Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

Cytokine xxx (2016) xxx–xxx

Contents lists available at ScienceDirect

Cytokine
journal homepage: www.journals.elsevier.com/cytokine

Molecular mechanisms of action of anti-TNF-a agents – Comparison


among therapeutic TNF-a antagonists
Hiroki Mitoma a,⇑, Takahiko Horiuchi b, Hiroshi Tsukamoto c, Naoyasu Ueda d
a
Department of Clinical Immunology and Rheumatology/Infectious Disease, Kyushu University Hospital, Fukuoka 812-8582, Japan
b
Department of Internal Medicine, Kyushu University Beppu Hospital, Beppu 874-0838, Japan
c
Department of Internal Medicine, Shin-Kokura Hospital, Kitakyushu 803-8505, Japan
d
Department of Internal Medicine, Miyazaki Prefectural Miyazaki Hospital, Miyazaki 880-8510, Japan

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

Article history: Tumor necrosis factor (TNF)-a is a potent pro-inflammatory and pathological cytokines in inflammatory
Received 2 May 2016 diseases such as rheumatoid arthritis and inflammatory bowel diseases. Anti-TNF-a therapy has been
Received in revised form 16 August 2016 established as an efficacious therapeutic strategy in these diseases. In clinical settings, three monoclonal
Accepted 16 August 2016
anti-TNF-a full IgG1 antibodies infliximab, adalimumab, and golimumab, PEGylated Fab’ fragment of
Available online xxxx
anti-TNF-a antibody certolizumab pegol, extracellular domain of TNF receptor 2/IgG1-Fc fusion protein
etanercept, are almost equally effective for rheumatoid arthritis. Although monoclonal full IgG1 antibod-
Keywords:
ies are able to induce clinical and endoscopic remission in inflammatory bowel diseases, certolizumab
Tumor necrosis factor-a
Anti-TNF-a therapy
pegol without Fc portion has been shown to be less effective for inflammatory bowel diseases compared
Inflammatory bowel disease to full IgG1 antibodies. In addition, there are no evidences that etanercept leads clinical remission in
Rheumatoid arthritis inflammatory bowel diseases. Besides the common effect of anti-TNF-a agents on neutralization of
soluble TNF-a, each anti-TNF-a agent has its own distinctive pharmacological properties which cause
the difference in clinical efficacies. Here we focus on the distinctions of action of anti-TNF-a agents espe-
cially in following points; (1) blocking ability against ligands, transmembrane TNF-a and lymphotoxin,
(2) effects toward transmembrane TNF-a-expressing cells, (3) effects toward Fcc receptor-expressing
cells, (4) degradation and distribution in inflamed tissue. Accumulating evidence will give us the idea
how to modify anti-TNF-a agents to enhance the clinical efficacy in inflammatory diseases.
Ó 2016 Elsevier Ltd. All rights reserved.

1. Introduction transgenic mice, tmTNF was shown to be sufficient to induce sys-


temic arthritis accompanied with synovial hyperplasia and inflam-
TNF-a is a major pro-inflammatory cytokine exerting pleiotro- mation [5,6]. tmTNF acts as a ligand by binding to TNF-a receptors
pic effects on various cell types by activating intracellular signaling as well as functions as a receptor that transmits outside-to-inside
nuclear factor kappa B (NF-kB), mitogen-activated protein kinase (reverse) signals into tmTNF-bearing cells (TNF-a-producing cells)
(MAPK), and caspases [1]. It plays a critical role for the pathogen- [7]. It is therefore considered that tmTNF plays a critical role in
esis of systemic inflammatory diseases such as rheumatoid arthri- local inflammation [8–13].
tis (RA), ankylosing spondylitis, psoriatic arthritis, inflammatory Anti-TNF-a agents have been successfully introduced into the
bowel diseases (IBD), and Behçet’s disease [2]. Not only a mature treatment of inflammatory diseases such as RA, juvenile idiopathic
form soluble TNF-a (sTNF), but also its precursor form transmem- arthritis, psoriatic arthritis, psoriasis, Crohn’s disease (CD), ulcera-
brane TNF-a (tmTNF), is involved in various inflammatory tive colitis (UC), ankylosing spondylitis, and Behçet’s disease
responses. tmTNF exerts its biological function in a cell-to-cell [14,15]. Infliximab, adalimumab and golimumab are full IgG1
contact manner, which is distinct from the feature of sTNF that monoclonal antibodies (mAbs) against human TNF-a. Infliximab
can act at the sites remote from TNF-a-producing cells [3,4]. In is a chimeric mouse/human anti-TNF-a monoclonal antibody
(mAb) composed of a murine variable region and a human IgG1
constant region. Adalimumab and golimumab are fully humanized
⇑ Corresponding author at: Department of Clinical Immunology and Rheumatol- anti-TNF-a mAbs, which is indistinguishable from the normal
ogy/Infectious Disease, Kyushu University Hospital, 3-1-1 Maidashi, Higashi-ku, human IgG1. Etanercept is a fusion protein composed of the 2
Fukuoka 812-8582, Japan.
extracellular portion of the human TNF receptor 2 (TNF-R2) (p75
E-mail address: mitoma@intmed1.med.kyushu-u.ac.jp (H. Mitoma).

http://dx.doi.org/10.1016/j.cyto.2016.08.014
1043-4666/Ó 2016 Elsevier Ltd. All rights reserved.

Please cite this article in press as: H. Mitoma et al., Molecular mechanisms of action of anti-TNF-a agents – Comparison among therapeutic TNF-a antag-
onists, Cytokine (2016), http://dx.doi.org/10.1016/j.cyto.2016.08.014
2 H. Mitoma et al. / Cytokine xxx (2016) xxx–xxx

TNF receptor) and the Fc portion (hinge, CH2 and CH3 domains) of agents toward tmTNF-expressing cells (TNF-a-producing cells),
human IgG1. Certolizumab is a Fab’ fragment of an anti-TNF-a (4) action of anti-TNF-a agents on Fcc receptors (FccR)-bearing
mAb and lacks the Fc portion. The hinge region of certolizumab cells, (5) distribution and degradation of anti-TNF-a agents in
is covalently linked to 2 cross-linked chains of a 20-kDa of poly- inflamed tissue. These knowledges would help us to treat refrac-
ethylene glycol, which is named as certolizumab pegol (Fig. 1) tory patients with inflammatory diseases better and to develop
[1,16]. more efficient therapeutic agents in the future.
Despite all of these anti-TNF-a agents are effective in RA, clini-
cal efficacies on CD and UC, are not equal among these agents [17].
2. Biological activities of soluble TNF and trasmembrane TNF
Specifically, anti-TNF-a full mAbs infliximab, adalimumab and
golimumab have a potential to induce clinical and endoscopic
TNF-a is generated as a precursor form called tmTNF, which is
remission in IBD, however soluble TNF-R2/IgG-Fc etanercept does
expressed as a type II polypeptide consisted of 233 amino acid
not. Even though head-to-head comparative studies have not been
residues (26-kDa) on cell surface of activated macrophages and
performed, PEGylated anti-TNF Fab’ fragment certolizumab pegol
lymphocytes as well as other cell types [39–45]. After cleavage
is considered to be less effective compared to full mAbs [18–20].
by such metalloproteinases as TNF-a-converting enzyme (TACE)
In addition, PEGylated dimeric extracellular human TNF receptor
between residues alanine76-valine77, sTNF of 157 amino acid resi-
1 (TNF-R1), onercept, was not efficacious against CD in a random-
dues (17-kDa) is released from the cell surface and mediates its
ized, double-blind, placebo-controlled trial [21]. Moreover, inflix-
biological activities through type 1 and type 2 TNF receptors
imab induces granulomatous infections like tuberculosis more
(TNF-R1 also known as TNFRSF1A, CD120a, and TNF-R2 also known
frequently than etanercept [22,23].
as TNFRSF1B, CD120b, respectively) [46–48]. sTNF is a homotrimer
Binding and neutralizing activities against sTNF are the critical
of 17-kDa monomers and tmTNF also exists as a homotrimer of 26-
and common mechanism of action of anti-TNF-a agents. On the
kDa monomers on the cell surface [49]. tmTNF also binds TNF-R1
other hand, recent studies have demonstrated that these agents
and TNF-R2, but its biological activities are supposed to be medi-
have additional biological effects against tmTNF- [24–33] and Fc
ated mainly through TNF-R2 [50]. After releasing sTNF by TACE
receptor-expressing cells [34,35]. In addition, there are distinct
cleavage, the residual cytoplasmic domain of tmTNF is additionally
features of anti-TNF-a agents in pharmacological half-life, distribu-
cleaved by homologues of signal peptide peptidase (SPPL) and
tion and degradation [36–38]. It would be helpful for the further
migrates into the nucleus of tmTNF-bearing cells and mediates
understanding of different clinical efficacies of anti-TNF-a agents
production of pro-inflammatory cytokines [51–53].
on IBD to discuss the biological functions of them comparatively.
TNF-R1 is constitutively expressed on almost all nucleated cells,
We would like to review the following issues: (1) biological activ-
whereas TNF-R2 is generally inducible and is preferentially
ities of sTNF and tmTNF, (2) blocking abilities of anti-TNF-a agents
expressed on endothelial cells and hematopoietic cells [1,54]. Both
against ligands tmTNF and lymphotoxin, (3) action of anti-TNF-a
TNF receptors form of preassembled homotrimers on the cell sur-
face and are capable of binding intracellular adaptor proteins that
lead to activation of intricate intracellular signaling processes and
mediate pleiotropic effects of TNF-a [55,56]. Receptor-mediated
effects of sTNF and tmTNF can lead to activation of NF-jB or to
apoptosis, depending on the metabolic state of the cell [1]. The sig-
naling pathways initiated by TNF-R2 that may be the preferential
receptor for tmTNF, are less characterized compared to TNF-R1.
However, TNF-R2 appears to have both shared and opposing effects
to TNF-R1 and may be actively involved in the pathogenesis of
inflammatory diseases [57].
tmTNF acts as a bipolar molecule that transmits signals not only
as a ligand, but also as a receptor in a cell-to-cell contact fashion.
tmTNF-a-bearing cells transmit signals to targets when cells
directly contact with TNF-R1- and/or TNF-R2-expressing cells.
The biological activities into tmTNF-bearing cells are transmitted
via TNF receptors through tmTNF, called as ‘‘outside-to-inside
signal” or ‘‘reverse signal” [4]. In contrast to well-characterized
functions of tmTNF as a ligand, the biological functions elicited
by outside-to-inside (reverse) signal have not completely been
clarified. However, it is supposed that outside-to-inside signaling
mediated by tmTNF contributes to the pleiotropy of this pro-
inflammatory cytokine and its fine tuning of immune responses
[7]. The biological activities of tmTNF as a receptor have been
demonstrated in T lymphocytes, monocytes/macrophages and nat-
ural killer (NK) cells in humans [8–13]. The elevation of intracellu-
lar calcium concentration in both human T cell lines and murine
macrophage cell lines was induced through tmTNF [58,59].

