You are on page 1of 9

ORIGINAL ARTICLE

Inhibition of EGFR Tyrosine Kinase by Erlotinib


Prevents Sclerodermatous Graft-Versus-Host Disease
in a Mouse Model
Florence Morin1,2,3, Niloufar Kavian1,2,3, Wioleta Marut1, Christiane Chéreau1, Olivier Cerles1,
Philippe Grange1, Bernard Weill1,2, Carole Nicco1,3 and Frédéric Batteux1,2,3

Chronic graft-versus-host disease (GVHD) follows allogeneic hematopoietic stem cell transplantation. It results
from alloreactive processes induced by minor histocompatibility antigen incompatibilities leading to the
activation of CD4 T cells and the development of fibrosis and inflammation of the skin and visceral organs and
autoimmunity that resemble systemic sclerosis. EGFR is a ubiquitous cell receptor deeply involved in cell
proliferation, differentiation, and motility. EGFR has recently been implicated in autoimmune and fibrotic
diseases. Therefore, we tested whether Erlotinib, an EGFR tyrosine kinase inhibitor, can prevent sclerodermatous
GVHD (Scl-GVHD). Scl-GVHD was induced in BALB/c mice by B10.D2 bone marrow and spleen cell
transplantation. Transplanted mice displayed severe clinical symptoms including alopecia, fibrosis of the skin
and visceral organs, vasculitis, and diarrhea. The symptoms were reversed in mice treated with Erlotinib. These
beneficial effects were mediated by the decreased production of activated/memory CD4+ T cells and the
reduction in T-cell infiltration of the skin and visceral organs along with a decrease in IFN-γ and IL-13 production
and autoimmune B-cell activation. The improvement provided by Erlotinib in the mouse model of Scl-GVHD
supplies a rationale for the evaluation of Erlotinib in the management of patients affected by chronic GVHD.
Journal of Investigative Dermatology (2015) 135, 2385–2393; doi:10.1038/jid.2015.174; published online 28 May 2015

INTRODUCTION and B cells from donor to induce chronic GVHD (Linhares


Chronic graft-versus-host disease (GVHD) is a side effect of et al., 2013; Nishimori et al., 2013; Socié and Ritz, 2014).
allogeneic hematopoietic stem cell transplantation, with Sclerodermatous GVHD (Scl-GVHD) represents 10 to 15% of
variable clinical presentations (Baird and Pavletic, 2006). cases of chronic GVHD (Vogelsang et al., 2003). This clinical
Chronic GVHD results from alloreactive processes induced form, similar to scleroderma, includes fibrotic changes and
by minor major histocompatibility complex incompatibilities chronic inflammation of the skin, lung, and gastrointestinal
between donor and recipient and involves donor-derived tract. Murine Scl-GVHD, produced by transplanting B10.D2
immune cells and host cell populations. Its pathophysiology is (H-2d) bone marrow and spleen cells across minor
less well known than acute GVHD (Ferrara et al., 2009). histocompatibility loci into lethally irradiated BALB/cJ (H-2d)
Chronic GVHD often mimics autoimmune diseases recipients, displays many clinical and histologic similarities
(Filipovich et al., 2005). Initially, chronic GVHD was associ- with scleroderma (Jaffee and Claman, 1983). Syngeneic
ated with a T helper type 2 (Th2)-type immune response, but (BALB/c into BALB/c) transplanted animals are used as
IFN and IL-17 seem implicated in its induction. Furthermore, controls. This animal model summarizes the clinical features
loss of immune tolerance by impaired thymic negative of human Scl-GVHD including fibrosis of the skin and inner
selection allows expansion and activation of autoreactive T organs and infiltrating cutaneous mononuclear cells (pre-
dominantly T cells) starting 14 days following bone marrow
1
INSERM U 1016, Institut Cochin, Université Paris Descartes Sorbonne Paris and splenocyte transplantation (Yamamoto, 2010).
Cité, Faculté de Médecine, Paris, France and 2Laboratoire d’Immunologie
biologique, Hôpital Cochin, AP-HP, Paris, France
We have observed in a mouse model of scleroderma that
angiotensin II elicits skin and visceral fibrosis that can be
Correspondence: Frédéric Batteux, Laboratoire d’Immunologie, INSERM U
1016, Institut Cochin, 8 rue Méchain, 75679 Paris Cedex 14, France. abrogated by angiotensin II receptor blockade using Irbesar-
E-mail: frederic.batteux@cch.aphp.fr tan (Marut et al., 2013). Angiotensin II increases fibroblast
3
These authors contributed equally to this work. proliferation, myofibroblasts differentiation, and the accumu-
Abbreviations: ALT, alanine aminotransferase; AST, aspartate lation of type I collagen through the phosphorylation of
aminotransferase; AU, arbitrary unit; eGFP, enhanced green fluorescent Smad3, a transcription factor implicated in the activation of
protein; pEGFR, phospho-EGFR; GVHD, graft-versus-host disease; Scl-GVHD,
sclerodermatous graft-versus-host disease the type I collagen gene, and through the activation of the
Received 26 August 2014; revised 3 April 2015; accepted 18 April 2015; nicotinamide adenine dinucleotide phosphate oxidase and the
accepted article preview online 4 May 2015; published online 28 May 2015 overproduction of reactive oxygen species (Yang et al., 2009).

