throbber
Review
`
`TRENDS in Molecular Medicine Vol.11 No.1 January 2005
`
`Dimethylfumarate for psoriasis: more
`than a dietary curiosity*
`
`Ulrich Mrowietz1 and Khusru Asadullah2
`
`1Department of Dermatology, University Clinic of Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
`2Schering AG, 13342 Berlin, Germany
`
`Swiss company Fumapharm (www.fumapharm.ch),
`became available [3]. Coating of the tablets enabled the
`liberation of FAEs in the small intestine and decreased the
`rate of adverse GI effects. A schedule of dosing was
`established on an empirical base, starting with the low-
`strength formulation for three weeks before the introduc-
`tion of high-strength tablets by increasing the number of
`tablets weekly. The maximum dose was defined as a total
`amount of 1.2 g FAE/day (two high-strength tablets three
`times a day), representing 720 mg dimethylfumarate
`(DMF). In 1994, this mixture of FAE was registered by
`the German drug administration (BfArM) as Fumadermw
`initial (low-strength tablets) and Fumadermw (high-
`strength tablets) for the systemic treatment of severe
`psoriasis.
`Since its official registration, Fumadermw has become
`the number one drug for the systemic therapy of psoriasis
`in Germany (approximately 66% of all prescriptions for
`systemic psoriasis therapy;
`‘EUROPSO patient survey
`
`(a)
`
`–
`
`–
`
`COO
`CH2
`CH2
`COO
`Succinate
`
`Succinatdehydrogenase
`
`FAD
`
`FADH 2
`
`–
`
`COO
`CH
`CH
`COO–
`Fumarate
`
`Fumarase
`
`OH
`
`H2O
`
`–
`
`COO
`CH
`CH2
`COO
`Malate
`
`–
`
`O
`
`R1
`
`O
`C7
`
`H
`
`C1
`
`C2
`
`H
`
`6
`C
`O
`
`R2
`
`O
`
`(b)
`
`CH3
`
`CH
`
`2
`
`O
`
`EHF
`
`O
`H
`C43
`C38
`
`H
`
`C3
`
`9
`
`OH
`
`42
`C
`O
`
`O
`
`CH3
`
`MHF
`O
`H
`C31
`C26
`
`H
`
`C2
`
`7
`
`OH
`
`30
`C
`O
`
`DMF
`O
`H
`C19
`C14
`
`O
`
`CH3
`
`H
`
`C1
`
`5
`
`18
`C
`O
`
`CH3
`
`O
`
`TRENDS in Molecular Medicine
`
`Figure 1. Fumarate and its esters. (a) Fumarate metabolism in the citric acid cycle.
`(b) The chemical structure of free fumaric acid (R1ZR2ZH) and the fumaric acid
`esters (R1ZR2ZAlkyl) dimethylfumarate (DMF), methylhydrogenfumarate (MHF)
`and ethylhydrogenfumarate (EHF).
`
`Fumaric acid esters (FAEs) have been used for the oral
`treatment of psoriasis since 1959 and have been
`registered for this indication in Germany since 1994.
`Dimethylfumarate (DMF) and its metabolite methyl-
`hydrogenfumarate (MHF) are the pharmacologically
`active compounds, with DMF being the main com-
`ponent of the marketed FAE-mixture. However, the
`mechanism of action of FAE is yet to be fully understood.
`It has been shown that DMF inhibits NFkB translocation,
`which leads to (i) the inhibition of pro-inflammatory
`cytokine production and adhesion molecule expression,
`(ii) the inhibition of dendritic cell differentiation and, at
`higher concentrations, (iii) the induction of apoptosis.
`Recent evidence also shows that these effects are
`mediated through the interference of the intracellular
`redox system by DMF. Here, the mode of action of FAE
`and its clinical use for psoriasis will be discussed.
`
`Introduction
`Fumaric acid is a simple-structured dicarbonic acid, which
`has an important role in the citric acid cycle in humans
`(Figure 1). Fumaric acid deficiencies, or other disorders
`related to a disturbed metabolism of this compound, are
`unknown as a cause of disease. Nevertheless, fumaric acid
`esters are the number one drug for the oral treatment of
`severe psoriasis in Germany.
