18
Development of a sea anemone toxin as an immunomodulator for therapy of autoimmune diseases Victor Chi a , Michael W. Pennington c , Raymond S. Norton d , Eric J. Tarcha e , Luz M. Londono e , Brian Sims-Fahey e , Sanjeev K. Upadhyay a , Jonathan T. Lakey b , Shawn Iadonato e , Heike Wulff f , Christine Beeton g , K. George Chandy a, * a Department of Physiology and Biophysics, UC Irvine, Irvine, CA 92697, USA b Department of Surgery, UC Irvine, Irvine, CA 92697, USA c Peptides International, 11621 Electron Drive, Louisville, KY 40299, USA d Medicinal Chemistry and Drug Action, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville 3052, Australia e Kineta Inc., 307 Westlake Ave North, Seattle, WA 98199, USA f Department of Pharmacology, UC Davis, Davis, CA 95616, USA g Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA article info Article history: Received 7 April 2011 Received in revised form 16 June 2011 Accepted 20 July 2011 Available online 12 August 2011 Keywords: Autoimmune ShK BgK Sea anemone Toxin T cell B cell Kv1.3 KCa3.1 Potassium channel CRAC Multiple sclerosis Rheumatoid arthritis Type-1 diabetes mellitus ShK-186 PAP-1 Psoriasis Transplant ShK-192 abstract Electrophysiological and pharmacological studies coupled with molecular identication have revealed a unique network of ion channelsdKv1.3, KCa3.1, CRAC (Orai1 þ Stim1), TRPM7, Cl swell din lymphocytes that initiates and maintains the calcium signaling cascade required for activation. The expression pattern of these channels changes during lymphocyte activation and differentiation, allowing the functional network to adapt during an immune response. The Kv1.3 channel is of interest because it plays a critical role in subsets of T and B lymphocytes implicated in autoimmune disorders. The ShK toxin from the sea anemone Stichodactyla helianthus is a potent blocker of Kv1.3. ShK-186, a synthetic analog of ShK, is being developed as a therapeutic for autoimmune diseases, and is scheduled to begin rst-in-man phase-1 trials in 2011. This review describes the journey that has led to the development of ShK-186. Ó 2011 Elsevier Ltd. All rights reserved. Ion channels were discovered in the immune system in 1984 when it became possible to record electrical signals from single lymphocytes (DeCoursey et al., 1984; Matteson and Deutsch, 1984). It is now clear that ve types of ion * Corresponding author. Department of Physiology and Biophysics, UC Irvine, Room 291 Irvine Hall, Irvine, CA 92697, USA. Tel.: þ1 949 824 7435; fax: þ1 949 824 3143. E-mail address: [email protected] (K.G. Chandy). Contents lists available at SciVerse ScienceDirect Toxicon journal homepage: www.elsevier.com/locate/toxicon 0041-0101/$ see front matter Ó 2011 Elsevier Ltd. All rights reserved. doi:10.1016/j.toxicon.2011.07.016 Toxicon 59 (2012) 529546

Development of a sea anemone toxin as an immunomodulator for therapy of autoimmune diseases

Embed Size (px)

Citation preview

e at SciVerse ScienceDirect

Toxicon 59 (2012) 529–546

Contents lists availabl

Toxicon

journal homepage: www.elsevier .com/locate/ toxicon

Development of a sea anemone toxin as an immunomodulatorfor therapy of autoimmune diseases

Victor Chi a, Michael W. Pennington c, Raymond S. Norton d, Eric J. Tarcha e, Luz M. Londono e,Brian Sims-Fahey e, Sanjeev K. Upadhyay a, Jonathan T. Lakey b, Shawn Iadonato e,Heike Wulff f, Christine Beeton g, K. George Chandy a,*

aDepartment of Physiology and Biophysics, UC Irvine, Irvine, CA 92697, USAbDepartment of Surgery, UC Irvine, Irvine, CA 92697, USAc Peptides International, 11621 Electron Drive, Louisville, KY 40299, USAdMedicinal Chemistry and Drug Action, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville 3052, AustraliaeKineta Inc., 307 Westlake Ave North, Seattle, WA 98199, USAfDepartment of Pharmacology, UC Davis, Davis, CA 95616, USAgDepartment of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA

a r t i c l e i n f o

Article history:Received 7 April 2011Received in revised form 16 June 2011Accepted 20 July 2011Available online 12 August 2011

Keywords:AutoimmuneShKBgKSea anemoneToxinT cellB cellKv1.3KCa3.1Potassium channelCRACMultiple sclerosisRheumatoid arthritisType-1 diabetes mellitusShK-186PAP-1PsoriasisTransplantShK-192

* Corresponding author. Department of PhysiologIrvine, Room 291 Irvine Hall, Irvine, CA 92697, USA. Tfax: þ1 949 824 3143.

E-mail address: [email protected] (K.G. Chandy).

0041-0101/$ – see front matter � 2011 Elsevier Ltddoi:10.1016/j.toxicon.2011.07.016

a b s t r a c t

Electrophysiological and pharmacological studies coupled with molecular identificationhave revealed a unique network of ion channelsdKv1.3, KCa3.1, CRAC (Orai1 þ Stim1),TRPM7, Clswelldin lymphocytes that initiates and maintains the calcium signaling cascaderequired for activation. The expression pattern of these channels changes duringlymphocyte activation and differentiation, allowing the functional network to adapt duringan immune response. The Kv1.3 channel is of interest because it plays a critical role insubsets of T and B lymphocytes implicated in autoimmune disorders. The ShK toxin fromthe sea anemone Stichodactyla helianthus is a potent blocker of Kv1.3. ShK-186, a syntheticanalog of ShK, is being developed as a therapeutic for autoimmune diseases, and isscheduled to begin first-in-man phase-1 trials in 2011. This review describes the journeythat has led to the development of ShK-186.

� 2011 Elsevier Ltd. All rights reserved.

y and Biophysics, UCel.: þ1 949 824 7435;

. All rights reserved.

Ion channels were discovered in the immune system in1984 when it became possible to record electrical signalsfrom single lymphocytes (DeCoursey et al., 1984; Mattesonand Deutsch, 1984). It is now clear that five types of ion

V. Chi et al. / Toxicon 59 (2012) 529–546530

channelsdthe potassium channels Kv1.3 and KCa3.1, theCa2þ-release activating Ca2þ (CRAC) channel encoded bythe Orai and STIM1 (stromal interacting protein 1) genes,TRPM7 involved in magnesium homeostasis, and Clswell(swelling-activated chloride channel)dconstitutea network in T lymphocytes that is vital for cellularhomeostasis, activation and differentiation (Cahalan andChandy, 2009). The coalescence of Kv1.3, KCa3.1 andCRAC channels at the immunological synapse (Beeton et al.,2006; Lioudyno et al., 2008; Nicolaou et al., 2007; Panyiet al., 2004) during antigen presentation has the potentialto generate local ionic accumulation or depletion, and tomediate trans-synaptic signaling by assembling intomolecular aggregates or signalosomes (Cahalan andChandy, 2009). These channels also regulate the Ca2þ

signaling required for lymphocyte activation by maintain-ing a balance between Ca2þ influx and Kþ efflux (Cahalanand Chandy, 2009). Of particular interest is the Kv1.3channel, which plays a critical functional role in effector-memory (TEM) cells and class-switched memory B cellsthat are implicated in diverse autoimmune diseases(Beeton et al., 2006; Wulff et al., 2003, 2004). Potent andselective channel blockers of Kv1.3 have been developed,which are effective in diverse animal models of immuno-logical disorders (Beeton et al., 2005, 2006; Norton et al.,2004; Pennington et al., 2009; Schmitz et al., 2005; Wulffand Pennington, 2007).

1. The clinical problem – autoimmune diseases

Nearly 80 different autoimmune disorders are known,affecting more than 125 million people worldwide. Auto-immune diseases involve virtually every organ system inthe body including joints (e.g. rheumatoid arthritis [RA],ankylosing spondylitis), the central nervous system(multiple sclerosis [MS]), endocrine organs (type-1 dia-betes mellitus [T1DM], Hashimoto’s thyroiditis)(Leyendeckers et al., 2002) and skin (psoriasis). Tissuedestruction is mediated by autoreactive (self-reactive)immune cells. The frequency of autoreactive lymphocytes(e.g. against myelin antigens in the central nervous system)is the same in healthy individuals as in patients withautoimmune diseases. However, healthy individuals do notdevelop autoimmune diseases because these potentiallyself-destructive cells are suppressed and maintained ina quiescent naïve state by regulatory T cells. Once anautoreactive T lymphocyte is triggered to proliferate and/orescapes regulation, the presence of the autoantigen in thebody causes the cell to undergo repeated stimulation untilit changes into a terminally differentiated cell called a TEM-effector, which contributes to tissue damage. Disease-associated autoreactive T cells in patients with MS(specific for myelin antigens), T1DM (specific for insulinand GAD65 antigens), RA (synovial T cells) or psoriasis areTEM-effector cells (Beeton et al., 2006; Fasth et al., 2004;Friedrich et al., 2000; Lovett-Racke et al., 1998; Miyazakiet al., 2008; Rus et al., 2005; Viglietta et al., 2002; Visserset al., 2004; Wulff et al., 2003). Autoreactive B cells simi-larly differentiate upon repetitive autoantigen stimulationinto class-switched memory B cells, which are implicatedin MS (Corcione et al., 2004), Hashimoto’s thyroiditis

(Leyendeckers et al., 2002), Sjorgen’s syndrome (Hansenet al., 2002), and systemic lupus erythematosis (Dornerand Lipsky, 2004; Jacobi et al., 2003). A therapeuticapproach that mutes or eliminates TEM-effectors and class-switched memory B cells without compromising theprotective immune response mediated by other lymphoidsubsets would have significant advantages over currenttherapies that broadly suppress the entire immuneresponse.

2. Why target KD channels in immune cells?

2.1. Kþ channels promote calcium influx in lymphocytes

Calcium signaling is essential for lymphocytes to acti-vate, synthesize and secrete cytokines (or antibodies),migrate in vivo, and proliferate. In the case of Tcells, antigenbinding to the T-cell receptor causes the generation of IP3,which releases Ca2þ from the ER Ca2þ store (Fig. 1).Depletion of the ER store triggers an ER protein calledSTIM1 to cluster under the plasma membrane and activateCa2þ influx through CRAC channels formed from Orai1subunits (Lioudyno et al., 2008; Luik et al., 2008; Park et al.,2009; Penna et al., 2008). The influx of Ca2þ raises thecytosolic Ca2þ concentration from a resting value of 50–100 nM into the low micromolar range (Fig. 1). Ca2þ influxis sustained by the counterbalancing efflux of Kþ throughKv1.3 or KCa3.1 (Fig. 1). The relative contribution of Kv1.3and KCa3.1 varies according to their relative expressionlevel, which changes depending on the state of differenti-ation and activation of lymphocytes (Cahalan and Chandy,2009; Chandy et al., 2004). This differential Kþ channelexpression pattern allows Ca2þ signaling to be targetedspecifically in different lymphocyte subsets.

2.2. Kv1.3 and KCa3.1 are differentially expressed duringlymphocyte activation and differentiation

The activation and differentiation scheme for human Tand B lymphocytes is shown in Fig. 2. Naïve CD4þ or CD8þ

human T cells differentiate into long-lived central memory(TCM) T cells (main memory pool), which then differentiateinto TEM cells on repeated stimulation. T cells at each stageactivate into effectorsdnaïve-effector, TCM-effector, TEM-effectordwhen they encounter antigen. CCR7, a chemokinereceptor required for homing to lymph nodes, is expressedon the cell surface of naïve and TCM cells as well as theirrespective effectors, whereas, TEM cells and TEM-effectorsdo not express CCR7 (Sallusto et al., 1999). Therefore, wedivide human Tcells into CCR7þ (naïve and TCM) and CCR7�

(TEM) subsets. Human B cells undergo a similar differenti-ation process. Naïve human B cells (IgDþCD27�) differen-tiate into early memory B cells (IgDþCD27þ), and onrepeated stimulation these cells differentiate into class-switched memory B cells (IgD�CD27þ) that express IgG,IgA or IgE on their cell surface (Agematsu et al., 2000;Nagumo et al., 2002). For the purpose of this discussion,human B cells are divided into IgDþ (naïve and earlymemory) and IgD- (class-switched memory) subsets.