© 2015 The Society for Investigative Dermatology www.jidonline.org 2385


F Morin et al.
Erlotinib Prevents Scl-GVHD

The latter phenomenon is responsible for EGFR phos- a Syngeneic GVH


phorylation. The transactivation of EGFR by angiotensin
initiates the proliferation and migration of fibroblasts, a
phenomenon that can be abrogated by AT1 antagonist
(Yahata et al., 2006). This link between EGFR and fibrosis
explains the attenuation of liver fibrosis and of the develop-
ment of hepatocellular carcinoma by EGFR inhibition using
Erlotinib (Fuchs et al., 2014).
Erlotinib is a potent inhibitor of EGFR tyrosine kinase used
b pEGFR
to treat advanced or metastatic non-small-cell lung cancer
after failure of first-line or second-line chemotherapy (Akita β-Actin
and Sliwkowski, 2003; Johnson et al., 2005; Robert, 2011). In Syn Syn Syn Syn GVH GVH GVH GVH
addition to its antiproliferative properties against tumor cells, Erlo Erlo Erlo Erlo
Erlotinib inhibits T-cell proliferation and Th1-cell–mediated pEGFR
immune response in vitro and in vivo through the inhibition of 0.8
*** *
the c-Raf/extracellular signal–regulated kinase cascade and
the Akt signaling pathway (Luo et al., 2011). 0.7
As GVHD involves both Th1 cell–mediated immune response
and fibrosis, we were prompted to investigate the effects of

AU
0.6
Erlotinib on the development of chronic Scl-GVHD in mice.

RESULTS 0.5
Phospho-EGFR expression in mouse ears from GVHD animals
and modulation by Erlotinib 0.4
Levels of phospho-EGFR (pEGFR) were assessed by immuno- Syn Syn-Erlo GVH GVH-Erlo
histochemistry on ear pinna sections and by western blot on
Figure 1. Effects of Erlotinib on mouse ear skin following graft-versus-host
lysates of ear lobes. pEGFR expression was greater in mouse disease (GVHD) induction and oral administration of Erlotinib (Erlo).
ear pinna that developed Scl-GVHD than in control mice (a) Immunohistochemical study of phospho-EGFR (pEGFR) on 7 μm ear skin
with a syngeneic graft (Figure 1a). This result was confirmed sections in syngeneic (Syn) BALB/c mice and in GVHD mice, revealed with
by western blot. pEGFR expression was increased by 32% diaminobenzidine tetrahydrochloride. (b) Western blot of pEGFR expression in
compared with syngeneic control mice (P = 0.043, Figure 1b). the ear skin of BALB/c mice submitted to a syngeneic or a nonsyngeneic graft.
Photographs were taken with a Nikon Eclipse 80i microscope. Original
Erlotinib decreased the expression of phospho-EGFR com- magnification × 50. Values in b are mean ± SEM. *P ≤ 0.05; ***P ≤ 0.001.
pared with untreated GVHD mice (0.67 ± 0.019 vs. 0.53 ± AU, arbitrary unit.
0.025 arbitrary unit (AU), P = 0.0027, Figure 1b).

Disease severity score Lymphocytic infiltration of the skin


The clinical score assessment started 10 days after transplan- Cellular infiltration of the skin of Erlotinib-treated GVHD
tation. At day 23, no difference between groups was mice was lower than in untreated animals (3.80 ± 0.55 vs.
observed. At day 26, the score of the GVHD group reached 12.86 ± 0.88, P = 0.0008, Figure 3a and b). Costaining with
1.11 ± 0.3 AU, whereas the score of the syngeneic group phycoerythrin-labeled anti-CD3 mAb showed that green
remained null (Po0.0001, Figure 2a). At the time of killing on donor cells were CD3-T lymphocytes (10.57 ± 0.48 vs.
day 33, a significant improvement in the Erlotinib group was 3.00 ± 0.30, Po0.0001, Figure 3a and b). IFN-γ production
observed versus untreated mice (2.39 ± 0.20 vs. 1.65 ± 0.10 by infiltrating lymphocytes was significantly increased in
AU, P = 0.047 at day 33, Figure 2a). GVHD mice compared with syngeneic mice (236.5 ± 73.3 vs.
5,828 ± 896 pg ml− 1, Po 0.0001, Figure 3c). Erlotinib sig-
Skin involvement nificantly decreased IFN-γ secretion (3,143 ± 463.7 vs.
At 7 days after transplantation, ear thickness was increased in 5,828 ± 896.0 pg ml − 1 in untreated animals, P = 0.0178,
GVHD groups compared with syngeneic groups (30.0 ± 0.4 Figure 3c). IL-13 secretion in the skin significantly increased
vs. 42.2 ± 0.8 μm, Po0.0001 at day 17, Figure 2b). From day in GVHD mice compared with syngeneic mice (1,420 ± 241
23, a decrease in earlobe thickening was observed in GVHD vs. 124 ± 32 pg ml − 1, P = 0.0002, Figure 3c). Erlotinib
mice treated with Erlotinib compared with untreated animals. decreased IL-13 secretion (792 ± 180 vs. 1,420 ± 241 pg ml − 1
At the time of killing, the mean ear pinna thickness was lower in untreated animals, P = 0.036, Figure 3c). These results have
in Erlotinib-treated GVHD mice than in untreated mice been confirmed by quantitative real-time reverse-transcrip-
(45.9 ± 1.2 vs. 35.8 ± 1.4 μm, Po0.0001, Figure 2b). Sirius tase–PCR for IFN-γ and IL-13. IFN-γ mRNAs were higher in
red staining showed an increased collagen deposition in the skin of GVHD mice than in syngeneic mice (7.69 ± 0.50-
GVHD mice pinna skin versus the three other groups foldvs. syngeneic mice, P = 0.0028, Figure 3d). Following
(Figure 2c). Erlotinib, IFN-γ mRNA levels in GVHD mice returned to

2386 Journal of Investigative Dermatology (2015), Volume 135


F Morin et al.
Erlotinib Prevents Scl-GVHD

a 3 b 55
Syn
Syn-Erlo

Ear thickness (μm)