`Fumaric acid is used as a nutritional additive in various
`forms in the food and farming industries, without
`untoward effects [1]. The clinical use of fumaric acid
`derivatives started in 1959 when the German chemist
`Schweckendiek, suffering from psoriasis, raised the
`hypothesis that the disease might be caused, at least in
`part, by disturbances in the citric acid cycle. He postulated
`that the exogenous supplementation of fumaric acid might
`reverse the pathological process. Schweckendiek tried to
`prove his hypothesis by treating himself with fumaric acid
`derivatives. Because of a potential irritant effect of free
`fumaric acid when taken orally, he used esters of fumaric
`acid. Gastrointestinal (GI) irritant effects, however, could
`not fully be circumvented. Taking a self-designed mixture
`of different fumaric acid esters (FAEs; see Glossary), his
`psoriasis cleared [2].
`To improve the pharmaceutical quality, a new product
`consisting of a defined mixture of FAEs, produced by the
`
`*Schering AG has no commercial interest in Fumadermw.
`Corresponding author: Mrowietz, U. (umrowietz@dermatology.uni-kiel.de).
`Available online 2 December 2004
`
`www.sciencedirect.com 1471-4914/$ - see front matter Q 2004 Elsevier Ltd. All rights reserved. doi:10.1016/j.molmed.2004.11.003
`
`Sawai (IPR2019-00789), Ex. 1019, p. 001
`
`

`

`44
`
`Review
`
`TRENDS in Molecular Medicine Vol.11 No.1 January 2005
`
`Glossary
`
`DMF: dimethylfumarate
`EHF: ethylhydrogenfumarate
`FAEs: fumaric acid esters
`MHF: methylhydrogenfumarate
`NFkB: nuclear factor kB
`
`2002’, www.europso.org). Clinical studies are limited in
`number; however, the reported efficacy in psoriasis is
`high, together with a favourable long-term safety profile
`(see below). The first data concerning combination treat-
`ment with FAEs were recently published which broadens
`the clinical usage in patients with severe psoriasis [4,5].
`Within the last few years, there has been increasing
`evidence that FAEs have potent immunomodulatory
`effects on various types of cells with importance not only
`for the psoriatic tissue reaction.
`
`Pathogenetic concepts of psoriasis
`Psoriasis is regarded as an immune disorder in which a
`putative antigen presented to T cells by antigen-presenting
`dendritic cells leads to the generation of specifically
`activated T cells [6]. According to the T-cell cytokine
`expression profile, psoriasis is classified as a Th1-type
`immune response.
`Because several other inflammatory diseases, such as
`rheumatoid arthritis, Crohn’s disease and multiple scler-
`osis, follow similar immunological pathways of T-cell
`activation, psoriasis can be regarded as a visible disease
`model. The crucial involvement of T-cells in the patho-
`genesis of psoriasis was highlighted by transplantation
`experiments of psoriasis skin onto severe combined
`immunodeficiency mice and investigations to analyse the
`T-cell receptor repertoire [7,8]. In addition to the inflam-
`matory component, the hyperproliferation of epidermal
`keratinocytes, together with a disturbed cellular differen-
`tiation (leading to the disease characteristic hyper-
`parakeratosis), seems to be dependent on a genetic
`susceptibility that is, among others, associated with the
`epidermal differentiation complex (psoriasis susceptibility
`locus; PSORS4) and corneodesmosin (PSORS1) [9].
`The mechanism of action of FAEs in psoriasis might,
`therefore, be of interest for future use in the treatment of
`diseases with a pathogenetic background similar to this
`chronic skin disorder. Here, a summary of the current
`knowledge about the clinical use of FAEs and its mode of
`action is given.
`
`Clinical use of FAE for the systemic treatment of
`psoriasis
`The consensus guidelines [3] recommend treating psoriasis
`patients with Fumadermw until there is an improvement of
`lesions or the patient is satisfied with the treatment
`outcome. A maintenance treatment to stabilize clinical
`improvement can be performed for up to two years. Clinical
`trials have shown a decrease of the psoriasis area and
`severity index (PASI) – a measure for psoriasis severity –
`between 50 and 80% after 12–16 weeks of therapy [10,11].
`Recently, Hoefnagel and colleagues [12] presented data
`about the long-term safety of Fumadermw therapy in
`
`www.sciencedirect.com
`
`patients treated continuously for up to 14 years. Notably,
`no increased risk for infections or malignancies could be
`found within this study population.
`In earlier clinical trials, the question of which of the
`different esters of fumaric acid is the active compound was
`addressed. In 1989, Nieboer et al. [13] published the
`results of an extensive study evaluating the clinical effects
`of different FAEs in various concentrations as mono-
`therapies and in combination in psoriasis patients. The
`data showed that ethylhydrogenfumarate (EHF) mono-
`therapy was not superior to placebo. However, by using
`DMF (240 mg/d), a 40% improvement was seen in the
`DMF-treated group after six weeks of therapy, whereas
`in the placebo-group, worsening of psoriasis was reported.
