ArticlePDF Available

Alterations in B- and circulating T-follicular helper cell subsets in immune thrombotic thrombocytopenic purpura

Authors:

Abstract and Figures

T follicular helper (Tfh) cells regulate development of antigen-specific B-cell immunity. We prospectively investigated B-cell and cTfh subsets in 45 immune TTP patients at presentation and longitudinally after rituximab (RTX). B-cell phenotype was altered at acute iTTP presentation with decreased transitional cells and postgerminal centre (post-GC) memory B cells and increased plasmablasts compared to healthy controls. A higher percentage of plasmablasts was associated with higher anti-ADAMTS13 IgG and lower ADAMTS13 antigen levels. In asymptomatic patients with ADAMTS13 relapse, there were increased naïve B cells and a global decrease in memory subsets, with a trend to increased plasmablasts. Total circulating Tfh (CD4+CXCR5+) and PD1+ Tfh cells were decreased at iTTP presentation. CD80 expression was decreased on IgD+ memory cells and double negative memory cells in acute iTTP. Longitudinal analysis: at repopulation after B cell depletion in de novo iTTP, post-GC and double negative memory B cells were reduced compared to pre-RTX. RTX did not cause alteration in cTfh frequency. The subsequent kinetics of naïve, transitional, memory B cells and plasmablasts did not differ significantly between patients who went on to relapse vs those who remained in remission. In summary, acute iTTP is characterised by dysregulation of B- and cTfh-cell homeostasis with depletion of post-GC memory cells and cTfh cells and increased plasmablasts. Changes in CD80 expression on B cells further suggest altered interactions with T cells.
Content may be subject to copyright.
American Society of Hematology
2021 L Street NW, Suite 900,
Washington, DC 20036
Phone: 202-776-0544 | Fax 202-776-0545
bloodadvances@hematology.org
Alterations in B- and circulating T-follicular helper cell subsets in immune
thrombotic thrombocytopenic purpura
Tracking no: ADV-2022-007025R1
Jin-Sup Shin (University College London Hospitals, United Kingdom) Maryam Subhan (University
College London, United Kingdom) Geraldine Cambridge (UCL, United Kingdom) Yanping Guo (Cancer
Institute, UCL, United Kingdom) Rens de Groot (University College London, United Kingdom) Marie
Scully (5National Institute for Health Research Cardiometabolic Programme, UCLH/UCL Cardiovascular
BRC, London, UK, United Kingdom) Mari Thomas (5National Institute for Health Research
Cardiometabolic Programme, UCLH/UCL Cardiovascular BRC, London, UK, United Kingdom)
Abstract:
T follicular helper (Tfh) cells regulate development of antigen-specific B-cell immunity. We
prospectively investigated B-cell and cTfh subsets in 45 immune TTP patients at presentation and
longitudinally after rituximab (RTX). B-cell phenotype was altered at acute iTTP presentation with
decreased transitional cells and postgerminal centre (post-GC) memory B cells and increased
plasmablasts compared to healthy controls. A higher percentage of plasmablasts was associated with
higher anti-ADAMTS13 IgG and lower ADAMTS13 antigen levels. In asymptomatic patients with ADAMTS13
relapse, there were increased naïve B cells and a global decrease in memory subsets, with a trend
to increased plasmablasts. Total circulating Tfh (CD4+CXCR5+) and PD1+ Tfh cells were decreased at
iTTP presentation. CD80 expression was decreased on IgD+ memory cells and double negative memory
cells in acute iTTP. Longitudinal analysis: at repopulation after B cell depletion in de novo iTTP,
post-GC and double negative memory B cells were reduced compared to pre-RTX. RTX did not cause
alteration in cTfh frequency. The subsequent kinetics of naïve, transitional, memory B cells and
plasmablasts did not differ significantly between patients who went on to relapse vs those who
remained in remission. In summary, acute iTTP is characterised by dysregulation of B- and cTfh-cell
homeostasis with depletion of post-GC memory cells and cTfh cells and increased plasmablasts.
Changes in CD80 expression on B cells further suggest altered interactions with T cells.
Conflict of interest: COI declared - see note
COI notes: M.S. has received speaker's fees and honoraria from Alexion, Sanofi, Novartis, and
Takeda and has received research funding from Takeda. M.T. has received speaker's fees and
honoraria from Sanofi and Bayer. The remaining authors declare no competing financial interests.
Preprint server: No;
Author contributions and disclosures: J.S. designed research, recruited patients, performed
laboratory testing, collected data, analysed data and wrote the manuscript. M.O.S. designed
research, recruited patients, performed laboratory testing, collected data, analysed data and wrote
the manuscript. G.C. designed research, analysed data and wrote the manuscript. Y.G. performed
laboratory testing, collected data and reviewed the manuscript. R.dG. designed research, analysed
data and wrote the manuscript. M.S. designed research, recruited patients, analysed data and wrote
the manuscript. M.T. designed research, recruited patients, analysed data and wrote the manuscript.
Non-author contributions and disclosures: No;
Agreement to Share Publication-Related Data and Data Sharing Statement: For original data, please
contact jin-sup.shin@nhs.net.
Clinical trial registration information (if any):
Downloaded from http://ashpublications.org/bloodadvances/article-pdf/doi/10.1182/bloodadvances.2022007025/1895989/bloodadvances.2022007025.pdf by guest on 11 May 2022
1
Alterations in B- and circulating T-follicular helper cell subsets in immune
thrombotic thrombocytopenic purpura
Authors
Jin-Sup Shin,1 Maryam Owais Subhan,2 Geraldine Cambridge,3 Yanping Guo,4 Rens de
Groot,2 Marie Scully,1,5 and Mari Thomas1,5
1Department of Haematology, University College London Hospital (UCLH), London, United Kingdom;
2Institute of Cardiovascular Science, University College London; 3Centre for Rheumatology Research,
University College London (UCL), London, United Kingdom; 4Cancer Research UK Flow Cytometry
Translational Technology Platform, Cancer Institute, UCL, London, United Kingdom; 5National
Institute for Health Research Cardiometabolic Programme, UCLH/UCL Cardiovascular BRC, London,
UK
Corresponding author contact information:
mari.thomas@nhs.net
Department of Haematology
University College London Hospitals
250 Euston Road, London
United Kingdom
NW1 2PG
Short title for the running head: B and cTfh cell changes in immune TTP
Word counts
- Text 3964
- Abstract 229
Figure/table count 6 figures 2 tables
Reference count 59
Downloaded from http://ashpublications.org/bloodadvances/article-pdf/doi/10.1182/bloodadvances.2022007025/1895989/bloodadvances.2022007025.pdf by guest on 11 May 2022
2
Key Points
Abnormal B-cell phenotype in acute iTTP with decreased transitional and
postgerminal centre memory cells and increased plasmablasts
Decreased total and PD1+ circulating T-follicular helper cells and changes in B-cell
CD80 expression suggest altered B-T–cell interactions
ABSTRACT
T follicular helper (Tfh) cells regulate development of antigen-specific B-cell immunity. We
prospectively investigated B-cell and cTfh subsets in 45 immune TTP patients at
presentation and longitudinally after rituximab (RTX). B-cell phenotype was altered at acute
iTTP presentation with decreased transitional cells and postgerminal centre (post-GC)
memory B cells and increased plasmablasts compared to healthy controls. A higher
percentage of plasmablasts was associated with higher anti-ADAMTS13 IgG and lower
ADAMTS13 antigen levels. In asymptomatic patients with ADAMTS13 relapse, there were
increased naïve B cells and a global decrease in memory subsets, with a trend to increased
plasmablasts. Total circulating Tfh (CD4+CXCR5+) and PD1+ Tfh cells were decreased at
iTTP presentation. CD80 expression was decreased on IgD+ memory cells and double
negative memory cells in acute iTTP. Longitudinal analysis: at repopulation after B cell
depletion in de novo iTTP, post-GC and double negative memory B cells were reduced
compared to pre-RTX. RTX did not cause alteration in cTfh frequency. The subsequent
kinetics of naïve, transitional, memory B cells and plasmablasts did not differ significantly
between patients who went on to relapse vs those who remained in remission. In summary,
acute iTTP is characterised by dysregulation of B- and cTfh-cell homeostasis with depletion
of post-GC memory cells and cTfh cells and increased plasmablasts. Changes in CD80
expression on B cells further suggest altered interactions with T cells.
