You are on page 1of 26

741860

research-article2017
TAH0010.1177/2040620717741860Therapeutic Advances in HematologyL Hill, A Alousi

Therapeutic Advances in Hematology Review

New and emerging therapies for acute and


Ther Adv Hematol

2018, Vol. 9(1) 21­–46

chronic graft versus host disease DOI: 10.1177/


https://doi.org/10.1177/2040620717741860
https://doi.org/10.1177/2040620717741860
2040620717741860

© The Author(s), 2017.


Reprints and permissions:
LaQuisa Hill, Amin Alousi, Partow Kebriaei, Rohtesh Mehta, Katayoun Rezvani http://www.sagepub.co.uk/
journalsPermissions.nav
and Elizabeth Shpall

Abstract: Graft versus host disease (GVHD) remains a major cause of morbidity and mortality
following allogeneic hematopoietic stem-cell transplantation (HSCT). Despite the use of
prophylactic GVHD regimens, a significant proportion of transplant recipients will develop
acute or chronic GVHD following HSCT. Corticosteroids are standard first-line therapy, but are
only effective in roughly half of all cases with ~50% of patients going on to develop steroid-
refractory disease, which increases the risk of nonrelapse mortality. While progress has
been made with improvements in survival outcomes over time, corticosteroids are associated
with significant toxicities, and many currently available salvage therapies are associated with
increased immunosuppression, infectious complications, and potential loss of the graft versus
leukemia (GVL) effect. Thus, there is an unmet need for development of newer treatment
strategies for both acute and chronic GVHD to improve long-term post-transplant outcomes
and quality of life for HSCT recipients. Here, we provide a concise review of major emerging
therapies currently being studied in the treatment of acute and chronic GVHD.

Keywords:  acute graft versus host disease, allogeneic stem-cell transplantation, chronic graft
versus host disease, GVHD

Received: 20 July 2017; revised manuscript accepted: 10 October 2017.

Introduction of grade III–IV acute GVHD between 1999 and Correspondence to:
Elizabeth Shpall
Allogeneic hematopoietic stem-cell transplanta- 2012.6 Overall survival (OS) and treatment- Department of Stem
tion (HSCT) is a potentially curative treatment related mortality (TRM) were also noted to have Cell Transplantation and
Cellular Therapy, The
for both benign and malignant hematologic con- improved during this time frame for patients University of Texas MD
ditions. Unfortunately, graft versus host disease treated with tacrolimus-based prophylactic regi- Anderson Cancer Center,
1515 Holcombe Boulevard,
(GVHD) remains a major cause of morbidity and mens. This is encouraging data for both clinicians Unit 0423, Houston,
mortality following HSCT. Despite prophylactic and HSCT patients, as despite increasing age of TX 77030-4000, USA
eshpall@mdanderson.org
treatment, acute GVHD (aGVHD) affects 30– transplant recipients, increasing use of alternative
LaQuisa Hill
70% of recipients and chronic GVHD (cGVHD) donors, and use of more reduced-intensity condi- Center for Cell and Gene
occurs in 20–50% of recipients depending on the tioning regimens over time, there has been Therapy, Baylor College
of Medicine, Houston,
type of transplant, patient characteristics, and improvement in transplant outcomes.7,8 As we TX, USA
GVHD prophylaxis regimen.1–5 GVHD is a sys- continue to investigate potentially more effective Amin Alousi
Partow Kebriaei
temic inflammatory condition primarily mediated preventive and treatment strategies, hopefully we Rohtesh Mehta
by the transplanted immune system that can lead can continue to make a meaningful impact on Katayoun Rezvani
Department of Stem
to severe multiorgan damage. The need for transplant outcomes. Cell Transplantation
increased and prolonged immunosuppression to and Cellular Therapy,
The University of Texas
treat GVHD, in addition to the immunosuppres- We briefly discuss current knowledge of emerging MD Anderson Cancer,
sive effects of the disease itself, increases the risk mechanistic targets for treatment of aGVHD and Houston, TX, USA
of: infection, organ impairment, poor quality of cGVHD, and novel therapies that are showing
life and ultimately, mortality. promising efficacy in both upfront and steroid-
refractory (SR) settings. However, most of these
A large registry analysis by Khoury and colleagues agents are still in early-phase clinical studies and
recently reported a 20% decline in the proportion have yet to be evaluated in large, late-phase

journals.sagepub.com/home/tah 21
Therapeutic Advances in Hematology 9(1)

randomized controlled trials. Many of these The risk score was recently refined, based on vali-
agents are also being investigated in preventive dated data from a multicenter cohort totaling 1723
trials; however, that is beyond the scope of this patients. Standard risk was defined as single-organ
review. involvement [stage 1–3 skin or stage 1–2 gastroin-
testinal (GI)] or two-organ involvement (stage 1–3
skin plus stage 1 GI or stage 1–3 skin plus stage
Acute graft versus host disease 1–4 liver); all others were defined as high risk
aGVHD is an immunologically mediated process (Table 1). Standard-risk patients had a day 28
due to mature donor T cells interacting with host overall response rate (ORR) rate of 69% versus
and donor antigen presenting cells (APCs), lead- 43% in high-risk patients (Table 2). The 6-month
ing to release of pro-inflammatory cytokines, NRM was 22% versus 44%, respectively.
which in turn results in the proliferation and hom-
ing of activated T cells to aGVHD target tissues, Levine and colleagues also recently validated the
ultimately causing host tissue damage.9–11 Recent Ann Arbor biomarker risk score based on plasma
comprehensive reviews of aGVHD biology have levels of tumor necrosis factor receptor-1 (TNFR-
be performed by Magenau and colleagues12 and 1), regenerating islet-derived 3-alpha (REG3α)
Zeiser and colleagues,13 and are beyond the scope and suppression of tumorigenicity 2 (ST2).22 A
of this review. A number of major histocompati- risk score of 3 demonstrated a 46% ORR com-
bility complex (MHC)-independent mechanisms pared with AA1/AA2 patients who had an 81%
have recently been implicated in GVHD patho- and 68% response rate, respectively. A risk score
genesis based on findings in murine models. These of 3 was also associated with a less durable
will be discussed in more detail below. response (defined as a complete response for ⩾6
months without recurrence of symptoms), higher
Corticosteroids (steroids) remain the first-line of likelihood of later developing SR lower GI GVHD
therapy for both aGVHD and cGVHD despite and had an NRM of 46% at 6 months regardless
suboptimal response rates of 40–60%.14–16 The of clinical severity of GVHD (Table 2). By utiliz-
likelihood of response to treatment in aGVHD ing new therapies based on risk stratification
decreases with increasing severity of disease.2 For models, we may be able to improve outcomes,
patients who develop SR disease, the long-term while minimizing toxicity.
prognosis is very poor, with a mortality rate of
approximately 70–80%,14,17 as response rates
with second-line treatments are low.2,18,19 To Steroid-free acute graft versus host disease
date, no second-line therapy has been proven therapy
superior to another20 for the treatment of
SR-aGVHD, and choice of therapy is often based Sirolimus
on patient specific characteristics, side-effect pro- Sirolimus (also known as rapamycin) is a mam-
file, and physician preference. malian target of rapamycin (mTOR) inhibitor
that has immunosuppressive activity in addition
Such suboptimal outcomes underscore the need to its antineoplastic properties.23 Inhibition of
for new treatment strategies. One newly proposed mTOR impairs T-cell signaling, and its use as a
treatment paradigm is risk stratifying patients for prophylactic agent and as a second-line agent for
treatment based on clinical staging of aGVHD SR-GVHD have been previously reported.24–27
and blood biomarkers. This has been proposed in However, a recent retrospective analysis evalu-
an attempt to spare those likely to respond to ster- ated the use of sirolimus monotherapy as upfront
oids from excessive toxicity, and to identify those treatment in 32 patients with newly diagnosed
who are less likely to respond and require aggres- aGVHD who were poor candidates for steroids
sive upfront therapy to improve nonrelapse mor- due to older age, high risk of toxicity, or at high
tality (NRM). The aGVHD risk score, which risk for relapse.28 A total of 27 patients were iden-
classifies patients into high risk or standard risk tified as Minnesota standard risk and five were
categories at the time of diagnosis, was developed high risk. Of note, no patients with grade IV
by the Minnesota group to identify patients aGVHD were included in this cohort. A total of
unlikely to respond to upfront treatment with 16 (50%) patients achieved a complete response
steroids.21 Patients with high risk (HR) GVHD (CR) with sirolimus versus 59% of patients from a
were less likely to respond to therapy and had a matched cohort treated with steroids. These data
twofold increased risk of TRM. indicate that use of sirolimus as a steroid-free

22 journals.sagepub.com/home/tah
Table 1. Graft versus host disease organ staging categories and graft versus host disease risk.16

GVHD categories grouped by day n Day 28 OR of day p value Relative risk p value Relative p value GVHD
28 CR/PR CR/PR 28 CR/PR of mortality risk of TRM risk
(95% CI) (95% CI) (95% CI)
Stage 1–3 skin only** 901 68% 1.0 1.0 1.0 StdR
Upper GI only 115 78% 1.6 (1.0–2.6) 0.04 0.6 (0.4–0.9) 0.02 0.5 (0.3–0.9) 0.03 StdR
Stage 1–2 lower GI only 100 73% 1.3 (0.8–2.1) 0.29 1.2 (0.8–1.7) 0.41 1.6 (1.0–2.3) 0.04 StdR
Stage 1–3 skin + upper GI 90 69% 1.0 (0.6–1.6) 0.89 0.9 (0.6–1.4) 0.64 0.9 (0.6–1.5) 0.71 StdR
Stage 1–3 skin + stage 1 lower GI 71 61% 0.7 (0.4–1.2) 0.16 1.1 (0.7–1.8) 0.60 1.3 (0.8–2.1) 0.35 StdR
Stage 1 lower GI + upper GI 64 64% 0.8 (0.5–1.4) 0.42 1.2 (0.7–1.9) 0.54 1.4 (0.9–2.4) 0.18 StdR
Stage 1–3 skin + stage 1–4 liver 51 71% 1.1 (0.6–2.1) 0.71 1.3 (0.8–2.0) 0.30 1.4 (0.9–2.3) 0.17 StdR
Stage 1–3 skin + stage 1 lower 62 61% 0.6 (0.4–0.9) 0.12 0.6 (0.3–1.1) 0.11 0.6 (0.3–1.3) 0.18 StdR
GI + upper GI
Stage 1–2 lower GI + stage 1–3 12 50% 0.4 (0.1–1.4) 0.15 2.9 (1.4–6.3) 0.005 3.2 (1.5–7.0) 0.003 HR
liver
Stage 1–3 skin + (stage 1–2 lower 23 35% 0.2 (0.1–0.6) 0.001 3.0 (1.7–5.1) <0.001 3.2 (1.8–5.6) <0.001 HR
GI or upper GI) + stage 1–3 liver
Stage 3 lower GI only 65 55% 0.5 (0.3–0.9) 0.02 2.1 (1.4–3.1) 0.003 2.7 (1.8–4.2) <0.001 HR
Stage 1–3 skin + stage 2 lower GI 54 52% 0.5 (0.3–0.9) 0.01 1.5 (1.0–2.3) 0.08 1.6 (1.0–2.6) 0.06 HR
Stage 3–4 lower GI + (stage 1–3 55 36% 0.2 (0.1–0.4) <0.001 2.5 (1.7–3.6) <0.001 3.0 (2.0–4.5) <0.001 HR
skin or liver stage 1–4)
Stage 1–4 liver alone 25 48% 0.3 (0.2–0.8) 0.01 1.0 (0.5–2.3) 0.96 1.7 (0.8–3.8) 0.19 HR
Stage 1–3 skin + stage 3–4 lower 13 8% 0.1 (0.01–0.3) 0.002 4.3 (2.3–8.2) <0.001 7.4 (3.6–15.2) <0.001 HR
GI + stage 1–4 liver
Stage 4 skin only 13 38% 0.3 (0.1–0.8) 0.02 0.8 (0.2–3.1) 0.71 2.2 (0.7–6.9) 0.16 HR
Stage 4 lower GI only 22 22% 0.1 (0.03–0.7) 0.02 3.5 (1.5–7.9) 0.003 3.1 (1.2–8.1) 0.02 HR
GVHD, graft versus host disease; CR, complete response; PR, partial response; CI, confidence interval; TRM, treatment-related mortality; GI, gastrointestinal; StdR, standard risk; HR,
high risk.

journals.sagepub.com/home/tah 23
L Hill, A Alousi et al.
Therapeutic Advances in Hematology 9(1)

Table 2.  Day 28 response rate and 6-month nonrelapse mortality by risk scoring systems.
Refined Day 28 RR 6-month NRM n
Minnesota aGVHD
Risk Score CR PR % 95% CI

StdR 48% 21% 22 20–24 1454


HR 27% 16% 44 38–50 269
Ann Arbor GVHD CR/PR
Risk Score
1 81%  8  3–16 74
2 68% 27 20–34 165
3 46% 46 33–58 61
aGVHD, acute graft versus host disease; CR, complete response; CI, confidence interval; HR, high risk; NRM, nonrelapse
mortality; PR, partial response; RR, response rate; StdR, standard risk.

