<<

Supplemental material to this article can be found at: http://jpet.aspetjournals.org/content/suppl/2016/12/29/jpet.116.238493.DC1

1521-0103/360/3/466–475$25.00 http://dx.doi.org/10.1124/jpet.116.238493 THE JOURNAL OF PHARMACOLOGY AND EXPERIMENTAL THERAPEUTICS J Pharmacol Exp Ther 360:466–475, March 2017 Copyright ª 2017 by The American Society for Pharmacology and Experimental Therapeutics

Factor XIIa as a Novel Target for : Target Engagement Requirement and Efficacy in a Rabbit Model of Microembolic Signals s

Christopher M. Barbieri, Xinkang Wang, Weizhen Wu, Xueping Zhou, Aimie M. Ogawa, Kim O’Neill, Donald Chu, Gino Castriota, Dietmar A. Seiffert, David E. Gutstein, and Zhu Chen In Vitro Pharmacology (C.M.B., A.M.O., K.O., D.C.) and Cardiometabolic Diseases (X.W., W.W., X.Z., G.C., D.A.S., D.E.G., Z.C.), Merck & Co., Inc., Kenilworth, New Jersey Downloaded from Received October 22, 2016; accepted December 22, 2016

ABSTRACT Factor XII (FXII) plays a critical role in thrombosis. nonhuman primate, and rabbit is more than 99.0%. The effects of What is unclear is the level of occupancy of FXIIa that is rHA-Mut-inf in carotid arterial thrombosis and microembolic signal jpet.aspetjournals.org needed for efficacy and the impact of FXIIa inhibition on cerebral (MES) in middle cerebral artery were assessed simultaneously in embolism. A selective activated FXII (FXIIa) inhibitor, recombi- rabbits. Dose-dependent inhibition was observed for both arterial nant human albumin-tagged mutant Infestin-4 (rHA-Mut-inf), thrombosis and MES. The ED50 of formation was was generated to address these questions. rHA-Mut-inf displayed 0.17 mg/kg i.v. rHA-Mut-inf for the integrated blood flow and potency comparable to the original wild-type HA-Infestin-4 (human 0.16 mg/kg for thrombus weight; the ED50 for MES was FXIIa inhibition constant 5 0.07 and 0.12 nM, respectively), with 0.06 mg/kg. Ex vivo aPTT tracked with efficacy. In summary, markedly improved selectivity against Factor Xa (FXa) and . our findings demonstrated that very high enzyme occupancy

rHA-Mut-inf binds FXIIa, but not FXII , and competitively will be required for FXIIa inhibitors, highlighting the at ASPET Journals on September 28, 2021 inhibits FXIIa activity. Its mode of action is hence akin to high potency and exquisite selectivity necessary for achieving typical small-molecule inhibitors. Plasma shift and aPTT studies efficacy in humans. Our MES studies suggest that targeting with rHA-Mut-inf demonstrated that calculated enzyme occupancy FXIIa may offer a promising strategy for prevention for FXIIa in achieving a putative aPTT doubling target in human, associated with thromboembolic events.

Introduction (Renné et al., 2005; Kleinschnitz et al., 2006; Müller et al., 2009; von Brühl et al., 2012), suggesting that FXII is critically Factor XII (FXII) is the initiator of the intrinsic coagulation involved in pathologic thrombus formation but is dispensable cascade and can be activated via contact with charged surfaces for hemostasis. Emerging studies also suggest that FXII is a to form FXIIa, which then activates FXI and plasma kalli- key mediator in host defense and additional immune- krein. Plasma can activate FXII in a reciprocal inflammatory responses, via its downstream kallikrein/kinin manner to further amplify contact activation (Renné and system (Schmaier, 2016) and interactions with endothelial Gailani, 2007; Renné et al., 2012; Björkqvist et al., 2014). and immune cells (Göbel et al., 2016; Long et al., 2016). FXII is Although human genetics and epidemiologic studies to date do thus a promising target for treating not only thrombotic not yield a clear relationship between FXII deficiency and the events but also a range of other disorders (Martini et al., risk of thrombosis, FXII deficiency in humans is not associated 2014; Göbel et al., 2016; Hopp et al., 2016; Nickel et al., 2016; with , despite marked activated partial Zamolodchikov et al., 2016) without conferring a bleeding risk. thromboplastin time (aPTT) prolongation (Lämmle et al., In recent years, inhibitors for FXII(a) have been generated 1991; Renné and Gailani, 2007). FXII knockout mice were and described in various preclinical models in vitro or in vivo. protected from arterial thrombosis, ischemic stroke, and These agents include monoclonal antibodies (Larsson et al., , while maintaining normal hemostasis 2014; Matafonov et al., 2014), natural peptide or inhibitors (Hagedorn et al., 2010; Yau et al., 2012), small- dx.doi.org/10.1124/jpet.116.238493. molecule inhibitors (Kleinschnitz et al., 2006; Baeriswyl et al., s This article has supplemental material available at jpet.aspetjournals.org. 2015), RNA aptamer (Woodruff et al., 2013), small interfering