3. Binding and neutralization of anti-TNF-a agents to soluble


Fig. 1. Structures of therapeutic anti-TNF-a agents. Infliximab is a mouse/human and transmembrane TNF-a
chimeric monoclonal anti-TNF-a IgG1 antibody. Adalimumab and golimumab are
fully humanized monoclonal anti-TNF-a IgG1 antibodies. Etanercept is a fusion
protein of the extracellular domain of human TNF-R2 and the Fc region of IgG1.
Infliximab binds both monomeric and trimeric forms of sTNF,
Certolizumab pegol is a PEGylated Fab’ fragment of humanized monoclonal anti- whereas etanercept binds only to the trimeric forms of sTNF [30].
TNF-a antibody. Each infliximab molecule is capable of binding two TNF-a

Please cite this article in press as: H. Mitoma et al., Molecular mechanisms of action of anti-TNF-a agents – Comparison among therapeutic TNF-a antag-
onists, Cytokine (2016), http://dx.doi.org/10.1016/j.cyto.2016.08.014
H. Mitoma et al. / Cytokine xxx (2016) xxx–xxx 3

molecules, and up to three infliximab molecules can bind each cannot be regulated by anti-TNF agents. Further studies are
TNF-a homotrimer. In contrast, etanercept is supposed to forms required to understand the benefits of lymphotoxin antagonists
one-to-one complex with a TNF-a homotrimer [30]. In fact, only in the treatment of IBD.
mAbs, but not etanercept, form large protein complexes in vitro
[60]. Although several groups examined binding activities of anti- 5. Functional properties of anti-TNF-a agents on
TNF-a agents to sTNF in different assay systems, all of infliximab, transmembrane TNF-a-expressing cells
adalimumab, etanercept and certolizumab pegol have almost sim-
ilar intrinsic binding properties to sTNF and nearly equal neutral- Considering the importance of tmTNF in the pathogenesis of
ization abilities against TNF receptor signaling [25,28]. Shealy D inflammatory diseases, distinct effects of anti-TNF-a agents on
and colleagues reported that the affinity of golimumab to sTNF is tmTNF may explain the difference in these clinical efficacies at
superior to those of infliximab and adalimumab. Golimumab is least in part. A number of groups reported head-to-head compar-
conformationally more stable, and the inhibitory ability of goli- ison of functional properties of these anti-TNF-a agents on
mumab against TNF-a-induced cytotoxicity and activation of tmTNF-expressing cells [25–28,30–33].
human endothelial cells is better than those of infliximab and adal-
imumab [31].
5.1. Complement-dependent cytotoxicity (CDC)
Infliximab, adalimumab, golimumab, etanercept and cer-
tolizumab pegol bind to tmTNF on tmTNF-transfected cells with
In the system using human Jurkat T lymphocytes [27,32], mur-
similar affinities that were lower compared to sTNF [25,28,31,
ine NS0 myeloma cells [28], murine Sp2/0 myeloma cells [25], or
61,62]. Other groups reported that etanercept does not bind tmTNF
Chinese hamster ovary (CHO) cells [61], CDC via anti-TNF-a agents
or binds much weakly compared to anti-TNF-a mAbs [33,34]. As
was analyzed. Infliximab, adalimumab and golimumab induced
with sTNF, up to three molecules of infliximab can bind one tmTNF,
CDC much more potently than etanercept [25,27,28,32,61]. On
one etanercept can bind one molecule of tmTNF, suggesting that
the other hand, certolizumab pegol did not have any CDC activities
anti-TNF-a mAbs forms large molecular complexes with tmTNF on
[28,32], which is caused by the absence of Fc portion of IgG1. From
the cell membrane [30]. Therefore, quantitative differences in
the structural point of view, lack of the activation of the comple-
binding between etanercept and anti-TNF-a mAbs may come from
ment cascade by etanercept seems to be intelligible as well. Inflix-
stoichiometric differences rather than distinctions of affinity. In
imab, adalimumab, golimumab and etanercept commonly possess
terms of inhibition of ligand activities of tmTNF, infliximab was sig-
the Fc portion of IgG1, whose CH2 domain activates the first com-
nificantly more potent than etanercept at blocking tmTNF-mediated
ponent of complement (C1) activation. Etanercept does carry this
E-selectin expression in human umbilical vein endothelial cell
CH2 domain of IgG1, but not the CH1 domain. A narrow region
(HUVEC) [30]. In a bioassay using human lung carcinoma cell line
of 23 amino acid residues within the CH1 domain serves as a plat-
A549, infliximab, adalimumab, and certolizumab pegol similarly
form of complement C3 activation [69,70], which was later con-
inhibited tmTNF-mediated cell death, however etanercept showed
firmed that three amino acid residues within the specific 23
about 2-fold less activity [28]. Taken together, effector functions of
amino acids are involved in the covalent attachment with C3
tmTNF as a ligand are inhibited by any of anti-TNF agents, although
[71–73]. Hence it is thus difficult for etanercept to make mem-
the activity of etanercept is weaker than the other agents.
brane attack complex of complement proteins (C5b-C9) for CDC
at least in vitro. When activated human peripheral blood mononu-
clear cells (PBMCs) were studied as target cells, none of infliximab,
4. Binding and neutralization of anti-TNF-a agents to
adalimumab and etanercept induced CDC [25], which may be due
lymphotoxin-a
to the use of different cell types from the above mentioned
experiments.
The homotrimer of lymphotoxin-a (LTa3) and heterotrimer of
two lymphotoxin-a and one lymphotoxin-b (LTa2b1) bind both
TNF-R1 and TNF-R2. Only etanercept, but not infliximab and 5.2. Antibody-dependent cell-mediated cytotoxicity (ADCC)
adalimumab, can bind and neutralize LTa3 and LTa2b1 [1,25,30].
One lymphotoxin-a also forms heterotrimers (LTa1b2) with two Infliximab, adalimumab, golimumab and etanercept showed
lymphotoxin-b which is much more dominant than LTa2b1 similar ADCC activities by using tmTNF-transfected Jurkat T lym-
in vivo. LTa1b2 binds lymphotoxin-b receptor (LTbR) but not TNF- phocytes as target cells and human PBMCs or NK cells as effector
receptors, hence etanercept is not able to neutralize LTa1b2 [63]. cells [32], while infliximab and adalimumab showed much more
LTa3 mediates proliferation and pro-inflammatory cytokine potent ADCC activities than etanercept in NS0 cells [28] or in
secretion of RA synovial fibroblasts and activates the inflammatory CHO cells [61]. Certolizumab pegol lacking Fc portion did not show
environment in human chondrocytes [64,65]. Therefore, the neu- any ADCC activity in any cell types as speculated [28]. The discrep-
tralization of LTa3 via etanercept is beneficial in patients with RA ancy in etanercept-induced ADCC is not clear, however it may be
to suppress inflammation [66]. However, anti-lymphotoxin-a caused by the different experimental conditions, such as species
mAb, pateclizumab, was not effective against RA patients with an and cell types of target cells, and expression levels of tmTNF. Inflix-
inadequate response to disease-modifying anti-rheumatic drugs, imab, adalimumab, golimumab and etanercept carry the CH2 and
indicating that blocking of lymphotoxin-a alone is not sufficient CH3 domain of human IgG1, whereas certolizumab pegol does
for controlling activities of RA [67]. not. These domains of IgG1 are involved in the binding to Fc recep-
The role of LTa3 in the pathogenesis of IBD, has not been well tors of effector cells [74], which leads to the lysis of target cells by
documented. LTa1b2 has been reported to regulate the develop- the secretion of granzyme B and perforin from effector cells.
ment of intestinal lymphoid organs, including Peyer’s patches
and mesenteric lymph nodes. The intestinal inflammation in 5.3. Outside-to-inside signaling (reverse signaling) into tmTNF-
chronic dextran sodium sulfate (DSS)-induced colitis is suppressed expressing cells
by inhibition of LTbR signaling. Conversely, the course of Citrobac-
ter rodentium-induced infectious colitis is more severe in mice Infliximab and adalimumab, but not etanercept, induced apop-
with impaired LTbR signaling [68]. LTbR signaling via LTa1b2 might tosis and cell cycle G0/G1 arrest in tmTNF-expressing Jurkat T lym-
play an important role in the intestinal inflammation, which phocytes that neither express TNF-R1 nor TNF-R2. This effect is