`It was concluded that the main active ingredient of
`Fumadermw is DMF.
`
`Adverse events related to FAE treatment
`The most common adverse event reported in all clinical
`trials with FAE is GI complaints. Clinical symptoms
`include diarrhoea (most common), stomach ache and
`cramps, increased frequency of stools, nausea and vomit-
`ing. GI-complaints are clearly related to the tablet
`formulation of FAE. With a newly developed microtablet
`formulation, the rate of GI complaints is decreased to
`below 6% (unpublished data*).
`Flush is another characteristic adverse event related to
`FAE-treatment. The symptoms are highly variable and
`are reported mainly as a transient feeling of warmth or
`heat normally lasting for minutes. Remarkably, these side
`effects frequently occur only during the initial phase of
`therapy. Other symptoms occurring as adverse events
`during FAE therapy are rare. There is no report about
`an increased frequency of infections, neither of bacterial
`(e.g. folliculitis) nor viral origin (e.g. herpes simplex,
`zoster or human papillomavirus-related infections, such
`as common warts, common cold or flu). There are no
`reports in the literature about the development of any
`form of malignancies (e.g. lymphoma, myeloproliferative
`disorders and solid tumours) during long-term treatment
`with FAE.
`FAE-induced leukocytopenia is frequently observed
`[10,14]. Severe lymphocytopenia might be a reason to
`terminate FAE-therapy. Although there are no published
`follow-up data in lymphopenic patients after FAE-with-
`drawal, there is a complete recovery up to baseline values
`within 6–12 weeks after termination of FAE-therapy
`(U. Mrowietz, unpublished data).
`A characteristic of FAE treatment is an increase in
`peripheral blood eosinophils [10,11]. Maximum eosinophil
`numbers are usually seen between week four and eight of
`therapy with a peak at week six. In individual patients, the
`percentage of eosinophils in the differential count can exceed
`25%. However, clinical symptoms or disorders related to
`eosinophilia have not been observed. When FAE-therapy is
`continued, eosinophil numbers normally decline and, in
`most patients, return to baseline levels [11].
`
`* Langner, A. et al. (2004) Results of a phase II study of a novel oral fumarate,
`BG-12, in the treatment of severe psoriasis. J. Eur. Acad. Dermatol. Venerol. 18, 798.
`Presented at the European Congress on Psoriasis, October 21–24 2004, Paris, France.
`
`Sawai (IPR2019-00789), Ex. 1019, p. 002
`
`

`

`Review
`
`TRENDS in Molecular Medicine Vol.11 No.1 January 2005
`
`45
`
`Pharmacokinetics of fumaric esters in humans
`There is little knowledge about the pharmacokinetics of
`FAEs in humans. FAEs are almost completely absorbed in
`the small intestine. The postulated main active ingredient of
`Fumadermw, DMF, is rapidly hydrolysed by ubiquitous
`esterases to methylhydrogenfumarate (MHF), its main
`metabolite. MHF is further metabolized into fumaric acid
`and finally into carbon dioxide and water. There is no evi-
`dence for an organ-specific toxicity due to drug metabolism.
`Metabolism is independent of cytochrome P-450-
`dependent pathways. This is of particular importance
`because psoriasis is frequently associated with other
`diseases, such as diabetes mellitus or coronary heart
`disease, requiring concomitant medications [15]. Drug
`interactions have not been reported.
`In a recent investigation in healthy subjects given
`one tablet containing 240 mg DMF and 95 mg calcium-
`monoethylfumarate, only MHF, the metabolite of DMF,
`could be detected in serum after a mean time of 120 min,
`when the tablet was taken under fasting conditions. DMF
`and free fumaric acid were below detection limit of the
`HPLC-system used [16].
`By using an intestinal tissue homogenate the half-life
`of DMF was less than 2 min. The enzymes responsible for
`DMF degradation are carboxylesterases rather than
`cholinesterases [17], but the significance of this finding
`is not yet clear.
`
`Mode of action
`FAEs can modulate the functions, including proliferation
`and mediator production and secretion, of several cell
`types. Can the various known effects of FAE be linked to a
`common mechanism of action?
