You are on page 1of 12

Journal of Pathology

J Pathol 2008; 214: 149–160


Published online in Wiley InterScience
(www.interscience.wiley.com) DOI: 10.1002/path.2287

Invited Review

TNF-mediated inflammatory disease


JR Bradley*
NIHR Cambridge Biomedical Research Centre, Addenbrooke’s Hospital, Cambridge, UK

*Correspondence to: Abstract


JR Bradley, NIHR Cambridge
Biomedical Research Centre, Box TNF was originally described as a circulating factor that can cause necrosis of tumours,
118, Addenbrooke’s Hospital, but has since been identified as a key regulator of the inflammatory response. This
Cambridge CB2 2QQ, UK. review describes the known signalling pathways and cell biological effects of TNF, and
E-mail: jrb1000@cam.ac.uk our understanding of the role of TNF in human disease. TNF interacts with two different
receptors, designated TNFR1 and TNFR2, which are differentially expressed on cells and
No conflicts of interest were
tissues and initiate both distinct and overlapping signal transduction pathways. These diverse
declared.
signalling cascades lead to a range of cellular responses, which include cell death, survival,
differentiation, proliferation and migration. Vascular endothelial cells respond to TNF by
undergoing a number of pro-inflammatory changes, which increase leukocyte adhesion,
transendothelial migration and vascular leak and promote thrombosis. The central role of
TNF in inflammation has been demonstrated by the ability of agents that block the action of
TNF to treat a range of inflammatory conditions, including rheumatoid arthritis, ankylosing
spondylitis, inflammatory bowel disease and psoriasis. The increased incidence of infection
in patients receiving anti-TNF treatment has highlighted the physiological role of TNF in
infectious diseases.
Copyright  2007 Pathological Society of Great Britain and Ireland. Published by John
Wiley & Sons, Ltd.
Keywords: tumour necrosis factor; signal transduction; inflammation; infection; arthritis;
therapy

Introduction their trimeric forms, and the two forms of TNF may
have distinct biological activities. TNFα converting
Tumour necrosis factor (TNF, also known as TNFα) enzyme (TACE, also known as ADAM-17) medi-
was identified in 1975 as an endotoxin-induced glyco- ates release of TNF from the cell surface [3], but is
protein, which caused haemorrhagic necrosis of sarco- involved in processing several cell-membrane asso-
mas that had been transplanted into mice [1]. Human ciated proteins, including TNF receptors, which are
tumour necrosis factor was cloned in 1985 [2], and released by its action to produce soluble froms that can
recombinant TNF was shown to induce the haem- neutralize the actions of TNF [4]. TACE may there-
orrhagic necrosis of transplanted methylcholanthrene- fore be either pro- or anti-inflammatory, depending on
induced sarcomas in syngeneic mice. TNF has since whether it acts on an effector (eg macrophage) or tar-
been implicated in a diverse range of inflammatory, get (eg endothelial) cell, releasing ligand or receptors,
infectious and malignant conditions, and the impor- respectively.
tance of TNF in inflammation has been highlighted by TNF is not usually detectable in healthy individ-
the efficacy of anti-TNF antibodies or administration uals, but elevated serum and tissue levels are found
of soluble TNF receptors (TNFRs) in controlling dis- in inflammatory and infectious conditions [5,6] and
ease activity in rheumatoid arthritis and other inflam- serum levels correlate with the severity of infections
matory conditions. [7,8]. Although cells of the monocyte/macrophage lin-
eage are the main source of TNF in inflammatory dis-
ease, a wide range of cells can produce TNF, including
Production of TNF mast cells, T and B lymphocytes, natural killer (NK)
cells, neutrophils, endothelial cells, smooth and car-
TNF is produced predominantly by activated macro- diac muscle cells, fibroblasts and osteoclasts.
phages and T lymphocytes as a 26 kDa protein,
pro-TNF, which is expressed on the plasma mem- TNF signal transduction
brane, where it can be cleaved in the extracellular
domain by the matrix metalloproteinases, which result TNF signal transduction pathways are complex and
in the release a soluble 17 kDA soluble form. Both still not fully understood. Regulation of the transcrip-
membrane-associated and soluble TNFs are active in tion factor NF-κB is a key component of TNF signal

Copyright  2007 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
www.pathsoc.org.uk
150 JR Bradley

Figure 1. Signalling pathways leading to the main cellular responses of TNF. Soluble TNF receptors or monoclonal anti-TNF
antibodies, which prevent TNF interacting with its receptors and activating these pathways, can be used to treat inflammatory
disease

transduction, but large-scale physical mapping in com- TNFR1 signals by recruitment of TNFR-associated-
bination with loss of function analysis using RNA death-domain protein (TRADD) [12]. TNFR1 is a
interference recently identified 221 molecular associa- type I transmembrane protein, which in resting cells
tions and 80 previously unknown interactors involved is predominantly sequestered in the Golgi appara-
in modulation of the TNF–NF-κB pathway alone [9]. tus, from where it can be mobilized to the cell sur-
All known responses to TNF are triggered by bind- face.
ing to one of two distinct receptors (Figure 1), des- The significance of the Golgi pool of TNFR1
ignated TNFR1 (also known as TNFRSF1A, CD120a, molecules is unclear. One hypothesis is that it may act
p55) and TNFR2 (also known as TNFRSF1B, CD120b, as a reservoir to increase surface receptor expression
p75), which are differentially regulated on various density, thereby sensitizing cells to the actions of
cell types in normal and diseased tissue [10]. The TNF. There is precedence for this idea in smooth
extracellular, ligand-binding domains of TNF recep- muscle cells, in which the TNF receptor family
tors contain cysteine-rich subdomains, characteristic member Fas localizes predominantly to the Golgi,
of members of the nerve growth factor/TNF recep- from where it can be translocated to the cell surface,
tor gene family. In contrast, the intracellular domains thereby sensitizing cells to Fas ligand-induced killing
of the two receptors show no sequence homology and [13].
are devoid of intrinsic enzyme activity, and activate Surface-expressed TNFR1 exist as trimers associ-
distinct signal transduction pathways by recruitment ated through pre-ligand assembly domains (PLADs)
of cytosolic proteins through specific protein–protein [14], which reside within the membrane distal
interaction domains [11]. The ability of TNFR1 and cysteine-rich domain. In unstimulated receptors the
TNFR2 to interact with both identical and unrelated cytoplasmic domain is pre-associated with a cyto-
molecules may explain their shared and diverse func- plasmic protein designated silencer of death domain
tions. Based on cell culture work and studies with (SODD) [15]. SODD is thought to prevent constitu-
receptor knockout mice, both the pro-inflammatory tive signalling of TNFR1, although mice congenitally
and the programmed cell death pathways that are acti- deficient in expression of the sodd gene display normal
vated by TNF, and associated with tissue injury, are TNFR1 signalling [16]. Both TNFR1 and SODD con-
largely mediated through TNFR1. The consequences tain ‘death domains’ (DDs), which are protein motifs
of TNFR2 signalling are less well characterized, but that interact with other DDs. SODD uses its DD to
TNFR2 has been shown to mediate signals that pro- bind to the DD contained within the cytosolic por-
mote tissue repair and angiogenesis. tion of TNFR1, preventing signalling. TNF binding to

J Pathol 2008; 214: 149–160 DOI: 10.1002/path


Copyright  2007 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
TNF-mediated inflammatory disease 151

