differentiated, CD57+allergen-
reactive, and allergen non-reactive CD8+T cellspossibly coupled with a
higher frequency of cytotoxic EM CD8+T cells, (ii) a lower frequency of
naive CD8+T cells, (iii) a lower expression of CD86 on APCs and/or a
lower frequency of CD86+APCs pre-OIT, and (iv) a lower frequency of
IFN-γ+CD4+memory T effectors post-OIT.
While many clinical studies have conrmed the efcacy of peanut
OIT in conferring desensitization, only a handful have tested the
induction of SU during the post-avoidance period. Moreover, the
immune mechanistic studies performed during a subset of such SU
testing trials primarily have been focused on serological readouts (i.e.,
peanut-specic IgE and IgG4), basophil activation, measurement of
Th2 cytokines, and changes in the frequency of total and peanut-
reactive CD4+Tregs and Teffectors14,1720. A recent study examining
longitudinal transcriptional changes and clonality of peanut-reactive
Th cell subsets from peanut OIT participants offers further rened
Fig. 4 | Higher frequency and expression of IFNγas an additional distinctive
featureof DS vs. SU participants.a Frequency of IL-4+CD4+memoryT effectorsat
week 104, frequenciesof IFNγ+CD4+memory T effectors at week104 and week 117,
and expression of IFNγin CD4+memory T effectors at week 104 in PMAi-stimulated
PBMCs from SU vs. DS [week 104 (PMAi stim:SU, n= 21, DS, n= 29); week 117 (PMAi
stim: SU, n= 17, DS, n=27)].bExpression ofIFNγin memory γδ TcellsamongPMAi-
stimulated week 104 PBMCs from SU vs. DS; expression of CXCR3 in memory and
naive γδ T cells amongunstimulated PBMCsat week 104 [week 104 (PMAi stim: SU,
n=21,DS, n= 29; unsti m: SU, n= 20, DS, n= 29); week 117 (PMAi stim and unstim:
SU, n= 17, DS, n= 27)]. cPlasma CXCL9 level compared between SU (n=8)andDS
(n= 13) at week 117. In the boxplots, medians are shown,and the hingesrepresent
the rst and third quartile. The whiskers are the smallest and largest values after
exclusion of outliers (greater than the 75th percentile plus 1.5 times the inter-
quartile range (IQR), or less than 25th percentile minus 1.5 times the IQR). qvalues
were computed using the χ2tests in mixed-effects models, adjusted for multiple
hypothesis testing using the Benjamini and Hochberg method. dAge of wheezing
onset for SU participants with asthma (n= 11) was compared with that for DS par-
ticipants (n=15).pvalue wascomputed using unpaired,two-sided, non-parametric
MannWhitney test. Source data are provided as a Source Data le.
Article https://doi.org/10.1038/s41467-022-34222-8
Nature Communications | (2022) 13:6646 7
Content courtesy of Springer Nature, terms of use apply. Rights reserved
insights into OIT-induced changes and features predictive of post-OIT
outcome in the context of CD4+T cells21. However, a comprehensive
evaluation of circulating immune cell frequencies and functional
markers, and the cumulative potential impact of any signicant
changes distinguishing SU in a given OIT cohort, has not been pre-
viously performed. Our analyses put forth such an evaluation by
employing high dimensional, multi-parametric tools such as mass
cytometry and Luminex. Our study thus provides unique insights into
immune players in the induction of OIT-induced tolerance.
Notably, we observed signicant differences beyond Th2 and Treg
subsets while comparing immunophenotypes of SU vs. DS participants
within our CyTOF dataset. In agreement with previous reports,
attenuation of Th2 phenotype was observed exclusively among active
participants post-OIT compared to placebo-treated participants, while
no change was noted in the frequencies of pr CD4+T cells and Tregs
post-OIT17,21. Interestingly, in addition to prototypical Th2 cytokines,
the frequency of a regulatory cytokine- IL-10+pr CD4+T cells was seen
to be diminished in active participants post-OIT. This might be
attributed to an initialautocrine IL-10 induction in pathogenic pr CD4+
T cells in response to peanut dose build-up followed by a marked
decrease by week 104 post-OIT22. However, CD4+T cell subset read-
outs did not signicantly distinguish SU vs. DS in our study or those
by others14,17. Instead, higher frequencies of naive CD8+and lower
frequencies of EM CD8+T cells, at BL and throughout the course of
OIT, identied those individuals with less likelihood of becoming SU.
Our preliminary data demonstrate that CD8+T cells from SU parti-
cipants have higher GzB-mediated cytolytic potential. Of note, GzB
has been shown to be critical for TCR-induced death of Th2 cells23,24
thus raising a possibility of EM CD8+cell sourced-GzB conferring SU
participants a selective advantage by regulating pathogenic peanut-
reactive Th2 proliferation, and thus peanut-specic IgE during
avoidance. The signicant negative correlation observed between
the BL frequency of EM CD8+cells and peanut-specic as well as Ara h
2-specic IgE reinforces this possibility. Investigations are underway
to evaluate how GzB from EM CD8+cells may preferentially target
immune cell subsets in the IgE pathway, and inuence of CD57
expression on GzB function in this context. The higher frequency of
naive CD8+T cells, despite higher expression of CD86 on APC subsets
and a higher level of IL-2 in peanut-stimulated PBMC culture super-
natants, observed in DS compared to SU participants at BL likely
implies a higher threshold of activation. Of note, our group has
recently reported hypomethylation of the IL-2 locus, and conse-
quently higher levels of IL-2, in peanut-stimulated PBMC culture
supernatants as one epigenetic readout distinguishing peanut-
allergic twins from their non-allergic counterparts25.
Concomitantly, a higher frequency and expression of CD57 on EM
and TEMRA CD8+T cells in DS participants suggests terminal differ-
entiation and immunosenescence. Early childhood viral exposure,
chronic in vivo inammation, and natural aging may drive such term-
inal differentiation. The speculation regarding early childhood viral
exposure also isconsistent with the signicantly lower age of wheezing
onset we diagnosed for DS participants (Fig. 4d).
CD57 and CD27 have reciprocal expression patterns in T cell
ontology26. Interestingly, terminally differentiated CD27-
CRTH2+CD161+memory allergen-reactive CD4+Tcells,termedTh2A
cells, have been strongly associated with allergies27.Despitebeing
frequently associated with senescence, CD57+CD8+T cells subsets
have been reported to be highly proinammatory26. We also observed
a higher frequency of IFNγ+CD4+T effectors amon g post-OIT PBMCs of
DS participants on PMA/Ionomycin stimulation. Thus, taken together,
preexistence of terminally differentiated, proinammatory CD8+and
CD4+subsets compounded with a high-activation-threshold, reluc-
tant-to-differentiateCD8+T cells in DS participants, likely pose
immune plasticity challenges. These challenges might be mitigated
during active dosing of OIT leading to successful desensitization,
although they can resurface post-avoidance, and are reected through
clinical relapse/reactivity to allergens.
There are limitations to our study. Firstly, not all immune subsets
of interest such as Tfh subsets21,28,Th2a
27 cells could be monitored
through our CyTOF panel. Secondly, a thorough longitudinal analysis
inclusive of PBMCs and plasma from additional time points over the
course of OIT was not feasible due to limited sample availability. In
addition, unlike manual gating, FlowSOM-based unsupervised clus-
tering analysis could not identify certain rare immune cell subsets as
clusters, particularly pr CD8+T cells. Semi-supervised cell type anno-
tation algorithm- CyAnno29 developed by our group will likely yield
a better output in this regard. In addition, the ML models used to
identify features for classication of SU vs. DS groups were based on
small sample size of training and testing datasets, which may have led
to model over-tting, and would be a major limitation of models
proposed in this study and features identied. To address this issue, we
simulated the real dataset 300 times and built elastic net models for
each of them. Here, the elastic netmodel was appliedbecause it uses L1
and L2 regularization penalties for model building and the algorithm
has been proven to be useful for similar cases where the number of
features are larger than the number of samples. To further minimize
the effect of model over-tting, we only prioritized the SU vs. DS
classication features that are conserved across 300 elastic net model.