Expression of Kv1.3 and KCa3.1 varies in CCR7þ andCCR7� T cells, and IgDþ and IgD� B cells, as summarized in

Fig. 1. Kv1.3 channels provide the counterbalancing Kþ efflux for Ca2þ entry into CCR7� TEM-effector cells. Top left of the figure is an antigen-presenting cell(APC), which processes and presents antigen via MHC to the T-cell receptor (TCR) on a TEM cell. A signaling cascade is initiated involving the production of IP3 andthe release of calcium from internal stores via the IP3-triggered IP3 receptor leading to an increase in cytosolic calcium to about 250 nM. Emptying of the storescauses calcium to enter T cells via CRAC channels, resulting in the calcium level rising to about 1 mM. The calcium signaling cascade initiates new gene expressionvia the calmodulin–calcineurin–NFAT pathway.

V. Chi et al. / Toxicon 59 (2012) 529–546 531

Table 1. In humans, quiescent naïve, TCM and TEM cellsexpress about 300 functional Kv1.3 channels and about 10–20 KCa3.1 channels per cell (Fig. 2). Their channel-expression-pattern changes upon activation into effectorcells (Ghanshani et al., 2000; Wulff et al., 2003, 2004).KCa3.1 expression increases to about 500 channels per cellwhen CCR7þ T cells (naïve and TCM cells) activate intoeffectors (naïve-effectors and TCM-effectors). Activation byionomycin alone (which triggers calcium signaling) doesnot up-regulate expression of KCa3.1, whereas activation byphorbol myristate acetate alone (which activates proteinkinase C) increases KCa3.1 levels via transcriptional acti-vation of Ikaros and AP-1 sites on the KCa3.1 promoter andnew synthesis of KCa3.1 channel protein (Ghanshani et al.,2000). CCR7� TEM cells, in contrast, up-regulate Kv1.3 toabout 1500 per cell (Wulff et al., 2003) when they areactivated into TEM-effectors (Fig. 2). This change in CCR7expression and channel phenotype can be demonstratedin vitro by challenging human T cells multiple times withantigen. Repetitive antigen stimulation causes a progres-sive decrease of CCR7 and KCa3.1 expression and anincrease in Kv1.3 levels, reflecting the differentiation fromCCR7þ naïve T cells into CCR7� TEM cells (Wulff et al., 2003).

A similar change in Kþ channel expression pattern isseen as rat T cells differentiate from naïve toTEM cells (Table1). Quiescent naïve rat T cells from normal spleen or lymphnode express fewer Kv1.3 channels (1–10 Kv1.3/cell) thanhuman naïve T cells and the same number of KCa3.1channels (10–20/cell). When activated, naïve rat T cells up-regulate both Kv1.3 (w200/cell) and KCa3.1 (w300/cell)and acquire a pattern similar to activated CCR7þ humannaïve-effector T cells (Beeton et al., 2001). Repeated antigen

stimulation of rat T cells causes them to differentiate into Tcells with a channel phenotype similar to human TEM-effectors (w1500 Kv1.3/cell; 20–100 KCa3.1/cell) (Beetonet al., 2001). Antigen-specific rat T cell lines are CCR7�

TEM-effectors and exhibit the Kv1.3high channel pattern(Beeton et al., 2006).

In Rhesus and Sootey Mangabey monkeys, naïve, TCMand TEM cells express 70–125 Kv1.3 channels and 5–10KCa3.1 channels per cell when quiescent (Table 1). Uponactivation, naïve and TCM cells up-regulate both Kv1.3 (250/cell) and KCa3.1 (200/cell) channels (Pereira et al., 2007),while TEM cells up-regulate only Kv1.3 (w1000 Kv1.3/cell;10–20 KCa3.1/cell). Similarly, T cells from cynomolgusmonkeys display an outward Kþ current with biophysicalproperties of Kv1.3 and sensitivity to the Kv1.3-specificinhibitor ShK-186 (data not shown).

Of clinical relevance, disease-associated autoreactive T-cell clones from patients with MS and T1DM are CCR7�

TEM-effectors that express high numbers of Kv1.3 (Beetonet al., 2006; Wulff et al., 2003). In patients with MS,myelin antigen-specific patient T cells were CCR7� TEM-effectors that express >1000 Kv1.3 channels per cell,whereas T cells specific for pancreatic antigens (insulin orGAD) from the same patients were CCR7þ T cells andexhibited the Kv1.3 pattern of naïve/TCM cells (Beeton et al.,2006). In the brains of patients withMS, CD3þ/CD4þ/CCR7�

TEM cells are abundant in the perivenular infiltrate andparenchymal infiltrate of the majority of MS plaques, andmany of these cells stained positively for Kv1.3 (Rus et al.,2005). In patients with RA, T cells from the synovial fluidof affected joints were CCR7� TEM cells with >1000 Kv1.3channels per cell, whereas T cells from the synovial fluid of

Fig. 2. Kþ Channel-expression patterns in human T and B cells, and sensitivity to block by Kv1.3 and KCa3.1 blockers. A, Numbers of Kv1.3 and KCa3.1 channels percell in CCR7þ naïve or CCR7þTCM cells as they activate into naïve-effectors or TCM-effectors, respectively, and in CCR7� TEM cells when the activate into TEM-effectors. Activation was with anti-CD3 antibody. In some experiments, the cells were washed, rested, reactivated with anti-CD3 antibody. The effect of Kv1.3-specific and KCa3.1-specific blockers on proliferation ([3H]-thymidine incorporation) during activation is shown. B, Kv1.3 and KCa3.1 channels per cell in IgDþ Bcells (naïve or early memory) and IgD� B cells (class-switched memory) following activation, and the effect of Kþ channel inhibitors.

V. Chi et al. / Toxicon 59 (2012) 529–546532

non-autoimmune osteoarthritis patients were CCR7þ Tcells with low numbers of Kv1.3 (Beeton et al., 2006).Synovial biopsies from affected RA joints showed thepresence of CCR7� TEM cells that stained positively forKv1.3. The Kv1.3 channel is therefore an interesting targetfor the treatment of diverse autoimmune diseases.

The biophysical and pharmacological properties of thevoltage-gated and calcium-activated Kþ channels in humanB cells are identical to those of cloned Kv1.3 and KCa3.1homotetramers and their expression pattern changesduring differentiation and activation as it does in T cells(Fig. 2) (Wulff et al., 2004). IgDþ B cells (naïve and earlymemory), like their T-cell counterparts (naive and TCMcells), up-regulate KCa3.1 upon activation, whereas IgD� Bcells (class-switched memory), like TEM cells, up-regulateKv1.3 upon activation (Wulff et al., 2004). Interestingly,quiescent class-switched memory B cells express much

higher Kv1.3 levels than quiescent TEM cells. It should benoted that these cells are probably not truly resting sincethey are enlarged in size and express other activationmarkers like CD86 (Wulff et al., 2004).

2.3. Pharmacological sensitivity to Kþ channel blockersparallels the Kþ channel expression pattern in lymphocytes

The switch in Kþ channel phenotype has importantfunctional consequences in T and B cells. Kv1.3 channelsregulate membrane potential and calcium signaling inresting CCR7þ T cells, but when they are activated intoeffector cells (naïve-effector and TCM-effector) KCa3.1 takesover this role (Chandy et al., 2004; Wulff et al., 2003).Consequently, selective blockers of Kv1.3 suppress activa-tion/proliferation (3H-thymidine incorporation) of CCR7þ Tcells (Beeton et al., 2006; Ghanshani et al., 2000;

Table 1Number of Kv1.3 and KCa3.1 channels per cell in T cells (A) and B cells (B) from different species.

A

Species Channel type (Channels/cell) Naïve CCR7+CD45RA+ TCM CCR7+CD45RA� TEM CCR7�CD45RA�

Resting Effector Resting Effector Resting Effector

Human (CD4+ and CD8+) Kv1.3 200–250 300–350 250 300–400 200–300 1500–1800KCa3.1 <5 500–550 20 500–600 20–35 40–50

Rhesus Macaque Kv1.3 75 250 105 w195 125 1085Kv1.3 5 200 7 190 15 16

Sooty Mangabey Kv1.3 235 w190 237 200 w230 1370KCa3.1 5 195 6 200 10 24

Rat Kv1.3 5 200 5 200 50 1500KCa3.1 15 300 15 300 10 60

Mouse Kv1.3 10–30 500 10–30 500 300 800KCa3.1 10 150 10 150 130 400

Dog Kv1.3 180 w195 NT NTKCa3.1 w5 w150

B

Species Channel type (Channels/cell) Naïve (IgD+CD27�) Early memory(IgD+CD27+)

Class-switched memory(IgD�CD27+)

Resting Activated Resting Activated Resting Activated

Human Kv1.3 100 90 255 180 2425 3170KCa3.1 5 595 10 700 65 70

V. Chi et al. / Toxicon 59 (2012) 529–546 533

Vennekamp et al., 2004; Wulff et al., 2003), although thesecells up-regulate KCa3.1 channels within 6–8 h and escapeKv1.3 blocker-inhibition while acquiring sensitivity toKCa3.1 blockade (Fig. 2). By contrast, CCR7� T cells (TEM andTEM-effector) depend exclusively on Kv1.3 channels toprovide the counterbalancing Kþ efflux needed to sustaincalcium signaling in the resting and effector stages (Beetonet al., 2005, 2006; Schmitz et al., 2005; Vennekamp et al.,2004; Wulff et al., 2003). These cells remain sensitive toinhibition by Kv1.3 blockers when they transit fromquiescent to TEM-effector cells and when TEM-effectorsundergo further activation (Fig. 2). Pharmacological sensi-tivity again parallels the Kþ channel expression pattern inhuman B cells. KCa3.1-specific inhibitors inhibit activation/proliferation (3H-thymidine incorporation) of IgDþ B cells(naive or early memory), while Kv1.3 blockers have noeffect (Wulff et al., 2004). By contrast, Kv1.3 blockerssuppress the proliferation of class-switched memory Bcells, while KCa3.1 inhibitors have no effect (Wulffet al., 2004).

2.4. Kv1.3 plays an important role in other immune cells

The Kv1.3 channel has also been implicated in themodulation of responses of other immune cell types. Kv1.3expression is increased during dendritic cell maturation,and the predominant outward current in mature dendriticcells is biophysically and pharmacologically consistent withKv1.3 (Mullen et al., 2006; Zsiros et al., 2009). Certainsubsets of NK cells that engage and destroy foreign cells,also express Kv1.3 channels, and their secretion of cytolyticgranules and their killing of target cells is dependent onKv1.3-regulation of membrane potential (Schlichter et al.,1986; Sidell et al., 1986). Kv1.3 in microglia contributes tothe respiratory burst in these cells (Khanna et al., 2001) andmicroglia-mediated neuronal killing (Fordyce et al., 2005).Thus, Kv1.3 inhibitors such as ShK-186 may have a role in

immune modulation beyond simply the TEM-effector andclass-switched memory B cell populations.

3. The discovery of ShK and development of ShK-186

Stichodactyla helianthus is a common species of seaanemone in the Caribbean Sea around Cuba. In 1995, OlgaCastaneda, Evert Karlsson, Alan Harvey, Reto Stöcklin andtheir colleagues found that S. helianthus extracts adminis-tered to mice by intraperitoneal injection, induced hyper-sensitivity to touch and sound, excessive salivation,lacrimation, sweating, motor incoordination and paralysis,reminiscent of poisoning by cholinesterase inhibitors(Castaneda and Harvey, 2009; Castaneda et al., 1995).However, a whole-animal study revealed weak cholines-terase inhibitor-activity in the extracts, suggesting thatthese toxic symptoms were due to activity againsta different molecular target. Realizing that the Kþ channel-blocking snake venom toxin dendrotoxin could producea similar toxicity pattern via enhanced cholinergicimpulses, these investigators used a screening assay basedon 125I-dendrotoxin binding to synaptosomes to identifya peptide, ShK, that blocked Kþ channels in culturedneurons (Castaneda et al., 1995). Soon after, Michael Pen-nington, William Kem, Ray Norton and their colleaguessynthesized the peptide (Pennington et al., 1995), deter-mined its three-dimensional structure (Tudor et al., 1996),and showed that ShK blocked the Kv1.3 channel in T cellswith low picomolar affinity (Pennington et al., 1996).

