Gu2014 Article AllogeneicMesenchymalStemCellT

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Clin Rheumatol (2014) 33:1611–1619

DOI 10.1007/s10067-014-2754-4

ORIGINAL ARTICLE

Allogeneic mesenchymal stem cell transplantation for lupus


nephritis patients refractory to conventional therapy
Fei Gu & Dandan Wang & Huayong Zhang & Xuebing Feng &
Gary S. Gilkeson & Songtao Shi & Lingyun Sun

Received: 10 March 2014 / Revised: 7 July 2014 / Accepted: 17 July 2014 / Published online: 14 August 2014
# International League of Associations for Rheumatology (ILAR) 2014

Abstract Allogeneic mesenchymal stem cell transplantation achieved renal remission during 12-month visit by MSCT.
(MSCT) has been shown to be clinically efficacious in the Eleven of 49 (22.4 %) patients experienced renal flare by the
treatment of various autoimmune diseases. Here, we analyzed end of 12 months after a previous remission. Renal activity
the role of allogeneic MSCT to induce renal remission in evaluated by British Isles Lupus Assessment Group (BILAG)
patients with active and refractory lupus nephritis (LN). This scores significantly declined after MSCT (mean ± SD, from
is an open-label and single-center clinical trial conducted from 4.48±2.60 at baseline to 1.09±0.83 at 12 months), in parallel
2007 to 2010 in which 81 Chinese patients with active and with the obvious amelioration of renal function. Glomerular
refractory LN were enrolled. Allogeneic bone marrow- or filtration rate (GFR) improved significantly 12 months after
umbilical cord-derived mesenchymal stem cells (MSCs) were MSCT (mean ± SD, from 58.55±19.16 to 69.51±27.93 mL/
administered intravenously at the dose of 1 million cells per min). Total disease activity evaluated by Systemic Lupus
kilogram of bodyweight. All patients were then monitored Erythematosus Disease Activity Index (SLEDAI) scores also
over the course of 12 months with periodic follow-up visits decreased after treatment (mean ± SD, from 13.11±4.20 at
to evaluate renal remission, as well as possible adverse events. baseline to 5.48±2.77 at 12 months). Additionally, the doses
The primary outcome was complete renal remission (CR) and of concomitant prednisone and immunosuppressive drugs
partial remission (PR) at each follow-up, as well as renal were tapered. No transplantation-related adverse event was
flares. The secondary outcome included renal activity score, observed. Allogeneic MSCT resulted in renal remission for
total disease activity score, renal function, and serologic in- active LN patients within 12-month visit, confirming its use as
dex. During the 12-month follow-up, the overall rate of sur- a potential therapy for refractory LN.
vival was 95 % (77/81). Totally, 60.5 % (49/81) patients
Keywords Lupus nephritis . Mesenchymal stem cells .
Transplantation
Fei Gu and Dandan Wang contributed equally to this paper.
Electronic supplementary material The online version of this article
(doi:10.1007/s10067-014-2754-4) contains supplementary material,
which is available to authorized users. Introduction

F. Gu : D. Wang : H. Zhang : X. Feng : L. Sun (*)


Department of Rheumatology and Immunology, The Affiliated
Systemic lupus erythematosus (SLE) is an autoimmune dis-
Drum Tower Hospital of Nanjing University Medical School, 321 order often characterized by the development of glomerulo-
Zhongshan Road, Nanjing, Jiangsu 210008, People’s Republic of nephritis [1]. The general consensus is that 60 % of lupus
China patients will develop clinically relevant nephritis at some time
e-mail: lingyunsun@nju.edu.cn
in the course of their illness, and lupus nephritis (LN) is a
G. S. Gilkeson major cause of mortality among patients [2]. Randomized
Devision of Rheumatology, Medical University of South Carolina, trials indicated that long-term use of a combination of steroids
Charleston, USA and immunosuppressive agents, such as cyclophosphamide
(CYC) or mycophenolate mofetil (MMF), was effective for
S. Shi
Center for Craniofacial Molecular Biology, University of Southern LN patients [3, 4]. However, the rate of side effects is high,
California School of Dentistry, Los Angeles, SC, USA and some nonresponders were reported [5]. For refractory
1612 Clin Rheumatol (2014) 33:1611–1619