`
`FAEs affect T-cell numbers
`Early studies clearly demonstrated that FAEs have
`profound effects on the T-cell system. In an open study
`using Fumadermw in 16 patients with severe psoriasis
`(PASIO20), flow-cytometric determination of leukocytes
`was performed weekly for 12 weeks [18]. Leukocytopenia
`was observed in 94% of the patients, with a mean
`reduction in leukocyte numbers of 26.6% (maximum
`C
`T cells decreased by 45.4%
`60%). The number of CD4
`C
`C
`T cells by 44.5%. In one patient, CD8
`cells
`and CD8
`decreased by 87%; however, clinical signs of immuno-
`suppression-like infections were not observed.
`In seven of 16 patients, absolute numbers of CD4
`T cells were w200/ml. There was no change in the
`CD4:CD8 ratio. HLA-DR-expression on T cells remained
`unaltered and there was no significant alteration in CD25
`expression (IL-2 receptor a-chain). The decrease in the
`number of peripheral T cells is associated with a reduction
`of the lesional T-cell infiltrate. An immunohistochemical
`study of 33 patients, from whom biopsies were taken
`before and after two, six and eight weeks of Fumadermw
`therapy, revealed a decrease in the epidermal inflamma-
`tory infiltrate of 74.4% after eight weeks of treatment,
`C
`T cells of 51.2% [19]. The
`with a decrease of CD4
`subepidermal
`infiltrate decreased by 52.1%, with a
`C
`cells of 84.4% after eight weeks.
`reduction in CD4
`C
`memory T cells slightly increased
`Interestingly, CD45RO
`
`C
`
`www.sciencedirect.com
`
`within the epidermis from 26.8% to 33%, in the sub-
`epidermal layer from 28.9% to 33.6% and decreased in the
`deeper dermis from 20.3% to 11.9% after eight weeks of
`Fumadermw therapy. Within the eight-week treatment
`period there was a complete clearing of neutrophil
`granulocytes from the skin in pre-treatment biopsies.
`Although treatment with fumarates lead to a profound
`decrease in the number of T cells in some patients, which
`might be a reason to stop therapy, adverse events related
`to a decreased immunosurveillance have not been
`observed in short term or long-term studies [12].
`In an in vitro study, the effect of MHF on T-cell cytokine
`production was analysed in greater detail [20]. The
`C
`authors demonstrated, in purified T cells (98% CD2
`)
`stimulated with antibodies against CD2, CD28 or a
`mixture of CD28 and CD3, that !200 mM MHF did not
`influence cell proliferation. MHF induced the production
`of the Th2-type cytokines IL-4 and IL-5 (EC50 100 mM), but
`was without effect on IL-2 or IFNg production, which are
`prominent Th1-type cytokines. The stimulating effect of
`MHF on IL-4 and IL-5 production was also shown for
`several different T-cell clones. By using radioactively
`labelled MHF and DMF, evidence for specific binding
`sites for MHF on T-cells was provided. In a separate set of
`C
`C
`CD45RO
`memory T cells were
`experiments, CD4
`stimulated with antibodies against CD2 and CD28 and
`the effect of MHF was evaluated. MHF upregulated the
`secretion of IL-4 2.5-fold and IL-5 threefold in stimulated
`memory T cells, without any effect on IFNg secretion. The
`induction of IL-4 and IL-5, but not of IFNg, by MHF was
`further seen in peripheral blood mononuclear cells
`(PBMCs) stimulated with purified protein derivative
`(PPD) as a pathogenic stimulus, mimicking an immune
`response against Mycobacterium tuberculosis. MHF
`enhanced the release of TNFa and IL-10 in lipopolysac-
`charide-stimulated monocytes without having an effect on
`IL-12 and IL-1RA secretion [21]. Newer data showed a
`reduced secretion of IFNg after the treatment of dendritic
`cells with MHF and subsequent T-cell stimulation [22].
`Goreschi et al. (unpublished data†) analysed the ex vivo
`Th1- and Th2-cytokine repertoire of T cells from patients
`with psoriasis treated with FAEs. They found a significant
`C
`suppression of the intracellular IFNg:IL-4 ratio in CD4
`T cells, which began after three weeks of therapy and
`paralleled clinical improvement.
`From these data,
`it was concluded that the main
`mechanism by which FAE induces the remission of
`psoriatic lesions is a shift of the immunological balance
`from a Th1- towards a Th2-like response. However, the
`prominent decrease in the number of T cells that is
`associated with FAE therapy prompted the investigation
`of the effect of these compounds on programmed cell
`death.
`
`FAE-induced apoptosis
`In U937-cells, Sebo¨ck et al.
`[23] were the first to
`demonstrate the potent apoptosis-inducing activity of
`
`† Goreschi, K. et al. (2002) Fumaric acid ester an anti-psoriatic drug abolishes the
`capacity of T cells to induce Th1-mediated autoimmune disease. Arch. Dermatol. Res.