ShK contains 35 amino acids including six cysteines thatform three disulfide bonds (Fig. 3) (Pohl et al., 1995). ShKblocks Kv1.3 with an IC50 of w10 pM (Kalman et al., 1998),but it also blocks two other channels with picomolarpotency (Kv1.1, Kv1.6) and three others (Kv1.2, Kv3.2 andKCa3.1) with nanomolar potency (Table 1). It was there-fore important to develop ShK analogs with improvedselectivity for Kv1.3 over the other channels. As a first step,

Fig. 3. A, Alignment of ShK and related sea anemone toxins: The six cysteines are in gray and the disulfide pairing is shown. Lys22 and Tyr23 are highlighted inblue. Lys22 occupies the lumen of the Kv1.3 channel pore. GenBank Accession numbers are listed next to each sequence. Many of these sea anemone toxinsequences have been characterized for Kþ channel-blocking activity (Castaneda et al., 1995; Cotton et al., 1997; Gendeh et al., 1997a, 1997b; Honma et al., 2008;Shiomi, 2009; Yamaguchi et al., 2011). B, A phylogenetic tree based on the multiple sequence alignment and generated with the PHYLIP program (http://www.genebee.msu.su/genebee.html): ShK (ShK-Tx) and toxins from six other sea anemones (S. mertensii [SmK-Tx], S. haddoni [ShaK-Tx], S. gigantean [SgK-Tx], T. aster[TaK-Tx], C. adhaesivum [CaK-Tx], H. magnifica [HhK-Tx]) have the same number of residues as ShK, share remarkable sequence similarity, and cluster in one sub-group. Two toxins from Heteractis magnifica (HmK-Tx1, HmK-Tx2) and the toxin from Anemonia erythraea (AnerK-Tx) have the same number of residues as ShK,and comprise a closely-related but distinct cluster. Toxins from three other sea anemones (B. granulifera [BgK-Tx], A. equine [AceqK-Tx], A. sulcata [AsKs-Tx])exemplified by the BgK toxin contain four extra residues between cysteines 3 and 4, and constitute a distinct, yet ShK-related group. (For interpretation ofthe references to color in this figure legend, the reader is referred to the web version of this article.)

V. Chi et al. / Toxicon 59 (2012) 529–546534

an Ala scan of non-Cys residues was utilized to identify ShKresidues required for interaction with Kv1.3 (Arg11, His19,Ser20, Lys22, Tyr23, Arg24); these residues were found to beclustered on one surface of the peptide (Pennington et al.,1996; Rauer et al., 1999) (Fig. 4). Complementary muta-genesis and doublemutant cycle analysis were then used tocharacterize the ShK-binding site on Kv1.3 (Kalman et al.,

1998; Lanigan et al., 2002). ShK binds to a shallow vesti-bule at the outer mouth of the Kv1.3 channel pore where itinteracts predominantly with two adjacent subunits of thechannel, and Lys22 occludes the pore lumen like a cork ina bottle (Fig. 4) (Kalman et al., 1998; Lanigan et al., 2002).

Guided by this knowledge, some 380 ShK analogs havebeen generated with the goal of developing a Kv1.3-specific

Fig. 4. A, Stereo view of the closest-to-average structure of ShK in solution (PDB id 1ROO) (Tudor et al., 1996). The three disulfide bridges (Cys3-Cys35, Cys12-Cys28,and Cys17-Cys32) are shown in yellow. B, Surface views of ShK with key residues colored as follows: Lys22 blue, Tyr23 magenta, His19 deep purple, Ser20 orangeand Arg24 cyan. Met21, which is susceptible to oxidation, is colored yellow and Arg1 is colored teal as a reference point. In the right hand view, Arg24 is not visibleand Arg1 lies at the far left of the structure; this orientation is similar to that of ShK-192 in part C. C, Structure of ShK-192 docked with a model of the pore-vestibule region of Kv1.3 (Pennington et al., 2009). Kv1.3 is shown in a ribbon representation, while ShK-192 is shown as a molecular surface, with all resi-dues colored wheat except Lys22, which is colored blue, Tyr23 magenta and the N-terminal extension pink. (For interpretation of the references to color in thisfigure legend, the reader is referred to the web version of this article.)

V. Chi et al. / Toxicon 59 (2012) 529–546 535

inhibitor. Our early work focused on the full-length peptideas a significant decrease in potency was observed if wetruncated ShK, made mono- and bis-disulfide analogs byreplacing its disulfide bridges with the isostere surrogatealpha amino butyric acid, or stabilized its helical motifs withlactam rings (Lanigan et al., 2002; Pennington et al., 1999).WhenLys22was replacedwith the shorter positively-chargedunnatural amino acid diaminopropionic acid (Dap), thisanalog, ShK-Dap22 (Kalman et al., 1998), blocked Kv1.3 withpicomolar potency and exhibited >75-fold selectivity forKv1.3 over Kv1.1 and other related channels (Table 2).Scientists at Amgen Inc., Thousand Oaks, California, havemade a number of interesting ShK analogs (Sullivan et al.,2010). One of these, ShK-Q16K-PEG[20K], has w1600-foldselectivity for Kv1.3 over Kv1.1 (Table 2). Another analogcontains the heavy chain of an antibody attached to a singleShK-Q16K; this peptibody exhibits>1400-fold selectivity forKv1.3 over Kv1.1 (Sullivan et al., 2010). Both these analogsmay last longer in the circulation than ShK. Our analogs thatare progressing to the clinic have modifications at the N-terminus of the peptide (Table 2). ShK-170 contains a L-phosphotyrosine attached via a aminoethyloxyethyloxy-acetyl (Aeea) linker to the a-amino group of Arg1 (Beetonet al., 2005). ShK-170 blocks Kv1.3 channels with an IC50 of69 pM, and it is 100-fold selective for Kv1.3 over Kv1.1, 260-fold selective over Kv1.6, 280-fold selective over Kv3.2,680-fold selective over Kv1.2, and >1000-fold selective over

all other channels tested including KCa3.1 (Table 2). Tostabilize the C-terminus of ShK-170 and improve itsmanufacturing aspects, we replaced the C-terminal carboxylwith an amide to minimize digestion by carboxypeptidases.The new analog, ShK-186 (Beeton et al., 2006), retains theselectivity and potency profile of ShK-170. Another analog,ShK-192, includes three stabilizing elements to minimizedegradation: the N-terminal phosphotyrosine was replacedwith the nonhydrolyzable phosphate mimetic para-phosphonophenylalanine (Ppa), Met21 was replaced withthe isosteric homolog norleucine to avoid methionineoxidation, and the C-terminal was amidated (Penningtonet al., 2009). ShK-192 blocks Kv1.3 with an IC50 of 140 pMandexhibits 30-fold selectivity over Kv3.2, 75-fold selectivityover Kv1.6 and 157-fold selectivity over Kv1.1 (Table 2).

4. ShK analogs are immunomodulatory

4.1. In vitro studies

ShK-186 inhibits Ca2þ signaling in human TEM clones ina dose-dependent fashion with an IC50 z 200 pM (Beetonet al., 2006). ShK-186 is significantly more effective insuppressing IL-2 and IFN-g production by human CCR7�

TEM cells than CCR7þ naïve/TCM cells, but it is less effectivein suppressing the production of TNF-a and IL-4 by eithersubset (Fig. 5) (Beeton et al., 2006). ShK-186 is z10-fold

Table 2The blocking potencies (Kd values) of ShK and selected analogs on a panel of Kþ channels is shown. For comparison, Kd values for two other Kv1.3 blockers,OSK1-(K16,D20) (Mouhat et al., 2005) and PAP-1 (Schmitz et al., 2005) are shown.

Channel ShK(Kd: nM)

ShK-Dap22

(Kd: nM)ShK-170/ShK-186(Kd: nM)

ShK-192(Kd: nM)

ShK-Q16K-PEG[20K](Kd: nM)

OSK1-(K16,D20)(Kd: nM)

PAP-1(Kd: nM)

Clofazimine(Kd: nM)

Kv1.1 0.028 1.8 7 22 997 0.40 65 >10,000Kv1.2 10 39 48 >100 639 3 250 >10,000Kv1.3 0.010 0.023 0.069 0.140 0.94 0.003 2 300Kv1.5 >100 >100 >100 >100 >10,000 >100 45 >10,000Kv1.6 0.2 10.5 18 10.5 466 >100 62 NDKv1.7 >100 >100 >100 ND >10,000 >100 98 NDKv2.1 ND ND >100 ND ND ND 3000 NDKv3.1 >100 >100 >100 ND ND >100 5000 >10,000Kv3.2 5 ND 20 4.2 ND 980 NDKv11.1 (HERG) >100 ND >100 >100 >10,000 >100 5000 NDKir2.1 >100 ND >100 ND ND ND 15,000 NDKCa2.1 >100 ND >100 ND ND >100 10,000 NDKCa2.3 >100 ND >100 ND ND >100 5000 NDKCa3.1 28 >100 115 >100 >10,000 >100 10,000 ND

V. Chi et al. / Toxicon 59 (2012) 529–546536

more effective in suppressing proliferation ([3H]thymidineincorporation) by human CCR7� TEM cells compared withCCR7þ T cells (Fig. 5) (Beeton et al., 2006). When these cellpopulations are activated for 48 h, rested, and restimulated,CCR7� TEM-effectors cells remain exquisitely sensitive toShK-186 inhibition (IC50 z 100 pM), whereas CCR7þ

effector cells (naïve-effector, TCM-effector) are resistant(Fig. 6). This “escape” by activated CCR7þ naïve/TCM cells isdue to up-regulation of KCa3.1 that provides the counter-balancing Kþ efflux in these cells in place of Kv1.3. Asignificant aspect of this finding is that Kv1.3 blockers,unlike current immunomodulatory therapies thatbroadly suppress the immune response, would selectivelysuppress CCR7� T cells (TEM/TEM-effectors) implicated in

Fig. 5. Effect of ShK-186 on cytokine production by human T cell subsets a

autoimmune diseases, but not CCR7þ T cells that are majormediators of immune protection. In support of this thera-peutic concept, ShK-186, preferentially suppresses disease-associated autoreactive CCR7� TEM cells (myelin- orGAD65-specific T cells or synovial T cells from affectedjoints) of MS and RA patients, respectively, withoutaffecting other T cell subsets from these patients (Beetonet al., 2006).

ShK analogs have similar effects on rat T cells. ShK-170suppressed myelin antigen-triggered proliferation of therat CCR7� TEM cell line, PAS (IC50 ¼ 0.08 nM), >100-foldmore effectively than mitogen-induced proliferation of ratsplenic T cells (IC50 ¼ 100 nM) (Beeton et al., 2005).Proliferation of the PAS cell line was also inhibited by

ctivated with anti-CD3 antibody. Adapted from Beeton et al. (2006).

Fig. 6. Effect of ShK-186 on proliferation ([3H]-thymidine incorporation) by human T cell subsets activated with anti-CD3 antibody (adapted from Beeton et al.,2006). The Kþ channel expression patterns of CCR7þ and CCR7� T cells are shownwhen they are at rest, when activated into effectors, and when reactivated. Thepharmacological sensitivity to ShK-186 depends on the channel expression pattern. ShK-186 suppresses CCR7þ naïve/TCM cells with an IC50 value of 2–5 nM,whereas CCR7� TEM cells are suppressed with an IC50 value ¼ 0.1–0.2 nM. The roughly 10-fold lower sensitivity of CCR7þ T cells is because they up-regulate KCa3.1during the activation process. When CCR7þ effectors are reactivated, they are insensitive to ShK-186 because their membrane potential is dominated by ShK-186-resistant KCa3.1 channels, whereas CCR7� effectors are suppressed with an IC50 ¼ 0.08–0.1 nM. Similar results are obtained with rat T cells. ShK-170 suppressesmitogen-induced proliferation of rat splenic T cells with an IC50 ¼ 100 nM, whereas it suppresses antigen-triggered proliferation of the rat CCR7� TEM cell line,PAS, with an IC50 ¼ 0.08 nM.

V. Chi et al. / Toxicon 59 (2012) 529–546 537

Amgen’s ShK-Q16K-PEG[20K] analog (IC50 ¼ 0.170 nM) andtheir ShK-Q16K-peptibody (IC50 ¼ 0.84 nM) (Sullivanet al., 2010).