ABBREVIATIONS: aPTT, activated partial thromboplastin time; AUC, area under the curve; FXa, Factor Xa; FXII, Factor XII; FXIIa, activated Factor

XIIa; HA, human albumin; koff, off kinetic parameter; kon, on kinetic parameter; Ly60, percentage of lysis at 60 minutes after maximum amplitude; MCA, middle cerebral artery; MES, microembolic signal; PT, prothrombin time; R, clotting time; rHA-Mut-inf, recombinant human albumin–tagged mutant Infestin-4; rHA-WT-inf, recombinant human albumin–tagged wild-type Infestin-4; SPR, surface plasmon resonance; TCD, transcranial Doppler; TEG, thromboelastography; TGA, generation assay; tPA, tissue .

466 FXIIa Inhibition by rHA-Mut-inf 467

RNA (Cai et al., 2015), and antisense oligonucleotide (Revenko transfection. The recombinant protein was captured from the cell et al., 2011). Although results with these various tool mole- culture supernatant by Blue Sepharose 6 Fast Flow resin and eluted in cules in aggregate strengthen the notion that targeting FXII 50 mM Tris with 3 M NaCl, pH 8.5. The purified protein was buffer ’ may provide a benefit in a range of disease settings, some of exchanged into Dulbecco s modified PBS (pH 7.0) and analyzed by the small-molecule or peptide inhibitors used in generating SDS-PAGE and Western blot with HA antibody (Abcam, Cambridge, MA) for confirmation. in vivo data have insufficient selectivity (Hansson et al., 2014; In Vitro Protease Inhibition Assays. Human FXIIa, plasma Korneeva et al., 2014; Xu et al., 2014), hampering the precise kallikrein, thrombin, activated , and plasmin were purchased understanding on target-based pharmacology. Despite the from Enzyme Research Laboratories (South Bend, IN). Human FVIIa, difficulty in achieving the required target selectivity and FXIa, and Xa were purchased from Sekisui Diagnostics (Lexington, potency using a small-molecule inhibitor, this modality poten- MA). Human FIXa was purchased from Haematologic Technologies tially maintains favorable attributes such as oral bioavailability (Essex Junction, VT). Recombinant human tissue plasminogen activa- and shorter pharmacokinetics that are unavailable in certain tor (tPA) was purchased from Hyphen Biomed, S.A.S. (Neuville-sur-Oise, other therapeutic classes. Because small-molecule drugs need to France). Cynomolgus macaque and rabbit FXIIa were prepared by balance among potency, selectivity, and pharmaceutical prop- Evotec, Inc (Princeton, NJ). The 7-amido-4-thifluoromethylcoumarin erties (e.g., permeability and bioavailability), understanding the containing fluorescence substrate CH3SO2-cyclohexyl-Gly-Gly-Arg-AFC was custom synthesized by CPC Scientific (Sunnyvale, CA), whereas levels of target engagement (e.g., enzyme occupancy, residence substrates N-CBZ-Gly-Pro-Arg-AFC and n-acetyl-Gly-Pro-Arg-AFC time) needed for efficacy is an important component in establish-