primary therapy would potentially be safe and JAK inhibition impairs the differentiation and
effective for aGVHD. The Bone Marrow function of dendritic cells, the most important
Transplant Clinical Trials Network (BMT CTN) APCs, causing reduced T-cell activation.34,35
1501 trial is a phase II trial investigating the use of These data support the potential role of JAK inhi-
single-agent sirolimus versus prednisone for front- bition for treatment of GVHD.
line treatment of standard-risk aGVHD by refined
Minnesota risk criteria, with further confirmation Ruxolitinib.  Ruxolitinib is a JAK 1/2 inhibitor that
based on the Ann Arbor biomarker risk score has been shown to have a significant role in signal-
[ClinicalTrials.gov identifier: NCT02806947]. ing pathways mediating inflammation in acute and
chronic GVHD,36 in addition to roles in adaptive
and innate immune responses. It has been shown
Kinase inhibitors that interferon gamma receptor (INFγR) signaling
is mediated via JAK1/JAK2 and is upregulated in
Janus kinases activated Tcells.37 T cells deficient in INFγR cause
The Janus kinase (JAK) family is composed of significantly less GVHD compared with wild-type
four tyrosine kinases that function as signal trans- T cells, and pharmacologic inhibition of JAK1/2
ducers of cytokine-mediated pathways to control in mouse models leads to less GVHD, in addition
cellular proliferation, survival and differentia- to preserving anti-leukemic effects previously
tion.29 JAKs associate with a variety of cytokine reported.37,38 Spoerl and colleagues showed that
receptors, and activation of JAK leads to phos- JAK 1/2 inhibition led to reduction of pro-inflam-
phorylation of signal proteins of the signal trans- matory cytokines and an increase in the frequency
ducers and activators of transcription (STAT) of regulatory T cells (Tregs) in mouse models with
family, which act as transcription factors for pro- improved survival in mice who developed acute
inflammatory genes.30 JAK1 and JAK2 are both GVHD and reduced histopathological grading.30
widely expressed, and their inactivation leads to Based on this observation, they trialled six patients
impairment of immune function via cytokine- with heavily treated SR-aGVHD with ruxolitinib.
induced intracellular signaling of lymphocytes.31 Two patients with GI GVHD had complete resolu-
tion of symptoms during treatment, one patient
While inhibition of JAK1/2 (nonspecific inhibi- with liver GVHD had complete normalization of
tion) significantly reduces T-cell function, recent bilirubin, and all four patients with skin GVHD
evidence suggests that specific inhibition of JAK1 had a response to ruxolitinib therapy. All patients
may be the primary mediator of response.32 More also had a reduction in serum levels of pro-inflam-
specific targeting may ameliorate off-target effects matory cytokines [interleukin-6 (IL-6) and IL-2R],
such as myelosuppression which is primarily and the amount of corticosteroids required.
mediated by inhibition of JAK2-associated recep-
tors required for hematopoietic cell development In a multicenter survey study done in centers
and proliferation.33 There is also evidence that throughout Europe and the US, 54 patients with

24 journals.sagepub.com/home/tah
L Hill, A Alousi et al.

Table 3.  Clinical trials of tyrosine kinase inhibitors (registered through ClinicalTrials.gov).

Trial number Study design Diagnosis Intervention Location Status


Ruxolitinib  
NCT02953678 Phase II SR-aGVHD Ruxolitinib + USA Recruiting
CCS
NCT03147742 Open label, SR-aGVHD / Ruxolitinib USA Recruiting
expanded access SR-cGVHD
NCT03112603 Phase III SR-cGVHD Ruxolitinib USA Not yet
versus BAT recruiting
NCT02997280 Phase II SR-aGVHD / Ruxolitinib Russia Recruiting
SR-cGVHD
NCT02396628 Phase II SR-aGVHD BAT Germany Recruiting
+/–ruxolitinib
NCT02913261 Phase III SR-aGVHD Ruxolitinib Europe, Asia, Recruiting
versus BAT Australia, Canada
SR, steroid refractory; aGVHD, acute graft versus host disease; cGVHD, chronic graft versus host disease; BAT, best
available therapy; CCS, corticosteroids.

SR-aGVHD (grade III or IV) were treated with off- placebo in combination with steroids is currently
label use of ruxolitinib39 at a dose of 5–10 mg orally being planned through the BMT CTN.
twice daily. The ORR was 81.5% (44/54) with 25
(46.3%) patients achieving a CR. The median time
to response was 1.5 (range 1–11) weeks, and only 3 Proteasome inhibitors
patients experienced a GVHD-relapse, suggesting Proteasome inhibitors (PIs) are drugs that inhibit
durable response was achievable. The 6-month sur- the proteasome, which are enzyme complexes
vival was 79%. Cytomegalovirus (CMV) reactiva- that are responsible for regulating protein turno-
tion and cytopenias were the most common adverse ver within cells. They have been shown to have
events (AEs) observed. CMV reactivation occurred immune-modulating effects in murine models of
in 33% of patients, all of which responded to antivi- aGVHD and cGVHD, including deletion of allo-
ral treatment. Grade III/IV cytopenias, a previously reactive T cells, inhibition of APCs, inhibition of
established side effect of ruxolitinib, occurred in IL-6, increased survival of Tregs, and decrease in
33% of patients. Several phase II and III trials are levels of B-cell activating factor (BAFF).41 Several
currently ongoing for SR-aGVHD (Table 3). PIs have been US Food and Drug Administration
(FDA) approved for the treatment of multiple
Itacitinib.  Results from a phase 1 trial of selective myeloma.
JAK1 inhibitor itacitinib (INCB039110) in
patients with aGVHD were reported at the 2016
American Society of Hematology Meeting Bortezomib
(ASH).40 Patients with grades II–IV aGVHD Bortezomib is a first-generation, reversible PI that
were randomized 1:1 to a dose of 200 or 300 mg directly induces apoptosis of tumor cells by sup-
orally once daily of INCB039110 combined with pressing tumor-survival pathways and arresting
steroids, and were stratified based on treatment- cell growth.42 It is a potent inhibitor of nuclear
naïve (n = 14) versus SR (n = 17) disease. Pre- factor-kappa B, which regulates the transcription
liminary results showed a day 28 ORR of 88.3% of multiple pro-inflammatory genes, and is impor-
when used in the first-line setting as treatment for tant for T-cell proliferation, activation and
aGVHD, and 64.7% for SR disease. GRAVI- survival.43
TAS-301 is a phase III trial currently recruiting
for front-line treatment of aGVHD with itacitinib Several preclinical and early-phase studies have
versus placebo in combination with steroids [Clin- shown bortezomib to be effective in preventing
icalTrials.gov identifier: NCT03139604]. A phase aGVHD,44–47 although in one murine study,
II trial of upfront use of a JAK1 inhibitor or delayed or prolonged administration was

journals.sagepub.com/home/tah 25
Therapeutic Advances in Hematology 9(1)

associated with severe colonic toxicity.45 Given is a phase II/III study for treatment of newly diag-
the critical role of T cells and pro-inflamma- nosed LGI aGVHD in combination with steroids
tory cytokines in the development of aGVHD, [ClinicalTrials.gov identifier: NCT02956122],
bortezomib would be a potential therapeutic and the other is an early-access trial for patients
candidate. with SR-aGVHD [ClinicalTrials.gov identifier:
NCT03172455]. A pilot study for use of ATT in
Results of a small phase II study [ClinicalTrials. SR-aGVHD has completed enrollment, but
gov identifier: NCT00408928] evaluating the use results are not yet available [ClinicalTrials.gov
of bortezomib for SR-aGVHD, the largest to date, identifier: NCT01700036].
were recently reported by Wagner and col-
leagues.48 Eleven patients were treated on study
with eight patients being evaluable for response. Interleukin-22
Two patients (25%) achieved a CR, and two IL-22 is a cytokine produced by cells from both
patients had a partial response (PR). However, 10 the adaptive and the innate system including
patients died, with the majority of deaths attrib- CD4+ T cells, CD8+ T cells, natural killer cells,
uted to infection (60%), followed by progressive and gamma-delta (γδ) T cells.53 It serves an essen-
disease (20%). One patient also died of hemor- tial role in host defense against extracellular path-
rhage and pulmonary complication, respectively. ogens, strengthens epithelial barrier function by
The authors were unable to determine if any of acting upon intestinal stem cells (ISCs), and helps
the deaths were related to the use of bortezomib. with tissue repair and wound healing. In addition
While bortezomib appears to have some activity in to its protective properties, it has pro-inflamma-
treating aGVHD, additional studies are needed to tory properties when produced in excess.53,54
better determine the safety and toxicity profile. Studies of murine models transplanted with
IL-22-deficient grafts showed loss of ISCs, devel-
opment of more severe aGVHD and increased
Cytokine modulation mortality compared with wild-type models.55,56
Lindemans and colleagues showed that treatment
Alpha-1 antitrypsin of murine models with IL-22 in vivo reduced
Alpha-1 antitrypsin (AAT) is a serine protease intestinal pathology and mortality from GVHD.57
inhibitor that is able to modulate immune and There is also a potential relationship between
inflammatory functions through alteration of IL-22 and the microbiome, as microbacteria has
cytokine profiles.49 Low AAT plasma levels in been shown to induce IL-22 secretion that then
human donors were found to be associated with a induces the production of antimicrobial pro-
higher rate of aGVHD in recipients50 and GVHD teins.57–59 There is currently a phase I/II clinical
severity increased with decreasing AAT levels as trial underway [ClinicalTrials.gov identifier:
well. An inverse relationship between AAT levels NCT02406651] evaluating the safety and feasi-
and GVHD was noted, with high levels of ATT bility of use of IL-22 in combination with corti-
exerting a protective effect against GVHD while costeroids for the treatment of newly diagnosed
maintaining, or even enhancing, graft versus leu- grade II–IV acute LGI GVHD.
kemia (GVL) activity50 and decreasing mortality
in mouse models.51 Donors treated with ATT
had an increase in Tregs, as well as a 50-fold Monoclonal antibodies
increase of IL-10, favoring an anti-inflammatory
profile. Natalizumab
Natalizumab is a humanized monoclonal anti-
Based on these observations, a prospective phase body (mAb) against α4-integrin containing adhe-
I/II dose-escalation study was performed to eval- sion molecules which are widely expressed on
uate the use of ATT in the treatment of leukocytes, primarily lymphocytes.60 Based on
SR-aGVHD. Twelve patients were enrolled in studies done in inflammatory bowel disease
the first two cohorts (all grade III or IV GVHD (IBD), it has been recognized that the endothe-
with stage 3–4 lower gastrointestinal (LGI) lium plays a key role in the pathogenesis of inflam-
involvement), and eight patients achieved a mation and in mucosal immune trafficking.61
response, with four patients achieving a CR.52 Natalizumab inhibits adhesion molecules, pre-
There are currently two clinical trials underway to venting leukocyte migration from the circulation
further evaluate the use of ATT for GVHD. One into inflamed gut mucosa.60,62 It is currently FDA

26 journals.sagepub.com/home/tah
L Hill, A Alousi et al.