4.2. Delayed-type hypersensitivity (DTH), an in vivo modelfor inflammation caused by skin-homing CCR7� TEM cells

We used the DTH model to determine the in vivoimmunomodulatory efficacy of ShK analogs. Lewis ratswere immunized with ovalbumin and complete Freund’sadjuvant, and a week later, one ear of each rat was injectedwith ovalbumin and the other ear with saline. Within 24 h,the ovalbumin-injected ear was significantly swollen, redand inflamed compared to the saline-injected ear. Oncedaily subcutaneous injections (in the flank) of ShK-170(10 mg/kg), ShK-186 (10 or 100 mg/kg) or ShK-192 (1, 10, or100 mg/kg) suppressed the DTH response compared to ratsadministered saline (Beeton et al., 2005; Matheu et al.,2008; Pennington et al., 2009). [Note that ShK-170 andShK-186 are identical, except for the C-terminal carboxyl in170 is replaced by an amide in 186].

Two-photon microscopy was then used to image skin-homing CCR7� TEM-effectors during DTH and assess theaction of ShK-186 (Matheu et al., 2008). Within 3 h ofantigen-challenge, GFP-labeled ovalbumin-specific CCR7�

TEM-effectorswerevisible inovalbumin-injectedearswherethey formed stable contacts with tissue antigen-presenting

cells. These TEM-effectors activated over the course of a day,enlarged and began crawling rapidly along collagen fibers.This behavior coincided with massive swelling of theovalbumin-injected ear. The saline-injected ear did notswell up, and very few ovalbumin-specific TEM-effectorswere seen. Treatmentwith ShK-186 (100mg/kgoncedaily bysubcutaneous injection) inhibited the DTH response byrendering CCR7� TEM-effectors immotile in the ear andpreventing their activation and enlargement (Matheu et al.,2008). The same dosing regimen had no effect on themotility of CCR7þ T cells in lymphoid tissue, and it did notaffect the motility of bystander cells. Prolonged immobili-zation of CCR7� TEM-effector cells in inflamed tissues due toKv1.3 blockade might prevent these cells from receivingactivation and survival signals, thus leading to TEM senes-cence via cytokine deprivation or the ‘death-by-neglect’mechanism. In support of this, Kv1.3 channel blockers(margatoxin and correolide) suppress DTH to tuberculin inminipigs, and 2–3 weeks following the cessation of treat-ment, DTH cannot be elicited by repeat challenge withtuberculin (Koo et al., 1999, 1997). This period of ‘remission’by Kv1.3 blocker therapy may be the result of the death-by-neglect mechanism whereby tuberculin-specific skin-homing CCR7� T cells are immobilized and then deleted atthe site of DTH. Note, Koo et al. (1999, 1997), attributed theremission following Kv1.3 blocker-therapy to cellulardepletion of the thymus.

V. Chi et al. / Toxicon 59 (2012) 529–546538

4.3. Experimental autoimmune encephalomyelitis (EAE) –a model for multiple sclerosis

Myelin-specific T cells in patients with MS are predom-inantly CCR7� TEM-effector cells with elevated Kv1.3expression (Beeton et al., 2006; Wulff et al., 2003). To testwhether such cellsmight bedisease-inducing,wegeneratedrat myelin-specific CCR7� TEM-effector cell lines andcompared their ability to induce experimental autoimmuneencephalomyelitis (EAE) following adoptive transfer tohealthy rats. Cells that expressed high numbers of Kv1.3channels induced severe adoptive EAE compared to cellswith lower numbers of Kv1.3 channels (Beeton et al., 2005,2001). Furthermore, ShK-170 suppressed myelin antigen-triggered proliferation of these Kv1.3high CCR7� TEM-effec-tors, and its daily administration (10 mg/kg/day subcuta-neous injection) both prevented and effectively treatedadoptive EAE (Beeton et al., 2005). In the same model,Amgen’s ShK-Q16K-PEG[20K] analog prevented adoptiveEAE in a dose-dependent manner when administered oncedaily by subcutaneous injection (Sullivan et al., 2010).

Most patients with MS exhibit a relapsing and remittingclinical course of disease. EAE with a similar relapsing-remitting pattern can be induced by immunizing DarkAgouti (DA) rats with autologous spinal cord in emulsionwith complete Freund’s adjuvant. This disease is termedchronic relapsing-remitting EAE (CR-EAE). T cells in thecentral nervous system of these rats are predominantlyCCR7� TEM/TEM-effectors during the chronic relapsing-remitting phase of the disease (Matheu et al., 2008). ShK-186 administered by once-daily subcutaneous injections(100 mg/kg/day) from the start of symptoms amelioratedthe disease by decreasing inflammatory cell-infiltrationinto the central nervous system and by decreasing demy-elination (Matheu et al., 2008). Over the last 4 years, wehave repeated these studies multiple times. In Fig. 7, wehave compiled our data and compared clinical scores in ratswith CR-EAE that were treated with once-daily subcuta-neous injections of vehicle (n ¼ 91), ShK-186 (100 mg/kg/day, n ¼ 53), or ShK-192 (100 mg/kg/day, n ¼ 20; 1 mg/kg/day, n ¼ 20). A repeated-measures two-way ANOVAstatistical test shows that ShK-186 and ShK-192 signifi-cantly reduced severity of disease (cumulative p < 0.001).

Fig. 7. ShK-186 and ShK-192 reduce disease severity in chronic relapsing-remittingShK186- and ShK-192-treatment in rats with CR-EAE. ShK-186 and ShK-192 wereTreatment was started at the onset of disease (clinical score ¼ 1) Cumulative p valuep < 0.001. B, Effect of treatment with ShK-192 at 100 and 1 mg/kg/day in rats with CRtail tone; 2 ¼ mild paraparesis and ataxia; 3 ¼ moderate paraparesis, tripping whil5 ¼ complete hind paralysis and incontinence; 6 ¼ moribund, difficulty breathing,

4.4. Pristane-induced arthritis – a model for rheumatoidarthritis and psoriatic arthritis

ShK-186 was evaluated in a third autoimmune diseasemodel. DA rats injected with pristane oil develop arthritiswithin 7–10 days, which is believed to be similar to RA andpsoriatic arthritis (Hultqvist et al., 2006). In saline-treatedrats, disease severity worsened continuously with time.Significant periostitis, erosion, and deformity were visiblein X-rays of the joints, and pathological evaluation revealedsevere synovitis, inflammatory cell infiltrate, and cartilageulceration. ShK-186, administered from the start of symp-toms by once-daily subcutaneous injections (100 mg/kg/day), significantly reduced the number of affected joints,and the rats showed significant improvement in radiolog-ical and histopathological findings (Beeton et al., 2006).

4.5. Autoimmune glomerulonephritis

In a rat model of anti-glomerular basement membraneglomerulonephritis, the majority of CD4þ T cells and someCD8þ T cells in the kidney were Kv1.3high TEM cells (Hyodoet al., 2010). Somemacrophages in the kidney infiltrate alsoexpressed Kv1.3. Rats treated with the Kv1.3 inhibitorPsora-4 showed less proteinuria and fewer crescenticglomeruli than rats administered vehicle (Hyodo et al.,2010). ShK-186 may therefore be useful in the treatmentof autoimmune kidney disease.

4.6. Inflammatory disorders of the skin

In a rat model of contact dermatitis, CD8þ TEM cellspredominated amongst T cells infiltrating the skin duringthe elicitation phase (Azam et al., 2007). PAP-1, a Kv1.3inhibitor (Table 2) (Schmitz et al., 2005), suppressed thisdisease by inhibiting the infiltration of CD8þ TEM cells andby reducing the production of the inflammatory cytokinesIFN-g, IL-2, and IL-17 (Azam et al., 2007). In studies onpsoriasis, more Kv1.3þ immune cells were found in skinfrom patients with psoriasis than in normal human skin(Gilhar et al., 2011). Grafting of human psoriatic skin on tobeige-SCIDmice produced a condition resembling psoriasis,and local injections of ShK ameliorated disease by

experimental autoimmune encephalomyelitis in DA rats. A, Effect of vehicle-,administered by once daily subcutaneous injections, both at 100 mg/kg/day.from repeated-measures two-tailed ANOVA for both ShK-186 and ShK-192 is-EAE (cumulative p < 0.001). Clinical scores: 0.5 ¼ distal tail limp; 1 ¼ loss ofe walking; 3.5 ¼ one hind limb paralyzed; 4 ¼ complete hind limb paralysis;does not eat or drink, euthanized immediately.

V. Chi et al. / Toxicon 59 (2012) 529–546 539

significantly reducing the number of Kv1.3þ infiltrating cells(Gilhar et al., 2011). Interestingly, clofazimine, a drug thattreats pustular psoriasis in humans (Chuaprapaisilp andPiamphongsant, 1978; Nair and Shereef, 1991), has beenshown to block Kv1.3 with specificity over closely-relatedchannels (Ren et al., 2008) (Table 2). Seven decades of clin-ical use has revealed clofazimine to be generally safe,although it causes reversible skin discoloration in manypatients, andprolonged therapywithhigh concentrations ofclofazimine results in the formation of drug crystals intissues, leading to eye problems, splenic infarction, and, inone case, death (Craythorn et al., 1986; McDougall et al.,1980; Parizhskaya et al., 2001; Mathew et al., 2006). Thesereports highlight the promise of Kv1.3 blockers such as ShK-186 for the treatment of inflammatory skin disorders.

4.7. Experimental autoimmune diabetes mellitus – a modelfor T1DM

Rats of the BB/Wor strain begin developing experi-mental autoimmune diabetes mellitus at the age of 70 days,and by 110 days disease-incidence is close to 100%. Once-daily oral administration of PAP-1 (50 mg/kg) significantlydelayed the onset of disease in rats and only 47% of the ratshad developed disease by 110 days (Beeton et al., 2006).Disease amelioration was associated with reduced infil-tration of pancreatic islets by T cells and macrophages, anddecreased b-cell destruction. Although Kv1.3 inhibitors arereported to increase glucose uptake inmouse adipocytes bystimulating GLUT4 translocation (Li et al., 2006; Xu et al.,2004), PAP-1, ShK or margatoxin did not increase basal orinsulin-stimulated glucose uptake by isolated cultured ratadipocytes, and did not enhance production of the insulin-sensitizing adipocyte hormone adiponectin by these cells(Beeton et al., 2006), indicating that PAP-1’s therapeuticactivity is likely to be via immunomodulation rather thana metabolic effect. ShK-186 (100 mg/kg once daily) delayedthe onset of diabetes in these rats by about 10 days (Wulffand Pennington, 2007), but did not decrease disease inci-dence (data not shown). Differences in pharmacokineticproperties might explain the difference in efficacy of PAP-1and ShK-186 in this model. Pharmacokinetic studies showthat PAP-1 (log P 4.03) concentrates in the pancreas (alipophilic organ) when compared to blood, and this accu-mulation may contribute to its therapeutic effectiveness(Beeton et al., 2006). ShK-186, a peptide with a net basiccharge is unlikely to concentrate in the pancreas.

Studies on Kv1.3�/� mice suggest that Kv1.3 blockersmight be effective in treating type-2 diabetes mellitus andobesity (Xu et al., 2003). Kv1.3�/� mice are more insulinsensitive and gain less weight on a high-fat diet than wild-type littermates, and blockade of Kv1.3 with margatoxin,a peptide fromscorpionvenom, increases peripheral insulinsensitivity inwild-type C57BLmice, and in db/db and ob/obmice with type 2 diabetes mellitus and obesity (Xu et al.,2003). One mechanism for these therapeutic effects is thatgenetic deletion of Kv1.3 or pharmacological blockade of thechannel promotes the translocation of the glucose trans-porter to the plasma membrane in adipose tissue and skel-etal muscle, thereby enhancing peripheral insulinsensitivity (Xu et al., 2004). However, a recent study

performed by scientists at Pfizer concluded that Kv1.3 is notinvolved in the modulation of peripheral insulin sensitivity(Straub et al., 2011). These authors demonstrated that Kv1.3wasnotexpressed inhumanadiposeor skeletalmuscle fromhealthy individuals or patients with type-2 diabetes melli-tus, Kv1.3 blockers (PAP-1 or MgTX) did not alter glucoseuptake into human skeletal muscle cells or mouse adipo-cytes, administration of PAP-1 failed to improve insulinsensitivity in hyperglycemic ob/obmice at free plasma PAP-1 concentrations that are specific for inhibition of Kv1.3, andinsulin sensitivity was only increased when PAP-1 concen-trationsweresufficient to inhibitotherKv1channels (Straubet al., 2011). In other studies, deletion of Kv1.3 in micelacking the melanocortin-4 receptor (MC4R�/�/Kv1.3�/�)caused them to live longer, have a lower bodyweight, andenjoy increased reproductive success compared toMC4R�/�

mice (Tucker et al., 2008). These changes were attributed todecreased fat deposition, reduced fasting leptin levels, andenhanced dark-phase locomotor activity and mass-specificmetabolism (Tucker et al., 2008).