cases, intravenous immunoglobulins, plasma exchange, Rheumatology criteria for the classification of SLE [24].
immunoabsorption, and multitarget therapy, such as the cal- Clinical disease activity of lupus nephritis was defined by at
cineurin inhibitor tacrolimus, are promising options [6–8], but least two of the following: (1) Laboratory tests documented
the present clinical studies are all limited to small sample size active lupus nephritis three consecutive times: decrease in
with short follow-up visits and lack of strict criteria by which renal function (serum creatinine >106 μmol/L), increase in
to accurately determine complete remission [9, 10]. Novel proteinuria (defined as more than 1.0 g of protein excretion in
methods of induction therapy for refractory LN include the a 24-h urine specimen), and deterioration in microscopic
use of monoclonal antibodies that (1) modulate and/or deplete hematuria (defined as >10 red cells per high power field) or
B cells, including anti-CD20 and anti-CD22 antibodies, re- the presence of cellular casts; (2) serologic abnormality (in-
spectively, or (2) interfere with the stimulatory effects of the crease of anti-double-stranded DNA (dsDNA) antibodies or
soluble factor B-lymphocyte stimulator, i.e., anti-BLyS hu- deterioration of hypocomplementemia); (3) renal biopsy
man monoclonal antibody, belimumab (BmAb). The number documenting lupus nephritis according to the International
of case reports is continuously increasing, and they have Society of Nephrology/Renal Pathology Society classification
shown that these biologic agents are effective in the treatment of active or active/chronic lupus nephritis in renal biopsy class
of refractory LN. However, treatment-related adverse events III, class IV-S or IV-G, class V, class III + V, or class IV + V
have appeared after long-term applications, and the relapse (within 3 months) [25]. All patients were resistant to at least
rates observed in different studies ranged from 38 to 87 % 6 months of therapy with CYC and/or MMF in combination
[11]. Additionally, these drugs cause a heavy economic bur- with corticosteroid, according to conventional treatment stan-
den for patients, especially in developing countries [12]. dard. Written informed consent was obtained from each pa-
Hematopoietic stem cell transplantation (HSCT) has been tient. This study was approved by the Ethics Committee at
reported as a promising therapy to achieve long-term remis- The Affiliated Drum Tower Hospital of Nanjing University
sion in refractory LN [13], but the rates of relapse and Medical School and was conducted in compliance with cur-
treatment-related toxicity are high as is the development of a rent Good Clinical Practice standards and in accordance with
secondary autoimmune disorder [14]. the principles set forth under the Declaration of Helsinki
Mesenchymal stem cells (MSCs) possess immunomodula- (1989).
tory and reparative properties through specific interactions
with immune cells that participate in both innate and adaptive Treatment protocol
immune responses. MSCs exposed to an inflammatory micro-
environment can downregulate active immune responses. Allogeneic bone marrow MSCs were isolated from bone
Clinical trials focusing on MSCs to treat graft-versus-host marrow aspirates obtained from healthy donors, i.e., members
disease (GVHD) and other diseases are underway [15, 16]. of the patient’s immediate family, and all signed informed
In the past 6 years, we have applied allogeneic MSC trans- consents. Briefly, 20 mL of bone marrow was aspirated on
plantation in treating patients with refractory autoimmune the posterior iliac crest. Bone marrow mononuclear cells were
disorders, including SLE, systemic sclerosis, myositis, multi- separated by Ficoll density centrifugation. Mononuclear cells
ple sclerosis, inflammatory bowel disease, and rheumatoid were cultured in a 175-cm2 flask (Corning) with low-glucose
arthritis, and the clinical efficacy is encouraging [17–23]. Dulbecco’s modified Eagle’s medium (DMEM-LG, Gibco)
These cases also suggested a safety profile of allogeneic containing 10 % fetal bovine serum (FBS) (HyClone) and 1 %
MSC transplantation (MSCT) administered intravenously penicillin-streptomycin (Gibco) in a humidified incubator at
in vivo. 37 °C under 5 % CO2. Nonadherent cells were removed when
In the present study, we analyze the 12-month clinical the medium was exchanged on the third day. When the pri-
outcome and safety profile of allogeneic MSCT in 81 patients mary MSCs had expanded to 80 % confluence, they were
with refractory LN, who did not respond to conventional harvested and expanded to reach treatment dose based on the
treatment regimens. body weight of the recipient [17].
Patients who did not have appropriate bone marrow donors
were infused with umbilical cord (UC)-derived MSCs. UC
Methods MSCs were prepared by the Stem Cell Center of Jiangsu
Province. The cords were rinsed by phosphate-buffered saline
Patient selection (PBS) in penicillin and streptomycin, and the cord blood was
removed during this process. The washed cords were cut into
This study is an open-label clinical trial. From March 2007 to 1-mm2-sized pieces and floated in DMEM-LG containing
October 2010, 81 patients with active lupus nephritis refrac- 10 % FBS and 1 % penicillin-streptomycin (Gibco). The
tory to standard therapies were enrolled. All enrolled patients pieces of cord were subsequently incubated at 37 °C in humid
had fulfilled at least 4 of 11 American College of air with 5 % CO2. Nonadherent cells were removed by
Clin Rheumatol (2014) 33:1611–1619 1613