`294, 28. Presented at the XXIX Annual meeting of the Arbeitgemeinschaft
`Dermatologische Forschung ADF, Ferbruary 28–March 2 2002, Berlin, Germany.
`
`Sawai (IPR2019-00789), Ex. 1019, p. 003
`
`

`

`46
`
`Review
`
`TRENDS in Molecular Medicine Vol.11 No.1 January 2005
`
`DMF. These data were confirmed in human monocyte-
`derived dendritic cells as an important regulator of specific
`T-cell activation. Zhu and Mrowietz [24] showed that DMF
`and MHF could completely prevent the GM-CSF- and
`IL-4-induced differentiation of monocytes into dendritic
`cells. DMF also potently induced apoptosis in these cells.
`Recently, the effect of FAEs on T-cell apoptosis was
`investigated in greater detail. In purified human T cells,
`DMF, but not MHF, could induce apoptosis. This was
`substantiated by measuring two independent markers
`[Apo2.7 expression and DNA fragmentation, using
`TUNEL (Tdt-mediated dUTP nick end labeling)]. T cells
`stimulated with IL-2, antibodies against OKT3 or both
`were more susceptible to DMF-mediated apoptosis com-
`pared with unstimulated cells. In parallel to an increase in
`Apo2.7 expression, DMF induced a dose-dependent down-
`regulation of anti-apoptotic Bcl2-expression [25].
`
`FAEs modulate cytokine production
`In a series of experiments, the effect of FAEs, notably
`DMF, on inflammatory mediator transcription and pro-
`duction was investigated. Using human PBMCs, DMF
`inhibited lipopolysaccharide-induced protein secretion of
`the chemokines IL-8, Mig and IP-10 in a dose-dependent
`manner without altering cell viability [27].
`Because keratinocytes themselves are actively produc-
`ing inflammatory mediators, such as chemokines, Stoof
`et al. [26] investigated the effect of DMF on phorbol-ester-
`or
`IFNg-stimulated normal human keratinocytes
`obtained from healthy human skin. They showed that
`DMF caused a dose-dependent inhibition of groa, IL-8,
`Mig, IP-10 and IP-9/I-TAC mRNA expression. Further-
`more, protein secretion of IL-8, Mig and IP-10 was
`decreased by the DMF treatment of keratinocytes.
`Ockenfels et al. [27] co-cultured normal human kera-
`tinocytes obtained from patients with psoriasis together
`with HUT78-cells (a T-cell-like cell line). DMF inhibited
`IFNg, IL-6 and TGFa release and increased IL-10 release
`in all co-culture experiments.
`
`FAE modulates adhesion molecule expression
`Because the inflammatory response in psoriasis is associ-
`ated with an increased expression of adhesion molecules
`on endothelial cells and keratinocytes [28], the effect of FAEs
`was investigated. Vandermeeren and co-workers [29]
`showed that DMF inhibited TNFa- or IL-1a-stimulated
`expression of the adhesion molecule ICAM-1 in human
`fibroblasts.
`In human umbilical vein endothelial cells (HUVECs),
`DMF blocked the TNFa-induced expression of the
`adhesion molecules ICAM-1, VCAM-1 and E-selectin
`[29]. Furthermore, the adherence of monocytic U937
`cells to TNFa- and IL-4-treated HUVECs was inhibited
`by DMF.
`
`FAEs inhibit NFkB activity
`Because the transcription of chemokines and adhesion
`factors, including IL-8 and E-selectin, is dependent upon
`NFkB, it is possible that DMF modulates this regulatory
`pathway. Additional evidence for a possible role of NFkB
`was provided by the observation that apoptosis is, at least
`
`www.sciencedirect.com
`
`in part, dependent on the modulation of NFkB activity as a
`regulator of cell survival. Bureau et al. [30] demonstrated
`the importance of constitutively active NFkB for the
`survival of quiescent mature immune cells, such as T and
`B cells, monocytes and macrophages and neutrophil
`granulocytes. The blockade of NFkB lead to apoptotic
`cell death in human macrophages, which depended on the
`activation of caspase 9, but not caspases 3 or 8 [31]. In
`human T cells, the inhibition of NFkB translocation was
`associated with apoptotic cell death independent of
`caspases 1 and 3 [32]. Ward et al. [33] found TNFa to be
`a survival
`factor for neutrophils linked to NFkB
`activation.