4.8. Transplantation

Of the approximately 20 billion T cells in human skin,about 85% are skin-homing CCR7� TEM cells (Clark, 2010),which are probably critical for the rejection of skin trans-plants. Kv1.3 blockers may therefore be effective in pre-venting the rejection of skin grafts. In support of this idea,clofazimine, an inhibitor of Kv1.3 channels, prevented therejection of grafted skin in an animal model (Ren et al.,2008). Human foreskin was grafted onto Pfb-Rag2�/�

mice that lack T, B and NK cells (Ren et al., 2008). After thegraft had healed, 100 � 106 human peripheral bloodlymphocytes from an unrelated donor were adoptivelytransferred into the same animals and rejection of the skingraft was monitored. The graft was rejected within 11 daysin vehicle-treated mice, but mice treated for 10 days withclofazimine did not reject the graft for 35 days, 20 daysafter the cessation of treatment (Ren et al., 2008).

In a study on kidney transplantation, ShK (80 mg/kg threetimes daily s.c.) in combination with the KCa3.1 blockerTRAM-34 (120 mg/kg/day i.p.) was as effective as cyclo-sporine A (5 mg/kg/day s.c.) in reducing infiltration ofa kidney graft (from Fisher to Lewis rats) by mononuclearcells, CD8þ T cells and macrophages (Grgic et al., 2009). Inanother study, ShK-186 (300 mg/kg/day twice daily s.c.) andTRAM-34 (20 mg/kg/day twice daily, i.p.) delayed by twodays rejection of pancreatic islets from Lewis rats that weregrafted under the kidney capsule of DA rats (Fig. 8). Incontrast, rats that received cyclosporine A (5 mg/kg s.c.), animmunomodulator that affects both CCR7þ and CCR7�

effectors, did not reject the transplanted islets for 60 days,and after cessation of the drug, islets were rejected in 2weeks (Fig. 8). Animals that received cyclosporine A for 21days followed by either vehicle or ShK-186 (100 mg/kg/days.c.), rejected islets in 2weeks indicating that ShK-186 alonedoes not prevent acute rejection (Fig. 8). Chronic rejection isbelieved to be mediated by CCR7� TEM cells, and should beprevented by Kv1.3 blockers. In fact, clofazimine has beenreported to prevent chronic graft-versus-disease in humansfollowing bone marrow transplantation (Lee et al., 1997).

Fig. 8. Effectiveness of ShK-186, TRAM-34 and cyclosporine A in preventing the rejection of transplanted pancreatic islets. A, Blood glucose levels from diabeticdark agouti (DA) rats transplanted with pancreatic islets from Lewis rats under the kidney capsule, that received either vehicle, ShK-186 (300 mg/kg twice dailys.c.) þ TRAM-34 (20 mg/kg twice daily i.p.), or cyclosporine A (5 mg/kg s.c.). Cyclosporine A was stopped on day-60. Red *: p < 0.01. Blue *: p < 0.001. Only twoblue * are shown next to the cyclosporine A data, but every day thereafter until day 60 was statistically significant at p < 0.001. B, Blood glucose levels fromtransplanted DA rats that were treated with Cyclosporine A for 21 days followed by ShK-186 (100 mg/kg/day) or vehicle. DA rats were made diabetic byStreptozotocin (20 mg/kg, i.p., administered a total of 5 times every third day). Freshly isolated islets (4000IEQ) from Lewis rats were transplanted under thekidney capsule of DA rats 21 days after they developed diabetes mellitus (two consecutive readings of over 350 mg/dL of blood glucose were considered to beindicative of diabetes). Rats became euglycemic following islet transplant. The recurrence of diabetes was considered a sign of graft rejection. (For interpretationof the references to color in this figure legend, the reader is referred to the web version of this article.)

V. Chi et al. / Toxicon 59 (2012) 529–546540

5. Pre-clinical development of ShK-186

5.1. Manufacture and formulation of ShK-186

ShK-186 has been manufactured under full GMP regu-lations at up to the 30 g scale using standard Fmoc-tBusolid-phase synthesis methods. Synthesis at this scaledemonstrates the robustness of the manufacturing process,

and is adequate to support clinical development throughthe expected range of human doses (approximately 1–15 mg/kg/week). Disulfide bond formation is achieved by airoxidation or using a glutathione exchange reaction. Themanufacturing process provides a consistently pure andpotent peptide with dissociation constants as measured onthe cloned Kv1.3 channel consistent with publishedreports. Formulation of ShK-186 takes advantage of the

V. Chi et al. / Toxicon 59 (2012) 529–546 541

inherent high degree of stability and solubility of this pol-ybasic peptide (Tudor et al., 1998). Ideal liquid formulationsare composed of phosphate-buffered isotonic solutionswith a pH optimum around 6.0. Small amounts of surfac-tants can be added to the formulation to enhance resistanceto peptide aggregation under various stress conditions. Theresulting formulations are resistant to aggregation, expo-sure to ultraviolet light and serial freeze–thaw cycles. Inaddition, and despite the presence of a single oxidizablemethionine in the peptide (Fig. 4), formulated ShK-186demonstrates good stability under forced oxidationconditions (0.01% hydrogen peroxide) for several hours.The resulting formulated drug product demonstrates long-term stability as a frozen or liquid formulation and isreadily appropriate for early clinical development of thedrug. The longer-term commercial development ofa subcutaneous formulation will take advantage of thedrug’s solubility and potency to permit the use of smalldose volumes and autoinjectors or needle-free injectionsystems.

5.2. Pharmacodynamic assay to measure ShK-186’simmunomodulatory effect in vivo

A critical feature of the development of new drugs withnovel mechanisms of action is the coordinate developmentof pharmacodynamic assays to measure and relate drugactivity with drug exposure in vivo. Antagonism of Kv1.3provides a special challenge due to its highly restrictedfunctional role in CCR7� T cells (TEM/TEM-effector) and thepaucity of this T cell subset in the vascular compartment(only 5–15% of the total T cells). An early attempt to char-acterize the pharmacodynamic activity of Kv1.3-selectiveinhibitors involved the ex vivo stimulation (PMA plus ion-omycin) of peripheral blood mononuclear cells from min-ipigs treated with the scorpion toxinmargatoxin (Koo et al.,1997). Drug treatment suppressed [3H]-thymidine incor-poration (Koo et al., 1999, 1997). This work was laterexpanded by Amgen to demonstrate that ex vivo stimula-tion of whole blood from cynomolgus monkeys treatedwith ShK analogs resulted in reduced expression of IL-2,IFN-g, and IL-17 (Sullivan et al., 2010). Our analysis of

Fig. 9. ShK-186 suppresses cytokine production in human T cells fromwhole blood.ShK-186 was most effective in suppressing the production of IL-2 and the Th1 cyteffective in inhibiting IL-4 (Th2 cells).

ex vivo stimulated human whole blood suggests thata direct effect on Kv1.3 channel blockade can be measuredusing stimulations that exclusively trigger calciumsignaling. For example, thapsigargin, an inhibitor of theSERCA pump, empties the ER Ca2þ store and triggers theactivation of CRAC channels on the cell surface, resulting inCa2þ influx into T cells. The ensuing calcium-signalingcascade stimulates cytokine production without up-regulating the KCa3.1 channel by CCR7þ T cells and theirescape from Kv1.3 blocker-inhibition. This permits theeffect of ShK-186 to be measured over the entire T cellpopulation. Fig. 9 shows that ShK-186 suppressedthapsigargin-induced production of cytokines by T cells inwhole blood at picomolar concentrations with thefollowing potency sequence: IL-2 ¼ Th1 (IFN-g) > Th17 (IL-17) > Th2 (IL-4). In a similar assay, ShK-Q16K-PEG[20K]inhibited IL-2 (IC50: 109 pM) and IFN-g (IC50: 240 pM)production by human T cells, and IL-17 production bycynomolgus monkey T cells (Sullivan et al., 2010). Th1 andTh17 cells are implicated in the pathophysiology of auto-immune diseases (Damsker et al., 2010; Hemdan et al.,2011; Miossec et al., 2009). These drugs have no apparenteffect on IL-6 or TNF-a production using the same system.

Amgen used the ex vivo assay to monitor the immuno-modulatory effect of their ShK-Q16K-PEG[20K] analog incynomolgus monkeys selected for high numbers of CD4þ

CCR7� TEM cells (Sullivan et al., 2010). Monkeys receivedfour once-weekly subcutaneous injections of the analog at0.5 mg/kg. IL-17 was profoundly suppressed, and thissuppression lasted for 2 weeks after cessation of treatment.

5.3. Pharmacokinetic studies

ShK analogs are clearly immunomodulatory if admin-istered once daily by subcutaneous injection. We useda patch-clamp bioassay to ascertain whether circulatinglevels of ShK analogs after subcutaneous injection weresufficient to inhibit TEM cells. ShK-170 was detectable inserum 5 min after a single subcutaneous injection of200 mg/kg, peak levels of 12 nMwere reached in 30min andthe level then fell to a baseline of approximately 0.3 nMover the next 7 h (Beeton et al., 2005). The disappearance of

T cells were stimulated with thapsigargin, an inhibitor of the ER SERCA pump.okine IFN-g. It was less effective in suppressing IL-17 (Th17 cells) and least

V. Chi et al. / Toxicon 59 (2012) 529–546542

ShK-170 from the blood could be fitted by a single expo-nential curve, and the circulating half-life was estimated tobe w50 min. After a 20-fold lower dose of ShK-170 (10 mg/kg), the peak serum concentration reached z500 pMwithin 30 min, and repeated once-daily administration ofthis dose resulted in steady-state levels of 0.3 nM (Beetonet al., 2005). This steady-state level suggests that thepeptide accumulates in the body on repeated administra-tion. The route of administrationdsubcutaneous or intra-venousddid not alter the steady-state level, indicating thatthis depot is not the skin, but rather lies elsewhere. ShK-186and ShK-192 are also cleared from the circulation witha half-life of 30–50 min, but sufficient amounts of thesepeptides to cause immune suppression of CCR7� TEM cells(0.2 nM) can be detected in the serum even 2 days aftersubcutaneous injection (Pennington et al., 2009). Theseresults suggest that the peptide undergoes rapid clearancefrom the circulatory compartment into peripheralcompartment(s), and then returns to the circulation ata steady-state level of w0.2–0.3 nM. Interestingly, ultrafil-tration followed by RP-HPLC shows that w90% of ShK-186is bound within 15 min to proteins in serum from rats,cynomolgus and humans (Fig. 10). Serum protein-boundShK-186 may constitute a reservoir of drug, althoughthere are likely to be other compartments in the body.These data are consistent with the behavior of otherpositively-charged peptide drugs, which demonstrate rapiddistribution from the central compartment and largeapparent volumes of distribution (Boswell et al., 2010).These effects are thought to be mediated by low-affinitycharge interactions between positively-charged drugs andabundant negatively charged surfaces throughout the body(Boswell et al., 2010; Omar et al., 1992). Studies with the D-diastereomer of ShK emphasize that renal clearance ratherthan proteolysis is the major determinant of elimination-half-life of ShK peptides (Beeton et al., 2008). More care-ful pharmacokinetic studies with LC–MS and/or ELISAmethods, or with radiolabeled ShK-186, are required tofurther characterize the pharmacokinetic properties of thepeptide.

Fig. 10. ShK-186 exhibits significant binding (90%) to proteins in human, rat(Sprague–Dawley) and Cynomolgus monkey serum. Sera were incubated for15 min at room temperature with ShK-186, before being subjected toultrafiltration (Millipore Centrifree Ultrafiltration Device – Cat#: 4104) fol-lowed by RP-HPLC. The sera were spiked with 1� or 10 ml/ml Halt� Proteaseand Phosphatase Inhibitor cocktail and 1� or 10 ml/ml EDTA (0.5 M) toprevent dephosphorylation of ShK-186.