washing. The medium was replaced every 3 days after the activity, BILAG score of renal system, and changes in renal
initial plating. When well-developed colonies of fibroblast- function, serum albumin, and anti-dsDNA antibody levels
like cells appeared after 10 days, the cultures were trypsinized pre- and post-MSCT.
and passaged into a new flask for further expansion [18].
Criteria for release of MSCs for clinical use included Safety assessments
absence of visible clumps; spindle-shaped morphology; and
absence of contamination by pathogens, as documented by The incidence and severity of adverse events (AEs) were
aerobic and anaerobic cultures, as well as by virus for hepatitis monitored. Serious adverse events (SAEs), infusion-related
B surface antigen, hepatitis B core antibody, hepatitis C virus AEs (an AE occurring during or within 24 h following MSC
antibody, human immunodeficiency virus antibodies I and II, infusion), and infection-related AEs were summarized inde-
cytomegalovirus IgM, and syphilis antibody, as determined by pendently. Transplantation-related mortality included all
enzyme-linked immunosorbent assay (ELISA); cell viability deaths associated with transplantation of MSCs, except those
greater than 92 %, as determined by trypan blue testing and related to recurrence of underlying disease.
immune phenotyping proving expression of CD73, CD105,
CD90, and CD29 (>90 %); and absence of CD45, CD34, Statistical analysis
CD14, CD79, and HLA-DR (<2 %). In vitro osteogenic and
adipogenic induction of MSCs was also assayed. We used Rates of overall survival, complete and partial renal remission,
good manufacturing practice (GMP) conditions and clinical and relapse at different follow-up visits were calculated by
grade reagents to prepare the cells. All patients received a using the Kaplan-Meier method and were statistically tested
single intravenous infusion of BM- or UC-derived MSCs (1 with the log-rank test. The renal BILAG index was used to
million cells per kilogram of body weight per infusion). assess response, and scores were converted to numeric values
After MSCT, all patients were examined during follow-up (A=9, B=3, C=1, D=0, E=0) to enable evaluation [28]. We
visits at 1, 3, 6, and 12 months or termination as a result of calculated heart rates (HR) and their 95 % confidence intervals
death. Standard physical examinations, including blood pres- (CIs) using the univariate Cox proportional hazards model.
sure, cutaneous, and skeletal manifestations, were performed Unpaired t tests, χ2 tests, and Fisher’s exact tests were used as
by the same rheumatologist. Laboratory assessments, includ- appropriate. All analyses were performed by statistical soft-
ing complete blood cell count, routine urine test, 24-h protein- ware (SPSS 16.0). Statistical significance was set at P<0.05.
uria, kidney and liver function, serum albumin, and anti-
dsDNA antibody, were performed at each visit at the same
laboratory. Disease activity was assessed by Systemic Lupus
Erythematosus Disease Activity Index (SLEDAI). Renal ac- Results
tivity index was evaluated by British Isles Lupus Assessment
Group (BILAG) score for renal system. Participant characteristics

Outcome Eighty-one patients (74 females and 7 males) were enrolled in


this trial, and the mean disease duration was 83.1 months (6–
The primary end points were rates of overall survival, com- 264). Patient characteristics are listed in Table 1. All patients
plete and partial renal remission, and relapse within 12 months. were refractory to conventional treatments and underwent
Complete remission was defined as the return to normal MSCT; they then completed 12 months of follow-up visits.
values of serum creatinine (<106 μmol/L), proteinuria Twenty-three patients (23/81, 28 %) were given bone marrow-
(<0.3 g/24 h), and urine sediment. Partial remission was derived MSC infusion (two patients had son as donor, two
defined as improvement of no less than 50 % in all abnormal patients had daughter as donor, three patients had father as
renal measurements, without worsening (within 10 %) of any donor, three patients had husband as donor, four patients had
measurement. A renal flare was defined as the recurrence of mother as donor, four patients had sister as donor, and five
serum creatinine (>33 % increase within a 1-month period patients had brother as donor), and the other 58 patients (58/
directly attributed to lupus and confirmed 1 week later) or 81, 72 %) received UC-derived MSCs.
proteinuria (at least threefold increase within a 3-month period
accompanied by microscopic hematuria), irrespective of prior Primary outcome
complete or partial remission. Treatment failure was defined
as a condition requiring higher doses of corticosteroid or Overall survival and disease remission
immunosuppressant for disease control or failure to reach
complete or partial remission within 6 months [26, 27]. Key The overall survival rate during the 12-month follow-up peri-
secondary end points included SLEDAI score for disease od was 95 % (77/81). Survival rate was not correlated with
1614 Clin Rheumatol (2014) 33:1611–1619

Table 1 Baseline characteristics of patients renal flare, and another 5 patients (5/35, 14.3 %; 35
Demographics patients were in renal remission at 6 months) underwent
relapse in the following 6 months. The overall rate of
Age (year) 31.6 (12–55) relapse within 12 months was 22.4 % (11/49) (Fig. 1b).
Women/men (n) 74/7 Renal flare was not correlated with age (P=0.966), dis-
Duration (month) 83.1 (6–264) ease duration (P=0.939), MSC source (P=0.862), base-
Renal BILAG [n (mean)] 81 (4.48±2.60) line SLEDAI score (P = 0.919), or baseline proteinuria
Proteinuria [n, (g/24 h)] 81 (2.74±1.20) levels (P=0.187) by Cox regression analysis, but it was
Serum creatinine [n (μmol/L)] 33 (196.27±99.01) significantly correlated with baseline serum creatinine
Serum albumin [n (g/dL)] 61 (2.58±0.47) levels (P=0.003, OR=1.773, 95 % CI 1.213–2.591). A
GFR [n (mL/min)] 27 (58.55±19.16) total of 32 patients (32/81, 39.5 %) did not achieve either
Renal biopsy [n (%)] 13 (16.05) complete or partial remission within 12 months, an out-
SLEDAI score [n (mean)] 81 (13.11±4.20) come that was defined as treatment failure.
Cutaneous involvement [n (%)] 59 (72.84)
Musculoskeletal involvement [n (%)] 57 (70.37)
Hematologic involvement [n (%)] 36 (44.44) Secondary outcomes
Neuropsychiatric involvement [n (%)] 4 (4.94)
Baseline prednisolone [n (%)] 81 (100) Lupus nephritis improvements
Hydroxychloroquine [n (%)] 43 (53.09)
Cyclophosphamide [n (%)] 66 (81.48) Lupus nephritis activity assessed by BILAG score de-
Mycophenolate mofetil [n (%)] 19 (23.46) creased significantly 1 month after MSCT (mean ± SD
4.48±2.60 vs. 1.70±1.05, P<0.01) and continued to im-
BILAG British Isles Lupus Assessment Group, SLEDAI Systemic Lupus prove at 3 months (1.19 ± 0.92, P < 0.01 vs. baseline
Erythematosus Disease Activity Index levels). The average renal BILAG score at 6 months was
1.29 ± 1.33, which demonstrated significant difference
disease duration (P=0.880) or the source of MSCs (P=0.265) compared to baseline levels and 1-month follow-up visit
by Cox regression analysis. The probability of complete re- (both P <0.01). Moreover, the BILAG score decreased
mission was 17.5 % (14/80) at 3 months, 18.2 % (14/77) at significantly by the 12-month follow-up visit (1.09 ±
6 months, and 23.4 % (18/77) at 12 months. A total 30.9 % 0.83, P < 0.01 vs. baseline levels and 1-month visit;
(25/81) of patients experienced complete remission during the Fig. 2a). All enrolled patients had defined proteinuria at
12 months. By Cox regression analysis, we found that com- baseline (>0.5 g/24 h), but proteinuria decreased from
plete remission was not correlated with age (P=0.475), dis- 2.74±1.20 to 2.06±1.04 g at 1 month, 1.74±1.21 g at
ease duration (P=0.480), MSC source (P=0.916), or baseline 3 months, 1.56±1.03 g at 6 months, and 1.52±1.04 g at
SLEDAI score (P=0.231), while it was significantly correlat- 12 months, with statistical differences at each follow-up
ed with baseline proteinuria levels (P=0.003, OR=0.517, visit (P < 0.001; Fig. 2b). There was no difference in
95 % CI 0.336–0.794) and baseline serum creatinine levels amelioration of proteinuria between bone marrow and
(P=0.047, OR=0.471, 95 % CI 0.224–0.990). UC-derived MSCTs (supplementary Fig. 1).
Within the 12-month follow-up period, partial remission Thirty-three patients (33/81, 41 %) had abnormal serum
was achieved in 22.5 % (18/80) of patients at 3 months, creatinine levels at baseline (mean ± SD, 195.9 ±
27.3 % (21/77) of patients at 6 months, and 20.8 % (16/77) 98.6 μmol/L). After allogeneic MSCT, serum creatinine
of patients at 12 months. Overall, 60.5 % (49/81) of patients declined to 186.0 ± 116.8 μmol/L at 1 month, 187.8 ±
achieved complete or partial remission during 12 months 160.9 μmol/L at 3 months, 172.9 ± 124.4 μmol/L at
(Fig. 1a). We also found that partial renal remission was not 6 months, and 162.3 ± 94.7 μmol/L at 12 months, with
correlated with age (P=0.711), duration (P=0.897), MSC statistical difference at 12 months (P=0.0057, Fig. 2c).
source (P=0.202), baseline SLEDAI score (P=0.566), or For 31 patients (31/81, 38 %) who had elevated serum urea
baseline serum creatinine levels (P=0.056) by Cox regression, nitrogen at baseline (mean±SD, 18.27±9.49 μmol/L), this
but it was statistically correlated with baseline proteinuria index improved significantly 3 and 12 months post-MSCT
levels (P=0.039, OR=0.762, 95 % CI 0.588–0.986). (15.12 ± 6.81 μmol/L at 3 months, P = 0.04 vs. baseline
level; 11.74 ± 4.66 μmol/L at 12 months, P < 0.001 vs.
Relapse baseline level, Fig. 2d). Furthermore, glomerular filtration
rate (GFR) improved significantly 12 months after alloge-
By the end of 6 months, 6 out of 32 patients (18.8 %; 32 neic MSC infusion (mean ± SD, from 58.55 ± 19.16 to
patients were in renal remission at 3 months) experienced 69.51±27.93 mL/min, n=27, P=0.046).
Clin Rheumatol (2014) 33:1611–1619 1615

Fig. 1 Cumulative probability of achieving a renal remission and b relapse for patients infused with mesenchymal stem cells (MSCs) during 12 months,
by Kaplan-Meier survival analysis

Total disease activity score and serologic ameliorations and the average dose of CYC was tapered from 0.80±0.19 to
0.46±0.19 g/month at 12 months (mean ± SD, P<0.001, n=
Total disease activity assessed by SLEDAI score improved 63, three patients died, Fig. 4b and Table 2), according to
significantly after allogeneic MSC infusions (mean ± SD, from disease remission, without the addition of other immunosup-
13.11±4.20 at baseline to 9.64±3.86 at 1 month; 7.59±3.04 at pressive drugs.
3 months; 6.12±2.90 at 6 months; 5.48±2.77 at 12 months, n=
81, P<0.01, Fig. 3a). In addition, the mean serum albumin
concentration for 61 patients increased (2.58±0.47 g/dL at Safety
baseline, 2.85±0.48 g/dL at 1 month, 3.24±0.51 g/dL at
3 months, 3.48±0.50 g/dL at 6 months, and 3.55±0.59 g/dL Serious adverse events
at 12 months, all P<0.01 vs. baseline levels, Fig. 3b) in parallel
with the decline of proteinuria. Serum anti-dsDNA antibody Four of 81 patients died after MSCT, all judged to be from
declined after MSCT without reaching statistical difference at events unrelated to treatment. One patient died 156 days after
each follow-up visit (mean ± SD, from 630.1±792.6 IU/mL at MSCT from disease relapse after an acute gastroenteritis and
baseline to 475.5±633.5 IU/mL at 1 month, 485.0±633.9 IU/ heart failure. Two other patients succumbed to disseminated
mL at 3 months, 466.5±583.4 IU/mL at 6 months, and 410.3± pulmonary infections and uncontrolled lupus nephritis 78 and
372.4 IU/mL at 12 months, n=18, P>0.05, Fig. 3c). 179 days after MSCT, respectively. One patient had relapse of
her lupus at 4 months, with pulmonary hypertension, and she
Maintenance therapy after mesenchymal stem cell died from right-sided heart failure 245 days after MSC
transplantation infusion.

Prednisone dosage was tapered 5–10 mg every 2 weeks in the


first month following transplantation according to clinical Infection-related adverse events
status and laboratory indicators of disease remission. At the
end of 12 months, 64 out of 77 patients (83 %) had their Two patients experienced enteritis and diarrhea 3 and 4 months
prednisone dosage tapered, and 59 out of 77 patients (77 %) after MSCT, respectively, with transient increase of serum
had their prednisone tapered to 5–10 mg per day (mean ± SD, creatinine, which was controlled by conventional treatments.
10.32±4.12 mg/day at 12 months vs. 22.24±12.78 mg/day at Two other patients had herpes virus infection 1 and 6 months
baseline, n=77, P<0.001, Fig. 4a). Sixty-six out of 81 patients after MSCT, respectively, with no renal flare. None of the AEs
(81 %) had CYC for immunosuppressive therapy at baseline, were considered to be related to MSC infusion.
1616 Clin Rheumatol (2014) 33:1611–1619

A ** B
** ***
** 8 ***
10
** ***
*
6 *** *

Proteinuria (g/24h)
8
Renal BILAG score

6
4

0
0

th
T

s
s

s
th

th

th
SC

on

on

on

on
T

th

M
th

th

th
SC

m
on

m
on

on

on

1
M

or
m

12
m

m
e

ef
1
or

12

B
ef
B

C D
** ***
*
1000 60

Serum urea nitrogen (micro-mol / litre)


Serum creatinine (micro-mol / litre)

800
*
40
600

400
20

200

0 0
T

th

s
T

th

th

th

th
th

th

SC
th
SC

on
on

on

on

on
on

on

on

M
M

m
m

m
m

e
e

1
1

or
or

12
3

12

ef
ef

B
B

Fig. 2 Baseline and repeated measurements of a renal BILAG score, b 24-h proteinuria, c serum creatinine, and d serum urea nitrogen for patients
infused with allogeneic mesenchymal stem cells (MSCs). *P<0.05 vs. pre-MSCT, **P<0.01 vs. pre-MSCT, ***P<0.001 vs. pre-MSCT

A
**
B ** C
** **
***
** ** * 4000
Serum anti-dsDNA antibody (IU/ml)

30 5
** *** ***
** ***
*** 3000
Serum albumin (g/dl)

**
4

**
SLEDAI score

20

2000
3

10

1000
2

0
0
1
T

th

s
T

th

th

th

th
SC
th

th

th
SC

on
T

th

s
on

on

on

on
th

th

th
SC
on

on

on

M
on

m
M

on

on

on

m
m

e
m

1
e

or
m

m
or

12
3

e
12

ef
or
ef

12

B
ef
B

Fig. 3 Measurements of a SLE Disease Activity Index score, b serum albumin, and c serum anti-dsDNA antibody for patients who underwent
allogeneic mesenchymal stem cell (MSC) transplantation. **P<0.01 vs. pre-MSCT
Clin Rheumatol (2014) 33:1611–1619 1617

A B
100 *** 2.0 ***
Dose of prednisone (mg/day)

Dose of CYC (gm/month)


80
1.5

60
1.0

40

0.5
20

0 0.0
Before MSCT 12 months Before MSCT 12 months
Fig 4 Doses of prednisone (a) and cyclophosphamide (CYC, b) were tapered for patients after allogeneic mesenchymal stem cell transplantation
(MSCT). ***P<0.001 vs. pre-MSCT