Downloaded from http://ashpublications.org/bloodadvances/article-pdf/doi/10.1182/bloodadvances.2022007025/1895989/bloodadvances.2022007025.pdf by guest on 11 May 2022
3
INTRODUCTION
Immune thrombotic thrombocytopenic purpura (iTTP) is a life-threatening thrombotic
microangiopathy mediated by an immunoglobulin G (IgG) antibody against the
metalloprotease ADAMTS13 that enhances its clearance or inhibits its VWF processing
activity.1 In iTTP, there is an incompletely understood loss of tolerance resulting in a shift
from immune homeostasis to autoimmunity. This involves dendritic cells which acquire
antigens derived from ADAMTS13 that activate cross-reactive naïve CD4+ T cells which
differentiate into autoreactive effector CD4+ T cells.2,3 Mature autoreactive B cells recirculate
into the germinal centre of secondary lymph nodes where they are stimulated by compatible
antigens in the presence of the autoreactive T helper cells and differentiate into
autoantibody-producing plasma cells or long-lived memory B cells.4
B cell depletion therapy with rituximab (RTX; a chimeric monoclonal antibody against the
pan-B cell marker, CD20) has been demonstrated to be effective in reducing relapse rates in
iTTP and prolonging disease-free survival in acute episodes, compared to PEX and steroid
alone.5-7 Giving rituximab pre-emptively in patients at high risk of a clinical relapse (based on
a fall in ADAMTS13 activity levels to<10-20%, i.e. ‘ADAMTS13 relapse’) reduces clinical
relapse rates compared to historical controls.8
A recent Genome Wide Association Study (GWAS) of iTTP confirmed associations with
SNPs at the HLA locus and identified a novel association on chromosome 3.9 The locus on
chromosome 3 contains five genes, one of which is the CD80 gene. CD80 is a co-stimulator
for T lymphocyte activation. After activation through the B-cell receptor (BCR) or IL4, B cells
express CD80, which interacts with CD28 on T cells to provide co-stimulation signals.10 The
HLA-DRB1*11 and DRB1*03 genes are known risk factors for iTTP, suggesting the MHC
class II protein variants they encode have optimal affinity for certain ADAMTS13 peptides
recognised by CD4+ T-cell receptors in iTTP patients.11,12 Two peptides derived from the
CUB-2 domain of ADAMTS13 are presented on HLA-DRB1*11 and HLA-DRB1*03
respectively and recognised by iTTP patient-derived CD4+ T cells.13,14
CD4+ T cells are pivotal in development of iTTP as CD4+ T cell help is required in the
production and affinity maturation of ADAMTS13-directed antibodies.3 Within the CD4+T cell
population, the T follicular helper (Tfh) subset are vital for supporting antibody-mediated
immune responses by providing co-stimulation signals through CD40L and IL-21 production,
which promote the growth, differentiation and class-switching of antigen-activated naïve B
cells.15-17 Tfh cells constitutively express the chemokine receptor CXCR5 which facilitates
Downloaded from http://ashpublications.org/bloodadvances/article-pdf/doi/10.1182/bloodadvances.2022007025/1895989/bloodadvances.2022007025.pdf by guest on 11 May 2022
4
migration into germinal centres (GC).18 Tfh cells express co-stimulatory molecules such as
inducible co-stimulatory molecule (ICOS) and immune-regulatory molecules such as
programmed death-1 (PD-1) as their transcription factors, which can be used to further
define Tfh cells.19 More specifically, PD-1+ICOS- Tfh cells seem to represent quiescent and
PD-1+ICOS+ Tfh cells recently activated memory Tfh cells20,21
Circulating Tfh (cTfh) cells also express the GC homing receptor CCR7.22 Expansion of cTfh
cells has been associated with the development of several autoimmune diseases.23-29
The aim of this study was to investigate the B cell and cTfh subset distribution in iTTP
patients, both at presentation and longitudinally after anti-CD20 therapy in relation to clinical
and laboratory parameters and B cell kinetics. The role of cTfh has not previously been
investigated in iTTP. The temporal relationships between B and T cell subpopulation at
critical stages throughout the course of disease will improve the understanding of the
pathogenesis of iTTP, potentially provide biomarkers to predict relapse and may identify new
avenues for therapeutic intervention.
METHODS
Patients and controls:
We prospectively enrolled iTTP patients from September 2018 - June 2021 through the
United Kingdom TTP Registry (database and Biobank of UK TTP - Multicentre Research
Ethics Committee [MREC]:08/H0810/54 and MREC:08/H0716/72). The study was conducted
according to the Declaration of Helsinki. Blood samples were obtained from age and sex-
matched healthy controls (HC), with no history of autoimmune disease or
immunosuppressant medication. Definitions of iTTP diagnosis, remission and relapse were
based on previous studies.30-32
Patients were divided into two groups: de novo acute iTTP episodes and asymptomatic
ADAMTS13 relapses (treated with pre-emptive RTX). Acute presentations of iTTP were
treated with PEX, corticosteroids and RTX (4-8 doses 375 mg/m2 to normalize ADAMTS13
activity). Time after RTX was defined as time since first RTX infusion. Caplacizumab was
given to 18/22 of patients.
Patients with previously diagnosed iTTP who were in clinical remission (normal platelet
count) but developed a new severe ADAMTS13 deficiency were referred to as ADAMTS13
relapse episodes.32 These patients received pre-emptive RTX (four doses of
Downloaded from http://ashpublications.org/bloodadvances/article-pdf/doi/10.1182/bloodadvances.2022007025/1895989/bloodadvances.2022007025.pdf by guest on 11 May 2022
5
200/500/375mg/m2 one week apart). Dosing varied at clinician discretion or as part of a
separate randomised control trial (Elective Rituximab in TTP trial;[REC]17/LO/1055).
Blood samples and PBMC isolation:
Heparinised blood samples collected from patients prior to initiation of RTX or other
immunosuppression and HC were analysed within 24 hours or frozen at -80˚C and stored for
later analysis. Samples were also collected one month and three months post RTX and then
three monthly thereafter until clinical or ADAMTS13 relapse, or end of study (whichever
came first). Peripheral blood mononuclear cells (PBMC) were isolated by diluting blood (1:1)
with phosphate-buffered saline(PBS) 1X and layered over Ficoll-Paque Premium 1084R
(Sigma-Aldrich).33
Flow cytometry
PBMC (approximately 1 ×106/sample) were incubated with conjugated antibodies for 20
minutes in the dark at room temperature. Cells were washed twice and resuspended in PBS
and samples were analysed on a Cytoflex S cytometer (Beckman Coulter) (Supplementary
Materials). Each sample was divided into two antibody panels: for B cell subsets and Tfh
cells (Supplementary Materials: Table 1a & 1b). Viability was determined using
LIVE/DEAD™ Fixable Near-IR Dead Cell Stain Kit (InvitrogenTM). Analysis was performed on
FlowJo software version 10 (FlowJo LLC).
B cell immunophenotyping and gating strategy
We used the mature B cell ‘Bm1–Bm5’ classification to identify B cell subsets based on co-
expression of IgD and CD38. B cell subsets include transitional B cells, naive B cells,
memory populations including IgD+ memory cells, post germinal centre (post-GC) cells,
double negative memory cells and plasmablasts.34,35 Antibodies used for the B cell panel
were CD19 PECy7, CD27 APC, IgD BV421, CD38 PE. Following on from our GWAS finding
that the CD80/POGLUT1 locus is associated with iTTP,9 we also analysed the level of CD80
expression (median fluorescence intensity or MFI) on B-cells subsets in iTTP.36 Gating
strategies are shown in Supplemental Figure 1.
Circulating Tfh immunophenotyping and gating strategy
Downloaded from http://ashpublications.org/bloodadvances/article-pdf/doi/10.1182/bloodadvances.2022007025/1895989/bloodadvances.2022007025.pdf by guest on 11 May 2022
6
The total cTfh population was defined by CD3+ T cells that expressed CD4+CXCR5+. The
different subsets within the total cTfh population were defined by expression of activation
markers: PD1+ cTfh, ICOS+ cTfh and PD1+ICOS+ cTfh. In the cTfh panel, PBMC were
stained with CD3 PECy7, CD4 PerCP, CXCR5 FITC, PD1 APC and ICOS BV421
antibodies. The gating strategy and representative plots are shown in Supplemental Figure
2.
ADAMTS13 assays
ADAMTS13 activity was analysed by fluorescence resonance energy transfer (FRETS
(normal range: 60%-123%).37 ADAMTS13 antigen levels were quantified using in-house
developed enzyme-linked immunosorbent assay, previously described (normal range: 74%
to 134%).38 ADAMTS-13 activity and antigen levels were expressed as a percentage relative
to pooled normal plasma (PNP). Anti-ADAMTS13 IgG levels were measured with in-house
ELISA (normal range <6.1%) with concentration of anti-ADAMTS13 IgG calculated as a
percentage relative to an index plasma from a patient with a high auto-antibody titre
(assigned a value of 100%).7
Statistical Analysis
Mann-Whitney U and Wilcoxon tests were used for paired and unpaired continuous
variables, respectively. The X2 and Fisher’s exact tests were performed for categorical
variables. Spearman correlation test was used to measure the possible relationship between
two variables of interest. Statistical analysis was performed using GraphPad Prism 8
(GraphPad Software Inc., La Jolla, CA, USA).
Data Sharing statement
For original data, please contact jin-sup.shin@nhs.net.
RESULTS
Patient demographics and ADAMTS13 biomarkers
Demographics of iTTP patients and HC and ADAMTS13 assay results are shown in
Supplemental Table 2a). There were 45 unique patients with iTTP involving 46 episodes: 22
de novo acute episodes and 24 asymptomatic ADAMTS13 relapses treated with pre-emptive
rituximab.
Downloaded from http://ashpublications.org/bloodadvances/article-pdf/doi/10.1182/bloodadvances.2022007025/1895989/bloodadvances.2022007025.pdf by guest on 11 May 2022
7
In acute iTTP episodes, 82% (18/22) cases had cardiac involvement and 50% (11/22)
neurological. 14% (3/22) of acute episodes required organ support on the intensive care unit.
All acute episodes were treated with PEX, corticosteroids and RTX. 82% (18/22) patients
received anti-VWF nanobody, Caplacizumab. Additional immunosuppression included
mycophenolate mofetil (27%, 6/22) and bortezomib (9%, 2/22). All but one of ADAMTS13
relapse episodes in this study had previously received rituximab treatment, either during an
acute presentation or previous ADAMTS13 relapse episode. In this cohort, the median
number of previous TTP episodes was 3 (range 1-8 episodes), with a median duration since
the most recent TTP episode being 21 months (range 13-191 months), (Supplemental Table
2b).