Amgen conducted pharmacokinetic studies with ShK-Q16K-PEG[20K] by the intravenous and subcutaneousroutes of administration in mice, rats, beagles, and cyn-omolgus monkeys at doses ranging from 0.2 to 2 mg/kg.Following a single injection of 2mg/kg ShK-Q16K-PEG[20K]to rats, peak levels of w1000 ng/ml were measured at 24 hand the level dropped to 1 ng/ml 7 days later (Sullivan et al.,2010). After a single subcutaneous injection of 0.5 mg/kg tocynomolgus monkeys, a peak level of w1000 ng/ml wasmeasured during the first 24 h and the level remained high(w100 ng/ml, w25 nM) 10 days later, indicating that thecirculating half-life of this analog is significantly longer inmonkeys than rats. Similar results were obtained withbeagle dogs.

5.4. Safety studies

We performed a number of non-GLP studies in rats todetermine the relative safety of ShK analogs. To determineif ShK-186 compromised the protective immune response,we examined whether rats treated with the peptide couldclear infections of rat-adapted influenza virus or Chlamydia,a common sexually transmitted disease. Dexamethasone,a steroid that broadly suppresses the immune response,was used as a positive control, and saline as a negativecontrol. Rats treatedwith saline or ShK-186 (100 mg/kg/day)cleared influenza infection in 4 days and Chlamydia infec-tion in 14 days, whereas clearance of these infections wassignificantly delayed in dexamethasone-treated rats(Matheu et al., 2008). The small molecule Kv1.3 blockerPAP-1 similarly did not delay the clearance of influenzavirus (Matheu et al., 2008). Thus, ShK-186, at concentra-tions that are therapeutic in DTH, EAE and PIA, does notcompromise the protective immune response against acuteinfectious agents.

ShK-170, ShK-186 and ShK-192 were not cytotoxic ona panel of mammalian cell lines, and were negative in theAmes test (Beeton et al., 2005; Pennington et al., 2009).These peptides exhibit 10,000-fold selectivity for Kv1.3over the HERG (Kv11.1) channel (Table 2), which underliesmany drug-induced cardiac arrhythmias, and ShK-170(10 mg/kg/day) did not alter heart rate variability parame-ters in rats (Beeton et al., 2005). ShK-186 administered oncedaily for 28 days to female Lewis rats by subcutaneousinjection (100 mg/kg or 500 mg/kg per day) did notperceptibly alter blood cell counts or serum chemistryparameters, and it did not cause any histopathologicalchanges in several tissues (brain, heart, lung, liver, kidney,gastrointestinal tract, pancreas, adrenal glands, urinarybladder, uterus, ovaries, thymus, spleen, mesenteric lymphnode and bonemarrow) (Beeton et al., 2006).We did detectlow titer anti-ShK-186 antibodies in these rats (Beetonet al., 2006), but it remains to be determined whetherthese antibodies are neutralizing or have the potential toreduce the long-term therapeutic effectiveness of thepeptide. In a study performed on cynomolgus monkeys atAmgen, no anti-peptide antibodies were detected in theanimals that received low or high doses of ShK-Q16K-PEG[20K], whereas animals that received an intermediatedoses of (0.5 mg/kg) developed such antibodies (Sullivanet al., 2010).

V. Chi et al. / Toxicon 59 (2012) 529–546 543

Amgen has conducted pharmacokinetic and toxicolog-ical studies on a number of ShK analogs in rats, dogs andmonkeys. Single intravenous doses were evaluated in ratsbetween 0.04 and 5 mg/kg. Repeat dose studies in ratsevaluated weekly or every-third-day dosing of 0.1–5.0 mg/kg by the subcutaneous route for up to twoweeks duration.Intravenous administration ShK-Q16K-PEG[20K], over alldoses and subcutaneous administration at �0.3 mg/kgproduced a consistent pattern of lethargy, weight loss,cyanosis, and vascular collapse in rats, with mortality inseveral of the dosing groups (Sullivan et al., 2010). Theseeffects were observed in all rat strains tested (single 5 mg/kg s.c. dose) but not in studies involving administration todog (single 0.2 mg/kg IV or 0.5–2.0 mg/kg s.c. dose), mouse(single 0.2 mg/kg IV dose, 0.6–5.0 mg/kg/day s.c. for 14days), or cynomolgus monkey (single 0.2 mg/kg IV dose,0.7 mg/kg s.c. every third day for two weeks or 0.1–2.0 mg/kg/week s.c. for two weeks) (Sullivan et al., 2010). Tele-metrized rats also showed a reduction in mean arterialblood pressure and increased heart rate following a single0.7 and 2.5 mg/kg s.c. injection (Sullivan et al., 2010).Clinical observations were coincident with a finding ofelevated serum histamine, leading the investigators tospeculate that the adverse observations were due to mastcell degranulation (Sullivan et al., 2010). Positively-chargeddrugs like ShK are known to mediate IgE-independentanaphylactoid reactions (Mathelier-Fusade, 2006). Insupport of this idea, high doses (micromolar concentrations)of ShK-Q16K-PEG[20K], the ShK-Q16K-peptibody and ShK-192 released histamine from rat peritoneal mast cells inculture aseffectivelyasknowndegranulators (e.g. compound48/80). Patch-clamp studies of rat peritoneal mast cells didnot reveal a Kv1.3 current, and the current in these cells wasnot sensitive to ShK analogs. This suggested a non-channel-dependent mechanism. Interestingly, these analogs did notinduce histamine release from human or mouse mast cells,suggesting that the effectwas species-specific (Sullivan et al.,2010). Furthermore, cynomolgus monkeys that received0.7mg/kgof ShK-Q16K-PEG[20K]every thirdday for2weeks,or 2 mg/kg once weekly for 2 weeks, or 0.5 mg/kg onceweekly for four weeks, gained weight normally, no electro-cardiogram changes were observed, and macroscopic andmicroscopic pathology analysis was normal. Total serumexposure to the peptide was significantly higher in theseanimals (meanAUCvalueof584,000ng�h/mlvalue) than inthe rat toxicologystudies. Basedupon theseobservations, it isunlikely that humans receiving ShK analogs will exhibithistamine-mediated responses, since human T cells are>10,000 times more sensitive to Kv1.3 blocker-inhibition byShK compared to the off-target degranulating effect onmastcells.

6. Overview and future directions

Three ion channelsdKv1.3, KCa3.1, Stim/Oraidregulatecalcium signaling during the activation process inlymphocytes. All three of these channels cluster at theimmunological synapse, the narrow cleft formed betweenthe antigen-presenting cell (e.g. dendritic cell or macro-phage or class-switched memory B cell) and the Tlymphocyte. Kv1.3 and KCa3.1 regulate the membrane

potential of lymphocytes and allow efflux of positively-charged ions (Kþ) to counterbalance the influx of Ca2þ

through CRAC (Stim1/Ora1) channels. The contribution ofKv1.3 and KCa3.1 to this process varies according to theirrelative expression level in CCR7þ and CCR7� T cells, orIgDþ and IgD� B cells. Following antigen stimulation,human CCR7þ T cells and IgDþ B cells up-regulate KCa3.1channels within 24 h, which then provides the counter-balancing Kþ efflux to sustain calcium signaling. In contrast,antigen stimulation of human CCR7� cells and IgD� B cells,the two lymphoid subsets implicated in autoimmunediseases, results in the up-regulation of Kv1.3. The sensi-tivity of these two subsets to Kv1.3 channel-blockade offersopportunities for subset-selective immunomodulation inautoimmune disorders and chronic rejection. Kv1.3blockers have been shown to be effective in numerousanimal models of autoimmune disease, and disease-associated autoreactive T cells in the blood and tissues ofpatients with MS, RA and T1DM are Kv1.3high CCR7� TEM-effectors. A synthetic derivative of a sea anemone peptide,ShK-186, blocks Kv1.3 at low picomolar concentrations andshows a high degree of selectivity for Kv1.3 over otherchannels. ShK-186 attenuates calcium signaling in CCR7�

TEM cells, suppresses cytokine production, proliferation andmigration of these cells, and is effective in amelioratingDTH, and reducing disease severity in animal models of MSand RA. Large-scale manufacture of this peptide has beenachieved, a pharmacodynamic method has been developedfor assessing immunomodulation in vivo and pharmacoki-netic studies are underway. We anticipate beginning phase1 human clinical trials with ShK-186 in 2011. Amgen hasmade PEGylated analogs of ShK and a peptibody of ShK, butthe development status of these molecules is not known.

Conflict of interest statement

The authors (KGC, RSN, MWP, HW, CB) are co-foundersof Airmid Inc., which has licensed a patent on PAP-1 fromthe University of California. Airmid has sub-licensed thepatent to Circassia Inc., who is developing the drug asa therapeutic for autoimmune disease (B S-F).

The authors (SI, ET, LL, B S-F) are co-founders of KinetaInc., which has licensed a patent on ShK-186 from theUniversity of California. Kineta is developing the drug asa therapeutic for autoimmune disease.

The authors (KGC, MWP, CB) are consultants to KinetaInc.

Acknowledgments

The work was supported by grant numbers: NIH RO1NS48252 (KGC), NIH 1R43AI085691 (SI, KGC), NIH RO1GM076063 (HW), UC Discovery 445160 (KGC, SI), IacoccaFoundation 485160 (SI, JTL, KGC), and fellowship supportfrom the National Health and Medical Research Council ofAustralia (RSN).

References

Agematsu, K., Hokibara, S., Nagumo, H., Komiyama, A., 2000. CD27:a memory B-cell marker. Immunol. Today 21, 204–206.

V. Chi et al. / Toxicon 59 (2012) 529–546544

Azam, P., Sankaranarayanan, A., Homerick, D., Griffey, S., Wulff, H., 2007.Targeting effector memory T cells with the small molecule Kv1.3blocker PAP-1 suppresses allergic contact dermatitis. J. Invest. Der-matol. 127, 1419–1429.

Beeton, C., Pennington, M.W., Wulff, H., Singh, S., Nugent, D., Crossley, G.,Khaytin, I., Calabresi, P.A., Chen, C.Y., Gutman, G.A., Chandy, K.G.,2005. Targeting effector memory T cells with a selective peptideinhibitor of Kv1.3 channels for therapy of autoimmune diseases. Mol.Pharmacol. 67, 1369–1381.

Beeton, C., Smith, B.J., Sabo, J.K., Crossley, G., Nugent, D., Khaytin, I., Chi, V.,Chandy, K.G., Pennington, M.W., Norton, R.S., 2008. The D-diaste-reomer of ShK toxin selectively blocks voltage-gated Kþ channels andinhibits T lymphocyte proliferation. J. Biol. Chem. 283, 988–997.

Beeton, C., Wulff, H., Barbaria, J., Clot-Faybesse, O., Pennington, M.,Bernard, D., Cahalan, M.D., Chandy, K.G., Beraud, E., 2001. Selectiveblockade of T lymphocyte Kþ channels ameliorates experimentalautoimmune encephalomyelitis, a model for multiple sclerosis. Proc.Natl. Acad. Sci. U. S. A. 98, 13942–13947.

Beeton, C.,Wulff, H., Standifer, N.E., Azam, P.,Mullen, K.M., Pennington,M.W.,Kolski-Andreaco, A., Wei, E., Grino, A., Counts, D.R., Wang, P.H.,Leehealey, C.J., Andrews, B.S., Sankaranarayanan, A., Homerick, D.,Roeck,W.W.,Tehranzadeh, J., Stanhope,K.L.,Zimin,P.,Havel, P.J.,Griffey, S.,Knaus,H.G.,Nepom,G.T.,Gutman,G.A., Calabresi, P.A., Chandy, K.G., 2006.Kv1.3 channels are a therapeutic target for T cell-mediated autoimmunediseases. Proc. Natl. Acad. Sci. U. S. A. 103, 17414–17419.

Boswell, C.A., Tesar, D.B., Mukhyala, K., Theil, F.P., Fielder, P.J., Khawli, L.A.,2010. Effects of charge on antibody tissue distribution and pharma-cokinetics. Bioconjug. Chem. 21, 2153–2163.