Discussion patients who were unresponsive to conventional immuno-


suppression treatments, especially previous regular intra-
MSCs provide stromal support for HSCs in the bone marrow venous CYC. In our previous studies, we reported the
compartment, as well as generate different cell types, such as effect of allogeneic MSCs on refractory SLE patients, in
osteoblasts, adipocytes, chondroblasts, and myoblasts [29, whom the changes of renal functional indices had also
30]. Recently, this cell type has been thought to possess wide been analyzed [17, 18]. However, the small number of
immunoregulatory properties on various immune cells and patients and short follow-up period limited further appli-
differential potential to multiple tissues [31, 32]. Many obser- cations. Thus, it was worthwhile studying the role of allo-
vations have illuminated the role of MSCs in acute or chronic geneic MSCT in LN patients. In the present group of
renal injury models [33, 34]. However, the clinical application patients, allogeneic MSCT could induce renal remission
of MSCs on human renal disease has been limited to prevent by the end of 12 months. Renal function obviously im-
acute renal transplant rejection [35], and to the best of our proved after MSCT. Furthermore, the total disease activity
knowledge, no study has reported on MSCT for human renal score evaluated by SLEDAI markedly improved after
disorder. MSCT. Thus, our data suggest that allogeneic MSCT is
Therefore, this open-label clinical study analyzes the role effective in inducing renal remission for both active and
of allogeneic MSCT as an alternative therapy in active LN refractory lupus nephritis within 12 months.
Further, Cox regression analysis demonstrated significant
Table 2 Treatments used pre- and post-allogeneic MSCT
correlations between complete renal remission and baseline
proteinuria, as well as serum creatinine levels, and partial renal
Drug and dosage No. of patients involved (n) remission was also correlated with baseline proteinuria.
Furthermore, renal flare was more likely to occur in patients
Pre-MSCT Post-MSCT
with higher baseline serum creatinine levels. All these data
Glucocorticoid (prednisone or its equivalence) indicate that baseline renal functional index can influence total
≥20 mg/day 49 6 prognosis by MSCT. Those who had higher proteinuria or
>10 and <20 mg/day 20 12 serum creatinine at baseline were less likely to achieve renal
≤10 mg/day 12 59 remission and had a higher incidence of renal flare. This
Cyclophosphamide (per month) observation was also confirmed by a recently published ani-
>0.8 g 7 0 mal model study performed by Chang et al. who found that
>0.6 and ≤0.8 g 45 8 UC blood-derived MSCT at an early age in lupus mice
>0.4 and ≤0.6 g 13 18 (2 months old) showed significant therapeutic effect on lupus
≤0.4 g 1 37 nephritis, as demonstrated by the inhibition of proteinuria,
Mycophenolate mofetil amelioration of survival rate, serum creatinine, as well as
1.5–2.0 g/day 15 4
glomerular proliferation, and sclerosis scores, while mice
≤1 g/day 4 8
treated at 6 months of age failed to achieve a satisfactory
clinical improvement [36]. These data support the view that
MSCT mesenchymal stem cell transplantation early MSCT may be more efficacious in lupus nephritis.
1618 Clin Rheumatol (2014) 33:1611–1619