Post germinal centre memory B cells are decreased and plasmablasts increased at
iTTP presentation. (Figure 1)
The total number of CD19+ B cells at presentation of acute iTTP or preceding pre-emptive
rituximab for ADAMTS13 relapse episodes was not significantly different to HC. At
presentation in acute iTTP episodes, post-GC memory cells were decreased compared to
healthy controls (HC 13.7% (range, 3.1-28.8%) vs acute iTTP 7.9% (range, 1.8-27.8%),
p=0.008), whereas plasmablasts were increased (HC 0.7% (range, 0.2-2.5%) vs acute iTTP
1.6% (range, 0.2-15.4%), p=0.007), (Figure 1).
B cell repopulation after RTX recapitulates ontogeny beginning with naïve B cell exit from the
bone marrow followed by gradual maturation of memory subsets over time. In ADAMTS13
relapse cases, there was an increased proportion of naïve B cells compared to healthy
controls and a trend to increased transitional cells and plasmablasts (Figure 1). There was a
marked reduction in all memory subsets.
Altered circulating T follicular helper cell subsets at iTTP presentation: decreased
total cTfh and PD1+ cTfh (Figure 2)
The relative proportions of CD4+ cTfh subsets in acute TTP and ADAMTS13 relapse were
determined and compared with HC (Figure 2). The frequency of total cTfh and PD1+ cTfh
were significantly reduced in acute iTTP compared to HC. This may be suggestive of
migration of circulating Tfh cells into germinal centers. There were no significant differences
seen in ICOS+ or PD1+ICOS+ cTfh cells. In ADAMTS13 relapses, no differences were seen
in total, PD1+ or PD1+ICOS+ cTfh cells. However, ICOS+ cTfh cells were increased in
patients compared to HC.
Downloaded from http://ashpublications.org/bloodadvances/article-pdf/doi/10.1182/bloodadvances.2022007025/1895989/bloodadvances.2022007025.pdf by guest on 11 May 2022
8
Relationship between ADAMTS13 parameters, circulating Tfh cells and B cell subsets
at iTTP presentation
In the acute iTTP group, median time to achieve a normal platelet count from presentation
was 4 days (range, 2-11) and median time to normalization of ADAMTS13 activity was 33
days (range 3-382; interquartile range, 22-124). At presentation in acute iTTP episodes, a
higher percentage of plasmablasts appears to be associated with lower antigen levels (r= -
0.41, p=0.055) (Figure 3). Indeed, a plasmablast level of >3% was associated with IgG
antibody level of >50% (Table 1). This may be due to increased production of anti-
ADAMTS13 IgG antibody from a larger number of plasmablasts, which in turn results in
increased ADAMTS13 clearance. In both acute iTTP cases and ADAMTS13 relapses, there
was no correlation between total cTfh cells and any of the B cell subsets (data not shown).
Expression of CD80 on B cell subsets at acute presentation and post RTX
In acute iTTP episodes, CD80 MFI was decreased in IgD+ memory cells and double
negative memory cells compared to HC (Figure 4). However, in ADAMTS13 relapse cases,
CD80 MFI was significantly increased in post-GC and double negative memory cells
compared to HC). A possible explanation for this difference between acute iTTP and
asymptomatic ADAMTS13 relapse may be that in asymptomatic falls in ADAMTS13 activity
prior to an acute clinical relapse, it is possible to detect activated memory B cell populations
due to less ‘background noise’ or before they marginate.
A summary of the B and T follicular helper cell immunophenotyping results at acute iTTP
presentation and ADAMTS13 relapse is shown in Table 2.
Longitudinal analysis: effect of RTX
Flow cytometric analysis of B cell subsets and cTfh cells was performed for patients pre-RTX
and longitudinally 1 month, 3 months, 6, months, 9 months, 12 months etc until end of
study/relapse. After RTX treatment, all 46 episodes achieved B cell depletion (defined by a
laboratory CD19+ count <0.005 ×109/L), except 1 patient in the ADAMTS13 relapse cohort
(Supplemental Figure 3 (a) – (f) for acute iTTP cases; data not shown for ADAMTS13
relapse episodes). In contrast, RTX did not cause any significant alterations in cTfh numbers
(Supplemental Figure 3 (g) – (k) for acute iTTP cases; data not shown for ADAMTS13
relapse episodes). B cell return occurred at median 10 months (range 6-14) months) in the
Downloaded from http://ashpublications.org/bloodadvances/article-pdf/doi/10.1182/bloodadvances.2022007025/1895989/bloodadvances.2022007025.pdf by guest on 11 May 2022
9
acute iTTP group and 8 months (range 0.25-15 months) in ADAMTS13 relapse group. The
timepoint of B cell return was not associated with ADAMTS13 or clinical relapse.
Comparison of B cell subsets frequencies prior to RTX treatment and at B cell return
We then compared two specific timepoints: pre-RTX and B cell return. Repopulation
following B cell depletion in the acute iTTP group demonstrated relatively higher proportion
of plasmablasts but a reduction of in post-GC and double negative memory B cells (Figure
5a). In the ADAMTS13 relapse group where all except 1 patient had received historical RTX,
plasmablasts were increased in frequency at B cell return compared to pre-treatment levels
but the distribution of B cell subsets was otherwise similar before and after re-treatment
(Figure 5b).
Longitudinal follow up: relapses vs sustained remissions
We prospectively investigated changes in B-cell subsets after RTX in 20 iTTP patients (16
from our initial cohort and 4 additional patients) longitudinally until a subsequent clinical or
ADAMTS13 relapse, and compared to patients who remained in remission over an
equivalent period.
There were 10 patients in the relapse cohort (3 followed after an acute episode, 7 after
elective rituximab) and 10 in the remission cohort (4 acute and 6 elective episodes at t=0).
Median age was 51y (range 38-81) and 45.5y (21-68) respectively. Median follow up was15
months (range 6-24). One patient in the relapse group did not achieve B-cell depletion
(CD19+ count 0.005x10^9/L). Time to B-cell return was similar: 8 months (4-13 months) in
relapses vs 8months in remission (0.25-15.5 months). All subsequent relapses were
ADAMTS13 relapses and occurred at a median of 14 months (9-25 months).
Longitudinal analysis showed that in both groups B-cell return after rituximab develops along
normal B-cell ontogeny. The kinetics of naïve, transitional, memory B-cells and plasmablasts
did not differ significantly between patients who subsequently went on to relapse vs those
who remained in sustained remission. (Figure 6) No alterations in B-cell subsets were
identified prior to a relapse.
Downloaded from http://ashpublications.org/bloodadvances/article-pdf/doi/10.1182/bloodadvances.2022007025/1895989/bloodadvances.2022007025.pdf by guest on 11 May 2022
10
DISCUSSION
Interactions between T and B cells which occur within germinal centres (GC) of secondary
lymphoid organs (SLO) are critical for the development of humoral immune responses. Tfh
cells, a distinct subset of T helper cells, have been recognised in recent years as a crucial
regulator of GC formation, B cell development and longterm humoral memory generation,
and have a significant role in the pathogenesis of autoimmune diseases.17 The mechanism
involved in the loss of tolerance and subsequent development of anti-ADAMTS13 antibodies
in iTTP patients is still largely unknown and we currently lack the ability to predict relapse
accurately. The observed association between the MHC class II allele HLA DRB1*11 and
development of iTTP implies a role for helper CD4+ T cells in the initiation of autoimmune
reactivity against ADAMTS13.11,12,39
This is the first prospective study to perform a comprehensive analysis of B cell subsets and
cTfh cell changes in iTTP before and after immunosuppressive therapy. B cell phenotype is
altered at acute iTTP presentation with decreased post-GC memory B cells and an increase
in plasmablasts in iTTP compared to healthy controls. Potential mechanisms for the reduced
proportion of memory B cells among circulating B cells at presentation of iTTP include 1)
some B cells dying in the early memory B cell stages and never becoming memory B cells,
2) hyperactivation of T and B cells in iTTP resulting in an elevated differentiation of memory
B cells into antibody-producing plasma cells or 3) migration of memory B cells into lymphoid
tissue.40-42 Similar reductions in the memory B cell compartments have been described in
sarcoidosis, systemic sclerosis and Sjogren’s syndrome.40,43-48
Comparing immunophenotype results between different studies is complex due to the variety
of staining and gating protocols used, and clinical and demographic characteristics of the
patient cohort, but some general conclusions can be made regarding iTTP in comparison to
other autoimmune diseases. We found an increased ratio of naïve to switched memory B
cells and higher plasmablasts in the acute patients. In contrast, in patients with active SLE
there is an apparent expansion of memory vs naïve B cell subsets correlating in part with
disease activity, but this is due to a naïve B cell lymphopaenia.49 Expansion of plasmablasts
also occurs in SLE but is relatively higher than we found in iTTP. In Sjogrens syndrome,
transitional and naïve B cells seem expanded compared to memory subsets, thus closer to
the iTTP result, but in RA patients naïve and memory populations are generally similar to
healthy age matched controls.50 More recently, attention has focussed on the double
negative (DN) (IgD-CD27-) B cell subset. In systemic sclerosis, SLE and RA, the relative
importance of this subset, especially in relation to therapy resistance, has been recognised.
Downloaded from http://ashpublications.org/bloodadvances/article-pdf/doi/10.1182/bloodadvances.2022007025/1895989/bloodadvances.2022007025.pdf by guest on 11 May 2022
11
The DN B cell population contains both activated and antibody secreting post germinal
centre B cells and very early switched B cells (with fewer mutated Ig transcripts).51,52 This
population was not different from HC in the iTTP patients in relation to relapse, but additional
studies would be needed to assess functional properties of B cells within this population in
iTTP.