were purchased from Sigma-Aldrich (St. Louis, MO). Downloaded from ing chemical tractability of the target. The monoclonal antibody All enzymatic reactions were carried out in 50 mM HEPES, 150 mM modality, although likely carrying a superior selectivity profile, NaCl, 5 mM CaCl2, and 0.1% polyethylene-glycol (8000 nominal is mechanistically distinct from small-molecule inhibitors and is molecular weight), at pH 7.4 and 25°C. The change in fluorescence limited in applicable patient settings due to its parenteral route during reaction progression was monitored using a Tecan m200 of administration. (TECAN, Männedorf, Switzerland) plate reader in kinetic mode set It has been observed that FXIIa can be localized to thrombi at 405 nm excitation and 500 nm emission with 9- and 20-nm and can directly interact with , thereby potentially bandwidths, respectively. For all tested, except for FXIIa, jpet.aspetjournals.org modulating clot structure and thrombus stability (Konings the slope of the fluorescence versus time plots was determined by linear regression and plotted versus rHA-Mut-inf concentration to et al., 2011; Kuijpers et al., 2014). Although FXII inhibition or determine IC , which was subsequently converted to inhibition loss of function markedly reduces thrombus formation in 50 constant (KI) using the Cheng-Prusoff equation. For rHA-Mut-inf various models and species, thrombi in FeCl -injured mesen- 3 and rHA-WT-inf inhibition of human FXIIa, KI and binding kinetic teric vessels of FXII knockout mice appeared unstable and parameters (kon and koff) were determined using global progress curve prone to embolization in intravital microscopy studies (Renné analysis for one-step slow and tight binding (Zhang and Windsor, et al., 2005), and FXIIa inhibitor treatment has resulted in 2013). Additional characterization of human, cynomolgus macaque, at ASPET Journals on September 28, 2021 increased shedding of emboli in an ultrasound-induced model and rabbit FXIIa activity was performed after a 2-hour preincubation of atherothrombosis in Apoe2/2 mice (Kuijpers et al., 2014). of the enzyme with either rHA-WT-inf or rHA-Mut-inf to enable the As FXII has been proposed to be a target for stroke and other enzyme-rHA-inf complex to reach equilibrium and was analyzed using thromboembolic disorders (e.g., device-mediated thrombosis global progress curve analysis for tight binding. Plasma Shift Studies. For plasma shift studies, standard FXIIa in extracorporeal membrane oxygenation and cardiopulmo- enzymatic assays were performed as described above except that nary bypass) (Dobrovolskaia and McNeil, 2015; Krupka et al., human, cynomolgus macaque, or rabbit FXIIa was preincubated with 2016), it is important to further understand the role of FXII in rHA-Mut-Inf in either the absence or presence of human, cynomolgus thrombus embolization. macaque, or rabbit 30% heat-inactivated plasma for 2 hours prior to In this report, we set out to examine the potency, selectivity, initiation of the reaction by the addition of substrate. It has been and mode of action of a previously reported FXIIa inhibitor, determined that the 30% heat-inactivated plasma retains protein Inf4mut15 (Campos et al., 2012). We then used this molecule binding activity but is no longer able to be activated for the to address the following two questions: 1) levels of target endogenous coagulation machinery (data not shown). Fluorescence engagement (calculated enzyme occupancy) needed for FXIIa versus time plots were analyzed from global progress curve analysis in generating efficacy; and 2) effects of FXIIa inhibition in a for one-step fast and tight binding (Zhang and Windsor, 2013). To determine the percentage unbound concentration of rHA-Mut- novel model of cerebral microembolic signals (MESs) induced inf in 100% plasma, % Free, the following equation was used: by FeCl3 injury of the carotid artery in rabbits (Zhou et al., 2016a, b). % 5 100 Free  %  PlasmaAssay % PlasmaTarget KI 2 app 1 1 % Â 2 1 Materials and Methods PlasmaAssay KI

