throbber
236
`
`Fingolimod Mitsubishi Pharma/Novartis
`Francis J Dumont
`
`Address
`421 Dogwood Avenue
`Egg Harbor Township
`NJ 08234
`USA
`Email: Francisj_dumont@yahoo.com
`
`IDrugs 2005 8(3):236-253
`© The Thomson Corporation ISSN 1369-7056
`
`Mitsubishi Pharma Corp and Novartis AG are developing
`fingolimod, an orally active
`immunosuppressant affecting
`lymphocyte re-circulation,
`for
`the potential prevention of
`transplant rejection and the treatment of autoimmune diseases,
`including multiple sclerosis. Fingolimod is a synthetic sphingosine
`analog that becomes phosphorylated in vivo and acts as a
`sphingosine-1-phosphate receptor agonist.
`
`Introduction
`Contemporary immunosuppressive therapies are largely
`unsatisfactory, which is in part due to the low therapeutic
`index of the two current mainstay immunosuppressants
`cyclosporin A (CsA) and tacrolimus. These drugs potently
`block lymphokine production by inhibiting calcineurin
`function during T-cell activation, but exert serious
`mechanism-based
`toxicity
`[371251],
`[402151],
`[505878].
`Significant adverse side effects also limit the utility of other
`drugs that suppress lymphocyte activation or proliferation
`at different
`levels,
`such as
`sirolimus, everolimus,
`leflunomide and mycophenolate mofetil (MMF) [371251],
`[483827]. The latter agents are useful in multi-drug regimens
`mitigating their own toxicity and that of calcineurin
`inhibitors or corticosteroids. However, there is a pressing
`need for immunosuppressants with novel modes of action
`and improved safety to provide for better prophylaxis of
`transplant rejection and more effective treatment of chronic
`autoimmune/inflammatory diseases (such as rheumatoid
`arthritis and multiple sclerosis (MS) [505878].
`
`researchers at
`(FTY-720), discovered by
`Fingolimod
`Yoshitomi Pharmaceutical Industries Ltd (now Mitsubishi
`Pharma Corp) [176944], [225279], may go some way toward
`fulfilling this need. While still not entirely elucidated, the
`mechanism of action of fingolimod appears to be quite
`unique since it reflects an alteration of the trafficking of
`lymphocytes rather than of their activation or proliferation
`[371332],
`[558907],
`[558908]. Numerous studies have
`demonstrated the ability of fingolimod to prolong allograft
`survival in rodents, dogs and non-human primates and to
`act synergistically with
`inhibitors of calcineurin or
`proliferation, without exerting major toxic effects [558907].
`The immunosuppressive activity of fingolimod was also
`established
`in various rodent models of autoimmune
`diseases
`[558907]. These encouraging preclinical data
`prompted the development of fingolimod, by Mitsubishi
`and Novartis AG, for potential use in transplant rejection
`and autoimmune diseases [522816], [538366]. Over recent
`years, phase I and II clinical trials in renal transplantation
`have provided preliminary evidence that fingolimod is well
`tolerated
`and
`efficacious when
`administered
`in
`
`
`
`Originator Mitsubishi Pharma Corp
`
`Licensee Novartis AG
`
`Status Phase III Clinical
`
`Indications Autoimmune disease, Cancer, Inflammatory
`bowel disease, Insulin-dependent diabetes, Multiple
`sclerosis, Myocarditis, Transplant rejection
`
`Actions Apoptosis inducer, Anticancer,
`Immunosuppressant, Lymphocyte trafficking modulator,
`Sphingosine kinase substrate, Sphingosine-1-phosphate
`receptor agonist
`
`Technology Oral formulation
`
`Synonym FTY-720
`Registry Nos: 162359-56-0, 162359-55-9
`
`
`
`
`
`OH
`
`OH
`
`NH2
`
`CH3
`
`
`
`
`combination with CsA [558910], [558912]. The preclinical
`and clinical studies described below were all carried out
`using oral delivery, unless otherwise specified.