`When the effect of DMF on the NFkB-pathway was
`analysed more closely, a significant inhibition of kB1/p50
`in the nucleus was found, whereas there was little influ-
`ence on IkBa, b and 3 or RelA/p65 or c-Rel. AP-1-mediated
`gene transcription remained unaltered [34].
`The effect of DMF on the NFkB cascade was further
`investigated using the TNFa- or VEGF-induced mRNA
`expression of tissue factor in human endothelial cells [35].
`DMF inhibited TNFa-, but not VEGF-, induced tissue factor
`expression. Further experiments revealed an inhibition of
`TNFa-mediated nuclear entry of p65. This was not due to
`the inhibition of TNFa-induced signalling to IkB, because
`IkBa phosphorylation and degradation were not altered
`[36]. By interfering with NFkB translocation, DMF acts not
`only as an inhibitor of TNFa-induced cellular functions but
`also as an inhibitor of TNFa-production, because its
`transcription is, at least in part, dependent on NFkB.
`Litjens et al. [22] provided evidence that, in addition to
`DMF, MHF inhibited LPS-induced NFkB activation in
`human dendritic cells.
`
`FAEs interfere with cellular redox-systems
`Barchowsky et al. [37], using porcine aortic endothelial
`cells, found an association between DMF-mediated inhi-
`bition of NFkB translocation and intracellular thiol levels.
`The phosphorylation and ubiquitination of IkB is depen-
`dent on the balance of reduced glutathione (GSH) and
`oxidized glutathione-disulfide (GSSG), which is influenced
`by intracellular reactive oxygen species (ROS) through
`redox-sensitive kinases [38].
`Data from the toxicology literature indicate a direct
`link between cell death by necrosis or apoptosis and
`intracellular levels of thiols. In a model of acetaminophen-
`induced liver toxicity, hepatocyte necrosis was observed in
`relation to decreased glutathione levels [39]. The addition
`of antioxidant glutathione-monoethylester shifted necrotic
`cell death to apoptosis. An increase of oxidized and a
`reduction of reduced glutathione lead to increased
`apoptosis in M14 melanoma cells, which was associated
`with a downregulation of the c-myc protooncogene [40].
`The effect of DMF on intracellular thiols is time
`dependent. The addition of DMF to Chinese hamster
`ovary cells for 5 min was followed by a decrease of
`intracellular glutathione to 10% of basal
`levels [41].
`When the activity of the glutathione-synthesis-limiting
`enzyme g-glutamine–cysteine synthase in guinea pig
`hippocampus cells was inhibited by the addition of the
`irreversible inhibitor buthioninsulfoximine, intracellular
`
`Sawai (IPR2019-00789), Ex. 1019, p. 004
`
`

`

`Review
`
`TRENDS in Molecular Medicine Vol.11 No.1 January 2005
`
`47
`
`levels of reduced glutathione were decreased by 30% [42].
`The subsequent addition of DMF further reduced gluta-
`thione to 4% of baseline levels. However, Duffy et al. [43]
`demonstrated an increase of glutathione in a retinal-
`neuroblastoma hybrid cell-line by DMF. This discrepancy
`was further investigated by using human retinal pigment
`epithelial cells. Nelson and co-workers [44] found a time-
`dependent effect of DMF on intracellular thiols: an initial
`decrease of glutathione followed by a sustained increase of
`more than twofold.
`These data were confirmed in animal experiments in
`which DMF was added to the diet of mice and rats for two
`weeks, leading to a significant increase of glutathione and
`quinone-reductase, which is another detoxifying enzyme
`that is found in various organs, such as the liver and small
`intestine [45].
`From these experiments, it can be concluded that DMF
`can significantly modulate intracellular levels of detoxify-
`ing enzyme systems, such as the GSH–GSSG system,
`upregulating reduced-glutathione levels after prolonged
`exposure or dietary supplementation. There are no data
`about the activity of MHF in these systems.
`
`FAEs: hypothesized molecular mode of action
`Taken together, the following hypothesis about the
`mechanism of action of DMF can be proposed: (i) DMF
`interferes with intracellular thiols resulting in increased
`levels of reduced glutathione after prolonged exposure,
`through an unknown mechanism; (ii) increased gluta-
`thione levels inhibit redox-sensitive kinases, resulting in
`(iii) an inhibition of the phosphorylation and ubiquitina-
`tion of IkB,
`leading to (iv) an inhibition of NFkB
`translocation (Figure 2). This pathway leads to the
`modulation of the NFkB-dependent cascades of inflam-
`matory cytokine production and adhesion molecule
`expression. Because DMF seems not to interfere with
`the basal cytokine expression that is necessary for
`immune defence reactions, the documented long-term
`safety regarding the lack of an increased risk of infections
`or tumour development might be explained.