TNFR1 may result in the release of SODD, allowing pro-caspase 3. The recruitment and activation of pro-
binding of a different DD-containing cytoplasmic pro- caspase 8 can be inhibited by cFLIP, and elevated
tein, TNFR-associated DD protein (TRADD). TRADD levels of cFLIP induced by NF-κB activation may pre-
initiates signalling by recruiting two additional pro- vent the activation of this death pathway [30]. TNFR1
teins, receptor interacting protein-1 (RIP-1), a ser- activates other signalling responses that are less clearly
ine/threonine kinase which binds to TRADD through defined at a molecular level. These include activa-
its own DD [17], and TNFR-associated factor-2 tion of the ras–raf–MEK1–ERK1,2 pathway [31] and
(TRAF-2) an E3 ubiquitin ligase [18]) that does not phosphatidylinositol-3 kinase (PI3K), which phospho-
contain a DD. Within minutes this complex is internal- rylates the membrane lipid phosphatidylinositol 4.5
ized [12], and the TRADD–RIP-1–TRAF2 complex diphosphate (PIP2 ), converting it to phosphatidylinos-
is released from TNFR1. Inhibitors of this process can itol 3, 4, 5 triphosphate (PIP3 ). PIP3 activates PDK-1,
interfere with signalling [19,20]. an enzyme that phosphorylates the kinase Akt, and
Subsequent signalling events involve recruitment also activates PDK-2, a complex containing mam-
and activation of different mitogen activated protein malian target of rapamycin (mTOR), rictor and SAPK-
kinase kinase kinases (MAP3Ks)]. RIP-1 is thought interacting protein (Sin)1, which is also involved in
to mediate recruitment of MEKK-3 and transforming activation of Akt [32]. ERK-1, -2 and Akt are gen-
growth factor-beta (TGFβ)-activated kinase (TAK)1, erally associated with cell survival and proliferation
which in turn, activate the β-subunit of the inhibitor [33].
of κB (IκB)] kinase (IKK) complex [21,22], leading to The signalling pathways initiated by TNFR2 are
phosphorylation of IκB proteins, signalling IκB ubiq- less clearly defined, but TNFR2 appears to sig-
uitination and proteosome-mediated degradation. Cys- nal both shared and opposing effects to TNFR1.
tosolic IκB proteins from a complex with the NF-κB TNFR2 lacks an intracellular death domain, but can
family transcription factors, masking nuclear localiza- interact with TRAFs. TRAF1 was initially identi-
tion signals within NF-κB, and IκB degradation allows fied as a novel 45 kDa protein that could be co-
NF-κB to enter the nucleus and initiate gene transcrip- immunoprecipitated with human TNFR2 transfected
tion. into the murine interleukin-2-dependent cytotoxic T
TRAF2 can also contribute to NF-κB activation, cell line CT6, and also from CT6 cell lysates by a
GST fusion protein containing the region of human
both through binding of the IKK complex [23] and
TNFR2 required for signal transduction [34]. At
through recruitment of inhibitor of cellular apop-
the same time, TRAF2 was identified as a novel
tosis proteins (cIAP)-1 and -2. cIAPs have cas-
56 kDa protein by using the yeast two-hybrid sys-
pase inhibitors that also have ubiquitin protein ligase
tem to detect proteins that interact directly with the
(E3)] activity and can participate in IκB degrada-
cytoplasmic domain of hTNFR2. Despite the co-
tion [24]. The TRADD–RIP-1–TRAF-2 complex can immunoprecipitation studies, only a very weak interac-
also recruit apoptosis-signalling kinase-1 (ASK-1)], a tion between TRAF1 and the cytoplasmic domains of
MAP3K that associates with TRAF2 [25] and activates hTNFR2 or mTNFR2 could be detected using the two-
MAP2Ks, including MEK-4 and -6 [26,27]. These hybrid system. This seeming contradiction was rec-
MEKs phosphorylate and activate c-Jun N-terminal onciled by the observations that a strong heteromeric
kinases (JNKs) and p38 MAPKs. Activated JNKs interaction occurred between TRAF1 and TRAF2, and
phosphorylate the amino terminal region of c-Jun, a that TRAF1 and TRAF2 could form homo- and het-
subunit of the transcription factor activating protein 1 erotypic dimers. Consequently, TRAF1 and TRAF2
(AP-1). Phosphorylation of the amino terminal region can associate with the cytoplasmic domain of TNFR2
of c-Jun by JNK is essential for interactions of this as a heterodimeric complex in which only TRAF2 con-
protein with cAMP-response-element-binding-protein- tacts the receptor directly. However, despite inherent
binding protein [CBP]/p300 and gene transcription. strong TRAF2 binding activity, ligand-dependent acti-
TNFR1 can also interact with FAN (factor associated vation of TNFR2 does not appear to deliver strong
with neutral SMase activation) to activate neutral sph- TRAF2-dependent signals, and TNFR2 has since been
ingomyelinase, which can generate ceramide from the found to have a TRAF2-binding site with high inher-
breakdown of plasma membrane sphingomyelin [28] ent signalling capabilities, designated T2bs-N, together
and initiate apoptosis through the mitochondrial path- with a carboxyl-terminal TRAF2-binding site des-
way [29]. ignated T2bs-C that prevents the delivery of sig-
In addition to mediating cell survival and pro- nals from T2bs-N [35]. This may provide a mech-
inflammatory signals through NF-κB and AP-1, anism for diverting TRAF2 from TNFR1 via T2bs-
TNFR1 can also initiate cell death signalling path- C without inducing TRAF2-mediated signals. Bind-
ways. This involves the binding of Fas-associated ing of TNF to TNFR2 can also limit signalling by
DD protein (FADD) to TRADD and the subsequent c-IAP1-dependent ubiquination and degradation of
recruitment of pro-caspase 8 by the TRADD–FADD TRAF2 and ASK1, terminating MAP3K signalling
complex. Autocatalytic activation of bound pro- [36].
caspase 8 releases activated caspase 8, which ini- TNFR2 can also activate endothelial/epithelial tyro-
tiates apoptotis through cleavage and activation of sine kinase (Etk), a cytosolic kinase implicated in

J Pathol 2008; 214: 149–160 DOI: 10.1002/path


Copyright  2007 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
152 JR Bradley

cell adhesion, migration, proliferation and survival, TNFR1 is more commonly down-regulated by these
independently of TRAF2. Etk is a novel regulator of stimuli [43–45]. The possibility of differential syn-
epithelial cell junctions and mediates the TNF-induced thesis of the receptors is further supported by charac-
phosphatidylinositol 3-kinase (PI3K)–Akt angiogenic terization of their promoter regions. The 5 -regulatory
pathway in vascular endothelial cells through Etk- region of TNFR1 possesses features of a housekeeping
mediated cross-talk with vascular endothelial growth promoter [46,47]. The inducibility of TNFR2 is sup-
factor receptor 2 (VEGFR2) [37]. TNF activates Etk ported by analysis of its promoter region, which has
through TNFR2 in a TRAF2-independent manner. a cAMP-response element and consensus elements for
TNFR2 associates with an inactive form of Etk in a a number of transcription factors, including NF-κB,
ligand-independent fashion through the C-terminal 16- AP-1, IRF-1 and GAS [42]. An additional factor that
amino acid sequence of TNFR2 and multiple domains is known to influence expression of TNF receptors
of Etk. TNF is thought to induce a conformational is shedding from the cell surface. TNF-binding pro-
change in TNFR2 that triggers unfolding of the teins, later characterized as soluble forms of the two
closed, inactive form of Etk. In endothelial cells, TNF molecular species of cell surface TNF receptors, were
induces assembly of a trimolecular complex contain- first purified from normal human urine [48]. Inflam-
ing TNFR2, Etk and vascular endothelial growth factor matory mediators are known to induce shedding of
receptor 2 (VEGFR2, also known as KDR or flk-1). TNFR1 from endothelial cells [49,50], and nitric oxide
Within this complex, there is a coordinate recipro- and hydrogen peroxide have been implicated in the
cal phosphorylation of Etk and VEGFR2, resulting in activation of a metalloproteinase involved in shed-
PI3K activation [37]. The appearance of phosphory- ding of TNFR1 [51]. The TNF receptor-associated
lated Etk is indicative of TNFR2 signalling [38]. periodic syndrome (TRAPS) is an autoinflammatory
The utilization of different signalling mechanisms syndrome characterized by episodes of fevers, with
by TNFR1 and TNFR2 is consistent with the ability severe localized inflammation [52]. TRAPS is associ-
of each receptor to signal distinct biological responses ated with heterozygous mutations in the extracellular
in cultured cells. Ligation of TNFR1 is both nec- domain of TNFR1, which are associated with receptor
essary and sufficient to induce cytotoxic and pro- misfolding, an inability to form soluble receptors and
inflammatory TNF responses, whereas TNFR2 may altered signalling [53].
promote cell activation, migration or proliferation.
Under certain circumstances, TNFR2 may contribute
to TNFR1 responses, particularly at low concentra- Cell biological effects of TNF in the
tions of TNF [39], consistent with the notion of ‘ligand inflammatory response
passing’, in which TNFR2 captures TNF and passes
it to TNFR1 [40]. Cooperation between the receptors Although TNF receptors are differentially expressed
may also be explained by the ligand-induced formation on a wide range of cells and tissues, many of the pro-
of TNF receptor heterocomplexes [41]. inflammatory effects of TNF can be explained on the
basis of TNF’s effects on vascular endothelium and
endothelial leukocyte interactions. In response to TNF,
Regulation of TNF receptors endothelial cells promote inflammation by displaying,
in a distinct temporal, spatial and anatomical pattern
Most cell lines and primary tissues co-express both [54–56], different combinations of adhesion molecules
TNF receptors, although TNFR2 is preferentially for leukocytes, including E-selectin, intercellular adhe-
expressed on cells of haematopoietic lineage [42]. TNF sion molecule-1 (ICAM-1) and vascular cell adhesion
receptor types show a much more limited pattern of molecule-1 (VCAM-1) [57,58]. In combination with
cellular expression in vivo, and are highly regulated the release of chemokines (including IL-8, MCP-1
by ischaemic or inflammatory tissue injury. For exam- and IP-10) [59], these responses lead to recruitment
ple, normal kidney appears largely devoid of TNFR2 of different populations of leukocytes independent of
molecules, and TNFR1 molecules are essentially con- antigen recognition. In addition, many of the clas-
fined to microvascular and glomerular endothelial sical features of inflammation can be produced by
cells. This restricted protein distribution is supported local effects of TNF on endothelial cells. TNF-induced
by in situ hybridization studies for receptor-encoding expression of cyclo-oxygenase 2 can increase EC pro-
mRNA. In kidney allografts undergoing rejection or duction of vasodilatory PGI2 resulting in vasodilata-
ischaemic injury, TNFR1 is lost from the endothelium tion [60], causing ‘rubor’ and ‘calor’ through increased
and there is new expression of TNFR2 in renal tubu- local blood flow. ‘Tumour’ can result from TNF-
lar epithelial cells. This regulated expression of TNF mediated increased vascular permeability, allowing the
receptors, which is recognized to occur in a wide range increased trans-endothelial passage of fluid and macro-
of tissues, is likely to result from changes in both the molecules to create oedema. In addition, TNF-induced
rate of synthesis and shedding of receptors. expression of pro-coagulant proteins, such as tissue
A number of stimuli, including TNF, IL-1, IL-10 factor, and down-regulation of anticoagulant protein,
and tissue plasminogen activator, increase expression such as thrombomodulin TNF, can cause intravascular
of TNFR2 through transcriptional activation, whereas thrombosis [61].

J Pathol 2008; 214: 149–160 DOI: 10.1002/path


Copyright  2007 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
TNF-mediated inflammatory disease 153

Physiological roles of TNF has been provided by the existence of viruses encoding
TNF-binding proteins in their genome [74]. Further-
One of the major biological roles of TNF is in the more, side-effects typically associated with viraemia,
host defence to bacterial, viral and parasitic infec- including fever, rigors, headache and fatigue, were
tions. Physiologically, TNF is important for the normal observed in early trials of TNF in cancer patients
response to infection, but inappropriate or excessive [75,76]. Indeed, although initially described as a
production can be harmful. TNF was originally identi- tumouricidal agent, toxicity has limited the role of
fied as an endotoxin lipopolysaccharide (LPS)-induced TNF as a chemotherapeutic agent for cancer. Further-
humoral mediator of murine cachexia, the syndrome more, TNF may under some circumstances contribute
of anorexia, weight loss and protein wasting that com- to carcinogenesis by promoting proliferation, inva-
plicates cancer and chronic infection and inflammation sion and metastasis of tumour cells [77]. However,
[62]. TNF was confirmed as the principal mediator of isolated limb perfusion with TNF is of value in palli-
the lethal effect of Escherichia coli-derived endotoxin ating patients with metastatic sarcoma and melanoma
by demonstrating that passive immunization of mice [78,79], providing tumour control and limb salvage for
with rabbit anti-serum to TNF protected mice from its the short survival of patients, and isolated hepatic per-
lethality [63]. Baboons passively immunized with a fusion with TNF has been used in patients with hepatic
neutralizing monoclonal anti-TNF antibody and sub- metastases [80].
sequently infused with a lethal dose of live E. coli Although TNF blockade failed to be of benefit in
were protected against shock, vital organ dysfunction, severe sepsis, the trials that were undertaken paved
persistent stress hormone release and death [64]. How- the way for its use in chronic inflammatory diseases
ever, although a recombinant, soluble fusion protein such as rheumatoid arthritis, which in turn has high-
that combines an extracellular portion of the human lighted the physiological roles of TNF in sepsis and
TNF receptor and the Fc portion of IgG (TNFR : Fc) malignancy.
neutralizes TNF and prevents death in animal mod-
els of bacteraemia and endotoxaemia, in patients with
septic shock, treatment with TNFR : Fc did not reduce Therapeutic agents for TNF blockade
mortality, and higher doses appeared to be associated
with increased mortality [65]. Three drugs, Humira (adalibumab), Remicade (inflix-
The importance of TNF as a modulator of the host imab) and Enbrel (etanercept) are currently licensed
response to infection has been confirmed by stud- as TNF-blocking agents and used to treat rheumatoid
ies using TNF receptor-deficient mice. Mice deficient arthritis and other inflammatory diseases, including
in TNFR1 are resistant to lethal dosages of either ankylosing spondylitis and Crohn’s disease. Etaner-
lipopolysaccharides or Staphylococcus aureus entero- cept is a recombinant human soluble fusion protein in
toxin B, but have severely impaired clearance of the which TNFR2 is coupled to the Fc portion of IgG.
intracellular bacterium Listeria monocytogenes and Infliximab is a human–murine chimeric IgG1 mon-
readily succumb to this infection [66,67]. The activity oclonal anti-TNF antibody. Adalibumab is a human
of TNF in endotoxin-induced lethal shock and innate anti-human TNF antibody produced by phage display.
resistance to Listeria appears to be independent of Other agents in clinical development have used PEGy-
TNFR2 [68]. TNF has since been shown to be essen- lation to couple a large molecular weight polyethylene
tial for the formation and maintenance of granulomas, glycol molecule to the TNF antagonist, with the aim of
which limit dissemination of Listeria and other infec- prolonging the half-life. PEG–sTNFR1 is a pegylated
tions, including mycobacteria [69], and a number of form of soluble TNFR1, and CDP-870 is a pegylated
granulomatous infections have been reported in asso- Fab of the humanized anti-TNF antibody CDP-571.
ciation with the use of TNF antagonists to treat human Different anti-TNF therapies may have different
inflammatory disease [70]. binding and pharmacokinetic profiles. The chimeric
Evidence also supports a key role for TNF in para- monoclonal antibody infliximab may be a more potent
sitic and viral infections. TNF levels are increased in inhibitor of TNFR2 signalling than the TNFR2–Fc
the serum of children with uncomplicated Plasmod- fusion protein entarecept. Infliximab binds transmem-
ium falciparum malaria, and markedly increased in brane TNF with higher avidity, forming more sta-
children with a fatal outcome from cerebral malaria, ble complexes and more effectively inhibiting the
leading to speculation that increased TNF production actions of transmembrane TNF than entarecept [81].
is a normal host response to P. falciparum infection, Transmembrane TNF is superior to soluble TNF in
but that excessive levels of production may predispose activating TNFR2 in various systems, including T
to cerebral malaria and a fatal outcome [8]. cell activation, thymocyte proliferation and granulo-
TNF appears to be central for the ICAM-1- cyte/macrophage colony-stimulating factor production
dependent recruitment of mononuclear cells and [82]. Thus, under certain conditions infliximab may be
microvascular damage that occurs in cerebral malaria more effective at blocking signalling through TNF2
[71]. Anti-TNF therapy inhibits fever in cerebral than etanercept.
malaria [72] but does not improve survival [73]. Sup- The US Food and Drug Administration (FDA)
port for a role of TNF in host defences against viruses has reported certain serious, but uncommon, adverse

J Pathol 2008; 214: 149–160 DOI: 10.1002/path


Copyright  2007 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
154 JR Bradley

events with all three approved anti-TNF agents Inflammatory bowel disease
(http://www.fda.gov/ohrms/dockets/ac/03/briefing/
3930B1 01 B-TNF.Briefing.htm), including serious TNF immunoreactivity is increased the lamina propria
bacterial infections, tuberculosis and certain oppor- in intestinal specimens from patients with Crohn’s
tunistic infections, demyelinating syndromes and sys- disease and ulcerative colitis [100,101], and mice
temic lupus erythaematosus-like reactions. In con- overexpressing TNF develop a Crohn’s disease-like
trolled studies of TNF blockade, more cases of lym- inflammatory bowel disease [90].
phoma occurred among patients receiving the agents Infliximab has been shown to be effective in induc-
than among control patients, and in open-labelled ing remission, and an effective maintenance ther-
uncontrolled studies the rate of malignancies in the apy for patients with Crohn’s disease with and
treatment group was several times higher than would without fistulas [102,103]. A randomized controlled
be expected in the general population. The avail- trial demonstrated that the humanized anti-TNF anti-
able data suggest that these adverse reactions may be body CDP571 was an effective for treatment of
related to TNF blockade and therefore represent class patients with moderate to severe Crohn’s disease
effects of these agents. [104], but subsequent studies showed that CDP571
is not effective for long-term treatment of unselected
patients with moderate to severe Crohn’s disease
Rheumatoid arthritis
[105].
Rheumatoid arthritis is a chronic autoimmune inflam- A systemic review of randomized controlled trials of
matory disorder affecting approximately 1% of the TNF-blocking agents in patients with active Crohn’s
population, characterized by inflammation of syn- disease found that infliximab and CDP571, but not
ovial tissue, leading to progressive damage, erosion etanercept, may be effective in inducing remission
of adjacent cartilage and bone and chronic disabil- [106].
ity. The inflammation is associated with accumu- The role of TNFα-blocking agents in ulcerative col-
lation of inflammatory cells, predominantly T cells itis is less clear, and recent studies have yielded con-
and macrophages, but also B cells, plasma cells and flicting results. A systematic review concluded that
dendritic cells. There is synovial hyperplasia and in patients with moderate to severe ulcerative coli-
angiogenesis is a prominent feature [83]. Many pro- tis whose disease is refractory to conventional treat-
inflammatory cytokines, including IL-1, IL-6, TNF ment using corticosteroids and/or immunosuppres-
and GM-CSF, are produced within the inflamed joint sive agents, infliximab is effective in inducing clin-
[84]. ical remission, inducing clinical response, promoting
Support for a dominant role of TNF was provided mucosal healing and reducing the need for colectomy,
by a number of in vitro and in vivo studies. Production at least in the short term [107].
of a range of pro-inflammatory cytokines by cultured
cells from the joints of patients with rheumatoid arthri-
tis can be down-regulated by a neutralizing antibody Ankylosing spondylitis
to TNF [85,86]. Neutralizing TNF with either anti-
TNF antibody or a recombinant soluble TNF recep- Ankylosing spondylitis is an inflammatory arthritis
tor ameliorates joint disease in a murine model of that predominantly affects the spine and sacroiliac
collagen-induced arthritis [87–89]. Deleting TNF AU- joints. It occurs more commonly in patients with
rich elements (AREs) from the mouse genome resulted Crohn’s disease. TNF has been detected in the sacro-
in profound temporal and spatial deregulation of TNF iliac joints of patients with ankylosing spondylitis
production, characterized by the persistent accumula- [108], particularly in early active disease [109], and
tion and decreased rates of decay of the mutant TNF elevated serum levels of TNF correlate with disease
mRNA. This deregulation resulted in overexpression activity [110].
of TNF and the development of chronic inflammatory A randomized, double-blind trial of etanercept
arthritis and inflammatory bowel disease [90]. showed that treatment with etanercept for 4 months
An open Phase I/II clinical trial of a murine-human resulted in rapid, significant and sustained improve-
chimeric neutralizing monoclonal antibody to TNF in ment in patients with ankylosing spondylitis [111].
20 patients with active rheumatoid arthritis demon- Subsequent studies have confirmed the efficacy and
strated that the treatment was safe and well toler- safety of etanercept in patients with active ankylos-
ated and resulted in significant clinical and laboratory ing spondlitis over 2 years of continuous treatment
improvements [91]. The efficacy of anti-TNF treat- [112,113], and have shown that etanercept is effec-
ments in rheumatoid arthritis was subsequently con- tive in patients with early onset ankylosing spondylitis
firmed in randomized controlled trials, which estab- before the age of 18 [114].
lished that methotrexate and anti-TNF treatment have Infliximab is also an effective agent in patients
a synergistic effect, and demonstrated that long- with active ankylosing spondylitis, for inducing and
term treatment is feasible [92–98], although loss of maintaining remission and readministration to treat
response may occur in about half of patients during relapse after discontinuation of long-term therapy
the first year [99]. [115,116].