50

Clinical score - AU
GVH
2 GVH-Erlo 45
*
40 ***
1 35
30
0 25
0 10 20 30 40 0 10 20 30 40
Days Days

c
Syngeneic Syngeneic Erlotinib

GVH GVH Erlotinib

Figure 2. Effects of Erlotinib (Erlo) on clinical features following graft-versus-host disease (GVHD) induction and oral administration of Erlotinib. (a) Evolution
of the GVHD score in the different groups during Erlotinib treatment. (b) Evolution of ear skin thickness during Erlotinib treatment. (c) Representative Sirius
red–dyed ear skin sections of 7 μm, showing fibrosis in syngeneic (Syn) BALB/c mice and in GVHD mice, treated by Erlotinib or not. Photographs were taken
with a Nikon Eclipse 80i microscope. Original magnification × 50. Values in a and b are mean ± SEM. *P ≤ 0.05; ***P ≤ 0.001. Bars represent mean ± SEM. AU,
arbitrary unit.

initial levels (1.03 ± 0.04-fold vs. 7.69 ± 0.50-fold in the 55.7 ± 5.85 UI l − 1, Po0.05, and AST: 464.71 ± 79.37 vs.
untreated GVHD group, P = 0.0029, Figure 3d). IL-13 mRNAs 160.33 ± 12.38 UI l − 1, Po0.01, Figure 4b and c). Erlotinib
were increased in GVHD mice compared with syngeneic decreased transaminase levels in GVHD mice (ALT: 104.88 ±
(3.69 ± 0.07-fold vs. syngeneic mice, Po0.0001, Figure 3d). 12.60 vs. 184.57 ± 31.15 UI l − 1, Po0.05, and AST: 291.25 ±
This level was decreased following Erlotinib compared with 17.04 vs. 464.71 ± 79.37 UI l − 1, Po0.05, Figure 4b and c).
untreated mice (0.52 ± 0.04-fold vs. 3.69 ± 0.07-fold in the Liver sections from untreated GVHD mice showed portal
untreated GVHD group, Po0.0001, Figure 3d). CCL17 inflammation and bile duct destruction (Figure 4a). GVHD
mRNAs were overexpressed in the skin of GVHD mice mice treated with Erlotinib displayed a significant reduction in
compared with syngeneic mice (3.34 ± 0.54-fold vs. syn- liver lesions compared with untreated GVHD animals
geneic mice, P = 0.0017). Erlotinib abolished the overexpres- (0.63 ± 0.12 vs. 2.01 ± 0.47 AU, Po0.05). Mice grafted with
sion of CCL17 in GVHD mice (0.71 ± 0.41-fold vs. syngeneic bone marrow and splenocytes, treated or not with
3.34 ± 0.54-fold in the untreated GVHD group, P = 0.0026, Erlotinib, showed no liver inflammation (0.25 ± 0.07 vs.
Figure 3d). Similarly, CCL21 mRNAs were higher in GVHD 0.19 ± 0.08 AU, P = nonsignificant).
mice than in syngeneic mice (6.51 ± 0.29-fold vs. syngeneic
mice, P = 0.0016; Figure 3d). This mRNA level was decreased Effects of Erlotinib on B- and T-cell responses
in Erlotinib-treated GVHD mice compared with untreated Anti-DNA topoisomerase I autoantibodies were increased
mice (1.23 ± 0.10-fold vs. 6.51 ± 0.29-fold in the untreated in GVHD mice compared with untreated syngeneic mice
GVHD group, P = 0.0020; Figure 3d). No difference in CCL27 (Po0.001, Figure 5a). Erlotinib significantly reduced the
mRNAs was found among the four groups (Figure 3d). levels of anti-DNA topoisomerase I autoantibodies (1.15 ±
0.23 vs. 2.65 ± 0.82 AU, Po0.001, Figure 5a). Furthermore,
Prevention of GVHD-associated liver disease by Erlotinib Erlotinib reduced the number of B cells by 43% in syngeneic
Serum alanine aminotransferase (ALT) and aspartate mice compared with the untreated group (Po0.001,
aminotransferase (AST) were higher in GVHD -untreated Figure 5b). No difference in B-cell counts was observed
mice than in syngeneic groups (ALT: 184.6 ± 31.15 vs. between treated and untreated GVHD mice.

www.jidonline.org 2387
F Morin et al.
Erlotinib Prevents Scl-GVHD

a c IFN-γ
8 000
GVH GVH Erlotinib *
6 000

pg ml–1
4 000
2 000
0

lo

rlo
VH
Sy

Er

-E
G
n-

VH
Sy

G
GVH GVH Erlotinib IL-13
3 000

pg ml–1
2 000
*
1 000

lo

rlo
VH
Sy

Er

-E
G
n-

VH
Sy

G
b d qRT-PCR
Syn
eGFP Anti-CD3 10 Syn-Erlo
15 GVH
**
Mean cell numbers

15 ***
Mean cell numbers

8 GVH-Erlo
***

Fold increase
**
10 10 6
4 *** **
5 5
2
0 0 0
rlo
rlo

13

1
VH

7
VH

7
L2
N

L1

L2
-E
-E

IL
IF
G
G

C
C

C
VH
VH

C
C

C
G
G

Figure 3. Effects of Erlotinib (Erlo) on mouse skin following graft-versus-host disease (GVHD) induction and oral administration of Erlotinib. (a) Representative
5 μm frozen skin sections with enhanced green fluorescent protein (eGFP) donor cells and phycoerythrin-conjugated anti-CD3 mAb staining. (b) Scores obtained
by enumerating eGFP- and anti-CD3-positive cells in 10 areas per frozen skin section. (c) Levels of IFN-γ and IL-13 produced by skin cells and used as
markers of T helper type 1 and 2 (Th1 and Th2) responses. (d) Levels of mRNAs for IFN-γ, IL-13, CCL17, CCL21, and CCL27 in the skin. Photographs
were taken with a Nikon Eclipse 80i microscope. Original magnification: × 100. Values in b, c, and d are mean ± SEM. *P ≤ 0.05; **P ≤ 0.01; ***P ≤ 0.001.
qRT-PCR, quantitative real-time reverse-transcriptase–PCR; Syn, syngeneic.

