You are on page 1of 12

Journal of Autoimmunity xxx (2016) 1e12

Contents lists available at ScienceDirect

Journal of Autoimmunity
journal homepage: www.elsevier.com/locate/jautimm

Review article

Immunomodulators in SLE: Clinical evidence and immunologic


actions
L. Durcan a, *, M. Petri b
a
Division of Rheumatology, University of Washington, Seattle, USA
b
Division of Rheumatology, Johns Hopkins University, School of Medicine, Baltimore, USA

a r t i c l e i n f o a b s t r a c t

Article history: Systemic lupus erythematosus (SLE) is a potentially fatal autoimmune disease. Current treatment stra-
Received 1 June 2016 tegies rely heavily on corticosteroids, which are in turn responsible for a significant burden of morbidity,
Received in revised form and immunosuppressives which are limited by suboptimal efficacy, increased infections and malig-
16 June 2016
nancies. There are significant deficiencies in our immunosuppressive armamentarium, making immu-
Accepted 21 June 2016
Available online xxx
nomodulatory therapies crucial, offering the opportunity to prevent disease flare and the subsequent
accrual of damage. Currently available immunomodulators include prasterone (synthetic dehy-
droeipandrosterone), vitamin D, hydroxychloroquine and belimumab. These therapies, acting via
Keywords:
SLE
numerous cellular and cytokine pathways, have been shown to modify the aberrant immune responses
Immunomodulation associated with SLE without overt immunosuppression.
Hydroxychloroquine Vitamin D is important in SLE and supplementation appears to have a positive impact on disease
Vitamin D activity particularly proteinuria. Belimumab has specific immunomodulatory properties and is an
Dehydroeipandrosterone effective therapy in those with specific serological and clinical characteristics predictive of response.
Belimumab Hydroxychloroquine is a crucial background medication in SLE with actions in many molecular path-
ways. It has disease specific effects in reducing flare, treating cutaneous disease and inflammatory ar-
thralgias in addition to other effects such as reduced thrombosis, increased longevity, improved lipids,
better glycemic control and blood pressure. Dehydroeipandrosterone is also an immunomodulator in SLE
which can have positive effects on disease activity and has bone protective properties.
This review outlines the immunologic actions of these drugs and the clinical evidence supporting their
use.
© 2016 Elsevier Ltd. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
2. Dehydroeipandrosterone . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
3. Vitamin D . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
4. Hydroxychloroquine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
5. B cell therapies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
6. Stem cell transplantation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
7. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
Support . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00

1. Introduction

* Corresponding author. Systemic lupus erythematosus (SLE) is a chronic, multisystem,


E-mail address: laurajanedurcan@hotmail.com (L. Durcan).

http://dx.doi.org/10.1016/j.jaut.2016.06.010
0896-8411/© 2016 Elsevier Ltd. All rights reserved.

Please cite this article in press as: L. Durcan, M. Petri, Immunomodulators in SLE: Clinical evidence and immunologic actions, Journal of
Autoimmunity (2016), http://dx.doi.org/10.1016/j.jaut.2016.06.010
2 L. Durcan, M. Petri / Journal of Autoimmunity xxx (2016) 1e12

autoimmune condition characterized by the presence of autoanti- prasterone.


bodies to nuclear material and immune complex deposition in Normal serum levels of dehydroeipandrosterone range from 1 to
involved tissues. Whilst numerous advances have been made in 50 nM. During fetal development, plasma dehydroepiandrosterone
unraveling the pathogenesis of this complex disease, it remains sulphate levels are 100 200 mg/dL (3 7 mM), falling rapidly after
incompletely understood. A multitude of cell types and molecules, birth and remaining low until adrenarche. Levels then increase
participating in many cellular mechanisms have been implicated in rapidly, followed by an age related decline [4]. This decline is
SLE. Abberancies in apoptotic pathways and in innate and adaptive possibly mediated by decrease in 17,20-lyase activity [5]. The rate of
immune mechanisms are found in patients with SLE, with genetic, decline of blood levels is in the region of 2% per year, by the 8th-9th
epigenetic, environmental and hormonal factors known to decade residual levels are 10e20% of their peak [6]. There are
contribute to the disease. There are a number of central events in gender differences to consider with higher levels in males [7]. In
the development of SLE, these include increased production of addition to these considerations there are genetic variations.
autoantibodies during apoptosis, decreased clearance of cellular Genome-wide association studies (GWAS) indicate that serum
debris with dysregulated handling and presentation. Subsequent levels of dehydroepiandrosterone sulphate are regulated at
disease activity and tissue damage is mediated by autoantibodies, approximately 60% by genotypes near these genes: BCL2L11,
immune complexes and complement activation with numerous ZKSCAN5, ARPC1A,TRIM4, HHEX, CYP2C9, BMF, and SULT2A1[8].
cytokine and interferon pathways implicated. The complexity of Dehydroeipandrosterone does not have a specific receptor. It
these disease mechanisms have meant that there are a multitude of can bind to steroid hormone receptors (reviewed by Triash et al. [9],
possible targets for immunomodulation in SLE. However, at pre- and by Webb et al. [5]) pregnane X receptor/steroid and xenobiotic
sent, there are few tools in our therapeutic armamentarium which receptor (PXR/SXR, NR1I2) [5]; estrogen receptors a and b,
can be considered immunomodulatory. For the most part, we rely androgen receptors [10]; peroxisome proliferator activated re-
on immunosuppressives, in particular for organ specific disease. ceptors [5]; and pregnane X receptor [11]. At most of these sites,
Improvements have been made in pharmacotherapy over the dehydroeipandrosterone acts as a partial agonist with weak affinity
past 50 years which have positively impacted upon the prognosis of due to competition for binding. Taking into account the fact that
SLE although, disappointingly, poor renal outcomes [1,2], cardio- dehydroeipandrosterone is itself a precursor for many of the higher
vascular disease and the accumulation of organ damage often affinity molecules, it is difficult to estimate the degree to which
incited by high dose prednisone remain major challenges. Thera- dehydroeipandrosterone itself is effective.
peutic advances include anti-malarials, corticosteroids, immuno- The principal regulator of dehydroeipandrosterone production,
suppressives, ace inhibitors, antibiotics, B-cell therapies, vitamin D is adrenocorticotropic hormone. This in turn depends on cortico-
supplementation and dehydroeipandrosterone (DHEA). Despite tropin releasing hormone of hypothalamic origin for regulation [3].
these therapies SLE continues to associate with premature mor- In adults, dehydroeipandrosterone levels peak in the morning,
tality and morbidity. Current strategies rely heavily on the immu- following the circadian pattern of ACTH secretion [12]. The bio-
nosuppressive properties of corticosteroids to control logical effects of dehydroeipandrosterone can be considered both
inflammation. Chronic and high dose corticosteroids associate with androgenic and estrogenic since it is a precursor of both. Labrie
significant morbidity and are responsible for much of the long-term et al. suggest that more than 30% of total androgen in men and over
damage accrual in SLE. Other immunosuppressives, such as myco- 90% of estrogen in postmenopausal women are derived from pe-
phenolate mofetil, methotrexate and azathioprine, are essential in ripheral conversion of dehydroeipandrosterone [13]. Elevated
the management of organ specific disease, however they are dehydroeipandrosterone contributes to disorders associated with
limited by efficacy, in particular in renal disease. hyperandrogenic states such as in polycystic ovarian disease and
Immunomodulating therapies that are not immunosuppressive, non-classical 21-hydroxylase deficient congenital adrenal hyper-
are a more attractive therapeutic option, offering the opportunity plasia [14]. Low levels have been associated with many age related
to modify the aberrant immune responses in SLE and thus prevent disorders and with multiple autoimmune conditions, including SLE.
inflammation and subsequent damage without the risks of infec- Women with SLE have been shown in numerous studies,
tion and malignancy. Current strategies, considered to have reviewed by McMurray et al., to have significantly depressed con-
immunomodulating properties, include hydroxychloroquine (and centrations of androgens and elevated levels of estradiol compared
other antimalarials), vitamin D, dehydroeipandrosterone and with both males with SLE and healthy controls [15]. In female pa-
certain B cell therapies. Stem cell transplantation is as of yet un- tients with SLE, levels of both dehydroeipandrosterone and dehy-
proven in randomized controlled studies for SLE but offers a droepiandrosterone sulphate are low [15e17]. Lahita et al.
fascinating perspective on immunomodulation and may, in the demonstrated low levels of all androgens in females with SLE with
future, be a therapeutic option for those with severe, life threat- the lowest amount of both metabolites in those with active disease
ening disease. Here we review current immunomodulating stra- [16]. The fact that SLE is commonly treated with corticosteroids has
tegies in SLE, their clinical efficacy and examine their mechanisms been considered to be a confounding factor due to inhibitory
of action. feedback mechanisms. However, steroid naïve SLE patients have
also been shown to have low levels of dehydroeipandrosterone
2. Dehydroeipandrosterone [16].
Dehydroeipandrosterone exerts anti-proliferative and anti-
Dehydroeipandrosterone is a weak androgenic steroid and with inflammatory effects, and modulates immune function. Praster-
its metabolite, dehydroepiandrosterone sulphate (DHEAS), is the one (synthetic dehydroeipandrosterone) therapy has been shown
most abundant adrenal steroid hormone. Dehydroeipandrosterone in small studies to be beneficial in depression [18] with promise in
is a precursor of both androgens and estrogens and is synthesized the management of the negative symptoms of schizophrenia
primarily by the adrenal cortex (zona reticularis) from 17 a- [19,20]. There is little evidence to lend support to the theory that it
hydroxypregnenolone. It can then be sulphated, at the 3b’-hydroxyl may have anti-aging effects. As it is known to have some andro-
group, into dehydroepiandrosterone sulphate in the adrenals and genic properties, supplementation has been associated with mild
in peripheral tissues, dehydroeipandrosterone is also metabolized virilization, acne, voice changes and terminal hair growth.
further into more active steroids including androstenedione, There is evidence that dehydroeipandrosterone has activity on
testosterone and estrogen [3]. In its drug form it is called multiple cytokine and immunologic pathways. Numerous studies

Please cite this article in press as: L. Durcan, M. Petri, Immunomodulators in SLE: Clinical evidence and immunologic actions, Journal of
Autoimmunity (2016), http://dx.doi.org/10.1016/j.jaut.2016.06.010
L. Durcan, M. Petri / Journal of Autoimmunity xxx (2016) 1e12 3