J Pathol 2008; 214: 149–160 DOI: 10.1002/path


Copyright  2007 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
TNF-mediated inflammatory disease 155

Psoriasis Patients with chronic inflammatory conditions such


as rheumatoid arthritis have an increased incidence
Psoriasis is an inflammatory skin disorder, in which an
of cardiovascular disease. Inflammatory mediators,
inflammatory cell infiltrate is associated with hyperk-
including TNF, have been implicated in this increased
eratotic lesions, giving rise to typical psoriatic plaques.
cardiovascular risk, and there is some evidence that
TNF, TNFR1 and TNFR2 are upregulated in dermal
anti-TNF therapy ameliorates this risk in patients with
blood vessels in involved skin from patients with pso-
rheumatoid arthritis [134].
riasis [117,118].
Randomized trials showed that infliximab resulted
in a rapid and significant improvement in psoriatic Respiratory disease
plaques [119,120]. Up to one-third of patients with TNF has been implicated in the pathophysiology of
psoriasis develop an inflammatory arthritis, which many inflammatory lung diseases, including chronic
can affect both spinal and peripheral joints. Early bronchitis, chronic obstructive pulmonary disease,
studies suggested a role for etanercept in the treatment acute respiratory distress syndrome and asthma [135].
of psoriatic arthritis, and subsequent studies have In asthma, TNF has been implicated in airway
shown that infliximab, etanercept and adalimumab are inflammation and remodelling, and may play a role
all effective treatments for the dermatological and in bronchial hyper-responsiveness. Leukocytes from
articular manifestations of psoriasis [121–124]. bronchiolar lavage of asthma patients have increased
release of TNF [136], and inhaled TNF increases air-
Disease of the central nervous system way responsiveness in normal subjects and is associ-
ated with a pulmonary neutrophil infiltration, assessed
In the central nervous system, TNF is produced pri- by induced sputum. To date there are no random-
marily by microglia and astrocytes in response to ized controlled trials of TNF blockade in asthma,
a wide range of pathological processes, including but patients with asthma who received infliximab for
infection, inflammatory disease, ischaemia and trau- rheumatoid arthritis have demonstrated a significant
matic injury [125]. However, TNF has been shown improvement, and an open label uncontrolled study of
to have both harmful and beneficial effects in the etanercept in 17 subjects with severe asthma demon-
injured brain [126]. Inhibition of TNF ameliorates strated an improvement in asthma symptoms, lung
ischaemic brain injury in mice [127], whereas mice function and bronchial hyper-responsiveness follow-
lacking TNF are highly susceptible to experimen- ing 12 weeks of entanercept [137]. A pilot study of
tal autoimmune encephalomyelitis, and treatment with patients with refractory asthma provided evidence for
TNF dramatically reduces disease severity [128]. TNF- a role of TNF, and demonstrated a beneficial effects
mediated protection against experimental autoimmune of etanercept on markers of asthma control [138].
encephalomyelitis does not require TNFR1, although
TNFR1 appears to be necessary for detrimental effects Renal disease
of TNF, which occur during the acute phase of the
disease [129]. In contrast, TNFR2 has been shown to TNF has been implicated in the pathogenesis of many
promote proliferation of oligodendrocyte progenitors renal diseases, including ischaemic renal injury, renal
and remyelination in a neurotoxicant murine model of transplant rejection and glomerulonephritis, which is
demyelination [130]. Neutralization of TNF failed to often part of a systemic vasculitis. In diseases associ-
benefit patients with relapsing–remitting multiple scle- ated with renal inflammation, different forms of TNF
rosis, and significantly increased exacerbations [131]. blockade vary in their efficacy and adverse effects,
and these differences may be attributed to different
effects on signalling though TNF receptor subtypes. In
Cardiovascular disease
acute renal injury, TNFR2-mediated cell proliferation
TNF has also been implicated in the pathogenesis may be important for tubular cell regeneration [38],
of a number of cardiovascular diseases, including whereas in proliferative forms of glomerulonephri-
atherosclerosis, myocardial infarction, heart failure, tis, TNFR1-mediated cytotoxicity and inhibition of
myocarditis and cardiac allograft rejection, and vas- TNFR2-mediated cellular proliferation may be more
cular endothelial cell responses to TNF may underlie desirable.
the vascular pathology in many of these conditions. TNFR2 is essential for the development of renal
However, clinical trials have demonstrated no clinical injury in a model of immune complex glomeru-
benefit of TNF blockade in congestive cardiac failure. lonephritis induced by anti-GBM antibody, raising the
This may be because TNFR1 and TNFR2 differen- possibility that selective blockade of TNFR2 may be a
tially regulate cardiac responses to TNF. In transgenic promising strategy for treatment of immune-mediated
mice with TNF-induced cardiomyopathy, ablation of glomerulonephritis [139].
the TNFR2 gene exacerbates heart failure and reduces As discussed above, infliximab may be more effec-
survival, whereas ablation of TNFR1 blunts heart fail- tive at blocking signalling through TNFR2 than etan-
ure and improves survival [132]. In cardiac allografts ercept. In support of this, infliximab is an effective
either TNF receptor is capable of mediating a response treatment for patients with refractory Wegener’s gran-
that will culminate in graft arterial disease [133]. ulomatosis [140], in which lymphocyte activation and

J Pathol 2008; 214: 149–160 DOI: 10.1002/path


Copyright  2007 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
156 JR Bradley