`
`Concluding remarks
`The empirically observed effect of FAE on the immuno-
`logically mediated skin disease psoriasis has led to the
`discovery of the potent immunomodulatory activity of the
`active compound DMF. FAEs, particularly DMF, might be
`regarded as potent immunomodulators with clinically
`proven effectiveness in psoriasis. From the present data,
`the inhibition of NFkB translocation and downstream
`NFkB-dependent pro-inflammatory pathways are the
`hallmarks of the mode of action of DMF. The interference
`with intracellular redox-systems might lead directly to the
`effects observed.
`Unfortunately, the product approved only in Germany
`(Fumadermw), being a mixture of different FAEs com-
`posed by empirical means, could not be licensed else-
`where. However, a new drug, termed BG-12, is currently
`in clinical development and consists of DMF as a
`monosubstance in a microtablet formulation.
`Future research should be directed towards determin-
`ing how FAEs, namely DMF and its metabolites, including
`
`www.sciencedirect.com
`
`DMF
`
`+
`
`–
`
`GSH:GSSG
`
`ROI
`
`+
`–
`Redox-sensitive kinases
`
`Phosphorylation of IκB
`
`Ubiquitination of IκB
`
`Cytosol
`
`Poly-ubiquitinated IκB
`is degraded by
`26S proteasome
`
`NF-κB activation
`
`
`
`TranslocationTranslocation
`
`Nucleus
`GSSG
`+ –
`NFκB
`Binding
`
`DMF
`
`+
`
`–
`
`GSH
`
`–
`DMF
`
`NFκB-site
`
`TRENDS in Molecular Medicine
`
`Figure 2. Proposed mode of action of DMF on NFkB-regulated gene transcription.
`The balance between oxidized glutathione (GSSG) and reduced glutathione (GSH)
`as well as the presence of reactive oxygen intermediates (ROI) regulates
`degradation of the inhibitor of kB (IkB) and subsequent activation of NFkB. DMF
`increases GSH, leading to an inhibition of NFkB-activation. Furthermore, DMF
`inhibits NFkB p65 subunit translocation from the cytosol into the nucleus, and by
`increasing GSH levels decreases the binding of NFkB to DNA.
`
`free fumaric acid, interact with intracellular structures
`and components at the molecular level. The establishment
`of more sophisticated methods for analysing FAE metab-
`olism in vivo and ex vivo is an essential prerequisite. The
`identification of the mode of action of FAEs in more detail
`might lead to the identification of promising new drug
`targets. More clinical studies are also needed; these
`should include larger patient populations over longer
`periods of time, to verify the suspected favourable long-
`term safety.
`
`References
`1 Blank, R. et al. (1999) Effect of fumaric acid and dietary buffering
`capacity on ileal and fecal amino acid digestibilities in early-weaned
`pigs. J. Anim. Sci. 77, 2974–2984
`2 Schweckendiek, W. (1959) Heilung von Psoriasis. Med. Monatschr. 13,
`103–104
`3 Mrowietz, U. et al. (1999) Treatment of severe psoriasis with fumaric
`acid esters: scientific background and guidelines for therapeutic use.
`Br. J. Dermatol. 141, 424–429
`4 Balasubramaniam, P. et al. (2004) Fumaric acid esters in severe
`psoriasis, including experience of use in combination with other
`systemic modalities. Br. J. Dermatol. 150, 741–746
`5 Ormerod, A.D. and Mrowietz, U. (2004) Fumaric acid esters, their
`place in the treatment of psoriasis. Br. J. Dermatol. 150, 630–632
`6 Prinz, J.C. (2001) Psoriasis vulgaris – a sterile antibacterial skin
`reaction mediated by cross-reactive T cells? An immunological view of
`the pathophysiology of psoriasis. Clin. Exp. Dermatol. 26, 326–332
`7 Boehncke, W.H. et al. (1996) Pulling the trigger on psoriasis. Nature
`379, 777
`8 Vollmer, S. et al. (2001) Dominant lesional T cell receptor rearrange-
`ments persist in relapsing psoriasis but are absent from nonlesional
`skin: evidence for a stable antigen-specific pathogenic T cell response
`in psoriasis vulgaris. J. Invest. Dermatol. 117, 1296–1301
`9 Elder, J.T. et al. (2001) The genetics of psoriasis 2001: the odyssey
`continues. Arch. Dermatol. 137, 1447–1454
`
`Sawai (IPR2019-00789), Ex. 1019, p. 005
`
`

`

`48
`
`Review
`
`TRENDS in Molecular Medicine Vol.11 No.1 January 2005
`
`(1994) Antipsoriatic effect of fumaric acid
`10 Altmeyer, P. et al.