approved for use in multiple sclerosis and Crohn’s clinical efficacy will need to be confirmed in phase
disease. Despite its proven efficacy in these dis- II/III trials. A multicenter phase II study recently
eases, use has been limited due to the possible opened and is recruiting [ClinicalTrials.gov iden-
side effect of progressive multifocal leukoenceph- tifier: NCT02993783].
alopathy (PML) – a potentially life-threatening
demyelinating neurologic disease. There has been
a reported incidence of 4.2 cases per 1000 patients Brentuximab vedotin
treated.63,64 Brentuximab vedotin (BV) is an anti-CD30 anti-
body–drug conjugate composed of the antimicro-
Acute GI GVHD is often propagated after intes- tubule agent monomethyl auristatin E (MMAE)
tinal damage from transplant-conditioning regi- and a human CD30 antibody. It has been
mens, leading to significant inflammation in the approved for the treatment of relapsed classical
gut lining. Therefore, it would seem plausible Hodgkin lymphoma, systemic anaplastic large
that natalizumab might be able to mitigate GI cell lymphoma and for postautologous HSCT
GVHD by preventing homing of leukocytes to the consolidation of those CD30+ diseases. CD30 is
GI tract. Patients are currently being recruited for a tumor necrosis factor receptor family member
two phase II studies evaluating the safety and effi- and is highly expressed on activated lymphocytes
cacy of natalizumab for treatment of GVHD. One with minimal expression on normal tissues.71
study is evaluating its use in combination with Infiltrating lymphocytes in chronic autoimmune
steroids for initial treatment of acute GI GVHD and inflammatory diseases such as rheumatoid
[ClinicalTrials.gov identifier: NCT02176031]. arthritis and systemic sclerosis have also been
The second study is evaluating the combination shown to express high levels of CD30.72,73 In pre-
for treatment of high-risk aGVHD (based on Ann clinical studies, Chen and colleagues demon-
Arbor risk score) with the primary outcome being strated that patients with aGVHD had a higher
rate of CR at day 28 [ClinicalTrials.gov identifier: percentage of CD30 expressing CD8+ T cells,
NCT02133924]. significantly higher plasma levels of soluble
CD30, and increased CD30+ lymphocytes in
affected intestinal tissue compared with patients
Vedolizumab without aGVHD.74
Vedolizumab is a mAb that specifically inhibits
α4β7 integrins located on activated T cells from Based on these results, a multicenter phase I trial
interacting with MAdCAM-1 (located on gut epi- was initiated to investigate the use of BV for the
thelium) preventing homing to the intestinal treatment of SR-aGVHD.75 Thirty-four patients
mucosa.65,66 It is currently FDA approved for the were treated in the dose-escalating study to deter-
treatment of Crohn’s disease and ulcerative coli- mine maximum tolerated dose (MTD). The
tis. Several preclinical murine studies have shown study initially had a schedule of weekly dosing for
that α4β7 integrins play an important role in the 3 weeks followed by maintenance dosing every 3
development of intestinal GVHD.67–69 A recent weeks for four additional doses. However, after
case series describing the off-label use of vedoli- treatment of six patients with death of two patients
zumab for treatment of SR-aGVHD of the GI from neutropenic sepsis, the study was revised to
tract showed promising results.70 Five patients dosing every 2 weeks for four doses with cohorts
with clinical grade IV GI GVHD were treated; of five patients per dose level (0.6 mg/kg, 1.0 mg/
four patients had isolated GI manifestations and kg, and 1.2 mg/kg). The dose-limiting toxicity
one patient had multiorgan involvement. (DLT) was defined at 0.8 mg/kg after one patient
developed sepsis and multiorgan failure. Grade
Four patients were able to discontinue systemic III toxicities included neutropenia, headache,
steroids after receiving three doses of vedoli- hypoxia, ileus and elevated bilirubin. No periph-
zumab, and two patients were off all immunosup- eral neuropathy of any grade was observed.
pressant medications at the time of last follow up.
Clinical responses were noted within 7–10 days The day 28 ORR was 38.2% with five (14.7%)
with patients receiving a variable number of infu- patients achieving a CR and eight (23.5%)
sions.70 The median time to follow up was 10 patients achieving a very good PR (VGPR). At
months, with four patients alive and receiving day 56, seven additional patients had achieved
outpatient care. No associated toxicities were CR; three of whom had previously achieved
mentioned. Safety and toxicity, in addition to VGPR. Responses were not significantly different

journals.sagepub.com/home/tah 27
Therapeutic Advances in Hematology 9(1)

for individual organ involvement: intestinal (n = improved response rates (76% versus 47% and
10; 42%), liver (n = 5; 45%), and skin (n = 5; 82% versus 68%, respectively; p = 0.03 for both).
28%), however the study was not designed to
assess efficacy. The OS at 6 and 12 months follow Hashmi and colleagues performed a systematic
up was 41% and 38%, respectively. Of those review and meta-analysis to assess response and
patients (n = 16) who did not respond to BV, survival in patients with SR-aGVHD treated with
only one remained alive at 12 months. Causes of MSCs.88 Thirteen nonrandomized studies includ-
death in the other patients were: GVHD (n = ing 336 patients were included, and six studies (n
11), infection (n = 3), and disease relapse (n = = 119) provided data on primary outcome of
1). Larger randomized trials are needed to con- 6-month survival following treatment with MSCs.
firm efficacy. The weighted 6-month survival was 63% [confi-
dence interval (CI) 50–74%] compared with the
Multiple mAbs targeting different pathways in weighted average of 49% from 29 non-MSC
the pathophysiology of aGVHD have been evalu- studies of American Society of Blood and Marrow
ated in the past. The responses and toxicities of Transplantation (ASBMT) that were used to for-
several of the older agents compared with newer mulate guidelines for recommended second-line
aAbs are summarized in Table 6. However, cross therapies. However, this must be interpreted with
comparison of these studies is limited due to caution as there were many limitations of the
multiple factors, including differences in study study, including differences in timing of adminis-
design (the majority of reports are retrospective tration of MSCs, heterogeneity of study popula-
series), varying definitions of SR-aGVHD, tions, variable definitions of SR disease, and
heterogeneous patient populations, variation in variability in the dose and quality of the MSC
time-to-response assessments, and use of succes- product. Thus, direct comparison of MSCs with
sive salvage agents. other second-line agents in prospective, rand-
omized trials are needed to reach a definitive con-
clusion on survival advantage. There are currently
Adoptive cell therapy four studies for upfront use and nine studies for
SR-aGVHD underway utilizing MSCs for treat-
Mesenchymal stromal cells ment of aGVHD (Table 5).
Mesenchymal stromal cells (MSCs) are pluripo-
tent stem cells that are able to differentiate into
multiple cell lineages of mesenchymal origin Microbiome restoration
including osteoblast, chondrocytes, and adipo-
cytes.76–79 They have been shown to inhibit B- Fecal microbiota transplantation
and T-cell activation, block the function of APCs, Alteration of intestinal homeostasis, via disrup-
inhibit natural killer cells, and increase tion and damage to the gastrointestinal epithe-
Tregs.78,80,81 MSCs can be isolated and expanded lium, has emerged as having a critical role in the
ex vivo from bone marrow, umbilical cord blood, development and severity of acute GVHD involv-
adipose tissue, and placenta.76,78,82 MSCs do not ing the GI tract.89,90 Dysbiosis, or loss of intesti-
express class II human histocompatibility leuko- nal diversity, has been associated with increased
cyte antigens (HLA) and therefore do not pro- TRM.91–93 Obligate anaerobes, particularly mem-
voke immunologic responses in HLA-mismatched bers of the Clostridiales order, have been identi-
recipients. fied as important mediators of intestinal
homeostasis by upregulation of intestinal Tregs
MSC therapies have been studied in phase I and and inhibition of inflammation by producing anti-
II trials for the treatment of SR-aGVHD with inflammatory cytokines.94 Increased abundance
overall responses ranging from 60–75%.83–86 of intestinal Blautia (a member of the Clostridia
Results from two multicenter, randomized phase class) has been associated with reduced GVHD
III trials led by Osiris Therapeutic were reported, mortality and improved OS.95
in abstract form only, on the use of MSCs for
treatment of de novo aGVHD and SR-aGVHD.82,87 Two recent small case series utilized fecal micro-
Neither trial reached its primary endpoint of biota transplants (FMTs) in an attempt to
durable CR ⩾28 days. However, patients with SR restore normal intestinal flora. The first study
liver and GI GVHD treated with Prochymal, the evaluated the safety and efficacy of FMT in
Osiris product, were reported to have significantly patients with SR or steroid-dependent aGVHD.

28 journals.sagepub.com/home/tah
L Hill, A Alousi et al.

Out of a total of four patients, three had achieved Chronic GVHD is a multiorgan disease associ-
complete resolution of symptoms at day 28 ated with significant immunodeficiency which
GVHD assessment, and one had a PR.96 In a makes treatment with immunosuppressive medi-
study evaluating three patients with SR-acute GI cations challenging due to the increased risk of
GVHD who received FMT on a compassionate severe, life-threatening infections.3–5 As previ-
use basis; two patients had resolution of all signs ously mentioned, steroids are the most widely
and symptoms of GVHD and one patient had used first-line therapy for treatment of moderate-
partial resolution.97 No significant severe AEs, to-severe cGVHD with or without the addition of
including infection, were attributed to the proce- other immunosuppressive agents.4 If patients fail
dure in either study. In the latter study, all three to respond or have progressive disease with ster-
patients ultimately died from non-GVHD causes oid therapy, second-line treatment is required.
with duration of response lasting 8–9 weeks at The response rate to second-line therapy is
time of death. Early-phase trials are currently in reported to be 25–50% based primarily on phase
development at MD Anderson Cancer Center II trials, with no single therapy being better than
and Memorial Sloan Kettering to determine if any other.104,105 Choice of therapy is generally
these results are reproducible. based on patient comorbidities, organs involved,
and physician experience.

Chronic graft versus host disease


While aGVHD is driven mainly by mature donor T Kinase inhibitors
cells, cGVHD has been discovered to involve a
more complex immune reaction with both T and B Janus kinase inhibitors
cells contributing to the underlying pathology. In addition to mechanisms previously elucidated
Although donor antibodies to recipient antigens are above, it has been demonstrated that patients
known to play a role in cGVHD, the exact mecha- with cGVHD have upregulated IFN-inducible
nisms of how these cells contribute to the underly- genes and elevated levels of INF-induced
ing pathology are still being investigated. BAFF has chemokines which are involved in CXCR3+-
been identified as a key regulator of B-cell homeo- mediated lymphocyte trafficking.106 JAK1/JAK2
stasis, and high levels have been shown to rescue are mediators of INFγR signaling and inhibition
self-reactive B cells from peripheral deletion,98 pro- of the JAK pathway results in decreased expres-
moting survival and differentiation. Sarantopoulos sion of CXCR3, reduced GVHD, and improved
and colleagues showed that patients with active survival in murine models.37 Data from preclini-
cGVHD had significantly elevated levels of BAFF cal studies and observational human studies have
and increased signaling through the ERK and AKT identified JAK-STAT inhibition as an exciting
pathways (both of which are activated via BAFF) therapeutic target.
which was associated with decreased apoptosis of
activated B cells.99,100 Ruxolitinib. In the previously mentioned multi-
center survey study, 41 patients with moderate-
In addition, distinct subsets of B cells known as to-severe SR-cGVHD were also included. The
regulatory B cells (Bregs) have been identified101 ORR was 85.4% with 32 (78%) patients achiev-
to have immunosuppressive effects via IL-10 pro- ing a PR and three patients achieving a CR. The
duction that inhibit CD4+ T-cell proliferation median time to response was 3 (range 1–25)
and cytokine production.102 Sarvaria and col- weeks, with responses noted in all involved organ
leagues demonstrated that patients with cGVHD systems. Only two patients (5.7%) experienced a
had reduced levels of circulating Bregs and GVHD relapse, and 14.6% (6/41) of patients had
impaired IL-10 production compared with no response. CMV reactivation was and cytope-
patients without cGVHD102,103 suggesting Breg nias were observed in 17% and 14.6% of patients,
therapy might be a potential therapeutic target. respectively. However, caution must be taken
In-depth reviews of cGVHD biology have been when interpreting these results due to potential
recently published by Im and colleagues41 and recall bias, as the reported response rates were
Reddy and colleagues.12 A better understanding based on retrospective chart review. Evaluation
of potential mechanisms contributing to the and assessment of responses in cGVHD, based on
pathophysiology of cGVHD has led to develop- the National Institutes of Health (NIH) consen-
ment of targeted therapies that will be discussed sus criteria,107 requires extensive examination and
below. detailed documentation in order to capture

journals.sagepub.com/home/tah 29
Therapeutic Advances in Hematology 9(1)

accurate responses. It often takes 4–6 weeks to Ibrutinib was recently granted FDA approval,
note objective improvement in cGVHD following the first drug to ever be approved for treatment
initiation of therapy, although they reported of cGVHD, based on results from a multicenter
observing improvements as early as 1 week after phase II study evaluating the safety and efficacy
treatment was started. of its use in patients with cGVHD who had failed
one to three lines of prior systemic therapy.113
Larger, prospective randomized controlled trials Forty-two patients with steroid-dependent or
are needed to determine the true response rate -resistant cGVHD were treated with a daily dose
based on standardized grading of responses, and of 420 mg of ibrutinib. The ORR was 67%
to determine the optimal dosing schedule and (28/42 patients; 21% CR; 45% PR). A total of
duration of treatment required for long-term con- 71% of responders had a sustained response of
trol of GVHD. Several phase II and III trials are at least 20 weeks. Approximately one third of
currently ongoing (Table 3). patients who responded were able to reduce the
dose of steroids to ⩽0.15 mg/kg/day during the
Baricitinib.  Baricitinib is an oral JAK1/2 inhibitor course of the study, and five patients were able
developed for the treatment of rheumatoid arthri- to completely discontinue steroid therapy.
tis (RA), atopic dermatitis, and systemic lupus Improvement occurred in multiple organ sys-
erythematosus (SLE). It received its first global tems, with 42% (5/12) of patients with at least
approval in February 2017 for refractory RA in three involved showing a response in at least
the European Union. There is a phase I/II three organs, which were associated with
study underway evaluating its use in the treat- improvement in quality of life.
ment of SR-cGVHD [ClinicalTrials.gov identi-
fier: NCT02759731]. Importantly, the drug was not without toxicity,
with the most common AEs observed being
fatigue (57%), diarrhea (36%), muscle spasms
Ibrutinib (29%), nausea (26%) and bruising (24%).
Ibrutinib is an irreversible inhibitor of Bruton’s Serious AEs occurred in 22 patients (52%) with
tyrosine kinase (BTK) and IL-2 inducible T-cell ⩾grade III occurring in 17 patients (40%). Two
kinase (ITK). BTK is primarily responsible for deaths were reported due to multilobular pneu-
the activation of signaling pathways involved in monia and bronchopulmonary aspergillosis.
B-cell proliferation, trafficking and adhesion, There is currently a phase III trial [ClinicalTrials.
while ITK is involved in the secretion of IL-2 and gov identifier: NCT02959944] underway evalu-
T-helper 2 cytokines. While the exact involve- ating the efficacy of corticosteroids plus ibruti-
ment of BTK and ITK in the pathogenesis of nib versus placebo for the frontline treatment of
cGVHD is not known, given the role of T- and new-onset moderate-to-severe cGVHD.
B-cell dysregulation in the development of
cGVHD,108–110 there have been investigations
into targeting the B-cell receptor (BCR) and Spleen tyrosine kinase inhibitors
T-cell receptor (TCR) signaling pathways. Spleen tyrosine kinase (Syk), predominantly
expressed in hematopoietic cells, has recently
In studies of murine models of cGVHD, treat- been identified as being necessary for both TCR
ment with ibrutinib was shown to delay disease signaling activation,114 and activation of BCR
progression, increase GVHD progression-free signaling pathways leading to B-cell proliferation
survival, and improve both clinical and pathologi- and survival.115 Syk activation also leads to acti-
cal findings in the T-cell mediated model. vation of multiple immune cells (mast cells, mac-
Improved pulmonary function and tissue immu- rophages, neutrophils) leading to production and
noglobulin deposition was noted in an alloanti- release of pro-inflammatory cytokines. Based on
body-driven model that induced bronchiolitis these early observations, Syk inhibitors were ini-
obliterans (BO).111 In contrast to murine studies tially tested in murine models and early stage
by Schutt and colleagues that showed prophylac- clinical trials of various autoimmune diseases
tic ibrutinib to be effective in preventing cGVHD such as SLE, RA, and IgA nephropathy with var-
in mice,112 Dubovsky and colleagues did not see ying degrees of success.116–120 Also, there are cur-
any reduction in cGVHD in their mouse model rently several phase I/II trials evaluating the
when treated with ibrutinib prophylactically start- safety and efficacy of two different orally availa-
ing day –2 through day 28.111 ble Syk inhibitors, fostamatinib and entospletinib