have reported improvements in immune function with dehy- subjects; 45 in the placebo group, 47 in the 100 mg group, and 45 in
droeipandrosterone supplementation including regulation of the the 200 mg group, 29%, 38%, and 51%, respectively, were responders
production of pro-inflammatory cytokines such as IL-2, IL-1, IL-6 (P ¼ 0.031 for 200 mg versus placebo).
and TNFa [4,21,22]. There is also some evidence that dehy- Two studies by Chang et al. evaluated dehydroeipandrosterone
droeipandrosterone can modulate the proinflammatory cytokine supplementation, firstly reporting on a multicenter international
profile associated with SLE, in particular IL-10 levels have been study evaluating 120 participants with mild to moderate SLE
shown to decrease with supplementation [22]. [22,33]. This work demonstrated no significant reduction in disease
In a murine SLE model (NZBA ~ NZW), decreased severity of activity measured by SLE activity measure (SLAM), but improve-
lupus-like disease was demonstrated with the administration of ments were seen in flare, with a 16% reduction, and in the patient
dehydroeipandrosterone with data pointing toward decreased global score, which decreased significantly (in keeping with an
antibody production [23]. However, the improved survival in SLE improved self-reported assessment of disease activity). This was
mouse models was demonstrated only in those who were supple- followed by the analysis of cytokines in a subgroup from a single
mented at 2 months and not in those who received synthetic center of Chinese patients. They observed no change in IL-1 and
dehydroeipandrosterone at 6 months [24], leading investigators to reduced IL-10 with many cytokines proving to be undetectable in
question whether there is a crucial point in SLE pathogenesis at this work.
which hormonal imbalances trigger a loss of self-tolerance. It is yet A subsequent large, multicenter, randomized, double-blind,
to be established in humans whether dehydroeipandrosterone placebo-controlled trial evaluating prasterone supplementation
levels are reduced before clinical disease onset. demonstrated no difference in SLEDAI or SLAM in an intention to
Dehydroeipandrosterone levels have been found to correlate treat analysis, but when those with disease activity, defined as
negatively with IL-6 [25] which is known to play an important role SLAM >7, SLEDAI >2, were considered there were more responders
in immune regulation and inflammation in both healthy in- in those who received dehydroeipandrosterone supplementation
dividuals and in SLE, amongst other autoimmune diseases. IL-6 is a compared with placebo (58.5% versus 44.5% (P ¼ 0.017)). There was
pleiotropic cytokine strongly implicated in particular in lupus less flare in the dehydroeipandrosterone group (with SLEDAI >2)
nephritis [26,27] and arthritis [28]. It shares several activities with and less worsening of patient global (10.9% versus 22.6%,
IL-1 and tumor necrosis factor alpha, which have also been impli- P ¼ 0.007).
cated in SLE, in the induction of pyrexia and the production of acute Consideration has also been given to whether dehydroeipan-
phase proteins. Whether IL-6 changes with supplementary DHEA in drosterone supplementation could influence bone metabolism.
SLE has not been evaluated. Hartkamp and colleagues addressed this question and found no
IL-10 is also implicated in the pathogenesis of SLE and serum change in overall bone mineral density in women, both pre and
from patients with SLE has been shown to stimulate IL-10 pro- post menopausal, with quiescent disease taking less than 10 mg
duction from peripheral blood mononuclear cells [29]. Chang et al. prednisone per day [34]. However, in those who were post-
evaluated changes in cytokine profiles in females with active SLE menopausal there was a mean increase in bone density of 1.80%
participating in a randomized controlled trial of 200 mg prasterone with prasterone therapy compared with a decrease of 2.32% in the
compared with placebo [22]. The levels of cytokines including placebo group. This suggested a protective effect on bone mineral
interleukin IL-1, IL-2, IL-4, and Il-10 were determined. A significant density in postmenopausal women with SLE. Mease et al. also
reduction in IL-10 was demonstrated in those taking dehy- evaluated the effect of prasterone on bone mineral density [35].
droeipandrosterone supplementation. The other cytokines were Significant differences between treatment groups (200 mg pras-
either undetectable, or in the case of IL-1, there was no difference terone and placebo) for percentage change in bone mineral density
demonstrated. for both the lumbar spine and total hip were present. At the lumbar
These observations led a number of investigators to evaluate the spine, there was a mean gain in bone mineral density of 1.7% in the
therapeutic utility of supplemental dehydroeipandrosterone in prasterone group compared to a mean loss of 1.1% in the placebo
women with SLE. The details of these clinical trials are outlined in group (p ¼ 0.003 between groups). At the hip, the mean gain was
Table 1. 2.0% with prasterone compared to a mean loss of 0.3% in the pla-
Prasterone therapy was first formally evaluated and reported cebo group (p ¼ 0.013). Among those who were postmenopausal,
upon in SLE by van Vollenhoven [30] in an uncontrolled, open- the mean bone density of the lumbar spine increased by 3.1% in the
label, single center study involving 10 patients in reciept of prasterone group compared to a decrease of 1.7% in the placebo
200 mg per day. No significant difference in SLE disease activity group (p ¼ 0.012 between groups). Sa nchez-Guerrero et al. [36] also
index (SLEDAI) was demonstrated. However, there was a significant evalauted the effect of supplementary dehydroepiandrosterone on
improvement in the physician global assessment of disease activity bone mineral density. This was a randomized controlled trial which
(PGA). This study was followed by a double-blind placebo had three arms; 200 mg prasterone, 100 mg and placebo. There was
controlled study involving 21 patients with ‘severe’ disease [31]. dose-dependent increase in bone mineral density at the lumbar
The primary end-point was clinical response, which was defined spine (at 18 months) in patients who received 200 versus 100 mg
prospectively. Patients were considered to be responders if they prasterone (p ¼ 0.021). For patients who received 200 mg, the gain
demonstrated stabilization of their major clinical manifestation at at the lumbar spine was 1.083± 0.512% (p ¼ 0.042). There was no
six months (as defined in the protocol). change in bone mineral density at the hip over 18 months with
In a subsequent multi-center, double blind randomized placebo- prasterone treatment.
controlled trial, 191 patients were randomized to receive praster- Dehydroeipandrosterone has been shown to improve fatigue in
one, either 100 mg, 200 mg or placebo [32]. This trial was under- other chronic diseases. Nordmark et al. evaluated the effect of
taken to evaluate the possibility that dehydroeipandrosterone supplementary dehydroeipandrosterone on fatigue and depression
supplementation could have steroid-sparing properties. The pri- in SLE. There was no difference demonstrated between the dehy-
mary end point was reduction in prednisone (or corticosteroid droeipandrosterone group and placebo, although interestingly,
equivalent) dose. There was no difference demonstrated between when those who believed they were taking prasterone were
the three groups. However, differences were demonstrated when considered there was a significant difference compared to those
patients with quiescent disease were excluded. In the group with who thought they were in the placebo arm [37].
disease activity (defined as SLEDAI score >2), comprised of 137 There are conclusive data that low blood levels of

Please cite this article in press as: L. Durcan, M. Petri, Immunomodulators in SLE: Clinical evidence and immunologic actions, Journal of
Autoimmunity (2016), http://dx.doi.org/10.1016/j.jaut.2016.06.010
Table 1
Clinical Trials in SLE involving Dehydroeipandrosterone.

Author & year Design N Patients Dose þ time Outcome Result

van Vollenhoven Single center 10 Female 200 mg SLEDAI SLEDAI: decreased (non
et al., 1994 [30] Open label Patient assessment significant)
Uncontrolled (0e100) Physician assessment:
Physician improved (P ¼ 0.04)
assessment (0 Patient assessment: unchanged
e100)
van Vollenhoven Number of centers 21 Female‘Severe’ 200mg/placebo ‘Responder Responder: DHEA 7/9 achieved
et al., 1999 [31] not stated, in San 6 months, analysis’ response, placebo 4/10 (non-
Fransisco area. 6 months open SLEDAI significant).
Double-blind label extension SLAM SLEDAI: No significant change
Randomized BMD SLAM: No significant change
Placebo-controlled BMD: Non significant increase
in lumbar spine.
Petri et al., 2002 Multi-center 191 Female Prednisone 100mg/200mg/ Sustained No reduction in prednisone
[32] Double-blind 10e30 mg placebo reduction <7.5 mg dose.
Randomized 7-9 months prednisone 200 mg: 55% responder
Placebo-controlled SLEDAI 100 mg: 44%
SF-36 Placebo:41%
KFSS SF-36: Non significant
Damage index KFSS: Non significant change
Chang et al., 2002 Multicenter 120 Female 200 mg/placebo SLAM SLAM: Non significant
[33] Randomized 24 weeks Flare Flare: 16% decrease
Double blind SLEDAI Patient global: Improved 5.5
Placebo controlled versus 5.4; P ¼ 0.005
Chang et al., 2004 Single center 32 Female Active SLE 200mg/placebo Il 10
[22] Randomized Chinese Il 1b
Double blind TNFa IL 10: Reduced (9.21e1.89 pg/
Placebo controlled ml with DHEA) IL1ß:
(subgroup of Chang UnchangedTNFa: Undectable
et al., 2002(22))
Petri et al., 2004 Multicenter 381 Female 200mg/placebo SLEDAI ITT analysis: No difference
[145] Randomized SLAM >7 SLAM SLEDAI: >2 Response: 58.5%
Double-blind SLEDAI KFSS versus 44.5% (P ¼ 0.017)
Placebo-controlled >2 (n ¼ 293) Patient global Flare: Less with SLEDAI>2
Time to flare Patient global: Less worsening
SLEDAI: Less worsening
Hartkamp et al., 2 centers 58: Initial DEXA Female 200 mg/placebo BMD BMD: No significant difference
2004 [34] Randomized 56: Both baseline 10 mg prednisone 12 months SLEDAI (Postmenopausal mean change
Double-blind and follow up DEXA was 1.80% with DHEA,
Placebo-controlled 2.32% with placebo.
Premenopausal: No change)
SLEDAI: No change
Mease et al., 2005 Multicenter 66: Initial DEXA Female 200mg/placebo BMD (% change) BMD: Lumbar spine, gain in
[35] Double- blind 55: Both baseline SLE 12 months SLEDAI BMD of 1.7% in versus loss of
Randomized and follow up DEXA 10 mg SLAM 1.1% (P ¼ 0.003).
Placebo-controlled prednisone: (6 KFSS Total hip, 2.0% gain versus a loss
months) SLICC damage of 0.3% with placebo
index (p ¼ 0.013)
Patient VAS No significant change in other
Physician VAS outcomes
Nordmark et al., 2 centers 41 Female 200mg/placebo SF-36 SF-36: Improved emotional
2005 [37] Double-blind >5 mg prednisone 6 months with 6 BMD components
Randomized Age 20-65 month open Body composition BMD: No increase
Placebo-controlled extension SLEDAI/mSLEDAI Body composition: Increased
MCS waist hip ratio
HSCL-56 SLEDAI: No change
PGWB mSLEDAI: No change
MCS: Improved
(unsustained)
HSCL-56: Improved
PGWB: Nonsignificant
Hartkamp et al., 2 centers 60 Female 200mg/placebo Fatigue: MFI Fatigue: No change
2010 [146] Double-blind No prednisone 12 months Depression: Zung Depression: No change
Randomized self-rating scale Well-being: No change
Placebo-controlled SF-36
Pain VAS
SLEDAI
nchez-Guerrero
Sa Multicenter 155 Female >5 mg 200mg/placebo in BMD BMD: Increased at lumbar
et al., 2008 [36] Double-blind prednisone phase 1, then spine, 200 mg dose, (non-
Randomized No osteoporosis/ 200mg/100 mg in significant at 6 months,
Placebo-controlled bisphosphonate extension phase significant at 18 months).
Calcium Maintenance of BMD at hip.
Vitamin D (100 mg no effect)

SLEDAI: Systemic lupus erythematosus disease activity index, DHEA: Dehydroeipandrosterone SLAM: Systemic lupus activity measure, BMD: Bone mineral density, SF-36:
Short form health survey, KFSS: Krupps fatigue severity scale, DEXA: Dual-energy X-ray absorptiometry, mSLEDAI: Modified systemic lupus erythematosus disease activ-
ity index, MCS: Mental component summary, HSCL-56: Hopkins symptom check list, PGWB: Psychological general wellbeing index, MFI: Multidimensional fatigue inventory,
VAS: Visual analogue scale.