Cahalan, M.D., Chandy, K.G., 2009. The functional network of ion channelsin T lymphocytes. Immunol. Rev. 231, 59–87.

Castaneda, O., Harvey, A.L., 2009. Discovery and characterization ofCnidarian peptide toxins that affect neuronal potassium ion channels.Toxicon 54, 1119–1124.

Castaneda, O., Sotolongo, V., Amor, A.M., Stocklin, R., Anderson, A.J.,Harvey, A.L., Engstrom, A., Wernstedt, C., Karlsson, E., 1995. Charac-terization of a potassium channel toxin from the Caribbean seaanemone Stichodactyla helianthus. Toxicon 33, 603–613.

Chandy, K.G., Wulff, H., Beeton, C., Pennington, M., Gutman, G.A.,Cahalan, M.D., 2004. Potassium channels as targets for specificimmunomodulation. Trends Pharmacol. Sci. 25, 280–289.

Chuaprapaisilp, T., Piamphongsant, T., 1978. Treatment of pustularpsoriasis with clofazimine. Br. J. Dermatol. 99, 303–305.

Clark, R.A., 2010. Skin-resident T cells: the ups and downs of on siteimmunity. J. Invest. Dermatol. 130, 362–370.

Corcione, A., Casazza, S., Ferretti, E., Giunti, D., Zappia, E., Pistorio, A.,Gambini, C., Mancardi, G.L., Uccelli, A., Pistoia, V., 2004. Recapitulationof B cell differentiation in the central nervous system of patients withmultiple sclerosis. Proc. Natl. Acad. Sci. U. S. A. 101, 11064–11069.

Cotton, J., Crest, M., Bouet, F., Alessandri, N., Gola, M., Forest, E.,Karlsson, E., Castaneda, O., Harvey, A., Vita, C., Menez, A., 1997. Apotassium-channel toxin from the sea anemone Bunodosoma gran-ulifera, an inhibitor for Kv1 channels. Revision of the amino acidsequence, disulfide-bridge assignment, chemical synthesis, and bio-logical activity. Eur. J. Biochem. 244, 192–202.

Craythorn, J.M., Swartz, M., Creel, D.J., 1986. Clofazimine-induced bull’s-eye retinopathy. Retina 6, 50–52.

Damsker, J.M., Hansen, A.M., Caspi, R.R., 2010. Th1 and Th17 cells:adversaries and collaborators. Ann. N. Y. Acad. Sci. 1183, 211–221.

DeCoursey, T.E., Chandy, K.G., Gupta, S., Cahalan, M.D., 1984. Voltage-gated Kþ channels in human T lymphocytes: a role in mitogenesis?Nature 307, 465–468.

Dorner, T., Lipsky, P.E., 2004. Correlation of circulating CD27high plasmacells and disease activity in systemic lupus erythematosus. Lupus 13,283–289.

Fasth, A.E., Cao, D., van Vollenhoven, R., Trollmo, C., Malmstrom, V., 2004.CD28nullCD4þ T cells-characterization of an effector memory T-cellpopulation in patients with rheumatoid arthritis. Scand. J. Immunol.60, 199–208.

Fordyce, C.B., Jagasia, R., Zhu, X., Schlichter, L.C., 2005. Microglia Kv1.3channels contribute to their ability to kill neurons. J. Neurosci. 25,7139–7149.

Friedrich, M., Krammig, S., Henze, M., Docke, W.D., Sterry, W.,Asadullah, K., 2000. Flow cytometric characterization of lesional Tcells in psoriasis: intracellular cytokine and surface antigen expres-sion indicates an activated, memory effector/type 1 immunopheno-type. Arch. Dermatol. Res. 292, 519–521.

Gendeh, G.S., Chung, M.C., Jeyaseelan, K., 1997a. Genomic structure ofa potassium channel toxin from Heteractis magnifica. FEBS Lett. 418,183–188.

Gendeh, G.S., Young, L.C., de Medeiros, C.L., Jeyaseelan, K., Harvey, A.L.,Chung, M.C., 1997b. A new potassium channel toxin from the seaanemone Heteractis magnifica: isolation, cDNA cloning, and functionalexpression. Biochemistry 36, 11461–11471.

Ghanshani, S., Wulff, H., Miller, M.J., Rohm, H., Neben, A., Gutman, G.A.,Cahalan, M.D., Chandy, K.G., 2000. Up-regulation of the IKCa1potassium channel during T-cell activation: molecular mechanismand functional consequences. J. Biol. Chem. 275, 37137–37149.

Gilhar, A., Bergman, R., Assay, B., Ullmann, Y., Etzioni, A., 2011. Thebeneficial effect of blocking Kv1.3 in the psoriasiform SCID mousemodel. J. Invest. Dermatol. 131, 118–124.

Grgic, I., Wulff, H., Eichler, I., Flothmann, C., Kohler, R., Hoyer, J., 2009.Blockade of T-lymphocyte KCa3.1 and Kv1.3 channels as novelimmunosuppression strategy to prevent kidney allograft rejection.Transplant. Proc. 41, 2601–2606.

Hansen, A., Odendahl, M., Reiter, K., Jacobi, A.M., Feist, E., Scholze, J.,Burmester, G.R., Lipsky, P.E., Dorner, T., 2002. Diminished peripheralblood memory B cells and accumulation of memory B cells in thesalivary glands of patients with Sjogren’s syndrome. Arthritis Rheum.46, 2160–2171.

Hemdan, N.Y., Birkenmeier, G., Wichmann, G., Abu El-Saad, A.M.,Krieger, T., Conrad, K., Sack, U., 2011. Interleukin-17-producing Thelper cells in autoimmunity. Autoimmun. Rev. 9, 785–792.

Honma, T., Kawahata, S., Ishida, M., Nagai, H., Nagashima, Y., Shiomi, K.,2008. Novel peptide toxins from the sea anemone Stichodactylahaddoni. Peptides 29, 536–544.

Hultqvist, M., Olofsson, P., Gelderman, K.A., Holmberg, J., Holmdahl, R.,2006. A new arthritis therapy with oxidative burst inducers. PLoSMed. 3, e348.

Hyodo, T., Oda, T., Kikuchi, Y., Higashi, K., Kushiyama, T., Yamamoto, K.,Yamada, M., Suzuki, S., Hokari, R., Kinoshita, M., Seki, S., Fujinaka, H.,Yamamoto, T., Miura, S., Kumagai, H., 2010. Voltage-gated potassiumchannel Kv1.3 blocker as a potential treatment for rat anti-glomerularbasement membrane glomerulonephritis. Am. J. Physiol. Ren. Physiol.299, F1258–F1269.

Jacobi, A.M., Odendahl, M., Reiter, K., Bruns, A., Burmester, G.R.,Radbruch, A., Valet, G., Lipsky, P.E., Dorner, T., 2003. Correlationbetween circulating CD27high plasma cells and disease activity inpatients with systemic lupus erythematosus. Arthritis Rheum. 48,1332–1342.

Kalman, K., Pennington, M.W., Lanigan, M.D., Nguyen, A., Rauer, H.,Mahnir, V., Paschetto, K., Kem, W.R., Grissmer, S., Gutman, G.A.,Christian, E.P., Cahalan, M.D., Norton, R.S., Chandy, K.G., 1998. ShK-Dap22, a potent Kv1.3-specific immunosuppressive polypeptide. J.Biol. Chem. 273, 32697–32707.

Khanna, R., Roy, L., Zhu, X., Schlichter, L.C., 2001. Kþ channels and themicroglial respiratory burst. Am. J. Physiol. Cell Physiol. 280, C796–C806.

Koo, G.C., Blake, J.T., Shah, K., Staruch, M.J., Dumont, F., Wunderler, D.,Sanchez, M., McManus, O.B., Sirotina-Meisher, A., Fischer, P., Boltz, R.C.,Goetz, M.A., Baker, R., Bao, J., Kayser, F., Rupprecht, K.M., Parsons, W.H.,Tong, X.C., Ita, I.E., Pivnichny, J., Vincent, S., Cunningham, P., Hora Jr., D.,Feeney, W., Kaczorowski, G., et al., 1999. Correolide and derivatives arenovel immunosuppressants blocking the lymphocyte Kv1.3 potassiumchannels. Cell. Immunol. 197, 99–107.

Koo, G.C., Blake, J.T., Talento, A., Nguyen, M., Lin, S., Sirotina, A., Shah, K.,Mulvany, K., Hora Jr., D., Cunningham, P., Wunderler, D.L.,McManus, O.B., Slaughter, R., Bugianesi, R., Felix, J., Garcia, M.,Williamson, J., Kaczorowski, G., Sigal, N.H., Springer, M.S., Feeney, W.,1997. Blockade of the voltage-gated potassium channel Kv1.3 inhibitsimmune responses in vivo. J. Immunol. 158, 5120–5128.

Lanigan, M.D., Kalman, K., Lefievre, Y., Pennington, M.W., Chandy, K.G.,Norton, R.S., 2002. Mutating a critical lysine in ShK toxin alters itsbinding configuration in the pore-vestibule region of the voltage-gated potassium channel, Kv1.3. Biochemistry 41, 11963–11971.

Lee, S.J., Wegner, S.A., McGarigle, C.J., Bierer, B.E., Antin, J.H., 1997. Treat-ment of chronic graft-versus-host disease with clofazimine. Blood 89,2298–2302.

Leyendeckers, H., Voth, E., Schicha, H., Hunzelmann, N., Banga, P.,Schmitz, J., 2002. Frequent detection of thyroid peroxidase-specificIgGþ memory B cells in blood of patients with autoimmune thyroiddisease. Eur. J. Immunol. 32, 3126–3132.

Li, Y., Wang, P., Xu, J., Desir, G.V., 2006. Voltage-gated potassium channelKv1.3 regulates GLUT4 trafficking to the plasma membrane via a Ca2þ-dependent mechanism. Am. J. Physiol. Cell Physiol. 290, C345–C351.

Lioudyno, M.I., Kozak, J.A., Penna, A., Safrina, O., Zhang, S.L., Sen, D.,Roos, J., Stauderman, K.A., Cahalan, M.D., 2008. Orai1 and STIM1moveto the immunological synapse and are up-regulated during T cellactivation. Proc. Natl. Acad. Sci. U. S. A. 105, 2011–2016.

V. Chi et al. / Toxicon 59 (2012) 529–546 545

Lovett-Racke, A.E., Trotter, J.L., Lauber, J., Perrin, P.J., June, C.H., Racke, M.K.,1998. Decreased dependence of myelin basic protein-reactive T cellson CD28-mediated costimulation in multiple sclerosis patients:a marker of activated/memory T cells. J. Clin. Invest. 101, 725–730.

Luik, R.M., Wang, B., Prakriya, M., Wu, M.M., Lewis, R.S., 2008. Oligo-merization of STIM1 couples ER calcium depletion to CRAC channelactivation. Nature 454, 538–542.

Mathelier-Fusade, P., 2006. Drug-induced urticarias. Clin. Rev. AllergyImmunol. 30, 19–23.

Matheu, M.P., Beeton, C., Garcia, A., Chi, V., Rangaraju, S., Safrina, O.,Monaghan, K., Uemura, M.I., Li, D., Pal, S., de la Maza, L.M., Monuki, E.,Flugel, A., Pennington, M.W., Parker, I., Chandy, K.G., Cahalan, M.D.,2008. Imaging of effector memory T cells during a delayed-typehypersensitivity reaction and suppression by Kv1.3 channel block.Immunity 29, 602–614.

Mathew, B.S., Pulimood, A.B., Prasanna, C.G., Ramakrishna, B.S., Chandy, S.J., 2006. Clofazimine induced enteropathy – a case highlighting theimportance of drug induced disease in differential diagnosis. Trop.Gastroenterol. 27, 87–88.

Matteson, D.R., Deutsch, C., 1984. K channels in T lymphocytes: a patchclamp study using monoclonal antibody adhesion. Nature 307,468–471.

McDougall, A.C., Horsfall, W.R., Hede, J.E., Chaplin, A.J., 1980. Splenicinfarction and tissue accumulation of crystals associated with the useof clofazimine (Lamprene; B663) in the treatment of pyodermagangrenosum. Br. J. Dermatol. 102, 227–230.

Miossec, P., Korn, T., Kuchroo, V.K., 2009. Interleukin-17 and type 17helper T cells. N. Engl. J. Med. 361, 888–898.

Miyazaki, Y., Iwabuchi, K., Kikuchi, S., Fukazawa, T., Niino, M., Hirotani, M.,Sasaki, H., Onoe, K., 2008. Expansion of CD4þCD28� T cells producinghigh levels of interferon-{gamma} in peripheral blood of patientswith multiple sclerosis. Mult. Scler. 14, 1044–1055.

Mouhat, S., Visan, V., Ananthakrishnan, S., Wulff, H., Andreotti, N.,Grissmer, S., Darbon, H., De Waard, M., Sabatier, J.M., 2005. Kþ

channel types targeted by synthetic OSK1, a toxin from Orthochirusscrobiculosus scorpion venom. Biochem. J. 385, 95–104.

Mullen, K.M., Rozycka, M., Rus, H., Hu, L., Cudrici, C., Zafranskaia, E.,Pennington, M.W., Johns, D.C., Judge, S.I., Calabresi, P.A., 2006.Potassium channels Kv1.3 and Kv1.5 are expressed on blood-derived dendritic cells in the central nervous system. Ann. Neu-rol. 60, 118–127.

Nagumo, H., Agematsu, K., Kobayashi, N., Shinozaki, K., Hokibara, S.,Nagase, H., Takamoto, M., Yasui, K., Sugane, K., Komiyama, A., 2002.The different process of class switching and somatic hypermutation;a novel analysis by CD27(�) naive B cells. Blood 99, 567–575.

Nair, L.V., Shereef, P.H., 1991. Successful treatment of generalized pustularpsoriasis with clofazimine. Int. J. Dermatol. 30, 151.

Nicolaou, S.A., Neumeier, L., Peng, Y., Devor, D.C., Conforti, L., 2007. TheCa2þ-activated Kþ channel KCa3.1 compartmentalizes in the immu-nological synapse of human T lymphocytes. Am. J. Physiol. CellPhysiol. 292, C1431–C1439.

Norton, R.S., Pennington, M.W., Wulff, H., 2004. Potassium channelblockade by the sea anemone toxin ShK for the treatment of multiplesclerosis and other autoimmune diseases. Curr. Med. Chem. 11, 3041–3052.

Omar, B.A., Flores, S.C., McCord, J.M., 1992. Superoxide dismutase:pharmacological developments and applications. Adv. Pharmacol.23, 109–161.

Panyi, G., Vamosi, G., Bacso, Z., Bagdany, M., Bodnar, A., Varga, Z.,Gaspar, R., Matyus, L., Damjanovich, S., 2004. Kv1.3 potassium chan-nels are localized in the immunological synapse formed betweencytotoxic and target cells. Proc. Natl. Acad. Sci. U. S. A. 101, 1285–1290.

Parizhskaya, M., Youssef, N.N., Di Lorenzo, C., Goyal, R.K., 2001. Clofazi-mine enteropathy in a pediatric bone marrow transplant recipient. J.Pediatr. 138, 574–576.

Park, C.Y., Hoover, P.J., Mullins, F.M., Bachhawat, P., Covington, E.D.,Raunser, S., Walz, T., Garcia, K.C., Dolmetsch, R.E., Lewis, R.S., 2009.STIM1 clusters and activates CRAC channels via direct binding ofa cytosolic domain to Orai1. Cell 136, 876–890.

Penna, A., Demuro, A., Yeromin, A.V., Zhang, S.L., Safrina, O., Parker, I.,Cahalan, M.D., 2008. The CRAC channel consists of a tetramer formedby Stim-induced dimerization of Orai dimers. Nature 456, 116–120.

Pennington, M.W., Byrnes, M., Zaydenberg, I., Khaytin, I., de Chastonay, J.,Krafte, D., Hill, R., Mahnir, V., Volberg, W., Gorczyca, W., Kem, W.R.,1995. Chemical synthesis and characterization of ShK toxin: a potentpotassium channel inhibitor from a sea anemone. Int. J. Pept. ProteinRes. 346, 354–358.

Pennigton, M.W., Lanigan, M.D., Mahnir, V., Rauer, H., McVaugh, C.T,Behm, D., Donaldson, D., Chandy, K.G., Kem, W.R., Norton, R.S., 1999.

Role of disulfide bonds in the structure and potassium channelblocking activity of ShK toxin. Biochemistry 38, 14549–14558.

Pennington, M.W., Mahnir, V., Khaytin, I., Zaydenberg, I., Byrnes, M.,Kem,W.R., 1996. An essential binding surface for ShK toxin interactionwith rat brain potassium channels. Biochemistry 35, 16407–16411.

Pennington,M.W., Beeton, C., Galea, C.A., Smith, B.J., Chi, V., Monaghan, K.P.,Garcia, A., Rangaraju, S., Giuffrida, A., Plank, D., Crossley, G., Nugent, D.,Khaytin, I., Lefievre, Y., Peshenko, I., Dixon, C., Chauhan, S., Orzel, A.,Inoue, T., Hu, X., Moore, R.V., Norton, R.S., Chandy, K.G., 2009. Engi-neering a stable and selective peptide blocker of the Kv1.3 channel in Tlymphocytes. Mol. Pharmacol. 75, 762–773.

Pereira, L.E., Villinger, F., Wulff, H., Sankaranarayanan, A., Raman, G.,Ansari, A.A., 2007. Pharmacokinetics, toxicity, and functional studiesof the selective Kv1.3 channel blocker 5-(4-phenoxybutoxy)psoralenin rhesus macaques. Exp. Biol. Med. (Maywood) 232, 1338–1354.

Pohl, J., Hubalek, F., Byrnes, M.E., Nielsen, K.R., Woods, A., Pennington, M.W., 1995. Assignment of the three disulfide bonds in ShK toxin:a potent potassium channel inhibitor from the sea anemone Sticho-dactyla helianthus. Lett. Pept. Sci. 1, 291–297.

Rauer, H., Pennington, M., Cahalan, M., Chandy, K.G., 1999. Structuralconservation of the pores of calcium-activated and voltage-gatedpotassium channels determined by a sea anemone toxin. J. Biol.Chem. 274, 21885–21892.

Ren, Y.R., Pan, F., Parvez, S., Fleig, A., Chong, C.R., Xu, J., Dang, Y., Zhang, J.,Jiang, H., Penner, R., Liu, J.O., 2008. Clofazimine inhibits human Kv1.3potassium channel by perturbing calcium oscillation in T lympho-cytes. PLoS ONE 3, e4009.

Rus, H., Pardo, C.A., Hu, L., Darrah, E., Cudrici, C., Niculescu, T., Niculescu, F.,Mullen, K.M., Allie, R., Guo, L., Wulff, H., Beeton, C., Judge, S.I., Kerr, D.A.,Knaus, H.G., Chandy, K.G., Calabresi, P.A., 2005. The voltage-gatedpotassium channel Kv1.3 is highly expressed on inflammatory infil-trates in multiple sclerosis brain. Proc. Natl. Acad. Sci. U. S. A. 102,11094–11099.

Sallusto, F., Lenig, D., Forster, R., Lipp, M., Lanzavecchia, A., 1999. Twosubsets of memory T lymphocytes with distinct homing potentialsand effector functions. Nature 401, 708–712.

Schlichter, L., Sidell, N., Hagiwara, S., 1986. Potassium channels mediatekilling by human natural killer cells. Proc. Natl. Acad. Sci. U. S. A. 83,451–455.

Schmitz, A., Sankaranarayanan, A., Azam, P., Schmidt-Lassen, K.,Homerick, D., Hansel, W., Wulff, H., 2005. Design of PAP-1, a selectivesmall molecule Kv1.3 blocker, for the suppression of effector memoryT cells in autoimmune diseases. Mol. Pharmacol. 68, 1254–1270.

Shiomi, K., 2009. Novel peptide toxins recently isolated from sea anem-ones. Toxicon 54, 1112–1118.

Sidell, N., Schlichter, L.C., Wright, S.C., Hagiwara, S., Golub, S.H., 1986.Potassium channels in human NK cells are involved in discrete stagesof the killing process. J. Immunol. 137, 1650–1658.

Straub, S.V., Perez, S.M., Tan, B., Coughlan, K.A., Trebino, C.E., Cosgrove, P.,Buxton, J.M., Kreeger, J.M., Jackson, V.M., 2011. Pharmacologicalinhibition of Kv1.3 fails to modulate insulin sensitivity in diabeticmice or human insulin-sensitive tissues. Am. J. Physiol. Endocrinol.Metab. (Epub date 2011/05/19).

Sullivan, J.K., Miranda, L.P., Gegg, C.V., Hu, S., Belouski, E.J., Murray, J.K.,Nguyen, H., Walker, K.W., Arora, T., Jacobsen, F.W., Li, Y., Boone, T.C.,2010. Selective and Potent Peptide Inhibitors of Kv1.3. WO 2010/108154A2. Amgen, p. 288.

Tucker, K., Overton, J.M., Fadool, D.A., 2008. Kv1.3 gene-targeted deletionalters longevity and reduces adiposity by increasing locomotion andmetabolism in melanocortin-4 receptor-null mice. Int. J. Obes. (Lond.)32, 1222–1232.

Tudor, J.E., Pallaghy, P.K., Pennington, M.W., Norton, R.S., 1996. Solutionstructure of ShK toxin, a novel potassium channel inhibitor from a seaanemone. Nat. Struct. Biol. 3, 317–320.

Tudor, J.E., Pennington, M.W., Norton, R.S., 1998. Ionisation behaviour andsolution properties of the potassium-channel blocker ShK toxin. Eur. J.Biochem. 251, 133–141.

Vennekamp, J., Wulff, H., Beeton, C., Calabresi, P.A., Grissmer, S.,Hansel, W., Chandy, K.G., 2004. Kv1.3-blocking 5-phenylalkoxypsoralens: a new class of immunomodulators. Mol.Pharmacol. 65, 1364–1374.

Viglietta, V., Kent, S.C., Orban, T., Hafler, D.A., 2002. GAD65-reactive T cellsare activated in patients with autoimmune type 1a diabetes. J. Clin.Invest. 109, 895–903.

Vissers, W.H., Arndtz, C.H., Muys, L., Van Erp, P.E., de Jong, E.M., van deKerkhof, P.C., 2004. Memory effector (CD45ROþ) and cytotoxic (CD8þ)T cells appear early in the margin zone of spreading psoriatic lesionsin contrast to cells expressing natural killer receptors, which appearlate. Br. J. Dermatol. 150, 852–859.

V. Chi et al. / Toxicon 59 (2012) 529–546546

Wulff, H., Calabresi, P.A., Allie, R., Yun, S., Pennington, M., Beeton, C.,Chandy, K.G., 2003. The voltage-gated Kv1.3 Kþ channel in effectormemory T cells as new target for MS. J. Clin. Invest. 111, 1703–1713.

Wulff, H., Knaus, H.G., Pennington, M., Chandy, K.G., 2004. Kþ channelexpression during B-cell differentiation: implications for immuno-modulation and autoimmunity. J. Immunol. 173, 776–786.

Wulff, H., Pennington, M., 2007. Targeting effector memory T-cells withKv1.3 blockers. Curr. Opin. Drug Discov. Devel. 10, 438–445.

Xu, J., Koni, P.A., Wang, P., Li, G., Kaczmarek, L., Wu, Y., Li, Y., Flavell, R.A.,Desir, G.V., 2003. The voltage-gated potassium channel Kv1.3 regulatesenergy homeostasis and body weight. Hum. Mol. Genet. 12, 551–559.

Xu, J., Wang, P., Li, Y., Li, G., Kaczmarek, L.K., Wu, Y., Koni, P.A., Flavell, R.A.,Desir, G.V., 2004. The voltage-gated potassium channel Kv1.3 regu-lates peripheral insulin sensitivity. Proc. Natl. Acad. Sci. U. S. A. 101,3112–3117.

Yamaguchi, Y., Hasegawa, Y., Honma, T., Nagashima, Y., Shiomi, K., 2011.Screening and cDNA cloning of Kv1 potassium channel toxins in seaanemones. Mar. Drugs 8, 2893–2905.

Zsiros, E., Kis-Toth, K., Hajdu, P., Gaspar, R., Bielanska, J., Felipe, A.,Rajnavolgyi, E., Panyi, G., 2009. Developmental switch of theexpression of ion channels in human dendritic cells. J. Immunol. 183,4483–4492.