glomerular cell proliferation are important in patho- 3. Black RA, Rauch CT, Kozlosky CJ, Peschon JJ, Slack
genesis. In contrast, etanercept is not effective for the JL, Wolfson MF, et al. A metalloproteinase disintegrin
that releases tumour-necrosis factor-alpha from cells. Nature
maintenance of remission in patients with Wegener’s 1997;385(6618):729–733.
granulomatosis, and its use is associated with an 4. Wang J, Al-Lamki RS, Zhang H, Kirkiles-Smith N, Gaeta ML,
increased incidence of solid tumours [141]. Thiru S, et al. Histamine antagonizes tumor necrosis factor
(TNF) signalling by stimulating TNF receptor shedding from
the cell surface and Golgi storage pool. J Biol Chem
Other inflammatory diseases 2003;278(24):21751–21760.
As the potential role of TNF blockade has become 5. Robak T, Gladalska A, Stepien H. The tumour necrosis factor
family of receptors/ligands in the serum of patients with
clear, its successful use has been reported in an
rheumatoid arthritis. Eur Cytokine Netw 1998;9(2):145–154.
increasing number of inflammatory conditions. These 6. Nurnberger W, Platonov A, Stannigel H, Beloborodov VB,
include juvenile rheumatoid arthritis [142]; therapy- Michelmann I, von Kries R, et al. Definition of a new score for
resistant sarcoidosis, a multisystemic disorder charac- severity of generalized Neisseria meningitidis infection. Eur J
terized histologically by the presence of granuloma- Pediatr 1995;154(11):896–900.
tous inflammation [143–145]; inflammatory myopa- 7. Waage A, Halstensen A, Espevik T. Association between tumour
necrosis factor in serum and fatal outcome in patients with
thies [146]; Behcet disease [147]; and inflammatory
meningococcal disease. Lancet 1987;1(8529):355–357.
eye disease [148]. 8. Kwiatkowski D, Hill AV, Sambou I, Twumasi P, Castracane J,
Manogue KR, et al. TNF concentration in fatal cerebral, non-fatal
cerebral, and uncomplicated Plasmodium falciparum malaria.
Summary Lancet 1990;336(8725):1201–1204.
9. Bouwmeester T, Bauch A, Ruffner H, Angrand PO, Bergamini
G, Croughton K, et al. A physical and functional map of the
Although the capacity of bacterial toxins to induce
human TNFα/NF-κB signal transduction pathway. Nat Cell Biol
tumour regression was first described at the end of 2004;6(2):97–105.
the nineteenth century, TNF was not identified as the 10. Al-Lamki RS, Wang J, Skepper JN, Thiru S, Pober JS,
tumouricidal agent until 1975. TNF was cloned in Bradley JR. Expression of tumor necrosis factor receptors
1984, and the last two decades have seen how molec- in normal kidney and rejecting renal transplants. Lab Invest
ular and cellular biological techniques, combined with 2001;81(11):1503–1515.
11. Ledgerwood EC, Pober JS, Bradley JR. Recent advances in
in vivo studies, have unravelled the complexity of
the molecular basis of TNF signal transduction. Lab Invest
TNF and its signalling pathways and the pathophys- 1999;79(9):1041–1050.
iological responses it triggers. This has led to an 12. Jones SJ, Ledgerwood EC, Prins JB, Galbraith J, Johnson DR,
understanding of its key role in inflammatory disease Pober JS, et al. TNF recruits TRADD to the plasma membrane
and perhaps the most remarkable example of transla- but not the trans-Golgi network, the principal subcellular location
tional medicine, as scientists have collaborated with of TNF-R1. J Immunol 1999;162(2):1042–1048.
13. Bennett M, Macdonald K, Chan SW, Luzio JP, Simari R,
clinicians and the pharmaceutical industry to develop
Weissberg P. Cell surface trafficking of Fas: a rapid mechanism
improved anti-inflammatory therapies. In turn, clinical of p53-mediated apoptosis. Science 1998;282(5387):290–293.
studies are providing valuable insights into the mech- 14. Chan FK, Chun HJ, Zheng L, Siegel RM, Bui KL,
anism of action of TNF, allowing the science to return Lenardo MJ. A domain in TNF receptors that mediates
to the laboratory to continue to address the many unan- ligand-independent receptor assembly and signalling. Science
swered questions. 2000;288(5475):2351–2354.
15. Jiang Y, Woronicz JD, Liu W, Goeddel DV. Prevention of
constitutive TNF receptor 1 signalling by silencer of death
Acknowledgement domains. Science 1999;283:543–546.
16. Endres R, Hacker G, Brosch I, Pfeffer K. Apparently normal
JRB is supported by the NIHR Cambridge Biomedical Research
tumor necrosis factor receptor 1 signalling in the absence of the
Centre, UK. silencer of death domains. Mol Cell Biol 2003;23(18):6609–6617.
17. Hsu H, Huang J, Shu HB, Baichwal V, Goeddel DV. TNF-
dependent recruitment of the protein kinase RIP to the TNF
Teaching materials receptor-1 signalling complex. Immunity 1996;4:387–396.
18. Takeuchi M, Rothe M, Goeddel DV. Anatomy of TRAF2.
Power Point slides of the figures from this Review may Distinct domains for nuclear factor-κB activation and association
with tumor necrosis factor signalling proteins. J Biol Chem
be found at the web address http://www.interscience. 1996;271:19935–19942.
wiley.com/jpages/0022-3417/suppmat/path.2287.html 19. Bradley JR, Johnson DR, Pober JS. Four different classes of
inhibitors of receptor-mediated endocytosis decrease TNF-
induced gene expression in human endothelial cells. J Immunol
References 1993;150:5544–5555.
20. Schutze S, Machleidt T, Adam D, Schwandner R, Wiegmann K,
1. Carswell EA, Old LJ, Kassel RL, Green S, Fiore N, Williamson Kruse ML, et al. Inhibition of receptor internalization by
B. An endotoxin-induced serum factor that causes necrosis of monodansylcadaverine selectively blocks p55 tumor necrosis
tumors. Proc Natl Acad Sci USA 1975;72(9):3666–3670. factor receptor death domain signalling. J Biol Chem
2. Pennica D, Hayflick JS, Bringman TS, Palladino MA, Goed- 1999;274:10203–10212.
del DV. Cloning and expression in Escherichia coli of the 21. Yang J, Lin Y, Guo Z, Cheng J, Huang J, Deng L, et al. The
cDNA for murine tumor necrosis factor. Proc Natl Acad Sci USA essential role of MEKK3 in TNF-induced NF-κB activation. Nat
1985;82(18):6060–6064. Immunol 2001;2:620–624.

J Pathol 2008; 214: 149–160 DOI: 10.1002/path


Copyright  2007 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
TNF-mediated inflammatory disease 157

22. Blonska M, Shambharkar PB, Kobayashi M, Zhang D, Saku- 41. Pinckard JK, Sheehan KC, Schreiber RD. Ligand-induced forma-
rai H, Su B, et al. TAK1 is recruited to the tumor necrosis factor- tion of p55 and p75 tumor necrosis factor receptor heterocom-
alpha (TNFα) receptor 1 complex in a receptor-interacting protein plexes on intact cells. J Biol Chem 1997;272(16):10784–10789.
(RIP)-dependent manner and cooperates with MEKK3, leading to 42. Santee SM, Owen-Schaub LB. Human tumor necrosis factor
NF-κB activation. J Biol Chem 2005;280:43056–43063. receptor p75/80 (CD120b) gene structure and promoter
23. Devin A, Lin Y, Yamaoka S, Li Z, Karin M, Liu Z. The α- and characterization. J Biol Chem 1996;271(35):21151–21159.
β-subunits of IκB kinase (IKK) mediate TRAF2-dependent IKK 43. Kalthoff H, Roeder C, Brockhaus M, Thiele HG, Schmiegel W.
recruitment to tumor necrosis factor (TNF) receptor 1 in response Tumor necrosis factor (TNF) up-regulates the expression of
to TNF. Mol Cell Biol 2001;21:3986–3994. p75 but not p55 TNF receptors, and both receptors mediate,
24. Chen ZJ. Ubiquitin signalling in the NF-κB pathway. Nat Cell independently of each other, up-regulation of transforming
Biol 2005;7:758–765. growth factor alpha and epidermal growth factor receptor mRNA.
25. Nishitoh H, Saitoh M, Mochida Y, Takeda K, Nakano H, J Biol Chem 1993;268(4):2762–2766.
Rothe M, et al. ASK1 is essential for JNK/SAPK activation by 44. Winzen R, Wallach D, Engelmann H, Nophar Y, Brakebusch C,
TRAF2. Mol Cell 1998;2(3):389–395. Kemper O, et al. Selective decrease in cell surface expression
26. Ichijo H, Nishida E, Irie K, ten Dijke P, Saitoh M, Moriguchi T, and mRNA level of the 55-kDa tumor necrosis factor receptor
et al. Induction of apoptosis by ASK1, a mammalian MAPKKK during differentiation of HL-60 cells into macrophage-like but
that activates SAPK/JNK and p38 signalling pathways. Science not granulocyte-like cells. J Immunol 1992;148(11):3454–3460.
1997;275:90–94. 45. Winzen R, Wallach D, Kemper O, Resch K, Holtmann H.
27. Nishitoh H, Saitoh M, Mochida Y, Takeda K, Nakano H, Selective up-regulation of the 75-kDa tumor necrosis factor
Rothe M, et al. ASK1 is essential for JNK/SAPK activation by (TNF) receptor and its mRNA by TNF and IL-1. J Immunol
TRAF2. Mol Cell 1998;2:389–395. 1993;150(10):4346–4353.
28. Adam-Klages S, Adam D, Wiegmann K, Struve S, Kolanus W, 46. Kemper O, Wallach D. Cloning and partial characterization of
Schneider-Mergener J, et al. FAN, a novel WD-repeat protein, the promoter for the human p55 tumor necrosis factor (TNF)
couples the p55 TNF-receptor to neutral sphingomyelinase. Cell receptor. Gene 1993;134(2):209–216.
1996;86:937–947. 47. Rothe J, Bluethmann H, Gentz R, Lesslauer W, Steinmetz M.
29. Slowik MR, De Luca LG, Min W, Pober JS. Ceramide is not a Genomic organization and promoter function of the murine
signal for tumor necrosis factor-induced gene expression but does tumor necrosis factor receptor beta gene. Mol Immunol
cause programmed cell death in human vascular endothelial cells. 1993;30(2):165–175.
Circ Res 1996;79:736–747. 48. Nophar Y, Kemper O, Brakebusch C, Englemann H, Zwang R,
30. Micheau O, Lens S, Gaide O, Alevizopoulos K, Tschopp J. NF- Aderka D, et al. Soluble forms of tumor necrosis factor receptors
κB signals induce the expression of c-FLIP. Mol Cell Biol (TNF-Rs). The cDNA for the type I TNF-R, cloned using
2001;21:5299–5305. amino acid sequence data of its soluble form, encodes both
31. Xu XS, Vanderziel C, Bennett CF, Monia BP. A role for c- the cell surface and a soluble form of the receptor. EMBO J
Raf kinase and Ha-Ras in cytokine-mediated induction of cell 1990;9(10):3269–3278.
adhesion molecules. J Biol Chem 1998;273:33230–343238. 49. Madge LA, Sierra-Honigmann MR, Pober JS. Apoptosis-
32. Jacinto E, Facchinetti V, Liu D, Soto N, Wei S, Jung SY, et al. inducing agents cause rapid shedding of tumor necrosis
SIN1/MIP1 Maintains rictor-mTOR complex integrity and factor receptor 1 (TNFR1). A nonpharmacological explanation
regulates Akt phosphorylation and substrate specificity. Cell for inhibition of TNF-mediated activation. J Biol Chem
2006;127:125–137. 1999;274(19):13643–13649.
33. Robinson MJ, Cobb MH. Mitogen-activated protein kinase 50. Bradley JR, Johnson DR, Pober JS. Endothelial activation by
pathways. Curr Opin Cell Biol 1997;9:180–186. hydrogen peroxide. Selective increases of intercellular adhesion
34. Rothe M, Wong SC, Henzel WJ, Goeddel DV. A novel family molecule-1 and major histocompatibility complex class I. Am J
of putative signal transducers associated with the cytoplasmic Pathol 1993;142(5):1598–1609.
domain of the 75 kDa tumor necrosis factor receptor. Cell 51. Hino T, Nakamura H, Abe S, Saito H, Inage M, Terashita K,
1994;78(4):681–692. et al. Hydrogen peroxide enhances shedding of type I soluble
35. Grech AP, Gardam S, Chan T, Quinn R, Gonzales R, Basten A, tumor necrosis factor receptor from pulmonary epithelial cells.
et al. Tumor necrosis factor receptor 2 (TNFR2) signalling Am J Resp Cell Mol Biol 1999;20(1):122–128.
is negatively regulated by a novel, carboxyl-terminal TNFR- 52. McDermott MF, Aksentijevich I, Galon J, McDermott EM,
associated factor 2 (TRAF2)-binding site. J Biol Chem Ogunkolade BW, Centola M, et al. Germline mutations in the
2005;280(36):31572–31581. extracellular domains of the 55 kDa TNF receptor, TNFR1,
36. Zhao Y, Conze DB, Hanover JA, Ashwell JD. Tumor necrosis define a family of dominantly inherited autoinflammatory
factor receptor 2 signalling induces selective c-IAP1-dependent syndromes. Cell 1999;97(1):133–144.
ASK1 ubiquitination and terminates mitogen-activated protein 53. Lobito AA, Kimberley FC, Muppidi JR, Komarow H, Jack-
kinase signalling. J Biol Chem 2007;282(11):7777–7782. son AJ, Hull KM, et al. Abnormal disulfide-linked oligomeriza-
37. Zhang R, Xu Y, Ekman N, Wu Z, Wu J, Alitalo K, et al. tion results in ER retention and altered signalling by TNFR1
Etk/Bmx transactivates vascular endothelial growth factor 2 mutants in TNFR1-associated periodic fever syndrome (TRAPS).
and recruits phosphatidylinositol 3-kinase to mediate the tumor Blood 2006;108(4):1320–1327.
necrosis factor-induced angiogenic pathway. J Biol Chem 54. Messadi DV, Pober JS, Fiers W, Gimbrone MA Jr, Murphy GF.
2003;278(51):51267–51276. Induction of an activation antigen on postcapillary venular
38. Al-Lamki RS, Wang J, Vandenabeele P, Bradley JA, Thiru S, endothelium in human skin organ culture. J Immunol
Luo D, et al. TNFR1- and TNFR2-mediated signalling pathways 1987;139:1557–1562.
in human kidney are cell type-specific and differentially 55. Petzelbauer P, Pober JS, Keh A, Braverman IM. Inducibility and
contribute to renal injury. FASEB J 2005;19(12):1637–1645. expression of microvascular endothelial adhesion molecules in
39. Slowik MR, De Luca LG, Fiers W, Pober JS. Tumor necrosis lesional, perilesional, and uninvolved skin of psoriatic patients. J
factor activates human endothelial cells through the p55 tumor Invest Dermatol 1994;103:300–305.
necrosis factor receptor but the p75 receptor contributes to 56. Bradley JR, Pober JS. Prolonged cytokine exposure causes a
activation at low tumor necrosis factor concentration. Am J Pathol dynamic redistribution of endothelial cell adhesion molecules to
1993;143(6):1724–1730. intercellular junctions. Lab Invest 1996;75(4):463–472.
40. Tartaglia LA, Pennica D, Goeddel DV. Ligand passing: the 57. Pober JS, Bevilacqua MP, Mendrick DL, Lapierre LA, Fiers W,
75-kDa tumor necrosis factor (TNF) receptor recruits TNF Gimbrone MA Jr. Two distinct monokines, interleukin 1 and
for signalling by the 55-kDa TNF receptor. J Biol Chem tumor necrosis factor, each independently induce biosynthesis
1993;268(25):18542–18548. and transient expression of the same antigen on the surface

J Pathol 2008; 214: 149–160 DOI: 10.1002/path


Copyright  2007 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
158 JR Bradley

of cultured human vascular endothelial cells. J Immunol 75. Creagan ET, Kovach JS, Moertel CG, Frytak S, Kvols LK. A
1986;136:1680–1687. phase I clinical trial of recombinant human tumor necrosis factor.
58. Munro JM, Pober JS, Cotran RS. Tumor necrosis factor and Cancer 1988;62(12):2467–2471.
interferon-g induce distinct patterns of endothelial activation and 76. Feinberg B, Kurzrock R, Talpaz M, Blick M, Saks S, Gutter-
associated leukocyte accumulation in skin of Papio anubis. Am man JU. A phase I trial of intravenously-administered recombi-
J Pathol 1989;135:121–133. nant tumor necrosis factor-alpha in cancer patients. J Clin Oncol
59. Rollins BJ, Yoshimura T, Leonard EJ, Pober JS. Cytokine- 1988;6(8):1328–1334.
activated human endothelial cells synthesize and secrete 77. Moore RJ, Owens DM, Stamp G, Arnott C, Burke F, East N,
a monocyte chemoattractant, MCP-1/JE. Am J Pathol et al. Mice deficient in tumor necrosis factor-alpha are resistant
1990;136:1229–1233. to skin carcinogenesis. Nat Med 1999;5(7):828–831.
60. Mark KS, Trickler WJ, Miller DW. Tumor necrosis factor-alpha 78. Grunhagen DJ, de Wilt JH, Graveland WJ, van Geel AN,
induces cyclooxygenase-2 expression and prostaglandin release Eggermont AM. The palliative value of tumor necrosis factor
in brain microvessel endothelial cells. J Pharmacol Exp Therap alpha-based isolated limb perfusion in patients with metastatic
2001;297(3):1051–1058. sarcoma and melanoma. Cancer 2006;106(1):156–162.
79. Grunhagen DJ, van Etten B, Brunstein F, Graveland WJ, van
61. Bevilacqua MP, Pober JS, Majeau GR, Fiers W, Cotran RS,
Geel AN, de Wilt JH, et al. Efficacy of repeat isolated limb
Gimbrone MA Jr. Recombinant tumor necrosis factor induces
perfusions with tumor necrosis factor alpha and melphalan for
procoagulant activity in cultured human vascular endothelium:
multiple in-transit metastases in patients with prior isolated limb
characterization and comparison with the actions of interleukin
perfusion failure. Ann Surg Oncol 2005;12(8):609–615.
1. Proc Natl Acad Sci USA 1986;83(12):4533–4537.
80. van Etten B, de Vries MR, van IMG, Lans TE, Guetens G,
62. Cerami A, Ikeda Y, Le Trang N, Hotez PJ, Beutler B. Weight
Ambagtsheer G, et al. Degree of tumour vascularity correlates
loss associated with an endotoxin-induced mediator from
with drug accumulation and tumour response upon TNFα-based
peritoneal macrophages: the role of cachectin (tumor necrosis isolated hepatic perfusion. Br J Cancer 2003;88(2):314–319.
factor). Immunol Lett 1985;11(3–4):173–177. 81. Scallon B, Cai A, Solowski N, Rosenberg A, Song XY, Shealy
63. Beutler BA, Milsark IW, Cerami A. Cachectin/tumor necrosis D, et al. Binding and functional comparisons of two types
factor: production, distribution, and metabolic fate in vivo. J of tumor necrosis factor antagonists. J Pharmacol Exp Therap
Immunol 1985;135(6):3972–3977. 2002;301(2):418–426.
64. Tracey KJ, Fong Y, Hesse DG, Manogue KR, Lee AT, 82. Grell M, Douni E, Wajant H, Lohden M, Clauss M, Maxeiner B,
Kuo GC, et al. Anti-cachectin/TNF monoclonal antibod- et al. The transmembrane form of tumor necrosis factor is the
ies prevent septic shock during lethal bacteraemia. Nature prime activating ligand of the 80 kDa tumor necrosis factor
1987;330(6149):662–664. receptor. Cell 1995;83(5):793–802.
65. Fisher CJ Jr, Agosti JM, Opal SM, Lowry SF, Balk RA, Sad- 83. Koch AE, Harlow LA, Haines GK, Amento EP, Unemori EN,
off JC, et al. Treatment of septic shock with the tumor necrosis Wong WL, et al. Vascular endothelial growth factor. A cytokine
factor receptor: Fc fusion protein. The Soluble TNF Receptor modulating endothelial function in rheumatoid arthritis. J
Sepsis Study Group. N Engl J Med 1996;334(26):1697–1702. Immunol 1994;152(8):4149–4156.
66. Pfeffer K, Matsuyama T, Kundig TM, Wakeham A, Kishihara K, 84. Feldmann M, Brennan FM, Maini RN. Role of cytokines in
Shahinian A, et al. Mice deficient for the 55 kDa tumor necrosis rheumatoid arthritis. Annu Rev Immunol 1996;14:397–440.
factor receptor are resistant to endotoxic shock, yet succumb to 85. Brennan FM, Chantry D, Jackson A, Maini R, Feldmann
L. monocytogenes infection. Cell 1993;73(3):457–467. M. Inhibitory effect of TNFα antibodies on synovial
67. Rothe J, Lesslauer W, Lotscher H, Lang Y, Koebel P, Kont- cell interleukin-1 production in rheumatoid arthritis. Lancet
gen F, et al. Mice lacking the tumour necrosis factor recep- 1989;2(8657):244–247.
tor 1 are resistant to TNF-mediated toxicity but highly 86. Butler DM, Maini RN, Feldmann M, Brennan FM. Modulation
susceptible to infection by Listeria monocytogenes. Nature of proinflammatory cytokine release in rheumatoid synovial
1993;364(6440):798–802. membrane cell cultures. Comparison of monoclonal anti TNFα
68. Peschon JJ, Torrance DS, Stocking KL, Glaccum MB, Otten C, antibody with the interleukin-1 receptor antagonist. Eur Cytokine
Willis CR, et al. TNF receptor-deficient mice reveal divergent Netw 1995;6(4):225–230.
roles for p55 and p75 in several models of inflammation. J 87. Williams RO, Feldmann M, Maini RN. Anti-tumor necrosis
Immunol 1998;160(2):943–952. factor ameliorates joint disease in murine collagen-induced
69. Roach DR, Bean AG, Demangel C, France MP, Briscoe H, arthritis. Proc Natl Acad Sci USA 1992;89(20):9784–9788.
Britton WJ. TNF regulates chemokine induction essential 88. Thorbecke GJ, Shah R, Leu CH, Kuruvilla AP, Hardison AM,
Palladino MA. Involvement of endogenous tumor necrosis factor
for cell recruitment, granuloma formation, and clearance of
alpha and transforming growth factor beta during induction of
mycobacterial infection. J Immunol 2002;168(9):4620–4627.
collagen type II arthritis in mice. Proc Natl Acad Sci USA
70. Wallis RS, Broder M, Wong J, Lee A, Hoq L. Reactivation of
1992;89(16):7375–7379.
latent granulomatous infections by infliximab. Clin Infect Dis
89. Piguet PF, Grau GE, Vesin C, Loetscher H, Gentz R, Less-
2005;41(suppl 3):S194–198.
lauer W. Evolution of collagen arthritis in mice is arrested
71. Rudin W, Eugster HP, Bordmann G, Bonato J, Muller M,
by treatment with anti-tumour necrosis factor (TNF) anti-
Yamage M, et al. Resistance to cerebral malaria in tumor necrosis body or a recombinant soluble TNF receptor. Immunology
factor-alpha/beta-deficient mice is associated with a reduction of 1992;77(4):510–514.
intercellular adhesion molecule-1 up-regulation and T helper type 90. Kontoyiannis D, Pasparakis M, Pizarro TT, Cominelli F, Kol-
1 response. Am J Pathol 1997;150(1):257–266. lias G. Impaired on/off regulation of TNF biosynthesis in mice
72. Kwiatkowski D, Molyneux ME, Stephens S, Curtis N, Klein N, lacking TNF AU-rich elements: implications for joint and gut-
Pointaire P, et al. Anti-TNF therapy inhibits fever in cerebral associated immunopathologies. Immunity 1999;10(3):387–398.
malaria. Qu J Med 1993;86(2):91–98. 91. Elliott MJ, Maini RN, Feldmann M, Long-Fox A, Charles P,
73. van Hensbroek MB, Palmer A, Onyiorah E, Schneider G, Katsikis P, et al. Treatment of rheumatoid arthritis with chimeric
Jaffar S, Dolan G, et al. The effect of a monoclonal antibody monoclonal antibodies to tumor necrosis factor alpha. Arthrit
to tumor necrosis factor on survival from childhood cerebral Rheumat 1993;36(12):1681–1690.
malaria. J Infect Dis 1996;174(5):1091–1097. 92. Elliott MJ, Maini RN, Feldmann M, Kalden JR, Antoni C,
74. Smith CA, Davis T, Anderson D, Solam L, Beckmann MP, Smolen JS, et al. Randomized double-blind comparison of
Jerzy R, et al. A receptor for tumor necrosis factor defines chimeric monoclonal antibody to tumour necrosis factor
an unusual family of cellular and viral proteins. Science alpha (cA2) versus placebo in rheumatoid arthritis. Lancet
1990;248(4958):1019–1023. 1994;344(8930):1105–1110.

J Pathol 2008; 214: 149–160 DOI: 10.1002/path


Copyright  2007 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
TNF-mediated inflammatory disease 159

93. Maini RN, Breedveld FC, Kalden JR, Smolen JS, Davis D, Mac- 109. Francois RJ, Neure L, Sieper J, Braun J. Immunohistological
farlane JD, et al. Therapeutic efficacy of multiple intravenous examination of open sacroiliac biopsies of patients with
infusions of anti-tumor necrosis factor alpha monoclonal anti- ankylosing spondylitis: detection of tumour necrosis factor alpha
body combined with low-dose weekly methotrexate in rheuma- in two patients with early disease and transforming growth
toid arthritis. Arthrit Rheumat 1998;41(9):1552–1563. factor beta in three more advanced cases. Ann Rheumat Dis
94. Moreland LW. Clinical and pharmacological experience with 2006;65(6):713–720.
etanercept. Exp Opin Invest Drugs 1999;8(9):1443–1451. 110. Lange U, Teichmann J, Stracke H. Correlation between plasma
95. Lipsky PE, van der Heijde DM, St Clair EW, Furst DE, TNFα, IGF-1, biochemical markers of bone metabolism, markers
Breedveld FC, Kalden JR, et al. Infliximab and methotrexate in of inflammation/disease activity, and clinical manifestations in
the treatment of rheumatoid arthritis. Anti-Tumor Necrosis Factor ankylosing spondylitis. Eur J Med Res 2000;5(12):507–511.
Trial in Rheumatoid Arthritis with Concomitant Therapy Study 111. Gorman JD, Sack KE, Davis JC Jr. Treatment of ankylosing
Group. N Engl J Med 2000;343(22):1594–1602. spondylitis by inhibition of tumor necrosis factor alpha. N Engl
96. van de Putte LB, Atkins C, Malaise M, Sany J, Russell AS, van J Med 2002;346(18):1349–1356.
Riel PL, et al. Efficacy and safety of adalimumab as monotherapy 112. Baraliakos X, Listing J, Rudwaleit M, Brandt J, Sieper J,
in patients with rheumatoid arthritis for whom previous disease Braun J. Radiographic progression in patients with ankylosing
modifying antirheumatic drug treatment has failed. Ann Rheumat spondylitis after 2 years of treatment with the tumour
Dis 2004;63(5):508–516. necrosis factor alpha antibody infliximab. Ann Rheumat Dis
97. Keystone EC, Kavanaugh AF, Sharp JT, Tannenbaum H, Hua Y, 2005;64(10):1462–1466.
Teoh LS, et al. Radiographic, clinical, and functional outcomes 113. Davis JC, van der Heijde DM, Braun J, Dougados M, Cush J,
of treatment with adalimumab (a human anti-tumor necrosis Clegg D, et al. Sustained durability and tolerability of etanercept
factor monoclonal antibody) in patients with active rheuma- in ankylosing spondylitis for 96 weeks. Ann Rheumat Dis
toid arthritis receiving concomitant methotrexate therapy: a 2005;64(11):1557–1562.
randomized, placebo-controlled, 52-week trial. Arthrit Rheumat 114. Inman RD, Clegg DO, Davis JC, Whitmore JB, Solinger A.
2004;50(5):1400–1411. Etanercept in adult patients with early onset ankylosing
98. Klareskog L, van der Heijde D, de Jager JP, Gough A, Kalden J, spondylitis. J Rheumatol 2006;33(8):1634–1636.
Malaise M, et al. Therapeutic effect of the combination of 115. Braun J, Brandt J, Listing J, Zink A, Alten R, Golder W,
etanercept and methotrexate compared with each treatment alone et al. Treatment of active ankylosing spondylitis with inflix-
in patients with rheumatoid arthritis: double-blind randomized imab: a randomized controlled multicentre trial. Lancet
controlled trial. Lancet 2004;363(9410):675–681. 2002;359(9313):1187–1193.
99. Buch MH, Bingham SJ, Bryer D, Emery P. Long-term inflix- 116. Baraliakos X, Listing J, Rudwaleit M, Brandt J, Alten R,
imab treatment in rheumatoid arthritis: subsequent outcome of Burmester G, et al. Safety and efficacy of readministration
initial responders. Rheumatology 2007;46(7):1153–1156. of infliximab after long-term continuous therapy and with-
100. Breese EJ, Michie CA, Nicholls SW, Murch SH, Williams CB, drawal in patients with ankylosing spondylitis. J Rheumatol
Domizio P, et al. Tumor necrosis factor alpha-producing cells 2007;34(3):510–515.
in the intestinal mucosa of children with inflammatory bowel 117. Kristensen M, Chu CQ, Eedy DJ, Feldmann M, Brennan FM,
disease. Gastroenterology 1994;106(6):1455–1466. Breathnach SM. Localization of tumour necrosis factor-alpha
101. Murch SH, Braegger CP, Walker-Smith JA, MacDonald TT. (TNFα) and its receptors in normal and psoriatic skin: epidermal
Location of tumour necrosis factor alpha by immunohis- cells express the 55 kDa but not the 75 kDa TNF receptor. Clin
tochemistry in chronic inflammatory bowel disease. Gut Exp Immunol 1993;94(2):354–362.
1993;34(12):1705–1709. 118. Ettehadi P, Greaves MW, Wallach D, Aderka D, Camp RD.
102. Hanauer SB, Feagan BG, Lichtenstein GR, Mayer LF, Elevated tumour necrosis factor-alpha (TNFα) biological activity
Schreiber S, Colombel JF, et al. Maintenance infliximab for in psoriatic skin lesions. Clin Exp Immunol 1994;96(1):146–151.
Crohn’s disease: the ACCENT I randomized trial. Lancet 119. Chaudhari U, Romano P, Mulcahy LD, Dooley LT, Baker DG,
2002;359(9317):1541–1549. Gottlieb AB. Efficacy and safety of infliximab monother-
103. Sands BE, Anderson FH, Bernstein CN, Chey WY, Feagan BG, apy for plaque-type psoriasis: a randomized trial. Lancet
Fedorak RN, et al. Infliximab maintenance therapy for fistulizing 2001;357(9271):1842–1847.
Crohn’s disease. N Engl J Med 2004;350(9):876–885. 120. Gottlieb AB, Evans R, Li S, Dooley LT, Guzzo CA, Baker D,
104. Sandborn WJ, Feagan BG, Hanauer SB, Present DH, Suther- et al. Infliximab induction therapy for patients with severe
land LR, Kamm MA, et al. An engineered human antibody plaque-type psoriasis: a randomized, double-blind, placebo-
to TNF (CDP571) for active Crohn’s disease: a random- controlled trial. J Am Acad Dermatol 2004;51(4):534–542.
ized double-blind placebo-controlled trial. Gastroenterology 121. Leonardi CL, Powers JL, Matheson RT, Goffe BS, Zitnik R,
2001;120(6):1330–1338. Wang A, et al. Etanercept as monotherapy in patients with
105. Sandborn WJ, Feagan BG, Radford-Smith G, Kovacs A, Enns R, psoriasis. N Engl J Med 2003;349(21):2014–2022.
Innes A, et al. CDP571, a humanized monoclonal antibody to 122. Antoni CE, Kavanaugh A, Kirkham B, Tutuncu Z, Burmester
tumour necrosis factor alpha, for moderate to severe Crohn’s GR, Schneider U, et al. Sustained benefits of infliximab
disease: a randomized, double blind, placebo controlled trial. Gut therapy for dermatologic and articular manifestations of
2004;53(10):1485–1493. psoriatic arthritis: results from the infliximab multinational
106. Akobeng AK, Zachos M. Tumor necrosis factor-alpha antibody psoriatic arthritis controlled trial (IMPACT). Arthrit Rheumat
for induction of remission in Crohn’s disease. Cochrane Database 2005;52(4):1227–1236.
Syst Rev http://mrw.interscience.wiley.com/cochrane/clsysrev/ 123. Genovese MC, Mease PJ, Thomson GT, Kivitz AJ, Perdok RJ,
articles/CD003574/frame.html. Weinberg MA, et al. Safety and efficacy of adalimumab in
107. Lawson MM, Thomas AG, Akobeng AK. Tumour necro- treatment of patients with psoriatic arthritis who had failed
sis factor alpha blocking agents for induction of remis- disease modifying antirheumatic drug therapy. J Rheumatol
sion in ulcerative colitis. Cochrane Database Syst Reviews 2007;34(5):1040–1050.
http://mrw.interscience.wiley.com/cochrane/clsysrev/articles/CD 124. Tyring S, Gordon KB, Poulin Y, Langley RG, Gottlieb AB,
005112/frame.html. Dunn M, et al. Long-term safety and efficacy of 50 mg of
108. Braun J, Bollow M, Neure L, Seipelt E, Seyrekbasan F, Herbst etanercept twice weekly in patients with psoriasis. Arch Dermatol
H, et al. Use of immunohistologic and in situ hybridization 2007;143(6):719–726.
techniques in the examination of sacroiliac joint biopsy specimens 125. Feuerstein GZ, Liu T, Barone FC. Cytokines, inflammation, and
from patients with ankylosing spondylitis. Arthrit Rheumat brain injury: role of tumor necrosis factor-alpha. Cerebrovasc
1995;38(4):499–505. Brain Metab Rev 1994;6(4):341–360.

J Pathol 2008; 214: 149–160 DOI: 10.1002/path


Copyright  2007 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
160 JR Bradley

126. Shohami E, Ginis I, Hallenbeck JM. Dual role of tumor necrosis 137. Howarth PH, Babu KS, Arshad HS, Lau L, Buckley M,
factor alpha in brain injury. Cytokine Growth Factor Rev McConnell W, et al. Tumour necrosis factor (TNFα) as a
1999;10(2):119–130. novel therapeutic target in symptomatic corticosteroid dependent
127. Nawashiro H, Martin D, Hallenbeck JM. Neuroprotective effects asthma. Thorax 2005;60(12):1012–1018.
of TNF binding protein in focal cerebral ischemia. Brain Res 138. Berry MA, Hargadon B, Shelley M, Parker D, Shaw DE,
1997;778(2):265–271. Green RH, et al. Evidence of a role of tumor necrosis factor alpha
128. Liu J, Marino MW, Wong G, Grail D, Dunn A, Bettadapura J, in refractory asthma. N Engl J Med 2006;354(7):697–708.
et al. TNF is a potent anti-inflammatory cytokine in autoimmune- 139. Vielhauer V, Stavrakis G, Mayadas TN. Renal cell-expressed
mediated demyelination. Nat Med 1998;4(1):78–83. TNF receptor 2, not receptor 1, is essential for the development
129. Kassiotis G, Kollias G. Uncoupling the proinflammatory from of glomerulonephritis. J Clin Invest 2005;115(5):1199–1209.
the immunosuppressive properties of tumor necrosis factor 140. Lamprecht P, Voswinkel J, Lilienthal T, Nolle B, Heller M,
(TNF) at the p55 TNF receptor level: implications for Gross WL, et al. Effectiveness of TNFα blockade with
pathogenesis and therapy of autoimmune demyelination. J Exp infliximab in refractory Wegener’s granulomatosis. Rheumatology
Med 2001;193(4):427–434. 2002;41(11):1303–1307.
130. Arnett HA, Mason J, Marino M, Suzuki K, Matsushima GK, 141. Etanercept plus standard therapy for Wegener’s granulomatosis.
Ting JP. TNFα promotes proliferation of oligodendrocyte pro- N Engl J Med 2005;352(4):351–361.
genitors and remyelination. Nat Neurosci 2001;4(11):1116–1122. 142. Gartlehner G, Hansen RA, Jonas BL, Thieda P, Lohr KN.
131. TNF neutralization in MS: results of a randomized, placebo- Biologics for the treatment of juvenile idiopathic arthritis: a
controlled multicenter study. The Lenercept Multiple Sclerosis systematic review and critical analysis of the evidence. Clin
Study Group and The University of British Columbia MS/MRI Rheumatol 2008;27:67–76.
Analysis Group. Neurology 1999;53(3):457–465. 143. Khanna D, Liebling MR, Louie JS. Etanercept amelio-
132. Higuchi Y, McTiernan CF, Frye CB, McGowan BS, Chan TO, rates sarcoidosis arthritis and skin disease. J Rheumatol
Feldman AM. Tumor necrosis factor receptors 1 and 2 2003;30(8):1864–1867.
differentially regulate survival, cardiac dysfunction, and 144. Ulbricht KU, Stoll M, Bierwirth J, Witte T, Schmidt RE. Suc-
remodeling in transgenic mice with tumor necrosis factor-alpha- cessful tumor necrosis factor alpha blockade treatment in therapy-
induced cardiomyopathy. Circulation 2004;109(15):1892–1897. resistant sarcoidosis. Arthrit Rheumat 2003;48(12):3542–3543.
133. Suzuki J, Cole SE, Batirel S, Kosuge H, Shimizu K, Isobe M, 145. Baughman RP, Drent M, Kavuru M, Judson MA, Costabel U,
et al. Tumor necrosis factor receptor-1 and -2 double deficiency du Bois R, et al. Infliximab therapy in patients with chronic
reduces graft arterial disease in murine cardiac allografts. Am J sarcoidosis and pulmonary involvement. Am J Resp Crit Care
Transpl 2003;3(8):968–976. Med 2006;174(7):795–802.
134. Wolfe F, Michaud K. Heart failure in rheumatoid arthritis: rates, 146. Efthimiou P. Tumor necrosis factor-alpha in inflammatory
predictors, and the effect of anti-tumor necrosis factor therapy. myopathies: pathophysiology and therapeutic implications. Semin
Am J Med 2004;116(5):305–311. Arthrit Rheumat 2006;36(3):168–172.
135. Mukhopadhyay S, Hoidal JR, Mukherjee TK. Role of TNFα in 147. Ribi C, Sztajzel R, Delavelle J, Chizzolini C. Efficacy of TNFα
pulmonary pathophysiology. Resp Res 2006;7:125. blockade in cyclophosphamide-resistant neuro-Behcet disease. J
136. Cembrzynska-Nowak M, Szklarz E, Inglot AD, Teodorczyk- Neurol Neurosurg Psychiat 2005;76(12):1733–1735.
Injeyan JA. Elevated release of tumor necrosis factor-alpha and 148. Sobrin L, Kim EC, Christen W, Papadaki T, Letko E, Foster CS.
interferon-gamma by bronchoalveolar leukocytes from patients Infliximab therapy for the treatment of refractory ocular
with bronchial asthma. Am Rev Resp Dis 1993;147(2):291–295. inflammatory disease. Arch Ophthalmol 2007;125(7):895–900.

J Pathol 2008; 214: 149–160 DOI: 10.1002/path


Copyright  2007 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.

You might also like