The current ML model based ndings nonetheless warrant validation
using a similar, independent dataset of food OIT study participants
tested for SU.
Higher levels of allergen- or allergen-component-specicIgEpre-
OIT have been reported in the past to predict a negative outcome
towards SU13,14. Frequency of naive CD8+T cells positively correlated
with peanut and Ara h 2-specic IgE, while abundance of CD57+pr CD8+
T cells negatively correlated with the Ara h 2-specic IgG/IgE ratio at
BL. Thus, the CD8+T cell differentiation status, along with the fre-
quency of IL-4+and IL-9+pr CD4+T cells, may be considered as
potential biomarkers predictive of SU following OIT. Prospective
peanut-allergic participants presenting with such skewed CD8+Tcell
differentiation may benet from continued peanut dosing. In addition,
adjunctive treatment with anti-IgE, which has been shown to reduce
the frequency of CXCR3+CD8+Tcells
30 could be helpful in certain
patients. Moreover, future research to examine the function of CD8+T
cell subsets in the context of food allergy pathogenesis and SU needs
to be extended to include regulatory CD8+Tcells
31 and
Tc2 subpopulations32,theidentication of which was beyond the
scope of our CyTOF panel.
The CyTOF and Luminex data presented here conrmed
attenuation of Th2 phenotype as well as reduction in proinammatory
cytokines in active participants at week 104 post-OIT. In addition, the
Luminex data also showed signicant changes in monocyte and DC
differentiation-related mediators such as GCSF, GMCSF and FLT3L, as
well as a reduction in tissue homeostasis factors viz. IL-22 and VEGF,
which warrant further investigation.
Taken together, this study demonstrates that frequencies of non-
Th2 immune cells, primarily CD8+T cell subsets, may dictate the like-
lihood of achieving sustained unresponsiveness following food
allergy OIT.
Methods
Study participants
The clinical research protocol for POISED study was approved by the
Division of Allergy, Immunology, and Transplantation (DAIT) and the
National Institute of Allergy and Infectious Diseases (NIAID) Allergy
and Asthma Data Safety Management Board, the DAIT/NIAID Clinical
Review Committee, the Stanford Institutional Review Board, and the
US Food and Drug Administration (FDA). Written informed consent
was obtained from adult participants or parents/ guardians of minor
participants along with assent from minor participants of age 7 years
Article https://doi.org/10.1038/s41467-022-34222-8
Nature Communications | (2022) 13:6646 8
Content courtesy of Springer Nature, terms of use apply. Rights reserved
and older. All compliant participants were given $30 gift cards as a
compensation for each study visit as consistent with Stanford IRB-
approved protocol. The study protocol, patient demographics, and
treatment outcomes have been described in detail previously8.Briey
120 peanut-allergic participants (age: 755 years) were randomized
into active vs. placebo group. Participants in the active arm (n=95)
were built up and maintained on 4000mg peanut protein through
week 104. Fifty-one of the total per-protocol active arm participants
were randomized to discontinue peanut consumption post-week 104
(peanut-0 group). DBPCFCs to 4000 mg peanut protein were done at
BL and weeks 104, 117, 130, 143, and 156. Participants in peanut-0
group,who passed oral food challenge (OFC) at week 117 (i.e., Peanut-0
Successes) were denoted as Sustained unresponsive, while those,
who failed week 117 OFC (i.e., Peanut-0 Failures) were termed as
Desensitized. Baseline demographics for all the participants in the
study have been previously published8. Supplementary Table 1 shows
baseline demographic characteristics for the participants per rando-
mization (i.e., active vs. placebo) and outcome (i.e., SU vs. DS), and for
the subset, whose samples were analyzed through Luminex.
Blood draws and processing
From each study participant, ~40 mL blood was drawn byvenipuncture
and collected in EDTA tubes at baseline (BL, i.e., during the initial
screening phase or at week 0 post-enrollment), and every 3 months
over the course of treatment. PBMCs and plasma isolated by Ficoll-
based density gradient centrifugation were frozen in aliquots, and
stored in liquid nitrogen at 80 °C, until thawing for respective assays.
In these analyses, we used PBMCs and plasma stored at BL, week 104,
and week 117.
Mass cytometry
Mass cytometry studies were performed on PBMC samples at BL, week
104, and week 117 as listed in Supplementary Table 2. PBMCs frozen
per time point per participant were thawed and rested overnight at
37 °C with 5% CO
2
in RPMI + 10% FBS, and Pen-Strep. Cells were
counted, and plated in 3 round-bottom wells of a 96-well plate at the
density of 3 × 106cells in culture with 300 μL RPMI, 5% FBS, Pen-Strep
per well. To evaluate the allergen (peanut)-induced response, PBMCs
in one of the wellswere stimulated with 200 μg/mL peanut solution for
24 h with the addition of brefeldin A (5μg/mL; BioLegend, San Diego,
CA) for the last 4 h. PBMCs in the second well were left unstimulated,
treated with brefeldin A for 4 h before harvesting and served as con-
trols. PBMCs in the third well were stimulated with 20 ng/mL PMA +1
μg/mL Ionomycin (Sigma-Aldrich) for4 h in the presenceof brefeldin A
to measure cytokine expression. The downstream steps comprising
cell harvesting, staining, were followed as previously described12.
Briey, the cells were harvested at the end of the incubation period,
washed with CyFACS buffer (0.1% bovine serum albumin, 0.1%
sodium azide, 2 mM EDTA in PBS), and stained with surface antibody
cocktail (Supplementary Data 1) for 30 min at room temperature in
dark followed by staining with live/dead 115-DOTA maleimide. The
cells were then washed with CyFACS buffer and barcoded using Cell-
ID 20-Plex Pd Barcoding Kit (Fluidigm Co., Product# 201060)
according to the manufacturers instructions. Barcoded cells were
xed with 2% paraformaldehyde overnight at 4 °C. Following a
CyFACS wash the next day, equal number of cells per sample were
pooled, permeabilized, and stained with intracellular antibody
cocktail (Supplementary Data 1) for 30 min at room temperature. The
cells were then washed twice with permeabilization buffer followed
by two washes with CyFACS buffer. Finally, the pooled samples were
stained with Ir interchelator, washed twice in CyFACS buffer, fol-
lowed by one wash in CyPBS and two further washes in MilliQ water.
Approximately 300,000 cells per sample in each pool were acquired
on a Helios mass cytometer (Fluidigm Co.). Data normalization,
concatenation and debarcoding were performed using CyTOF
software (Fluidigm Co.). Data were analyzed by manual gating as well
as FlowSOM-based unsupervised clustering analysis.
Manual gating on normalized, de-barcoded data les was carried
out using FlowJo v10 (FlowJo LLC., Ashland, OR). Gating scheme used
to identify and analyze CD3+and CD3-immune cell subsets has been
previously illustrated30 as well as depicted in Supplementary Fig. 8a, b.
Workow illustrations were generated using BioRender application.
The FlowSOM-based unsupervised computational analysis was
performed separately for peanut-stimulated, unstimulated, and PMA/
ionomycin-stimulated samples at independent time points (i.e., week
0, week 104, and week 117)33. CD127 was excluded to minimize batch
effects, and the remaining lineage markers were transformed using
arcsinh function (inverse hyperbolic sine; cofactor of 5), followed by
their further normalization with landmark alignment procedure using
the warpSet function from the owStats R package (version 4.4.0)34.In
each analysis 50,000 live cells were randomly selected and subjected
to FlowSOM-based clustering with default parameters and with a xed
number of 30 clusters. A dened set of lineage markers were used for
cell clustering (Supplementary Table 3). Distinct clusters represented
as a heatmap (R package pheatmap, version 1.0.12)35 were manually
annotated by visual inspection of median expression of lineage mar-
kers per cluster. In addition, 100,000 cells were randomly selected
from the predicted clusters for further visualization (R package
ggplot2 version 3.3.3)36 by applying the non-linear dimensionality
reduction technique- Uniform Manifold Approximation and Projection
(UMAP) at the lineage marker levels using the R package UMAP (ver-
sion 0.2. 7.0, default parameters except min_dist = 0.25)37.
Signicant differences in frequency (i.e.,proportionor abundance
within a given parent population) of cell subsets and median intensity
(i.e., expression) of functional markers within a cell subset were eval-
uated by manual gating as well as unsupervised clustering analysis. For
both these approaches, pvalues were computed using the χ2tests in
mixed-effects models, wherein dened groups were used as xed-
effects and batch number as random-effects using lmerTest R
package38. The computed pvalues were adjusted to control the FDR
during multiple hypothesis testing using the Benjamini and Hochberg
method. Readouts with pvalue < 0.05 and adjusted pvalue (i.e., Q
value) <0.1 have been reported as signicant in this analysis. Any
extreme outlier data point(s) were identied and removed using
identify_outliers() function (coef = 3.0) from rstatix R package (Ver-
sion 0.7.0)39.
Luminex assay
Luminex analysis was performed on a convenience sample subset
(Supplementary Table 1). PBMCs were thawed, rested for 6 h, and
plated in 2 round-bottom wells of a 96-well plate at the density of
1.4 × 106cells in culture with 200 μL RPMI, 5% FBS, Pen-Strep per well.
PBMCs in one of the wells were stimulated with 200μg/mL peanut
solution for 18 h, while the ones in the other well were left unstimu-
lated for that duration. A total of 130 μL culture supernatant was har-
vested per well in prelabeled microfuge tubes and stored at 80 °C
until performing the assay. Culture supernatants were probed with
Human 48-plex Cytokine/Chemokine magnetic bead panel, while
plasma samples were tested using a combination of 48-plex and 23-
plex Cytokine/Chemokine magnetic bead panel (MilliporeSigma, Bur-
lington, MA). While comparing the readouts (MFIs), pvalues were
computed using Wilcoxon signed-rank sum test (unpaired and paired
where Time is used as group), and adjusted for multiple hypothesis
testing using Benjamini and Hochberg method. The rstatix R package
was used to identify and remove any extreme outlier data point(s)39.
Evaluation of Granzyme B expression by ow cytometry
Week 52 PBMCs from 5 SU and DS participants each were thawed and
rested overnight. Approximately 0.3 million PBMCs were stimulated
with 20 ng/mL PMA and 1 μg/mL Ionomycin for 4 h, and equivalent
Article https://doi.org/10.1038/s41467-022-34222-8
Nature Communications | (2022) 13:6646 9
Content courtesy of Springer Nature, terms of use apply. Rights reserved
number of PBMCs per participant was left unstimulated. Brefeldin A
(5 μg/mL) and Monensin (2 μM) was added to PBMCs during PMAi
stimulation as well as to unstimulated PBMCs for 4 h prior to har-
vesting. On harvesting, cells were stained with antibodies listed in
Supplementary Data 1. Data were acquired on CyTek Dx10 ow cyt-
ometer and analyzed using FlowJo v10 (FlowJo LLC., Ashland, OR).
Spearman correlations
Spearman correlations between mass cytometry and serological
parameters were performed for SU and DS participants. The correla-
tion coefcient (Rho; p) and associated pvalue signicance was com-
puted using cor.test() R function. Correlations with ǀpǀ> 0.4 and
p< 0.001 were considered as signicant and were shortlisted for sub-
sequent evaluation. The resulting correlations were plotted using
ggplot R function and linear regression line with 95% condence
interval shading was computed using geom_smooth() R function
(method = lm).
Machine learning analysis
To identify the BL mass cytometry (CyTOF) parameters that distin-
guish SU participants from DS, we applied a Machine learning (ML)
approach on BL simulated datasets (n= 300; see Supplementary
Fig. 6a, b). Data simulation was used to compensate for the small
sample size (n= 46 at BL) available for model training as well as to
obtain condence intervals for model performance evaluation, using
the simstudy R package40. Here, the data were simulated (300 times)
from manually gated frequencies and marker expression of cell types
from BL CyTOF samples with a set of constraints, including sample
size, mean, and variance per participant p,i.e.,X
p
~N(μ,σ2). Each of the
simulated dataset had the same sample size as the original BL dataset
with overall same mean and variance of CyTOF parameters in the
respective participant (i.e., 46 samples and 123 CyTOF parameters).
For ML prediction, we used elastic net algorithm whichis a regularized
logistic regression method that uses a linear and weighted combina-
tion of L1 and L2 regularization of the lasso and ridge methods41.This
ML algorithm is considered as suitable for the datasets wherein there
are more features (123 features) than instances (i.e., 46 participants).
Here, the glmnet R package (version 4.1-2) was used for executing
elastic net, wherein the model parameters were tuned using k-fold
cross-validation42. Finally, each of the given simulated dataset was used
for building elastic net model and the feature importance matrix were
computed for respective dataset. The model accuracy was evaluated
by comparing the predicted outcome with expected outcome, and
after 300 runs, the performance statistics, including specicity, sen-
sitivity, and area under the curve (AUC) were also evaluated. Robust
CyTOF parameters in each elastic net model were then dened as
those features with score greater than 80 (arbitrary selected threshold;
max(score) = 100 and min(score) = 0). The cumulative importance
scoreforeachfeaturewasdened as:
Cumulative importance= Pi=iterations
i=1 δ
iterations ×100 ð1Þ
Where, iteration is the number of times (=300) simulated datasets
were modeled, and δis the feature score computed for each model,
and its value in each iteration iis dened as:
δ=1, if δi,>80
1, if δi0, 80
ð2Þ
Reporting summary
Further information on research design is available in the Nature
Research Reporting Summary linked to this article.
Data availability
The raw unlabeled POISED Mass Cytometry dataset les generated in
this study in FCS 3.0 format, associated metadata, and the gating
schema have been deposited in the FlowRepository database under
accession codes FR-FCM-Z4MA and FR-FCM-Z2V9. The processed CSV
les are provided in the Source Data le, which is available on the
dryad digital repository under the DOI accession 10.5061/
dryad.95x69p8p6. Source Data are provided with this paper.
References
1. Chinthrajah, R. S. et al. Oral immunotherapy for peanut allergy: the
pro argument. World Allergy Organ. J. 13, 100455 (2020).
2. Tang,M.L.K.,Lozinsky,A.C.&Loke,P.Peanutoralimmunotherapy:
state of the art. Immunol. Allergy Clin. North Am. 40,97110
(2020).
3. Patrawala, M., Shih, J., Lee, G. & Vickery, B. Peanut oral immu-
notherapy: a current perspective. Curr. Allergy asthma Rep. 20,
14 (2020).
4. Yu, W., Freeland, D. M. H. & Nadeau, K. C. Food allergy: immune
mechanisms, diagnosis and immunotherapy. Nat. Rev. Immunol. 16,
751765 (2016).
5. Kulis,M.D.,Patil,S.U.,Wambre,E.&Vickery,B.P.Immune
mechanisms of oral immunotherapy. J. Allergy Clin. Immunol. 141,
491498 (2018).
6. Tordesillas, L. & Berin, M. C. Mechanisms of oral tolerance. Clin. Rev.
Allergy Immunol. 55,107117 (2018).
7. Schoos, A. M. et al. Immunological outcomes of allergen-specic
immunotherapy in food allergy. Front. Immunol. 11,568598
(2020).
8. Chinthrajah, R. S. et al. Sustained outcomes in oral immunotherapy
for peanut allergy (POISED study): a large, randomised, double-
blind, placebo-controlled, phase 2 study. Lancet (Lond., Engl.) 394,
14371449 (2019).
9. Yu,W.etal.Allergen-specic CD8(+) T cells in peanut-allergic
individuals. J. Allergy Clin. Immunol. 143,19481952 (2019).
10. Yu, W., Zhou, X., Lyu, S. C., Davis, M. M. & Nadeau, K. C. Regulation
of peanut-specic CD8(+) T cells from nonallergic individuals. J.
Allergy Clin. Immunol. 147,385387.e381 (2021).
11. Zhou, X. et al. A positive feedback loop reinforces the allergic
immune response in human peanut allergy. J. Exp. Med. 218,
e20201793 (2021).
12. Neeland, M. R. et al. Mass cytometry reveals cellular ngerprint
associated with IgE+ peanut tolerance and allergy in early life. Nat.
Commun. 11,1091(2020).
13. Tsai,M.,Mukai,K.,Chinthrajah,R.S.,Nadeau,K.C.&Galli,S.J.
Sustained successful peanut oral immunotherapy associated with
low basophil activation and peanut-specicIgE.J. Allergy Clin.
Immunol. 145,885896.e886 (2020).
14. Vickery, B. P. et al. Sustained unresponsiveness to peanut in sub-
jects who have completed peanut oral immunotherapy. J. Allergy
Clin. Immunol. 133, 468475 (2014).
15. Suárez-Fariñas, M. et al. Predicting development of sustained
unresponsiveness to milk oral immunotherapy using epitope-
specic antibody binding proles. J. Allergy Clin. Immunol. 143,
10381046 (2019).
16. Ray,A.,Camiolo,M.,Fitzpatrick,A.,Gauthier,M.&Wenzel,S.E.Are
we meeting the promise of endotypes and precision medicine in
asthma? Physiol. Rev. 100,9831017 (2020).
17. Blumchen, K. et al. Oral peanut immunotherapy in children with
peanut anaphylaxis. J. Allergy Clin. Immunol. 126,8391.e81
(2010).
18. Syed, A. et al. Peanut oral immunotherapy results in increased
antigen-induced regulatory T-cell function and hypomethylation of
forkhead box protein 3 (FOXP3). J. Allergy Clin. Immunol. 133,
500510 (2014).
Article https://doi.org/10.1038/s41467-022-34222-8
Nature Communications | (2022) 13:6646 10
Content courtesy of Springer Nature, terms of use apply. Rights reserved
19. Narisety, S. D. et al. A randomized, double-blind, placebo-
controlled pilot study of sublingual versus oral immunotherapy for
the treatment of peanut allergy. J. Allergy Clin. Immunol. 135,
12751282 (2015). e12711276.
20. Vickery,B.P.etal.Earlyoralimmunotherapy in peanut-allergic
preschool children is safe and highly effective. J. Allergy Clin.
Immunol. 139,173181.e178 (2017).
21. Monian, B. et al. Peanut oral immunotherapydifferentiallysup-
presses clonally distinct subsets of T helper cells. J. Clin. Invest. 132,
e150634 (2022).
22. Wambre, E. Effect of allergen-specic immunotherapy on
CD4+ T cells. Curr. Opin. Allergy Clin. Immunol. 15,
581587 (2015).
23. Devadas, S. et al. Granzyme B is critical for T cell receptor-induced
cell death of type 2 helper T cells. Immunity 25,237247
(2006).
24. Sharma, V., Delgado, M. & Ganea, D. Granzyme B, a new player in
activation-induced cell death, is down-regulated by vasoactive
intestinal peptide in Th2 but not Th1 effectors. J. Immunol. (Baltim.,
Md.: 1950) 176,97110 (2006).
25. Zhou, X. et al. Targeted DNA methylation proling reveals epige-
netic signatures in peanut allergy. JCI Insight 6, e143058 (2021).
26. Larbi, A. & Fulop, T. From truly naïveto exhausted senescent
T cells: when markers predict functionality. Cytometry A 85,
2535 (2014).
27. Wambre, E. et al. A phenotypically and functionally distinct human
TH2 cell subpopulation is associated with allergic disorders. Sci.
Transl. Med. 9, eaam9171 (2017).
28. Gowthaman, U. et al. Identication of a T follicular helper cell
subset that drives anaphylactic IgE. Science (New York, N.Y.) 365,
eaaw6433 (2019).
29. Kaushik, A. et al. CyAnno: a semi-automated approach for cell type
annotation of mass cytometry datasets. Bioinformatics (Oxford,
England) 37,41644171 (2021).
30. Manohar, M. et al. Immune changes beyond Th2 pathways during
rapid multifood immunotherapy enabled with omalizumab. Allergy
76, 28092826 (2021).
31. Yu, Y. et al. Recent advances in CD8(+) regulatory T cell research.
Oncol. Lett. 15,81878194 (2018).
32. Gelfand,E.W.&Hinks,T.S.C.Istherearolefortype2CD8(+)Tcells
in patients with steroid-resistant asthma? J. Allergy Clin. Immunol.
144,648650 (2019).
33. Van Gassen, S. et al. FlowSOM: using selforganizing maps for
visualization and interpretation of cytometry data. Cytom. Part A 87,
636645 (2015).
34. Hahne, F. owStats: Statistical methods for the analysis of ow
cytometry data. (2022). http://www.github.com/RGLab/owStats.
35. Kolde, R. pheatmap: Pretty Heatmaps. (2019). https://cran.rproject.
org/web/packages/pheatmap/index.html.
36. Wickham, H., Chang, W. & Wickham, M. H. Package ggplot2.
Create Elegant Data Visualisations Using the Grammar of Graphics.
Version 2,1189 (2016).
37. McInnes, L., Healy, J. & Melville, J. Umap: uniform manifold
approximation and projection for dimension reduction. Preprint at
arXiv:1802.03426 (2018).
38. Kuznetsova, A., Brockhoff, P. B. & Christensen, R. H. lmerTest
package: tests in linear mixed effects models. J. Stat. Softw. 82,
126 (2017).
39. Kassambara, A. rstatix: pipe-friendly framework for basic statistical
tests. (2020). https://cran.rproject.org/web/packages/rstatix/
index.html.
40. Goldfeld, K. & Wujciak-Jens, J. simstudy: Illuminating research
methods through data generation. J. Open Source Softw. 5,
2763 (2020).
41. Zou, H. & Hastie, T. Regularization and variable selection via the
elastic net. J. R. Stat. Soc.: Ser. B (Stat. Methodol.) 67,
301320 (2005).
42. Friedman, J., Hastie, T. & Tibshirani, R. Regularization Paths for
Generalized Linear Models via Coordinate Descent. J Stat Softw. 33,
122 (2010).
Acknowledgements
This work was supported through the Sean N. Parker Center for Allergy
and Asthma Research at Stanford University and NIAID AADCRC grant
U19AI10420. We thank our patients and their families for their partici-
pation and the staff members of the Sean N. Parker Center for their
support, Drs Yael Rosenberg-Hasson, Ramona Hoh, and Anita Kant for
their advice, and Dr Vanitha Sampath for her help with reviewing and
compiling this manuscript.
Author contributions
K.C.N., S.J.G., S.D.B., and H.T.M. designed the study. R.S.C., S.B.S., and
L.E.K. steered and oversaw trial recruitment, progress, and clinical data
management. S.G. optimized mass cytometry panel. D.D. performed
mass cytometry on all the PBMC samples. M.M. devised manual gating
scheme and analytical strategy. E.D. and M.M. performed manual gating.
X.H. performed all the experimental steps and panel selection for
Luminex analysis. A.F. and M.M. evaluated Granzyme B output from
CD8+T cell subsets using ow cytometry. A.K. analyzed and plotted all
the data in the manuscript including machine learning analysis. S.A.,
M.D., and R.T. guided data analysis, visualizations, and the statistical
evaluations listed in the manuscript. A.K., M.M., R.D.K., and K.C.N. wrote
the manuscript. D.D. and X.H. contributed equally to this work as second
authors. S.J.G. was the overall PI of the NIH-funded U19 study. S.J.G.,
S.D.B., H.T.M., M.T., S.A., M.D., R.T., and W.Y. critically reviewed the
manuscript.
Competing interests
K.C.N. reports grants from National Institute of Allergy and Infec-
tious Diseases (NIAID), National Heart, Lung, and Blood Institute
(NHLBI), National Institute of Environmental Health Sciences
(NIEHS), and Food Allergy Research & Education (FARE); stock
options from IgGenix, Seed Health, ClostraBio, and ImmuneID; is a
Director of World Allergy Organization (WAO), Advisor at Cour
Pharma, Consultant for Excellergy, Red tree ventures, Eli Lilly, and
Phylaxis, Co-founder of Before Brands, Alladapt, Latitude, and
IgGenix; and National Scientic Committee member at Immune
Tolerance Network (ITN), and National Institutes of Health (NIH)
clinical research centers, outside the submitted work; patents
include, Mixed allergen composition and methods for using the
same,Granulocyte-based methods for detecting and monitoring
immune system disorders,Methods and Assays for Detecting and
Quantifying Pure Subpopulations of White Blood Cells in Immune
System Disorders,and Methods of isolating allergen-specic
antibodies from humans and uses thereof. R.S.C. reports grants
from NIAID, CoFAR, Aimmune, DBV Technologies, Astellas,
Regeneron, Stanford Maternal and Child Health Research Institute
(MCHRI), and FARE. She is an Advisory Board Member at Alladapt
Therapeutics, Novartis, Genentech, Sano, Allergenis, and Nutricia.
S.B.S. reports grants from NIH, Regeneron, DBV, AIMMUNE,
Novartis, CoFAR, and FARE. She receives personal fees from Astra
Zeneca, DBV, and honoraria from FARE. S.J.G. reports grants from
the NIH. S.D.B. has consulted for Regeneron, Sanoand Novartis
on topics unrelated to this study, owns shares in AbCellera, has
patents awarded or submitted related to immunoglobulin gene and
protein analysis, and reports grants from the NIH. S.A. reports
grants from the NIH. All others authors declare no competing
interests.
Article https://doi.org/10.1038/s41467-022-34222-8
Nature Communications | (2022) 13:6646 11
Content courtesy of Springer Nature, terms of use apply. Rights reserved
Additional information
Supplementary information The online version contains
supplementary material available at
https://doi.org/10.1038/s41467-022-34222-8.
Correspondence and requests for materials should be addressed to
Kari C. Nadeau.
Peer review information Nature Communications thanks the anon-
ymous reviewer(s) for their contribution to the peer review of this work.
Reprints and permissions information is available at
http://www.nature.com/reprints
Publishers note Springer Nature remains neutral with regard to jur-
isdictional claims in published maps and institutional afliations.
Open Access This article is licensed under a Creative Commons
Attribution 4.0 International License, which permits use, sharing,
adaptation, distribution and reproduction in any medium or format, as
long as you give appropriate credit to the original author(s) and the
source, provide a link to the Creative Commons license, and indicate if
changes were made. The images or other third party material in this
article are included in the articles Creative Commons license, unless
indicated otherwise in a credit line to the material. If material is not
included in the articles Creative Commons license and your intended
use is not permitted by statutory regulation or exceeds the permitted
use, you will need to obtain permission directly from the copyright
holder. To view a copy of this license, visit http://creativecommons.org/
licenses/by/4.0/.
© The Author(s) 2022
Article https://doi.org/10.1038/s41467-022-34222-8
Nature Communications | (2022) 13:6646 12
Content courtesy of Springer Nature, terms of use apply. Rights reserved
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com
... The potential mechanisms behind this cell reduction remain unclear, although deletion, anergy, and exhaustion may all contribute (44). There was a notable decrease in the frequencies of IL-4 + , IL-9 + , and IL-10 + peanut-reactive CD4 + T cells among total CD4 + T cells in peanutallergic individuals following OIT (47). Additionally, it was discovered that, compared with desensitization, higher frequencies of both IL-4 + CD4 + T cells and IFN-g + CD4 + T cells post-OIT displayed a significant negative correlation with sustained unresponsiveness (47). ...
... There was a notable decrease in the frequencies of IL-4 + , IL-9 + , and IL-10 + peanut-reactive CD4 + T cells among total CD4 + T cells in peanutallergic individuals following OIT (47). Additionally, it was discovered that, compared with desensitization, higher frequencies of both IL-4 + CD4 + T cells and IFN-g + CD4 + T cells post-OIT displayed a significant negative correlation with sustained unresponsiveness (47). Meanwhile, there was a significant reduction in the Th2-polarization surrogate marker OX40 expression in CD4 + T cells in peanut-allergic individuals after desensitization (47). ...
... Additionally, it was discovered that, compared with desensitization, higher frequencies of both IL-4 + CD4 + T cells and IFN-g + CD4 + T cells post-OIT displayed a significant negative correlation with sustained unresponsiveness (47). Meanwhile, there was a significant reduction in the Th2-polarization surrogate marker OX40 expression in CD4 + T cells in peanut-allergic individuals after desensitization (47). The study by Michael Kulis et al. (48) demonstrated a significant increase in peanut-responsive CD4 + T cells during the first four and eight months of peanut OIT in both high-and low-dose groups compared to baseline. ...
Article
Full-text available
Nowadays, the management of food allergies has increasingly moved from conventional oral immunotherapy (OIT) to low-dose OIT or low-dose OIT utilizing hypoallergenic foods. This shift is largely because the latter appears to induce oral tolerance with fewer adverse effects than the former. However, the mechanisms underpinning such differences remain unclear. To better understand these mechanisms, we conducted a comparative study scrutinizing the mechanisms of OIT, especially those of low-dose desensitization. We also summarized articles on low-dose OIT and low-dose OIT using hypoallergenic foods. We examined the efficacy, safety, and immunological parameters of low-dose OIT and those of low-dose OIT with hypoallergenic foods with the aim of shedding some light on low-dose OIT and its therapeutic application in inducing oral tolerance for individuals with food allergies.
... ↓ Th2 genes and ↑ type 1 IFN expression by CD4+ T cells from children who achieve SU post probiotic and peanut OIT [25] ↓ CD4+ cell frequency after 3 months egg OIT, returning to baseline by 8 months OIT Naïve γδ Tregs after 24 weeks peanut OIT are transcriptionally similar to those found in healthy children [21] Memory γδ Tregs retain Th2 genes (OX40R, GITR) and have upregulated immunosuppressive genes (CD69, ISG20, CTLA4, TGFB). Indicates early modulation of Th2 gene expression in memory γδ Tregs due to peanut OIT [21] ↓ IFNγ expression in memory γδ T cells at week 104 peanut OIT (PBMCs stimulated with PMA/Ionomycin) linked to SU [27] Continuous increase in γδ T during first 8 months of egg OIT ↓ Th2 cytokines in PBMCs post-therapy: IL-13, IL-9 (unstimulated) and IL-4 (PMA/Ionomycin-stimulated) [30] ↑ in allergen specific IgG4 ratios by week 36 of therapy [30] ↓ IL-17, IL-1β, MCP-1, IL-12p40, GM-CSF, and FLT3L in unstimulated PBMCs, and ↓ IL-17, IL-8, and MIP1α in stimulated PBMCs by week 36 with combined peanut OIT and OIT [30] Peanut-IgG4, Ara h 1-IgG4, and Ara h 2-IgG4 levels increase then decrease without returning to baseline [31] Peanut ↓ peanut-IgE, Ara h 1-IgE, Ara h 2-IgE, SPT, and peanut IgE: total IgE ratios during peanut OIT [31] Participants who discontinue OIT have ↑ peanut-IgE and Ara h 2-IgE at 12 months versus participants who continue treatment [31] Participants who tolerated the final challenge post-omalizumab + OIT had lower Ara h 1-3 IgE and higher Ara h 2/6 IgG4 compared to the failure group [32] Over time, CD3/CD28-stimulated PBMCs showed a reduced Th2 cytokine profile (IL-5, IL-13, IL-9) and increased IFNγ, indicating a shift in T cell polarity due to OIT [33] Dupilumab monotherapy ↓ ps-IgE and ↑ ps-IgG4/IgE ratio [34] Dupilumab ...
... Ashley et al., 25 found that children (aged 1-10 years, n = 62) undergoing probiotic and peanut OIT who achieved remission/ sustained unresponsiveness (SU), defined as passing a 3950 mg cumulative peanut protein food challenge 2-6 weeks after ceasing treatment, have a distinct rewiring in Th2/IFN gene modules. This included a loss of Th2 gene expression and upregulation of type I interferon genes in purified CD4+ T cells when exposed to crude peanut protein in vitro, while CD4+ T cells from children who received a placebo for both OIT and probiotic treatment (remained allergic) retained a Th2 profile (IL-4, IL-9, IL-13, IL-31).Kaushik et al.27 examined PBMCs from participants in the POISED study (median participant age: 5 years, n = 120) at several time points throughout peanut OIT. They found a consistent reduction in Th2-polarized peanut-reactive CD4+ T cells throughout treatment, and corresponding reductions in the expression of type 2 proteins IL-4, IL-5, IL-9, and IL-13. ...
Article
Full-text available
IgE‐mediated food allergies are common and can be life‐threatening, especially for children. With increasingly rapid advances in immunological technologies, including the ability to profile highly complex immune features from small sample volumes, our understanding of the immune mechanisms that underpin the development of food allergies continues to grow. This also extends to the immune mechanisms associated with the outcomes of oral immunotherapy (OIT). This review focuses on studies within the past 5 years related to immune signatures associated with food allergy in childhood, immune responses that determine reaction severities to offending allergens, immune alterations that occur during OIT in children, and immune effects of adjunct therapies including omalizumab, dupilumab, and abrocitinib. We conclude by providing a perspective on current evidence and directions for future research that will enable new prediction and screening tools and facilitate the development of effective curative strategies.
... [1][2][3] The mechanistic analysis of our phase 2 peanut OIT POISED trial cohort samples 4 demonstrates that a higher likelihood of achieving SU is associated with a higher frequency of effector memory (EM; CD45RA − CCR7 − ) CD8 + T cells. 5 We therefore further investigated the transcriptional and protein expression distinctions among CD8 + T cells in a subset of SU vs. desensitized alone (DS) POISED participants 4 ( Figure 1A; Table S1). ...
... We bulk-sorted total CD8 + T cells from thawed, peanut- We previously showed that EM CD8 + T cells from SU participants express lower CD57 and higher Granzyme B (GzB) compared to DS participants. 5 Since GNLY featured as a differentially expressed transcript in SU vs. DS at baseline as well as week 117, we sought to evaluate Granulysin (Gnly) expression among SU vs. DS participants' CD8 + T cells at baseline in conjunction with GzB and CD57 using spectral flow cytometry ( Figure 1A; Table S2). Interestingly, significant distinctions were identified in total CD8 + T cell subsets among peanut-stimulated PBMCs, but not in peanut-reactive (CD69 + ) CD8 + T cells. ...
... Notably, although CD8 + T cells have been only occasionally described in patient cohorts or animal studies in the allergy fields [38][39][40][41] , the percentage of CCR4 + CXCR3¯type-2 cytotoxic CD8 + T cells (CD8 + Tc2, Fig. 3d, e) 42 among living cells was also higher in VAP vs. HC at each time point including baseline, while the percentage of CCR4¯CXCR3 + type-1 CD8 + T cells (CD8 + Tc1) was modestly but significantly decreased ( Supplementary Fig. 3A). CD8 + Tc2-like cells, referred to as the direct parent gate of CD8 + Tc2 ( Supplementary Fig. 1), were also among the two significantly higher immune subsets in PAP vs. HC at several time points (Supplementary Figs. ...
Article
Full-text available
Allergen-specific immunotherapy (AIT) induces immune tolerance, showing the highest success rate (>95%) for insect venom while a much lower chance for pollen allergy. However, the molecular switches leading to successful durable tolerance restoration remain elusive. The primary outcome of this observational study is the comprehensive immunological cellular characterization during the AIT initiation phase, whereas the secondary outcomes are the serological and Th2-cell-type-specific transcriptomic analyses. Here we apply a multilayer-omics approach to reveal dynamic peripheral immune landscapes during the AIT-initiation phase in venom allergy patients (VAP) versus pollen-allergic and healthy controls. Already at baseline, VAP exhibit altered abundances of several cell types, including classical monocytes (cMono), CD4⁺ hybrid type 1-type 17 cells (Th1-Th17 or Th1/17) and CD8⁺ counterparts (Tc1-Tc17 or Tc1/17). At 8-24 h following AIT launch in VAP, we identify a uniform AIT-elicited pulse of late-transitional/IL-10-producing B cells, IL-6 signaling within Th2 cells and non-inflammatory serum-IL-6 levels. Sequential induction of activation and survival protein markers also immediately occur. A disequilibrium between serum IL-6 and cMono in VAP baseline is restored at day seven following AIT launch. Our longitudinal analysis discovers molecular switches during initiation-phase insect-venom AIT that secure long-term outcomes. Trial number: NCT02931955.
... We systematically screened the FlowRepository database for large, high-quality CyTOF datasets (MIFlowCyt score > 0.5) uploaded within the last 5 years. The largest multidimensional CyTOF dataset meeting these criteria was utilized for our work which has been previously published 1 (24). The dataset comprises of 126,873,075 cells, distributed in 779 human peripheral blood mononuclear cell (PBMC) samples that were collected from 120 intent-to-treat participants, divided into three different groups (Peanut stimulated, Unstimulated, and PMA/Ionomycinstimulated). ...
Article
Full-text available
High-dimensional cell phenotyping is a powerful tool to study molecular and cellular changes in health and diseases. CyTOF enables high-dimensional cell phenotyping using tens of surface and intra-cellular markers. To utilize the full potential of CyTOF, we need advanced clustering and machine learning methodologies to enable automated gating of the complex data. Here we show that critical modifications to a machine learning based FlowSOM package and precise parameter optimization can enable us to reliably analyze the complex CyTOF data. We show the impact of key parameters on clustering outcomes while addressing bugs within the publicly available package. We modified the FlowSOM pipeline to fix the bugs, enable scalability to handle large datasets and perform parameter optimization. We further validated this modified pipeline on a substantial external immunological dataset demonstrating the need of data-specific tailored parameter optimization to ensure reliable definition and interrogation of immune cell populations associated with immune disorders.
... In contrast, in PBMCs, we observed a decrease in this population in both groups (p < 0.05 vs. saline), which may be due to a fluctuation in the immune response or a weakening of its regulatory capacity. CD8 + Tregs may also play an inhibitory role in the inflammatory response, although CD8 + T cells are uncommonly thought to be involved in IgE-mediated food allergy [42][43][44]. CD8 is a membrane glycoprotein that binds to the major histocompatibility complex class I (MHC class I) proteins. It is mainly found on cytotoxic T lymphocytes but also on dendritic cells, NK cells, and double-positive thymocytes. ...