`derivatives. Results of a multicenter double-blind study in 100
`patients. J. Am. Acad. Dermatol. 30, 977–981
`11 Mrowietz, U. et al. (1998) Treatment of psoriasis with fumaric acid
`esters. Results of a prospective multicenter study. Br. J. Dermatol.
`138, 456–460
`12 Hoefnagel, J.J. et al. (2003) Long-term safety aspects of systemic
`therapy with fumaric acid esters in severe psoriasis. Br. J. Dermatol.
`149, 363–369
`13 Nieboer, C. et al. (1989) Systemic therapy with fumaric acid derivates:
`new possibilities in the treatment of psoriasis. J. Am. Acad. Dermatol.
`20, 601–608
`14 Ho¨xtermann, S. et al. (1998) Fumaric acid esters suppress peripheral
`CD4- and CD8-positive lymphocytes in psoriasis. Dermatology 196,
`223–230
`15 Henseler, T. and Christophers, E. (1995) Disease concomitance in
`psoriasis. J. Am. Acad. Dermatol. 32, 982–986
`16 Litjens, N.H. et al. (2004) Pharmacokinetics of oral fumarates in
`healthy subjects. Br. J. Clin. Pharmacol. 58, 429–432
`17 Werdenberg, D. et al. (2003) Presystemic metabolism and intestinal
`absorption of antipsoriatic fumaric acid esters. Biopharm. Drug
`Dispos. 24, 259–273
`18 Altmeyer, P. et al. (1996) Verlaufsbeobachtungen der Lymphozyten-
`subpopulationen bei Psoriasis-Patienten unter oraler Therapie mit
`Fumaraten. Aktuel. Dermatol. 22, 272–277
`19 Bacharach-Buhles, M. et al. (1994) Fumaric acid esters (FAEs)
`suppress CD15- and ODP 4-positive cells in psoriasis. Acta Derm.
`Venereol. Suppl. (Stockh.) 186, 79–82
`20 De Jong, P. et al. (1996) Selective stimulation of T helper 2 cytokine
`responses by the anti-psoriasis agent monomethylfumarate. Eur.
`J. Immunol. 26, 2067–2074
`21 Asadullah, K. et al. (1997) Influence of monomethylfumarate on
`monocytic cytokine formation-explanation for adverse and thera-
`peutic effects in psoriasis? Arch. Dermatol. Res. 289, 623–630
`22 Litjens, N.H. et al. (2004) Monomethylfumarate affects polarization of
`monocyte-derived dendritic cells resulting in down-regulated Th1
`lymphocyte responses. Eur. J. Immunol. 34, 565–575
`23 Sebo¨k, B. et al. (2000) Dimethylfumarat ist unter den Fumadermw-
`Inhaltsstoffen der sta¨ rkste Induktor von Apoptose-Pha¨ nomenen in
`lympho-histiozyta¨ ren U-937-Zellen. HGZ. Hautkr. 75, 1–5
`24 Zhu, K. and Mrowietz, U.
`(2001) Inhibition of dendritic cell
`differentiation by fumaric acid esters. J. Invest. Dermatol. 116,
`203–208
`25 Treumer, F. et al. (2003) Dimethylfumarate is a potent inducer of
`apoptotic cell death in human T cells. J. Invest. Dermatol. 121,
`1383–1388
`26 Stoof, T.J. et al. (2001) The antipsoriatic drug dimethylfumarate
`strongly suppresses chemokine production in human keratinocytes
`and peripheral blood mononuclear cells. Br. J. Dermatol. 144,
`1114–1120
`27 Ockenfels, H.M. et al. (1998) The antipsoriatic agent dimethylfuma-
`rate immunomodulates T-cell cytokine secretion and inhibits cyto-
`kines of the psoriatic cytokine network. Br. J. Dermatol. 139, 390–395
`
`28 Veale, D. et al. (1995) Immunolocalization of adhesion molecules in
`psoriatic arthritis, psoriatic and normal skin. Br. J. Dermatol.
`132, 32–38
`29 Vandermeeren, M. et al. (1997) Dimethylfumarate is an inhibitor of
`cytokine-induced E-selectin, VCAM-1, and ICAM-1 expression in
`human endothelial cells. Biochem. Biophys. Res. Commun. 234, 19–23
`30 Bureau, F. et al. (2002) Constitut

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket