throbber
Clin Transplant 2006: 20 (Suppl. 17): 17–24
`
`Copyright ª Blackwell Munksgaard 2006
`
`FTY720 (fingolimod) in renal
`transplantation
`
`Budde K, Schu¨ tz M, Glander P, Peters H, Waiser J, Liefeldt L, Neumayer
`H-H, Bo¨ hler T. FTY720 (fingolimod) in renal transplantation.
`Clin Transplant 2006: 20 (Suppl. 17): 17–24. ª Blackwell Munksgaard, 2006
`
`Abstract: FTY720 (Fingolimod) is a novel immunomodulator with a mode
`of action that is completely different from classical immunosuppressants.
`FTY is a structural and functional analogue of the natural serum lipid,
`sphingosine, and is the first in a new class of drugs called sphingosine
`1-phosphate receptor (S1P-R) modulators. This review discusses the recent
`findings on the mechanism of action, preclinical models and outlines the
`results of the ongoing clinical development program. FTY is highly effective
`in prolonging allograft survival in preclinical models of transplantation and
`in experimental models of autoimmune diseases. In clinical trials, this novel
`compound was investigated in de novo renal transplantation and in multiple
`sclerosis. Pharmacokinetics are characterized by a prolonged absorption
`phase, a large volume of distribution, and a long elimination half-life. FTY
`induces a rapid and transient decrease in lymphocyte counts, which sup-
`ports the modulatory effects of the drug on lymphocyte sequestration. The
`most common adverse event was asymptomatic transient bradycardia, a
`pharmacodynamic effect modulated by atrial S1 P-R. FTY failed to show
`an improvement in efficacy for the prevention of renal allograft rejection in
`two large phase III studies. FTY treatment regimens were associated with
`impaired renal function and the development of macula edema. Conse-
`quently, the further development in renal transplantation was stopped.
`Because initial clinical studies strongly suggest that FTY is highly effective
`in multiple sclerosis FTY is now being explored in phase III studies for the
`treatment of demyelinating diseases, Ongoing studies in multiple sclerosis
`are eagerly awaited because they may provide novel therapeutic options for
`patients with autominnue diseases.
`
`Klemens Buddea, Manuela
`Schu¨ tza, Petra Glandera, Harm
`Petersa, Johannes Waisera, Lutz
`Liefeldta, Hans-H. Neumayera and
`Torsten Bo¨ hlerb
`
`aDepartment of Nephrology, Charite´
`Universitatsmedizin Berlin, Germany and
`bINSERM U466, Institut Louis Bugnard, CHU
`Rangueil, Toulouse, France
`
`Key words: FTY720 – fingolimod – sphingosine 1
`– phosphate receptor – renal transplantation –
`multiple sclerosis
`
`Corresponding author: Klemens Budde MD,
`Department of Nephrology, Charite´, Universi-
`ta¨tsmedizin Berlin, Schumannstr. 20/21, 10098
`Berlin, Germany.
`Tel.: +49 30 450 514 002;
`fax: +49 30 450 514 902;
`e-mail: klemens.budde@charite.de
`
`Current immunosuppressive protocols based on
`calcineurin inhibitors (CNI), anti-metabolites and
`steroids allow one yr renal graft survival rates of
`more than 90% (1). Despite these improvements in
`the prevention of solid organ graft rejection during
`the last decade, there remains a medical need for
`new regimens that minimize the frequent drug-
`related toxicities associated with these agents (2).
`Organ and bone marrow toxicity, severe gastroin-
`testinal side-effects, pharmacokinetic variability
`and chronic allograft dysfunction necessitate the
`development of novel drugs for new immunosup-
`pressive strategies in transplantation medicine. The
`current immunosuppressives have a narrow thera-
`peutic window and are frequently associated with
`clinical signs of overimmunosuppression such as
`malignancies and opportunistic infections (3, 4). In
`
`addition, the transplant community is now seeking
`for better control of cardiovascular risk factors,
`because cardiovascular disease is the main cause of
`death in patients after successful kidney transplan-
`tation (5, 6).
`The new therapeutic agent, FTY720 (FTY,
`fingolimod) (7), may address some of the medical
`needs by providing a therapeutic alternative in
`combination therapy. FTY, a structural and
`functional analogue of the natural serum lipid,
`sphingosine, is the first in a new class of drugs
`called sphingosine 1-phosphate receptor (S1P-R)
`modulators (8, 9). FTY is a novel immunomod-
`ulator with a mode of action that appears to be
`completely different from classical
`immunosup-
`pressants: FTY does not impair T- and B-cell
`activation, proliferation and effector function in
`
`17
`
`Apotex v. Novartis
`IPR2017-00854
`NOVARTIS 2030
`
`

`

`Budde et al.
`
`infection (10). It is
`response to systemic viral
`highly effective in prolonging allograft survival in
`preclinical models of
`transplantation. Further-
`more, FTY acts synergistically with CNIs and
`proliferation inhibitors in these models, suggesting
`that use of FTY in combination with classical
`immunosuppressants may be a promising new
`option for transplant patients. FTY has a unique
`safety profile that may not overlap with currently
`available immunosuppressants. Therefore it offers
`the opportunity to explore synergy in efficacy with
`other agents and avoid their related toxicities.
`FTY is currently being investigated in de novo
`renal transplant population (11, 12) as well as in
`other therapeutic areas, such as multiple sclerosis
`(MS) (13). The aim of this review is to discuss the
`recent findings on the mechanism of action,
`preclinical models and to briefly outline the
`results of the ongoing clinical development pro-
`gram of this drug.
`
`Mechanism of action and preclinical development
`
`Soon after the discovery of the high efficacy of the
`synthetic sphingolipid FTY to prevent allograft
`rejection in animal models it became evident that
`the mechanism of action of FTY differs completely
`from all known conventional immunosuppressive
`drugs (14). FTY does not inhibit proliferation or
`activation of lymphocytes (15). First animal experi-
`ments with FTY showed a reversible reduction of
`lymphocyte counts in the peripheral blood (3) and
`later also in humans (4). The first assumption was
`that peripheral lymphopenia is caused by apoptosis
`of
`lymphocytes
`(16). However, apoptosis of
`lymphocytes could not be detected in FTY720
`treated renal allograft recipients and took place at
`supra-pharmacological
`concentrations of FTY
`in vitro (17). Another hypothesis suggested that
`FTY induced the sequestration of lymphocytes
`from the peripheral blood compartment
`into
`lymphoid tissue, because the FTY induced periph-
`eral
`lymphopenia was associated with increased
`lymphocyte counts in lymph nodes (LN), Peyer’s
`patches and thymus (18, 19). Using adoptive
`transferred lymphocytes it was demonstrated that
`previously labelled lymphocytes return back into
`the circulation after cessation of FTY treatment
`(20). This effect was sensitive to pertussis toxin,
`which indicates a G-protein coupled signal trans-
`duction for FTY. In lymphocyte cultures, FTY
`exerted a differential expression of proteins linked
`to cytoskeleton/cell motility, to cell adhesion and
`vesicle trafficking (21). By synthesis of a chiral
`FTY analogue,
`the modulation of
`lymphocyte
`trafficking could be dissected from the FTY
`
`18
`
`induced lymphocyte apoptosis (22). Pinchewer
`(10) showed that FTY treatment does not affect
`expansion, memory and anti-viral activity of the
`immune response. Therefore FTY can be regarded
`rather as an ÔimmunomodulatorÕ than an Ôimmu-
`nosuppressantÕ.
`The molecular structure of the sphingolipid FTY
`resembles
`the natural occurring sphingosine-1
`molecule. Similar to sphingosine FTY is phos-
`phorylated by sphingosine kinases to FTY-P (23,
`24) and the phosphorylated form is the active
`metabolite binding with higher affinity as its
`natural ligand to the S1P-R (13, 25, 26), formerly
`named endothelial differentiation gene (EDGs).
`Importantly, FTY-P does not solely elicit agonistic
`responses after binding to the S1P-R. After binding
`to S1P-R FTY-P down-regulates the receptor
`expression,
`thus leading to a functional S1P-
`antagonism (27).
`Early in vitro experiments indicated that chem-
`okines and its receptors might be involved in the
`mechanism of action of FTY.
`Indeed, FTY
`modulates the migratory response toward chem-
`okines in lymphocyte culture experiments (28).
`Foerster observed a delayed lymphopenia after
`administration of FTY to CCR7 knock-out mice
`suggesting chemokine receptor CCR7 dependent
`and independent mechanisms
`(29). FTY may
`render lymphocytes more susceptible to the chem-
`okine-driven migration into LN. Naive lympho-
`cytes entering the LN through high endothelial
`venules (HEV) down-regulate their S1P-R in order
`to stay and interact with antigen presenting cells in
`the LN (30). In order to migrate from LN back
`into the circulation lymphocytes need to up-regu-
`late their S1P-R. High S1P-concentrations in the
`blood create a concentration gradient between
`blood and LN, and S1P serves as one of the most
`powerful chemoattractants for S1P-R expressing
`lymphocytes in LN. The group of Cyster (31)
`showed that down-regulation of S1P-R on lympho-
`cytes reduced the egress of lymphocytes from LNs
`into the blood stream, thus causing peripheral
`lymphopenia. By permanent down-regulation of
`the S1P-R FTY interferes with the egress of
`activated lymphocytes,
`resulting in peripheral
`lymphopenia and a profound decrease of periph-
`eral effector lymphocytes. Because only naive
`CD62L-expressing lymphocytes
`encounter LN
`through HEV, FTY does not interfere with already
`activated effector cells or memory T-cells from the
`peripheral blood by this mechanism.
`In line with the proposed mechanism of action of
`FTY, experiments performed in non-human pri-
`mates (32) and humans (11) revealed that the
`severity of FTY-induced lymphopenia varies
`
`

`

`between different lymphocyte subsets. T cells were
`more reduced than B-cells, CD4 more than CD8
`T-cells, however, granulocytes and monocytes were
`not affected by FTY. Patients treated with FTY
`had a predominant reduction of CD62L-positive
`cells, while CCR5-positive T-cells were affected to
`a lesser degree (33). It is conceivable that the
`remaining T-cells in the periphery can infiltrate the
`graft and mediate graft rejection (34). Interestingly,
`FTY concentrations needed to induce lymphope-
`nia are lower than those to yield optimal rejection
`prophylaxis in transplant models. This observation
`suggests, that the anti-rejection properties of FTY
`are mediated at least in part by other mechanisms
`besides the effects on lymphocyte homing. S1P-R
`are also expressed on other cell types like dendritic
`cells, heart muscle, and endothelial cells, however
`these cells express a different pattern of S1P-R
`subtypes (35). Recently, it has been shown that
`FTY interferes with effector functions of human
`monocyte-derived DC (36, 37), providing new
`insights into the immunomodulatory mechanism
`of FTY.
`The S1P-R expression on atrial myocytes ex-
`plains the FTY effect on heart rate (38). Recent
`data indicated that direct agonism of FTY-P on
`S1P-R expressed in atrial myocytes of the heart is
`responsible for the negative chronotropic effects of
`FTY. FTY has been shown to activate cardiac
`G-protein-gated potassium channels IKACh (39).
`The reduction in heart rate observed following
`FTY is thought to be mediated by activation of
`these channels and the transient nature of the effect
`may be due to agonist-induced homologous desen-
`sitization involving S1P-R internalization (38).
`Furthermore, FTY exerts profound effects on
`endothelial cell function and permeability (40, 41),
`as evidenced by the fact that the S1P-Rs were first
`identified as receptors involved in endothelial
`differentiation. Endothelial cell
`layers serve as
`selective barriers regulating the transmigration
`from macromolecules and lymphocytes. It has
`been demonstrated that FTY affects lymphocyte
`transmigration explaining reduced lymphocyte
`infiltrates in grafted organs. The effects of FTY
`on endothelial cells could also account for the
`observed protective effects of FTY in models of
`chronic graft rejection. On the other hand one
`might speculate that the effects of FTY on endo-
`thelial cell permeability are related to some of the
`observed side-effects in FTY treated patients.
`The research on the biology of S1P-R is just in
`the beginning and new derivatives of FTY-P with
`better S1P-R subtype specificity may help to better
`elucidate these mechanism and eventually help to
`avoid some of the observed side-effects.
`
`FTY720 (fingolimod) in renal transplantation
`
`FTY was preclinically investigated in several
`animal species and in numerous transplantation
`models (42). The most prominent pharmacody-
`namic effect in all animal models tested was the
`induction of reversible lymphopenia. FTY was
`shown to prevent skin, kidney, liver and heart in
`models of allograft rejection (7, 9, 42). FTY exerts
`synergism with all other tested immunosuppres-
`sants including CNIs, proliferation inhibitors and
`mycophenolate to prevent acute and chronic
`allograft rejection. An overlap and therefore a
`synergistic increase of the incidence and severity of
`side-effects of FTY with the conventional drugs
`has not been observed. Furthermore, FTY may
`attenuate ischaemia-reperfusion injury in hepatic
`and renal transplantation (43, 44). The potential of
`FTY to treat autoimmune disease was also con-
`firmed in several experimental models for MS,
`auto-immune diabetes,
`lupus nephritis and Thy
`1-induced mesangial
`proliferative
`glomerulo-
`nephritis (45).
`
`First clinical experience with FTY in phase I and II
`trials
`
`Early phase I and phase II studies (4, 11, 46) have
`shown that FTY is well tolerated in renal trans-
`plant recipients and provides effective rejection
`prophylaxis (12) with minimal addition to the
`side-effect burden. The pharmacokinetics, phar-
`macodynamics and safety of FTY have been
`investigated in two randomized, double-blind,
`placebo-controlled, phase I studies involving stable
`renal transplant patients (at least one-yr post-
`transplantation) maintained on a cyclosporin A
`(CsA)-based regimen.
`
`Pharmacokinetics
`
`These studies revealed unique pharmacokinetics
`characterized by a prolonged absorption phase,
`with an elimination half-life >100 h, independent
`of dose. Consistent with the long half-life of FTY
`(47), almost all subjects took around four wk of
`daily dosing to reach steady-state concentrations.
`Maximum plasma concentration (Cmax) and area
`under the curve (AUC) were dose-proportional,
`with low inter-subject variability. Furthermore,
`FTY had an unusually high apparent volume of
`distribution (>1400 L) and a relatively low appar-
`ent oral clearance (median 158 mL/min). Addi-
`tional pharmacokinetic data were obtained from a
`phase II study, confirming the pharmacokinetic
`results from phase I studies. Again, FTY exhibited
`a long terminal half-life of around 200 h, a high
`volume of distribution (>3000 L) and a low
`
`19
`
`

`

`Budde et al.
`
`clearance (10.8 L/h) in de novo renal allograft
`recipients. The high volume of distribution is
`consistent with the lipophilic character of the drug
`and suggests widespread tissue distribution, e.g., in
`cellular membranes throughout
`the body. The
`prolonged half-life of FTY suggests that the agent
`can be given once daily, with little fluctuation over
`the dosing interval at steady state. Furthermore,
`the low inter-subject variability observed following
`administration of FTY indicates consistent absorp-
`tion and disposition of the drug, which should
`allow for a simple, standardized dosing regimen for
`all subjects, without
`the need for blood level
`monitoring or individualized dose titration.
`
`Pharmacodynamics
`
`Animal transplantation models have consistently
`shown that FTY, administered at pharmacological
`doses, induces a decrease in lymphocyte count (3,
`48, 49). This pharmacodynamic effect of FTY has
`also been observed in all human studies. Admin-
`istration of a single dose of FTY to stable renal
`transplant patients resulted in a transient decrease
`in lymphocyte count within four h of administra-
`tion in all subjects (4, 11). This pharmacodynamic
`effect of FTY exhibited non-linear dose-depend-
`ence, with the most prolonged and intensive
`reduction in lymphocyte count being observed in
`the highest dose group. The reduction in lympho-
`cyte counts is much faster than the pharmacoki-
`netics of the drug with a long terminal half-life
`might predict. Furthermore, the effect of FTY on
`lymphocyte count was pan-lymphocytic, affecting
`all principal lymphocyte subsets, with the greatest
`effects being observed on CD4+ and naı¨ ve T cells,
`while monocyte and granulocyte counts remain
`unaffected (11). A marked decline in peripheral
`lymphocytes (of around 85% of baseline) has also
`been demonstrated following multiple doses of
`FTY in stable renal transplant patients (11, 46) and
`in de novo allograft recipients following transplan-
`tation (12). After cessation of treatment, recovery
`was evident within three d after the last dose, with
`a trend toward complete recovery to baseline
`values in all dose groups four wk after last dose
`(47). All doses of FTY appeared to affect all
`lymphocyte subsets tested to a similar degree. In
`contrast to other immunosuppressive agents, mul-
`tiple doses of FTY had no effect on blood
`granulocytes, monocytes or eosinophils, and also
`had no effect on the number of erythrocytes or
`platelets (11). The predominant effect of FTY on
`CD4+ and naı¨ ve T cells observed in these phar-
`macodynamic studies supports the potential mod-
`ulatory effect of the drug on sequestration of these
`
`20
`
`cells into lymph nodes and Peyer’s patches (50).
`The decrease in PBL occurs immediately after the
`first dose, persists throughout the entire period of
`treatment and recovers slowly and completely after
`treatment discontinuation. Furthermore, the find-
`ings of these studies suggest that pharmacokinetics
`only partially explain the pharmacodynamic re-
`sponse to FTY. Consequently, pharmacodynamic
`monitoring, by measuring lymphocyte number,
`may be a useful predictor of the response to FTY
`in future trials (47).
`
`Safety
`
`FTY has been found to have an excellent tolerab-
`ility profile in these early phase I and II trials:
`overall, FTY was well tolerated following single-
`dose and multiple dose administration (4, 12, 46).
`The most common adverse event was bradycardia.
`Already in the first in human study, an increased
`frequency of bradycardia was observed in patients
`receiving higher doses of FTY and in patients with
`mild bradycardia at baseline (4). Similar to lymp-
`hopenia, heart rates reached a nadir around four to
`eight h after FTY administration and lasted for up
`to 24 h. The transient reduction in heart rate was
`asymptomatic and required no clinical interven-
`tion; all patients recovered without sequelae. In
`subsequent studies in de novo transplant patients
`about 25% of patients, this negative chronotropic
`effect
`led to reports of
`transitory bradycardia
`events that were mild or moderate in severity
`(12). The vast majority of bradycardia adverse
`events (70%) as reported by the investigators were
`reported within 24 h after the first dose and 85%
`within the first 48 h after the first drug adminis-
`tration. These events were asymptomatic in most
`of the patients, with no changes of blood pressure
`and no other cardiac abnormality, and none
`resulted in clinical
`sequelae. Bradycardia was
`reversible spontaneously (in about 60% of the
`cases) or following administration of atropine
`or b2-adrenergic agents. Only 1.6% of treated
`patients discontinued FTY because of bradycardia.
`FTY has not been associated with any other
`cardiac events in clinical trials to date, and the
`incidences of cardiac failure and myocardial
`infarction observed in a phase II trial with the
`agent were similar to those reported in the litera-
`ture (5, 6). FTY did not appear to cause any other
`specific adverse events, electrocardiogram or phys-
`ical examination changes or laboratory abnormal-
`ities. No additional organ toxicity was observed in
`these phase I and II studies. Despite some preclin-
`ical data suggesting lung toxicity, in these early
`clinical trials, there has been no evidence of an
`
`

`

`increased rate of pulmonary adverse events and no
`abnormalities in the pulmonary tests performed in
`renal transplant patients treated with FTY (4). In
`conclusion, from the clinical trials in phase I and
`II, it appeared that there were no safety concerns,
`and the transient effect of FTY on heart rate with
`treatment initiation is asymptomatic, self limiting
`and manageable and is not associated with any
`measurable increase in mortality or cardiac mor-
`bidity.
`
`Ecacy and safety in recent clinical trials
`
`To date, more than 1000 subjects have received
`FTY (2.5 or 5.0 mg/d)
`in combination with
`cyclosporine and corticosteroids in the context of
`de novo renal transplantation with a mean follow-
`up of >12 months after transplantation. FTY has
`not been examined in maintenance patients. Over-
`all, FTY was well tolerated and has demonstrated
`efficacy in the prevention of acute graft rejection in
`human kidney transplantation.
`The efficacy of FTY in preventing acute rejection
`in de novo renal transplant recipients has been
`evaluated in two phase II studies (12, 51). These
`studies have demonstrated that FTY 2.5 mg plus
`full dose CsA microemulsion (NeoralÒ; FDN
`Novartis Pharma, Basel, Switzerland) and 5 mg
`plus reduced dose Neoral (RDN) provides equiv-
`alent efficacy to mycophenolate mofetil (MMF)
`with conventional Neoral dose – currently consid-
`ered the standard of care in de novo renal trans-
`plantation. Doses of FTY below 1 mg in
`combination with CsA were associated with a
`slightly higher incidence of rejection compared to
`standard therapy with MMF (12). Furthermore,
`FTY was well tolerated and did not appear to be
`associated with nephrotoxicity, bone marrow tox-
`icity or other side-effects commonly observed with
`immunosuppressant therapies (4, 12). As men-
`tioned above, bradycardia was the most prominent
`side-effect in these studies. Because of the favour-
`able safety profile and the good efficacy two large
`phase III studies were initiated in order to thor-
`oughly evaluate the safety and efficacy of this
`compound in comparison to mycophenolate in a
`large cohort of de novo renal allograft recipients
`with low to medium immunological risk.
`Two large phase III clinical trials compared the
`efficacy and safety of two doses of FTY (2.5 and
`5 mg) plus either RDN or FDN with MMF plus
`FDN in adult de novo renal transplant recipients
`(56, 57). Both 12-month phase III studies have
`shown that FTY 2.5 mg plus FDN provides com-
`parable efficacy for the prevention of acute rejection
`in de novo renal transplant recipients compared
`
`FTY720 (fingolimod) in renal transplantation
`
`with MMF plus FDN. The higher incidence of
`rejection observed with the FTY 5 mg plus RDN
`regimen suggests that FTY combined with CsA
`minimization (reduction ‡50%) may not provide
`sufficient prophylaxis from acute rejection.
`Surprisingly, renal allograft function was lower
`for the two FTY treatment arms compared with the
`MMF arm. Although the mean creatinine clearance
`increased over time in both FTY treatment groups,
`the rate and absolute increase was significantly lower
`in the FTY arm compared with MMF. Preliminary
`analysis suggests that renal function remains relat-
`ively stable with no evidence of progressive deteri-
`oration. The reason for this impairment is unknown
`and is currently under investigation, and could result
`from both sub-clinical
`immune injury and non-
`immunologic drug effects. Initial post hoc analysis of
`a population at low immunological risk without
`clinical evidence for rejection revealed also lower
`renal function in this subpopulation of patients
`suggesting direct or indirect non-immunological
`effects of FTY on renal function.
`Additional side-effects observed more frequently
`with FTY in these large phase III studies were a
`mild increase in airway resistance without evidence
`of progressive changes in lung volumes over time.
`Moderate elevations in ALT and bilirubin levels
`were noted with FTY compared with MMF; these
`differences are not
`thought
`to be of clinical
`significance. The two FTY regimens were also
`associated with twofold higher incidence of macu-
`lar oedema compared to the control group with
`MMF. The clinical significance of this effect is
`currently unclear and further detailed characteri-
`zation is ongoing.
`Given the fact that FTY offers no improvement
`in efficacy, however, may pose the patients at risk
`for these previously unknown side-effects of this
`novel class of drugs, it became clear that more
`analyses are needed to fully investigate this inter-
`esting drug and the causes of renal dysfunction in
`de novo renal transplant recipients. Especially the
`impaired renal function and the development of
`macula oedema are new unexpected and unex-
`plained side-effects, observed in the phase III trials,
`which need further thorough investigation, and
`have led to the temporary discontinuation of all
`ongoing trials in renal transplantation.
`
`Multiple sclerosis
`
`The remarkable effects of the drug in all animal
`models for autoimmune diseases tested so far
`provide a strong rationale for testing this com-
`pound in the clinical
`setting of autoimmune
`diseases. Especially the impressive results obtained
`
`21
`
`

`

`Budde et al.
`
`with FTY in EAE, the animal model of human
`MS, shows the potential of FTY for the treatment
`of demyelinating diseases (52, 53). The exact
`pathophysiology of MS remains unclear but an
`autoimmune response directed against central
`neuronal system antigens is suspected (54). Current
`treatment options consist of interferon, which may
`reduce relapse rates, and also may reduce disease
`progression (54). However to date, no effective
`treatment is available to prevent or cure MS. The
`convincing results from the animal models provi-
`ded the rationale for developing FTY in the clinical
`indication of MS. As recently reported (55), a
`phase II trial using 1.25 and 5 mg FTY showed a
`highly significant reduction (55%) in relapse rates.
`In addition, time to first relapse was significantly
`prolonged and 86% remained relapse free over six
`month compared with 70% in the placebo group.
`Inflammatory disease activity was reduced dramat-
`ically by 80% over six month treatment as evalu-
`ated by MRI lesions. Furthermore, new disease
`activity was reduced by more than two thirds
`compared with placebo (55). The oral application
`form and a less toxic profile makes FTY a
`promising candidate for the treatment of MS.
`Importantly, safety data from this trial did not
`reveal any nephrotoxicity and no cases of macula
`oedema have been reported in this setting so far,
`suggesting that especially the combination therapy
`with cyclosporine in the setting of renal transplan-
`tation might be responsible for the observed
`adverse events like nephrotoxicity and macula
`oedema. Thus, in conclusion phase II clinical data
`suggest that FTY will be a safe and effective
`treatment option for MS, and large prospective
`phase III trials are ongoing to confirm the exciting
`phase II results in relapsing MS.
`
`Conclusions
`
`The availability of agents with different mecha-
`nisms of action for preventing graft rejection that
`can be combined safely will be essential for the
`future of organ transplantation if further improve-
`ments in outcomes post-transplant are to be
`achieved. In contrast to classical immunosuppres-
`sants, FTY has no anti-proliferative activity at
`therapeutic dosing and rather acts as a Ôimmuno-
`modulatorÕ, suggesting that this agent may be
`useful to augment the immunosuppressive actions
`of other drugs. After phosphorylation FTY-P
`exerts its effects via binding to S1P-R on various
`cell types including lymphocytes, dendritic cells,
`endothelial cells and atrial myocytes. FTY leads to
`down-regulation of S1P-R, which causes lympho-
`cyte trapping within LN. Preclinical studies with
`
`22
`
`FTY have demonstrated that the agent is highly
`effective in a wide range of animal models of
`transplantation. Initial clinical studies with FTY
`suggested that this novel immunomodulator is also
`effective in human transplant recipients, producing
`a marked reduction in blood lymphocyte count
`following single and multiple dosing. Furthermore,
`FTY in combination with CsA was well tolerated
`in phase I and II studies in renal transplantation,
`with no significant safety issues except transient
`and asymptomatic bradycardia. Because phase II
`studies demonstrated in combination therapy with
`cyclosporine similar efficacy of FTY compared to
`standard therapy with mycophenolate, a large
`phase III program was initiated exploring the
`ability of FTY to provide equivalent efficacy to
`classical immunosuppressive regimens in the pre-
`vention of acute rejection. Although these studies
`demonstrated similar efficacy with respect
`to
`rejection prophylaxis of FTY in combination with
`regular cyclosporine doses, no clear benefit over
`conventional therapy with mycophenolate could be
`established. Furthermore,
`it was discovered in
`these pivotal studies that FTY therapy was asso-
`ciated with renal impairment and macula oedema
`and consequently, the clinical programme for the
`development of FTY in renal transplantation was
`paused.
`The FTY is also being investigated in patients
`with MS. However, the MS population differs
`considerably from the renal transplant population
`and the agent will be used at lower doses than in the
`renal
`transplant setting, and as monotherapy.
`Thus, the findings of the current study conducted
`in renal transplant recipients cannot be extrapola-
`ted to the use of FTY in MS patients. Furthermore,
`the drug represents a new modality that might lead
`to a quantum improvement in the treatment of
`autoimmune disorders. In conclusion,
`the full
`analysis of the ongoing studies is eagerly awaited,
`which might provide new insights on its novel
`mechanism of action and will allow to better define
`its role in the treatment of renal allograft recipients
`and patients with autoimmune diseases like MS.
`
`References
`
`1. Nishikawa K, Terasaki PI. Annual trends and triple
`therapy 1991–2000. Clin Transpl 2001: 247.
`2. Halloran PF. Immunosuppressive drugs for kidney
`transplantation. N Engl J Med 2004: 351: 2715.
`3. Suzuki S, Enosawa S, Kakefuda T et al. A novel im-
`munosuppressant, FTY720, with a unique mechanism of
`action, induces long-term graft acceptance in rat and dog
`allotransplantation. Transplantation 1996: 61: 200.
`4. Budde K, Schmouder RL, Brunkhorst R et al. First
`human trial of FTY720, a novel immunomodulator, in
`
`

`

`stable renal transplant patients. J Am Soc Nephrol 2002:
`13: 1073.
`5. Jardine AG, Fellstrom B, Logan JO et al. Cardiovas-
`cular risk and renal transplantation: post hoc analyses of
`the Assessment of Lescol
`in Renal Transplantation
`(ALERT) Study. Am J Kidney Dis 2005: 46: 529.
`6. Lentine KL, Brennan DC, Schnitzler MA. Incidence
`and predictors of myocardial
`infarction after kidney
`transplantation. J Am Soc Nephrol 2005: 16: 496.
`7. Kahan BD. FTY720: a new immunosuppressive agent
`with novel mechanism(s) of action. Transplant Proc 1998:
`30: 2210.
`8. Clemens JJ, Davis MD, Lynch KR, Macdonald TL.
`Synthesis of 4(5)-phenylimidazole-based analogues of
`sphingosine-1-phosphate and FTY720: discovery of potent
`S1P1 receptor agonists. Bioorg Med Chem Lett 2005: 15:
`3568.
`9. Kunzendorf U, Ziegler E, Kabelitz D. FTY720–the
`first compound of a new promising class of imunosup-
`pressive drugs. Nephrol Dial Transplant 2004: 19: 1677.
`10. Pinschewer DD, Ochsenbein AF, Odermatt B et al.
`FTY720 immunosuppression impairs effector T cell per-
`ipheral homing without affecting induction, expansion,
`and memory. J Immunol 2000: 164: 5761.
`11. Budde KL, Schmouder R, Nashan B et al. Pharmaco-
`dynamics of single doses of the novel immunosuppressant
`FTY720 in stable renal transplant patients. Am J Trans-
`plant 2003: 3: 846.
`12. Tedesco-Silva H, Mourad G, Kahan BD et al.
`FTY720, a novel immunomodulator: efficacy and safety
`results from the first phase 2A study in de novo renal
`transplantation. Transplantation 2004: 77: 1826.
`13. Brinkmann V, Davis MD, Heise CE et al. The immune
`modulator FTY720 targets sphingosine 1-phosphate re-
`ceptors. J Biol Chem 2002: 277: 21453.
`14. Kiuchi M, Adachi K, Kohara T et al. Synthesis and bio-
`logical
`evaluation of 2,2-disubstituted 2-aminoethan-
`ols:analogues of FTY720. Bioorg Med Chem Lett 1998: 8:
`101.
`15. Wang ME, Tejpal N, Qu X et al. Immunosuppressive
`effects of FTY720 alone or in combination with cyclo-
`sporine and/or sirolimus. Transplantation 1998: 65: 899.
`16. Suzuki S, Li XK, Enosawa S, Shinomiya T. A new im-
`munosuppressant, FTY720,
`induces
`bcl-2-associated
`apoptotic cell death in human lymphocytes. Immunology
`1996: 89: 518.
`17. Bo¨ hler T, Waiser J, Schu¨ tz M et al. FTY 720 Mediates
`reduction of lymphocyte counts in human renal allograft
`recipients by
`an apoptosis-independent mechanism.
`Transpl Int 2000: 13: 311.
`18. Chiba K, Yanagawa Y, Masubuchi Y et al. FTY720,
`a novel
`immunosuppressant,
`induces sequestration of
`circulating mature lymphocytes by acceleration of lym-
`

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