Similarly, CD4 T cells decreased by 40% in the Erlotinib- inhibitor Erlotinib prevents skin and visceral lesions through
treated syngeneic group compared with the untreated group an inhibition of the allogeneic response.
(Po0.05, Figure 5c). There was no effect on CD8 cell Experimental chronic GVHD shares characteristic features
numbers in treated and untreated syngeneic mice (Figure 5d). with scleroderma, including skin and visceral fibrosis,
If the levels of CD4 and CD8 T cells were significantly inflammation with systemic sclerosis, and autoimmunity.
decreased in untreated GVHD mice compared with untreated Erlotinib improves the clinical outcome of sublethally
syngeneic animals (93 and 87%, respectively Po0.001, irradiated BALB/c mice transplanted with B10.D2 hemato-
Figure 5c and d), no significant difference between the levels poietic cells. Fibrosis, alopecia, and vasculitis are clearly
of CD4 and of CD8 T cells was observed between treated and reduced in treated versus untreated mice. EGFR is a
untreated GVHD animals (Figure 5c and d ). There was an ubiquitous cell receptor involved in proliferation, differentia-
increase in the percentages of CD4 and CD8 memory T cells tion, and cell motility (Schneider and Wolf, 2009). This
within CD4 and CD8 T-cell subsets in GVHD mice compared receptor has a role in tissue fibrosis through the control of
with syngeneic animals (1.6 ± 1.4% vs. 83 ± 3%, Po 0.001, fibroblast proliferation and migration (Pastore et al., 2008).
Figure 5e and 50 ± 17% vs. 83 ± 7%, Po0.001, Figure 5f). Moreover, EGFR signaling promotes fibrosis through a trans-
Erlotinib did not influence the percentage of CD4+ and CD8+ forming growth factor-β–dependent mechanism, implicating
T cells in the syngeneic group (Figure 5e and f). Erlotinib activation of smad2/3 in various tissues like kidney or skin.
induced an increase in percent CD4 naive T cells in GVHD This could explain why EGFR inhibition by Erlotinib attenu-
mice compared with untreated mice (45% vs. 17 ± 1.6 ± ates liver fibrosis and the development of hepatocellular
1.4%, Po0.001, Figure 5e), whereas it had no effect on CD8 carcinoma (Fuchs et al., 2014).
naive T cells. Liver involvement is a common feature in human GVHD
that also occurs in the Scl-GVHD model (Jaffee and Claman,
DISCUSSION 1983; Lee et al., 2002). Indeed, in GVHD mice, we found
Here we show that Scl-GVHD is associated with an activation a periportal lymphocytic infiltration associated with an
of the EGFR pathway whose inhibition by the tyrosine kinase increase in serum transaminase levels. Erlotinib decreased

2388 Journal of Investigative Dermatology (2015), Volume 135


F Morin et al.
Erlotinib Prevents Scl-GVHD

a Syngeneic Syngeneic Erlotinib

GVH GVH Erlotinib

b 250 c
600 *
*
200

AST (UI I–1)


ALT (UI I–1)

400
150

100
200
50

0 0
Syn Syn-Erlo GVH GVH-Erlo Syn Syn-Erlo GVH GVH-Erlo
Figure 4. Effects of Erlotinib (Erlo) on liver involvement following graft-versus-host disease (GVHD) induction and oral administration of Erlotinib.
(a) Hematoxylin–eosin staining of liver sections. GVH and GVH Erlo groups showed mixed lymphomonocytic infiltrations around the portal vein with a major
reduction in the GVH Erlo group. (b) Serum aspartate aminotransferase (AST) levels in various experimental groups. (c) Serum alanine aminotransferase (ALT)
levels in various experimental groups. Photographs were taken using a Nikon Eclipse 80i microscope. The arrows showed mixed lymphocytic infiltrations around
the portal vein. Original magnification × 100. Values in b and c are mean ± SEM. *P ≤ 0.05. Syn, syngeneic.

transaminase levels and reduced the periportal lymphocytic natural killer cells. CCL17 produced by dendritic cells,
infiltrate. Thus, in addition to its direct antifibrotic properties keratinocytes, fibroblasts, and endothelial cells (Saeki and
(Fuchs et al, 2014), Erlotinib inhibits the EGFR pathway and, Tamaki, 2006) has been implicated in various skin diseases
consecutively, abrogates tissue infiltration and inflammation such as atopic dermatitis and bullous pemphigoid (Furudate
of the liver. et al., 2014; Hamilton et al., 2014). CCL21 is the ligand of
The pathophysiology of chronic GVHD is still poorly CCR7 expressed by T cells, B cells, and dendritic cells
understood. However, evidence suggests that, unlike acute (Comerford et al., 2013). It has been implicated in contact
GVHD that is CD8+ T-cell dependent, the events observed in allergic dermatitis (Eberhard et al., 2004).
chronic GVHD depend on the activation of donor CD4+ In addition to its antiproliferative properties against tumor
T cells specific of minor histocompatibility antigen (Korngold cells, Erlotinib exerts inhibitory effects on T-cell proliferation.
and Sprent, 1978; Kansu, 2004; Shlomchik, 2007; Chu and Erlotinib causes G0/G1 arrest and inhibits the secretion of IL-2
Gress, 2008). After transplantation of spleen and bone and the phosphorylation of c-Raf, extracellular signal–
marrow cells, naive CD4+ T cells from B10.D2 initiate the regulated kinase, and Akt (Luo et al., 2011). Erlotinib acts as
disease in irradiated BALB/c mice. Activated donor CD4+ other antiproliferative molecules like steroids, cyclosporine,
T cells infiltrate the skin and induce fibrosis (Korngold and sirolimus, or mycophenolate that are treatments for chronic
Sprent, 1978; Kansu, 2004; Chu and Gress, 2008). In our GVHD (Wolff et al., 2010, 2011 ). Erlotinib impairs Th1-
hands, the beneficial effect of Erlotinib was associated with a mediated immune response as it inhibits the secretion of IFN-γ
reduction in the percentages of effector memory CD4+ T cells by activated T cells in vitro and ameliorates in vivo picryl
(CD69 −CD4+CD44highCD62Llow) but not of CD8+ T cells in chloride–induced ear contact dermatitis (Luo et al., 2011).
GVHD mice. IFN-γ is involved in the pathophysiology of chronic GVHD
This reduction is associated with a diminution in infiltrating (Korholz et al., 1997; Bruggen et al., 2014). It induces an
T cells in the skin and with a profound decrease in CCL17 and overexpression of HLA and adhesion molecules in target
CCL21 expression. Increased CCL17 mRNAs have already tissues and favors the recruitment of macrophages. Therefore,
been shown by Zhou et al. (2007) in murine Scl-GVHD. its inhibition by Erlotinib can participate in the improvement
CCL17 is the ligand of CCR4 on Th2 cells, basophils, and of GVHD. Recently, the immunomodulating role of Erlotinib

www.jidonline.org 2389
F Morin et al.
Erlotinib Prevents Scl-GVHD

a Anti-DNA Topoisomerase b Chronic GVHD is associated with the production of


I Abs B cells
4 20 autoantibodies consecutive to the production of Th2 cyto-
*** *** kines (Ruzek et al., 2004; Kavian et al., 2012). In line with

×106 B cells
3 15
previous reports, we observed a production of anti-DNA-
AU

2 NS 10
topoisomerase I antibodies in Scl-GVHD mice that was
1 5
NS decreased by Erlotinib. We conclude that Erlotinib targets
0 rlo 0 CD4+ T cells, blocks T- and B-cell cooperation in Scl-GVHD
n

lo

rlo
lo
VH

mice, and prevents the development of autoimmunity in

vH
Sy

Sy
Er

Er
-E

-E
G
G
n-

n-
VH

vH
our model.
Sy

Sy

G
G

c d This work highlights the role of the EGF receptor pathway


CD4 T cells CD8 T cells
1 500 400 in the pathophysiology of Scl-GVHD. The inhibition of EGFR
NS
* phosphorylation by Erlotinib reduces tissue inflammation and
×106 T cells

×106 T cells

300
1 000
200 fibrosis, limits the production of specific autoantibodies, and
500 NS prevents the production of CD4+ memory T cells. Inhibition of
NS 100
the EGFR pathway acts on the three main components of Scl-
0
0
GVHD: fibrosis, inflammation, and autoimmunity. These
n
n

lo

rlo

rlo
lo
vH

vH
Sy
Sy

Er

Er

mechanisms explain the beneficial effects of Erlotinib


-E

-E
G

G
n-

n-
vH

vH
Sy

Sy

observed and evidence the possible usefulness of this


G

e CD4 naive/memory+naive f CD8 naive/memory+naive molecule for the treatment of chronic GVHD in humans.
T cells T cells
100 NS 100 NS
80 80
MATERIALS AND METHODS
NS Animals, cells, and chemicals
60 *** 60
%
%

40 40
Six-week-old female BALB/c mice (Janvier Laboratory, Le Genest
20 20
Saint Isle, France) and male B10.D2-enhanced green fluorescent
0 0
protein (eGFP) mice (gift from Colette Kanellopoulos-Langevin,
CDTA–CNRS–Orléans, France) were used in all experiments.
rlo

rlo
lo
n

rlo
vH

vH
Sy

Sy
Er

-E

-E
-E
G

G
n-

Animals received humane care according to the guidelines of our


vH

vH
vH
Sy

G
G

institution (Inserm and Université Paris Descartes—CEEA34 Ethics


Figure 5. Inhibition of the production of autoantibodies in mice with graft- committee). All mice were housed in ventilated cages with sterile
versus-host disease (GVHD) systemic sclerosis treated in vivo with Erlotinib
food and water ad libitum. All cells were cultured as reported
(Erlo). Inhibition of the production of T cells with sclerodermatous GVHD (Scl-
previously (Kavian et al., 2010). All chemicals were obtained
GVHD) treated in vivo with Erlotinib. (a) Levels of anti-topoisomerase I
antibodies (Abs), as measured by ELISA. (b) Splenic B-cell numbers, as from Sigma-Aldrich (St Louis, MO), except for Erlotinib (Roche
assessed by flow cytometry. (c) Splenic CD4 T-cell numbers, as assessed by Registration, Meylan, France).
flow cytometry. (d) Splenic CD8 T-cell numbers, as assessed by flow
cytometry. (e) Ratio between naive and memory CD4 cells. (f) Ratio between Experimental procedure for induction of GVHD in BALB/c mice
naive and memory CD8 cells. Values in a–f are mean ± SEM. *P ≤ 0.05; GVHD following splenocyte and bone marrow transplantation was
***P ≤ 0.001. AU, arbitrary unit; NS, nonsignificant; Syn, syngeneic.
induced in BALB/c mice (H-2d; Janvier Laboratory) by grafting cells
from 7- to 8-week-old male B10.D2-eGFP mice (H-2d; Janvier
Laboratory), as previously described (Kavian et al., 2012). Briefly,
was observed following the treatment of head and neck recipient mice were lethally irradiated with 750 cGy from a
squamous cell carcinoma by an EGFR inhibitor that induces Gammacel [137Cs] source. After 3 hours, they were injected
the production of transforming growth factor-β and intravenously with donor spleen cells (5.106 per recipient) and
prostaglandin E2 by the tumor cells that attenuate antitumor bone marrow cells (2.106 per recipient), previously treated with a
immune responses (Kumai et al., 2014). Therefore, the hypotonic solution of potassium acetate for red blood cell lysis and
immunomodulating effects of Erlotinib associate intrinsic suspended in RPMI-1640. A control group of BALB/c recipient mice
effects on T-cell signaling and extrinsic effects through received syngeneic BALB/c spleen and bone marrow cells (syngeneic
paracrine immunomodulating signals from another target bone marrow transplantation, referred to as control animals).
cell population. In addition to IFN-γ, IL-13 is also highly
expressed in the skin of GVHD mice. Erlotinib decreases the Treatment of Scl-GVHD mice with Erlotinib
production of IL-13 in their skin. Following the graft of B10. Scl-GVHD and control mice were randomized and treated per os
D2 lymphoid cells into irradiated BALB/c mice, an increase in 5 days a week for 4 weeks with either Erlotinib (50 mg kg − 1 per day)
the expression of type 2 cytokines in the skin of Scl-GVHD or vehicle (methylcellulose 1%) alone, beginning on day 7 after bone
mice has already been observed (Zhou et al., 2007). marrow transplantation (9 mice per group). Animals were killed by
Moreover, in other chronic GVHD models, induction of cervical dislocation 7 weeks after transplantation.
GVHD fibrosis of the skin and of visceral organs requires type
2 immune responses. Consequently, the reduced production Disease severity score
of IL-13 in our model probably contributes to the amelioration To determine the incidence and severity of disease, we assigned
of skin fibrosis. a score to each mouse using the following criteria: 0: no external

2390 Journal of Investigative Dermatology (2015), Volume 135


F Morin et al.
Erlotinib Prevents Scl-GVHD

sign; 1: piloerection on back and underside, 1: hunched posture microscopy (Olympus BX60) by a pathologist who was blinded to
or lethargy; 0.5: alopecia o1 cm2; 1: alopecia 41 cm2; 1: vasculitis the experimental group assignment. Degree of inflammation of
(one or more purpural lesions on ears or tail); and 1: eyelid sclerosis coded microscopic sections of liver was graded from 0 to 4, as
(blepharophymosis). The severity score is the sum of these values and previously described (Nagler et al., 2000).
ranges from 0 (unaffected) to a maximum of 6. The incidence and
severity score were recorded every week by two blinded scientists. Serum AST and ALT activities
Serum activity of AST and ALT were used as markers of hepatocyte
Skin thickness cytolysis. AST and ALT activities were quantified using a standard
Skin thickness of the right and left ears lobes of mice was measured clinical automatic analyser (Modular PP, Roche Diagnostics, Meylan,
each week with a digital calliper and expressed in mm. France).

Histopathological analysis Immunohistochemistry of skin tissue sections


Liver and skin pieces fixed in formol were embedded in paraffin. Frozen skin tissue sections of 5 μm thickness were stained for 1 hour
Then, 5-mm-thick tissue sections were stained with hematoxylin and at room temperature with 1:50 phycoerythrin-labeled hamster
eosin or Picrosirius Red. Slides were examined by standard bright- anti-mouse CD3 (clone 145-2C11; BD Bioscience, San Jose, CA) or
field microscopy (Olympus BX60, Rungis, France) by a pathologist with the respective phycoerythrin-labeled control isotype (BD
who was blinded to the experimental group assignment. Bioscience). After extensive washing in phosphate-buffered saline,
slides were viewed with an Olympus microscope equipped with an
Skin collagen content epifluorescence system to view eGFP- and phycoerythrin-labeled
Skin pieces were dilacerated using a scalpel, put into tubes, thawed, cells. Images were collected using a digital camera with × 400
and mixed with pepsin (1:10 weight ratio) and 0.5 M acetic acid magnification objective. Hematopoietic donor cells appear in green
overnight at room temperature under stirring. The assay of collagen (B10.D2-eGFP) and in red for CD3+ T cells.
content was based on the quantitative dye-binding Sircol method
(Biocolor, Belfast, Ireland). Determination of IFN-γ and IL-13 production in the skin
Punch biopsies (6 mm diameter) from the skin of each mouse were
Immunohistochemistry of pEGFR proteins in mouse ear skin incubated for 72 hours at 37 °C in complete RPMI medium with
Sections of ear pinna, 7 μm thick, were placed onto slides, 5 μg ml − 1 concanavalin A. Supernatants were collected and frozen
deparaffinized, and then rehydrated. Antigen retrieval, endogenous at − 80 °C. Cytokine concentrations from 1:4 diluted supernatants
peroxidase blocking, and nonspecific binding blocking were were measured by ELISA. “Mouse IFN gamma ELISA Ready-SET-Go!”
performed as previously described (Marut et al., 2013). Sections and “Mouse IL-13 ELISA Ready-SET- Go!” kits were purchased from
were then incubated with 1:50 rabbit polyclonal anti-pEGFR (Y1092) eBioscience (San Diego, CA). Concentrations were calculated from a
(ab40815, Abcam, Paris, France) overnight at 4 °C. Incubation with standard curve according to the manufacturer’s protocol. Results are
horseradish peroxidase–labeled secondary goat anti-rabbit and expressed in pg ml − 1.
staining with diaminobenzidine tetrahydrochloride were performed
as previously described (Marut et al., 2013). Pictures were taken with Determination of IL-13, IFN-γ, CCL17, CCL21, and CCL27
a digital camera on a BX60 microscope (Olympus), using DP mRNA levels in the skin
Manager Software, at × 100 magnification. Immunostaining was RNA isolation was realized as described by Grange et al. (2009).
analyzed using ImageJ software (version 1.36; National Institutes of Amplification was achieved with 50 ng of total RNA for each sample.
Health, Bethesda, MD) for quantitative assessment of staining Real-time PCR was carried out with iTaq Universal SYBR Green
positivity. Results are expressed as percentages of positive skin area One-Step Kit (Bio-Rad, Hercules, CA). We used the 5′-GCTTGCC
analyzed with ImageJ software. TTGGTGGTCTCGCC-3′ and 5′-GGGCTACACAGAACCCGCCA-3′
primers for IL-13, the 5′-CAGCAACAGCAAGGCGAAAAAGG-3′ and
Western blot of pEGFR proteins in mouse ears skin 5′-TTTCCGCTTCCTGAGGCTGGAT-3′ primers for IFN-γ, the 5′-GG
Skin fragments of earlobes were incubated with 250 μl of RIPA buffer TACCATGAGGTCACTTCAGA-3′ and 5′-CACTCTCGGCCTACATT
for western blot. Protein extracts (30 μg total protein) were resolved as GGT-3′ primers for CCL17, the 5′-GGGAAACAAAGCCCCGG-3′
previously described (Marut et al., 2013). Blots were incubated and 5′-GCTGTGTCTGTTCAGTTCTCTTG-3′ primers for CCL21, the
overnight at 4 °C with a 1:500 rabbit polyclonal anti-pEGFR (Y1092) 5′-CTGCTGAGGAGGATTGTC-3′ and 5′-CACGACAGCCTGGAG
(ab40815, Abcam). Revelation and analysis were performed as GTGA-3′ primers for CCL27, and the 5′-TGCCGAGGATTTGG
previously described (Marut et al., 2013). Results are expressed as AAAAAG-3′ and 5′-CCCCCCCTTGAGCACACAG-3′ primers for
AU, defined as the ratio between the densitometric intensity of the HRPT that was used as a control.
bands of the proteins tested and bands labeled with mouse anti-β-
actin mAb. Serum anti-DNA topoisomerase I autoantibodies
Levels of anti-DNA topoisomerase I IgG antibodies were assayed by
Liver inflammation ELISA using purified calf thymus DNA topoisomerase I bound to the
At the time of killing, livers were removed from all mice in both wells of a microtiter plate (Abnova, Taipei City, Taiwan). Mouse
groups and kept in formol. Fixed liver pieces were embedded in serum (1:4) and 1:1,000 anti-murine Ig horseradish peroxidase
paraffin and stained with hematoxylin and eosin by standard (Dako, Glostrup, Denmark) secondary antibody were used as
procedure. Slides were examined by standard bright-field previously described (Servettaz et al., 2009; Kavian et al., 2012).

www.jidonline.org 2391
F Morin et al.
Erlotinib Prevents Scl-GVHD

Flow cytometric analysis of spleen cell subsets Grange PA, Raingeaud J, Calvez V et al. (2009) Nicotinamide inhibits
Cell suspensions from spleens were prepared after hypotonic lysis of Propionibacterium acnes-induced IL-8 production in keratinocytes
through the NF-kappaB and MAPK pathways. J Dermatol Sci 56:
erythrocytes in potassium acetate solution. Cells were incubated with 106–12
the appropriate labeled antibody at 4 °C for 30 minutes in phosphate- Hamilton JD, Suárez-Fariñas M, Dhingra N et al. (2014) Dupilumab improves
buffered saline with 2% normal fetal calf serum. Flow cytometry was the molecular signature in skin of patients with moderate-to-severe atopic
performed using a FACS Fortessa flow cytometer (BD Biosciences), dermatitis. J Allergy Clin Immunol 134:1293–300
according to standard techniques. The mAbs used in this study Jaffee BD, Claman HN (1983) Chronic graft-versus-host disease (GVHD) as a
model for scleroderma. I. Description of model systems. Cell Immunol 77:
were anti-B220-PECy7, anti-CD11b-biotin and Streptavidin-PerCP, 1–12
anti-CD11c-PE on one hand and anti-CD69-BV421, anti-CD44-PE, Johnson JR, Cohen M, Sridhara R et al. (2005) Approval summary for erlotinib
anti-CD62L-APC, anti-CD4-PerCP, and anti-CD8-PE-Cy7 (BD for treatment of patients with locally advanced or metastatic non-small
Biosciences) on the other hand. Data were analyzed with FlowJo cell lung cancer after failure of at least one prior chemotherapy regimen.
Clin Cancer Res Off J Am Assoc Cancer Res 11:6414–21
software (Tree Star, Ashland, OR).
Kansu E (2004) The pathophysiology of chronic graft-versus-host disease. Int J
Hematol 79:209–15
Statistical analysis Kavian N, Servettaz A, Mongaret C et al. (2010) Targeting ADAM-17/notch
All quantitative data are expressed as mean ± SEM. Data were signaling abrogates the development of systemic sclerosis in a
compared using one-way analysis of variance followed by murine model. Arthritis Rheum 62:3477–87
Dunn’s multiple comparison test. Student’s unpaired t-test was used Kavian N, Marut W, Servettaz A et al. (2012) Arsenic trioxide prevents murine
sclerodermatous graft-versus-host disease. J Immunol 188:5142–9
when comparing two groups. All analyses were carried out using the
Korholz D, Kunst D, Hempel L et al. (1997) Decreased interleukin 10 and
GraphPad Prism 5.0 statistical software package (San Diego, CA),
increased interferon-gamma production in patients with chronic graft-
and P-values of o0.05 were considered significant. versus-host disease after allogeneic bone marrow transplantation. Bone
Marrow Transplant 19:691–5
Korngold R, Sprent J (1978) Lethal graft-versus-host disease after bone
CONFLICT OF INTEREST marrow transplantation across minor histocompatibility barriers in mice.
The authors state no conflict of interest. Prevention by removing mature T cells from marrow. J Exp Med 148:
1687–98
ACKNOWLEDGMENTS Kumai T, Oikawa K, Aoki N et al. (2014) Tumor-derived TGF-β and
We thank A Colle for typing the manuscript and N Saidu for reviewing the prostaglandin E2 attenuate anti-tumor immune responses in head and
manuscript. This work was supported by grants from University Paris neck squamous cell carcinoma treated with EGFR inhibitor. J Transl Med
Descartes. 12:265
Lee SJ, Klein JP, Barrett AJ et al. (2002) Severity of chronic graft-versus-host
disease: association with treatment-related mortality and relapse. Blood
REFERENCES 100:406–14
Akita RW, Sliwkowski MX (2003) Preclinical studies with Erlotinib (Tarceva). Linhares YPL, Pavletic S, Gale RP (2013) Chronic GVHD: Where are we?
Semin Oncol 30:15–24 Where do we want to be? Will immunomodulatory drugs help? Bone
Baird K, Pavletic SZ (2006) Chronic graft versus host disease. Curr Opin Marrow Transplant 48:203–9
Hematol 13:426–35 Luo Q, Gu Y, Zheng W et al. (2011) Erlotinib inhibits T-cell-mediated immune
Bruggen MC, Klein I, Greinix H et al. (2014) Diverse T-cell responses response via down-regulation of the c-Raf/ERK cascade and Akt signaling
characterize the different manifestations of cutaneous graft-versus-host pathway. Toxicol Appl Pharmacol 251:130–6
disease. Blood 123:290–9 Marut W, Kavian N, Servettaz A et al. (2013) Amelioration of systemic