`
`Synthesis and SAR
`Fingolimod is a synthetic sphingosine analog initially
`generated by the chemical modification of myriocin (ISP-1),
`a natural product from the ascomycete Isaria sinclairii
`[371535]. Myriocin, first described as an antifungal antibiotic
`in 1972 [371531], was re-discovered more than 20 years later
`as an immunosuppressive metabolite. Although potently
`immunosuppressive
`in vivo, myriocin
`caused
`fatal
`gastrointestinal toxicity [371535], and various synthetic
`derivatives were thus tested to identify a safer compound.
`Simplified
`2-alkyl-2-amino-1,3-propanediol
`structures
`demonstrated
`reduced
`toxicity while
`retaining
`immunosuppressive activity [176999], [186975], [225279],
`[226420]. Insertion of a phenyl ring into the alkyl side chain
`led to the development of fingolimod, which exhibited
`improved immunosuppressive activity and safety [176944],
`[225279]. Synthesis of additional analogs demonstrated that
`while the length of the hydrophobic alkyl chain is not
`critical, the position of the phenyl ring is highly important
`for activity, with the optimum length between the phenyl
`ring and the quaternary carbon being two carbon atoms
`[371349],
`[377090]. None of
`these analogs proved
`pharmacologically superior to fingolimod [371349], [377090].
`Of
`the
`two
`hydroxymethyl
`groups present
`in
`the hydrophilic portion of
`fingolimod, only
`the
`pro-S hydroxymethyl group appeared essential
`for
`immunosuppressive activity [371349], [377090]. Moreover,
`only the R-enantiomer configuration at the chiral carbon of a
`fingolimod analog was
`immunosuppressive
`[371349],
`[377090]. Procedures for the synthesis of fingolimod and its
`
`SUN - IPR2017-01929, Ex. 1018, p. 1 of 18
`
`

`

`Fingolimod Dumont 237
`
`chiral analogs and corresponding phosphates have been
`described [378422], [477725], [530769], [558913], [558914],
`[559658], [579915].
`
`Preclinical Development
`Although fingolimod was initially reported to potently
`inhibit mouse T-cell proliferation in mixed-lymphocyte
`cultures [176944], this was not confirmed in subsequent
`studies [558965]. At concentrations up to 1 µM, fingolimod
`did not substantially affect either proliferation or interleukin
`(IL)-2 production of antigen- or mitogen-stimulated rat
`[371352], [371354] or human T-cells [371356], nor did it
`inhibit IL-2-driven T-cell growth [371352]. In this respect, the
`action of fingolimod clearly differed from CsA and
`tacrolimus (which inhibit IL-2 production) [371338] and
`sirolimus (which
`inhibits IL-2-dependent proliferation)
`[371358]. Sub-micromolar concentrations of fingolimod
`nevertheless exerted a synergistic effect with CsA and
`sirolimus in suppressing T-cell proliferation in vitro [371356].
`At higher concentrations (> 4 to 5 µM), fingolimod alone
`induced apoptosis of mature T-cells [371360], [371362],
`thymocytes [371363] and non-lymphoid cells [371363],
`[371364], [558969], [558970]. Fingolimod also promoted
`apoptosis of
`lymphocytes
`[371365]
`and
`enhanced
`superantigen-mediated T-cell deletion in vivo [371368], but,
`as discussed below, it is highly improbable that such effects
`contribute to the mechanism of immunosuppressive action
`of fingolimod.
`
`Despite its weak immunosuppressive activity in vitro,
`fingolimod proved to be a potent immunosuppressant in
`rodent models of graft rejection. Administration of the
`compound at > 0.1 mg/kg dose-dependently prolonged the
`survival of skin [212189], [242515], heart [212190] and liver
`[371356] allografts in rats. Fingolimod was efficacious when
`administered at 0.5 or 1 mg/kg for 2 to 5 weeks in rats with
`small bowel allotransplant known to elicit a strong rejection
`response [371371], [558982], [558983]. At higher doses
`(3 mg/kg),
`fingolimod significantly augmented
`limb
`[371369] and
`joint [371370] allograft survival in rats.
`Immunosuppression with fingolimod also protected corneal
`allograft from rejection [558984], [558985], and promoted
`long-term pancreatic islet allograft survival and function
`[371506], [477727], [477833] in mice and rats. In cardiac
`transplantation models, fingolimod not only prolonged the
`survival of the allograft [371372], but also reduced the
`development of graft atherosclerosis associated with chronic
`rejection [371372], [558986]. In these studies, fingolimod
`treatment was usually
`initiated
`the day before
`transplantation and continued for several weeks thereafter.
`In some instances fingolimod was also effective when
`administered only for 2 days, either from the time of
`transplantation of heart [371352] or liver [371377] allograft,
`or before transplantation of kidney allograft [371378].
`Moreover, fingolimod prolonged liver allograft survival in
`rats if given at 5 mg/kg only on days 3 and 4 post-
`transplantation [371377]. This dose also reversed ongoing
`acute rejection of cardiac allograft if administered on day 3
`to 7 post-transplantation in mice [558987]. Fingolimod
`treatment (0.5 mg/kg/day), delayed to 20 weeks after
`transplantation, ameliorated chronic allograft nephropathy
`induced by CsA (1.5 mg/kg/day) in a renal transplantation
`model [558988]. However, fingolimod failed to prolong skin
`
`allograft survival in rats when administered from day 4
`post-transplantation
`[378421], and overall,
`fingolimod
`proved more potent if given before transplantation, rather
`than post-operatively only. Importantly, fingolimod was
`also active in transplantation models in larger species. At a
`dose of 5 mg/kg, fingolimod, administered for only 2 days
`prior to transplantation, delayed the rejection of renal
`allograft in dogs [371377]. Chronic treatment at lower doses
`also prolonged the survival of renal and liver allografts in
`dogs [233473], [371379], [371380], [584577]. Furthermore,
`once-daily administration of fingolimod at a dose of
`3 mg/kg/day, initiated at least 2 days before transplantation
`and continued thereafter, extended renal allograft survival
`by 33 to 85 days in cynomolgus monkeys [477762].
`
`In addition to benefits derived from lone administration,
`fingolimod displayed
`strong
`synergy with
`other
`immunosuppressive agents
`in various
`transplantation
`models. This was first demonstrated for skin allograft in
`rats, and cardiac allograft in rats and dogs, in which low
`doses of fingolimod potentiated the effect of sub-therapeutic
`doses of CsA [212189], [212190], [212191]. Subsequent
`studies replicated this observation in rat models of skin,
`cardiac, small bowel or liver transplantation [371356],
`[371390], [371391], [371394], [477824] and in mouse models
`of
`cardiac
`transplantation
`[371372],
`[477825]. The
`combination of fingolimod with CsA proved highly effective
`in rat models of small bowel transplantation, where it
`prevented graft rejection and graft-versus-host reaction
`[371371], as well as cardiac transplantation, where it
`abrogated chronic rejection [558986]. Fingolimod plus CsA
`treatment prevented graft vessel disease in a rat carotid
`artery transplantation model [477821], [477831]. Moreover,
`fingolimod (1 or 3 mg/kg) administered every day or every
`other day in combination with CsA (15 mg/kg) inhibited the
`rejection of porcine islet xenografts in rats, while treatment
`with either drug alone was ineffective [558992]. Synergistic
`effects between
`fingolimod and CsA were
`further
`documented in canine models of kidney [371377], [371392],
`[371396] and liver [371380] allotransplantation. Similarly,
`fingolimod (0.1 to 0.3 mg/kg/day) given intravenously or
`orally synergized with sub-therapeutic doses of CsA (10 to
`30 mg/kg/day) to markedly prolong renal allograft survival
`in cynomolgus monkeys [371394], [477762]. Rejection-free
`graft survival was extended to between 32 and 101 days
`with this low-dose, combined-treatment regimen [477762].
`Fingolimod was also demonstrated to synergize with low
`doses of tacrolimus in preventing rejection of skin [371398],
`heart [371398], [371401] and liver [371403] allografts in rats.
`Furthermore, the combination of fingolimod (5 mg/kg) with
`tacrolimus (1 mg/kg) significantly improved survival of
`rat-to-hamster skin xenografts [371404]. However, in a liver
`transplantation model
`in dogs,
`the combination of
`fingolimod (0.1 mg/kg) with tacrolimus (0.5 mg/kg) was
`less effective than tacrolimus alone and caused mortality
`from
`infectious
`complication
`due
`to
`over-
`immunosuppression [371380]. This suggested that careful
`dose adjustment would be needed if fingolimod and
`tacrolimus were to be used together in clinical regimens.
`Fingolimod was further demonstrated to exert synergistic
`effects when administered with immunosuppressants other
`than calcineurin inhibitors. For example, the combination of
`fingolimod with sirolimus or everolimus resulted in potent
`
`SUN - IPR2017-01929, Ex. 1018, p. 2 of 18
`
`

`

`[559040],
`[477724],
`(EAE)
`[477832], encephalomyelitis
`[559041], [559042] and type 1 diabetes [371388], [371411],
`[371433],
`[492753],
`[559045].
`Chronic
`fingolimod
`administration prevented the spontaneous development of
`autoimmune diabetes in NOD mice [559046], [559047] and
`slowed the progression of systemic lupus erythematosus-
`like syndrome in MRL-lpr/lpr mice [559048]. It must be
`noted that fingolimod treatment generally needed to be
`initiated before, or at the time of disease induction, to be
`effective in these models. However, the pathology of EAU
`could still be significantly reduced when the drug was
`administered after disease onset [371444], and in the case of
`thyroiditis induced by neonatal thymectomy and irradiation,
`fingolimod significantly reversed ongoing autoimmune
`disease [371438]. Furthermore, administration of fingolimod
`(3 mg/kg/day ip) to SJL mice with established relapsing-
`remitting EAE, a chronic disease
`that mimics
`the
`predominant form of human MS, resulted in a rapid and
`sustained improvement in the clinical status of the mice,
`which was maintained as long as dosing was continued
`[559042]. Similarly, in IL-10 gene knockout mice, a model of
`inflammatory bowel disease,
`fingolimod significantly
`decreased the severity of colitis when administered for
`4 weeks after disease onset [559051]. While such data
`suggest a role for fingolimod monotherapy in the treatment
`of autoimmunity,
`it appears probable
`that, as
`for
`transplantation, the drug may be of greater utility when
`combined with other immunosuppressive agents. This
`possibility has not been explored in the studies published so
`far, but is suggested in patent application WO-2004028521
`(described below).
`
` A
`
` striking feature of the in vivo action of fingolimod,
`invariably observed in the aforementioned studies, was the
`induction (at immunosuppressive doses) of a marked
`decrease in the number of peripheral blood lymphocytes
`(PBLs). For example, a single-dose administration of
`fingolimod (0.1 mg/kg) in rats reduced PBL counts by
`> 90% between 3 and 24 h, with a return to baseline level
`within a week [371354], [396003]. In baboons or cynomolgus
`monkeys receiving fingolimod (0.1 or 0.3 mg/kg/day),
`peripheral lymphopenia occurred as soon as 4 h after
`treatment, reaching 60 to 80% by 24 to 48 h [371447],
`[417322]. This effect was somewhat more rapid and
`pronounced on T-cells than B-cells, with CD4+ cells being
`more greatly reduced than CD8+ cells [371354], [371447],
`[417322]. In cynomolgus monkeys that were chronically
`treated with fingolimod, PBL counts decreased to ~ 30 and
`14% of pretreatment values at doses of 0.03 and 3.0
`mg/kg/day, respectively and only ~ 4% of peripheral CD4+
`T-cells were refractory to depletion by the drug, compared
`with ~ 30% for CD8+ T-cells [477762]. As a correlate of this
`peripheral lymphopenia, fingolimod reduced the infiltration
`of allografts by T-cells [371483], [371489], [558982], [558985],
`especially
`if
`the drug was administered before
`this
`infiltration occurred [378421]. Significantly diminished T-cell
`infiltration of autoimmune disease target organs was
`similarly documented along with PBL depletion in animal
`models of autoimmunity,
`following
`treatment with
`fingolimod [371388], [559040], [559047]. Interestingly, in the
`transplantation studies, the few T-cells present in the grafts
`of fingolimod-treated animals expressed IL-2 and interferon
`(IFN)γ mRNA [371483], whereas these cytokines were
`
`238 IDrugs 2005 Vol 8 No 3
`
`suppression of allograft rejection in rats [371356], [396003],
`mice [477828] and monkeys [477762], [558993]. In the latter
`model,
`the
`triple-daily
`combination of
`fingolimod
`(0.1 mg/kg)/CsA (10 mg/kg)/everolimus (0.25 mg/kg)
`resulted in further increased graft survival (from 47 to > 100
`days) compared with either drug given alone or in double
`combination [477762]. The co-administration of a low, non-
`toxic dose of mycophenolate sodium (10 mg/kg/day) with
`low doses of fingolimod (0.03 or 0.1 mg/kg/day) was also
`synergistic and prolonged heart allograft survival in rats
`[477726], [477728]. Moreover, fingolimod synergized with
`the blockade of CD28-mediated T-cell co-stimulation by
`CTLA-4-immunoglobulin (Ig) to prevent cardiac allograft
`rejection [559003] or obliterative bronchiolitis in tracheal
`transplantation [559005].
`
`Fingolimod facilitated the induction of tolerance to allografts
`in experimental systems involving the administration of
`allochimeric class I major histocompatibility complex (MHC)
`antigen [371409], or intrathymic injection of donor splenic
`cells [371505], in rats. In contrast, fingolimod prevented
`tolerance induction by donor-specific blood transfusion in
`intestinal transplantation [396001], [559007] or by an anti-
`CD4 mAb in a rat kidney transplantation model [559008].
`However, establishment of transplant tolerance was not
`influenced by fingolimod co-treatment in other models
`[559007], [559009].
`
`Several studies revealed that fingolimod may help alleviate
`grafted organ damage due to ischemia-reperfusion (IR)
`injury, a significant problem in clinical transplantation. This
`was observed in rat models involving cold preservation of
`kidney graft in which fingolimod treatment of recipients,
`either
`immediatedly (1 mg/kg
`iv) [559019] or 24 h
`(0.5 mg/kg po) [559026] prior to reperfusion ameliorated the
`morphological and
`functional consequences of post-
`transplant IR injury. A protective role of fingolimod
`(1 mg/kg iv) was also suggested in renal IR injury models in
`mice [409000], [559032]. Similarly, fingolimod pretreatment
`diminished the biochemical and histological manifestations
`of tissue injury in rat models of warm hepatic IR [477817],
`[559036], although an earlier study reported that such a
`treatment may aggravate IR-induced liver injury [371381].
`More recently, in a rat model of segmental hepatic ischemia,
`fingolimod (1 mg/kg iv) prevented hepatocyte apoptosis
`and decreased the acute phase inflammatory response in
`both normal and cirrhotic livers when administered 20 min
`before ischemia and 10 min before reperfusion [584578].
`
`Fingolimod inhibited various other T-cell-mediated immune
`responses in rodents, in addition to transplant rejection.
`These
`included graft-versus-host
`reactions
`[212192],
`[474285],
`contact
`allergy
`[477810],
`delayed-type
`hypersensitivity [371388], acute viral myocarditis [559576]
`and airway inflammation induced by adoptive transfer of
`Th1 or Th2 cells [530768]. Fingolimod prevented the
`spontaneous development of dermatitis in NC/Nga mice, a
`model for human atopic dermatitis [559575]. Importantly,
`fingolimod proved efficacious at suppressing several
`experimentally induced autoimmune diseases in mice or
`rats,
`including myocarditis
`[371443],
`experimental
`autoimmune uveoretinitis
`(EAU)
`[371444],
`thyroiditis
`[371438], collagen- and adjuvant-induced arthritis [371538],
`
`SUN - IPR2017-01929, Ex. 1018, p. 3 of 18
`
`

`

`Fingolimod Dumont 239
`
`[559497], [559499], which are known to play prominent roles
`in lymphocyte homing to secondary lymphoid organs
`[559052]. Although other chemokine receptors, such as
`CCR2 and CXCR4, may participate in the action of
`fingolimod [431191], [559502], [559577], further observations
`indicated
`that
`fingolimod-induced
`lymphocyte
`sequestration resulted from an inhibition of lymphocyte
`emigration from LN and PP rather than from an enhanced
`attraction to these organs [558908]. In this respect, it is worth
`noting that lymphocyte re-circulation to the blood requires
`lymphocytes to enter the thoracic duct lymph (TDL) after
`their transit through secondary lymphoid organs [371457],
`[559052]. Interestingly, fingolimod treatment decreased
`lymphocyte counts in the TDL to a greater extent than in the
`blood
`[371354]. Histological analyses of LN
`from
`fingolimod-treated mice revealed an accumulation of
`lymphocytes on the abluminal side of the lymphatic
`endothelium, along with an emptying of lymphatic sinuses,
`indicating that lymphocyte egress into lymph was blocked
`[558965], [559503]. This may result in an inhibition of both
`the re-circulation of naïve T-cells and the release of antigen-
`activated T-cells from the draining lymph node to lymph
`and to the blood compartment [558965]. In addition,
`fingolimod inhibited the passage of mature T-cells from the
`thymus into blood [371477], [559504].
`
` A
`
` breakthrough in understanding the pharmacological
`effects of fingolimod came from the discovery that the drug
`is rapidly phosphorylated in vivo. Furthermore, the resulting
`phosphorylated
`fingolimod
`(fingolimod-P)
`inhibited
`lymphocyte re-circulation and acted as a potent agonist on
`several members of the sphingosine-1-phosphate (S1P)
`receptor family, namely S1P1, S1P3, S1P4 and S1P5 [477724],
`[559503]. Upon binding their natural sphingolipid ligands,
`such cell-surface G protein-coupled receptors have been
`reported to elicit a variety of responses in diverse cell types
`[559505], [559507]. S1P1 receptors, which are expressed on
`lymphocytes and endothelial cells [559508], appear to be the
`most
`important S1P
`receptors with
`regard
`to
`the
`immunosuppressive action of fingolimod. This was first
`suggested by structure-activity analyses of semi-selective
`S1P receptor agonists [559509], [559510]. Moreover, a potent
`S1P1-selective agonist, structurally unrelated to S1P and
`fingolimod-P,
`induced peripheral
`lymphopenia
`by
`preventing the entry of lymphocytes into lymph, in a
`manner similar to fingolimod [559511]. A role for S1P1 in
`lymphocyte trafficking was further demonstrated by the
`observation that mice
`lacking this receptor on their
`lymphocytes exhibited an almost complete absence of
`T-cells, and severe deficiency of B-cells, in their blood
`[559513]. Moreover, S1P1-negative T-
`and B-cells,
`transferred to a normal host, accumulated in secondary
`lymphoid organs from which they failed to exit [559513]. A
`study conducted with knockout mice revealed that selective
`deletion of S1P1 in T-cells produced a block in the egress of
`mature T-cells from the thymus into the periphery [559515].
`Most importantly, exposure of normal lymphocytes to
`fingolimod-P in vivo or in vitro downregulated expression of
`S1P1 through internalization and rapid degradation, thereby
`inducing an S1P1-negative phenotype [559513], [559520].
`This implied that fingolimod-P behaves as a partial S1P1
`agonist in lymphocytes, rather than a full agonist as
`originally thought [477724], [559503]. Therefore, expression
`
`tacrolimus-treated
`in CsA- or
`markedly suppressed
`recipients [371403], [371483]. The combination of fingolimod
`with either CsA or tacrolimus abrogated both the T-cell
`infiltration and cytokine mRNA expression in the graft
`[371403], [371483], which may account for the synergism in
`the graft protection mentioned above. Furthermore, studies
`with fingolimod analogs revealed that their ability to cause
`lymphopenia correlated well with
`their efficacy
`in
`promoting rat skin allograft survival [377090]. There was
`also a close correlation between the degree of circulating
`lymphocyte depletion and heart allograft survival in rats
`treated with low doses of fingolimod (0.01 to 0.1 mg/kg) in
`conjunction with everolimus [396003]. This further suggests
`that lymphopenia and the associated reduction of graft
`infiltrating
`lymphocytes play a crucial role
`in
`the
`immunosuppressive effect of fingolimod.
`
`It was initially proposed that the lymphopenia caused by
`fingolimod reflected apoptotic cell death [371360], [371452],
`[371507]. However,
`the
`blood
`concentrations
`of
`immunosuppressive doses of fingolimod proved to be
`> 2-fold lower than those required to induce apoptosis
`[371354]. In addition, apoptotic cells could not be detected in
`the PBL of baboons following treatment with low doses of
`fingolimod [371447]. Similarly, apoptosis rates were not
`increased in the PBL of patients receiving fingolimod
`[438642]. In mice, dye-labeled lymphocytes that had been
`depleted from the blood after fingolimod treatment (0.3
`mg/kg) reappeared after cessation of treatment [371454],
`and there was no evidence for deletion of antiviral memory
`cells by the drug [371388]. Moreover, an S-enantiomer
`analog of fingolimod, which did not induce lymphopenia,
`proved
`as
`potent
`as
`the
`lymphopenia-inducing
`R-enantiomer analog in causing lymphocyte apoptosis in
`vitro [477818]. Therefore, lymphocyte apoptosis is unlikely to
`play a significant role in fingolimod-induced lymphopenia.
`
`An alternative and more plausible explanation for the
`lymphopenic effect of fingolimod was provided by the
`finding that, concomitant with a reduction of PBL numbers,
`the drug increased lymphocyte numbers in the peripheral
`and mesenteric lymph nodes (LN) and in Peyer's patches
`(PP), but not in spleen [371354], [371455], [371473]. This
`suggested
`that
`fingolimod altered
`the
`trafficking of
`lymphocytes such that they became sequestered in LN and
`PP. One possibility could be that fingolimod accelerated the
`homing of lymphocytes to these tissues, a process known to
`involve both
`specialized
`adhesion molecules
`and
`chemokines [371457], [559052]. While antibodies directed
`against lymphocyte homing molecules such as CD62L,
`CD49d and CD11a interfered with fingolimod-induced
`lymphocyte sequestration, expression of these molecules
`was not altered by the drug [371354], [371462]. Furthermore,
`evidence was obtained
`that
`fingolimod
`can
`act
`independently of CD62L [559481]. An augmentation of
`lymphocyte responses to homing chemokines was also
`postulated
`to mediate fingolimod-induced
`lymphocyte
`sequestration
`in LN and PP [371332], [477822]. This
`hypothesis was based on the observation that nanomolar
`concentrations of the drug stimulated T-cell chemotaxis to
`certain chemokines in vitro [559484]. However, fingolimod
`proved capable of producing lymphocyte sequestration in
`mice lacking the chemokine receptors CCR7 and CXCR5
`
`SUN - IPR2017-01929, Ex. 1018, p. 4 of 18
`
`

`

`in rats, monkeys and humans, giving rise to the biologically
`active
`fingolimod-P metabolite
`[477724],
`[559503].
`Phosphorylation of fingolimod also occurred
`in cells
`incubated with the compound [559503], and the MDR-1
`multidrug transporter appeared to mediate the efflux of
`fingolimod-P from cells [559545]. Moreover, fingolimod-P
`was
`detected
`in
`plasma
`following
`intravenous
`administration of fingolimod in rats, and of the two chiral
`forms of a fingolimod analog, the R-enantiomer was readily
`phosphorylated, whereas the S-enantiomer exhibited only
`trace phosphorylation in rat blood [477724], [559503], which
`was
`consistent with
`the R-enantiomer being
`the
`immunosuppressive isomer [377090], [477818]. Following
`oral administration of fingolimod, the blood level of
`fingolimod-P exceeded that of the parent compound by up
`to 4-fold [477724]. Fingolimod was phosphorylated in vitro
`by both sphingosine kinase type 1 (SPHK1) and type 2
`(SPHK2), but SPHK2 was more effective, suggesting that it
`may be the relevant enzyme in vivo [530765], [559546],
`[585265]. While phosphorylation of fingolimod appeared
`reversible in vivo [477724], the compound was irreversibly
`metabolized by hepatic oxidation
`to carboxylic acid
`derivatives devoid of immunosuppressive activity that were
`excreted in urine and feces [371344], [413804]. A study using
`human liver microsomes suggested that CYP4F3, or another
`closely related form of P450 enzyme, was the primary
`catalyst of fingolimod oxidation, which resulted in the
`formation of two metabolic products [371490]. Since none of
`the major drug-metabolizing P450 enzymes appear to be
`involved in this metabolism, interactions of fingolimod with
`potential co-medications such as CsA,
`tacrolimus or
`sirolimus are unlikely. This is consistent with earlier
`findings in dogs [371377], and with data from human
`studies that demonstrated the lack of pharmacokinetic or
`pharmacodynamic cross-interference between fingolimod
`and CsA [559547], [559548]. However, a potentiation of
`fingolimod exposure by CsA co-administration was noted in
`cynomolgus monkeys [417322].
`
` A
`
` pharmacokinetic study in baboons revealed that when
`administered as a single oral dose, fingolimod (0.3 mg/kg)
`displayed a Cmax of 2.16 ng/ml, an AUC(0-48 h) of 77.9 ng.h/ml
`and a t1/2 of 36 ± 12 h. Upon repeated dosing at 0.03 mg/kg,
`the compound accumulated over time to reach a stable
`blood concentration (0.72 ng/ml) by days 7 to 9 [371447]. In
`cynomolgus monkeys treated with fingolimod as a single
`dose either orally (0.1 or 1 mg/kg) or intravenously
`(0.1 mg/kg),
`a
`linear
`three-compartment model
`characterized the time course of fingolimod concentrations
`with a t1/2 of ~ 31 h, a Cl of ~ 0.53 l/h/kg and a
`bioavailability of ~ 38% [448400]. This long terminal t1/2 most
`likely reflected extensive tissue distribution and binding of
`the compound, particularly as the steady state Vd of the
`drug was also notably large (Vdss = 15 l/kg) [448400].
`
`The pharmacokinetics of fingolimod (0.25 to 3.5 mg) were
`analyzed following the administration of single doses in
`renal transplant patients (n = 20) [477737]. Fingolimod
`displayed a prolonged absorption phase with a Tmax of 12 to
`24 h and an elimination t1/2 ranging from 89 to 157 h, which
`was dose-independent. The Cmax (0.16 to 2.8 ng/ml) and
`AUC (28 to 434 ng.h/ml) were proportional to the dose,
`with low interpatient variability. Moreover, fingolimod
`
`240 IDrugs 2005 Vol 8 No 3
`
`of functional S1P1 by lymphocytes appears to be required
`for their egress from the thymus and secondary lymphoid
`organs. The egress-blocking effects of fingolimod and
`consequent depletion of T- and B-cells from the circulation
`may thus result from the inactivation of this receptor by the
`fingolimod-P metabolite. In addition, the downregulation of
`S1P1 by fingolimod on marginal zone B-cells, a unique
`subset of sessile B-cells with a partially activated phenotype,
`was shown to cause their rapid relocalization to lymphoid
`follicles [559540]. This indicated that the action of the drug
`may extend beyond a blockade of lymphocyte egress and
`affect lymphoid tissue compartmentalization. Fingolimod
`was further demonstrated to suppress the antibody response
`to a T-dependent antigen by inhibiting the formation of
`germinal centers in peripheral lymphoid tissues of mice
`[559543].
`
`Evidence was obtained that fingolimod may also alter the
`barrier function of the vascular endothelium. Fingolimod-P
`(10 nM) induced calcium mobilization and MAP kinase
`activation, and promoted survival and adherens junction
`assembly in endothelial cells in vitro [515624], [530766],
`[559544]. Consistent with these observations in vitro, the
`enhanced vascular permeability
`for macromolecules
`induced by vascular endothelial growth factor (VEGF) in
`vivo was inhibited by fingolimod treatment (10 µg) in mice
`[530766]. Whether such an effect contributes
`to
`the
`aforementioned ability of the drug to attenuate graft vessel
`disease [477831] and IR injury [559036] remains to be
`investigated.
`
`Fingolimod was also demonstrated to exert antitumor
`effects, albeit through still poorly defined mechanisms. At
`micromolar concentrations in vitro, the drug inhibited
`proliferation and promoted the apoptosis of various human
`or mouse tumor cell lines, including glioma [558969],
`bladder cancer [558970] breast cancer [452318], [585263],
`prostate cancer [585260], [585264], hepatoma [514900],
`[515624], [585261] and myeloma [574214], while affecting
`normal cell counterparts to a lesser extent. Tumor cell death
`induced by fingolimod may be due to activation of
`pro-apoptotic signaling pathways [585263], [585264], and/or
`inhibition of anti-apoptotic pathways,
`such as
`the
`phosphoinositide 3-kinase/Akt pathway [585261], but there
`is no evidence that this could be mediated via S1P receptors.
`An intriguing possibility, which remains to be confirmed, is
`that high concentrations of
`fingolimod may
`inhibit
`sphingosine ki

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