Article
Full-text available
Background/objectives: Cow's milk allergy is one of the most common food allergies in children, and its pathomechanism is still under investigation. Recently, an increasing number of studies have linked food allergy to intestinal barrier dysfunction. The present study aimed to investigate changes in the intestinal microenvironment during the development of β-lactoglobulin (β-lg) allergy under conditions of early intestinal dysfunction. Methods: BALB/c mice received intraperitoneal β-lg with Freund's adjuvant, followed by oral β-lg while receiving dextran sulphate sodium salt (DSS) in their drinking water (0.2% w/v). The immunized group without DSS and the groups receiving saline, oral β-lg, or DSS served as controls. Results: The study showed that the immunization effect was greater in mice with mild intestinal barrier dysfunction. Although DSS did not affect the mice's humoral response to β-lg, in combination with β-lg, it significantly altered their cellular response, affecting the induction and distribution of T cells in the inductive and peripheral tissues and the activation of immune mediators. Administration of β-lg to sensitized mice receiving DSS increased disease activity index (DAI) scores and pro-inflammatory cytokine activity, altered the distribution of claudins and zonulin 1 (ZO-1) in the colonic tissue, and negatively affected the balance and activity of the gut microbiota. Conclusions: The research model used appears attractive for studying food allergen sensitization, particularly in relation to the initial events leading to mucosal inflammation and the development of food hypersensitivity.
Article
Background Oral immunotherapy is an established approach to desensitize the immune system in the context of allergic disease; however, the only currently approved product is for peanut allergy. ADP101 is a novel, pharmaceutical-grade, multifood oral immunotherapy in development to simultaneously treat single or multiple food allergies, containing allergenic proteins from 15 foods in equal parts by protein weight. Objective The phase 1/2 Harmony trial (NCT04856865) evaluated efficacy and safety of ADP101 in participants with qualifying allergy to 1 to 5 foods in ADP101, defined as dose-limiting symptoms with a ≤100 mg challenge dose during double-blind, placebo-controlled food challenge (DBPCFC). Methods Participants were randomized to low-dose (1500 mg/d; 100 mg protein per food) or high-dose (4500 mg/d; 300 mg protein per food) ADP101, or matched placebo, with dose escalation followed by daily maintenance dosing over 40 weeks. The primary endpoint was the proportion of participants tolerating a ≥600 mg challenge dose of a single qualifying food without dose-limiting symptoms at the Week 40 Exit DBPCFC (ie, responders). Results In the primary analysis population (61 pediatric participants aged 4-17 years), a greater response rate was observed in both the high-dose ADP101 (55.0%) and low-dose ADP101 (38.1%) groups compared with pooled placebo (20.0%) (nominal P = .048, P = .306, respectively; adjusted for multiple comparisons, P = .097, P = .306, respectively). Desensitization to ≥2 foods was observed in individuals with multiple food allergies, as was desensitization at levels over 600 mg. ADP101-treated participants showed an overall reduction in skin-prick test reactivity, with an increase in maximum tolerated dose across the majority of foods tested. Adverse events were mostly mild or moderate, with no life-threatening events or deaths. Conclusions The study did not meet its primary endpoint, but ADP101 demonstrated a favorable safety profile and increased the reactive threshold in DBPCFC in pediatric participants with single or multiple food allergies across multiple endpoints, warranting further clinical investigation.
Article
The mechanisms of tolerance induction in food allergies involve the establishment of oral tolerance along with the development of digestive and mucosal barrier functions. When food antigens are degraded to amino acids, their antigenicity disappears and no immune response is elicited. The mucosal barrier is composed of mucus, intestinal epithelial cells, immunoglobulin A, and antimicrobial peptides that block the entry of antigens in the intestinal tract. Oral tolerance is a physiological phenomenon that suppresses the immune response to antigen recognition in the gastrointestinal tract, and is modulated by antigen-presenting cells, regulatory T cells, and inhibitory cytokines. Besides the intestinal tract, the liver and tonsils are other organs that induce immune tolerance; the intestinal microbiota also contributes to the induction of immune tolerance. The mechanisms of oral immunotherapy are complex, involving both acquired and innate immunity, and negative regulation of the immune response by regulatory T cells and interleukin-10 are particularly important. High doses of antigens induce clonal anergy but may enhance intestinal inflammation and allergic reactions and interfere with tolerance. Therefore, more detailed mechanisms must be elucidated to optimize oral immunotherapy protocols and develop new therapies.
Article
IgE-mediated food allergy (IgE-FA) occurs due to a breakdown in immune tolerance that leads to a detrimental type 2 helper T cell (T H 2) adaptive immune response. While the processes governing this loss of tolerance are incompletely understood, several host-related and environmental factors impacting the risk of IgE-FA development have been identified. Mounting evidence supports the role of an impaired epithelial barrier in the development of IgE-FA, with exposure of allergens through damaged skin and gut epithelium leading to the aberrant production of alarmins and activation of T H 2-type allergic inflammation. The treatment of IgE-FA has historically been avoidance with acute management of allergic reactions, but advances in allergen-specific immunotherapy and the development of biologics and other novel therapeutics are rapidly changing the landscape of food allergy treatment. Here, we discuss the pathogenesis and immunobiology of IgE-FA in addition to its diagnosis, prognosis, and treatment. Expected final online publication date for the Annual Review of Immunology, Volume 42 is April 2024. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Article
Full-text available
Food allergy affects an estimated 8% of children in the US. Oral immunotherapy (OIT) is a recently approved treatment, with outcomes ranging from sustained tolerance to food allergen to no apparent benefit. The immunological underpinnings that influence clinical outcomes of OIT still remain largely unresolved. Using single-cell RNA sequencing and paired TCRα/β sequencing, we assessed the transcriptomes of CD154+ and CD137+ peanut-reactive T helper cells from 12 peanut-allergic patients longitudinally throughout OIT. We observed expanded populations of cells expressing Th1, Th2, and Th17 signatures that further separated into six clonally distinct subsets. Four of these subsets demonstrated convergence of TCR sequences, suggesting antigen-driven T cell fate. Over the course of OIT, we observed suppression of Th2 and Th1 gene signatures in effector clonotypes but not Tfh-like clonotypes. Positive outcomes were associated with stronger suppression of Th2 signatures in Th2A-like cells, while treatment failure was associated with the expression of baseline inflammatory gene signatures that were present in Th1 and Th17 populations and unmodulated by OIT. These results demonstrate that differential clinical responses to OIT are associated both with pre-existing characteristics of peanut-reactive CD4+ T cells and with suppression of a subset of Th2 cells.
Article
Full-text available
Food allergies are a leading cause of anaphylaxis, and cellular mechanisms involving antigen presentation likely play key roles in their pathogenesis. However, little is known about the response of specific antigen-presenting cell (APC) subsets to food allergens in the setting of food allergies. Here, we show that in peanut-allergic humans, peanut allergen drives the differentiation of CD209+ monocyte-derived dendritic cells (DCs) and CD23+ (FcєRII) myeloid dendritic cells through the action of allergen-specific CD4+ T cells. CD209+ DCs act reciprocally on the same peanut-specific CD4+ T cell population to reinforce Th2 cytokine expression in a positive feedback loop, which may explain the persistence of established food allergy. In support of this novel model, we show clinically that the initiation of oral immunotherapy (OIT) in peanut-allergic patients is associated with a decrease in CD209+ DCs, suggesting that breaking the cycle of positive feedback is associated with therapeutic effect.
Article
Full-text available
Background Multifood oral immunotherapy (mOIT) with adjunctive anti‐IgE (omalizumab, XOLAIR®) treatment affords safe, effective, and rapid desensitization to multiple foods, although the specific immune mechanisms mediating this desensitization remain to be fully elucidated. Methods Participants in our phase 2 mOIT trial (NCT02643862) received omalizumab from baseline to week 16 and mOIT from week 8 to week 36. We compared the immune profile of PBMCs and plasma taken at baseline, week 8, and week 36 using high‐dimensional mass cytometry, component‐resolved diagnostics, the indirect basophil activation test, and Luminex. Results We found (i) decreased frequency of IL‐4⁺ peanut‐reactive CD4⁺ T cells and a marked downregulation of GPR15 expression and CXCR3 frequency among γδ and CD8⁺ T‐cell subsets at week 8 during the initial, omalizumab‐alone induction phase; (ii) significant upregulation of the skin‐homing receptor CCR4 in peanut‐reactive CD4⁺ T and Th2 effector memory (EM) cells and of cutaneous lymphocyte‐associated antigen (CLA) in peanut‐reactive CD8⁺ T and CD8⁺ EM cells; (iii) downregulation of CD86 expression among antigen‐presenting cell subsets; and (iv) reduction in pro‐inflammatory cytokines, notably IL‐17, at week 36 post‐OIT. We also observed significant attenuation of the Th2 phenotype post‐OIT, defined by downregulation of IL‐4 peanut‐reactive T cells and OX40 in Th2EM cells, increased allergen component‐specific IgG4/IgE ratio, and decreased allergen‐driven activation of indirectly sensitized basophils. Conclusions This exploratory study provides novel comprehensive insight into the immune underpinnings of desensitization through omalizumab‐facilitated mOIT. Moreover, this study provides encouraging results to support the complex immune changes that can be induced by OIT.
Article
Full-text available
DNA methylation (DNAm) has been shown to play a role in mediating food allergy, however, the mechanism by which it does so is poorly understood. In this study, we used targeted NextGen bisulfite sequencing to evaluate DNAm levels in 125 targeted highly informative genomic regions containing 602 CpG sites on 70 immune-related genes to understand whether DNAm can differentiate peanut allergy (PA) vs non-allergy (NA). We found PA-associated DNAm signatures associated with 12 genes (7 novel to food allergy, 3 associated with Th1/Th2, and 2 associated with innate immunity) as well as DNAm signature combinations with superior diagnostic potential compared to serum peanut specific-IgE for PA vs. NA. Further, we found that following peanut protein stimulation, peripheral blood mononuclear cell (PBMCs) from PA participants showed increased production of cognate cytokines compared to NA participants. The varying responses between PA and NA participants may be associated with the interaction between the modification of DNAm and the interference of environment. Using Euclidean distance analysis, we found that the distances of methylation profile comprising 12 DNAm signatures between PA and NA pairs in monozygotic (MZ) twins were smaller than that in randomly paired genetically unrelated individuals, suggesting that PA related DNAm signatures may be associated with genetic factors.
Article
Full-text available
IgE-mediated food allergies are caused by adverse immunologic responses to food proteins. Allergic reactions may present locally in different tissues such as skin, gastrointestinal and respiratory tract and may result is systemic life-threatening reactions. During the last decades, the prevalence of food allergies has significantly increased throughout the world, and considerable efforts have been made to develop curative therapies. Food allergen immunotherapy is a promising therapeutic approach for food allergies that is based on the administration of increasing doses of culprit food extracts, or purified, and sometime modified food allergens. Different routes of administration for food allergen immunotherapy including oral, sublingual, epicutaneous and subcutaneous regimens are being evaluated. Although a wealth of data from clinical food allergen immunotherapy trials has been obtained, a lack of consistency in assessed clinical and immunological outcome measures presents a major hurdle for evaluating these new treatments. Coordinated efforts are needed to establish standardized outcome measures to be applied in food allergy immunotherapy studies, allowing for better harmonization of data and setting the standards for the future research. Several immunological parameters have been measured in food allergen immunotherapy, including allergen-specific immunoglobulin levels, basophil activation, cytokines, and other soluble biomarkers, T cell and B cell responses and skin prick tests. In this review we discuss different immunological parameters and assess their applicability as potential outcome measures for food allergen immunotherapy that may be included in such a standardized set of outcome measures.
Article
Full-text available
Food allergy (FA) is a growing public health problem with personal, social, nutritional, and economic consequences. In the United States, it is estimated that 8% of children and 10.8% of adults have food allergies. Allergies to peanuts are particularly worrisome as unlike allergies to other allergenic foods, such as milk and egg, which are commonly outgrown by 5 or 10 years of age, 80% of peanut allergies persist into adulthood. The first drug for peanut allergy, Palforzia, was approved by the US Food and Drug Administration (FDA) in January 2020. For other food allergies, the current standard of care for the management of FA is suboptimal and is limited to dietary elimination of the offending allergen, vigilance against accidental ingestion, and treatment of allergic reactions with antihistamines and epinephrine. However, dietary avoidance can be challenging, and it is estimated that approximately 40% of patients with food allergies report at least one food allergy-related emergency department in their lifetime. Reactions, even from minimal exposures, can be life-threatening. Oral immunotherapy (OIT) has been the best researched therapeutic approach for treating FA over the last decade, with clinical trials investigating its efficacy, safety, and ability to improve participants' quality of life (QoL). A number of studies and meta-analyses have shown that OIT treatment is effective in raising the threshold of reactivity to peanuts and other foods in addition to producing a measurable serum immune response to such therapy. Although OIT-related adverse events (AEs) are common during treatment, serious reactions are rare. In fact, while the majority of patients experience AEs related to dosing, most continue daily dosing in hopes of achieving protection against the culprit food. Moreover, the majority of participants report improvement of QoL after OIT and are positive about undergoing OIT. These results show patients' commitment to OIT and their optimism regarding the benefits of treatment. As a first step in therapeutic options to protect from reactions to unintentional ingestion of allergenic foods, and importantly, to address the many psychosocial aspects of living with FA, OIT shows promise. Future research will focus on identifying optimal OIT regimens that maintain protection after therapy and allow for regular food consumption without allergic symptoms. Education and informed shared decision making between patients and providers are essential in optimizing current therapy regimens.
Article
Full-text available
Purpose of the Review Peanut oral immunotherapy (OIT) is one of the most studied experimental therapies for food allergy. With the recently FDA-approved peanut product, Palforzia, the goal of this article is to review the most recent data from clinical trials, discuss recent trends, and anticipate future developments. Recent Findings The latest research suggests that peanut OIT could be a promising option for peanut-allergic patients, with the majority of participants in research studies achieving the primary efficacy endpoint of desensitization, as well as sustained unresponsiveness in select populations. Some studies also showed improvements in food allergy-related quality of life. However, peanut OIT is not without risk or side effects, including potentially serious allergic reactions. Summary Future research will need to evaluate the short- and long-term effectiveness of the therapy in the real-world setting, predictors of important treatment outcomes, and the use of adjunctive therapies that may mitigate some of these allergic reactions.
Article
Motivation For immune system monitoring in large-scale studies at the single-cell resolution using CyTOF, (semi-)automated computational methods are applied for annotating live cells of mixed cell types. Here, we show that the live cell pool can be highly enriched with undefined heterogeneous cells, i.e., ‘ungated’ cells, and that current semi-automated approaches ignore their modeling resulting in misclassified annotations. Result We introduce ‘CyAnno’, a novel semi-automated approach for deconvoluting the unlabeled cytometry dataset based on a machine learning framework utilizing manually gated training data that allows the integrative modeling of ‘gated’ cell types and the ‘ungated’ cells. By applying this framework on several CyTOF datasets, we demonstrated that including the ‘ungated’ cells can lead to a significant increase in the precision of the ‘gated’ cell types prediction. CyAnno can be used to identify even a single cell type, including rare cells, with higher efficacy than current state-of-the-art semi-automated approaches. Availability The CyAnno is available as a python script with a user-manual and sample dataset at https://github.com/abbioinfo/CyAnno Supplementary information Supplementary data are available at Bioinformatics online.
Article
Short summary: Peanut-specific CD8+ T cells in nonallergic individuals are not deleted, but have an expansion block that can be released by impairing regulatory T cell associated signaling pathways.