Please cite this article in press as: L. Durcan, M. Petri, Immunomodulators in SLE: Clinical evidence and immunologic actions, Journal of
Autoimmunity (2016), http://dx.doi.org/10.1016/j.jaut.2016.06.010
L. Durcan, M. Petri / Journal of Autoimmunity xxx (2016) 1e12 5

dehydroeipandrosterone associate with disease activity in SLE in Medicine as <20 ng/mL and insufficiency as 21e29 ng/mL [44].
numerous populations and various age groups. Clinical trials have, Although there is no consensus in SLE specifically on the optimum
for the most part, demonstrated improvements in disease activity vitamin D level, for adult patients at risk of fractures, falls, auto-
in those with disease activity, although these studies are difficult to immune disease or cardiovascular disease, a 25(OH)D level of at
pool due to differences in dosing regimens and time-frames. Safety least 30e40 ng/ml has been recommended [45]. Given the risks of
data from these studies were reassuring. Supplementation has not skin cancer associated with UV exposure, (and in SLE, the risk of
been evaluated in male patients, in whom deficiency is uncommon disease flare) supplementation is via the oral route. Serum levels
relative to females with SLE. There is no evidence to suggest that higher than 150 ng/mL have been associated with vitamin D
males would benefit from prasterone therapy. Further, in post intoxication characterized by hypercalcemia, hypercalciuria, and
menopausal women there is concern that the administration of an calcifications [45].
exogenous source of estrogen could increase the risk of hormone Vitamin D insufficiency and deficiency have been implicated in
sensitive malignancies such as uterine and breast cancer. certain malignancies, cardiovascular disease and many autoim-
Evidence in human SLE for the immunomodulating properties mune conditions, including SLE, rheumatoid arthritis and multiple
of dehydroepiandrosterone are at present limited to improvements sclerosis [41,46e49]. In general populations, vitamin D insuffi-
in some cytokine profiles. It is however likely based on animal data ciency is common and increases in prevalence with distance from
that there are other mechanisms impacted upon by prasterone the equator, although cultural practices in which clothing
therapy. There is promising data that disease activity is improved completely shields the skin makes deficiency commonplace,
by supplementing dehydroepiandrosterone, this effect is demon- regardless of latitude [41]. The prevalence of low levels of vitamin D
strated in particular in patient reported measures, which are in SLE has been reported, in both adults and children in multiple
crucially important in this chronic disease. Evidence also exists that ethnic populations at different times of the year, at between 36.8%
dehydroeipandrosterone can protect against bone loss. As fragility and 75% [49e55]. The reasons for these low levels have not been
fractures are the most commonly reported item on the American fully elucidated. Decreased UV exposure in those with SLE
College of Rheumatology/Systemic Lupus International Collabo- compared with controls and the influence of medications, predni-
rating Clinics damage index [38], bone health is of crucial impor- sone and antimalarials, have been implicated without conclusion.
tance in this population. The time frame for the bone density The presence of photosensitivity, which may imply enhanced sun
studies is relatively short. It is likely that longer durations of ther- avoidance behaviors, has been found to associate with critically low
apy and follow-up are necessary to determine if these changes are levels [52]. Renal disease is also a significant contributing factor.
meaningful and to establish whether they translate into a reduction The presence of renal disease was found, by Kamen et al. to be the
in fractures. At present, there are insufficient data to determine strongest predictor of deficiency with an odds ratio of 13.3 [52].
whether dehydroeipandrosterone has any influence on fatigue in There are also general differences in metabolism in individuals with
SLE. darker skin due to decreased cutaneous conversion of vitamin D to
its more active form following UVB exposure [56]. As SLE is more
3. Vitamin D common and more severe in non-caucasians this has also been
thought to influence levels. It does not appear that dietary intake is
Vitamin D, 1,25-dihydroxyvitamin D, is a steroid hormone, different in SLE compared to control populations [57].
principally known for its roles in bone health and calcium ho- In its active form, vitamin D has numerous immunologic func-
meostasis, now also recognized for its immunomodulatory prop- tions mediated through binding to specific nuclear vitamin D re-
erties. The chemical structure of vitamin D and its role in the ceptors. These are present in most cells of the innate and adaptive
metabolic bone disease, rickets, were first described in the 1930’s immune system. Vitamin D receptors are expressed in monocytes,
[39]. Rickets, in children, and its adult equivalent, osteomalacia, are activated macrophages, dendritic cells, natural killer cells, T and B
caused by very low levels of vitamin D. This is, for the most part, cells. Activation of these receptors has potent anti-proliferative,
due to inadequate UV exposure. Malabsorption syndromes, renal pro-differentiative, and immunomodulatory functions which can
failure and poor intake can also play a role. As vitamin D is a fat- both suppress and enhance the immune response.
soluble vitamin there is evidence that certain GI surgeries, In vitro, vitamin D blocks B cell proliferation and differentiation,
including gastric bypass, decrease absorption [40]. and suppresses immunoglobulin production [58,59]. It can atten-
In humans, vitamin D is mainly synthesized in the skin uate the expression of pro inflammatory cytokines, induced by
following ultraviolet B (UVB) exposure (wavelength, 290e315 nm) stimulation of toll-like receptor 3,4, 7 and 8 [60].
with a minority coming from dietary sources (<10%) [41]. It has two It can decrease T cell proliferation and shift maturing T cells
major forms, firstly ergocalciferol (commonly known as vitamin away from Th1 toward Th2 and regulatory T cell phenotypes.
D2), acquired from ultraviolet (UV) irradiation and secondly, Vitamin D has been shown to suppress dendritic cell differentia-
cholecalciferol (known as vitamin D3), which is made in the skin tion. This is of particular importance in autoimmune conditions,
and acquired in food sources [41]. Both D2 and D3 forms can be including SLE due to the central role played by dendritic cells in the
used for food fortification and supplementation. Vitamin D is bio- maintenance of self-tolerance [61]. Aberrant interferon production
logically inert and requires hydroxylation, by D-25- hydroxylase is also implicated in SLE disease activity and pathogenesis. Low
(25-OHase) to 25-hydroxyvitamin D (25(OH)D). This represents the levels of vitamin D have been associated with an increased inter-
major circulating vitamin D metabolite and is the most reliable feron gene signature in patients with SLE (via the inhibition of
parameter in establishing levels [42]. 25(OH)D then requires a dendritic cell maturation) [62]. Contrarily, vitamin D supplemen-
further hydroxylation step, by 25(OH)D-1-OHase, to form its bio- tation did not diminish the interferon signature in a placebo-
logically active form which is 1,25-dihydroxyvitamin D (1,25(OH) controlled study [63]. In further support of vitamin D as an
2D). This process is controlled by parathyroid hormone and the immunoregulator are the findings that vitamin D supplementation
phosphaturic hormone fibroblast growth factor [43]. associates with down regulation of the Th1 immune response and
The highest concentration of 25(OH)D, in humans, is in plasma the proliferation of activated B cells with up regulation of regula-
(usually measured in serum), but the largest pool of 25(OH)D is in tory T cells.
adipose tissue and muscle. In the general population, for skeletal A suggestion for the immunomodulatory effects of vitamin D in
health, vitamin D deficiency has been defined by the Institute of human SLE arises from a large body of data demonstrating an

Please cite this article in press as: L. Durcan, M. Petri, Immunomodulators in SLE: Clinical evidence and immunologic actions, Journal of
Autoimmunity (2016), http://dx.doi.org/10.1016/j.jaut.2016.06.010
6 L. Durcan, M. Petri / Journal of Autoimmunity xxx (2016) 1e12

inverse relationship between serum levels and disease activity. whereby vitamin D exerts this effect is incompletely understood,
Observational studies have, in the majority of cases, demonstrated but it may be the result of an increase in T regulatory cells. Vitamin
an inverse relationship between serum vitamin D levels and SLE D is a safe therapy in SLE and therapy should be considered
disease activity, as measured by SLEDAI [64e69], SLAM[70] and essential in those with deficiency and insufficiency. Whether
British Isles Lupus Assessment Group (BILAG) [71] (in diverse vitamin D supplementation can impact upon cardiovascular out-
populations at varying latitudes). The interventional studies, which comes will require further study.
have evaluated the effect of supplementation, are outlined in
Table 2. 4. Hydroxychloroquine
Most data indicate a modest beneficial effect on disease activity
with vitamin D supplementation (see Table 3). This effect is difficult Hydroxychloroquine is an essential tool in the medical man-
to quantify as studies in SLE utilize many different supplementation agement of SLE with numerous disease-specific and longitudinal
protocols, over various time frames. Supplementation regimens benefits [83e89]. It appears to work through numerous mecha-
varied from 600 iu to 7500 iu daily (50,000 iu weekly in addition to nisms in SLE, mediating subtle immunomodulation without
daily 400 iu). Ruiz- Irastorza et al. found that 600e800 iu over 24 causing immunosuppression. Hydroxychloroquine has been shown
months had no effect on disease activity measured by SLEDAI [72]. in multiple, diverse, SLE populations to associate with improved
Higher doses were utilized in a subsequent study by Terrier and survival [84,90,91] and specifically has been shown to be effective
colleagues [73]. The SLEDAI decreased (2.9 ± 2.5 to 2.6 ± 2.5) which in the treatment of cutaneous disease [92], arthritis [87], with an
was not statistically significant. In the Hopkins Lupus Cohort, there augmenting effect on the efficacy of mycophenolate mofetil in the
was 0.2 improvement in SLEDAI per each 20 ng/ml increase in management of nephritis [89]. Further, hydroxychloroquine has
vitamin D [74]. The dose of vitamin D was 50,000 iu per week with been shown to decrease thrombosis in those with positive anti
400 iu per day. Although statistically significant, the change in phospholipid antibodies [93e95] and to improve pregnancy out-
disease activity is small and of unclear clinical relevance. The comes for women with SLE, with and without antiphospholipid
physician global assessment of disease activity also improved with antibodies [86,96,97]. In addition to disease specific benefits in SLE,
a modest improvement (4%) in proteinuria as measured by the hydroxychloroquine has been shown to have lipid lowering prop-
urine protein to creatinine ratio. In an Egyptian population erties [98,99], anti-thrombotic effects [100] and hypoglycemic ac-
receiving 2000 iu daily, serum levels correlated with disease ac- tions [101]. It can decrease progression to SLE in undifferentiated
tivity [75] and in juvenile SLE, 50,000 iu per week associated with a connective tissue disease [102] and in women with the Ro (SSa)
decrease in disease activity [76]. As there were no serious adverse antibody it decreases the risk of congenital heart block [103,104].
events reported in these studies and the safety of vitamin D has Hydroxychloroquine is an antimalarial medication. This class in-
been widely reported upon, we consider vitamin D an essential, cludes chloroquine and quinacrine, which can also be used in the
safe therapy which has, at least, a modest beneficial effect on dis- treatment of SLE. As hydroxychloroquine is the cornerstone of the
ease activity. medical management of SLE with a multitude of known benefits,
The mechanisms whereby vitamin D exerts these effects are we will focus on this drug.
incompletely understood. An increase in regulatory T cells with The mechanism of action of antimalarial drugs in SLE includes
supplementation was found by Andreoli et al. [77] and by Terrier many molecular pathways [105e109]. Hydroxychloroquine is a
et al. [73]. There was no change in interferon signature demon- weak base is thought to work, in part by increasing lysosomal pH in
strated with supplemental vitamin D [63]. It was theorized antigen presenting cells. This interferes with phagocytosis and
vitamin D would impact upon was fatigue. Fatigue, measured by causes disruption in the presentation of self-antigens [110,111]. T
visual analogue scale was found to improve with vitamin D sup- cell responses have been shown to be altered by these medications
plementation at 400e600 iu daily, but in adolescents, fatigue did and numerous cytokines are inhibited (IL-1, Il-2, Il-6, IL-17, IL-22,
not improve with therapy (measured by the kids fatigue severity interferon alpha and tumor necrosis factor alpha) [105e109]. The
scale). beneficial effects of hydroxychloroquine, in particular, may be via
Vitamin D deficiency has been associated with increased car- the inhibition of toll-like receptor activation. The endosomal acid-
diovascular disease in the general population [78e80]. Patients ification resulting from hydroxychloroquine therapy results in
with SLE are also known to have enhanced cardiovascular risk, decreased signaling of toll-like receptors 3,7,8 and 9 [112]. In turn
which contributes to mortality. The relationship, in SLE, between the reduced toll-like receptor signaling results in decreased acti-
vitamin D and cardiovascular risk factors has been evaluated, vation of dendritic cell and the reduction of interferon production
including in a large international inception cohort [69]. It was [113], amongst other mechanisms [100].
found that those with replete vitamin D levels were less likely to Hydroxychloroquine is effective in the treatment of SLE. Much of
have hypertension and dyslipidemia. Adverse lipid profiles have the data associating hydroxychloroquine with improved outcomes
also been demonstrated with low vitamin D levels [68]. Both Wu arises from observational studies. The clinical trials of hydroxy-
et al. (2009) and Reynolds et al. (2012) have also demonstrated a chloroquine in SLE are outlined in Table 2. In a randomized, double
significant association with insulin resistance and low vitamin D. It blind drug withdrawal study, patients were either continued on
is unclear whether there is data to link low vitamin D levels and the therapy or switched to a placebo. The risk of flare in those switched
development of carotid plaque as the results of Reynolds et al. [67] to placebo increased by 2.5 with withdrawal of hydroxychloroquine
and Ravenell et al. [81] conflict. It has also been recently reported [114]. However, contrarily, it did not reduce flares in a large trial
that vitamin D supplementation improves endothelial repair evaluating belimumab [115] and data are conflicting regarding the
mechanisms, dysregulation of which may contribute to the attainment of therapeutic hydroxychloroquine blood levels and
enhanced cardiovascular risk associated with SLE [82]. disease control. Costedoat-Chalumeau et al. found lower hydroxy-
Vitamin D has long been known for its importance in bone chloroquine level in those with active disease and that lower
health. There is now also ample evidence that vitamin D is baseline levels were predictive of flare [116]. In those with cuta-
important in SLE. The data indicate a modest effect on disease ac- neous lupus, hydroxychloroquine levels were significantly higher in
tivity with perhaps a greater impact on renal parameters. This is of patients with complete remission [117]. However, no relationship
particular importance as poor renal outcomes are a major challenge between blood levels and SLE flare was found in a subsequent
with current immunosuppressive therapies. The mechanism clinical trial [118]. In the Hopkins Lupus Cohort, there was a

Please cite this article in press as: L. Durcan, M. Petri, Immunomodulators in SLE: Clinical evidence and immunologic actions, Journal of
Autoimmunity (2016), http://dx.doi.org/10.1016/j.jaut.2016.06.010
L. Durcan, M. Petri / Journal of Autoimmunity xxx (2016) 1e12 7

Table 2
Vitamin D supplementation studies in SLE.

Author & year Design N Patients þ location Dose Outcome Result

Ruiz-Irastorza et al., Longitudinal 80 SLE 600-800iu/day SLEDAI Fatigue: VAS, 4.1 versus 3.3 P ¼ 0.015.
2010 [72] Observational Spain 24 months SDI SLEDAI: No effect
(prospective- Fatigue SDI: No effect
cohort) (VAS)
Single center
Terrier et al., 2012 Single center 20 SLE 100,000Iu weekly for 4 Safety SLEDAi: 2.9 ± 2.5 to 2.6 ± 2.5 at 2
[73] Open-label France weeks, then 100,000 SLEDAI months, non significant.
monthly for 6 months B cells Anti dsDNA: Decreased at 2 and 6
T cells months.
Cytokines CD4: Non significant increase
CD*: Decreased in frequency but not in
number.
T regs: Increased
Petri et al., 2013 Longitudinal 1006 SLE 50,000 iu weekly þ 400 iu SLEDAI SLEDAI: Significant decrease.
[74] Cohort USA calcium/vitamin D daily Physician global (0 Physician global: Improved significantly
(37% African e3) UPCR: 20-ng/ml
American) UPCR increase in the 25(OH)D value was
associated with a 4% decrease in UPCR
Abou-Raya et al., Randomized (2:1) 267 SLE 2000iu daily/placebo SLEDAI SLEDAI: Correlated negatively with
2013 [75] Egypt vitamin D. SLEDAI improved with
supplementation.
Andreoli et al., 2015 Randomized 34 SLE 300,000 bolus, 50,000iu T cell and Promotion of regulatory T cells
[77] Unblinded Italy monthly compared with B cell populations Production of Th2 cytokines
Piantoni et al., 2015 25,000iu monthly SLE serology Serology: Unchanged
[147]
(reported in two
parts)
Arno w et al., 2015 Randomized 54 SLE 2000iu, 4000iu/placebo Interferon No effect on interferon signature
[63] Double blind USA (54% African signature
Placebo controlled American)
Lima et al., 2015 Randomized 50 Juvenile SLE 50,000 iu weekly versus K-FSS SLEDAI: Improved (P ¼ 0.01)
[76] Double blind Brazil (30% non- placebo SLEDAI ECLAM: Improved (P ¼ 0.006)
Placebo controlled caucasian) ECLAM

SLEDAI: SLE disease activity index, SDI: ACR/SLICC damage index, UPCR: Urine protein to Creatinine ratio, VAS: visual analogue scale, ECLAM: European Consensus Lupus
Activity Measurement, K-FSS: Kids fatigue severity scale.

statistically significant trend towards higher disease activity in Hydroxychloroquine is considered central to the management of
those who had low hydroxychloroquine blood levels. However, cutaneous disease [92] and it is also helpful for inflammatory ar-
within individual analysis over time did not demonstrate an thralgias [120]. Numerous other benefits include decreased
improvement in disease activity once therapeutic levels were thrombosis [93], increased survival [84], improved lipid profiles
attained [119]. [85,98,99] and lower blood glucose [101].
In terms of organ specific effects, increased rates of renal Pregnancy outcomes are improved in those with SLE taking
remission have been demonstrated in patients treated with hydroxychloroquine [86,96] and in particular support of its effect as
hydroxychloroquine (in addition to immunosuppressives). In fact, an immunomodulator, there is some data that it decreases the risk
nephritis patients treated with hydroxychloroquine with myco- of congenital heart block in children of Ro positive mothers
phenolate mofetil, had a remission rate which was 5 times higher [103,104]. Autoantibody-associated congenital heart block is a
than those who were treated with mycophenolate alone [89]. serious condition which arises from passively acquired antibodies

Table 3
Hydroxychloroquine clinical trials in SLE.

Author & year Design N Patients Dose þ Time Outcome Result

Williams et al., 1996 [87]. Randomized, 73 SLE 48 weeks Joint count Improved patient reported pain
Double blind Arthritis/Arthralgia Disease activity
Placebo-controlled Pain (patient reported)
Levy et al., 2001 [148]. Randomized 26 SLE & DLE Pregnancy duration SLEPDAI Decreased prednisone in HCQ
Placebo-controlled Birth outcomes group
Improved SLEPDAI
Higher birth weight
The Canadian Randomized 47 SLE 6 months Flare Relative risk of flare 6.1 times
Hydroxychloroquine Double blind higher with withdrawal of HCQ.
Study Group [114]. Placebo-controlled
withdrawal study
Costedoat-Chalumeau et al. Randomized 171 SLE (with HCQ level 7 months SLEDAI No difference in SLEDAI with
2013 [118] Double-blind <1000 ng/ml) Flare attaining therapeutic blood
Placebo controlled level (p ¼ 0.70)

SLE: Systemic lupus erythematosus, DLE: Discoid lupus erythematosus, SLEPDAI: Systemic lupus erythematosus pregnancy disease activity index, HCQ: Hydroxychloroquine,
SLEDAI: Systemic lupus erythematosus disease activity index.

Please cite this article in press as: L. Durcan, M. Petri, Immunomodulators in SLE: Clinical evidence and immunologic actions, Journal of
Autoimmunity (2016), http://dx.doi.org/10.1016/j.jaut.2016.06.010
8 L. Durcan, M. Petri / Journal of Autoimmunity xxx (2016) 1e12

that target the fetal cardiac conduction system. With a Ro antibody, [128]), BAFF (belimumab [129], blisibimod [130], tabalumab [131],
the risk of having a pregnancy complicated by congenital heart briobacept, atacicept [132]) and a proliferation-inducing ligand
block is in the region of 1e2% and the risk of recurrence in a sub- (APRIL) (atacicept [132]). Despite robust preclinical and mecha-
sequent pregnancy increases to around 18% [121]. Izmirly et al. nistic data, these agents have been disappointing in clinical trials
demonstrated less congenital heart block in babies exposed to and have not, with the exception of belimumab, been approved for
hydroxychloroquine compared with controls [103]. In those with a SLE. Some, such as rituximab, are considered immunosuppressive,
previous history of having a child with cardiac neonatal lupus, the as they lead to B-cell depletion.
risk of recurrence has also been shown to be decreased with Belimumab is unique in showing clinical efficacy in large ran-
hydroxychloroquine therapy [104]. The recurrence rate in fetuses domized controlled studies and in its subsequent FDA approval
exposed to HCQ was 7.5% versus 21.2% in the unexposed group. This [129,133]. It is a fully humanized monoclonal antibody that spe-
effect is thought to be mediated via toll like receptors [122]. cifically binds to soluble BAFF, preventing its interaction with re-
In further support of the role of hydroxychloroquine as an ceptors, resulting in a reduction in the numbers of peripheral
immunomodulator, there is evidence that therapy associates with naive, transitional and activated B cells. Unlike rituximab, it does
delayed onset of SLE (in patients who have less than 4 American not deplete B-cell populations. Two large, phase III, multi-center,
College of Rheumatology classification criteria). Those treated with prospective, randomized, controlled trials have compared beli-
hydroxychloroquine had a longer time between the first clinical mumab with placebo in SLE [129,133]. Navarra et al. evaluated 867
symptom and the diagnosis of SLE and less autoantibodies [102]. patients in Latin America, Asia-Pacific and Eastern Europe. All had
Hydroxychloroquine is a well-tolerated medication and side active disease with a SLEDAI of greater than 6. They were ran-
effects are few. However, there are increasing concerns regarding domized to receive belimumab 1 mg/kg, 10 mg/kg, or placebo. The
hydroxychloroquine related retinopathy, in particular in light of outcome measure was the Systemic lupus erythematosus
new screening methods, which are thought to have increased responder index (SRI), a composite measure designed to evaluate
sensitivity [123]. Current American Academy of Ophthalmology overall and organ specific disease activity. It is composed of the
guidelines advise monitoring, beyond the dilated retinal exami- SLEDAI, BILAG and PGA. Significantly higher rates of response were
nation and automated visual field testing, in an attempt to identify noted with belimumab 1 mg/kg and 10 mg/kg than with placebo
toxicity early [124]. The sensitivity and specificity of these tests are 125 at week 52. More patients had their SLEDAI score reduced by
not yet known for hydroxychloroquine related retinal toxicity. Thus at least 4 points during 52 weeks with belimumab at both doses
the true prevalence of retinal deposition may differ from what was than with placebo. Better outcomes were also observed for PGA
previously reported. Other toxicities, cardiac and neuromyoapthic and BILAG with belimumab treatment. Furie et al. [129] demon-
are rare. These are reviewed in detail by Costedoat-Chalumeau et al. strated similar results and accordingly, belimumab received FDA
[111]. approval.
Hydroxychloroquine is a crucial immunomodulatory medicine Pooled data from these studies indicate that belimumab pro-
in SLE with innumerous disease specific and longitudinal benefits. motes normalization of serological abnormalities, with reversal of
Clinical data point toward a reduction in flare with therapy, hypocomplementemia and decreased autoantibodies (anti double
amongst other effects, but at present the mechanism for many of stranded DNA, anti Sm, anticardiolipin and anti ribosomal P). Both
the long-term benefits are incompletely understood. For the pre- dosing regimens associated with significant reductions in the
vention of SLE, it is the only medication with any known effect. In numbers of CD20 B cells and in multiple B cell and plasma cell
women with Ro antibodies, it is the only known strategy to help subsets, including naive and activated B cells, as well as in
prevent congenital heart block. CD20 þCD138 þ plasmablasts whilst preserving the memory B cell
subset and T cell populations, supporting its status as a specific
5. B cell therapies immunomodulatory therapy [134]. Post-hoc analyses demon-
strated that this medication is of most utility in those with high
B cells play an important pathologic role in SLE. Abnormal B cell disease activity despite standard therapy, low complement levels,
proliferation, maturation, prolonged life-span of auto reactive antibodies to double stranded DNA and corticosteroid use [135]. It
clones, and autoantibody production are known to be present in may also be of use in renal disease [136] although specific clinical
SLE, and to associate with immune dysregulation and breakdown of trials are yet to be reported.
self-tolerance [125]. B cells are involved in several specific pro-
inflammatory mechanisms in SLE, including T cell antigen presen- 6. Stem cell transplantation
tation, cytokine release and autoantibody formation. Self-antigens
are presented on the cell surface to auto-reactive T cells, trig- Autologous and allogeneic stem cell transplantation have been
gering B cells into autoantibody production and propagating their reported as having therapeutic benefit in SLE. In severe SLE, re-
function as antigen presenting cells. These cells then release fractory to conventional therapy, stem cell transplantation can
proinflammatory cytokines which are implicated in SLE including associate with sustained clinical remission, with rates ranging from
interferon a, IL-6 and IL-10, B-cell activating factor (BAFF), TNF a 50 to 70%, associated with normalization of many immunologic
and a proliferation inducing ligand (APRIL) [125]. These pathways changes [137,138]. This is a strategy that offers treatment-free
contribute to clinical manifestations by inciting inflammation, remission. However, it has not been evaluated in any randomized
causing tissue damage and immune complex deposition. As a result controlled studies and associates with considerable short-term
of these abnormalities, B cell therapies have always been consid- morbidity and in some cases mortality [137,139,140]. Transplant
ered attractive immunosuppressive and immunomodulatory regi- mortality has ranged from none to as high as 25% [137]. As such, in
mens. Although these agents do suppress immune function, they the absence of a controlled study, these therapies are unlikely to
also can have a long lasting effect on B cell populations and disease become available outside of the research setting.
mechanisms. Belimumab in particular, has only mildly immuno- Stem cell transplantation, both autologous and allogeneic, offers
suppressive properties and functions for the most part as an a fascinating and instructive insight into SLE disease mechanisms
immunomodulatory agent. and can be considered the ultimate immunomodulating therapy.
B-cell strategies which have been considered in SLE target CD- Following transplantation, autoantibodies consistently decreases
20 (ocrelizumab [126], rituximab [127]), CD-22 (epratuzumab or disappear with normalization of T cell responses [141] with the

Please cite this article in press as: L. Durcan, M. Petri, Immunomodulators in SLE: Clinical evidence and immunologic actions, Journal of
Autoimmunity (2016), http://dx.doi.org/10.1016/j.jaut.2016.06.010
L. Durcan, M. Petri / Journal of Autoimmunity xxx (2016) 1e12 9

disappearance of plasmablasts and return of healthy levels of reg- [13] F. Labrie, A. Belanger, J. Simard, Van Luu-The, C. Labrie, DHEA and peripheral
androgen and estrogen formation: intracinology, Ann. N. Y. Acad. Sci. 774
ulatory T cells [142]. There is a CD4þFoxP3þ regulatory T cell in
(1995) 16e28.
transplanted patients that seems to inhibit abnormal T cell re- [14] M.O. Goodarzi, E. Carmina, R. Azziz, DHEA, DHEAS and PCOS, J. Steroid Bio-
sponses [143]. There is evidence that allogeneic mesenchymal stem chem. Mol. Biol. 145 (2015) 213e225.
cell transplant can reverse changes in the T cell-interferon axis [15] R.W. McMurray, W. May, Sex hormones and systemic lupus erythematosus:
review and meta-analysis, Arthritis Rheum. 48 (8) (2003) 2100e2110.
which has also been though to contribute to SLE pathogenesis [16] R.G. Lahita, H.L. Bradlow, E. Ginzler, S. Pang, M. New, Low plasma androgens
[144]. in women with systemic lupus erythematosus, Arthritis Rheum. 30 (3)
(1987) 241e248.
[17] T. Suzuki, N. Suzuki, E.G. Engleman, Y. Mizushima, T. Sakane, Low serum
7. Conclusion levels of dehydroepiandrosterone may cause deficient IL-2 production by
lymphocytes in patients with systemic lupus erythematosus (SLE), Clin. Exp.
Immunol. 99 (2) (1995) 251e255.
SLE is a potentially fatal autoimmune disease characterized by [18] O.M. Wolkowitz, V.I. Reus, E. Roberts, F. Manfredi, T. Chan, W.J. Raum, et al.,
autoantibodies and immune dysregulation resulting in multiorgan Dehydroepiandrosterone (DHEA) treatment of depression, Biol. Psychiatry
injury. Current treatment strategies for the most part are immu- 41 (3) (1997) 311e318.
[19] M.S. Ritsner, R.D. Strous, Neurocognitive deficits in schizophrenia are asso-
nosuppressive and are limited by efficacy and side effects, such as
ciated with alterations in blood levels of neurosteroids: a multiple regression
increased infections and long term toxicities. Immunomodulatory analysis of findings from a double-blind, randomized, placebo-controlled,
therapies offer the opportunity to prevent disease activity and crossover trial with DHEA, J. Psychiatr. Res. 44 (2) (2010) 75e80.
[20] R.D. Strous, R. Stryjer, R. Maayan, G. Gal, D. Viglin, E. Katz, et al., Analysis of
decrease the accrual of damage.
clinical symptomatology, extrapyramidal symptoms and neurocognitive
Hydroxychloroquine is a crucial background medication in SLE dysfunction following dehydroepiandrosterone (DHEA) administration in
and has actions on numerous cell types in many molecular path- olanzapine treated schizophrenia patients: a randomized, double-blind
ways. It has disease specific effects in reducing flare, treating placebo controlled trial, Psychoneuroendocrinology 32 (2) (2007) 96e105.
[21] J.S. Dillon, Dehydroepiandrosterone, dehydroepiandrosterone sulfate and
cutaneous disease and inflammatory arthralgias in addition to related steroids: their role in inflammatory, allergic and immunological
innumerous other effects such as reduced thrombosis, increased disorders, Curr. Drug Targets Inflamm. Allergy 4 (3) (2005) 377e385.
longevity, improved lipids, better glycemic control and blood [22] D.M. Chang, S.J. Chu, H.C. Chen, S.Y. Kuo, J.H. Lai, Dehydroepiandrosterone
suppresses interleukin 10 synthesis in women with systemic lupus erythe-
pressure. Dehydroeipandrosterone is also an immunomodulator in matosus, Ann. Rheum. Dis. 63 (12) (2004) 1623e1626.
SLE which can have positive effects on disease activity and has bone [23] J.A. Lucas, S.A. Ahmed, M.L. Casey, P.C. MacDonald, Prevention of autoanti-
protective properties. Vitamin D is now known to be important in body formation and prolonged survival in New Zealand black/New Zealand
white F1 mice fed dehydroisoandrosterone, J. Clin. Invest 75 (6) (1985)
SLE and supplementation appears to have a positive impact on 2091e2093.
disease activity including proteinuria. Belimumab has specific [24] S.D. Norton, L.L. Harrison, R. Yowell, B.A. Araneo, Administration of dehy-
immunomodulatory properties and is an effective therapy in those droepiandrosterone sulfate retards onset but not progression of autoimmune
disease in NZB/W mice, Autoimmunity 26 (3) (1997) 161e171.
with the serological and clinical characteristics predictive of
[25] R.H. Straub, K. Lehle, H. Herfarth, M. Weber, W. Falk, J. Preuner, et al., De-
response. Stem cell transplantation is a fascinating therapy in SLE, hydroepiandrosterone in relation to other adrenal hormones during an acute
but is not yet proven in controlled studies and associates with inflammatory stressful disease state compared with chronic inflammatory
disease: role of interleukin-6 and tumour necrosis factor, Eur. J. Endocrinol.
significant morbidity and in some cases, mortality.
146 (3) (2002) 365e374.
[26] E. Peterson, A.D. Robertson, W. Emlen, Serum and urinary interleukin-6 in
Support systemic lupus erythematosus, Lupus 5 (6) (1996) 571e575.
[27] M. Iwano, K. Dohi, E. Hirata, N. Kurumatani, Y. Horii, H. Shiiki, et al., Urinary
levels of IL-6 in patients with active lupus nephritis, Clin. Nephrol. 40 (1)
The Hopkins Lupus Cohort is supported by NIH AR 43727. (1993) 16e21.
[28] E.M. Ball, D.S. Gibson, A.L. Bell, M.R. Rooney, Plasma IL-6 levels correlate with
clinical and ultrasound measures of arthritis in patients with systemic lupus
References erythematosus, Lupus 23 (1) (2014) 46e56.
[29] J. Ro€nnelid, A. Tejde, L. Mathsson, K. Nilsson-Ekdahl, B. Nilsson, Immune
[1] C.C. Mok, Towards new avenues in the management of lupus glomerulone- complexes from SLE sera induce IL10 production from normal peripheral
phritis, Nat. Rev. Rheumatol. 12 (4) (2016 Apr) 221e234, http://dx.doi.org/ blood mononuclear cells by an FcgammaRII dependent mechanism: impli-
10.1038/nrrheum.2015. cations for a possible vicious cycle maintaining B cell hyperactivity in SLE,
[2] M.M. Ward, Changes in the incidence of end-stage renal disease due to lupus Ann. Rheum. Dis. 62 (1) (2003) 37e42.
nephritis, 1982-1995, Arch. Intern Med. 160 (20) (2000) 3136e3140. [30] R.F. van Vollenhoven, E.G. Engleman, J.L. McGuire, An open study of dehy-
[3] L.N. Parker, Control of adrenal androgen secretion, Endocrinol. Metab. Clin. droepiandrosterone in systemic lupus erythematosus, Arthritis Rheum. 37
North Am. 20 (2) (1991) 401e421. (9) (1994) 1305e1310.
[4] C.C. Chen, C.R. Parker, Adrenal androgens and the immune system, Semin. [31] R.F. van Vollenhoven, J.L. Park, M.C. Genovese, J.P. West, J.L. McGuire,
Reprod. Med. 22 (4) (2004) 369e377. A double-blind, placebo-controlled, clinical trial of dehydroepiandrosterone
[5] S.J. Webb, T.E. Geoghegan, R.A. Prough, K.K. Michael Miller, The biological in severe systemic lupus erythematosus, Lupus 8 (3) (1999) 181e187.
actions of dehydroepiandrosterone involves multiple receptors, Drug Metab. [32] M.A. Petri, R.G. Lahita, R.F. Van Vollenhoven, J.T. Merrill, M. Schiff,
Rev. 38 (1e2) (2006) 89e116. E.M. Ginzler, et al., Effects of prasterone on corticosteroid requirements of
[6] R.H. Derksen, Dehydroepiandrosterone (DHEA) and systemic lupus erythe- women with systemic lupus erythematosus: a double-blind, randomized,
matosus, Semin. Arthritis Rheum. 27 (6) (1998) 335e347. placebo-controlled trial, Arthritis Rheum. 46 (7) (2002) 1820e1829.
[7] W.E. Rainey, B.R. Carr, H. Sasano, T. Suzuki, J.I. Mason, Dissecting human [33] D.M. Chang, J.L. Lan, H.Y. Lin, S.F. Luo, Dehydroepiandrosterone treatment of
adrenal androgen production, Trends Endocrinol. Metab. 13 (6) (2002) women with mild-to-moderate systemic lupus erythematosus: a multi-
234e239. center randomized, double-blind, placebo-controlled trial, Arthritis Rheum.
[8] L. Vandenput, C. Ohlsson, Genome-wide association studies on serum sex 46 (11) (2002) 2924e2927.
steroid levels, Mol. Cell Endocrinol. 382 (1) (2014) 758e766. [34] A. Hartkamp, R. Geenen, G.L. Godaert, M. Bijl, J.W. Bijlsma, R.H. Derksen, The
[9] A.M. Traish, H.P. Kang, F. Saad, A.T. Guay, Dehydroepiandrosterone (DHEA)ea effect of dehydroepiandrosterone on lumbar spine bone mineral density in
precursor steroid or an active hormone in human physiology, J. Sex. Med. 8 patients with quiescent systemic lupus erythematosus, Arthritis Rheum. 50
(11) (2011) 2960e2982 quiz 83. (11) (2004) 3591e3595.
[10] F. Chen, K. Knecht, E. Birzin, J. Fisher, H. Wilkinson, M. Mojena, et al., Direct [35] P.J. Mease, E.M. Ginzler, O.S. Gluck, M. Schiff, A. Goldman, M. Greenwald, et
agonist/antagonist functions of dehydroepiandrosterone, Endocrinology 146 al., Effects of prasterone on bone mineral density in women with systemic
(11) (2005) 4568e4576. lupus erythematosus receiving chronic glucocorticoid therapy, J. Rheumatol.
[11] S.L. Ripp, J.L. Fitzpatrick, J.M. Peters, R.A. Prough, Induction of CYP3A 32 (4) (2005) 616e621.
expression by dehydroepiandrosterone: involvement of the pregnane X re- [36] J. Sa nchez-Guerrero, H.E. Fragoso-Loyo, C.M. Neuwelt, D.J. Wallace,
ceptor, Drug Metab. Dispos. 30 (5) (2002) 570e575. E.M. Ginzler, Y.R. Sherrer, et al., Effects of prasterone on bone mineral density
[12] F. Hammer, D.G. Drescher, S.B. Schneider, M. Quinkler, P.M. Stewart, in women with active systemic lupus erythematosus receiving chronic
B. Allolio, et al., Sex steroid metabolism in human peripheral blood mono- glucocorticoid therapy, J. Rheumatol. 35 (8) (2008) 1567e1575.
nuclear cells changes with aging, J. Clin. Endocrinol. Metab. 90 (11) (2005) [37] G. Nordmark, C. Bengtsson, A. Larsson, F.A. Karlsson, G. Sturfelt, L. Ro €nnblom,
6283e6289. Effects of dehydroepiandrosterone supplement on health-related quality of

Please cite this article in press as: L. Durcan, M. Petri, Immunomodulators in SLE: Clinical evidence and immunologic actions, Journal of
Autoimmunity (2016), http://dx.doi.org/10.1016/j.jaut.2016.06.010
10 L. Durcan, M. Petri / Journal of Autoimmunity xxx (2016) 1e12

life in glucocorticoid treated female patients with systemic lupus erythe- erythematosus, Osteoporos. Int. 20 (3) (2009) 427e433.
matosus, Autoimmunity 38 (7) (2005) 531e540. [65] H. Amital, Z. Szekanecz, G. Szücs, K. Danko , E. Nagy, T. Csepa
ny, et al., Serum
[38] D.D. Gladman, M.B. Urowitz, C.H. Goldsmith, P. Fortin, E. Ginzler, C. Gordon, concentrations of 25-OH vitamin D in patients with systemic lupus erythe-
et al., The reliability of the systemic lupus international collaborating clinics/ matosus (SLE) are inversely related to disease activity: is it time to routinely
american College of Rheumatology damage index in patients with systemic supplement patients with SLE with vitamin D? Ann. Rheum. Dis. 69 (6)
lupus erythematosus, Arthritis Rheum. 40 (5) (1997) 809e813. (2010) 1155e1157.
[39] G. Wolf, The discovery of vitamin D: the contribution of Adolf Windaus, [66] S.S. Yeap, A.Z. Othman, A.A. Zain, S.P. Chan, Vitamin D levels: its relationship
J. Nutr. 134 (6) (2004) 1299e1302. to bone mineral density response and disease activity in premenopausal
[40] L.A. Peterson, X. Zeng, C.P. Caufield-Noll, M.A. Schweitzer, T.H. Magnuson, Malaysian systemic lupus erythematosus patients on corticosteroids, Int. J.
K.E. Steele, Vitamin D status and supplementation before and after bariatric Rheum. Dis. 15 (1) (2012) 17e24.
surgery: a comprehensive literature review, Surg. Obes. Relat. Dis. 12 (3) [67] J.A. Reynolds, S. Haque, J.L. Berry, P. Pemberton, L.S. Teh, P. Ho, et al., 25-
(2016) 693e702. Hydroxyvitamin D deficiency is associated with increased aortic stiffness
[41] M.F. Holick, Vitamin D deficiency, N. Engl. J. Med. 357 (3) (2007) 266e281. in patients with systemic lupus erythematosus, Rheumatol. Oxf. 51 (3)
[42] H.F. DeLuca, Overview of general physiologic features and functions of (2012) 544e551.
vitamin D, Am. J. Clin. Nutr. 80 (6 Suppl) (2004) 1689Se1696S. [68] C.C. Mok, D.J. Birmingham, H.W. Leung, L.A. Hebert, H. Song, B.H. Rovin,
[43] M. Iruretagoyena, D. Hirigoyen, R. Naves, P.I. Burgos, Immune response Vitamin D levels in Chinese patients with systemic lupus erythematosus:
modulation by vitamin D: role in systemic lupus erythematosus, Front. relationship with disease activity, vascular risk factors and atherosclerosis,
Immunol. 6 (2015) 513. Rheumatol. Oxf. 51 (4) (2012) 644e652.
[44] A.C. Ross, J.E. Manson, S.A. Abrams, J.F. Aloia, P.M. Brannon, S.K. Clinton, et al., [69] A. Lertratanakul, P. Wu, A. Dyer, M. Urowitz, D. Gladman, P. Fortin, et al., 25-
The 2011 report on dietary reference intakes for calcium and vitamin D from hydroxyvitamin D and cardiovascular disease in patients with systemic
the Institute of Medicine: what clinicians need to know, J. Clin. Endocrinol. lupus erythematosus: data from a large international inception cohort,
Metab. 96 (1) (2011) 53e58. Arthritis Care Res. Hob. 66 (8) (2014) 1167e1176.
[45] J.C. Souberbielle, J.J. Body, J.M. Lappe, M. Plebani, Y. Shoenfeld, T.J. Wang, et [70] A. Becker, R. Fischer, M. Schneider, Bone density and 25-OH vitamin D serum
al., Vitamin D and musculoskeletal health, cardiovascular disease, autoim- level in patients with systemic lupus erythematosus, Z Rheumatol. 60 (5)
munity and cancer: recommendations for clinical practice, Autoimmun. Rev. (2001) 352e358.
9 (11) (2010) 709e715. [71] Z.S. Bonakdar, L. Jahanshahifar, F. Jahanshahifar, A. Gholamrezaei, Vitamin D
[46] M.F. Holick, Vitamin D: important for prevention of osteoporosis, cardio- deficiency and its association with disease activity in new cases of systemic
vascular heart disease, type 1 diabetes, autoimmune diseases, and some lupus erythematosus, Lupus 20 (11) (2011) 1155e1160.
cancers, South Med. J. 98 (10) (2005) 1024e1027. [72] G. Ruiz-Irastorza, S. Gordo, N. Olivares, M.V. Egurbide, C. Aguirre, Changes in
[47] A. Ascherio, K.L. Munger, K.C. Simon, Vitamin D and multiple sclerosis, Lancet vitamin D levels in patients with systemic lupus erythematosus: effects on
Neurol. 9 (6) (2010) 599e612. fatigue, disease activity, and damage, Arthritis Care Res. Hob. 62 (8) (2010)
[48] S. Patel, T. Farragher, J. Berry, D. Bunn, A. Silman, D. Symmons, Association 1160e1165.
between serum vitamin D metabolite levels and disease activity in patients [73] B. Terrier, N. Derian, Y. Schoindre, W. Chaara, G. Geri, N. Zahr, et al., Resto-
with early inflammatory polyarthritis, Arthritis Rheum. 56 (7) (2007) ration of regulatory and effector T cell balance and B cell homeostasis in
2143e2149. systemic lupus erythematosus patients through vitamin D supplementation,
[49] K. Müller, N.J. Kriegbaum, B. Baslund, O.H. Sørensen, M. Thymann, Arthritis Res. Ther. 14 (5) (2012) R221.
K. Bentzen, Vitamin D3 metabolism in patients with rheumatic diseases: low [74] M. Petri, K.J. Bello, H. Fang, L.S. Magder, Vitamin D in systemic lupus ery-
serum levels of 25-hydroxyvitamin D3 in patients with systemic lupus er- thematosus: modest association with disease activity and the urine protein-
ythematosus, Clin. Rheumatol. 14 (4) (1995) 397e400. to-creatinine ratio, Arthritis Rheum. 65 (7) (2013) 1865e1871.
[50] L.L. Ritterhouse, S.R. Crowe, T.B. Niewold, D.L. Kamen, S.R. Macwana, [75] A. Abou-Raya, S. Abou-Raya, M. Helmii, The effect of vitamin D supple-
V.C. Roberts, et al., Vitamin D deficiency is associated with an increased mentation on inflammatory and hemostatic markers and disease activity in
autoimmune response in healthy individuals and in patients with systemic patients with systemic lupus erythematosus: a randomized placebo-
lupus erythematosus, Ann. Rheum. Dis. 70 (9) (2011) 1569e1574. controlled trial, J. Rheumatol. 40 (3) (2013) 265e272.
[51] T.B. Wright, J. Shults, M.B. Leonard, B.S. Zemel, J.M. Burnham, Hypovitami- [76] G.L. Lima, J. Paupitz, N.E. Aikawa, L. Takayama, E. Bonfa, R.M. Pereira, Vitamin
nosis D is associated with greater body mass index and disease activity in D supplementation in adolescents and young adults with juvenile systemic
pediatric systemic lupus erythematosus, J. Pediatr. 155 (2) (2009) 260e265. lupus erythematosus for improvement in disease activity and fatigue scores:
[52] D.L. Kamen, G.S. Cooper, H. Bouali, S.R. Shaftman, B.W. Hollis, G.S. Gilkeson, a randomized, double-blind, placebo-controlled trial, Arthritis Care Res. Hob.
Vitamin D deficiency in systemic lupus erythematosus, Autoimmun. Rev. 5 68 (1) (2016) 91e98.
(2) (2006) 114e117. [77] L. Andreoli, F. Dall’Ara, S. Piantoni, A. Zanola, N. Piva, M. Cutolo, et al., A 24-
[53] A.M. Huisman, K.P. White, A. Algra, M. Harth, R. Vieth, J.W. Jacobs, et al., month prospective study on the efficacy and safety of two different monthly
Vitamin D levels in women with systemic lupus erythematosus and fibro- regimens of vitamin D supplementation in pre-menopausal women with
myalgia, J. Rheumatol. 28 (11) (2001) 2535e2539. systemic lupus erythematosus, Lupus 24 (4e5) (2015) 499e506.
[54] G. Ruiz-Irastorza, M.V. Egurbide, J.I. Pijoan, M. Garmendia, I. Villar, [78] D.H. Kim, S. Sabour, U.N. Sagar, S. Adams, D.J. Whellan, Prevalence of
A. Martinez-Berriotxoa, et al., Effect of antimalarials on thrombosis and hypovitaminosis D in cardiovascular diseases (from the national health and
survival in patients with systemic lupus erythematosus, Lupus 15 (9) (2006) nutrition examination survey 2001 to 2004), Am. J. Cardiol. 102 (11) (2008)
577e583. 1540e1544.
[55] C.C. Mok, Vitamin D and systemic lupus erythematosus: an update, Expert [79] K.E. Poole, N. Loveridge, P.J. Barker, D.J. Halsall, C. Rose, J. Reeve, et al.,
Rev. Clin. Immunol. 9 (5) (2013) 453e463. Reduced vitamin D in acute stroke, Stroke 37 (1) (2006) 243e245.
[56] C.E. Powe, M.K. Evans, J. Wenger, A.B. Zonderman, A.H. Berg, M. Nalls, et al., [80] J. Kendrick, G. Targher, G. Smits, M. Chonchol, 25-Hydroxyvitamin D defi-
Vitamin D-binding protein and vitamin D status of black Americans and ciency is independently associated with cardiovascular disease in the Third
white Americans, N. Engl. J. Med. 369 (21) (2013) 1991e2000. National Health and Nutrition Examination Survey, Atherosclerosis 205 (1)
[57] L.T. Hiraki, K.L. Munger, K.H. Costenbader, E.W. Karlson, Dietary intake of (2009) 255e260.
vitamin D during adolescence and risk of adult-onset systemic lupus ery- [81] R.L. Ravenell, D.L. Kamen, J.D. Spence, B.W. Hollis, T.J. Fleury, M.G. Janech, et
thematosus and rheumatoid arthritis, Arthritis Care Res. Hob. 64 (12) (2012) al., Premature atherosclerosis is associated with hypovitaminosis D and
1829e1836. angiotensin-converting enzyme inhibitor non-use in lupus patients, Am. J.
[58] S. Chen, G.P. Sims, X.X. Chen, Y.Y. Gu, P.E. Lipsky, Modulatory effects of 1,25- Med. Sci. 344 (4) (2012) 268e273.
dihydroxyvitamin D3 on human B cell differentiation, J. Immunol. 179 (3) [82] R.A. Furie, M.A. Petri, D.J. Wallace, E.M. Ginzler, J.T. Merrill, W. Stohl, et al.,
(2007) 1634e1647. Novel evidence-based systemic lupus erythematosus responder index,
[59] C. Aranow, Vitamin D and the immune system, J. Investig. Med. 59 (6) (2011) Arthritis Rheum. 61 (9) (2009) 1143e1151.
881e886. [83] A randomized study of the effect of withdrawing hydroxychloroquine sulfate
[60] S. Ojaimi, N.A. Skinner, B.J. Strauss, V. Sundararajan, I. Woolley, in systemic lupus erythematosus, N. Engl. J. Med. 324 (3) (1991) 150e154.
K. Visvanathan, Vitamin D deficiency impacts on expression of toll-like re- [84] G.S. Alarco  n, G. McGwin, A.M. Bertoli, B.J. Fessler, J. Calvo-Ale
n, H.M. Bastian,
ceptor-2 and cytokine profile: a pilot study, J. Transl. Med. 11 (2013) 176. et al., Effect of hydroxychloroquine on the survival of patients with systemic
[61] S.U. Monrad, K. Rea, S. Thacker, M.J. Kaplan, Myeloid dendritic cells display lupus erythematosus: data from LUMINA, a multiethnic US cohort (LUMINA
downregulation of C-type lectin receptors and aberrant lectin uptake in L), Ann. Rheum. Dis. 66 (9) (2007) 1168e1172.
systemic lupus erythematosus, Arthritis Res. Ther. 10 (5) (2008) R114. [85] E. Cairoli, M. Rebella, N. Danese, V. Garra, E.F. Borba, Hydroxychloroquine
[62] I. Ben-Zvi, C. Aranow, M. Mackay, A. Stanevsky, D.L. Kamen, L.M. Marinescu, reduces low-density lipoprotein cholesterol levels in systemic lupus ery-
et al., The impact of vitamin D on dendritic cell function in patients with thematosus: a longitudinal evaluation of the lipid-lowering effect, Lupus 21
systemic lupus erythematosus, PLoS One 5 (2) (2010) e9193. (11) (2012) 1178e1182.
[63] C. Aranow, D.L. Kamen, M. Dall’Era, E.M. Massarotti, M.C. Mackay, [86] M.E. Clowse, L. Magder, F. Witter, M. Petri, Hydroxychloroquine in lupus
F. Koumpouras, et al., Randomized, double-blind, placebo-controlled trial of pregnancy, Arthritis Rheum. 54 (11) (2006) 3640e3647.
the effect of vitamin D3 on the interferon signature in patients with systemic [87] H.J. Williams, M.J. Egger, J.Z. Singer, R.F. Willkens, K.C. Kalunian, D.O. Clegg, et
lupus erythematosus, Arthritis Rheumatol. 67 (7) (2015) 1848e1857. al., Comparison of hydroxychloroquine and placebo in the treatment of the
[64] V.Z. Borba, J.G. Vieira, T. Kasamatsu, S.C. Radominski, E.I. Sato, M. Lazaretti- arthropathy of mild systemic lupus erythematosus, J. Rheumatol. 21 (8)
Castro, Vitamin D deficiency in patients with active systemic lupus (1994) 1457e1462.