Chu Y-W, Gress RE (2008) Murine models of chronic graft-versus-host disease: fibrosis in mice by angiotensin II receptor blockade. Arthritis Rheum 65:
insights and unresolved issues. Biol Blood Marrow Transplant J Am Soc 1367–77
Blood Marrow Transplant 14:365–78 Nagler A, Pines M, Abadi U et al. (2000) Oral tolerization ameliorates liver
Comerford I, Harata-Lee Y, Bunting MD et al. (2013) A myriad of functions disorders associated with chronic graft versus host disease in mice.
and complex regulation of the CCR7/CCL19/CCL21 chemokine axis Hepatology 31:641–8
in the adaptive immune system. Cytokine Growth Factor Rev 24: Nishimori H, Maeda Y, Tanimoto M (2013) Chronic graft-versus-host disease:
269–83 disease biology and novel therapeutic strategies. Acta Med Okayama 67:
Eberhard Y, Ortiz S, Ruiz Lascano A et al. (2004) Up-regulation of the 1–8
chemokine CCL21 in the skin of subjects exposed to irritants. BMC Pastore S, Mascia F, Mariani V et al. (2008) The epidermal growth factor
Immunol 5:7 receptor system in skin repair and inflammation. J Invest Dermatol 128:
Ferrara JLM, Levine JE, Reddy P et al. (2009) Graft-versus-host disease. Lancet 1365–74
373:1550–61 Robert J (2011) [Tyrosine kinase inhibitors]. Bull Cancer (Paris) 98:1321–34
Filipovich AH, Weisdorf D, Pavletic S et al. (2005) National Institutes of Health Ruzek MC, Jha S, Ledbetter S et al. (2004) A modified model of graft-versus-
consensus development project on criteria for clinical trials in chronic host-induced systemic sclerosis (scleroderma) exhibits all major aspects of
graft-versus-host disease: I. Diagnosis and staging working group report. the human disease. Arthritis Rheum 50:1319–31
Biol Blood Marrow Transplant J Am Soc Blood Marrow Transplant 11:
945–56 Saeki H, Tamaki K (2006) Thymus and activation regulated chemokine (TARC)/
CCL17 and skin diseases. J Dermatol Sci 43:75–84
Fuchs BC, Hoshida Y, Fujii T et al. (2014) Epidermal growth factor receptor
inhibition attenuates liver fibrosis and development of hepatocellular Schneider MR, Wolf E (2009) The epidermal growth factor receptor ligands at
carcinoma. Hepatol Baltim Md 59:1577–90 a glance. J Cell Physiol 218:460–6
Furudate S, Fujimura T, Kambayashi Y et al. (2014) Comparison of CD163+ Servettaz A, Goulvestre C, Kavian N et al. (2009) Selective oxidation of DNA
CD206+ M2 macrophages in the lesional skin of bullous pemphigoid and topoisomerase 1 induces systemic sclerosis in the mouse. J Immunol 182:
pemphigus vulgaris: the possible pathogenesis of bullous pemphigoid. 5855–64
Dermatology 229:369–78 Shlomchik WD (2007) Graft-versus-host disease. Nat Rev Immunol 7:340–52

2392 Journal of Investigative Dermatology (2015), Volume 135


F Morin et al.
Erlotinib Prevents Scl-GVHD

Socié G, Ritz J (2014) Current issues in chronic graft-versus-host disease. Blood Yahata Y, Shirakata Y, Tokumaru S et al. (2006) A novel function of angiotensin
124:374–84 II in skin wound healing. Induction of fibroblast and keratinocyte migration
Vogelsang GB, Lee L, Bensen-Kennedy DM (2003) Pathogenesis and treatment by angiotensin II via heparin-binding epidermal growth factor (EGF)-like
of graft-versus-host disease after bone marrow transplant. Annu Rev Med growth factor-mediated EGF receptor transactivation. J Biol Chem 281:
54:29–52 13209–16

Wolff D, Gerbitz A, Ayuk F et al. (2010) Consensus conference on clinical Yamamoto T (2010) Animal model of systemic sclerosis. J Dermatol 37:26–41
practice in chronic graft-versus-host disease (GVHD): first-line and Yang F, Chung ACK, Huang XR et al. (2009) Angiotensin II induces connective
topical treatment of chronic GVHD. Biol Blood Marrow Transplant 16: tissue growth factor and collagen I expression via transforming growth
1611–28 factor-beta-dependent and -independent Smad pathways: the role
Wolff D, Schleuning M, von Harsdorf S et al. (2011) Consensus of Smad3. Hypertension 54:877–84
Conference on Clinical Practice in Chronic GVHD: second-line treatment Zhou L, Askew D, Wu C et al. (2007) Cutaneous gene expression by DNA
of chronic graft-versus-host disease. Biol Blood Marrow Transplant 17: microarray in murine sclerodermatous graft-versus-host disease, a model
1–17 for human scleroderma. J Invest Dermatol 127:281–92

www.jidonline.org 2393

You might also like