Please cite this article in press as: H. Mitoma et al., Molecular mechanisms of action of anti-TNF-a agents – Comparison among therapeutic TNF-a antag-
onists, Cytokine (2016), http://dx.doi.org/10.1016/j.cyto.2016.08.014
4 H. Mitoma et al. / Cytokine xxx (2016) xxx–xxx

mediated by reverse signaling through tmTNF, independently of been shown to contribute to the host defense against acute
complement and NK cells [26]. Despite the similar binding abilities Mycobacterium tuberculosis infection in humans. CD8+CCR7-
of anti-TNF-a mAbs to tmTNF, golimumab induced a significantly CD45RA+ effector memory T lymphocytes express granulysin and
weaker apoptosis than infliximab and adalimumab [32]. The rea- mediate an antimicrobial activity against Mycobacterium tuberculo-
son for this discrepancy is not clear, however one possible reason sis. This subset of T lymphocytes expressed tmTNF and bound
might be the difference of epitopes among these antibodies. tmTNF infliximab. Infliximab led them to be susceptible to complement-
forms trimer on the cell surface, antibody can bind two TNF tri- mediated lysis and the resultant reduced an antimicrobial activity
mers, leading to the accumulation of tmTNF on the cell membrane. [80].
Supportively, cross-linking of etanercept bound to the cell-surface Atreya R and colleagues raised another possibility for the induc-
tmTNF with anti-human IgG antibody resulted in an increased tion of apoptosis in activated T lymphocytes. Infliximab, adali-
apoptosis, which indicates that the multimer formation of tmTNF mumab and certolizumab pegol, but not etanercept, were able to
on the cell surface is essential for the initiation of the subsequent induce apoptosis of T lymphocytes in IBD only when mucosal
intracellular signals [26]. c-Jun N-terminal protein kinase (JNK) CD4+ T lymphocytes expressing TNF-R2 were co-cultured with
activation followed by up-regulation of p21WAF1/CIP1, Bax and Bak tmTNF-expressing CD14+ intestinal macrophages. The induction
as well as reactive oxygen species (ROS) accumulation are impor- of apoptosis was dependent on inhibition of TNF-R2 signaling in
tant intracellular signaling events for the apoptosis and cell cycle CD4+ T lymphocytes. They proposed that CD14+ macrophages
arrest. In addition, site-directed mutagenesis revealed that three protected CD4+ T lymphocytes from apoptosis through TNF-R2
serine residues in the cytoplasmic domain of tmTNF are essential survival signaling via tmTNF [81].
for these biologic effects [26]. The amino acid sequence of the Even though identification of precise mechanisms of apoptosis
intracellular domain of tmTNF is well conserved in different spe- induction is extremely difficult, it has been established that
cies. Moreover, all the three serine residues were conserved among activated T lymphocytes or macrophages involved in pathogenesis
different species, indicating that these residues are critical for or host defense fall into apoptosis in vivo after infusion of anti-TNF-
physiological functions of tmTNF [59]. a mAbs.

7. Expansion of regulatory T lymphocytes by anti-TNF-a


6. Induction of apoptosis by anti-TNF-a antibodies in lamina
antibodies through enhancement TNF receptor 2 signaling
propria T lymphocytes of Crohn’s disease, in psoriatic CD8+ T
cells, and in effector T cells against Mycobacterium tuberculosis
The role of TNF on the expansion and suppressive function of
regulatory T lymphocytes (Tregs) is still controversial in human
ten Hove T and colleagues studied induction of apoptosis in vivo
because a large number of contradictory findings have been
in ten patients with refractory CD via the biopsy of colon just
reported [82–90]. On the other hand, several studies have shown
before and 24 h after the infusion of infliximab. A significant
that signaling through TNF-R2 is important for function of Tregs
increase in CD3 and TUNEL positive cells within the colonic biopsy
in murine systems [91–94]. Nguyen DX and colleagues examined
specimen was detected 24 h after infusion of infliximab. In this
effects of anti-TNF-a agents on human Tregs in RA and health con-
study, detectable change of phenotype or apoptosis in peripheral
trols [95]. Monocytes from patients with RA expressed more
blood T lymphocyte was not observed [75]. They also tested apop-
tmTNF on their surface than those from healthy controls. Adali-
tosis induction at 24 h after infusion of infliximab by administra-
mumab, but not etanercept, bound tmTNF on monocytes from RA
tion of technetium labeled annexin V into infliximab-treated
and further enhanced expression levels of tmTNF. This effect was
patients, following visualization of apoptotic cells in vivo using a
not observed in monocytes from healthy controls. Surprisingly,
single-photon emission computer tomography. Annexin V uptake
adalimumab promoted the interaction between tmTNF on mono-
was specifically enhanced in the diseased intestine 24 h after infu-
cytes and TNF-R2 on Tregs in RA, resulting in the expansion of
sion of infliximab. Analysis of the mucosal biopsy specimens iden-
functional Foxp3-positive Tregs, which suppress Th17 lympho-
tified lamina propria CD4+ T cells as target cells of annexin V
cytes. Etanercept did not expand Tregs at all in this RA system.
undergoing apoptosis [76]. These facts suggest that infliximab
Infliximab, golimumab, and certolizumab pegol were not included
induced apoptosis of activated T lymphocytes in inflamed tissue.
in this study. Precise mechanisms how adalimumab paradoxically
Bedini C and colleagues investigated the apoptosis induction in
promoted TNF-R2 signaling mediated by tmTNF were not analyzed
psoriatic skin-homing T-lymphocytes. Psoriatic CD8+ T lympho-
in this study, thus further studies are required to understand the
cytes, but not CD4+ T lymphocytes, were susceptible to
significance of this effect in inflammatory diseases.
infliximab-induced apoptosis in vitro [77]. Caprioli F and colleagues
also investigated apoptosis in lamina propria in IBD patients
treated with infliximab. Infliximab-induced downregulation of 8. Functional properties of anti-TNF-a agents to
macrophages was significantly associated with both endoscopic monocytes/macrophages through Fcc receptors
responses to the therapy and achievement of mucosal healing.
The reduction of lamina propria CD68+ macrophages was associ- All of anti-TNF-a agents except certolizumab pegol contain Fc
ated with an increased rate of macrophage apoptosis [78]. In the portion of IgG1, hence these agents were speculated to exert sig-
clinical settings, it is likely that infliximab induces apoptosis at nals through Fc receptors. During mixed lymphocyte reaction
least in part by tmTNF-mediated effects; CDC, ADCC and/or (MLR), infliximab and adalimumab, but not etanercept and cer-
outside-to-inside signaling even though several groups reported tolizumab pegol, reduced proliferation of T lymphocytes. This
that peripheral PBMCs from healthy donors did not show the effect was lost by blocking of Fc receptor using control IgG or by
induction of apoptosis in vitro. deletion of Fc portion of anti-TNF-a mAbs. In addition,
Infliximab was associated with twofold to eightfold greater certolizumab-IgG with Fc portion did inhibit T cell proliferation,
risks of granulomatous infections such as tuberculosis, listeriosis indicating that Fc portion is required for this inhibitory function.
and histoplasmosis than etanercept in post-marketing surveillance In MLR, anti-TNF-a mAbs induced generation of a new population
in the USA [79]. The increased incidence of tuberculosis in patients of macrophages in an Fc-dependent manner. These macrophages
treated with infliximab or adalimumab compared to etanercept expressed the regulatory macrophage marker CD206, and had an
was also reported in other European countries [22]. tmTNF has immunosuppressive phenotype, producing anti-inflammatory

Please cite this article in press as: H. Mitoma et al., Molecular mechanisms of action of anti-TNF-a agents – Comparison among therapeutic TNF-a antag-
onists, Cytokine (2016), http://dx.doi.org/10.1016/j.cyto.2016.08.014
H. Mitoma et al. / Cytokine xxx (2016) xxx–xxx 5

cytokine interleukin (IL)-10 [34]. Importantly, they also showed agents to FcRn [36]. Certolizumab pegol lacking Fc portion was
that the induction of regulatory macrophages was obtained in CD confirmed not to have the binding ability to FcRn at all [103]. Goli-
patients with mucosal healing after treatment with infliximab, mumab was reported to have the binding ability to FcRn, however
but not in CD patients without mucosal healing. Authors discussed the comparative study with the other anti-TNF-a agents was not
that the different effect between infliximab and etanercept was performed [31].
caused by lack of binding ability of etanercept to tmTNF on acti- Effects of binding with sTNF on the affinities of infliximab, adal-
vated T lymphocytes [96]. It is still not clear why sTNF/etanercept imumab and etanercept to FcRn were examined by the surface
complexes is not enough to induce regulatory macrophages plasmon resonance sensorgrams. The affinity of infliximab–sTNF
through Fc receptor despite the presence of IgG1-Fc region. Arora complex or adalimumab–sTNF complex to FcRn was lower than
T and colleagues reported that upon addition of sTNF, anti-TNF-a that of infliximab alone or adalimumab alone, respectively [36].
mAbs, but not etanercept, showed significant increased binding Interestingly, these results are opposite to the bindig affinity of
ability to Fc receptors due to the formation of large protein com- anti-TNF-a mAbs to Fcc receptors [61]. The affinity of etaner-
plexes by mAbs [61]. Thus, increased binding ability of anti-TNF- cept–sTNF complex to FcRn was comparable with that of etaner-
a mAbs to Fc receptors mediated by sTNF/tmTNF recognition cept alone. These results suggest that for anti–TNF-a mAbs,
might be an important factor for the induction of regulatory binding with target molecules decreases the affinity to FcRn.
macrophages. Therefore, the anti–TNF-a mAbs complexed with TNF-a will be
On the other hand, Wojtal KA and colleagues reported unfavor- degraded more rapidly than anti–TNF-a mAbs free from TNF-a
able effects of anti-TNF-a mAbs through FccRI (CD64) in IBD in vivo.
patients. Infliximab/TNF complexes activate Fc receptors on PBMCs
through tyrosine phosphorylation and induce the expression of
10. Proteolytic cleavage of anti-TNF-a agents by tissue
pro-inflammatory cytokines such as MCP-1, GM-CSF, IL-6 and IL-
metalloproteinases
8. Adalimumab showed the similar response, but certolizumab
pegol or F(ab’)2 fragments of infliximab did not have this activity,
All of infliximab, adalimumab, golimumab, certolizumab pegol
confirming that Fc portion is required for this function. Etanercept
and etanercept include the hinge region of human IgG1, which is
was not included in this study, hence we are not able to compare
recognized by proteases such as matrix metalloproteinase
this activity between antibodies and a soluble receptor. Colonic
(MMP)-3 and MMP-12. Biancheri P and colleagues reported that
mRNA expression levels of CD64 were significantly increased in
these metalloproteinases cleave infliximab and adalimumab into
the inflamed tissues from non-responders of infliximab, compared
Fc domain and Fab domain in vitro. Etanercept is cleaved into Fc
to responders of infliximab, indicating that CD64 signaling might
domain and extracellular domain of TNF-R2 by these proteases.
be one of mechanisms of non-responsiveness of anti-TNF-a mAbs
Certolizumab pegol and golimumab are not examined in this
in CD [35].
study. Free F(ab’)2 fragments of infliximab and adalimumab after
Monocytes/macrophages express several kinds of activating Fc
proteolytic cleavage by MMP-3 or MMP-12 still can antagonize
receptors FccRI (CD64), FccRIIA (CD32), FccRIIIA (CD16) and one
functions of sTNF. On the other hand, soluble TNF-R2 isolated from
inhibitory receptor FccRIIB (CD32) [97]. IgG1 can bind all of
etanercept by these proteases lost the activity of neutralization of
these receptors, hence transmitting signaling in monocytes/
sTNF. The precise mechanism of decreased neutralization ability
macrophages is complicated. The expression level of each receptor
of etanercept after cleavage has been unknown. Dimeric form of
will affect the output of Fc receptor signaling, therefore anti-TNF-a
extracellular domain of TNF-R2 in etanercept might be broken
mAbs exert an anti-inflammatory effect in some conditions and do
after cleavage, resulting in generation of monomer of TNF-R2.
a pro-inflammatory effect in other conditions. FccR polymor-
Inflamed intestinal mucosal homogenates from patients with CD,
phisms have been associated with clinical response to TNF-a
but not ones from health controls, cleaved these three anti-TNF-
antagonists in patients with IBD [98–101], therefore, FcR signaling
a agents at hinge region. The authors also reported that the
might be one of important mechanisms of action of anti-TNF-a full
presence of anti-hinge autoantibody in sera from patients
mAbs.
with CD. Cleaved hinge region become neo-antigen, leading to
the generation of autoantibodies against human IgG1. Sera from
non-responders to infliximab showed higher titer of anti-hinge
9. Serum half-life and binding ability of anti-TNF-a agents to
antibodies than one from responders to infliximab [37]. Although
neonatal Fc receptors
it was not examined whether these anti-hinge autoantibodies
decrease neutralization abilities of anti-TNF-a agents against
Etanercept shows a shorter serum half-life (3–5.5 days) than
TNF-a, proteolytic cleavage of anti-TNF-a agents by mucosal
mAbs of infliximab (8–10 days), adalimumab (10–20 days), goli-
metalloproteinases and anti-hinge autoantibodies might be one
mumab (9–15 days) and certolizumab pegol (14 days) in vivo [1].
of mechanisms which contribute to the unresponsiveness to anti-
Neonatal Fc receptors (FcRn) bind to the Fc domain of IgG at acidic
TNF-a agents in IBD.
pH in the endosome and protect IgG from the ongoing catabolic
activities, thereby contributing to the long serum half-life of IgG
[36]. The binding affinity of anti-TNF-a mAbs infliximab and adal- 11. Distribution of anti-TNF-a agents into the inflamed tissue
imumab to FcRn was 10-fold higher than that of etanercept, result-
ing in the difference of serum half-life among anti-TNF-a agents Delivery of drugs at the site of inflammation is another impor-
in vivo. Authors speculate that the glycosylation at the C- tant factor for the efficacy of treatment in inflammatory diseases.
terminus of TNF-R2 region might induce the Fc conformational Palframan R and colleagues studied the distribution of infliximab,
change or interference with the binding between Fc portion and adalimumab and certolizumab pegol labeled with the low molecu-
FcRn by the steric hindrance, resulting in the reduction of bindig lar weight dye to the inflamed joints of mice with collagen-induced
affinity of etanercept to FcRn [102]. Supportively, the affinity of arthritis using a biofluorescence imaging. Etanercept was not
etanercept and mAbs became comparable after papain digestion included in this study. All of anti-TNF-a mAbs studied distributed
at hinge region, suggesting that differences in amino acid more effectively into inflamed joints than non-inflamed joints. The
sequences or posttranslational modification of the Fc portion did ratio of distribution of certolizumab pegol into the inflamed joint
not contribute to the difference in the binding affinity of these compared with the normal joint was greater than that observed

Please cite this article in press as: H. Mitoma et al., Molecular mechanisms of action of anti-TNF-a agents – Comparison among therapeutic TNF-a antag-
onists, Cytokine (2016), http://dx.doi.org/10.1016/j.cyto.2016.08.014
6 H. Mitoma et al. / Cytokine xxx (2016) xxx–xxx

Table 1
Pleiotropic mechanisms action of anti-TNF-a agents. Very strong, +++; strong, ++; weak, +; no effect; –; not determined, n.d.; soluble TNF-a; sTNF; tmTNF; transmembrane TNF-a;
lymphotoxin-a3, LTa3; complement-dependent cytotoxicity, CDC; antibody-dependent cell-mediated cytotoxicity, ADCC; regulatory T lymphocytes, Tregs; Fcc receptors, FccR;
neonatal Fc receptors, FcRn; matrix metalloproteinases, MMPs.

Actions of anti-TNF-a agents Full lgG1 mAb PEGylated TNF Reference


Fab’ receptor 2
Infliximab Adalimumab Golimumab Certolizumab Etanercept
(a) Binding and neutralization of
(1) sTNF ++ ++ +++ ++ ++ [25,28,30,31]
(2) tmTNF ++ ++ ++ ++ + [25,28,30,31,33,34,61,62]
(3) LTa3 – – – – ++ [30]
(b) Action toward tmTNF-expressing cells
(1) CDC ++ ++ ++ – + [25,27,28,32,61]
(2) ADCC ++ ++ ++ – +/++ [27,28,32,61]
(3) Apoptosis via reverse signaling ++ ++ + – – [26,32]
(c) Expansion of Tregs n.d. ++ n.d. n.d. – [95]
(d) Action toward FccR-bearing cells
(1) Induction of regulatory macrophages ++ ++ n.d. – – [34,96]
(2) Induction of pro-inflammatory cytokines ++ + n.d. – n.d. [35]
(e) Escape from catabolism through FcRn ++ ++ ++ – + [31,36,102,103]
(f) Cleavage by MMPs/loss of neutralization activity against sTNF +/ +/ n.d. n.d. +/+ [37]
(g) Distribution into the inflamed tissue + + n.d. ++ n.d. [38]

with infliximab and adalimumab. In addition, the duration of expo- [3] C. Perez, I. Albert, K. DeFay, N. Zachariades, L. Gooding, M. Kriegler, A
nonsecretable cell surface mutant of tumor necrosis factor (TNF) kills by cell-
sure in the inflamed versus normal tissue was more prolonged for
to-cell contact, Cell 63 (2) (1990) 251–258.
certolizumab pegol than for both infliximab and adalimumab [38]. [4] T. Horiuchi, H. Mitoma, S. Harashima, H. Tsukamoto, T. Shimoda,
The further studies including all anti-TNF-a agents are expected in Transmembrane TNF-alpha: structure, function and interaction with anti-
experimental mice models of IBD such as the DSS-induced colitis TNF agents, Rheumatology (Oxford, England) 49 (7) (2010) 1215–1228.
[5] L. Alexopoulou, M. Pasparakis, G. Kollias, A murine transmembrane tumor
and the 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced colitis. necrosis factor (TNF) transgene induces arthritis by cooperative p55/p75 TNF
receptor signaling, Eur. J. Immunol. 27 (10) (1997) 2588–2592.
[6] S. Georgopoulos, D. Plows, G. Kollias, Transmembrane TNF is sufficient to
12. Conclusions induce localized tissue toxicity and chronic inflammatory arthritis in
transgenic mice, J. Inflamm. 46 (2) (1996) 86–97.
[7] G. Eissner, W. Kolch, P. Scheurich, Ligands working as receptors: reverse
We have reviewed the diversity of functions of anti-TNF-a signaling by members of the TNF superfamily enhance the plasticity of the
agents introduced to the treatment of inflammatory diseases immune system, Cytokine Growth Factor Rev. 15 (5) (2004) 353–366.
(Table 1). Mechanisms of action of biological preparations are not [8] G. Eissner, S. Kirchner, H. Lindner, W. Kolch, P. Janosch, M. Grell, P. Scheurich,
R. Andreesen, E. Holler, Reverse signaling through transmembrane TNF
solely neutralization of target proteins, but also mediate additional
confers resistance to lipopolysaccharide in human monocytes and
signals through unexpected receptors such as tmTNF and/or Fc macrophages, J. Immunol. (Baltimore, Md.: 1950) 164 (12) (2000) 6193–6198.
receptors. Biological functions of cytokines are complicated, hence [9] S. Harashima, T. Horiuchi, N. Hatta, C. Morita, M. Higuchi, T. Sawabe, H.
it is possible that blocking of cytokines induces bipolar effects Tsukamoto, T. Tahira, K. Hayashi, S. Fujita, Y. Niho, Outside-to-inside signal
through the membrane TNF-alpha induces E-selectin (CD62E) expression on
in vivo. Moreover, pharmacological properties of anti-TNF-a agents activated human CD4+ T cells, J. Immunol. (Baltimore, Md.: 1950) 166 (1)
such as the distribution to diseased tissues and the degradation by (2001) 130–136.
proteases also contribute to the effects of biological preparations. [10] M. Rossol, U. Meusch, M. Pierer, S. Kaltenhauser, H. Hantzschel, S. Hauschildt,
U. Wagner, Interaction between transmembrane TNF and TNFR1/2 mediates
Understanding of mechanisms of action of anti-TNF-a agents and the activation of monocytes by contact with T cells, J. Immunol. (Baltimore,
the correlation with clinical aspects might enable us to select the Md.: 1950) 179 (6) (2007) 4239–4248.
appropriate biological agents in individual cases and to develop [11] N.K. Vudattu, E. Holler, P. Ewing, U. Schulz, S. Haffner, V. Burger, S. Kirchner, R.
Andreesen, G. Eissner, Reverse signalling of membrane-integrated tumour
new biological preparations for inflammatory diseases. necrosis factor differentially regulates alloresponses of CD4+ and CD8+ T cells
against human microvascular endothelial cells, Immunology 115 (4) (2005)
536–543.
Conflict of interest [12] L. Xin, J. Wang, H. Zhang, W. Shi, M. Yu, Q. Li, X. Jiang, F. Gong, K. Gardner, Q.Q.
Li, Z. Li, Dual regulation of soluble tumor necrosis factor-alpha induced
The authors declare no conflict of interest. activation of human monocytic cells via modulating transmembrane TNF-
alpha-mediated ’reverse signaling’, Int. J. Mol. Med. 18 (5) (2006) 885–892.
[13] M. Yu, W. Shi, J. Zhang, L. Niu, Q. Chen, D. Yan, T. Liu, W. Jing, X. Jiang, F. Wei,
Acknowledgements B. Yin, W. Zhang, Q. Li, Z. Li, Influence of reverse signaling via membrane TNF-
alpha on cytotoxicity of NK92 cells, Eur. J. Cell Biol. 88 (3) (2009) 181–191.
[14] C. Blandizzi, P. Gionchetti, A. Armuzzi, R. Caporali, S. Chimenti, R. Cimaz, L.
This work was supported by in part by Grants-in-Aid for Scien- Cimino, G. Lapadula, P. Lionetti, A. Marchesoni, A. Marcellusi, F.S. Mennini, C.
tific Research from the Ministry of Education, Culture, Sports, Salvarani, G. Girolomoni, The role of tumour necrosis factor in the
pathogenesis of immune-mediated diseases, Int. J. Immunopathol.
Science and Technology of Japan (26893188). Pharmacol. 27 (1 Suppl) (2014) 1–10.
[15] A. Arida, K. Fragiadaki, E. Giavri, P.P. Sfikakis, Anti-TNF agents for Behcet’s
disease: analysis of published data on 369 patients, Semin. Arthritis Rheum.
References
41 (1) (2011) 61–70.
[16] L.M. Sedger, M.F. McDermott, TNF and TNF-receptors: from mediators of cell
[1] D. Tracey, L. Klareskog, E.H. Sasso, J.G. Salfeld, P.P. Tak, Tumor necrosis factor death and inflammation to therapeutic giants – past, present and future,
antagonist mechanisms of action: a comprehensive review, Pharmacol. Ther. Cytokine Growth Factor Rev. 25 (4) (2014) 453–472.
117 (2) (2008) 244–279. [17] A.D. Levin, M.E. Wildenberg, G.R. van den Brink, Mechanism of action of Anti-
[2] J.R. Bradley, TNF-mediated inflammatory disease, J. Pathol. 214 (2) (2008) TNF therapy in inflammatory bowel disease, J. Crohn’s Colitis 10 (8) (2016)
149–160. 989–997.

Please cite this article in press as: H. Mitoma et al., Molecular mechanisms of action of anti-TNF-a agents – Comparison among therapeutic TNF-a antag-
onists, Cytokine (2016), http://dx.doi.org/10.1016/j.cyto.2016.08.014
H. Mitoma et al. / Cytokine xxx (2016) xxx–xxx 7

[18] W. Da, J. Zhu, L. Wang, Y. Lu, Efficacy and safety of certolizumab pegol for infliximab in the inflamed paws of mice with collagen-induced arthritis, J.
Crohn’s disease: a systematic review and meta-analysis, Adv. Therapy 30 (5) Immunol. Methods 348 (1–2) (2009) 36–41.
(2013) 541–553. [39] C. Agostini, R. Sancetta, A. Cerutti, G. Semenzato, Alveolar macrophages as a
[19] W.J. Sandborn, B.G. Feagan, S. Stoinov, P.J. Honiball, P. Rutgeerts, D. Mason, R. cell source of cytokine hyperproduction in HIV-related interstitial lung
Bloomfield, S. Schreiber, Certolizumab pegol for the treatment of Crohn’s disease, J. Leukoc. Biol. 58 (5) (1995) 495–500.
disease, New Engl. J. Med. 357 (3) (2007) 228–238. [40] L. Armstrong, D.R. Thickett, S.J. Christie, H. Kendall, A.B. Millar, Increased
[20] W.J. Sandborn, S. Schreiber, B.G. Feagan, P. Rutgeerts, Z.H. Younes, R. expression of functionally active membrane-associated tumor necrosis factor
Bloomfield, G. Coteur, J.P. Guzman, G.R. D’Haens, Certolizumab pegol for in acute respiratory distress syndrome, Am. J. Respir. Cell Mol. Biol. 22 (1)
active Crohn’s disease: a placebo-controlled, randomized trial, Clin. (2000) 68–74.
Gastroenterol. Hepatol.: Official Clin. Practice J. Am. Gastroenterol. Assoc. 9 [41] G. Caron, Y. Delneste, J.P. Aubry, G. Magistrelli, N. Herbault, A. Blaecke, A.
(8) (2011) 670–678.e3. Meager, J.Y. Bonnefoy, P. Jeannin, Human NK cells constitutively express
[21] P. Rutgeerts, W.J. Sandborn, R.N. Fedorak, D. Rachmilewitz, D. Tarabar, P. membrane TNF-alpha (mTNFalpha) and present mTNFalpha-dependent
Gibson, O. Haagen Nielsen, G. Wild, S. Schreiber, C. Pena Rossi, M. Zignani, cytotoxic activity, Eur. J. Immunol. 29 (11) (1999) 3588–3595.
Onercept for moderate-to-severe Crohn’s disease: a randomized, double- [42] M. Fishman, Cytolytic activities of activated macrophages versus
blind, placebo-controlled trial, Clin. Gastroenterol. Hepatol.: Official Clin. paraformaldehyde-fixed macrophages; soluble versus membrane-associated
Practice J. Am. Gastroenterol. Assoc. 4 (7) (2006) 888–893. TNF, Cell. Immunol. 137 (1) (1991) 164–174.
[22] R.S. Wallis, Tumour necrosis factor antagonists: structure, function, and [43] T. Horiuchi, C. Morita, H. Tsukamoto, H. Mitoma, T. Sawabe, S. Harashima, Y.
tuberculosis risks, Lancet. Infect. Dis 8 (10) (2008) 601–611. Kashiwagi, S. Okamura, Increased expression of membrane TNF-alpha on
[23] R.S. Wallis, S. Ehlers, Tumor necrosis factor and granuloma biology: activated peripheral CD8+ T cells in systemic lupus erythematosus, Int. J. Mol.
explaining the differential infection risk of etanercept and infliximab, Med. 17 (5) (2006) 875–879.
Semin. Arthritis Rheum. 34 (5 Suppl1) (2005) 34–38. [44] M. Kresse, M. Latta, G. Kunstle, H.M. Riehle, N. van Rooijen, H. Hentze, G.
[24] S. Derer, A. Till, R. Haesler, C. Sina, N. Grabe, S. Jung, S. Nikolaus, T. Kuehbacher, J. Tiegs, M. Biburger, R. Lucas, A. Wendel, Kupffer cell-expressed membrane-
Groetzinger, S. Rose-John, P.C. Rosenstiel, S. Schreiber, MTNF reverse signalling bound TNF mediates melphalan hepatotoxicity via activation of both TNF
induced by TNFalpha antagonists involves a GDF-1 dependent pathway: receptors, J. Immunol. (Baltimore, Md.: 1950) 175 (6) (2005) 4076–4083.
implications for Crohn’s disease, Gut 62 (3) (2013) 376–386. [45] R. Peck, M. Brockhaus, J.R. Frey, Cell surface tumor necrosis factor (TNF)
[25] Z. Kaymakcalan, P. Sakorafas, S. Bose, S. Scesney, L. Xiong, D.K. Hanzatian, J. accounts for monocyte- and lymphocyte-mediated killing of TNF-resistant
Salfeld, E.H. Sasso, Comparisons of affinities, avidities, and complement target cells, Cell. Immunol. 122 (1) (1989) 1–10.
activation of adalimumab, infliximab, and etanercept in binding to soluble [46] T.P. Birkland, J.P. Sypek, D.J. Wyler, Soluble TNF and membrane TNF expressed
and membrane tumor necrosis factor, Clin. Immunol. (Orlando, Fla.) 131 (2) on CD4+ T lymphocytes differ in their ability to activate macrophage
(2009) 308–316. antileishmanial defense, J. Leukoc. Biol. 51 (3) (1992) 296–299.
[26] H. Mitoma, T. Horiuchi, N. Hatta, H. Tsukamoto, S. Harashima, Y. Kikuchi, J. [47] R.A. Black, C.T. Rauch, C.J. Kozlosky, J.J. Peschon, J.L. Slack, M.F. Wolfson, B.J.
Otsuka, S. Okamura, S. Fujita, M. Harada, Infliximab induces potent anti- Castner, K.L. Stocking, P. Reddy, S. Srinivasan, N. Nelson, N. Boiani, K.A.
inflammatory responses by outside-to-inside signals through Schooley, M. Gerhart, R. Davis, J.N. Fitzner, R.S. Johnson, R.J. Paxton, C.J.
transmembrane TNF-alpha, Gastroenterology 128 (2) (2005) 376–392. March, D.P. Cerretti, A metalloproteinase disintegrin that releases tumour-
[27] H. Mitoma, T. Horiuchi, H. Tsukamoto, Y. Tamimoto, Y. Kimoto, A. Uchino, K. necrosis factor-alpha from cells, Nature 385 (6618) (1997) 729–733.
To, S. Harashima, N. Hatta, M. Harada, Mechanisms for cytotoxic effects of [48] J.K. Lazdins, M. Grell, M.R. Walker, K. Woods-Cook, P. Scheurich, K.
anti-tumor necrosis factor agents on transmembrane tumor necrosis factor Pfizenmaier, Membrane tumor necrosis factor (TNF) induced cooperative
alpha-expressing cells: comparison among infliximab, etanercept, and signaling of TNFR60 and TNFR80 favors induction of cell death rather than
adalimumab, Arthritis Rheum. 58 (5) (2008) 1248–1257. virus production in HIV-infected T cells, J. Exp. Med. 185 (1) (1997) 81–90.
[28] A. Nesbitt, G. Fossati, M. Bergin, P. Stephens, S. Stephens, R. Foulkes, D. Brown, [49] P. Tang, M.C. Hung, J. Klostergaard, Human pro-tumor necrosis factor is a
M. Robinson, T. Bourne, Mechanism of action of certolizumab pegol homotrimer, Biochemistry 35 (25) (1996) 8216–8225.
(CDP870): in vitro comparison with other anti-tumor necrosis factor alpha [50] M. Grell, E. Douni, H. Wajant, M. Lohden, M. Clauss, B. Maxeiner, S.
agents, Inflamm. Bowel Dis. 13 (11) (2007) 1323–1332. Georgopoulos, W. Lesslauer, G. Kollias, K. Pfizenmaier, P. Scheurich, The
[29] M. Ringheanu, F. Daum, J. Markowitz, J. Levine, S. Katz, X. Lin, J. Silver, Effects transmembrane form of tumor necrosis factor is the prime activating ligand
of infliximab on apoptosis and reverse signaling of monocytes from healthy of the 80 kDa tumor necrosis factor receptor, Cell 83 (5) (1995) 793–802.
individuals and patients with Crohn’s disease, Inflamm. Bowel Dis. 10 (6) [51] A. Domonkos, A. Udvardy, L. Laszlo, T. Nagy, E. Duda, Receptor-like properties
(2004) 801–810. of the 26 kDa transmembrane form of TNF, Eur. Cytokine Netw. 12 (3) (2001)
[30] B. Scallon, A. Cai, N. Solowski, A. Rosenberg, X.Y. Song, D. Shealy, C. Wagner, 411–419.
Binding and functional comparisons of two types of tumor necrosis factor [52] R. Fluhrer, G. Grammer, L. Israel, M.M. Condron, C. Haffner, E. Friedmann, C.
antagonists, J. Pharmacol. Exp. Ther. 301 (2) (2002) 418–426. Bohland, A. Imhof, B. Martoglio, D.B. Teplow, C. Haass, A gamma-secretase-
[31] D.J. Shealy, A. Cai, K. Staquet, A. Baker, E.R. Lacy, L. Johns, O. Vafa, G. Gunn 3rd, like intramembrane cleavage of TNFalpha by the GxGD aspartyl protease
S. Tam, S. Sague, D. Wang, M. Brigham-Burke, P. Dalmonte, E. Emmell, B. SPPL2b, Nat. Cell Biol. 8 (8) (2006) 894–896.
Pikounis, P.J. Bugelski, H. Zhou, B.J. Scallon, J. Giles-Komar, Characterization of [53] E. Friedmann, E. Hauben, K. Maylandt, S. Schleeger, S. Vreugde, S.F.
golimumab, a human monoclonal antibody specific for human tumor Lichtenthaler, P.H. Kuhn, D. Stauffer, G. Rovelli, B. Martoglio, SPPL2a and
necrosis factor alpha, mAbs 2 (4) (2010) 428–439. SPPL2b promote intramembrane proteolysis of TNFalpha in activated dendritic
[32] N. Ueda, H. Tsukamoto, H. Mitoma, M. Ayano, A. Tanaka, S. Ohta, Y. Inoue, Y. cells to trigger IL-12 production, Nat. Cell Biol. 8 (8) (2006) 843–848.
Arinobu, H. Niiro, K. Akashi, T. Horiuchi, The cytotoxic effects of certolizumab [54] P. Vandenabeele, W. Declercq, R. Beyaert, W. Fiers, Two tumour necrosis
pegol and golimumab mediated by transmembrane tumor necrosis factor factor receptors: structure and function, Trends Cell Biol. 5 (10) (1995) 392–
alpha, Inflamm. Bowel Dis. 19 (6) (2013) 1224–1231. 399.
[33] J.M. Van den Brande, H. Braat, G.R. van den Brink, H.H. Versteeg, C.A. Bauer, I. [55] F.K. Chan, H.J. Chun, L. Zheng, R.M. Siegel, K.L. Bui, M.J. Lenardo, A domain in
Hoedemaeker, C. van Montfrans, D.W. Hommes, M.P. Peppelenbosch, S.J. van TNF receptors that mediates ligand-independent receptor assembly and
Deventer, Infliximab but not etanercept induces apoptosis in lamina propria signaling, Science (New York, N.Y.) 288 (5475) (2000) 2351–2354.
T-lymphocytes from patients with Crohn’s disease, Gastroenterology 124 (7) [56] D.J. MacEwan, TNF ligands and receptors–a matter of life and death, Br. J.
(2003) 1774–1785. Pharmacol. 135 (4) (2002) 855–875.
[34] A.C. Vos, M.E. Wildenberg, M. Duijvestein, A.P. Verhaar, G.R. van den Brink, D. [57] I. Carpentier, B. Coornaert, R. Beyaert, Function and regulation of tumor
W. Hommes, Anti-tumor necrosis factor-alpha antibodies induce regulatory necrosis factor receptor type 2, Curr. Med. Chem. 11 (16) (2004) 2205–2212.
macrophages in an Fc region-dependent manner, Gastroenterology 140 (1) [58] M. Higuchi, K. Nagasawa, T. Horiuchi, M. Oike, Y. Ito, M. Yasukawa, Y. Niho,
(2011) 221–230. Membrane tumor necrosis factor-alpha (TNF-alpha) expressed on HTLV-I-
[35] K.A. Wojtal, G. Rogler, M. Scharl, L. Biedermann, P. Frei, M. Fried, A. Weber, J.J. infected T cells mediates a costimulatory signal for B cell activation–
Eloranta, G.A. Kullak-Ublick, S.R. Vavricka, Fc gamma receptor CD64 characterization of membrane TNF-alpha, Clin. Immunol. Immunopathol. 82
modulates the inhibitory activity of infliximab, PLoS ONE 7 (8) (2012) (2) (1997) 133–140.
e43361. [59] A.D. Watts, N.H. Hunt, Y. Wanigasekara, G. Bloomfield, D. Wallach, B.D.
[36] T. Suzuki, A. Ishii-Watabe, M. Tada, T. Kobayashi, T. Kanayasu-Toyoda, T. Roufogalis, G. Chaudhri, A casein kinase I motif present in the cytoplasmic
Kawanishi, T. Yamaguchi, Importance of neonatal FcR in regulating the serum domain of members of the tumour necrosis factor ligand family is implicated
half-life of therapeutic proteins containing the Fc domain of human IgG1: a in ’reverse signalling’, EMBO J. 18 (8) (1999) 2119–2126.
comparative study of the affinity of monoclonal antibodies and Fc-fusion [60] T. Kohno, L.T. Tam, S.R. Stevens, J.S. Louie, Binding characteristics of tumor
proteins to human neonatal FcR, J. Immunol. (Baltimore, Md.: 1950) 184 (4) necrosis factor receptor-Fc fusion proteins vs anti-tumor necrosis factor
(2010) 1968–1976. mAbs, J. Investig. Dermatol. Symp. Proc./The Society for Investigative
[37] P. Biancheri, R.J. Brezski, A. Di Sabatino, A.R. Greenplate, K.L. Soring, G.R. Dermatology, Inc. [and] European Society for Dermatological Research 12
Corazza, K.B. Kok, L. Rovedatti, A. Vossenkamper, N. Ahmad, S.A. Snoek, S. (1) (2007) 5–8.
Vermeire, P. Rutgeerts, R.E. Jordan, T.T. MacDonald, Proteolytic cleavage and [61] T. Arora, R. Padaki, L. Liu, A.E. Hamburger, A.R. Ellison, S.R. Stevens, J.S. Louie,
loss of function of biologic agents that neutralize tumor necrosis factor in the T. Kohno, Differences in binding and effector functions between classes of
mucosa of patients with inflammatory bowel disease, Gastroenterology 149 TNF antagonists, Cytokine 45 (2) (2009) 124–131.
(6) (2015) 1564–1574.e3. [62] H. Mitoma, T. Horiuchi, H. Tsukamoto, Binding activities of infliximab and
[38] R. Palframan, M. Airey, A. Moore, A. Vugler, A. Nesbitt, Use of biofluorescence etanercept to transmembrane tumor necrosis factor-alpha, Gastroenterology
imaging to compare the distribution of certolizumab pegol, adalimumab, and 126 (3) (2004) 934–935. author reply 935-6.