Please cite this article in press as: L. Durcan, M. Petri, Immunomodulators in SLE: Clinical evidence and immunologic actions, Journal of
Autoimmunity (2016), http://dx.doi.org/10.1016/j.jaut.2016.06.010
L. Durcan, M. Petri / Journal of Autoimmunity xxx (2016) 1e12 11

[88] R.G. Espinola, S.S. Pierangeli, A.E. Gharavi, E.N. Harris, A.E. Ghara, Hydroxy- [111] N. Costedoat-Chalumeau, B. Dunogue , N. Morel, V. Le Guern, G. Guettrot-
chloroquine reverses platelet activation induced by human IgG anti- Imbert, Hydroxychloroquine: a multifaceted treatment in lupus, Presse Med.
phospholipid antibodies, Thromb. Haemost. 87 (3) (2002) 518e522. 43 (6 Pt 2) (2014) e167ee180.
[89] N. Kasitanon, D.M. Fine, M. Haas, L.S. Magder, M. Petri, Hydroxychloroquine [112] R. Lafyatis, M. York, A. Marshak-Rothstein, Antimalarial agents: closing the
use predicts complete renal remission within 12 months among patients gate on Toll-like receptors? Arthritis Rheum. 54 (10) (2006) 3068e3070.
treated with mycophenolate mofetil therapy for membranous lupus [113] J. An, J.J. Woodward, T. Sasaki, M. Minie, K.B. Elkon, Cutting edge: antima-
nephritis, Lupus 15 (6) (2006) 366e370. larial drugs inhibit IFN-b production through blockade of cyclic GMP-AMP
[90] B.J. Fessler, G.S. Alarco  n, G. McGwin, J. Roseman, H.M. Bastian, synthase-DNA interaction, J. Immunol. 194 (9) (2015) 4089e4093.
A.W. Friedman, et al., Systemic lupus erythematosus in three ethnic groups: [114] A randomized study of the effect of withdrawing hydroxychloroquine sulfate
XVI. Association of hydroxychloroquine use with reduced risk of damage in systemic lupus erythematosus. The Canadian Hydroxychloroquine Study
accrual, Arthritis Rheum. 52 (5) (2005) 1473e1480. Group, N. Engl. J. Med. 324 (3) (1991) 150e154.
[91] E.J. Sutton, J.E. Davidson, I.N. Bruce, The systemic lupus international [115] M.A. Petri, R.F. van Vollenhoven, J. Buyon, R.A. Levy, S.V. Navarra, R. Cervera,
collaborating clinics (SLICC) damage index: a systematic literature review, et al., Baseline predictors of systemic lupus erythematosus flares: data from
Semin. Arthritis Rheum. 43 (3) (2013) 352e361. the combined placebo groups in the phase III belimumab trials, Arthritis
[92] A. Kuhn, V. Ruland, G. Bonsmann, Cutaneous lupus erythematosus: update of Rheum. 65 (8) (2013) 2143e2153.
therapeutic options part I, J. Am. Acad. Dermatol 65 (6) (2011) e179ee193. [116] N. Costedoat-Chalumeau, Z. Amoura, J.S. Hulot, H.A. Hammoud, G. Aymard,
[93] M. Petri, Use of hydroxychloroquine to prevent thrombosis in systemic lupus P. Cacoub, et al., Low blood concentration of hydroxychloroquine is a marker
erythematosus and in antiphospholipid antibody-positive patients, Curr. for and predictor of disease exacerbations in patients with systemic lupus
Rheumatol. Rep. 13 (1) (2011) 77e80. erythematosus, Arthritis Rheum. 54 (10) (2006) 3284e3290.
[94] R. Kaiser, C.M. Cleveland, L.A. Criswell, Risk and protective factors for [117] C. France s, A. Cosnes, P. Duhaut, N. Zahr, B. Soutou, S. Ingen-Housz-Oro, et al.,
thrombosis in systemic lupus erythematosus: results from a large, multi- Low blood concentration of hydroxychloroquine in patients with refractory
ethnic cohort, Ann. Rheum. Dis. 68 (2) (2009) 238e241. cutaneous lupus erythematosus: a French multicenter prospective study,
[95] M.G. Tektonidou, K. Laskari, D.B. Panagiotakos, H.M. Moutsopoulos, Risk Arch. Dermatol 148 (4) (2012) 479e484.
factors for thrombosis and primary thrombosis prevention in patients with [118] N. Costedoat-Chalumeau, L. Galicier, O. Aumaitre, C. Frances, V. Le Guern,
systemic lupus erythematosus with or without antiphospholipid antibodies, F. Liote, et al., Hydroxychloroquine in systemic lupus erythematosus: results
Arthritis Rheum. 61 (1) (2009) 29e36. of a French multicentre controlled trial (PLUS Study), Ann. Rheum. Dis. 72
[96] M. Petri, D. Howard, J. Repke, Frequency of lupus flare in pregnancy. The (11) (2013) 1786e1792.
Hopkins lupus pregnancy center experience, Arthritis Rheum. 34 (12) (1991) [119] L. Durcan, W.A. Clarke, L.S. Magder, M. Petri, Hydroxychloroquine blood
1538e1545. levels in SLE: clarifying dosing controversies and improving adherence,
[97] A. Mekinian, M.G. Lazzaroni, A. Kuzenko, J. Alijotas-Reig, A. Ruffatti, P. Levy, J. Rheumatol. 42 (11) (2015 Nov) 2092e2097, http://dx.doi.org/10.3899/
et al., The efficacy of hydroxychloroquine for obstetrical outcome in anti- jrheum.150379.
phospholipid syndrome: data from a European multicenter retrospective [120] H.J. Williams, M.J. Egger, J.Z. Singer, R.F. Willkens, K.C. Kalunian, D.O. Clegg, et
study, Autoimmun. Rev. 14 (6) (2015) 498e502. al., Comparison of hydroxychloroquine and placebo in the treatment of the
[98] G. Kerr, M. Aujero, J. Richards, H. Sayles, L. Davis, G. Cannon, et al., Associ- arthropathy of mild systemic lupus erythematosus, J. Rheumatol. 21 (8)
ations of hydroxychloroquine use with lipid profiles in rheumatoid arthritis: (1994) 1457e1462.
pharmacologic implications, Arthritis Care Res. (Hoboken) 66 (11) (2014 [121] N. Gleicher, U. Elkayam, Preventing congenital neonatal heart block in
Nov) 1619e1626, http://dx.doi.org/10.1002/acr.22341. offspring of mothers with anti-SSA/Ro and SSB/La antibodies: a review of
[99] L. Durcan, D.A. Winegar, M.A. Connelly, J.D. Otvos, L.S. Magder, M. Petri, published literature and registered clinical trials, Autoimmun. Rev. 12 (11)
Longitudinal evaluation of lipoprotein variables in systemic lupus erythe- (2013) 1039e1045.
matosus reveals adverse changes with disease activity and prednisone and [122] R.M. Clancy, D. Alvarez, E. Komissarova, F.J. Barrat, J. Swartz, J.P. Buyon, Ro60-
more favorable profiles with hydroxychloroquine therapy, J. Rheumatol. 43 associated single-stranded RNA links inflammation with fetal cardiac fibrosis
(4) (2016 Apr) 745e750, http://dx.doi.org/10.3899/jrheum.150437. via ligation of TLRs: a novel pathway to autoimmune-associated heart block,
[100] D.J. Wallace, V.S. Gudsoorkar, M.H. Weisman, S.R. Venuturupalli, New in- J. Immunol. 184 (4) (2010) 2148e2155.
sights into mechanisms of therapeutic effects of antimalarial agents in SLE, [123] R.B. Melles, M.F. Marmor, The risk of toxic retinopathy in patients on long-
Nat. Rev. Rheumatol. 8 (9) (2012) 522e533. term hydroxychloroquine therapy, JAMA Ophthalmol. 132 (12) (2014)
[101] H.C. Gerstein, K.E. Thorpe, D.W. Taylor, R.B. Haynes, The effectiveness of 1453e1460.
hydroxychloroquine in patients with type 2 diabetes mellitus who are re- [124] M.F. Marmor, U. Kellner, T.Y. Lai, J.S. Lyons, W.F. Mieler, Revised recom-
fractory to sulfonylureasea randomized trial, Diabetes Res. Clin. Pract. 55 (3) mendations on screening for chloroquine and hydroxychloroquine retinop-
(2002) 209e219. athy, Ophthalmology 118 (2) (2011) 415e422.
[102] J.A. James, X.R. Kim-Howard, B.F. Bruner, M.K. Jonsson, M.T. McClain, [125] T. Do€rner, C. Giesecke, P.E. Lipsky, Mechanisms of B cell autoimmunity in SLE,
M.R. Arbuckle, et al., Hydroxychloroquine sulfate treatment is associated Arthritis Res. Ther. 13 (5) (2011) 243.
with later onset of systemic lupus erythematosus, Lupus 16 (6) (2007) [126] E.F. Mysler, A.J. Spindler, R. Guzman, M. Bijl, D. Jayne, R.A. Furie, et al., Effi-
401e409. cacy and safety of ocrelizumab in active proliferative lupus nephritis: results
[103] P.M. Izmirly, M.Y. Kim, C. Llanos, P.U. Le, M.M. Guerra, A.D. Askanase, et al., from a randomized, double-blind, phase III study, Arthritis Rheum. 65 (9)
Evaluation of the risk of anti-SSA/Ro-SSB/La antibody-associated cardiac (2013) 2368e2379.
manifestations of neonatal lupus in fetuses of mothers with systemic lupus [127] J.T. Merrill, C.M. Neuwelt, D.J. Wallace, J.C. Shanahan, K.M. Latinis, J.C. Oates,
erythematosus exposed to hydroxychloroquine, Ann. Rheum. Dis. 69 (10) et al., Efficacy and safety of rituximab in moderately-to-severely active
(2010) 1827e1830. systemic lupus erythematosus: the randomized, double-blind, phase II/III
[104] P.M. Izmirly, N. Costedoat-Chalumeau, C.N. Pisoni, M.A. Khamashta, systemic lupus erythematosus evaluation of rituximab trial, Arthritis Rheum.
M.Y. Kim, A. Saxena, et al., Maternal use of hydroxychloroquine is associated 62 (1) (2010) 222e233.
with a reduced risk of recurrent anti-SSA/Ro-antibody-associated cardiac [128] D.J. Wallace, C. Gordon, V. Strand, K. Hobbs, M. Petri, K. Kalunian, et al., Ef-
manifestations of neonatal lupus, Circulation 126 (1) (2012) 76e82. ficacy and safety of epratuzumab in patients with moderate/severe flaring
[105] B.E. van den Borne, B.A. Dijkmans, H.H. de Rooij, S. le Cessie, C.L. Verweij, systemic lupus erythematosus: results from two randomized, double-blind,
Chloroquine and hydroxychloroquine equally affect tumor necrosis factor- placebo-controlled, multicentre studies (ALLEVIATE) and follow-up, Rheu-
alpha, interleukin 6, and interferon-gamma production by peripheral blood matol. Oxf. 52 (7) (2013) 1313e1322.
mononuclear cells, J. Rheumatol. 24 (1) (1997) 55e60. [129] R. Furie, M. Petri, O. Zamani, R. Cervera, D.J. Wallace, D. Tegzova , et al.,
[106] J. Bondeson, R. Sundler, Antimalarial drugs inhibit phospholipase A2 acti- A phase III, randomized, placebo-controlled study of belimumab, a mono-
vation and induction of interleukin 1beta and tumor necrosis factor alpha in clonal antibody that inhibits B lymphocyte stimulator, in patients with
macrophages: implications for their mode of action in rheumatoid arthritis, systemic lupus erythematosus, Arthritis Rheum. 63 (12) (2011) 3918e3930.
Gen. Pharmacol. 30 (3) (1998) 357e366. [130] R.A. Furie, G. Leon, M. Thomas, M.A. Petri, A.D. Chu, C. Hislop, et al., A phase 2,
[107] J.C. Silva, H.A. Mariz, L.F. Rocha, P.S. Oliveira, A.T. Dantas, A.L. Duarte, et al., randomised, placebo-controlled clinical trial of blisibimod, an inhibitor of B
Hydroxychloroquine decreases Th17-related cytokines in systemic lupus cell activating factor, in patients with moderate-to-severe systemic lupus
erythematosus and rheumatoid arthritis patients, Clin. (Sao Paulo) 68 (6) erythematosus, the PEARL-SC study, Ann. Rheum. Dis. 74 (9) (2015)
(2013) 766e771. 1667e1675.
[108] K. Sacre, L.A. Criswell, J.M. McCune, Hydroxychloroquine is associated with [131] D.A. Isenberg, M. Petri, K. Kalunian, Y. Tanaka, M.B. Urowitz, R.W. Hoffman, et
impaired interferon-alpha and tumor necrosis factor-alpha production by al., Efficacy and safety of subcutaneous tabalumab in patients with systemic
plasmacytoid dendritic cells in systemic lupus erythematosus, Arthritis Res. lupus erythematosus: results from ILLUMINATE-1, a 52-week, phase III,
Ther. 14 (3) (2012) R155. multicentre, randomised, double-blind, placebo-controlled study, Ann.
[109] R. Willis, A.M. Seif, G. McGwin, L.A. Martinez-Martinez, E.B. Gonz alez, Rheum. Dis. 75 (2) (2016) 323e331.
N. Dang, et al., Effect of hydroxychloroquine treatment on pro-inflammatory [132] D. Isenberg, C. Gordon, D. Licu, S. Copt, C.P. Rossi, D. Wofsy, Efficacy and
cytokines and disease activity in SLE patients: data from LUMINA (LXXV), a safety of atacicept for prevention of flares in patients with moderate-to-
multiethnic US cohort, Lupus 21 (8) (2012) 830e835. severe systemic lupus erythematosus (SLE): 52-week data (APRIL-SLE
[110] H.K. Ziegler, E.R. Unanue, Decrease in macrophage antigen catabolism caused randomised trial), Ann. Rheum. Dis. 74 (11) (2015) 2006e2015.
by ammonia and chloroquine is associated with inhibition of antigen pre- [133] S.V. Navarra, R.M. Guzma n, A.E. Gallacher, S. Hall, R.A. Levy, R.E. Jimenez, et
sentation to T cells, Proc. Natl. Acad. Sci. U. S. A. 79 (1) (1982) 175e178. al., Efficacy and safety of belimumab in patients with active systemic lupus

Please cite this article in press as: L. Durcan, M. Petri, Immunomodulators in SLE: Clinical evidence and immunologic actions, Journal of
Autoimmunity (2016), http://dx.doi.org/10.1016/j.jaut.2016.06.010
12 L. Durcan, M. Petri / Journal of Autoimmunity xxx (2016) 1e12

erythematosus: a randomised, placebo-controlled, phase 3 trial, Lancet 377 Depletion of autoreactive immunologic memory followed by autologous
(9767) (2011) 721e731. hematopoietic stem cell transplantation in patients with refractory SLE in-
[134] W. Stohl, F. Hiepe, K.M. Latinis, M. Thomas, M.A. Scheinberg, A. Clarke, et al., duces long-term remission through de novo generation of a juvenile and
Belimumab reduces autoantibodies, normalizes low complement levels, and tolerant immune system, Blood 113 (1) (2009) 214e223.
reduces select B cell populations in patients with systemic lupus erythe- [143] L. Zhang, A.M. Bertucci, R. Ramsey-Goldman, R.K. Burt, S.K. Datta, Regulatory
matosus, Arthritis Rheum. 64 (7) (2012) 2328e2337. T cell (Treg) subsets return in patients with refractory lupus following stem
[135] R.F. van Vollenhoven, M.A. Petri, R. Cervera, D.A. Roth, B.N. Ji, C.S. Kleoudis, et cell transplantation, and TGF-beta-producing CD8þ Treg cells are associated
al., Belimumab in the treatment of systemic lupus erythematosus: high with immunological remission of lupus, J. Immunol. 183 (10) (2009)
disease activity predictors of response, Ann. Rheum. Dis. 71 (8) (2012) 6346e6358.
1343e1349. [144] D. Wang, X. Feng, L. Lu, J.E. Konkel, H. Zhang, Z. Chen, et al., A CD8 T cell/
[136] M.A. Dooley, F. Houssiau, C. Aranow, D.P. D’Cruz, A. Askanase, D.A. Roth, et indoleamine 2,3-dioxygenase axis is required for mesenchymal stem cell
al., Effect of belimumab treatment on renal outcomes: results from the phase suppression of human systemic lupus erythematosus, Arthritis Rheumatol.
3 belimumab clinical trials in patients with SLE, Lupus 22 (1) (2013) 63e72. 66 (8) (2014) 2234e2245.
[137] G.G. Illei, R. Cervera, R.K. Burt, A. Doria, F. Hiepe, D. Jayne, et al., Current state [145] M.A. Petri, P.J. Mease, J.T. Merrill, R.G. Lahita, M.J. Iannini, D.E. Yocum, et al.,
and future directions of autologous hematopoietic stem cell transplantation Effects of prasterone on disease activity and symptoms in women with
in systemic lupus erythematosus, Ann. Rheum. Dis. 70 (12) (2011) active systemic lupus erythematosus, Arthritis Rheum. 50 (9) (2004)
2071e2074. 2858e2868.
[138] L. Sun, Stem cell transplantation: progress in Asia, Lupus 19 (12) (2010) [146] A. Hartkamp, R. Geenen, G.L. Godaert, M. Bijl, J.W. Bijlsma, R.H. Derksen,
1468e1473. Effects of dehydroepiandrosterone on fatigue and well-being in women with
[139] A.M. Marmont, Immune ablation with stem-cell rescue: a possible cure for quiescent systemic lupus erythematosus: a randomised controlled trial, Ann.
systemic lupus erythematosus? Lupus 2 (3) (1993) 151e156. Rheum. Dis. 69 (6) (2010) 1144e1147.
[140] D. Jayne, J. Passweg, A. Marmont, D. Farge, X. Zhao, R. Arnold, et al., Autol- [147] S. Piantoni, L. Andreoli, M. Scarsi, A. Zanola, F. Dall’Ara, C. Pizzorni, et al.,
ogous stem cell transplantation for systemic lupus erythematosus, Lupus 13 Phenotype modifications of T-cells and their shift toward a Th2 response in
(3) (2004) 168e176. patients with systemic lupus erythematosus supplemented with different
[141] A.E. Traynor, J. Schroeder, R.M. Rosa, D. Cheng, J. Stefka, S. Mujais, et al., monthly regimens of vitamin D, Lupus 24 (4e5) (2015) 490e498.
Treatment of severe systemic lupus erythematosus with high-dose chemo- [148] R.A. Levy, V.S. Vilela, M.J. Cataldo, R.C. Ramos, J.L. Duarte, B.R. Tura, et al.,
therapy and haemopoietic stem-cell transplantation: a phase I study, Lancet Hydroxychloroquine (HCQ) in lupus pregnancy: double-blind and placebo-
356 (9231) (2000) 701e707. controlled study, Lupus 10 (6) (2001) 401e404.
[142] T. Alexander, A. Thiel, O. Rosen, G. Massenkeil, A. Sattler, S. Kohler, et al.,

Please cite this article in press as: L. Durcan, M. Petri, Immunomodulators in SLE: Clinical evidence and immunologic actions, Journal of
Autoimmunity (2016), http://dx.doi.org/10.1016/j.jaut.2016.06.010

You might also like