30 journals.sagepub.com/home/tah
L Hill, A Alousi et al.

(GS-9973), for the treatment of relapsed/refrac- shown to increase regulatory T cells via phospho-
tory B-cell malignancies. rylation of STAT5.123,127 Recently, Flynn and
colleagues were able to show reversal of cGVHD
Syk inhibitors have also shown efficacy in animal manifestations in two murine models after tar-
and human studies of both acute and chronic geted inhibition of ROCK2, and further validated
GVHD. Leonhardt and colleagues showed the KD025-mediated effects on STAT3 and
decreased aGVHD, improved survival and STAT5 phosphorylation shifting the balance
reduced inflammatory cytokine profile (IL-6, from a pro-inflammatory balance.130 Based on
INF-γ, MCP-1) in mice treated with fostamatinib these promising preclinical findings, there is cur-
compared with those treated with cyclosporine A rently a phase II trial underway to evaluate the
114 without affecting T-cell cytolytic function. In a safety, tolerability, and activity of KD025 for the
murine model of sclerodermatous cGVHD, treatment of SR-cGVHD [ClinicalTrials.gov
administration of fostamatinib led to decreased identifier: NCT02841995].
fibrosis and a reduction in severity of scleroder-
matous changes.121 Deletion of the Syk gene in
murine donor bone marrow cells and in vivo inhi- Immune checkpoint blockade
bition with fostamatinib both showed reversibility
of chronic lung GVHD manifestations in a mouse Cytotoxic T-lymphocyte associated protein-4
model with BO.115 Inhibition of Syk also led to Cytotoxic T-lymphocyte associated protein-4
apoptosis in vitro of human B cells from patients (CTLA-4) is a negative regulator of T-cell
with active cGVHD. immune function. T-cell activation requires mul-
tiple costimulatory signals, one of which is the
Based on results from early-phase clinical trials in binding of CD28 on T-cells with CD80 or CD86
autoimmune diseases and preclinical data in on APCs. CTLA-4 is homologous to CD28, but
cGVHD, entospletinib is currently being evalu- binds with a greater affinity and avidity than
ated in a phase II trial in combination with sys- CD28. It is expressed on activated T cells and
temic corticosteroids for first-line treatment of binds to CD80/CD86, thereby blocking the inter-
cGVHD [ClinicalTrials.gov identifier: action with CD28, and inhibits T-cell prolifera-
NCT02701634]. Fostamatinib is currently being tion, differentiation and survival.131 Abatacept is a
tested for prevention of cGVHD [ClinicalTrials. selective costimulation modulator composed of
gov identifier: NCT02611063]. human CTLA-4 and a fragment of the fragment
crystallizable (Fc) domain of human immuno-
globulin-G1. Given the role that activated T cells
Rho kinase inhibitors play in GVHD, inhibition of T-cell activation via
Rho-associated protein kinase (ROCK) is a ser- blockade of costimulatory molecules, distinct
ine-threonine kinase primarily involved in regu- from direct inhibition of CTLA-4, could be a
lating cytoskeleton and cellular functions via potential therapeutic option.
phosphorylation of various downstream sub-
strates.122,123 Two isoforms have been identified: Results from a phase I trial evaluating the use of
ROCK1 and ROCK2, sharing more than 90% abatacept in patients with SR-cGVHD were pre-
homology within their kinase domain. Recent sented at the 2016 ASH conference.132 A total of
studies suggest that the ROCK2 isoform may 17 patients were enrolled and 16 were treated
play a specific role in the development of autoim- with a planned total of six doses of abatacept. In
munity, as inhibition of ROCK2 in mice and all, 7/16 (44%) evaluable patients had a PR based
human T-cells was effective in decreasing pro- on the NIH consensus criteria. Prednisone dos-
duction of IL-17 and IL-21, both pro-inflamma- age was decreased by ~51% by 1 month following
tory cytokines that have been identified as completion of the sixth dose. Severe AEs included
mediators of autoimmune disorders such as RA one grade IV pulmonary infection and three grade
and SLE.123–127 III cases, all of which resolved. Other low-grade
events included gastritis, diarrhea, fatigue, rash,
KD025 is an orally available selective ROCK2 and skin pain. The trial is currently ongoing.
inhibitor that downregulates phosphorylation of Overall, abatacept was well tolerated and based
STAT3 which is a transcription factor necessary on these promising results, a phase II trial is being
for the induction and expression of IL-17 and planned. There are also several trials underway
IL-21.127–129 Inhibition of ROCK2 was also using it in the preventive setting.

journals.sagepub.com/home/tah 31
Therapeutic Advances in Hematology 9(1)

Cytokine modulation the median steroid dose from 50 mg/day to 20


mg/day by week 15 (p < 0.001). However, pro-
Interleukin-2 gression of cGVHD was observed after cessation
IL-2 is a cytokine necessary for Treg develop- of bortezomib. While these results are encourag-
ment, expansion, activity, and survival.133–135 As ing, lack of a comparator arm makes it difficult to
previously mentioned, Tregs play an important discern the true impact of adding bortezomib to
role in preventing cGVHD. Koreth and col- steroids. Larger, well-designed randomized trials
leagues administered daily low-dose subcutane- are needed to determine if there is a true benefit.
ous IL-2 to 29 patients with SR-cGVHD at three There is currently a phase II trial underway for
different dose levels (0.3 × 106; 1 × 106 or 3 × upfront treatment of bronchiolitis obliterans
106 IU/m2) for a total of 8 weeks.136 In all, 12 of [ClinicalTrials.gov identifier: NCT01163786].
23 evaluable patients had major responses in mul-
tiple sites. Tregs were increased in all patients In a single-arm pilot study of bortezomib in
with minimal effect on conventional CD4+ T patients with steroid-dependent, -intolerant, or
cells.136,137 The highest dose level of 3 × 106 refractory cGVHD, five of six (83%) patients
IU/m2 was not well tolerated due to persistent achieved a PR defined as a decrease in ⩾1 point
grade I constitutional symptoms (fever, malaise on a 0–3-point scale per NIH consensus crite-
and arthralgias) requiring 50% dose reduction.136 ria.145 Most responders were also able to reduce
Overall, the therapy was safe and well tolerated at the dose or number of immunosuppressive thera-
the lower dose levels. pies being used during treatment with borte-
zomib. Flow cytometry revealed a significant
In a phase II study by the same authors, 35 decrease in B cells and an increase in Tregs.
patients with SR-cGVHD were treated with daily These findings suggest that proteasome inhibition
IL-2 for 12 weeks with 61% of evaluable patients may be a potential targeted therapy but additional
having a clinical response in multiple organ sites, studies with longer follow up are needed. A phase
including liver, skin, GI tract, lung, and joint/ II trial is underway for SR-cGVHD [ClinicalTrials.
muscle/fascia.138 The Treg cell numbers in the gov identifier: NCT01158105].
blood were again noted to be significantly ele-
vated compared with baseline, with no significant
change in conventional T cells. Several clinical Carfilzomib
trials are now recruiting to evaluate the use of Carfilzomib is an irreversible, second-generation
IL-2 alone or in combination with Tregs or extra- PI that potently inhibits the 26S proteasome. It
corporeal photopheresis (ECP) for treatment of has been shown to have fewer off-target side
SR-cGVHD (Table 4). effects, specifically, less peripheral neuropathy,
compared with bortezomib.146 It is currently
being studied for treatment of SR-cGVHD in a
Proteasome inhibitors phase II study [ClinicalTrials.gov identifier:
NCT02491359].
Bortezomib
As previously mentioned, bortezomib inhibits
nuclear factor-kappa B which is important for Ixazomib
B-cell development, activation, and survival in Ixazomib is a reversible second-generation oral
addition to T cells.139 Prior studies have shown PI that inhibits the 20S proteasome.146 It has
that bortezomib has inhibitory effects on acti- been well tolerated in patients treated with mul-
vated B cells and plasma cells,140,141 and case tiple myeloma, with the main toxicity being mild
reports of multiple myeloma patients treated with GI effects. It is being tested in a phase II trial
bortezomib for relapsed disease or maintenance for SR-cGVHD [ClinicalTrials.gov identifier:
therapy following HSCT showed encouraging NCT02513498].
improvement in cGVHD symptoms.142–144 A
phase II trial evaluating use of bortezomib in
combination with steroids for initial therapy of Anti-CD20 monoclonal antibodies
cGVHD reported an ORR at week 15 of 80% The role of B-cell dysregulation in the underlying
with two (10%) complete responses and 14 (70%) pathophysiology of cGVHD, as described above,
PRs in 20 evaluable patients.139 Responses were has led to the identification of B-cells as a thera-
seen in most organs, and it was possible to taper peutic target in treating cGVHD.

32 journals.sagepub.com/home/tah
L Hill, A Alousi et al.

Table 4.  Clinical trials of interleukin-2 and regulatory T cells (registered through ClinicalTrials.gov).

Trial number Study design Diagnosis Intervention Location Status


NCT01937468 Phase I SR-cGVHD LD IL-2 + Tregs Dana Farber Recruiting
NCT02340676 Phase II SR-cGVHD LD IL-2 + ECP Dana Farber Recruiting
NCT03007238 Phase II SR-cGVHD LD IL-2 + ECP City of Hope Recruiting
NCT02318082 Phase II SR-cGVHD Dose-escalated Dana Farber Recruiting
IL-2
NCT02749084 Phase I/II SR-cGVHD Donor Treg DLI Italy Recruiting
NCT02385019 Phase I/II SR-cGVHD Donor Treg Portugal Recruiting
NCT02519816 Phase II SR-cGVHD TCD + Treg Canada Recruiting
Expansion
SR, steroid refractory; cGVHD, chronic graft versus host disease; DLI, donor lymphocyte infusion; ECP, extracorporeal
photopheresis; LD IL-2, low-dose interleukin-2; TCD, T-cell depletion; Tregs, regulatory T cells.

Table 5.  Clinical trials utilizing mesenchymal stem cells (registered through ClinicalTrials.gov).

Trial identifier Study design Diagnosis Intervention Location Status


NCT00603330 Phase II SR-aGVHD MSC Germany Recruiting
NCT02770430 Phase II SR-aGVHD MSC Brazil Recruiting
NCT02055625 Phase I/II Oral cGVHD MSC Sweden Recruiting
NCT02379442 Phase I/II aGVHD MSC + CCS NIH Recruiting
NCT03158896 Phase I HR or SR- MSC USA Not yet
aGVHD Recruiting
NCT02923375 Phase I SR-aGVHD MSC Australia, Recruiting
UK
NCT02241018 Phase I/II SR-aGVHD Anti-CD25 + CNI China Recruiting
+/– MSC
NCT02291770 Phase III cGVHD First-line tx +/– China Recruiting
MSC
NCT02687646 Phase I/II SR-aGVHD MSC Spain Recruiting
NCT02032446 Phase I/II SR-aGVHD MSC Italy Recruiting
NCT02359929 Phase I SR-aGVHD / MSC Emory Recruiting
SR-cGVHD
NCT02336230 Phase III SR-aGVHD MSC USA Recruiting
SR, steroid refractory for NCT03158896; aGVHD, acute graft versus host disease; cGVHD, chronic graft versus host
disease; HR, high risk; CCS, corticosteroids; CNI, calcineurin inhibitor; MSC, mesenchymal stem cells; tx, treatment;
NIH, National Institutes of Health.

Rituximab is a chimeric mouse/human IgG anti- approved for treatment of CD20 positive B-cell
CD20 antibody that mediates B-cell lysis via anti- malignancies. Most data reported on its use in
body-dependent cellular cytotoxicity (ADCC), cGVHD are based on case reports, small early-
complement-dependent cytotoxicity (CDC), and phase trials, or retrospective studies; with ORRs
induction of apoptosis. It is currently FDA ranging from 50% to 86% reported.147 The best

journals.sagepub.com/home/tah 33
Table 6.  Responses and toxicities of conventional therapies compared with new monoclonal antibodies.

Drug/treatment Study design Line of tx n ORR CR OS Toxicities Reference


Anti-CD52  
Alemtuzumab Retrospective SR-aGVHD 18 83% 33% 55% Infections 78% Gómez-Almaguer
Grade II–IV CMV 61% et al.168
Neutropenia 43%
Therapeutic Advances in Hematology 9(1)

  Retrospective GI SR-aGVHD 20 70% 35% 50% (1 Bacterial 40% Schnitzler et al.169


Grade III–IV year) Aspergillosis 35%
EBV 15%
CMV frequent
  Phase II SR-aGVHD 10 55% 20% 0% Infections 80% Martínez et al.170
Grade III–IV CMV 70%
Cytopenias 60%
  Phase II SR-aGVHD 18 99% 28% 35% Bacterial 61% Schub et al.171
Grade III–IV Viral 44%
Fungal 22%
CMV 56%
EBV 11%
Anti-TNF  
Infliximab Retrospective SR-aGVHD Grade 21 67% 62% 38% (21 Bacterial 81% Couriel et al.172
II–IV mos) Fungal 48%
Viral 67%
  Retrospective SR-aGVHD Grade 32 59% 19% 68% Infections 72% Patriarca et al.173
II–IV CMV 41%
Etanercept Retrospective SR-aGVHD Grade 13 46% 31% 69% Bacterial 14% Busca et al.174
II–IV 8 62% 12% Fungal 19%
SR-cGVHD CMV 48%
  Phase I/II SR-cGVHD 10 60% 10% – Cause of death: Chiang et al.175
infection 10%

34 journals.sagepub.com/home/tah
Table 6. (Continued)
Drug/treatment Study design Line of tx n ORR CR OS Toxicities Reference
Immunmodulatory  
Mycophenolate mofetil Phase II SR-aGVHD 13 31% 15% 33% (2 Gastrointestinal Kim et al.176
Grade II–IV 13 77% – years) 27%
SR-cGVHD 54% (2 Infections 23%
years) CMV 11%
  Retrospective SR-aGVHD 10 60% 0% 70% Infection 67% Krejci et al.177
Grade II–III 11 64% – – Hematologic 29%
SR-cGVHD
  Phase II SR-aGVHD 19 47% 31% 16% Cause of death: Furlong et al.178
Grade II–IV Infection 32%
Sirolimus Pilot SR-aGVHD 21 57% 24% 33% TCP 33% Benito et al.26
Grade III–IV Neutropenia 19%
TG 38%
TMA 23%
  Retrospective SR-aGVHD 34 76% 44% 44% TG 71% Hoda et al.179
TMA 21%
  Phase II SR-cGVHD 35 63% 17% 57% (3 Infections 77% Couriel et al.24
years) TMA 11%
Renal 66%
TG 18%
Pentostatin Phase I SR-aGVHD 23 77% 63% 26% TCP 4% Bolaños-Meade
Grade II–IV Infection 9% et al.180
  Retrospective SR-aGVHD 12 50% 33% 10% Bacterial 50% Pidala et al.181
Grade II–IV Fungal 25%
Viral 8%
CMV 25%
  Phase II SR-cGVHD 58 55% – 70% (2 Infection 20% Jacobsohn et al.182
years)
Cellular  
photoimmunotherapy
Extracorporeal Pilot SR-aGVHD 21 67% 60% 57% Anemia 90% Greinix et al.183
photopheresis Grade II–IV TCP 71%
CMV 52%

(Continued)

journals.sagepub.com/home/tah 35
L Hill, A Alousi et al.
Table 6. (Continued)
Drug/treatment Study design Line of tx n ORR CR OS Toxicities Reference
  Phase I/II SR-aGVHD 9 78% 56% 56% (8 Hypotension Salvaneschi
Grade II–IV 14 64% 29% months) Anemia et al.184
SR-cGVHD 79% (36 CMV reactivation
months)
Therapeutic Advances in Hematology 9(1)

  Retrospective SR-aGVHD 23 – 52% 48% Hypotension Perfetti et al.185


Grade II–IV Anemia
TCP
  Retrospective SR-cGVHD 71 61% 20% 53% Anemia Couriel et al.186
Thrombocytopenia
Anti-α4β7 integrin  
Vedolizumab Case series SR-aGVHD 6 100% – 4/6 pts None reported Floisand et al.70
GI grade IV alive at
median
f/u 10
months
Anti-CD30  
Brentuximab vedotin Phase I SR-aGVHD 34 38% 15% 38% Neutropenia Chen et al.75
Elevated bilirubin
Headache
Ileus
Anti-CTLA4  
Abetacept Phase I SR-cGVHD 16 44% (PR) – – Pulmonary infection Nahas et al.132
Diarrhea/gastritis
Rash
aGVHD, acute graft versus host disease; cGVHD, chronic graft versus host disease; CMV, cytomegalovirus; CR, complete response; EBV, Epstein-Barr virus; f/u, follow up; GI,
gastrointestinal; ORR, overall response rate; OS, overall survival; PR, partial response; PTLD, post-transplant lymphoproliferative disease; pts, patients; ref, reference; SR, steroid
refractory; TCP, thrombocytopenia; TG, triglyceridemia; TMA, thrombotic microangiopathy; TNF, tumor necrosis factor; tx, treatment.

36 journals.sagepub.com/home/tah
L Hill, A Alousi et al.

response was seen primarily in patients with cuta- GVHD following HLA-matched allogeneic
neous and oral involvement. Rituximab has also HSCT.155 Several phase I/II studies have also
been demonstrated to be effective for GVHD shown that infusion of native donor Tregs and
prophylaxis in a phase II study that showed a ex vivo-expanded umbilical-cord-blood-derived
reduction in steroid-requiring cGVHD and Tregs are associated with a lower incidence of
improved OS in patients receiving peripheral aGVHD and improved immune reconstitution
blood grafts.148 However, randomized trials are compared with historical controls.156–158 In one
still needed to confirm these findings. study, no patients who received Treg infusion
developed cGVHD at 1 year versus 14% in the
control group.158 Parmar and colleagues were
Ofatumumab able to show in animal models that infusion of
Ofatumumab is a fully humanized IgG1 kappa sec- fucosylated Tregs to improve engraftment and
ond-generation monoclonal antibody against CD20. homing lead to longer in vivo persistence and
Compared with rituximab, it has more potent CDC decreased aGVHD at a lower cell dose compared
leading to increased B-cell destruction due to a with untreated Tregs.159
higher binding affinity.149 Ofatumumab is currently
FDA approved for treatment of chronic lymphocytic Studies of Tregs in the setting of cGVHD have
leukemia. Pidala and colleagues recently demon- shown reduced frequency of circulating Tregs
strated safety of ofatumumab in combination with compared with patients without cGVHD and nor-
steroids for the treatment of newly diagnosed mod- mal controls; however, the function of the cells
erate-to-severe cGVHD.150 Twelve patients were remains normal.154,160,161 The first in-human use
enrolled and two infusion reactions (grade II and III, of adoptive transfer of ex vivo-expanded Tregs was
respectively) were observed. No grade IV infusion performed in two patients; one with SR-aGVHD
reactions, constitutional symptoms, or ⩾ grade III and the other with SR-cGVHD.162 The patient
organ toxicities were observed; and hence no dose- with cGVHD had significant improvement in
limiting toxicities occurred. A phase II study is cur- GVHD symptoms and was able to reduce the
rently in progress to assess efficacy as primary amount of immunosuppressive therapy required.
therapy in cGVHD [ClinicalTrials.gov identifier: Theil and colleagues evaluated use of adoptive
NCT01680965]. Treg transfer in five patients with SR-cGVHD;
two patients had clinical improvement in GVHD
symptoms while the other three had stable disease
Obinutuzumab as the best response.163 No significant toxicity or
Obinutuzumab is a second-generation anti-CD20 GVHD exacerbations were observed.
monoclonal antibody currently FDA approved for
the treatment of follicular lymphoma. Based on the One potential limiting factor of widespread use of
promising activity of B-cell depletion for preven- adoptive Treg therapy is the need to manufacture
tion of cGVHD with rituximab, a phase II study sufficient numbers of Tregs for infusion without
using obinutuzumab for prevention of cGVHD contaminating the product with other T-cell subsets,
following peripheral blood stem-cell transplanta- such as effector T cells, which could affect the
tion is currently recruiting [ClinicalTrials.gov response. This requires laboratory expertise to per-
identifier: NCT02867384]. form complex methods for both cell purification and
expansion.164 Larger prospective randomized stud-
ies are still needed to determine the benefit and long-
Adoptive cell therapy term outcomes. See Table 4 for ongoing clinical
trials.
Regulatory T cells
Tregs are a subset of peripheral CD4+ T cells
(~5–10%) that are imperative for the develop- Mesenchymal stromal cells
ment of immune tolerance in healthy individu- Response rates in treatment of SR-cGVHD have
als.151–154 Reduction or loss of this population of been less robust,86,165,166 except notably in patients
T cells is associated with loss of immune toler- with sclerodermatous GVHD. The best reported
ance and development of autoimmunity. Rezvani response rate was seen in four patients with exten-
and colleagues reported that donor Treg content sive skin scleroderma and ulceration who had sig-
is predictive of aGVHD post-transplant, with a nificant improvement after four to eight treatments
high Treg content associated with a lower risk of with MSCs.167 Lack of clear benefit in cGVHD

journals.sagepub.com/home/tah 37
Therapeutic Advances in Hematology 9(1)

may be due to significant heterogeneity in multi- Conflict of interest statement


ple aspects between studies.81 Well-designed ran- The authors declare that there is no conflict of
domized studies with standardized methods of interest.
source procurement, expansion techniques, and
dosing schedules are needed to further clarify the
role of MSCs in the treatment of GVHD. There References
are currently three trials underway evaluating    1. Wingard JR, Majhail NS, Brazauskas R,
MSCs for treatment of cGVHD (Table 5). et al. Long-term survival and late deaths after
allogeneic hematopoietic cell transplantation.
J Clin Oncol 2011; 29: 2230–2239.
Conclusion    2. Pidala J and Anasetti C. Glucocorticoid-
As the number of allogeneic HSCT continues to refractory acute graft-versus-host disease. Biol
increase each year, the importance and need for Blood Marrow Transplant 2010; 16:
effective therapies to treat aGVHD and cGVHD 1504–1518.
and thereby improve outcomes for this potentially
   3. Horwitz ME and Sullivan KM. Chronic graft-
curative therapy cannot be over-emphasized.
versus-host disease. Blood Rev 2006; 20:
Responses to primary therapy with corticoster- 15–27.
oids are often suboptimal and treatment can be
complicated by significant side effects, including    4. Bhushan V and Collins RH Jr. Chronic graft-
diabetes, hypertension, osteoporosis, myopathy, versus-host disease. JAMA 2003; 290:
and avascular necrosis. Available second-line 2599–603.
treatments have even lower response rates, lead-    5. Lee SJ, Vogelsang G and Flowers ME. Chronic
ing to poor outcomes in acute GVHD, and pro- graft-versus-host disease. Biol Blood Marrow
longed morbidity with delayed immune recovery Transplant 2003; 9: 215–233.
with chronic GVHD. Currently, there is a desper-    6. Khoury HJ, Wang T, Hemmer MT, et al.
ate need for new treatment options for this mor- Improved survival after acute graft-
bid complication of transplant. It would be ideal versus-host disease diagnosis in the
to identify targeted therapies that are not as modern era. Haematologica 2017; 102:
broadly immunosuppressive, which can be used 958–966.
prior to the development of severe and irreversi-    7. Hahn T, McCarthy PL, Hassebroek A, et al.
ble tissue damage, and can provide durable Significant improvement in survival after
responses without impacting graft versus tumor allogeneic hematopoietic cell transplantation
(GVT) effects. Continued understanding of the during a period of significantly increased use,
underlying immune mechanisms and pathways older recipient age, and use of unrelated donors.
that are involved in the pathophysiology of J Clin Oncol 2013; 31: 2437–2449.
GVHD should yield development of more effec-    8. El-Jawahri A, Li S, Antin JH, et al. Improved
tive therapeutics. treatment-related mortality and overall survival
of patients with grade IV acute GVHD in the
While these novel therapies have been tested in modern years. Biol Blood Marrow Transplant
preclinical models and have shown promising 2016; 22: 910–918.
results in early phase studies, larger multicenter
   9. Matte CC, Liu J, Cormier J, et al. Donor APCs
studies are needed to determine true efficacy, are required for maximal GVHD but not for
preferably in comparison with current available GVL. Nat Med 2004; 10: 987–992.
therapies and long-term outcomes. Many of these
agents are also being studied for prevention of   10. Koyama M, Kuns RD, Olver SD, et al. Recipient
GVHD, which would be ideal as long as GVT is nonhematopoietic antigen-presenting cells are
sufficient to induce lethal acute graft-versus-host
not compromised. These therapies should help to
disease. Nat Med 2011; 18: 135–142.
improve symptoms and prevent progression of
GVHD, to enhance patient quality of life and   11. Paczesny S, Hanauer D, Sun Y, et al. New
ultimately improve OS. perspectives on the biology of acute GVHD.
Bone Marrow Transplant 2010; 45: 1–11.
Funding   12. Magenau J, Runaas L and Reddy P. Advances
This research received no specific grant from any in understanding the pathogenesis of graft-
funding agency in the public, commercial, or not- versus-host disease. Br J Haematol 2016; 173:
for-profit sectors. 190–205.

38 journals.sagepub.com/home/tah
L Hill, A Alousi et al.

  13. Zeiser R, Socie G and Blazar BR. Pathogenesis   25. Johnston LJ, Brown J, Shizuru JA, et al.
of acute graft-versus-host disease: From Rapamycin (sirolimus) for treatment of chronic
intestinal microbiota alterations to donor T cell graft-versus-host disease. Biol Blood Marrow
activation. Br J Haematol 2016; 175: 191–207. Transplant 2005; 11: 47–55.
  14. Weisdorf D, Haake R, Blazar B, et al. Treatment   26. Benito AI, Furlong T, Martin PJ, et al.
of moderate/severe acute graft-versus-host disease Sirolimus (rapamycin) for the treatment of
after allogeneic bone marrow transplantation: steroid-refractory acute graft-versus-host
an analysis of clinical risk features and outcome. disease. Transplantation 2001; 72: 1924–1929.
Blood 1990; 75: 1024–1030.   27. Lutz M and Mielke S. New perspectives on
  15. MacMillan ML, Weisdorf DJ, Wagner JE, et al. the use of mTOR inhibitors in allogeneic
Response of 443 patients to steroids as primary haematopoietic stem cell transplantation and
therapy for acute graft-versus-host disease: graft-versus-host disease. Br J Clin Pharmacol
comparison of grading systems. Biol Blood 2016; 82: 1171–1179.
Marrow Transplant 2002; 8: 387–394.   28. Pidala J, Tomblyn M, Nishihori T, et al.
  16. MacMillan ML, Robin M, Harris AC, et al. A Sirolimus demonstrates activity in the primary
refined risk score for acute graft-versus-host therapy of acute graft-versus-host disease
disease that predicts response to initial therapy, without systemic glucocorticoids. Haematologica
survival, and transplant-related mortality. Biol 2011; 96: 1351–1356.
Blood Marrow Transplant 2015; 21: 761–767.   29. Yamaoka K, Saharinen P, Pesu M, et al. The
  17. Levine JE, Logan B, Wu J, et al. Graft-versus- Janus kinases (Jaks). Genome Biol 2004; 5: 253.
host disease treatment: predictors of survival.   30. Spoerl S, Mathew NR, Bscheider M, et al.
Biol Blood Marrow Transplant 2010; 16: Activity of therapeutic JAK 1/2 blockade in
1693–1699. graft-versus-host disease. Blood 2014;123:
3832–3842.
  18. Deeg HJ. How I treat refractory acute GVHD.
Blood 2007; 109: 4119–4126.   31. O’Shea JJ and Murray PJ. Cytokine signaling
modules in inflammatory responses. Immunity
  19. Castilla-Llorente C, Martin PJ, McDonald 2008; 28: 477–487.
GB, et al. Prognostic factors and outcomes of
severe gastrointestinal GVHD after allogeneic   32. Perner F, Schnoder TM, Ranjan S, et al.
hematopoietic cell transplantation. Bone Marrow Specificity of JAK-kinase inhibition determines
Transplant 2014; 49: 966–971. impact on human and murine T-cell function.
Leukemia 2016; 30: 991–995.
  20. Martin PJ, Rizzo JD, Wingard JR, et al. First-
and second-line systemic treatment of acute   33. Murray PJ. The JAK-STAT signaling pathway:
graft-versus-host disease: recommendations of input and output integration. J Immunol 2007;
the American Society of Blood and Marrow 178: 2623–2639.
Transplantation. Biol Blood Marrow Transplant   34. Heine A, Held SA, Daecke SN, et al. The
2012; 18: 1150–1163. JAK-inhibitor ruxolitinib impairs dendritic cell
  21. MacMillan ML, DeFor TE and Weisdorf DJ. function in vitro and in vivo. Blood 2013; 122:
What predicts high risk acute graft-versus-host 1192–1202.
disease (GVHD) at onset?: identification of   35. Betts BC, Abdel-Wahab O, Curran SA, et al.
those at highest risk by a novel acute GVHD Janus kinase-2 inhibition induces durable
risk score. Br J Haematol 2012; 157: 732–741. tolerance to alloantigen by human
dendritic cell-stimulated T cells yet preserves
  22. Levine JE, Braun TM, Harris AC, et al. A
immunity to recall antigen. Blood 2011; 118:
prognostic score for acute graft-versus-host
5330–5339.
disease based on biomarkers: a multicentre
study. Lancet Haematol 2015; 2: e21–e29.   36. Hechinger AK, Smith BA, Flynn R, et al.
Therapeutic activity of multiple common
  23. Kirken RA and Wang YL. Molecular actions of
gamma-chain cytokine inhibition in acute and
sirolimus: sirolimus and mTor. Transplant Proc
chronic GVHD. Blood 2015; 125: 570–580.
2003; 35(Suppl. 3): 227S–230S.
  37. Choi J, Ziga ED, Ritchey J, et al. IFNγR
  24. Couriel DR, Saliba R, Escalon MP, et al.
signaling mediates alloreactive T-cell trafficking
Sirolimus in combination with tacrolimus and
and GVHD. Blood 2012; 120: 4093–4103.
corticosteroids for the treatment of resistant
chronic graft-versus-host disease. Br J Haematol   38. Choi J, Cooper ML, Alahmari B, et al.
2005; 130: 409–417. Pharmacologic blockade of JAK1/JAK2 reduces

journals.sagepub.com/home/tah 39
Therapeutic Advances in Hematology 9(1)

GVHD and preserves the graft-versus-leukemia marrow transplantation model. Blood 2011;
effect. PloS One 2014; 9: e109799. 118: 5031–5039.
  39. Zeiser R, Burchert A, Lengerke C, et al.   50. Marcondes AM, Karoopongse E, Lesnikova M,
Ruxolitinib in corticosteroid-refractory graft- et al. alpha-1-antitrypsin (AAT)-modified donor
versus-host disease after allogeneic stem cell cells suppress GVHD but enhance the GVL
transplantation: a multicenter survey. Leukemia effect: a role for mitochondrial bioenergetics.
2015; 29: 2062–2068. Blood 2014; 124: 2881–2891.
  40. Schroeder MA, Khoury HJ, Jagasia M, et al. A   51. Tawara I, Sun Y, Lewis EC, et al. Alpha-1-
phase I trial of Janus Kinase (JAK) inhibition antitrypsin monotherapy reduces graft-versus-
with INCB039110 in acute graft-versus-host host disease after experimental allogeneic bone
disease (aGVHD). Blood 2016; 128: abstract marrow transplantation. Proc Natl Acad Sci U S
390. A 2012; 109: 564–569.
  41. Im A, Hakim FT and Pavletic SZ. Novel targets   52. Marcondes AM, Hockenbery D, Lesnikova
in the treatment of chronic graft-versus-host M, et al. Response of steroid-refractory acute
disease. Leukemia 2017; 31: 543–554. GVHD to alpha1-antitrypsin. Biol Blood Marrow
Transplant 2016; 22: 1596–601.
  42. Moreau P, Richardson PG, Cavo M, et al.
Proteasome inhibitors in multiple myeloma: 10   53. Rutz S, Eidenschenk C and Ouyang W. IL-22,
years later. Blood 2012; 120: 947–959. not simply a Th17 cytokine. Immunol Rev 2013;
252: 116–132.
  43. Finn PW, Stone JR, Boothby MR, et al.
Inhibition of NF-κB-dependent T cell activation   54. Lamarthee B, Malard F, Gamonet C, et al.
abrogates acute allograft rejection. J Immunol Donor interleukin-22 and host type I interferon
2001; 167: 5994–6001. signaling pathway participate in intestinal graft-
  44. Sun K, Welniak LA, Panoskaltsis-Mortari versus-host disease via STAT1 activation and
A, et al. Inhibition of acute graft-versus-host CXCL10. Mucosal Immunol 2016; 9: 309–321.
disease with retention of graft-versus-tumor   55. Hanash AM, Dudakov JA, Hua G, et al.
effects by the proteasome inhibitor bortezomib. Interleukin-22 protects intestinal stem cells
Proc Natl Acad Sci U S A 2004; 101: 8120– from immune-mediated tissue damage and
8125. regulates sensitivity to graft versus host disease.
  45. Sun K, Wilkins DE, Anver MR, et al. Immunity 2012; 37: 339–350.
Differential effects of proteasome inhibition   56. Couturier M, Lamarthee B, Arbez J, et al.
by bortezomib on murine acute graft-versus- IL-22 deficiency in donor T cells attenuates
host disease (GVHD): delayed administration murine acute graft-versus-host disease mortality
of bortezomib results in increased GVHD- while sparing the graft-versus-leukemia effect.
dependent gastrointestinal toxicity. Blood 2005; Leukemia 2013; 27: 1527–1537.
106: 3293–3299.
  57. Lindemans CA, Calafiore M, Mertelsmann
  46. Vodanovic-Jankovic S, Hari P, Jacobs P, et al. AM, et al. Interleukin-22 promotes intestinal-
NF-κB as a target for the prevention of graft- stem-cell-mediated epithelial regeneration.
versus-host disease: comparative efficacy of Nature 2015; 528: 560–564.
bortezomib and PS-1145. Blood 2006; 107:
827–834.   58. Sabat R, Ouyang W and Wolk K. Therapeutic
opportunities of the IL-22-IL-22R1 system. Nat
  47. Koreth J, Stevenson KE, Kim HT, et al. Rev Drug Discov 2014; 13: 21–38.
Bortezomib, tacrolimus, and methotrexate for
prophylaxis of graft-versus-host disease after   59. Wolk K, Witte E, Wallace E, et al. IL-22
reduced-intensity conditioning allogeneic stem regulates the expression of genes responsible for
cell transplantation from HLA-mismatched antimicrobial defense, cellular differentiation,
unrelated donors. Blood 2009; 114: 3956–3959. and mobility in keratinocytes: a potential role in
psoriasis. Eur J Immunol 2006; 36:
  48. Wagner JL, Mookerjee B, Filicko-O’Hara JE,
1309–1323.
et al. Bortezomib for steroid refractory acute
graft versus host disease (GVHD). Biol Blood   60. Beniwal-Patel P and Saha S. The role of
Marrow Transplant 18: S370. integrin antagonists in the treatment of
inflammatory bowel disease. Expert Opin Biol
  49. Marcondes AM, Li X, Tabellini L, et al.
Ther 2014; 14: 1815–1823.
Inhibition of IL-32 activation by alpha-1
antitrypsin suppresses alloreactivity and   61. Danese S, Semeraro S, Marini M, et al.
increases survival in an allogeneic murine Adhesion molecules in inflammatory bowel

40 journals.sagepub.com/home/tah
L Hill, A Alousi et al.

disease: therapeutic implications for gut serological and clinical features of the disease.
inflammation. Dig Liver Dis 2005; 37: 811–818. J Rheumatol 2000; 27: 698–702.
  62. Rommer PS, Dudesek A, Stuve O, et al.   74. Chen YB, McDonough S, Hasserjian R, et al.
Monoclonal antibodies in treatment of multiple Expression of CD30 in patients with acute
sclerosis. Clin Exp Immunol 2014; 175: 373– graft-versus-host disease. Blood 2012; 120:
384. 691–696.
  63. Schwab N, Schneider-Hohendorf T, Melzer N,   75. Chen YB, Perales MA, Li S, et al. Phase 1
et al. Natalizumab-associated PML: challenges multicenter trial of brentuximab vedotin for
with incidence, resulting risk, and risk steroid-refractory acute graft-versus-host
stratification. Neurology 2017; 88: disease. Blood 2017; 129: 3256–3261.
1197–1205.
  76. Kekre N and Antin JH. Emerging drugs for
  64. TYSABRI (natalizumab) US prescribing graft-versus-host disease. Expert Opin Emerg
information. Cambridge, MA: Biogen Inc., Drugs 2016; 21: 209–218.
2016.
  77. Kebriaei P and Robinson S. Treatment of graft-
  65. Lam MC and Bressler B. Vedolizumab versus-host-disease with mesenchymal stromal
for ulcerative colitis and Crohn’s disease: cells. Cytotherapy 2011; 13: 262–268.
results and implications of GEMINI studies.
  78. Aggarwal S and Pittenger MF. Human
Immunotherapy 2014; 6: 963–971.
mesenchymal stem cells modulate allogeneic
  66. Neurath MF. Current and emerging therapeutic immune cell responses. Blood 2005; 105:
targets for IBD. Nat Rev Gastroenterol Hepatol 1815–1822.
2017; 14: 269–278.
  79. Jiang Y, Jahagirdar BN, Reinhardt RL, et al.
  67. Dutt S, Ermann J, Tseng D, et al. L-selectin Pluripotency of mesenchymal stem cells derived
and beta7 integrin on donor CD4 T cells from adult marrow. Nature 2002; 418:
are required for the early migration to host 41–49.
mesenteric lymph nodes and acute colitis of
  80. Di Nicola M, Carlo-Stella C, Magni M, et al.
graft-versus-host disease. Blood 2005; 106:
Human bone marrow stromal cells suppress
4009–4015.
T-lymphocyte proliferation induced by cellular
  68. Petrovic A, Alpdogan O, Willis LM, et al. or nonspecific mitogenic stimuli. Blood 2002;
LPAM (alpha 4 beta 7 integrin) is an important 99: 3838–3843.
homing integrin on alloreactive T cells in the
  81. Rizk M, Monaghan M, Shorr R, et al.
development of intestinal graft-versus-host
Heterogeneity in studies of mesenchymal
disease. Blood 2004; 103: 1542–1547.
stromal cells to treat or prevent graft-versus-host
  69. Li B, New JY, Yap EH, et al. Blocking disease: a scoping review of the evidence. Biol
L-selectin and alpha4-integrin changes donor Blood Marrow Transplant 2016; 22: 1416–1423.
cell homing pattern and ameliorates murine
  82. Kebriaei P and Robinson S. Mesenchymal
acute graft versus host disease. Eur J Immunol
stem cell therapy in the treatment of acute and
2001; 31: 617–624.
chronic graft versus host disease. Front Oncol
  70. Fløisand Y, Lundin KE, Lazarevic V, et al. 2011; 1: 16.
Targeting integrin α4β7 in steroid-refractory
  83. Le Blanc K, Rasmusson I, Sundberg B,
intestinal graft-versus-host disease. Biol Blood
et al. Treatment of severe acute graft-versus-
Marrow Transplant 2017; 23: 172–175.
host disease with third party haploidentical
  71. Francisco JA, Cerveny CG, Meyer DL, et al. mesenchymal stem cells. Lancet 2004; 363:
cAC10-vcMMAE, an anti-CD30-monomethyl 1439–1441.
auristatin E conjugate with potent and selective
  84. Le Blanc K, Frassoni F, Ball L, et al.
antitumor activity. Blood 2003; 102:
Mesenchymal stem cells for treatment of
1458–1465.
steroid-resistant, severe, acute graft-versus-host
  72. Gerli R, Pitzalis C, Bistoni O, et al. CD30+ disease: a phase II study. Lancet 2008; 371:
T cells in rheumatoid synovitis: mechanisms 1579–1586.
of recruitment and functional role. J Immunol
  85. Von Bonin M, Stolzel F, Goedecke A, et al.
2000; 164: 4399–4407.
Treatment of refractory acute GVHD with
  73. Ihn H, Yazawa N, Kubo M, et al. Circulating third-party MSC expanded in platelet lysate-
levels of soluble CD30 are increased in patients containing medium. Bone Marrow Transplant
with localized scleroderma and correlated with 2009; 43: 245–251.

journals.sagepub.com/home/tah 41
Therapeutic Advances in Hematology 9(1)

  86. Ringden O, Uzunel M, Rasmusson I, et al. steroid-resistant acute graft-versus-host disease


Mesenchymal stem cells for treatment of of the gut. Blood 2016; 128: 2083–2088.
therapy-resistant graft-versus-host disease.
  97. Spindelboeck W, Schulz E, Uhl B, et al.
Transplantation 2006; 81: 1390–1397.
Repeated fecal microbiota transplantations
  87. Martin BJ, Uberti J, Soiffer R, et al. Prochymal attenuate diarrhea and lead to sustained changes
improves response rates in patients with steroid- in the fecal microbiota in acute, refractory
refractory acute graft versus host disease gastrointestinal graft-versus-host-disease.
(SR-GVHD) involving the liver and gut: results Haematologica 2017; 102: e210–e213.
of a randomized, placebo-controlled, multicenter
phase III trial in GVHD. Orlando, FL: American   98. Thien M, Phan TG, Gardam S, et al. Excess
Society of Bone Marrow Transplantation, 2010. BAFF rescues self-reactive B cells from
peripheral deletion and allows them to enter
  88. Hashmi S, Ahmed M, Murad MH, et al. forbidden follicular and marginal zone niches.
Survival after mesenchymal stromal cell therapy Immunity 2004; 20: 785–798.
in steroid-refractory acute graft-versus-host
disease: systematic review and meta-analysis.   99. Sarantopoulos S and Ritz J. Aberrant B-cell
Lancet Haematol 2016; 3: e45–e52. homeostasis in chronic GVHD. Blood 2015;
125: 1703–1707.
  89. Taur Y, Xavier JB, Lipuma L, et al. Intestinal
domination and the risk of bacteremia in 100. Allen JL, Fore MS, Wooten J, et al. B cells
patients undergoing allogeneic hematopoietic from patients with chronic GVHD are activated
stem cell transplantation. Clin Inf Dis 2012; 55: and primed for survival via BAFF-mediated
905–914. pathways. Blood 2012; 120: 2529–2536.

  90. Jenq RR, Ubeda C, Taur Y, et al. Regulation of 101. Mizoguchi A and Bhan AK. A case for
intestinal inflammation by microbiota following regulatory B cells. J Immunol 2006; 176:
allogeneic bone marrow transplantation. J Exp 705–710.
Med 2012; 209: 903–911.
102. Khoder A, Sarvaria A, Alsuliman A, et al.
  91. Holler E, Butzhammer P, Schmid K, et al. Regulatory B cells are enriched within the IgM
Metagenomic analysis of the stool microbiome memory and transitional subsets in healthy
in patients receiving allogeneic stem cell donors but are deficient in chronic GVHD.
transplantation: loss of diversity is associated Blood 2014; 124: 2034–2045.
with use of systemic antibiotics and more
103. Sarvaria A, Basar R, Mehta RS, et al. IL-
pronounced in gastrointestinal graft-versus-host
10+ regulatory B cells are enriched in cord
disease. Biol Blood Marrow Transplant 2014; 20:
blood and may protect against cGVHD after
640–645.
cord blood transplantation. Blood 2016; 128:
  92. Shono Y, Docampo MD, Peled JU, et al. 1346–1361.
Increased GVHD-related mortality with
104. Martin PJ, Inamoto Y, Carpenter PA, et al.
broad-spectrum antibiotic use after allogeneic
Treatment of chronic graft-versus-host disease:
hematopoietic stem cell transplantation in
past, present and future. Korean J Hematol
human patients and mice. Sci Transl Med 2016;
2011; 46: 153–163.
8: 339ra71.
105. Wolff D, Bertz H, Greinix H, et al. The
  93. Taur Y, Jenq RR, Perales MA, et al. The effects
treatment of chronic graft-versus-host disease:
of intestinal tract bacterial diversity on mortality
consensus recommendations of experts from
following allogeneic hematopoietic stem cell
Germany, Austria, and Switzerland. Dtsch
transplantation. Blood 2014; 124: 1174–1182.
Arztebl Int 2011; 108: 732–740.
  94. Atarashi K, Tanoue T, Oshima K, et al. Treg
106. Hakim FT, Memon S, Jin P, et al. Upregulation
induction by a rationally selected mixture of
of IFN-inducible and damage-response
Clostridia strains from the human microbiota.
pathways in chronic graft-versus-host disease. J
Nature 2013; 500: 232–236.
Immunol 2016; 197: 3490–503.
  95. Jenq RR, Taur Y, Devlin SM, et al. Intestinal
107. Filipovich AH, Weisdorf D, Pavletic S, et al.
Blautia is associated with reduced death from
National Institutes of Health consensus
graft-versus-host disease. Biol Blood Marrow
development project on criteria for clinical trials
Transplant 2015; 21: 1373–1383.
in chronic graft-versus-host disease: I. Diagnosis
  96. Kakihana K, Fujioka Y, Suda W, et al. Fecal and staging working group report. Biol Blood
microbiota transplantation for patients with Marrow Transplant 2005; 11: 945–956.

42 journals.sagepub.com/home/tah
L Hill, A Alousi et al.

108. Srinivasan M, Flynn R, Price A, et al. 120. Kim MJ, McDaid JP, McAdoo SP, et al. Spleen
Donor B-cell alloantibody deposition and tyrosine kinase is important in the production of
germinal center formation are required for proinflammatory cytokines and cell proliferation
the development of murine chronic GVHD in human mesangial cells following stimulation
and bronchiolitis obliterans. Blood 2012; 119: with IgA1 isolated from IgA nephropathy
1570–1580. patients. J Immunol 2012; 189: 3751–3758.
109. Kapur R, Ebeling S and Hagenbeek A. B-cell 121. Le Huu D, Kimura H, Date M, et al. Blockade
involvement in chronic graft-versus-host of Syk ameliorates the development of murine
disease. Haematologica 2008; 93: sclerodermatous chronic graft-versus-host
1702–1711. disease. J Dermatol Sci 2014; 74: 214–221.
110. Allen JL, Tata PV, Fore MS, et al. Increased 122. Riento K and Ridley AJ. Rocks: multifunctional
BCR responsiveness in B cells from patients kinases in cell behaviour. Nat Rev Mol Cell Biol
with chronic GVHD. Blood 2014; 123: 2108– 2003; 4: 446–456.
2115.
123. Zanin-Zhorov A, Flynn R, Waksal SD, et al.
111. Dubovsky JA, Flynn R, Du J, et al. Ibrutinib Isoform-specific targeting of ROCK proteins in
treatment ameliorates murine chronic immune cells. Small GTPases 2016; 7: 173–177.
graft-versus-host disease. J Clin Invest 2014;
124. Biswas PS, Gupta S, Chang E, et al.
124: 4867–4876.
Phosphorylation of IRF4 by ROCK2
112. Schutt SD, Fu J, Nguyen H, et al. Inhibition of regulates IL-17 and IL-21 production and the
BTK and ITK with Ibrutinib is effective in the development of autoimmunity in mice. J Clin
prevention of chronic graft-versus-host disease Invest 2010; 120: 3280–3295.
in Mice. PLoS One 2015; 10: e0137641.
125. Garrett-Sinha LA, John S and Gaffen SL.
113. Miklos D, Cutler CS, Arora M, et al. Ibrutinib IL-17 and the Th17 lineage in systemic lupus
for chronic graft-versus-host disease after erythematosus. Curr Opin Rheumatol 2008; 20:
failure of prior therapy. Blood 2017, https://doi. 519–525.
org/10.1182/blood-2017-07-793786 (accessed
126. Ettinger R, Kuchen S and Lipsky PE. The role
18 September 2017).
of IL-21 in regulating B-cell function in health
114. Leonhardt F, Zirlik K, Buchner M, et al. Spleen and disease. Immunol Rev 2008; 223: 60–86.
tyrosine kinase (Syk) is a potent target for
127. Zanin-Zhorov A, Weiss JM, Nyuydzefe MS,
GvHD prevention at different cellular levels.
et al. Selective oral ROCK2 inhibitor down-
Leukemia 2012; 26: 1617–1629.
regulates IL-21 and IL-17 secretion in human
115. Flynn R, Allen JL, Luznik L, et al. Targeting T cells via STAT3-dependent mechanism. Proc
Syk-activated B cells in murine and human Natl Acad Sci U S A 2014; 111: 16814–16819.
chronic graft-versus-host disease. Blood 2015;
128. Korn T, Bettelli E, Oukka M, et al. IL-17
125: 4085–4094.
and Th17 Cells. Annu Rev Immunol 2009; 27:
116. Deng GM, Liu L, Bahjat FR, et al. Suppression 485–517.
of skin and kidney disease by inhibition of
129. Chen Z, Laurence A and O’Shea JJ. Signal
spleen tyrosine kinase in lupus-prone mice.
transduction pathways and transcriptional
Arthritis Rheum 2010; 62: 2086–2092.
regulation in the control of Th17 differentiation.
117. Bahjat FR, Pine PR, Reitsma A, et al. An orally Semin Immunol 2007; 19: 400–408.
bioavailable spleen tyrosine kinase inhibitor 130. Flynn R, Paz K, Du J, et al. Targeted Rho-
delays disease progression and prolongs survival associated kinase 2 inhibition suppresses
in murine lupus. Arthritis Rheum 2008; 58: murine and human chronic GVHD through a
1433–1444. Stat3-dependent mechanism. Blood 2016; 127:
118. Braselmann S, Taylor V, Zhao H, et al. R406, an 2144–2154.
orally available spleen tyrosine kinase inhibitor 131. Buchbinder EI and Desai A. CTLA-4 and
blocks fc receptor signaling and reduces immune PD-1 pathways: similarities, differences, and
complex-mediated inflammation. J Pharmacol implications of their inhibition. Am J Clin Oncol
Exp Ther 2006; 319: 998–1008. 2016; 39: 98–106.
119. Smith J, McDaid JP, Bhangal G, et al. A spleen 132. Nahas MR, Soiffer RJ, Alyea EP, et al. Phase
tyrosine kinase inhibitor reduces the severity 1 clinical trial evaluating abatacept in patients
of established glomerulonephritis. J Am Soc with steroid-refractory chronic graft versus host
Nephrol 2010; 21: 231–236. disease. Blood 2016; 128: abstract 387.

journals.sagepub.com/home/tah 43
Therapeutic Advances in Hematology 9(1)

133. Malek TR and Bayer AL. Tolerance, not 145. Pai CC, Chen M, Mirsoian A, et al. Treatment
immunity, crucially depends on IL-2. Nat Rev of chronic graft-versus-host disease with
Immunol 2004; 4: 665–674. bortezomib. Blood 2014; 124: 1677–1688.
134. Nelson BH. IL-2, regulatory T cells, and 146. Naymagon L and Abdul-Hay M. Novel agents
tolerance. J Immunol 2004; 172: 3983–3988. in the treatment of multiple myeloma: a review
about the future. J Hematol Oncol 2016; 9: 52.
135. Chinen T, Kannan AK, Levine AG, et al. An
essential role for the IL-2 receptor in Treg cell 147. Busca A. The use of monoclonal antibodies
function. Nat Immunol 2016; 17: 1322–1333. for the treatment of graft-versus-host disease
136. Koreth J, Matsuoka K, Kim HT, et al. following allogeneic stem cell transplantation.
Interleukin-2 and regulatory T cells in graft- Expert Opin Biol Ther 2011; 11: 687–697.
versus-host disease. N Engl J Med 2011; 365: 148. Cutler C, Kim HT, Bindra B, et al. Rituximab
2055–2066. prophylaxis prevents corticosteroid-requiring
137. Matsuoka K, Koreth J, Kim HT, et al. Low- chronic GVHD after allogeneic peripheral blood
dose interleukin-2 therapy restores regulatory stem cell transplantation: results of a phase 2
T cell homeostasis in patients with chronic trial. Blood 2013; 122: 1510–1517.
graft-versus-host disease. Sci Transl Med 2013; 149. Teeling JL, French RR, Cragg MS, et al.
5: 179ra43. Characterization of new human CD20
138. Koreth J, Kim HT, Jones KT, et al. Efficacy, monoclonal antibodies with potent cytolytic
durability, and response predictors of low-dose activity against non-Hodgkin lymphomas. Blood
interleukin-2 therapy for chronic graft-versus- 2004; 104: 1793–1800.
host disease. Blood 2016; 128: 130–137. 150. Pidala J, Kim J, Betts BC, et al. Ofatumumab
139. Herrera AF, Kim HT, Bindra B, et al. A phase in combination with glucocorticoids for
II study of bortezomib plus prednisone for primary therapy of chronic graft-versus-host
initial therapy of chronic graft-versus-host disease: phase I trial results. Biol Blood Marrow
disease. Biol Blood Marrow Transplant 2014; 20: Transplant 2015; 21: 1074–1082.
1737–1743. 151. Piccirillo CA and Shevach EM. Naturally-
140. Mulder A, Heidt S, Vergunst M, et al. occurring CD4+CD25+ immunoregulatory T
Proteasome inhibition profoundly affects cells: central players in the arena of peripheral
activated human B cells. Transplantation 2013; tolerance. Semin Immunol 2004; 16: 81–88.
95: 1331–1337. 152. Baecher-Allan C, Brown JA, Freeman GJ,
et al. CD4+CD25high regulatory cells in
141. Neubert K, Meister S, Moser K, et al. The
human peripheral blood. J Immunol 2001; 167:
proteasome inhibitor bortezomib depletes
1245–1253.
plasma cells and protects mice with lupus-like
disease from nephritis. Nat Med 2008; 14: 153. Stephens LA, Mottet C, Mason D, et al.
748–755. Human CD4(+)CD25(+) thymocytes and
peripheral T cells have immune suppressive
142. El-Cheikh J, Michallet M, Nagler A, et al. High
activity in vitro. Eur J Immunol 2001; 31:
response rate and improved graft-versus-host
1247–1254.
disease following bortezomib as salvage therapy
after reduced intensity conditioning allogeneic 154. Sakaguchi S, Sakaguchi N, Asano M, et al.
stem cell transplantation for multiple myeloma. Immunologic self-tolerance maintained by
Haematologica 2008; 93: 455–458. activated T cells expressing IL-2 receptor
alpha-chains (CD25). Breakdown of a single
143. Mateos-Mazon J, Pérez-Simón JA, Lopez O, mechanism of self-tolerance causes various
et al. Use of bortezomib in the management autoimmune diseases. J Immunol 1995; 155:
of chronic graft-versus-host disease among 1151–1164.
multiple myeloma patients relapsing after
allogeneic transplantation. Haematologica 2007; 155. Rezvani K, Mielke S, Ahmadzadeh M, et al.
92: 1295–1296. High donor FOXP3-positive regulatory T-cell
(Treg) content is associated with a low risk of
144. Todisco E, Sarina B, Castagna L, et al. GVHD following HLA-matched allogeneic
Inhibition of chronic graft-vs-host disease SCT. Blood 2006; 108: 1291–1297.
with retention of anti-myeloma effects by
the proteasome inhibitor bortezomib. Leuk 156. Di Ianni M, Falzetti F, Carotti A, et al.
Lymphoma 2007; 48: Tregs prevent GVHD and promote immune
1015–1018. reconstitution in HLA-haploidentical

44 journals.sagepub.com/home/tah
L Hill, A Alousi et al.

transplantation. Blood 2011; 117: 168. Gómez-Almaguer D, Ruiz-Argüelles GJ, del


3921–3928. Carmen Tarín-Arzaga L, et al. Alemtuzumab
for the treatment of steroid-refractory acute
157. Brunstein CG, Miller JS, Cao Q, et al. Infusion
graft-versus-host disease. Biol Blood Marrow
of ex vivo expanded T regulatory cells in adults
Transplant 2008; 14: 10–15.
transplanted with umbilical cord blood: safety
profile and detection kinetics. Blood 2011; 117: 169. Schnitzler M, Hasskarl J, Egger M, et al.
1061–1070. Successful treatment of severe acute intestinal
graft-versus-host resistant to systemic and
158. Brunstein CG, Miller JS, McKenna DH, et al. topical steroids with alemtuzumab. Biol Blood
Umbilical cord blood-derived T regulatory cells Marrow Transplant 2009; 15: 910–918.
to prevent GVHD: kinetics, toxicity profile, and
clinical effect. Blood 2016; 127: 1044–1051. 170. Martínez C, Solano C, Ferrá C, et al.
Alemtuzumab as treatment of steroid-refractory
159. Parmar S, Liu X, Najjar A, et al. Ex vivo acute graft-versus-host disease: results of a
fucosylation of third-party human regulatory phase II study. Biol Blood Marrow Transplant
T cells enhances anti-graft-versus-host disease 2009; 15: 639–642.
potency in vivo. Blood 2015; 125: 1502–1506.
171. Schub N, Günther A, Schrauder A, et al.
160. Zorn E, Kim HT, Lee SJ, et al. Reduced Therapy of steroid-refractory acute GVHD with
frequency of FOXP3+ CD4+CD25+ CD52 antibody alemtuzumab is effective. Bone
regulatory T cells in patients with chronic graft- Marrow Transplant 2011; 46: 143–147.
versus-host disease. Blood 2005; 106: 2903–
2911. 172. Couriel D, Saliba R, Hicks K, et al. Tumor
necrosis factor-alpha blockade for the treatment
161. Jiang S, Lechler RI, He XS, et al. Regulatory of acute GVHD. Blood 2004; 104: 649–654.
T cells and transplantation tolerance. Hum
Immunol 2006; 67: 765–776. 173. Patriarca F, Sperotto A, Damiani D, et al.
Infliximab treatment for steroid-refractory acute
162. Trzonkowski P, Bieniaszewska M, Juscinska graft-versus-host disease. Haematologica 2004;
J, et al. First-in-man clinical results of the 89: 1352–1359.
treatment of patients with graft versus
host disease with human ex vivo expanded 174. Busca A, Locatelli F, Marmont F, et al.
CD4+CD25+CD127- T regulatory cells. Clin Recombinant human soluble tumor necrosis
Immunol 2009; 133: 22–26. factor receptor fusion protein as treatment
for steroid refractory graft-versus-host disease
163. Theil A, Tuve S, Oelschlagel U, et al. Adoptive following allogeneic hematopoietic stem cell
transfer of allogeneic regulatory T cells into transplantation. Am J Hematol 2007; 82: 45–52.
patients with chronic graft-versus-host disease.
Cytotherapy 2015; 17: 473–486. 175. Chiang KY, Abhyankar S, Bridges K, et al.
Recombinant human tumor necrosis factor
164. Cutler CS, Koreth J and Ritz J. Mechanistic receptor fusion protein as complementary
approaches for the prevention and treatment treatment for chronic graft-versus-host disease.
of chronic GVHD. Blood 2017; 129: Transplantation 2002; 73: 665–667.
22–29.
176. Kim JG, Sohn SK, Kim DH, et al. Different
165. Muller I, Kordowich S, Holzwarth C, et al. efficacy of mycophenolate mofetil as salvage
Application of multipotent mesenchymal treatment for acute and chronic GVHD after
stromal cells in pediatric patients following allogeneic stem cell transplant. Eur J Haematol
allogeneic stem cell transplantation. Blood Cells 2004; 73: 56–61.
Mol Dis 2008; 40: 25–32.
177. Krejci M, Doubek M, Buchler T, et al.
166. Lucchini G, Introna M, Dander E, et al. Mycophenolate mofetil for the treatment of
Platelet-lysate-expanded mesenchymal acute and chronic steroid-refractory graft-
stromal cells as a salvage therapy for severe versus-host disease. Ann Hematol 2005; 84:
resistant graft-versus-host disease in a pediatric 681–685.
population. Biol Blood Marrow Transplant 2010;
16: 1293–1301. 178. Furlong T, Martin P, Flowers ME, et al.
Therapy with mycophenolate mofetil for
167. Zhou H, Guo M, Bian C, et al. Efficacy of bone refractory acute and chronic GVHD. Bone
marrow-derived mesenchymal stem cells in the Marrow Transplant 2009; 44: 739–748.
treatment of sclerodermatous chronic graft-
versus-host disease: clinical report. Biol Blood 179. Hoda D, Pidala J, Salgado-Vila N, et al.
Marrow Transplant 2010; 16: 403–412. Sirolimus for treatment of steroid-refractory

journals.sagepub.com/home/tah 45
Therapeutic Advances in Hematology 9(1)

acute graft-versus-host disease. Bone Marrow treatment of severe steroid-refractory acute


Transplant 2010; 45: 1347–1351. graft-versus-host disease: a pilot study. Blood
2000; 96: 2426–2431.
180. Bolaños-Meade J, Jacobsohn DA, Margolis
J, et al. Pentostatin in steroid-refractory acute 184. Salvaneschi L, Perotti C, Zecca M, et al.
graft-versus-host disease. J Clin Oncol 2005; 23: Extracorporeal photochemotherapy for
2661–2668. treatment of acute and chronic GVHD in
181. Pidala J, Kim J, Roman-Diaz J, et al. Pentostatin childhood. Transfusion 2001; 4110:
as rescue therapy for glucocorticoid-refractory 1299–1305.
acute and chronic graft-versus-host disease. Ann 185. Perfetti P, Carlier P, Strada P, et al.
Transplant 2010; 15: 21–29. Extracorporeal photopheresis for the treatment
182. Jacobsohn DA, Chen AR, Zahurak M, et al. of steroid refractory acute GVHD. Bone Marrow
Phase II study of pentostatin in patients with Transplant 2008; 42: 609–617.
corticosteroid-refractory chronic graft-versus-
Visit SAGE journals online 186. Couriel DR, Hosing C, Saliba R, et al.
host disease. J Clin Oncol 2007; 25: 4255–4261.
journals.sagepub.com/ Extracorporeal photochemotherapy for the
home/tah
183. Greinix HT, Volc-Platzer B, Kalhs P, et al. treatment of steroid-resistant chronic GVHD.
SAGE journals Extracorporeal photochemotherapy in the Blood 2006; 107: 3074–3080.

46 journals.sagepub.com/home/tah

You might also like