Please cite this article in press as: H. Mitoma et al., Molecular mechanisms of action of anti-TNF-a agents – Comparison among therapeutic TNF-a antag-
onists, Cytokine (2016), http://dx.doi.org/10.1016/j.cyto.2016.08.014
8 H. Mitoma et al. / Cytokine xxx (2016) xxx–xxx

[63] N.H. Ruddle, Lymphotoxin and TNF: how it all began-a tribute to the [84] G. Boschetti, S. Nancey, F. Sardi, X. Roblin, B. Flourie, D. Kaiserlian, Therapy
travelers, Cytokine Growth Factor Rev. 25 (2) (2014) 83–89. with anti-TNFalpha antibody enhances number and function of Foxp3(+)
[64] F. Calmon-Hamaty, B. Combe, M. Hahne, J. Morel, Lymphotoxin alpha regulatory T cells in inflammatory bowel diseases, Inflamm. Bowel Dis. 17 (1)
stimulates proliferation and pro-inflammatory cytokine secretion of (2011) 160–170.
rheumatoid arthritis synovial fibroblasts, Cytokine 53 (2) (2011) 207–214. [85] E. Toubi, A. Kessel, Z. Mahmudov, K. Hallas, M. Rozenbaum, I. Rosner,
[65] C. Buhrmann, P. Shayan, B.B. Aggarwal, M. Shakibaei, Evidence that TNF-beta Increased spontaneous apoptosis of CD4+CD25+ T cells in patients with active
(lymphotoxin alpha) can activate the inflammatory environment in human rheumatoid arthritis is reduced by infliximab, Ann. N. Y. Acad. Sci. 1051
chondrocytes, Arthritis Res. Ther. 15 (6) (2013) R202. (2005) 506–514.
[66] P. Neregard, A. Krishnamurthy, S. Revu, M. Engstrom, E. af Klint, A.I. Catrina, [86] X. Valencia, G. Stephens, R. Goldbach-Mansky, M. Wilson, E.M. Shevach, P.E.
Etanercept decreases synovial expression of tumour necrosis factor-alpha Lipsky, TNF downmodulates the function of human CD4+CD25hi T-regulatory
and lymphotoxin-alpha in rheumatoid arthritis, Scand. J. Rheumatol. 43 (2) cells, Blood 108 (1) (2006) 253–261.
(2014) 85–90. [87] Z. Huang, B. Yang, Y. Shi, B. Cai, Y. Li, W. Feng, Y. Fu, L. Luo, L. Wang, Anti-TNF-
[67] W.P. Kennedy, J.A. Simon, C. Offutt, P. Horn, A. Herman, M.J. Townsend, M.T. alpha therapy improves Treg and suppresses Teff in patients with rheumatoid
Tang, J.L. Grogan, F. Hsieh, J.C. Davis, Efficacy and safety of pateclizumab arthritis, Cell. Immunol. 279 (1) (2012) 25–29.
(anti-lymphotoxin-alpha) compared to adalimumab in rheumatoid arthritis: [88] S. Nadkarni, C. Mauri, M.R. Ehrenstein, Anti-TNF-alpha therapy induces a
a head-to-head phase 2 randomized controlled study (The ALTARA Study), distinct regulatory T cell population in patients with rheumatoid arthritis via
Arthritis Res. Ther. 16 (5) (2014) 467. TGF-beta, J. Exp. Med. 204 (1) (2007) 33–39.
[68] T.W. Spahn, M.K. Muller, W. Domschke, T. Kucharzik, Role of lymphotoxins in [89] H. Nie, Y. Zheng, R. Li, T.B. Guo, D. He, L. Fang, X. Liu, L. Xiao, X. Chen, B. Wan,
the development of Peyer’s patches and mesenteric lymph nodes: relevance Y.E. Chin, J.Z. Zhang, Phosphorylation of FOXP3 controls regulatory T cell
to intestinal inflammation and treatment, Ann. N. Y. Acad. Sci. 1072 (2006) function and is inhibited by TNF-alpha in rheumatoid arthritis, Nat. Med. 19
187–193. (3) (2013) 322–328.
[69] J.M. Shohet, L. Bergamaschini, A.E. Davis, M.C. Carroll, Localization of the [90] B. Zaragoza, X. Chen, J.J. Oppenheim, A. Baeyens, S. Gregoire, D. Chader, G.
human complement component C3 binding site on the IgG heavy chain, J. Gorochov, M. Miyara, B.L. Salomon, Suppressive activity of human regulatory
Biol. Chem. 266 (28) (1991) 18520–18524. T cells is maintained in the presence of TNF, Nat. Med. 22 (1) (2016) 16–17.
[70] J.M. Shohet, P. Pemberton, M.C. Carroll, Identification of a major binding site [91] X. Chen, X. Wu, Q. Zhou, O.M. Howard, M.G. Netea, J.J. Oppenheim, TNFR2 is
for complement C3 on the IgG1 heavy chain, J. Biol. Chem. 268 (8) (1993) critical for the stabilization of the CD4+Foxp3+ regulatory T. cell phenotype in
5866–5871. the inflammatory environment, J. Immunol. (Baltimore, Md.: 1950) 190 (3)
[71] L. Vidarte, C. Pastor, S. Mas, A.B. Blazquez, V. de los Rios, R. Guerrero, F. (2013) 1076–1084.
Vivanco, Serine 132 is the C3 covalent attachment point on the CH1 domain [92] M. Chopra, S.S. Riedel, M. Biehl, S. Krieger, V. von Krosigk, C.A. Bauerlein, C.
of human IgG1, J. Biol. Chem. 276 (41) (2001) 38217–38223. Brede, A.L. Jordan Garrote, S. Kraus, V. Schafer, M. Ritz, K. Mattenheimer, A.
[72] F. Vivanco, E. Munoz, L. Vidarte, C. Pastor, The covalent interaction of C3 with Degla, A. Mottok, H. Einsele, H. Wajant, A. Beilhack, Tumor necrosis factor
IgG immune complexes, Mol. Immunol. 36 (13–14) (1999) 843–852. receptor 2-dependent homeostasis of regulatory T cells as a player in TNF-
[73] A. Sahu, M.K. Pangburn, Covalent attachment of human complement C3 to induced experimental metastasis, Carcinogenesis 34 (6) (2013) 1296–1303.
IgG. Identification of the amino acid residue involved in ester linkage [93] Y. Grinberg-Bleyer, D. Saadoun, A. Baeyens, F. Billiard, J.D. Goldstein, S.
formation, J. Biol. Chem. 269 (46) (1994) 28997–29002. Gregoire, G.H. Martin, R. Elhage, N. Derian, W. Carpentier, G. Marodon, D.
[74] S. Siberil, C.A. Dutertre, C. Boix, E. Bonnin, R. Menez, E. Stura, S. Jorieux, W.H. Klatzmann, E. Piaggio, B.L. Salomon, Pathogenic T cells have a paradoxical
Fridman, J.L. Teillaud, Molecular aspects of human FcgammaR interactions protective effect in murine autoimmune diabetes by boosting Tregs, J. Clin.
with IgG: functional and therapeutic consequences, Immunol. Lett. 106 (2) Investig. 120 (12) (2010) 4558–4568.
(2006) 111–118. [94] F.E. McCann, D.P. Perocheau, G. Ruspi, K. Blazek, M.L. Davies, M. Feldmann, J.L.
[75] T. ten Hove, C. van Montfrans, M.P. Peppelenbosch, S.J. van Deventer, Dean, A.A. Stoop, R.O. Williams, Selective tumor necrosis factor receptor I
Infliximab treatment induces apoptosis of lamina propria T lymphocytes in blockade is antiinflammatory and reveals immunoregulatory role of tumor
Crohn’s disease, Gut 50 (2) (2002) 206–211. necrosis factor receptor II in collagen-induced arthritis, Arthritis Rheumatol.
[76] J.M. Van den Brande, T.C. Koehler, Z. Zelinkova, R.J. Bennink, A.A. te Velde, F.J. (Hoboken, N.J.) 66 (10) (2014) 2728–2738.
ten Cate, S.J. van Deventer, M.P. Peppelenbosch, D.W. Hommes, Prediction of [95] D.X. Nguyen, M.R. Ehrenstein, Anti-TNF drives regulatory T cell expansion by
antitumour necrosis factor clinical efficacy by real-time visualisation of paradoxically promoting membrane TNF-TNF-RII binding in rheumatoid
apoptosis in patients with Crohn’s disease, Gut 56 (4) (2007) 509–517. arthritis, J. Exp. Med. 213 (7) (2016) 1241–1253.
[77] C. Bedini, F. Nasorri, G. Girolomoni, O. Pita, A. Cavani, Antitumour necrosis [96] A.C. Vos, M.E. Wildenberg, I. Arijs, M. Duijvestein, A.P. Verhaar, G. de Hertogh,
factor-alpha chimeric antibody (infliximab) inhibits activation of skin- S. Vermeire, P. Rutgeerts, G.R. van den Brink, D.W. Hommes, Regulatory
homing CD4+ and CD8+ T lymphocytes and impairs dendritic cell function, macrophages induced by infliximab are involved in healing in vivo and
Br. J. Dermatol. 157 (2) (2007) 249–258. in vitro, Inflamm. Bowel Dis. 18 (3) (2012) 401–408.
[78] F. Caprioli, F. Bose, R.L. Rossi, L. Petti, C. Vigano, C. Ciafardini, L. Raeli, G. [97] P. Sondermann, D.E. Szymkowski, Harnessing Fc receptor biology in the
Basilisco, S. Ferrero, M. Pagani, D. Conte, G. Altomare, G. Monteleone, S. design of therapeutic antibodies, Curr. Opin. Immunol. 40 (2016) 78–87.
Abrignani, E. Reali, Reduction of CD68+ macrophages and decreased IL-17 [98] E. Louis, Z. El Ghoul, S. Vermeire, S. Dall’Ozzo, P. Rutgeerts, G. Paintaud, J.
expression in intestinal mucosa of patients with inflammatory bowel disease Belaiche, M. De Vos, A. Van Gossum, J.F. Colombel, H. Watier, Association
strongly correlate with endoscopic response and mucosal healing following between polymorphism in IgG Fc receptor IIIa coding gene and biological
infliximab therapy, Inflamm. Bowel Dis. 19 (4) (2013) 729–739. response to infliximab in Crohn’s disease, Aliment. Pharmacol. Ther. 19 (5)
[79] R.S. Wallis, M.S. Broder, J.Y. Wong, M.E. Hanson, D.O. Beenhouwer, (2004) 511–519.
Granulomatous infectious diseases associated with tumor necrosis factor [99] E.J. Louis, H.E. Watier, S. Schreiber, J. Hampe, F. Taillard, A. Olson, N. Thorne,
antagonists, Clin. Infect. Dis.: Official Publ. Infect. Dis. Soc. Am. 38 (9) (2004) H. Zhang, J.F. Colombel, Polymorphism in IgG Fc receptor gene FCGR3A and
1261–1265. response to infliximab in Crohn’s disease: a subanalysis of the ACCENT I
[80] H. Bruns, C. Meinken, P. Schauenberg, G. Harter, P. Kern, R.L. Modlin, C. study, Pharmacogenet. Genomics 16 (12) (2006) 911–914.
Antoni, S. Stenger, Anti-TNF immunotherapy reduces CD8+ T cell-mediated [100] R. Moroi, K. Endo, Y. Kinouchi, H. Shiga, Y. Kakuta, M. Kuroha, Y. Kanazawa, Y.
antimicrobial activity against Mycobacterium tuberculosis in humans, J. Clin. Shimodaira, T. Horiuchi, S. Takahashi, T. Shimosegawa, FCGR3A-158
Investig. 119 (5) (2009) 1167–1177. polymorphism influences the biological response to infliximab in Crohn’s
[81] R. Atreya, M. Zimmer, B. Bartsch, M.J. Waldner, I. Atreya, H. Neumann, K. disease through affecting the ADCC activity, Immunogenetics 65 (4) (2013)
Hildner, A. Hoffman, R. Kiesslich, A.D. Rink, T.T. Rau, S. Rose-John, H. Kessler, J. 265–271.
Schmidt, M.F. Neurath, Antibodies against tumor necrosis factor (TNF) induce [101] D. Ternant, Z. Berkane, L. Picon, V. Gouilleux-Gruart, J.F. Colombel, M. Allez, E.
T-cell apoptosis in patients with inflammatory bowel diseases via TNF Louis, G. Paintaud, Assessment of the influence of inflammation and FCGR3A
receptor 2 and intestinal CD14(+) macrophages, Gastroenterology 141 (6) genotype on infliximab pharmacokinetics and time to relapse in patients
(2011) 2026–2038. with Crohn’s disease, Clin. Pharmacokinet. 54 (5) (2015) 551–562.
[82] F.S. Kleijwegt, S. Laban, G. Duinkerken, A.M. Joosten, A. Zaldumbide, T. Nikolic, [102] C.H. Lee, D.K. Choi, H.J. Choi, M.Y. Song, Y.S. Kim, Expression of soluble and
B.O. Roep, Critical role for TNF in the induction of human antigen-specific functional human neonatal Fc receptor in Pichia pastoris, Protein Expr. Purif.
regulatory T cells by tolerogenic dendritic cells, J. Immunol. (Baltimore, Md.: 71 (1) (2010) 42–48.
1950) 185 (3) (2010) 1412–1418. [103] U. Mahadevan, D.C. Wolf, M. Dubinsky, A. Cortot, S.D. Lee, C.A. Siegel, T.
[83] M. Nagar, J. Jacob-Hirsch, H. Vernitsky, Y. Berkun, S. Ben-Horin, N. Amariglio, I. Ullman, S. Glover, J.F. Valentine, D.T. Rubin, J. Miller, M.T. Abreu, Placental
Bank, Y. Kloog, G. Rechavi, I. Goldstein, TNF activates a NF-jB-regulated cellular transfer of anti-tumor necrosis factor agents in pregnant patients with
program in human CD45RA- regulatory T cells that modulates their suppressive inflammatory bowel disease, Clin. Gastroenterol. Hepatol.: Official Clin.
function, J. Immunol. (Baltimore, Md.: 1950) 184 (7) (2010) 3570–3581. Practice J. Am. Gastroenterol. Assoc. 11 (3) (2013) 286–292. quiz e24.

Please cite this article in press as: H. Mitoma et al., Molecular mechanisms of action of anti-TNF-a agents – Comparison among therapeutic TNF-a antag-
onists, Cytokine (2016), http://dx.doi.org/10.1016/j.cyto.2016.08.014

You might also like