Over the last decade, our understanding of the pathophysiology of chronic graft-versus-host disease (cGVHD) has improved considerably. In this spotlight, we discuss emerging insights into the pathophysiology of cGVHD with a focus on B cells. First, we summarize supporting evidence derived from mouse and human studies. Next, novel cGVHD therapy approaches that target B cells will be covered to provide treating physicians with an overview of the rationale behind the emerging armamentarium against cGVHD.

Chronic graft-versus-host disease (cGVHD) is a major complication in patients undergoing allogeneic hematopoietic cell transplantation (allo-HCT), leading to reduced patient-reported quality of life1  and nonrelapse mortality.2  Risk factors for cGVHD development include prior acute GVHD, donor peripheral blood stem-cell grafts, HLA disparity, female donors for male recipients, and recipient age.3  Clinical cGVHD can involve classical acute GVHD epithelial target tissues (intestinal tract, liver, skin, lung) and any other organ system, including oral, esophageal, musculoskeletal, joint, fascial, ocular, hair and nails, lymphohematopoietic system, and genital tissues.4  The pleiotrophic symptoms resulting from such broad organ involvement made past diagnosis and scoring difficult. The 2005 and revised 2014 National Institutes of Health (NIH) criteria have brought greater consistency to terminology and methods for cGVHD diagnosis and staging.4,5 

To identify and validate novel targets in cGVHD, numerous mouse models are used. However, individual cGVHD mouse models cannot reproduce all features of cGVHD seen in patients (as reviewed by Zeiser and Blazar6,7 ), who present with a heterogeneous disease spectrum. Most models have 1 or 2 dominant cGVHD manifestations involving limited numbers of organs. These different manifestations of cGVHD depend on several factors, including the cytokines that are released. Some of these cytokines or their receptors are attractive targets to treat cGVHD. For instance, an anti–interleukin 2 (IL-2) receptor common γ chain neutralizing monoclonal antibody (mAb) reduced cGVHD,8  a result that may be based on a broad inhibitory effect on multiple cytokine receptors. Also, targeting of individual cytokines such as IL-17 was active against cGVHD.9  Additionally, the type and degree of donor and recipient genetic disparity in models suggest that the antigens recognized by B and T cells as well as the number of donor T cells transferred can dictate cGVHD phenotypes. Thus, mechanistic studies of multiple models when feasible are ideal.10 

Under normal conditions, B cells contribute to adaptive immunity by producing antibodies, secreting cytokines, and presenting antigen. B-cell activation begins when an antigen is recognized via the B-cell receptor (BCR). Activated B cells participate in a 2-step differentiation process that yields both short-lived plasmablasts for immediate protection against a pathogen and long-lived plasma cells and memory B cells for persistent protection.11  Together with BCR signaling, B-cell activating factor (BAFF) determines B-cell fate/survival. Comparable to the normal B-cell activation process, the first step in the pathogenesis of cGVHD is the recognition of antigen via the BCR (Figure 1A step 1). In contrast to the normal situation, B cells exhibit BCR hyperresponsiveness in cGVHD as shown in mouse models.12-14  After activation, pathogenic B cells expand (Figure 1A step 2) and are strongly affected by soluble factors in the microenvironment such as IL-4, IL-17,9  IL-21,12,15  and BAFF16  (Figure 1A step 3). This process is connected to the formation of GCs in cooperation with donor Tfhs. GC B cells undergo somatic hypermutation that can favor cGVHD by increasing the frequency of B cells capable of producing antibody to antigens that trigger BCR.

Figure 1.

The role of B cells in cGVHD. (A) Different steps of cGVHD development. Step 1: antigen (Ag)-presenting cells (APCs) present auto- and alloantigens and prime B cells. Direct activation of B cells via Ag or Ag/Ab complexes. APCs prime B cells against major histocompatibility complexes/peptides or neoantigens (eg, Y chromosome–encoded genes). This is enhanced in certain B-cell subgroups by hyperreactive BCR signaling. In addition to B-cell activation by APCs, there is likely also direct BCR activation via Ag or Ab/Ag complexes. Step 2: expansion of auto- and alloreactive B cells. Step 3: activated T follicular helper cells (Tfhs) produce IL-21 and cell-surface costimulatory molecules that lead to germinal center (GC) formation, which is not counterbalanced by sufficient T follicular regulatory cells (Tfrs). CD4 T helper cells produce IL-4, which promotes Ab class switch in autoreactive B cells. Stroma cells produce BAFF, which promotes B-cell activation. Step 4: plasma cells and plasma blasts produce high amounts of immunoglobulin. Deposition of immunoglobulin G (IgG) can lead to macrophage activation and organ damage. IgG-induced macrophage activation may contribute to cGVHD via secretion of proinflammatory cytokines by macrophages such as IL-6, which promotes B-cell survival and maintains inflammation. (B) Strategies to target B cells in cGVHD. The sketch shows a B cell and the mode of action of multiple immunosuppressive strategies that directly act on B cells or plasma cells in the context of cGVHD. The summary of translation of each approach is provided in Table 1. BTK, Bruton tyrosine kinase; ITK, IL-2–inducible kinase; MMF, mycophenolate mofetil; mTOR, mammalian target of rapamycin; MTX, methotrexate; ROCK2, ρ-GTPase kinase-2; SYK, splenic tyrosine kinase.

Figure 1.

The role of B cells in cGVHD. (A) Different steps of cGVHD development. Step 1: antigen (Ag)-presenting cells (APCs) present auto- and alloantigens and prime B cells. Direct activation of B cells via Ag or Ag/Ab complexes. APCs prime B cells against major histocompatibility complexes/peptides or neoantigens (eg, Y chromosome–encoded genes). This is enhanced in certain B-cell subgroups by hyperreactive BCR signaling. In addition to B-cell activation by APCs, there is likely also direct BCR activation via Ag or Ab/Ag complexes. Step 2: expansion of auto- and alloreactive B cells. Step 3: activated T follicular helper cells (Tfhs) produce IL-21 and cell-surface costimulatory molecules that lead to germinal center (GC) formation, which is not counterbalanced by sufficient T follicular regulatory cells (Tfrs). CD4 T helper cells produce IL-4, which promotes Ab class switch in autoreactive B cells. Stroma cells produce BAFF, which promotes B-cell activation. Step 4: plasma cells and plasma blasts produce high amounts of immunoglobulin. Deposition of immunoglobulin G (IgG) can lead to macrophage activation and organ damage. IgG-induced macrophage activation may contribute to cGVHD via secretion of proinflammatory cytokines by macrophages such as IL-6, which promotes B-cell survival and maintains inflammation. (B) Strategies to target B cells in cGVHD. The sketch shows a B cell and the mode of action of multiple immunosuppressive strategies that directly act on B cells or plasma cells in the context of cGVHD. The summary of translation of each approach is provided in Table 1. BTK, Bruton tyrosine kinase; ITK, IL-2–inducible kinase; MMF, mycophenolate mofetil; mTOR, mammalian target of rapamycin; MTX, methotrexate; ROCK2, ρ-GTPase kinase-2; SYK, splenic tyrosine kinase.

Close modal

IL-4 produced by CD4 T cells promotes B-cell immunoglobulin isotype switching,17-19  allowing daughter cells from the same activated B cell to produce secreted pathogenic IgG in cGVHD mice.12,17  Tfhs produce IL-21, which can promote auto- and alloreactive B-cell activation and survival along with increased local BAFF levels in cGVHD.16  Although the role of GCs in cGVHD initiation is likely to be important in many cGVHD mouse models, GCs were found not to be required for disease development in a recent report,20  possibly reflecting the wide clinical spectrum of cGVHD in patients. In a consecutive step, activated B cells can promote tissue injury via antibody and cytokine production and release, leading to clinical manifestations of cGVHD (Figure 1A step 4). IgG-induced macrophage activation may contribute to cGVHD via secretion of proinflammatory cytokines such as IL-6 and IL-22,21  which maintain inflammation. Tissue stiffness in cGVHD can be enhanced by copious immunoglobulin production and deposition together with fibroblast-derived extracellular matrix molecules including collagen and proteoglycans (Figure 1A step 4).

Pathogenic B-cell activation is found in various autoimmune diseases including systemic lupus erythematosus, multiple sclerosis, rheumatoid arthritis, type 1 diabetes, and others as well as in cGVHD.17,19  During cGVHD, donor B cells and T cells mount a coordinated response to both allogeneic and autologous antigens, which leads to their expansion (Figure 1A steps 1 and 2). Allogeneic antigens include minor histocompatibility antigens22,23  that are typically expressed or processed intracellularly and presented as peptides by major histocompatibility complex molecules. These include Y chromosome proteins/peptides in male recipients of female donor grafts, as well as cell membrane antigens, the former correlating with cGVHD by multivariable logistic regression analysis.24  Autoantigens are antigens on donor hematopoietic cells, which can be found for example on megakaryocytes or platelets. In agreement with the concept of recognition of autoantigens, patients can develop autoimmune thrombocytopenia after allo-HCT, which is mediated by antibodies produced by donor B cells and directed against donor platelets.

BAFF promotes B-cell survival and activation (Figure 1A step 3) and is significantly increased in plasma of patients with cGVHD.25,26  BAFF and BCR-associated signaling work in concert to promote activation and survival of B cells from patients with cGVHD.27  In those with cGVHD, B cells exhibit increased BCR responsiveness27  via increased proximal BCR intracellular signaling molecules SYK and B-cell linker (BLNK).28  In that context, it is important to understand which cell-intrinsic mechanisms enhance BCR responses. A novel observation here is that BCR responses to surrogate antigen were markedly increased when NOTCH2 was also activated.29  Intrinsic differences in important transcription factors like IRF4 contributed to NOTCH2 expression and responsiveness. How extrinsic factors like BAFF and intrinsic molecular pathways like NOTCH promote BCR-activated B cells is currently not clear but is an area of active investigation.

Tfhs can support antihost antibody production.30  This process typically takes place in GCs, areas of lymph nodes where B cells are activated in mice,30  but where this occurs in cGVHD in patients remains unknown. As in patients with autoimmune disease, this process also may occur in extrafollicular locations. Antigen targets of B-cell responses in cGVHD remain largely unknown, but ultimately, both auto- and alloimmune B-cell responses can occur. Lack of sufficient T regulatory cells (Tregs) in patients with cGVHD can contribute to impaired peripheral tolerance.31  Tregs are capable of selectively killing B cells,32  and their deficiency would predispose to a failure to control pathogenic B cells. Although human memory Tregs expand after allo-HCT, they cannot compensate for the lack of naïve Tregs, because of short telomeres and increased apoptosis.33  cGVHD tissue stiffness and organ dysfunction are likely supported by cooperation between B cells and macrophages, leading to fibroblast activation; however, so far there is no direct evidence for this interaction (Figure 1A step 4).

Under homeostatic conditions, multiple mechanisms prevent pathogenic B-cell function via central (thymic) and peripheral tolerance. In patients undergoing allo-HCT, uncontrolled expansion and immunoglobulin production by B cells possibly occurs because of thymic dysfunction. Impaired thymic function is caused by aging, conditioning regimen toxicity, calcineurin inhibitors, alloreactive T cells, and immunoglobulin deposition.19,34  Alloreactive T cells contribute to the process by depleting thymic dendritic cells, medullary thymic epithelial cells (TECs), and cortical TECs.34,35  A recent report also suggests pathologic antibodies target TECs in a cGVHD model.15,18  GVHD affects both positive selection by cortical TECs and negative selection by thymic B cells and cortical TECs,34,36  which allows potentially pathogenic CD4+ T cells to escape from tolerization or deletion before peripheral export37,38  and impedes the development of Tregs that contribute to peripheral tolerance.

Mouse studies revealed that peripheral immune tolerance to recipient tissues after transplantation is mediated by Tregs, Tfrs representing Tregs that migrate to the GCs,12  regulatory B cells,39  type 1 regulatory T cells,40  and invariant natural killer T cells.41-43  Tregs and Tfrs negatively regulate B-cell responses and cGVHD,43  and B regulatory cells that release IL-10 have been shown to ameliorate sclerodermatous cGVHD severity.44  In agreement with these mouse studies, analysis in patients with cGVHD suggests that B cells with a regulatory phenotype are both decreased and inactive.39,45  Increased T-cell help decreases self-regulation by B cells by promoting aberrant B-cell generation. Additionally, the absence of robust recovery of the peripheral B-cell compartment results in excess BAFF and promotion of autoreactive B cells that can cooperate to overwhelm peripheral tolerance mechanisms in those with cGVHD.46  Additionally, thymic T-cell generation, negative selection of antihost reactive T cells, thymic Treg production, and peripheral Treg survival are severely reduced in patients with cGVHD.31,47,48 

B-cell depletion with anti-CD20 antibodies was performed in preclinical models and patients.12,49,50  Anti-CD20 mAbs administered in the prophylactic setting reduced murine cGVHD, whereas established cGVHD was nonresponsive.12,49  In the clinical setting, the anti-CD20 mAb rituximab conferred some efficacy in patients with steroid-refractory cGVHD (SR-cGVHD),51  with attenuation of cGVHD in those patients who robustly recovered B cells.46,52  A prospective phase 2 trial showed that naïve B cells (PD-L1hi) were significantly reduced at cGVHD diagnosis but increased after rituximab treatment.50  To target plasma cells, different drugs that have been successfully used in the treatment of multiple myeloma such as pomalidomide53  were tested in cGVHD (Table 1; Figure 1B). IL-6 was shown to contribute to cGVHD. Because IL-6 is known to promote plasma blast and plasma-cell survival,54  further study of IL-6 and B cells is warranted. The anti–IL-6 receptor mAb tocilizumab is being investigated in a clinical trial as therapy for cGVHD.55  In other diseases, IL-6 also has a known role in promotion of collagen deposition and extracellular matrix production by fibroblasts.56 

Table 1.

Targeting B cells in cGVHD

Target nameNormal functionName of drugs testedSpecies analyzedEvidence for role in cGVHDReference
BTK and ITK Downstream of BCR activation Ibrutinib Mouse Yes 14,58  
Human Yes 59  
CD20 B-cell surface antigen Rituximab Mouse Yes (effective only in prevention) 49  
Human Yes 46,50,-52  
CD30 B cells express CD30 Brentuximab Human Clinical trials ongoing 74  
IL-6 receptor IL-6 induces proliferation of pre–B cells Tocilizumab Human Clinical trials ongoing 55  
JAK 1/2 JAK1/2 mediate downstream effects of cytokine and chemokine receptors in B cells63  Ruxolitinib Mouse Yes 64  
Retrospective clinical data Yes 64  
Prospective phase 3 trial ongoing Clinical trials ongoing 65  
Plasma cells Production of immunoglobulin that causes organ damage in cGVHD Pomalidomide Human Clinical trials ongoing 53  
Proteasome Activation of the proteasome is important in plasma cells Bortezomib Mouse Yes 75  
Human Clinical trials ongoing 76  
Proteasome Activation of the immunoproteasome is important in plasma cells Carfilzomib Human Clinical trials ongoing 77  
ROCK2 T-cell activation with pSTAT3 and pSTAT5 effects B-cell migration KD025 Mouse Yes 61  
Decreases Tfhs  
Human Clinical trials ongoing 62  
SYK Downstream of BCR activation Cell migration Endocytosis Entospletinib Fostamatinib Mouse Yes 10,13  
Clinical trials ongoing 57  
Target nameNormal functionName of drugs testedSpecies analyzedEvidence for role in cGVHDReference
BTK and ITK Downstream of BCR activation Ibrutinib Mouse Yes 14,58  
Human Yes 59  
CD20 B-cell surface antigen Rituximab Mouse Yes (effective only in prevention) 49  
Human Yes 46,50,-52  
CD30 B cells express CD30 Brentuximab Human Clinical trials ongoing 74  
IL-6 receptor IL-6 induces proliferation of pre–B cells Tocilizumab Human Clinical trials ongoing 55  
JAK 1/2 JAK1/2 mediate downstream effects of cytokine and chemokine receptors in B cells63  Ruxolitinib Mouse Yes 64  
Retrospective clinical data Yes 64  
Prospective phase 3 trial ongoing Clinical trials ongoing 65  
Plasma cells Production of immunoglobulin that causes organ damage in cGVHD Pomalidomide Human Clinical trials ongoing 53  
Proteasome Activation of the proteasome is important in plasma cells Bortezomib Mouse Yes 75  
Human Clinical trials ongoing 76  
Proteasome Activation of the immunoproteasome is important in plasma cells Carfilzomib Human Clinical trials ongoing 77  
ROCK2 T-cell activation with pSTAT3 and pSTAT5 effects B-cell migration KD025 Mouse Yes 61  
Decreases Tfhs  
Human Clinical trials ongoing 62  
SYK Downstream of BCR activation Cell migration Endocytosis Entospletinib Fostamatinib Mouse Yes 10,13  
Clinical trials ongoing 57  

Presented in alphabetical order.

Several small-molecule inhibitors are now in the pipeline, building upon the observation that patients with cGVHD have hyperreactive BCR signaling via the BCR proximal tyrosine kinase SYK. SYK was found to be upregulated in cGVHD B cells in mice12,13  and patients.28  SYK inhibition reduced established murine cGVHD, was associated with reduced GC responses, and activated CD80/86+ dendritic cell responses10  and induced apoptosis in B cells of patients with cGVHD.10,13,28  On the basis of these promising findings, the SYK inhibitor entospletinib, recently granted US Food and Drug Administration (FDA) orphan drug status, is being studied as first-line treatment with steroids.57  Further downstream of the BCR is BTK. In B cells of patients with cGVHD, phosphorylated BTK was present in the absence of in vitro stimulation by anti-IgM.14  In agreement with a role of BTK, cGVHD severity was reduced in murine recipients receiving donor B cells lacking BTK or ibrutinib that targets BTK.14  Ibrutinib additionally inhibits ITK,14  and in a cGVHD model where T cells lacked ITK, cGVHD was reduced.14  On the basis of these findings, it is likely, but not formally proven, that both BTK and ITK inhibition are critical to the efficacy of ibrutinib in cGVHD. In patients with cGVHD, ibrutinib reduced murine sclerodermatous and multiorgan system cGVHD as well as T- and B-cell activation.14,58  Guided by these preclinical data, an open-label phase 2 study evaluated the safety and efficacy of ibrutinib in patients with active cGVHD with SR-cGVHD.59  At a median follow-up of 13.9 months, best overall response was 67% (sustained ≥20 weeks in 71% of responders).59  On the basis of these clinical data and upon the foundations of the applied NIH consensus criteria from 2005, ibrutinib was FDA approved for SR-cGVHD.

With better understanding of the role of B cells in cGVHD pathogenesis, multiple additional strategies have been developed that deplete B cells, reduce their activation via manipulation of BCR-downstream events, or inhibit their migration toward inflammatory sites. Other agents also potentially target cytokine-mediated B-cell differentiation or survival. In normal mice and healthy volunteers, in vitro Tfh generation depends upon the ROCK2.60  In both murine sclerodermatous and multiorgan system cGVHD models, ROCK2 inhibition with KD025 ameliorated ongoing cGVHD, was associated with reduced Tfhs resulting from inhibition of pSTAT3 and IL-21 production, and increased Tfrs as a result of augmentation of pSTAT5 signaling.61  A phase 2a KD025 trial to treat SR-cGVHD62  is ongoing. BCR stimulation also activates JAK2/STAT3 signaling.63  In mice, JAK1/2 blockade with ruxolitinib inhibited multiple murine cGVHD features.64  Clinical responses were reported in a survey of patients with SR-cGVHD treated with ruxolitinib.64  On the basis of these promising results, a phase 3 multicenter ruxolitinib trial for treating SR-cGVHD65  is in progress. How the B-cell compartment is affected by these agents is unclear.

Pirfenidone inhibits TGF-β receptor signaling; downregulates NLRP3 inflammasomes, growth factors, and procollagen I and II; and is FDA approved for treating idiopathic pulmonary fibrosis. Pirfenidone treatment of established murine cGVHD restored pulmonary function and reversed lung fibrosis and was associated with reduced pulmonary macrophage infiltration and TGF-β production.66  How B cells are affected by agents that block fibrotic pathways requires further investigation.

Autoreactive B-cell regulation is mediated via Tregs. Tregs have the capacity to control recipient reactive B cells, with their expansion and survival dependent upon IL-2 production by T effector cells.67  Thus, low-dose IL-2 infusion has been tested as cGVHD treatment. A phase 1/2 study showed that exogenous IL-2 increased Tregs and improved cGVHD.68,69  On the basis of the defects in Tregs reported for patients with cGVHD, 31,48  a clinical study analyzed the feasibility and efficacy of human expanded Tregs administered to patients with cGVHD.70  The study reported that 2 of 5 treated patients achieved a complete remission.

Recent advances in our understanding of the role of B cells in cGVHD pathogenesis have paved the way for novel strategies that target activation, expansion, survival, and Ab production of B cells. Studies are urgently needed, because the first-line gold standard for cGVHD therapy remains steroids, which have multiple severe adverse effects. Both mouse and human studies of B-cell pathways have been a major driver in testing the aforementioned novel therapies. These drugs were in some instances already clinically applied in other diseases. In spite of their potential clinical benefit, an important clinical consideration is that cGVHD is connected to overall reduction in relapse.71  Thus, overly intensive cGVHD prevention may lead to reduced graft-versus-leukemia activity. Clinical judgment, the application of the NIH criteria for cGVHD diagnosis and scoring,4,72  novel cGVHD biomarkers,73  and measurement tools will be essential to make clinical meaningful progress in cGVHD treatment via B-cell targeting.

Contribution: All authors contributed equally to the writing of the manuscript.

Conflict-of-interest disclosure: R.Z. has received honorarium from Novartis and research funding from Jazz Pharma; S.S. has had a consultant/advisory role with Gilead and Pharmacyclics; and B.R.B. has had a consultant/advisory role with Tobira Therapeutics, Vulcan Capital, Idera Pharma, Sidley Austin LLP, Merck Sharpe & Dohme Corp, Merck Serono, Fate Therapeutics, Bristol-Myers Squibb, Sidley Austin, Kadmon Pharmaceuticals Inc, Kymab Scientific, Five Prime Therapeutics, Vitae Pharmaceuticals Inc, and Flx Bio; received research funding from Kadmon Corporation; and held patents/royalties/other intellectual property as an individual (no company).

Correspondence: Robert Zeiser, Department of Hematology, Oncology and Stem Cell Transplantation, University Medical Center Freiburg, Freiburg, D-79106 Freiburg, Germany; e-mail: robert.zeiser@uniklinik-freiburg.de.

1.
Pidala
J
,
Kurland
B
,
Chai
X
, et al
.
Patient-reported quality of life is associated with severity of chronic graft-versus-host disease as measured by NIH criteria: report on baseline data from the Chronic GVHD Consortium
.
Blood
.
2011
;
117
(
17
):
4651
-
4657
.
2.
Arai
S
,
Jagasia
M
,
Storer
B
, et al
.
Global and organ-specific chronic graft-versus-host disease severity according to the 2005 NIH Consensus Criteria
.
Blood
.
2011
;
118
(
15
):
4242
-
4249
.
3.
Flowers
ME
,
Inamoto
Y
,
Carpenter
PA
, et al
.
Comparative analysis of risk factors for acute graft-versus-host disease and for chronic graft-versus-host disease according to National Institutes of Health consensus criteria
.
Blood
.
2011
;
117
(
11
):
3214
-
3219
.
4.
Jagasia
MH
,
Greinix
HT
,
Arora
M
, et al
.
National Institutes of Health Consensus Development Project on criteria for clinical trials in chronic graft-versus-host disease: I. The 2014 Diagnosis and Staging Working Group report
.
Biol Blood Marrow Transplant
.
2015
;
21
(
3
):
389
-
401.e1
.
5.
Filipovich
AH
,
Weisdorf
D
,
Pavletic
S
, et al
.
National Institutes of Health consensus development project on criteria for clinical trials in chronic graft-versus-host disease: I. Diagnosis and staging working group report
.
Biol Blood Marrow Transplant
.
2005
;
11
(
12
):
945
-
956
.
6.
Zeiser
R
,
Blazar
BR
.
Preclinical models of acute and chronic graft-versus-host disease: how predictive are they for a successful clinical translation?
Blood
.
2016
;
127
(
25
):
3117
-
3126
.
7.
Zeiser
R
,
Blazar
BR
.
Pathophysiology of chronic graft-versus-host disease and therapeutic targets
.
N Engl J Med
.
2017
;
377
(
26
):
2565
-
2579
.
8.
Hechinger
AK
,
Smith
BA
,
Flynn
R
, et al
.
Therapeutic activity of multiple common γ-chain cytokine inhibition in acute and chronic GVHD
.
Blood
.
2015
;
125
(
3
):
570
-
580
.
9.
Forcade
E
,
Paz
K
,
Flynn
R
, et al
.
An activated Th17-prone T cell subset involved in chronic graft-versus-host disease sensitive to pharmacological inhibition
.
JCI Insight
.
2017
;
2
(
12
):
e92111
.
10.
Flynn
R
,
Allen
JL
,
Luznik
L
, et al
.
Targeting Syk-activated B cells in murine and human chronic graft-versus-host disease
.
Blood
.
2015
;
125
(
26
):
4085
-
4094
.
11.
Nutt
SL
,
Hodgkin
PD
,
Tarlinton
DM
,
Corcoran
LM
.
The generation of antibody-secreting plasma cells
.
Nat Rev Immunol
.
2015
;
15
(
3
):
160
-
171
.
12.
Flynn
R
,
Du
J
,
Veenstra
RG
, et al
.
Increased T follicular helper cells and germinal center B cells are required for cGVHD and bronchiolitis obliterans
.
Blood
.
2014
;
123
(
25
):
3988
-
3998
.
13.
Le Huu
D
,
Kimura
H
,
Date
M
, et al
.
Blockade of Syk ameliorates the development of murine sclerodermatous chronic graft-versus-host disease
.
J Dermatol Sci
.
2014
;
74
(
3
):
214
-
221
.
14.
Dubovsky
JA
,
Flynn
R
,
Du
J
, et al
.
Ibrutinib treatment ameliorates murine chronic graft-versus-host disease
.
J Clin Invest
.
2014
;
124
(
11
):
4867
-
4876
.
15.
Young
JS
,
Wu
T
,
Chen
Y
, et al
.
Donor B cells in transplants augment clonal expansion and survival of pathogenic CD4+ T cells that mediate autoimmune-like chronic graft-versus-host disease
.
J Immunol
.
2012
;
189
(
1
):
222
-
233
.
16.
Jia
W
,
Poe
JC
,
Su
H
, et al
.
Recipient-derived BAFF and alloantigen synergistically activate B cells in murine chronic GVHD [abstract]
.
Blood
.
2016
;
128
(
22
).
Abstract 498
.
17.
Jin
H
,
Ni
X
,
Deng
R
, et al
.
Antibodies from donor B cells perpetuate cutaneous chronic graft-versus-host disease in mice
.
Blood
.
2016
;
127
(
18
):
2249
-
2260
.
18.
Sarantopoulos
S
.
Antibodies are back for thymic attack in cGVHD
.
Blood
.
2016
;
127
(
18
):
2170
-
2171
.
19.
Srinivasan
M
,
Flynn
R
,
Price
A
, et al
.
Donor B-cell alloantibody deposition and germinal center formation are required for the development of murine chronic GVHD and bronchiolitis obliterans
.
Blood
.
2012
;
119
(
6
):
1570
-
1580
.
20.
Deng
R
,
Hurtz
C
,
Song
Q
, et al
.
Extrafollicular CD4+ T-B interactions are sufficient for inducing autoimmune-like chronic graft-versus-host disease
.
Nat Commun
.
2017
;
8
(
1
):
978
.
21.
Gartlan
KH
,
Bommiasamy
H
,
Paz
K
, et al
.
A critical role for donor-derived IL-22 in cutaneous chronic GVHD [published online ahead of print 23 September 2017]
.
Am J Tranplant
.
doi:10.1111/ajt.14513
.
22.
Zorn
E
,
Miklos
DB
,
Floyd
BH
, et al
.
Minor histocompatibility antigen DBY elicits a coordinated B and T cell response after allogeneic stem cell transplantation
.
J Exp Med
.
2004
;
199
(
8
):
1133
-
1142
.
23.
Porcheray
F
,
Miklos
DB
,
Floyd
BH
, et al
.
Combined CD4 T-cell and antibody response to human minor histocompatibility antigen DBY after allogeneic stem-cell transplantation
.
Transplantation
.
2011
;
92
(
3
):
359
-
365
.
24.
Miklos
DB
,
Kim
HT
,
Miller
KH
, et al
.
Antibody responses to H-Y minor histocompatibility antigens correlate with chronic graft-versus-host disease and disease remission
.
Blood
.
2005
;
105
(
7
):
2973
-
2978
.
25.
Sarantopoulos
S
,
Stevenson
KE
,
Kim
HT
, et al
.
High levels of B-cell activating factor in patients with active chronic graft-versus-host disease
.
Clin Cancer Res
.
2007
;
13
(
20
):
6107
-
6114
.
26.
Sarantopoulos
S
,
Stevenson
KE
,
Kim
HT
, et al
.
Altered B-cell homeostasis and excess BAFF in human chronic graft-versus-host disease
.
Blood
.
2009
;
113
(
16
):
3865
-
3874
.
27.
Allen
JL
,
Fore
MS
,
Wooten
J
, et al
.
B cells from patients with chronic GVHD are activated and primed for survival via BAFF-mediated pathways
.
Blood
.
2012
;
120
(
12
):
2529
-
2536
.
28.
Allen
JL
,
Tata
PV
,
Fore
MS
, et al
.
Increased BCR responsiveness in B cells from patients with chronic GVHD
.
Blood
.
2014
;
123
(
13
):
2108
-
2115
.
29.
Poe
JC
,
Jia
W
,
Su
H
, et al
.
An aberrant NOTCH2-BCR signaling axis in B cells from patients with chronic GVHD
.
Blood
.
2017
;
130
(
19
):
2131
-
2145
.
30.
Forcade
E
,
Kim
HT
,
Cutler
C
, et al
.
Circulating T follicular helper cells with increased function during chronic graft-versus-host disease
.
Blood
.
2016
;
127
(
20
):
2489
-
2497
.
31.
Zorn
E
,
Kim
HT
,
Lee
SJ
, et al
.
Reduced frequency of FOXP3+ CD4+CD25+ regulatory T cells in patients with chronic graft-versus-host disease
.
Blood
.
2005
;
106
(
8
):
2903
-
2911
.
32.
Zhao
DM
,
Thornton
AM
,
DiPaolo
RJ
,
Shevach
EM
.
Activated CD4+CD25+ T cells selectively kill B lymphocytes
.
Blood
.
2006
;
107
(
10
):
3925
-
3932
.
33.
Kawano
Y
,
Kim
HT
,
Matsuoka
K
, et al
.
Low telomerase activity in CD4+ regulatory T cells in patients with severe chronic GVHD after hematopoietic stem cell transplantation
.
Blood
.
2011
;
118
(
18
):
5021
-
5030
.
34.
Wu
T
,
Young
JS
,
Johnston
H
, et al
.
Thymic damage, impaired negative selection, and development of chronic graft-versus-host disease caused by donor CD4+ and CD8+ T cells
.
J Immunol
.
2013
;
191
(
1
):
488
-
499
.
35.
Tivol
E
,
Komorowski
R
,
Drobyski
WR
.
Emergent autoimmunity in graft-versus-host disease
.
Blood
.
2005
;
105
(
12
):
4885
-
4891
.
36.
Yamano
T
,
Nedjic
J
,
Hinterberger
M
, et al
.
Thymic B cells are licensed to present self antigens for central T cell tolerance induction
.
Immunity
.
2015
;
42
(
6
):
1048
-
1061
.
37.
Dertschnig
S
,
Hauri-Hohl
MM
,
Vollmer
M
,
Holländer
GA
,
Krenger
W
.
Impaired thymic expression of tissue-restricted antigens licenses the de novo generation of autoreactive CD4+ T cells in acute GVHD
.
Blood
.
2015
;
125
(
17
):
2720
-
2723
.
38.
Sakoda
Y
,
Hashimoto
D
,
Asakura
S
, et al
.
Donor-derived thymic-dependent T cells cause chronic graft-versus-host disease
.
Blood
.
2007
;
109
(
4
):
1756
-
1764
.
39.
Khoder
A
,
Sarvaria
A
,
Alsuliman
A
, et al
.
Regulatory B cells are enriched within the IgM memory and transitional subsets in healthy donors but are deficient in chronic GVHD
.
Blood
.
2014
;
124
(
13
):
2034
-
2045
.
40.
Zhang
P
,
Lee
JS
,
Gartlan
KH
, et al
.
Eomesodermin promotes the development of type 1 regulatory T (TR1) cells
.
Sci Immunol
.
2017
;
2
(
10
).
41.
Du
J
,
Paz
K
,
Thangavelu
G
, et al
.
Invariant natural killer T cells ameliorate murine chronic GVHD by expanding donor regulatory T cells
.
Blood
.
2017
;
129
(
23
):
3121
-
3125
.
42.
Zhang
C
,
Todorov
I
,
Zhang
Z
, et al
.
Donor CD4+ T and B cells in transplants induce chronic graft-versus-host disease with autoimmune manifestations
.
Blood
.
2006
;
107
(
7
):
2993
-
3001
.
43.
McDonald-Hyman
C
,
Flynn
R
,
Panoskaltsis-Mortari
A
, et al
.
Therapeutic regulatory T-cell adoptive transfer ameliorates established murine chronic GVHD in a CXCR5-dependent manner
.
Blood
.
2016
;
128
(
7
):
1013
-
1017
.
44.
Le Huu
D
,
Matsushita
T
,
Jin
G
, et al
.
Donor-derived regulatory B cells are important for suppression of murine sclerodermatous chronic graft-versus-host disease
.
Blood
.
2013
;
121
(
16
):
3274
-
3283
.
45.
de Masson
A
,
Bouaziz
JD
,
Le Buanec
H
, et al
.
CD24(hi)CD27+ and plasmablast-like regulatory B cells in human chronic graft-versus-host disease
.
Blood
.
2015
;
125
(
11
):
1830
-
1839
.
46.
Sarantopoulos
S
,
Stevenson
KE
,
Kim
HT
, et al
.
Recovery of B-cell homeostasis after rituximab in chronic graft-versus-host disease
.
Blood
.
2011
;
117
(
7
):
2275
-
2283
.
47.
Weinberg
K
,
Blazar
BR
,
Wagner
JE
, et al
.
Factors affecting thymic function after allogeneic hematopoietic stem cell transplantation
.
Blood
.
2001
;
97
(
5
):
1458
-
1466
.
48.
Alho
AC
,
Kim
HT
,
Chammas
MJ
, et al
.
Unbalanced recovery of regulatory and effector T cells after allogeneic stem cell transplantation contributes to chronic GVHD
.
Blood
.
2016
;
127
(
5
):
646
-
657
.
49.
Johnston
HF
,
Xu
Y
,
Racine
JJ
, et al
.
Administration of anti-CD20 mAb is highly effective in preventing but ineffective in treating chronic graft-versus-host disease while preserving strong graft-versus-leukemia effects
.
Biol Blood Marrow Transplant
.
2014
;
20
(
8
):
1089
-
1103
.
50.
Malard
F
,
Labopin
M
,
Yakoub-Agha
I
, et al
.
Rituximab-based first-line treatment of cGVHD after allogeneic SCT: results of a phase 2 study
.
Blood
.
2017
;
130
(
20
):
2186
-
2195
.
51.
Cutler
C
,
Miklos
D
,
Kim
HT
, et al
.
Rituximab for steroid-refractory chronic graft-versus-host disease
.
Blood
.
2006
;
108
(
2
):
756
-
762
.
52.
Cutler
C
,
Kim
HT
,
Bindra
B
, et al
.
Rituximab prophylaxis prevents corticosteroid-requiring chronic GVHD after allogeneic peripheral blood stem cell transplantation: results of a phase 2 trial
.
Blood
.
2013
;
122
(
8
):
1510
-
1517
.
53.
Pomalidomide for chronic graft-versus-host disease
. https://clinicaltrials.gov/ct2/show/NCT01688466
. Accessed 1 February 2018
.
54.
Jourdan
M
,
Cren
M
,
Robert
N
, et al
.
IL-6 supports the generation of human long-lived plasma cells in combination with either APRIL or stromal cell-soluble factors
.
Leukemia
.
2014
;
28
(
8
):
1647
-
1656
.
55.
Tocilizumab for chronic graft-versus-host disease treatment
. https://clinicaltrials.gov/ct2/show/NCT02174263
. Accessed 1 February 2018
.
56.
Novobrantseva
TI
,
Majeau
GR
,
Amatucci
A
, et al
.
Attenuated liver fibrosis in the absence of B cells
.
J Clin Invest
.
2005
;
115
(
11
):
3072
-
3082
.
57.
Efficacy and tolerability of entospletinib in combination with systemic corticosteroids as first-line therapy in adults with chronic graft versus host disease (cGVHD). https://clinicaltrials.gov/ct2/show/NCT02701634. Accessed 1 February 2018
.
58.
Schutt
SD
,
Fu
J
,
Nguyen
H
, et al
.
Inhibition of BTK and ITK with Ibrutinib Is Effective in the Prevention of Chronic Graft-versus-Host Disease in Mice
.
PLoS One
.
2015
;
10
(
9
):
e0137641
.
59.
Miklos
D
,
Cutler
CS
,
Arora
M
, et al
.
Ibrutinib for chronic graft-versus-host disease after failure of prior therapy
.
Blood
.
2017
;
130
(
21
):
2243
-
2250
.
60.
Weiss
JM
,
Chen
W
,
Nyuydzefe
MS
, et al
.
ROCK2 signaling is required to induce a subset of T follicular helper cells through opposing effects on STATs in autoimmune settings
.
Sci Signal
.
2016
;
9
(
437
):
ra73
.
61.
Flynn
R
,
Paz
K
,
Du
J
, et al
.
Targeted Rho-associated kinase 2 inhibition suppresses murine and human chronic GVHD through a Stat3-dependent mechanism
.
Blood
.
2016
;
127
(
17
):
2144
-
2154
.
62.
A study to evaluate the safety, tolerability, and activity of KD025 in subjects with chronic graft versus host disease
. https://clinicaltrials.gov/ct2/show/NCT02841995
. Accessed 1 February 2018
.
63.
Rozovski
U
,
Wu
JY
,
Harris
DM
, et al
.
Stimulation of the B-cell receptor activates the JAK2/STAT3 signaling pathway in chronic lymphocytic leukemia cells
.
Blood
.
2014
;
123
(
24
):
3797
-
3802
.
64.
Zeiser
R
,
Burchert
A
,
Lengerke
C
, et al
.
Ruxolitinib in corticosteroid-refractory graft-versus-host disease after allogeneic stem cell transplantation: a multicenter survey
.
Leukemia
.
2015
;
29
(
10
):
2062
-
2068
.
65.
A study of ruxolitinib vs best available therapy (BAT) in patients with steroid-refractory chronic graft vs. host disease (GvHD) after bone marrow transplantation (REACH3). https://clinicaltrials.gov/ct2/show/NCT03112603. Accessed 1 February 2018
.
66.
Du
J
,
Paz
K
,
Flynn
R
, et al
.
Pirfenidone ameliorates murine chronic GVHD through inhibition of macrophage infiltration and TGF-β production
.
Blood
.
2017
;
129
(
18
):
2570
-
2580
.
67.
Zeiser
R
,
Nguyen
VH
,
Beilhack
A
, et al
.
Inhibition of CD4+CD25+ regulatory T-cell function by calcineurin-dependent interleukin-2 production
.
Blood
.
2006
;
108
(
1
):
390
-
399
.
68.
Koreth
J
,
Matsuoka
K
,
Kim
HT
, et al
.
Interleukin-2 and regulatory T cells in graft-versus-host disease
.
N Engl J Med
.
2011
;
365
(
22
):
2055
-
2066
.
69.
Matsuoka
K
,
Koreth
J
,
Kim
HT
, et al
.
Low-dose interleukin-2 therapy restores regulatory T cell homeostasis in patients with chronic graft-versus-host disease
.
Sci Transl Med
.
2013
;
5
(
179
):
179ra43
.
70.
Theil
A
,
Tuve
S
,
Oelschlägel
U
, et al
.
Adoptive transfer of allogeneic regulatory T cells into patients with chronic graft-versus-host disease
.
Cytotherapy
.
2015
;
17
(
4
):
473
-
486
.
71.
Baron
F
,
Labopin
M
,
Niederwieser
D
, et al
.
Impact of graft-versus-host disease after reduced-intensity conditioning allogeneic stem cell transplantation for acute myeloid leukemia: a report from the Acute Leukemia Working Party of the European group for blood and marrow transplantation
.
Leukemia
.
2012
;
26
(
12
):
2462
-
2468
.
72.
Lee
SJ
,
Wolff
D
,
Kitko
C
, et al
.
Measuring therapeutic response in chronic graft-versus-host disease. National Institutes of Health consensus development project on criteria for clinical trials in chronic graft-versus-host disease: IV. The 2014 Response Criteria Working Group report
.
Biol Blood Marrow Transplant
.
2015
;
21
(
6
):
984
-
999
.
73.
Paczesny
S
,
Hakim
FT
,
Pidala
J
, et al
.
National Institutes of Health Consensus Development Project on criteria for clinical trials in chronic graft-versus-host disease: III. The 2014 Biomarker Working Group report
.
Biol Blood Marrow Transplant
.
2015
;
21
(
5
):
780
-
792
.
74.
Phase I trial of brentuximab vedotin for refractory chronic graft-vs.-host disease (GVHD)
. https://clinicaltrials.gov/ct2/show/NCT01940796
. Accessed 1 February 2018
.
75.
Pai
CC
,
Chen
M
,
Mirsoian
A
, et al
.
Treatment of chronic graft-versus-host disease with bortezomib
.
Blood
.
2014
;
124
(
10
):
1677
-
1688
.
76.
Pai
CC
,
Hsiao
HH
,
Sun
K
, et al
.
Therapeutic benefit of bortezomib on acute graft-versus-host disease is tissue specific and is associated with interleukin-6 levels
.
Biol Blood Marrow Transplant
.
2014
;
20
(
12
):
1899
-
1904
.
77.
Carfilzomib in treating patients with chronic graft-versus-host disease
. https://clinicaltrials.gov/ct2/show/NCT02491359
. Accessed 1 February 2018
.
Sign in via your Institution