The relative contribution of different naive and antigen-experienced B cell subsets to
pathogenesis and response to therapies will depend on the position and circumstances
underlying specific breaches of tolerance and thus ultimately depend on the specificity of the
autoreactive B cell receptor(s). The different patterns we have described in iTTP compared
to other systemic autoimmune diseases indicate a lack of apparent expansion of memory
populations in the peripheral blood and thus the probability of rapid sequestration or
expansion of autoantibody producing clones within tissues or lymphoid organs.
At acute iTTP presentation, we also demonstrate a novel association between higher
plasmablast frequency and higher ADAMTS13 IgG antibody level and a trend towards
reduced ADAMTS13 antigen levels. This suggests a potential underlying mechanism for
iTTP development, where rapid differentiation to auto-antibody producing plasmablasts
leads to increased production anti-ADAMTS13 IgG which in turn results in increased
ADAMTS13 clearance.
In asymptomatic patients with ADAMTS13 relapse, alterations in B cell subsets prior to pre-
emptive RTX therapy were pronounced with significantly increased naïve B cell population,
global decrease in all memory subsets and trend towards increased plasmablasts. These
subset alterations most likely represent changes related to historic RTX therapy with long-
term suppressive effects of RTX on memory cell subsets. This also explains the largely
similar distribution of B cell subsets before and after pre-emptive RTX treatment. Following B
cell depletion therapy, B cells repopulate primarily from bone marrow-derived naive B cells,
with delayed regeneration of memory B cells.53,54 Poor memory B cell reconstitution may
reflect long-term effects of RTX on B cells, described both after organ transplantation and in
other autoimmune diseases.55-57
Total cTfh (CD4+CXCR5+) and PD1+ Tfh cells were decreased in acute iTTP patients at
presentation compared to HC. A similar decrease in cTfh has been observed in sarcoidosis
with infiltration of Tfh into skin lesions, suggesting that cTfh are recruited into affected sites.47
In acute iTTP, a potential explanation for the concomitant decrease in post-GC memory cells
and cTfh cells may be that they are localising in germinal centres within lymphoid tissue. In
Downloaded from http://ashpublications.org/bloodadvances/article-pdf/doi/10.1182/bloodadvances.2022007025/1895989/bloodadvances.2022007025.pdf by guest on 11 May 2022
12
contrast, in asymptomatic patients with ADAMTS13 relapse, there were no significant
differences in total cTfh cells and PD1+ cTfh. This may be because this is a different stage
of the disease process or possibly that the effect of previous Rituximab alters the interplay of
B and cTfh cells. Tfh helper T cells utilise CXCR5 positivity to access germinal centres (GC).
In GC, they produce IL21 which plays a key role in class switch and affinity maturation
leading the generation of memory B cells and plasma cells from antigen-activated naïve B
cells. Other peripheral blood helper T cells (CXCR5-; CXCR2 and PD1+) can also produce
IL21 and have been identified as a source of extrafollicular T cell help to B cells in patients
with other autoimmune diseases.58 Although there are no frank inflammatory sites or
lymphoid clusters in iTTP, it is possible that aberrant interactions between ADAMTS13-
specific memory B cells and other T helper cells may occur in extrafollicular sites in spleen
for example.
We also compared B and cTfh frequencies at 2 different timepoints following RTX infusion:
prior to RTX and at B cell return. B cell depletion was achieved in all patients by 1 month.
Subsequent B cell reconstitution occurred at 10 months in the acute iTTP cohort and 8
months in the asymptomatic ADAMTS13 relapse cohort. At B cell return after a de novo
acute iTTP episode, plasmablast levels were significantly raised with a trend towards
increased transitional and naïve cells. Importantly, no patients relapsed at the time of B cell
return. The time interval between B cell return and relapse suggests that additional
differentiation and selection of specific autoreactive B cell clones from naive populations may
play a key role, although expansion of ADAMTS13 specific memory B cell populations in
lymphoid tissues to critical levels cannot be ruled out. RTX did not cause any significant
alterations in cTfh frequency.
Our data also reveals altered expression of CD80 on B cells in iTTP, with decreased
expression in IgD+ memory cells and double negative memory cells in acute iTTP episodes
and increased expression in post-GC memory and double negative memory cells in
ADAMTS13 relapses, prior to RTX therapy. CD80 and CD86 are expressed on antigen
presenting cells such as dendritic cells and activated B cells, and have the capacity to
stimulate or inhibit T cell responses through their receptors CD28 and cytotoxic T
lymphocyte-associated antigen 4 (CTLA-4) respectively.10 Dendritic cells exposed to
ADAMTS13 have previously been shown to present ADAMTS13 peptides, with preferential
HLA-DRB1* 11–dependent presentation of CUB2-derived peptides, which the authors
hypothesised may contribute to the onset of acquired TTP by stimulating low-affinity self-
reactive CD4+ T cells.2,3
Downloaded from http://ashpublications.org/bloodadvances/article-pdf/doi/10.1182/bloodadvances.2022007025/1895989/bloodadvances.2022007025.pdf by guest on 11 May 2022
13
Manipulation of the CD80/CD86 pathway has shown efficacy in the clinical setting, with
Abatacept (a CTLA-4 immunoglobulin which targets autoimmune B cells by reducing
CD80/CD86 expression) approved for the treatment of rheumatoid arthritis.59 Of interest, the
CD80 locus was identified in the GWAS for iTTP and contained a SNP in the 3’ UTR that
was in high linkage with the lead SNP.9 Further investigation into the link between
possession of this SNP with translation and expression of CD80 on different cell types is
ongoing. Additional studies are required to understand how these differences in expression
could translate into functional properties of CD80/86 in iTTP patients.
In the longitudinal analysis, no alterations in B-cell subsets were identified prior to a relapse,
suggesting temporal relationships between B sub-populations cannot be used as a
biomarker to better predict relapses.
Taken together, our findings give a novel insight into the role of B and cTfh cells in the
development of iTTP. We propose that de novo acute iTTP is characterised by dysregulation
of B and cTfh cell homeostasis with decreased circulating subsets of GC memory cells and
cTfh cells and an increased frequency of plasmablasts in the circulation, perhaps reflecting
the increase in cognate interaction between antigen-specific T and B cell populations in
secondary lymphoid tissue. Changes in the frequency of CD80 on B cells suggests altered
interactions with T cells. The finding of alteration in CD80 expression further supports the
recent novel association between iTTP and five alleles within a haploblock on chromosome
3 – one of which encodes CD80.9 Although our studies did not address the questions of
antigen selectivity and exquisite specificity of the autoimmune response to ADAMTS13 in
iTTP, the novel findings will direct further functional analysis, and provide potentially
interesting targets for further research and therapeutics in iTTP.
AUTHORSHIP CONTRIBUTIONS
Contributions: J.S. designed research, recruited patients, performed laboratory testing,
collected data, analysed data and wrote the manuscript. M.O.S. designed research,
recruited patients, performed laboratory testing, collected data, analysed data and wrote the
manuscript. G.C. designed research, analysed data and wrote the manuscript. Y.G.
performed laboratory testing, collected data and reviewed the manuscript. R.dG. designed
research, analysed data and wrote the manuscript. M.S. designed research, recruited
Downloaded from http://ashpublications.org/bloodadvances/article-pdf/doi/10.1182/bloodadvances.2022007025/1895989/bloodadvances.2022007025.pdf by guest on 11 May 2022
14
patients, analysed data and wrote the manuscript. M.T. designed research, recruited
patients, analysed data and wrote the manuscript.
DISCLOSURE OF CONFLICTS OF INTEREST
M.S. has received speaker’s fees and honoraria from Alexion, Sanofi, Novartis, and Takeda
and has received research funding from Takeda. M.T. has received speaker’s fees
and honoraria from Sanofi and Bayer. The remaining authors declare no competing financial
interests.
REFERENCES
1. Kremer Hovinga JA, Coppo P, Lämmle B, Moake JL, Miyata T, Vanhoorelbeke K. Thrombotic
thrombocytopenic purpura. Nature reviews Disease primers. 2017;3:17020-17020.
2. Sorvillo N, van Haren SD, Kaijen PH, et al. Preferential HLA-DRB1*11-dependent presentation
of CUB2-derived peptides by ADAMTS13-pulsed dendritic cells. Blood. 2013;121(17):3502-3510.
3. Verbij FC, Turksma AW, de Heij F, et al. CD4+ T cells from patients with acquired thrombotic
thrombocytopenic purpura recognize CUB2 domain-derived peptides. Blood. 2016;127(12):1606-
1609.
4. Carsetti R, Rosado MM, Wardmann H. Peripheral development of B cells in mouse and man.
Immunol Rev. 2004;197:179-191.
5. Westwood JP, Webster H, McGuckin S, McDonald V, Machin SJ, Scully M. Rituximab for
thrombotic thrombocytopenic purpura: benefit of early administration during acute episodes and
use of prophylaxis to prevent relapse. J Thromb Haemost. 2013;11(3):481-490.
6. Froissart A, Buffet M, Veyradier A, et al. Efficacy and safety of first-line rituximab in severe,
acquired thrombotic thrombocytopenic purpura with a suboptimal response to plasma exchange.
Experience of the French Thrombotic Microangiopathies Reference Center. Crit Care Med.
2012;40(1):104-111.
7. Scully M, Cohen H, Cavenagh J, et al. Remission in acute refractory and relapsing thrombotic
thrombocytopenic purpura following rituximab is associated with a reduction in IgG antibodies to
ADAMTS-13. Br J Haematol. 2007;136(3):451-461.
8. Westwood JP, Thomas M, Alwan F, et al. Rituximab prophylaxis to prevent thrombotic
thrombocytopenic purpura relapse: outcome and evaluation of dosing regimens. Blood Adv.
2017;1(15):1159-1166.
9. Stubbs M, Coppo P, Cheshire C, et al. Identification of a novel genetic locus associated with
immune mediated thrombotic thrombocytopenic purpura. Haematologica. 2021.
10. Sansom DM, Manzotti CN, Zheng Y. What's the difference between CD80 and CD86? Trends
Immunol. 2003;24(6):314-319.
11. SCULLY M, BROWN J, PATEL R, MCDONALD V, BROWN CJ, MACHIN S. Human leukocyte
antigen association in idiopathic thrombotic thrombocytopenic purpura: evidence for an
immunogenetic link. Journal of Thrombosis and Haemostasis. 2010;8(2):257-262.
12. COPPO P, BUSSON M, VEYRADIER A, et al. HLA-DRB1*11: a strong risk factor for acquired
severe ADAMTS13 deficiency-related idiopathic thrombotic thrombocytopenic purpura in
Caucasians. Journal of Thrombosis and Haemostasis. 2010;8(4):856-859.
Downloaded from http://ashpublications.org/bloodadvances/article-pdf/doi/10.1182/bloodadvances.2022007025/1895989/bloodadvances.2022007025.pdf by guest on 11 May 2022
15
13. Laurent G, Sandrine D, Ivan P, et al. The ADAMTS131239–1253 peptide is a dominant HLA-
DR1-restricted CD4+ T-cell epitope. Haematologica. 2017;102(11):1833-1841.
14. Johana H, Silvia DA, Nuno AGG, et al. Dissecting the pathophysiology of immune thrombotic
thrombocytopenic purpura: interplay between genes and environmental triggers. Haematologica.
2018;103(7):1099-1109.
15. Breitfeld D, Ohl L, Kremmer E, et al. Follicular B helper T cells express CXC chemokine
receptor 5, localize to B cell follicles, and support immunoglobulin production. J Exp Med.
2000;192(11):1545-1552.
16. Kim CH, Rott LS, Clark-Lewis I, Campbell DJ, Wu L, Butcher EC. Subspecialization of CXCR5+ T
cells: B helper activity is focused in a germinal center-localized subset of CXCR5+ T cells. J Exp Med.
2001;193(12):1373-1381.
17. Crotty S. Follicular helper CD4 T cells (TFH). Annu Rev Immunol. 2011;29:621-663.
18. Rasheed AU, Rahn HP, Sallusto F, Lipp M, Müller G. Follicular B helper T cell activity is
confined to CXCR5(hi)ICOS(hi) CD4 T cells and is independent of CD57 expression. Eur J Immunol.
2006;36(7):1892-1903.
19. Webb LMC, Linterman MA. Signals that drive T follicular helper cell formation. Immunology.
2017;152(2):185-194.
20. He J, Tsai LM, Leong YA, et al. Circulating precursor CCR7(lo)PD-1(hi) CXCR5(+) CD4(+) T cells
indicate Tfh cell activity and promote antibody responses upon antigen reexposure. Immunity.
2013;39(4):770-781.
21. Locci M, Havenar-Daughton C, Landais E, et al. Human circulating PD-1+CXCR3-CXCR5+
memory Tfh cells are highly functional and correlate with broadly neutralizing HIV antibody
responses. Immunity. 2013;39(4):758-769.
22. Morita R, Schmitt N, Bentebibel SE, et al. Human blood CXCR5(+)CD4(+) T cells are
counterparts of T follicular cells and contain specific subsets that differentially support antibody
secretion. Immunity. 2011;34(1):108-121.
23. Ma J, Zhu C, Ma B, et al. Increased frequency of circulating follicular helper T cells in patients
with rheumatoid arthritis. Clin Dev Immunol. 2012;2012:827480.
24. Zhu C, Ma J, Liu Y, et al. Increased frequency of follicular helper T cells in patients with
autoimmune thyroid disease. J Clin Endocrinol Metab. 2012;97(3):943-950.
25. Simpson N, Gatenby PA, Wilson A, et al. Expansion of circulating T cells resembling follicular
helper T cells is a fixed phenotype that identifies a subset of severe systemic lupus erythematosus.
Arthritis Rheum. 2010;62(1):234-244.
26. Zhang X, Liu S, Chang T, et al. Intrathymic Tfh/B Cells Interaction Leads to Ectopic GCs
Formation and Anti-AChR Antibody Production: Central Role in Triggering MG Occurrence. Mol
Neurobiol. 2016;53(1):120-131.
27. Gensous N, Charrier M, Duluc D, et al. T Follicular Helper Cells in Autoimmune Disorders.
Front Immunol. 2018;9:1637.
28. Park HJ, Kim DH, Lim SH, et al. Insights into the role of follicular helper T cells in
autoimmunity. Immune Netw. 2014;14(1):21-29.
29. Ueno H. T follicular helper cells in human autoimmunity. Curr Opin Immunol. 2016;43:24-31.
30. Scully M, Cataland S, Coppo P, et al. Consensus on the standardization of terminology in
thrombotic thrombocytopenic purpura and related thrombotic microangiopathies. J Thromb
Haemost. 2017;15(2):312-322.
31. Zheng XL, Vesely SK, Cataland SR, et al. ISTH guidelines for the diagnosis of thrombotic
thrombocytopenic purpura. Journal of Thrombosis and Haemostasis. 2020;18(10):2486-2495.
32. Cuker A, Cataland SR, Coppo P, et al. Redefining outcomes in immune TTP: an international
working group consensus report. Blood. 2021;137(14):1855-1861.
33. Turner RJ, Geraghty NJ, Williams JG, et al. Comparison of peripheral blood mononuclear cell
isolation techniques and the impact of cryopreservation on human lymphocytes expressing CD39
and CD73. Purinergic Signal. 2020;16(3):389-401.
Downloaded from http://ashpublications.org/bloodadvances/article-pdf/doi/10.1182/bloodadvances.2022007025/1895989/bloodadvances.2022007025.pdf by guest on 11 May 2022
16
34. Bohnhorst J, Bjørgan MB, Thoen JE, Natvig JB, Thompson KM. Bm1-Bm5 classification of
peripheral blood B cells reveals circulating germinal center founder cells in healthy individuals and
disturbance in the B cell subpopulations in patients with primary Sjögren's syndrome. J Immunol.
2001;167(7):3610-3618.
35. Shi Y, Agematsu K, Ochs HD, Sugane K. Functional analysis of human memory B-cell
subpopulations: IgD+CD27+ B cells are crucial in secondary immune response by producing high
affinity IgM. Clin Immunol. 2003;108(2):128-137.
36. Agematsu K, Hokibara S, Nagumo H, Komiyama A. CD27: a memory B-cell marker. Immunol
Today. 2000;21(5):204-206.
37. Kokame K, Nobe Y, Kokubo Y, Okayama A, Miyata T. FRETS-VWF73, a first fluorogenic
substrate for ADAMTS13 assay. Br J Haematol. 2005;129(1):93-100.
38. Alwan F, Vendramin C, Vanhoorelbeke K, et al. Presenting ADAMTS13 antibody and antigen
levels predict prognosis in immune-mediated thrombotic thrombocytopenic purpura. Blood.
2017;130(4):466-471.
39. John ML, Hitzler W, Scharrer I. The role of human leukocyte antigens as predisposing and/or
protective factors in patients with idiopathic thrombotic thrombocytopenic purpura. Ann Hematol.
2012;91(4):507-510.
40. Bohnhorst JO, Thoen JE, Natvig JB, Thompson KM. Significantly depressed percentage of
CD27+ (memory) B cells among peripheral blood B cells in patients with primary Sjögren's syndrome.
Scand J Immunol. 2001;54(4):421-427.
41. Hansen A, Reiter K, Ziprian T, et al. Dysregulation of chemokine receptor expression and
function by B cells of patients with primary Sjögren's syndrome. Arthritis & Rheumatism.
2005;52(7):2109-2119.
42. Bohnhorst JØ, Bjørgan MB, Thoen JE, Jonsson R, Natvig JB, Thompson KM. Abnormal B Cell
Differentiation in Primary Sjögren's Syndrome Results in a Depressed Percentage of Circulating
Memory B Cells and Elevated Levels of Soluble CD27 That Correlate with Serum IgG Concentration.
Clinical Immunology. 2002;103(1):79-88.
43. Lee N-S, Barber L, Akula SM, Sigounas G, Kataria YP, Arce S. Disturbed Homeostasis and
Multiple Signaling Defects in the Peripheral Blood B-Cell Compartment of Patients with Severe
Chronic Sarcoidosis. Clinical and Vaccine Immunology. 2011;18(8):1306-1316.
44. Kudryavtsev I, Serebriakova M, Starshinova A, et al. Imbalance in B cell and T Follicular
Helper Cell Subsets in Pulmonary Sarcoidosis. Scientific Reports. 2020;10(1):1059.
45. Binard A, Le Pottier L, Devauchelle-Pensec V, Saraux A, Youinou P, Pers J-O. Is the blood B-
cell subset profile diagnostic for Sjögren syndrome? Annals of the Rheumatic Diseases.
2009;68(9):1447-1452.
46. Ibrahem HM. B cell dysregulation in primary Sjögren's syndrome: A review. Jpn Dent Sci Rev.
2019;55(1):139-144.
47. Ly NTM, Ueda-Hayakawa I, Nguyen CTH, Okamoto H. Exploring the imbalance of circulating
follicular helper CD4+ T cells in sarcoidosis patients. Journal of Dermatological Science.
2020;97(3):216-224.
48. Szabó K, Jámbor I, Szántó A, et al. The Imbalance of Circulating Follicular T Helper Cell
Subsets in Primary Sjögren’s Syndrome Associates With Serological Alterations and Abnormal B-Cell
Distribution. Frontiers in Immunology. 2021;12:789.
49. Odendahl M, Jacobi A, Hansen A, et al. Disturbed peripheral B lymphocyte homeostasis in
systemic lupus erythematosus. J Immunol. 2000;165(10):5970-5979.
50. Hansen A, Odendahl M, Reiter K, et al. Diminished peripheral blood memory B cells and
accumulation of memory B cells in the salivary glands of patients with Sjögren's syndrome. Arthritis
Rheum. 2002;46(8):2160-2171.
51. Wei C, Anolik J, Cappione A, et al. A new population of cells lacking expression of CD27
represents a notable component of the B cell memory compartment in systemic lupus
erythematosus. J Immunol. 2007;178(10):6624-6633.
Downloaded from http://ashpublications.org/bloodadvances/article-pdf/doi/10.1182/bloodadvances.2022007025/1895989/bloodadvances.2022007025.pdf by guest on 11 May 2022
17
52. Soto L, Ferrier A, Aravena O, et al. Systemic sclerosis patients present alterations in the
expression of molecules involved in B cell regulation. Frontiers in Immunology. 2015;6.
53. Leandro MJ, Cambridge G, Ehrenstein MR, Edwards JC. Reconstitution of peripheral blood B
cells after depletion with rituximab in patients with rheumatoid arthritis. Arthritis Rheum.
2006;54(2):613-620.
54. Cambridge G, Stohl W, Leandro MJ, Migone TS, Hilbert DM, Edwards JC. Circulating levels of
B lymphocyte stimulator in patients with rheumatoid arthritis following rituximab treatment:
relationships with B cell depletion, circulating antibodies, and clinical relapse. Arthritis Rheum.
2006;54(3):723-732.
55. Bemark M, Holmqvist J, Abrahamsson J, Mellgren K. Translational Mini-Review Series on B
cell subsets in disease. Reconstitution after haematopoietic stem cell transplantation - revelation of
B cell developmental pathways and lineage phenotypes. Clin Exp Immunol. 2012;167(1):15-25.
56. Roll P, Palanichamy A, Kneitz C, Dorner T, Tony HP. Regeneration of B cell subsets after
transient B cell depletion using anti-CD20 antibodies in rheumatoid arthritis. Arthritis Rheum.
2006;54(8):2377-2386.
57. Anolik JH, Barnard J, Owen T, et al. Delayed memory B cell recovery in peripheral blood and
lymphoid tissue in systemic lupus erythematosus after B cell depletion therapy. Arthritis Rheum.
2007;56(9):3044-3056.
58. Makiyama A, Chiba A, Noto D, et al. Expanded circulating peripheral helper T cells in
systemic lupus erythematosus: association with disease activity and B cell differentiation.
Rheumatology (Oxford). 2019;58(10):1861-1869.
59. Lorenzetti R, Janowska I, Smulski CR, et al. Abatacept modulates CD80 and CD86 expression
and memory formation in human B-cells. J Autoimmun. 2019;101:145-152.
Downloaded from http://ashpublications.org/bloodadvances/article-pdf/doi/10.1182/bloodadvances.2022007025/1895989/bloodadvances.2022007025.pdf by guest on 11 May 2022
18
TABLES
Table 1: A high plasmablast frequency is associated with a higher ADAMTS13 IgG
antibody level at acute iTTP presentation.
Plasmablasts <3% (n=5) Plasmablasts >3%
(n=17)
pvalue *
ADAMTS13 antigen %
(median (range))
5 (0.4 - 86) 1.9 (0.6 - 2.2) 0.4
ADAMTS13 IgG
antibody % (median
(range))
32 (2-113) 64 (52 - 127) 0.02
*Mann Whitney U test
Table 2: Summary of B and cTfh cell subsets at acute iTTP presentation and
ADAMTS13 relapse compared to healthy controls
B and cTfh cell subsets at iTTP presentation
compared to healthy controls (HC)
Acute iTTP
presentation
(n=22)
ADAMTS13
relapse (n=24)
B cell subsets (% CD19+ B cells):
Transitional cells (IgD+CD38++)
Naive cells (IgD+CD38+)
IgD+ memory cells (IgD+CD38-)
Post GC memory cells (IgDCD38+)
Double-negative memory cells (IgDCD38)
Plasmablasts (IgDCD38++)
CD80 expression on B cell subsets:
IgD+ memory cells (IgD+CD38-)
Post GC memory cells (IgDCD38+)
Double-negative memory cells (IgDCD38)
Circulating T follicular helper cells (% CD3+CD4+ cells):
CD4+ CXCR5+ (Total cTfh cells)
CD4+ CXCR5+ PD1+
CD4+ CXCR5+ ICOS+
↓ decreased compared to HC; → no difference compared to HC; ↑ increased compared to
HC
Downloaded from http://ashpublications.org/bloodadvances/article-pdf/doi/10.1182/bloodadvances.2022007025/1895989/bloodadvances.2022007025.pdf by guest on 11 May 2022
19
FIGURE LEGENDS
Figure 1: Percentages of (a) transitional cells (IgD+CD38++), (b) naïve cells (IgD+CD38+)
(c) IgD+ memory B cells (IgD+ CD38-), (d) post germinal centre memory B cells (IgD-
CD38+) and (e) double negative memory B cells (IgD-CD38-) and (f) plasmablasts (igD-
CD38++) in patients with acute iTTP episodes (n=22), ADAMTS13 relapse (n=24) and HC
(n=27).
A13 relapse, ADAMTS13 relapse; Post-GC memory, post germinal centre memory cells; DN
memory cells, double negative memory cells
Figure 2: Percentages of (a) total cTfh (CD4+CXCR5+), (b) PD1+ cTfh
(CD4+CXCR5+PD1+), (c) ICOS+ cTfh (CD4+CXCR5+ICOS+) and (d) PD1+ICOS+ cTfh
(CD4+CXCR5+PD1+ICOS+) in patients with acute iTTP episodes (n=34), ADAMTS13
relapse (n=27) and HC (n=27). A13 relapse, ADAMTS13 relapse.
Figure 3: Relationship between ADAMTS13 antigen levels and plasmablast frequency.
Spearman correlation analysis was performed and p<0.05 indicates that the difference is
statistically significant.
Figure 4: Analysis of CD80 expression (median fluorescence intensity) on B cell subsets
defined by IgD/CD38. (a) Acute iTTP cases and (b) ADAMTS13 relapse cases.
Figure 5: Pairwise comparison of B cell subset frequencies before and after rituximab by
Wilcoxon signed–rank test. a) Acute iTTP episodes, pairwise comparison of 10 patients, b)
ADAMTS13 relapse episodes, pairwise comparison of 13 patients.
Figure 6: Longitudinal changes in B-cell subsets after RTX therapy for an acute iTTP
episode or ADAMTS13 relapse (ADAMTS13 activity 15%) until subsequent ADAMTS13
relapse, and compared to patients who remained in remission over an equivalent period.
Downloaded from http://ashpublications.org/bloodadvances/article-pdf/doi/10.1182/bloodadvances.2022007025/1895989/bloodadvances.2022007025.pdf by guest on 11 May 2022
(a) (b) (c) (d) (e) (f)
Figu re 1
Figure 1
Downloaded from http://ashpublications.org/bloodadvances/article-pdf/doi/10.1182/bloodadvances.2022007025/1895989/bloodadvances.2022007025.pdf by guest on 11 May 2022
(a) (b) (c) (d)
(
a
)
)
Figu re 2
Figure 2
Downloaded from http://ashpublications.org/bloodadvances/article-pdf/doi/10.1182/bloodadvances.2022007025/1895989/bloodadvances.2022007025.pdf by guest on 11 May 2022
0 20 40 60 80 100
0
1
2
3
4
5
9
10
15
16
Correlation: ADAMTS13 Antigen
& Plasmablasts
ADAMTS13 anti
g
en
(
%
)
Plasmablasts (%)
r= -0.41
p= 0.055
F
i
gur e 3
Figure 3
Downloaded from http://ashpublications.org/bloodadvances/article-pdf/doi/10.1182/bloodadvances.2022007025/1895989/bloodadvances.2022007025.pdf by guest on 11 May 2022
(a) Acute iTTP episodes (b) ADAMTS13 relapse
Figure 4
Figure 4
Downloaded from http://ashpublications.org/bloodadvances/article-pdf/doi/10.1182/bloodadvances.2022007025/1895989/bloodadvances.2022007025.pdf by guest on 11 May 2022
(a) (b) (c) (d) (e) (f)
(
c
)
(
e
(
f
)
(a) (b) (c) (d) (e) (f)
F
i
gur e 5 a
)
Figu re 5 b)
Figure 5
Downloaded from http://ashpublications.org/bloodadvances/article-pdf/doi/10.1182/bloodadvances.2022007025/1895989/bloodadvances.2022007025.pdf by guest on 11 May 2022
Figure 6
Downloaded from http://ashpublications.org/bloodadvances/article-pdf/doi/10.1182/bloodadvances.2022007025/1895989/bloodadvances.2022007025.pdf by guest on 11 May 2022
... iTTP is an autoantibody-mediated phenomenon that functions as a Type II immune reaction, and certain aspects of lymphocyte populations-although heretofore not well studied-are characteristic. Recent research indicates that T-follicular helper (Tfh) cells have a yet to be determined role in TTP pathogenesis, and that furthermore, there could be defined differences in these cell populations between new-onset and relapsed iTTP [52]. In effect, new-onset TTP is characterized by the overexpression of and hyperfunction of the memory capabilities of Tfh cells. ...