Based on these findings, the clinical efficacy in LN patients cannot draw any conclusion concerning the role of allogeneic
appears promising; however, mechanisms underlying the ef- MSCT in patients of other ethnic backgrounds, indicating the
fects of MSCT in vivo remain to be elucidated. To address urgent need for a controlled randomized study with subject of
this, we must first understand the homing of infused MSCs. In diverse ethnic origins.
an acute kidney injury model, Togel et al. have shown that In conclusion, our data suggest that treatment with alloge-
administered MSCs were predominantly detected in the glo- neic MSCT is efficacious for patients with active and refrac-
meruli and attached peritubular capillaries by in vivo micros- tory lupus nephritis, showing a favorable safety profile and
copy [37]. Our previous study also demonstrated that the positive results within 12 months.
infused MSCs could home to the kidney of MRL/lpr lupus
mice to facilitate therapeutic potential in vivo, while no in-
Acknowledgments The authors thank Professor Wanjun Chen (Muco-
fused MSCs were detected in normal mice [38]. In an animal
sal Immunology Unit, Oral Infection and Immunity Branch, National
model of acute tubular epithelial injury, Herrera et al. also Institute of Dental and Craniofacial Research, National Institutes of
reported the tubular localization of several MSC-GFP + cells, Health, MD, USA) for helpful comments. This work was supported by
with significant accumulation even after 21 days of infusion the Major International (Regional) Joint Research Project (no.
81120108021, to Dr. Sun), National Natural Science Foundation of China
[33]. All the data indicate that the infused MSCs could,
(no. 81273304, to Dr. Sun), and Jiangsu Province Kejiao Xingwei Pro-
indeed, home to the injured kidney. Second, it is necessary gram (to Dr. Sun), Jiangsu Provincial Natural Science Foundation
to understand how MSCs afford local renal amelioration. (BK20140098, to Dr. Wang), and Jiangsu Provincial Health Department
Some studies have shown that the infused MSCs could differ- Youth Foundation (Q201411, to Dr. Wang)and National Natural Science
Foundation of China (no. 81302557, to Dr. Gu).
entiate into functional tubular cells, which could physically
replace lost kidney cells and participate in renal repair in vivo Conflict of interest Fei Gu, none; Dandan Wang, none; Huayong
[34]. In contrast, primary paracrine factors can also medi- Zhang, none; Xuebing Feng, none; Gary S. Gilkeson, none; Songtao
ate the protective effects of MSCs. For example, soluble Shi, none; Lingyun Sun, none.
factors, such as hepatocyte growth factor (HGF), insulin
growth factor (IGF)-1, and vascular endothelial growth
factor (VEGF), are involved in the antiapoptotic and
immunomodulatory effect in the local microenvironment References
[37, 39, 40]. Finally, apart from local immunoregulation
and tissue repair, MSCs can provide general effect in the 1. Mills JA (1994) Systemic lupus erythematosus. N Engl J Med
recipient. A large number of studies found that serum 330(26):1871–1879
proinflammatory cytokines, such as TNF-α, interferon 2. Ward MM (2000) Changes in the incidence of end-stage renal disease
due to lupus nephritis, 1982-1995. Arch Intern Med 160(20):3136–
gamma (IFN-γ), IL-6, and IL-1β, significantly decreased 3140
after MSCT, while the anti-inflammatory cytokine 3. Austin HA 3rd, Klippel JH, Balow JE et al (1986) Therapy of lupus
transforming growth factor beta (TGF-β) and regulatory nephritis. Controlled trial of prednisone and cytotoxic drugs. N Engl J
T cells increased, suggesting that MSCs can facilitate Med 314(10):614–619
4. Boumpas DT, Austin HA 3rd, Vaughn EM et al (1992) Controlled
immunobalance in vivo [36, 41]. Our animal studies trial of pulse methylprednisolone versus two regimens of pulse
showed that UC-MSCT could inhibit renal MCP-1 and cyclophosphamide in severe lupus nephritis. Lancet 340(8822):
HMGB-1 expressions but upregulate Foxp3+ Treg cells in 741–745
lupus mice models [38]. Moreover, the clinical studies did 5. Houssiau FA, Vasconcelos C, D’Cruz D et al (2002)
Immunosuppressive therapy in lupus nephritis: the Euro-Lupus
confirm the immunoregulation of Treg and cytokines like Nephritis Trial, a randomized trial of low-dose versus high-dose
IL-4 and IFN-γ, as well as TGF-β, by MSCT in lupus intravenous cyclophosphamide. Arthritis Rheum 46(8):2121–2131
patients [17, 18], suggesting that immunomodulation is 6. Zandman-Goddard G, Blank M, Shoenfeld Y (2009) Intravenous
the most important mechanism for MSCT in treating immunoglobulins in systemic lupus erythematosus: from the bench
to the bedside. Lupus 18(10):884–888
lupus nephritis. 7. Jayne DR, Gaskin G, Rasmussen N, Abramowicz D, Ferrario
The present study has some limitations. First, this is not a F, Guillevin L et al (2007) Randomized trial of plasma ex-
controlled and randomized study. We still lack of a group of change or high-dosage methylprednisolone as adjunctive ther-
patients with conventional therapies, but not combined with apy for severe renal vasculitis. J Am Soc Nephrol 18(7):
2180–2188
allogeneic MSC infusion. Therefore, the current data only 8. Bao H, Liu ZH, Xie HL, Hu WX, Zhang HT, Li LS (2008)
provide evidence that allogeneic MSCT could induce renal Successful treatment of class V+IV lupus nephritis with multitarget
remission on the basis of other drugs taken by patients en- therapy. J Am Soc Nephrol 19(10):2001–2010
rolled in this study. Still, such evidence is sufficient to indicate 9. Glassock RJ (2008) Multitarget therapy of lupus nephritis: base hit or
home run? J Am Soc Nephrol 19(10):1842–1844
the use of allogeneic MSCT as a treatment method for refrac- 10. Pons-Estel GJ, Serrano R, Plasín MA, Espinosa G, Cervera R (2011)
tory LN. Second, this is a single-center clinical study, and all Epidemiology and management of refractory lupus nephritis.
patients enrolled were of Asian ethnicity. Therefore, we Autoimmun Rev 10(11):655–663
Clin Rheumatol (2014) 33:1611–1619 1619

11. Bertsias G, Boumpas DT (2008) Update on the management of lupus 27. Houssiau FA, D’Cruz D, Sangle S et al (2010) Azathioprine versus
nephritis: let the treatment fit the patient. Nat Clin Pract Rheumatol mycophenolate mofetil for long-term immunosuppression in lupus
4(9):464–472 nephritis: results from the MAINTAIN Nephritis Trial. Ann Rheum
12. Aringer M, Burkhardt H, Burmester GR et al (2012) Current state of Dis 69(12):2083–2089
evidence on “off label therapeutic options for systemic lupus erythe- 28. Ehrenstein MR, Conroy SE, Heath J, Latchman DS, Isenberg DA
matosus, including biological immunosuppressive agents, in (1995) The occurrence, nature and distribution of flares in a cohort of
Germany, Austria, and Switzerland—a consensus report. Lupus patients with systemic lupus erythematosus: a rheumatological view.
21(4):386–401 Br J Rheumatol 34(3):257–260
13. Burt RK, Traynor A, Statkute L et al (2006) Nonmyeloablative 29. Méndez-Ferrer S, Michurina TV, Ferraro F et al (2010) Mesenchymal
hematopoietic stem cell transplantation for systemic lupus erythema- and haematopoietic stem cells form a unique bone marrow niche.
tosus. JAMA 295(5):527–535 Nature 466(7308):829–834
14. Nagy A, Quaggin SE (2010) Stem cell therapy for the kidney: a 30. Singer NG, Caplan AI (2011) Mesenchymal stem cells: mechanisms
cautionary tale. J Am Soc Nephrol 21(7):1070–1072 of inflammation. Ann Rev Pathol 6:457–478
15. Djouad F, Bouffi C, Ghannam S, Noël D, Jorgensen C (2009) 31. Nauta AJ, Fibbe WE (2007) Immunomodulatory properties of mes-
Mesenchymal stem cells: innovative therapeutic tools for rheumatic enchymal stromal cells. Blood 110(10):3499–3506
diseases. Nat Rev Rheum 5(7):392–399 32. Hoogduijn MJ, Popp F, Verbeek R et al (2010) The immunomodu-
16. English K, French A, Wood K (2010) Mesenchymal stromal cells: latory properties of mesenchymal stem cells and their use for immu-
facilitators of successful transplantation? Cell Stem Cell 7(4):431–442 notherapy. Int Immunopharm 10(12):1496–1500
17. Liang J, Zhang H, Hua B et al (2010) Allogenic mesenchymal stem 33. Herrera MB, Bussolati B, Bruno S et al (2004) Mesenchymal stem
cells transplantation in refractory systemic lupus erythematosus: a cells contribute to the renal repair of acute tubular epithelial injury. Int
pilot clinical study. Ann Rheum Dis 69(8):1423–1429 J Mol Med 14(6):1035–1041
18. Sun L, Wang D, Liang J et al (2010) Umbilical cord mesenchymal 34. Li K, Han Q, Yan X, Liao L, Zhao RC (2010) Not a process of simple
stem cell transplantation in severe and refractory systemic lupus vicariousness, the differentiation of human adipose-derived mesen-
erythematosus. Arthritis Rheum 62(8):2467–2475 chymal stem cells to renal tubular epithelial cells plays an important
19. Akiyama K, Chen C, Wang D et al (2012) Mesenchymal stem cell role in acute kidney injury repairing. Stem Cells Dev 19(8):1267–
induced immunoregulation involves Fas Ligand/Fas-mediated T cell 1275
apoptosis. Cell Stem Cell 10(5):544–555 35. Tan J, Wu W, Xu X et al (2012) Induction therapy with autologous
20. Wang D, Zhang H, Cao M et al (2011) Efficacy of allogeneic mesenchymal stem cells in living-related kidney transplants: a ran-
mesenchymal stem cell transplantation in patients with drug- domized controlled trial. JAMA 307(11):1169–1177
resistant polymyositis and dermatomyositis. Ann Rheum Dis 70(7): 36. Chang JW, Hung SP, Wu HH et al (2011) Therapeutic effects of
1285–1288 umbilical cord blood-derived mesenchymal stem cell transplantation
21. Liang J, Zhang H, Hua B et al (2009) Allogenic mesenchymal stem in experimental lupus nephritis. Cell Transplant 20(2):245–257
cells transplantation in treatment of multiple sclerosis. Mult Scler 37. Tögel F, Hu Z, Weiss K, Isaac J, Lange C, Westenfelder C (2005)
15(5):644–646 Administered mesenchymal stem cells protect against ischemic acute
22. Liang J, Zhuang H, Wang D et al (2012) Allogenic mesenchymal renal failure through differentiation-independent mechanisms. Am J
stem cell transplantation in seven patients with refractory inflamma- Physiol Renal Physiol 289(1):F31–F42
tory bowel disease. Gut 61(3):468–469 38. Gu Z, Akiyama K, Ma X et al (2010) Transplantation of umbilical
23. Liang J, Li X, Zhang H et al (2012) Allogeneic mesenchymal stem cord mesenchymal stem cells alleviates lupus nephritis in MRL/lpr
cells transplantation in patients with refractory RA. Clin Rheumatol mice. Lupus 19(13):1502–1514
31(1):157–161 39. Chen Y, Qian H, Zhu W et al (2011) Hepatocyte growth factor
24. Smith EL, Shmerling RH (1999) The American College of modification promotes the amelioration effects of human umbilical
Rheumatology criteria for the classification of systemic lupus ery- cord mesenchymal stem cells on rat acute kidney injury. Stem Cells
thematosus: strengths, weaknesses, and opportunities for improve- Dev 20(1):103–113
ment. Lupus 8(8):586–595 40. Villanueva S, Ewertz E, Carrión F et al (2011) Mesenchymal stem
25. Weening JJ, D’Agati VD, Schwartz MM et al (2004) The classifica- cell injection ameliorates chronic renal failure in a rat model. Clin Sci
tion of glomerulonephritis in systemic lupus erythematosus revisited. (Lond) 121(11):489–499
J Am Soc Nephrol 15(2):241–250 41. Semedo P, Correa-Costa M, Antonio Cenedeze M et al (2009)
26. Ginzler EM, Dooley MA, Aranow C et al (2005) Mycophenolate Mesenchymal stem cells attenuate renal fibrosis through immune
mofetil or intravenous cyclophosphamide for lupus nephritis. N Engl modulation and remodeling properties in a rat remnant kidney model.
J Med 353(21):2219–2228 Stem Cells 27(12):3063–3073

You might also like