... In contrast, during relapse, the primed immune system is less able to mount a response. This is especially evidenced by a decrease during relapse of double negative IgD -CD38memory B cells [52]. Similarly, activated plasmablasts are significantly higher in new-onset iTTP as compared to healthy controls but not in relapsed patients [52,53]. ...
... This is especially evidenced by a decrease during relapse of double negative IgD -CD38memory B cells [52]. Similarly, activated plasmablasts are significantly higher in new-onset iTTP as compared to healthy controls but not in relapsed patients [52,53]. Thus, it has been hypothesized that increased plasmablasts increase the clearance rate of ADAMTS13 in new TTP patients due to first-time antibody production [52]. ...
Article
Full-text available
Thrombotic thrombocytopenic purpura (TTP) is an uncommon, but potentially disabling or even deadly, thrombotic microangiopathy with a well-studied mechanism of ADAMTS13 deficiency or dysfunction. While established treatments are largely effective, the standard ADAMTS13 testing required to definitively diagnose TTP may cause delays in diagnosis and treatment, highlighting the need for rapid and effective diagnostic methods. Additionally, the heterogeneous presentation and varied inciting events of TTP suggest more variation in its mechanism than previously thought, implying three potential pathways rather than the accepted two. The recent discovery of ADAMTS13 conformation as a potential contributor to TTP in addition to the proposal of using the absolute immature platelet count (A-IPC) as a biomarker, present novel areas for monitoring and treatment. A-IPC in particular may serve as a more rapid and accurate diagnostic test to distinguish TTP from non-TTP TMAs and to monitor treatment response and relapse. These considerations highlight the need to further study TTP in order to improve best practices and patient care.
... As in other autoimmune disorders, iTTP is characterized by a loss of tolerance resulting in a shift to autoimmunity [2]. Antigens derived from ADAMTS13 molecules, processed by dendritic cells, activate cross-reactive naïve CD41 T cells, which, in turn, differentiate into autoreactive effector CD41 T cells [3,4]. ...
... Autoreactive B cells recirculate into the germinal center (GC) of secondary lymph nodes, stimulated by antigens and auto reactive T helper cells and differentiate into autoAb-producing plasma cells or long-lived memory B cells [5]. Shin and colleagues performed an analysis of B cell subsets and circulating follicular T helper (cfTh) cell changes in iTTP [2]. A decreased number of post-GC memory B cells, an increased number of plasma blasts and a reduction of cfTh compared to healthy controls were found in the acute phase of iTTP. ...
... the germinal center (GC) of secondary lymph nodes, stimulated by antigens and auto reactive T helper cells and differentiate into autoAb-producing plasma cells or long-lived memory B cells [5]. Shin and colleagues performed an analysis of B cell subsets and circulating follicular T helper (cfTh) cell changes in iTTP [2]. A decreased number of post-GC memory B cells, an increased number of plasma blasts and a reduction of cfTh compared to healthy controls were found in the acute phase of iTTP. ...
Article
Full-text available
Thrombotic thrombocytopenic purpura (TTP) is a fatal disease in which platelet-rich microthrombi cause end-organ ischemia and damage. TTP is caused by markedly reduced ADAMTS13 (a disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13) activity. ADAMTS13 autoantibodies (autoAbs) are the major cause of immune TTP (iTTP), determining ADAMTS13 deficiency. The pathophysiology of such autoAbs as well as their prognostic role are continuous objects of scientific studies in iTTP fields. This review aims to provide clinicians with the basic information and updates on autoAbs’ structure and function, how they are typically detected in the laboratory and their prognostic implications. This information could be useful in clinical practice and contribute to future research implementations on this specific topic.
... Patients with multiple sclerosis exhibited significantly elevated PD1 + cTfh cells frequencies; 36 months after autologous haematopoietic stem cell transplantation (AHSCT), PD1 + cTfh cells frequencies were significantly reduced, consistent with normal subjects, suggesting that AHSCT can control the disease by modulating PD1 + cTfh cells expression in patients with multiple sclerosis [109] . Patients with acute immune thrombotic thrombocytopenic purpura had decreased GC memory B cells, total cTfh cells, PD1 + cTfh cells, and increased frequency of circulating plasma cells, while dysregulation of B cell and cTfh cells homeostasis may be caused by hyperactivation of T and B cells leading to differentiation of memory B cells to antibody-producing plasmablast and migration of circulating Tfh cells into the GC [110] . ...
Article
Full-text available
The follicular helper T cells are derived from CD4+T cells, promoting the formation of germinal centers and assisting B cells to produce antibodies. This review describes the differentiation process of Tfh cells from the perspectives of the initiation, maturation, migration, efficacy, and subset classification of Tfh cells, and correlates it with autoimmune disease, to provide information for researchers to fully understand Tfh cells and provide further research ideas to manage immune-related diseases.
Article
Immune thrombotic thrombocytopenic purpura (iTTP) is a microangiopathic hemolytic anemia (MAHA) underpinned by autoreactivity against the von Willebrand factor (vWF) cleaving protease, ADAMTS13 (adisintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13). Autoantibody mediated ADAMTS13 inhibition leads to the accumulation of ultra-large vWF multimers which activate platelets and endothelium to initiate microvascular thrombosis. In the absence of urgent therapeutic intervention, iTTP is rapidly fatal due to cumulative organ dysfunction including catastrophic neurological and cardiac sequalae. Therapeutic plasma exchange (TPE) is the mainstay of initial therapy and aims to remove pathological autoantibodies and ultra-large vWF multimers while replenishing ADAMTS13. Immunosuppression is an important treatment adjunct, as attainment of remission and successful TPE cessation is strongly associated with suppression of anti-ADAMTS13 antibody production. More recently, caplacizumab, an antibody fragment blocking the interaction between vWF multimers and platelets, has been incorporated into acute TTP management to mitigate end-organ damage while awaiting suppression of anti-ADAMTS13 activity. In most cases, remission is achieved using corticosteroids alone or in combination with the B-cell depleting antibody, rituximab. However, some patients are refractory to front-line immunosuppression in the context of ‘inhibitor boosting’ whereby the exposure to homologous plasma exacerbates the underlying autoimmune flare. As such cases have been observed in the context of likely effective B-cell depletion, it has been hypothesised that plasma cells (i.e., terminally differentiated B-cells) may provide a therapy-resistant nidus of anti-ADAMTS13 production as has been demonstrated in other autoimmune disease settings. Autoreactive plasma cells can be targeted by conventional and novel therapeutics, including those developed for malignant plasma cells in the context of multiple myeloma. Here we review the rationale and evidence for plasma cell directed therapy in refractory TTP, with a focus on the proteasome inhibitor, bortezomib, and the CD38 monoclonal antibody, daratumumab.
Article
Full-text available
Since B-cell hyperactivity and pathologic antibody response are key features in the immunopathogenesis of primary Sjögren’s syndrome (pSS), the role of follicular T helper (TFH) cells as efficient helpers in the survival and differentiation of B cells has emerged. Our aim was to investigate whether a change in the balance of circulating (c)TFH subsets and follicular regulatory T (TFR) cells could affect the distribution of B cells in pSS. Peripheral blood of 38 pSS patients and 27 healthy controls was assessed for the frequencies of cTFH cell subsets, TFR cells, and certain B cell subpopulations by multicolor flow cytometry. Serological parameters, including anti-SSA, anti-SSB autoantibodies, immunoglobulin, and immune complex titers were determined as part of the routine diagnostic evaluation. Patients with pSS showed a significant increase in activated cTFH cell proportions, which was associated with serological results. Frequencies of cTFH subsets were unchanged in pSS patients compared to healthy controls. The percentages and number of cTFR cells exhibited a significant increase in autoantibody positive patients compared to patients with seronegative pSS. The proportions of transitional and naïve B cells were significantly increased, whereas subsets of memory B cells were significantly decreased and correlated with autoantibody production. Functional analysis revealed that the simultaneous blockade of cTFH and B cell interaction with anti-IL-21 and anti-CD40 antibodies decreased the production of IgM and IgG. Imbalance in TFH subsets and TFR cells indicates an ongoing over-activated humoral immune response, which contributes to the characteristic serological manifestations and the pathogenesis of pSS.
Article
Full-text available
Immune thrombotic thrombocytopenic purpura (iTTP) is an ultra-rare, life-threatening disorder, mediated through severe ADAMTS13 deficiency causing multi-system micro-thrombi formation, and has specific human leukocyte antigen associations. We undertook a large genome-wide association study to investigate additional genetically distinct associations in iTTP. We compared two iTTP patient cohorts with controls, following standardized genome-wide quality control procedures for single-nucleotide polymorphisms and imputed HLA types. Associations were functionally investigated using expression quantitative trait loci (eQTL), and motif binding prediction software. Independent associations consistent with previous findings in iTTP were detected at the HLA locus and in addition a novel association was detected on chromosome 3 (rs9884090, P=5.22x10-10, odds ratio 0.40) in the UK discovery cohort. Meta-analysis, including the French replication cohort, strengthened the associations. The haploblock containing rs9884090 is associated with reduced protein O-glycosyltransferase 1 (POGLUT1) expression (eQTL P<0.05), and functional annotation suggested a potential causative variant (rs71767581). This work implicates POGLUT1 in iTTP pathophysiology and suggests altered post-translational modification of its targets may influence disease susceptibility.
Article
Full-text available
CD39 and CD73 are ecto-nucleotidases present on human peripheral blood mononuclear cells (PBMCs) and are emerging biomarkers on these cells in various disorders including cancer. Many factors influence PBMC quality, so it is essential to validate sample processing methods prior to incorporation in clinical studies. This study examined the impact of both PBMC cryopreservation and PBMC isolation using SepMate density gradient centrifugation on CD39 and CD73 expressing subsets. First, PBMCs were isolated from the peripheral blood of 11 healthy donors by routine Ficoll-Paque density gradient centrifugation, cryopreserved and compared with freshly isolated PBMCs by flow cytometry. The proportions of T and B cells expressing combinations of CD39 and CD73 were relatively stable over 6-month cryopreservation, although some T cell combinations revealed small but significant changes. Second, peripheral blood was collected from six healthy donors to compare PBMCs isolated by SepMate or Ficoll-Paque density gradient centrifugation. Compared with Ficoll-Paque, the more rapid SepMate method yielded 9.1% less PBMCs but did not alter cell viability or proportions of T and B cells expressing combinations of CD39 and CD73. The present study reveals that cryopreservation is suitable for studying T and B cells expressing combinations of CD39 and CD73. However, caution should be exercised when observing small differences in these cryopreserved subsets between different cohorts. Further, SepMate and Ficoll-Paque methods of PBMC isolation show similar results for T and B cell subset analysis; however, SepMate is a faster and easier approach.
Article
Full-text available
Background Despite an increase in our understandings of pathogenesis of thrombotic thrombocytopenic purpura (TTP), the approaches for initial diagnosis and management of TTP vary significantly. Objective The evidence‐based guidelines of the International Society of Thrombosis and Haemostasis (ISTH) are intended to support patients, clinicians, and other healthcare professionals in their decisions about the initial diagnosis and management of acute TTP. Methods In June 2018, ISTH formed a multidisplinary panel that included hematologists, intensive care physician, nephrologist, clinical pathologist, biostatistician, and patient representatives, as well as a methodology team from McMaster University. The panel composition was designed to minimize the potential conflicts of interests. The panel used the GRADE approach and PICO framework to develop and grade their recommendations. Public comments were sought and incorporated in the final document. Results The panel agreed on 3 recommendations covering the initial diagnosis with emphasis on the importance of ADAMTS13 testing (e.g. activity and anti‐ADAMTS13 IgG or inhibitor) and assessment of the pretest probability of TTP by clinical assessment and/or the risk assessment models like PLASMIC or French score. The panel noted how availability and turn‐around‐time of ADAMTS13 test results might affect early diagnosis and management, in particular the use of caplacizumab. Conclusions There is a lack of high‐quality evidence to support strong recommendations for the initial diagnosis and management of a suspected TTP. The panel emphasized the importance of obtaining ADAMTS13 testing in a proper clinical context. Future research should focus on how to monitor and act on ADAMTS13 levels during remission.
Article
Full-text available
Background: Sarcoidosis is a systemic granulomatous disease characterized by the combination of Th1 and Th17 responses. Recently, several arguments have suggested a potential involvement of B cells as well as T cells in the pathogenesis of sarcoidosis. Follicular helper CD4+ T (TFH) cells are specialized in interacting with and helping B cells, and play a crucial role in the formation of germinal centers. Objective: We sought to explore the status of TFH cells and investigate their possible pathogenic role in sarcoidosis. Methods: TFH cells and B cells in peripheral blood were examined by flow cytometry, and serum samples were studied by cytokine arrays. Immunohistochemistry was performed to check for the presence of TFH cells in sarcoidosis skin lesions. Gene expression in isolated TFH cells was analyzed by quantitative RT-PCR. Results: The proportion of circulating TFH cells was decreased. CD4+CXCR5+ TFH cells were observed in cutaneous lesions in sarcoidosis. Gene expression in circulating TFH cells and serum cytokine concentrations related to Th17 were increased in sarcoidosis patients. Gene expressions of B cell differentiation cytokines in TFH cells were not altered in sarcoidosis patients. Conclusion: We herein describe a decrease of circulating TFH cells and their migration to affected tissues. Circulating TFH cells are one of the potential cell types capable of producing IL-17 and enhancing Th17 responses, and may promote the chronic inflammation. We could not demonstrate a direct linkage between the imbalance of TFH cells and abnormal B cell differentiation in sarcoidosis.
Article
Full-text available
Sarcoidosis is a systemic granulomatous disease that develops due to the Th1, Th17 and Treg lymphocytes disturbance. There is an assumption, that B cells and follicular T-helper (Tfh) cells may play an important role in this disorder, as well as in several other autoimmune diseases. The aim of this study was to determine CD19+ B cells subset distribution in the peripheral blood and to define disturbance in the circulating Tfh cells subsets in patients with sarcoidosis. The prospective comparative study was performed in 2016–2018, where peripheral blood B cell subsets and circulating Tfh cell subsets were analyzed in 37 patients with primarily diagnosed sarcoidosis and 35 healthy donors using multicolor flow cytometry. In the results of our study we found the altered distribution of peripheral B cell subsets with a predominance of “naïve” (IgD + CD27−) and activated B cell (Bm2 and Bm2′) subsets and a decreased frequency of memory cell (IgD+ CD27+ and IgD− CD27+) in peripheral blood of sarcoidosis patients was demonstrated. Moreover, we found that in sarcoidosis patients there are increased levels of B cell subsets, which were previously shown to display regulatory capacities (CD24+++ CD38+++ and CD5 + CD27−). Next, a significantly higher proportion of CXCR5-expressing CD45RA − CCR7+ Th cells in patients with sarcoidosis in comparison to the healthy controls was revealed, that represents the expansion of this memory Th cell subset in the disease. This is the first study to demonstrate the association between the development of sarcoidosis and imbalance of circulating Tfh cells, especially CCR4− and CXCR3-expressing Tfh subsets. Finally, based on our data we can assume that B cells and Tfh2- and Tfh17-like cells – most effective cell type in supporting B-cell activity, particularly in antibody production – may be involved in the occurrence and development of sarcoidosis and in several other autoimmune conditions. Therefore, we can consider these results as a new evidence of the autoimmune mechanisms in the sarcoidosis development.
Article
Full-text available
Primary Sjögren's syndrome is a chronic autoimmune disorder of unknown etiology and is characterized by progressive focal lymphocytic infiltration of the lacrimal and salivary glands. Comparison of B cell subsets from the peripheral blood and salivary glands of patients with primary Sjögren's syndrome and those from healthy individuals shows dysregulation and derangement of B cell subsets in both peripheral circulation and in inflamed glandular tissues. This dysregulation is expressed as a decrease in the percentage of CD27+ memory B cells in peripheral blood and an increase in the CD27+ memory B cells in the affected glands. Further, the overall percentage of long-lived autoantibodies-producing plasma cells within the affected glands is increased. In the last two decades, several studies have shown growing evidences that B cells play multiple roles in primary Sjögren's syndrome pathophysiology, and that dysregulation of these cells may actually play a central role in the disease development.
Article
Immune-mediated thrombotic thrombocytopenic purpura (iTTP) is a potentially fatal thrombotic microangiopathy caused by autoantibody-mediated severe deficiency of ADAMTS13. Standardized definitions of response, exacerbation, remission and relapse were initially proposed in 2003 and modified by the International Working Group (IWG) for TTP in 2017. These definitions, which have been widely used in clinical practice and research, are based primarily on the platelet count and are benchmarked against the timing of discontinuation of therapeutic plasma exchange (TPE). They do not incorporate ADAMTS13 activity or the temporizing effects of caplacizumab, a novel anti-von Willebrand factor (VWF) nanobody, on the platelet count. In light of these limitations, the IWG aimed to develop revised consensus outcome definitions that incorporate ADAMTS13 activity and the effects of anti-VWF therapy using an estimate-talk-estimate approach. The updated definitions distinguish clinical remission and clinical relapse (defined primarily by platelet count) from ADAMTS13 remission and ADAMTS13 relapse (defined by ADAMTS13 activity). The revised definitions of exacerbation and remission are benchmarked against not only the timing of discontinuation of TPE, but also of anti-VWF therapy. Retrospective validation of the revised definitions is described, though they remain to be prospectively validated. Clinical implications of the updated outcome definitions are also discussed and an example of their application to clinical practice is provided in order to highlight their clinical relevance.
Article
Objective: Peripheral helper T (TPH) cells are a recently identified Th cell subset that promotes B cell differentiation and antibody production in inflamed tissues. This study investigated circulating TPH cells to determine their involvement in systemic lupus erythematosus (SLE). Methods: Peripheral blood mononuclear cells collected from SLE patients and healthy individuals were analysed. TPH cells were identified as CD3+CD4+CD45RA-CXCR5- cells with a high expression of PD-1. The frequency, activation status and subsets of TPH cells were evaluated by flow cytometry. The production of IL-21 was assessed by intracellular staining and the association of TPH cells with disease activity and B cell populations was determined. Results: Circulating TPH cells, identified as CD3+CD4+CD45RA-PD-1highCXCR5- cells were increased in the peripheral blood of SLE patients compared with controls. Circulating TPH cells produced similar amounts of IL-21 compared with follicular Th cells. The expansion and activation of TPH cells were correlated with SLE disease activity. Activated TPH cells, particularly Th1-type TPH cells, were associated with the promotion of B cell differentiation in SLE patients. Conclusion: The association of TPH cells with disease activity suggests the involvement of extrafollicular T-B cell interactions in the pathogenesis of SLE. TPH cells promote autoantibody production in aberrant lymphoid organs and therefore might be a novel therapeutic target in autoantibody-producing disorders.