Expression and Purification of Human Albumin-Tagged Where % PlasmaTarget is the target percentage plasma (100% in these Mutant Infestin-4 and Recombinant Human Albumin–Tagged studies), % PlasmaAssay is the percentage plasma used in the assay % – PlasmaAssay Wild-Type Infestin-4. Purified recombinant human albumin (HA) (30% in these studies), and KI 2 app is the plasma-shifted KI. tagged wild-type Infestin-4 (rHA-WT-inf) and recombinant HA-tagged Surface Plasmon Resonance. For surface plasmon resonance mutant Infestin-4 (rHA-Mut-inf) were custom generated by Genscript (SPR) experiments, rHA-Mut-inf was immobilized to a standard CM3 (Piscataway, NJ) based on the previously reported Infestin-4 (Campos sensor chip in a BIAcore T200 SPR instrument (GE Healthcare, et al., 2004b) and Inf4mut15 (P2–P49 sequence TRRFVA) (Campos Piscataway, NJ) using the Amine Coupling Kit and the supplied pH et al., 2012) cDNA sequences, respectively. Recombinant expression 5.0 sodium acetate buffer to a density of ∼2000 response units. The methods were as reported earlier (Xu et al., 2014). Briefly, the kinetics of binding of FXIIa and FXII zymogen were monitored using recombinant pUC57 plasmids containing HA-linker ((Gly-Gly-Ser)3)- the single-cycle kinetic method. Running buffer was 10 mM HEPES WT- or Mut-inf were transiently transfected into HEK293 cells. Cell sodium salt, 150 mM NaCl, 5 mM CaCl2, and 0.005% P-20 at pH 7.4. culture supernatant was harvested for purification on day 6 post- Solutions of FXIIa and FXII at 0.1, 1, 10, 30, and 100 nM were 468 Barbieri et al. sequentially injected at a flow rate of 25 ml/min for 240 seconds each. using area under the curve (AUC), calculated by the trapezoidal Dissociation was monitored for 3600 seconds after the last injection. rule, and expressed as a percent of control blood flow as described Data were analyzed using the 1:1 interaction model and the heterol- previously (Wong et al., 2008; Zhou et al., 2016b). At end of study (i.e., ogous ligand model in the BIAcore analysis software (GE Healthcare 60 minutes after FeCl3 injury), the wet thrombus weight was measured Life Sciences). Both fits yielded similar results for the dominant using a balance with a detection limit of 0.001 mg (Mettler Toledo binding event. Excellence Plus XP Series Analytical Balances; Mettler-Toledo, LLC, aPTT, Prothrombin Time, Thrombin Generation Assay, and Columbus, OH). Thromboelastography Assay. In vitro and ex vivo aPTT (triggered The clinical SONARATM TCD System (Nicolet Natus Neurology by aPTT-XL, Pacific Hemostasis, Middletown, VA) and prothrombin Inc., Middleton, WI) was used for continuously monitoring the blood time (PT) (triggered by TriniCLOT PT Excel; Tcoag, Bray, Ireland) flow velocity and MES in the ipsilateral MCA for 60 minutes upon were measured as described before (Xu et al., 2014; Cai et al., 2015). FeCl3 injury. A customized pulse-wave 2-MHz probe (optical density, Thrombin generation assay (TGA) triggered by 0.83 mM ellagic acid 11.3 mm; 90 mm long; focused at 12–25 mm; MTB Medizentechnik (r2 Diagnostics, South Bend, IN) was performed using standard Basler AG, Regensdorf, Switzerland) was fixed by a flexible-arm methods as reported previously (Xu et al., 2014; Cai et al., 2015). magnetic-base holder (McMASTER-CARR, Princeton, NJ) at the Briefly, rHA-Mut-inf at different concentrations was spiked into 60 ml posterior end of zygomatic bone of the rabbits, at an angle of ∼80° of citrated plasma (human, cynomolgus macaque, or rabbit) and against the buccal surface. The MCA was insonated at a depth between incubated at 37°C for 1 hour; a 15-ml trigger was then added to the 19 and 22 mm as described in detail previously (Zhou et al., 2016a). MES plasma and incubated for an additional 10 minutes at 37°C. Fifteen (defined as high-intensity transient signals) was saved and further

microliters of FluCa was subsequently injected into the samples to validated based on the criteria defined by the International Consensus Downloaded from initiate thrombin generation. Thrombinoscope software (Thrombino- Committee as described previously (Ringelstein et al., 1998; Zhou et al., scope, Maastricht, The Netherlands) was used to calculate peak thrombin 2016a,b). as well as additional parameters [lag time, endogenous thrombin Vehicle (10 mM phosphate buffer, pH 7.0) or various doses of rHA- potential, time to peak, and slope (peak divided by the difference Mut-inf (0.1–1.0 mg/kg) were administrated at 2 ml/kg, as a bolus, between lag and time to peak)]. through the ear vein 20 minutes prior to the FeCl3 injury. Doses Thromboelastography (TEG) analysis was carried out on the for rHA-Mut-inf were selected based on its comparable potency to

thromboelastograph coagulation analyzer model 5000 (Haemonetics, rHA-WT-inf in vitro and in our pilot in vivo studies (not shown), and jpet.aspetjournals.org Braintree, MA) as reported previously (Xu et al., 2013). Briefly, rHA- the potency of rHA-WT-inf in our previous studies in rabbits (Xu et al., WT-inf or rHA-Mut-inf at various concentrations was spiked into 2014). freshly collected citrated human whole blood and incubated at room Ex Vivo Measurement of rHA-Mut-inf Plasma Concentrations temperature for 10 minutes. Three hundred forty microliters of the and Clotting Time Assays. Plasma levels of rHA-Mut-inf were blood sample was then transferred into the TEG sample cup preloaded measured by a standard liquid chromatography-mass spectrometry with 20 ml of Ca, 3.4 ml of ellagic acid, and 1 ml of tPA, gently mixed, method as reported previously (Xu et al., 2014) based on the peptide with the TEG recording initiated right after. The final concentration of sequence of Mut-inf (Inf4mut15) (Campos et al., 2012). ellagic acid in the reaction system was 0.031 mM. The final concen- Ex vivo plasma aPTT and PT were determined by standard at ASPET Journals on September 28, 2021 tration of tPA in the reaction system was 1.5 nM, and this concentra- methods using aPTT-XL (Pacific Haemostasis, Waltham, MA) and tion of tPA had been precharacterized as effectively eliciting a TriniCLOT PT Excel (Tcoag) on a KC4 Delta coagulation analyzer phase in the donor’s blood sample without apparently (Tcoag). affecting the coagulation parameters (data not shown). R (reaction Statistical Analysis. Data are presented as the mean 6 S.E. and time), Ly60 (percentage of lysis at 60 minutes after maximum analyzed using one-way analysis of variance followed by Bonferroni amplitude), and additional parameters were recorded by the machine post hoc test in GraphPad Prism (version 7; GraphPad, La Jolla, CA) and analyzed. for comparison among groups with different treatment concentrations Rabbit MES Studies. Male New Zealand White rabbits (weight, or doses. ED50 was determined by a nonlinear four-parameter dose- 2.4–3.0 kg; age range, 10–13 weeks of age; Charles River Canada, response curve fit using GraphPad Prism under the dose ranges Saint-Constant, QC, Canada) were used for the current studies. All of described in each figure legend. Results were considered significant at the procedures were conducted in accordance with the Guide for the P , 0.05. Care and Use of Laboratory Animals as adopted and promulgated by the U.S. National Institutes of Health, and approved by the Animal Care and Use Committee of Merck & Co., Inc. (Kenilworth, NJ). Results The rabbit model of cerebral MES was induced by FeCl3 injury of rHA-Mut-inf Is a Highly Potent and Selective the carotid artery as described in detail previously (Zhou et al., Inhibitor of FXIIa. Despite its picomolar potency for FXIIa 2016a,b). Briefly, animals were anesthetized with a cocktail (50 mg/kg inhibition, the original rHA-WT-inf carries off-target activity i.m. ketamine HCl; Pfizer Inc., New York, NY; and 5 mg/kg xylazine; LLOYD Inc., Shenandoah, IA) as a nonrecovery procedure. The left on several other coagulation and fibrinolytic factors, including common carotid artery was surgically exposed. A Doppler flow probe inhibitory activity on Factor Xa (FXa) and plasmin (Campos (Model 1.5 or 2.0 PRB; Transonic Systems, Ithaca, NY) connected to a et al., 2004b; Xu et al., 2014; Kolyadko et al., 2015). To fully flowmeter (Model T403; Transonic Systems) was used to measure the understand the selectivity profile of rHA-Mut-inf, we exam- artery blood flow with continuous data acquisition by a PowerLab ined its impacts on the enzymatic activities of a comprehen- 16/35 and LabChart Pro system (AD Instruments, Colorado Springs, sive panel of human and rabbit that include CO). Thrombosis was induced by applying two pieces of filter paper coagulation factors, fibrinolytic factors, and . We chose (diameter, 7.4 mm; thickness above and beneath the vessel, 0.5 mm; to determine the apparent KI values for all of the enzymes Life Technologies, Grand Island, NY) presaturated with 30% FeCl3 tested (Table 1; Supplemental Fig. 1) to accurately assess the (anhydrous 98%; ACROS Organics/Thermo Fisher Scientific, Wal- occupancy of rHA-Mut-inf for these enzymes in various tham, MA) to the adventitial surface of the vessel. A piece of para-film was put underneath the vessel to protect the surrounding tissue from studies. To facilitate a direct comparison of the activity of injury. The filter papers were removed after 5 minutes followed by the these enzymes, we used similar fluorescently labeled tripep- washout of residue FeCl3 with warm saline. The blood flow was tide substrates for Michaelis-Menten constant (KM) determi- monitored for 60 minutes from the time of FeCl3 application (as time nations. For the human enzymes, no less than 4000-fold zero). Integrated carotid blood flow over 60 minutes was quantitated selectivity was observed for any off-target human enzyme FXIIa Inhibition by rHA-Mut-inf 469

TABLE 1 rHA-Mut-inf is highly selective for FXIIa over other enzymes involved in coagulation and fibrinolysis

b Enzyme Organism KI Fold Selectivity Substrate Substrate KM

nM mM FXIIa Human 0.07a 1 Acetyl-K-P-R-AFC 0.1 0.78 Rabbit 0.22a 1 0.1 0.72 FXa Human 30,000 430,000 Acetyl-K-P-R-AFC 0.1 2.6 Rabbit 700 3200 0.08 2.3 FIIa Human .5000 .77,000 Acetyl-K-P-R-AFC 0.05 0.05 Rabbit .15,000 .146,000 0.08 0.08 FXIa Human 9000 130,000 Z-G-P-R-AFC 0.1 0.1 Rabbit .26,000 .120,000 0.08 0.60 tPA Human .8100 .120,000 Cyclohexyl G-G-R-AFC 0.25 1.1 Rabbit .26,000 .120,000 0.25 1.4 Plasmin Human 1650 24,000 Acetyl-K-P-R-AFC 0.1 3.5 Rabbit 3300 15,000 0.08 0.94 Kallikrein Human .8500 .120,000 Acetyl-K-P-R-AFC 0.1 0.58 Rabbit 3530 16,000 0.08 0.88 FVIIa Human 98,200 1,000,000 Acetyl-K-P-R-AFC 0.1 5.4 Rabbit .29,000 .130,000 0.08 2.6 Downloaded from FIXa Human .9500 .140,000 Cyclohexyl G-G-R-AFC 0.25 .5mM aPC Human .9100 .130,000 Cyclohexyl G-G-R-AFC 0.25 2.5 uPA Human .7600 .110,000 Cyclohexyl G-G-R-AFC 0.25 0.80 Rabbit .26,000 .120,000 Acetyl-K-P-R-AFC 0.08 0.53 Trypsin Human 285 4000 Acetyl-K-P-R-AFC 0.025 0.025

Acetyl-K-P-R-AFC, n-acetyl-Gly-Pro-Arg-AFC; uPA, ; Z-G-P-R-AFC, N-CBZ-Gly-Pro-Arg-AFC. a As described in the legend to Fig 2. rHA-Mut-inf inhibition of FXIIa is time dependent, and KI was determined using global analysis of reaction progress curves to determine KI and binding kinetics parameters. All other enzymatic assays jpet.aspetjournals.org displayed zero-order kinetics in the presence of rHA-Mut-inf and were analyzed using standard methods. b Fold selectivity is the difference in KI of rHA-Mut-inf for the indicated enzyme vs. the KI of rHA-Mut-inf for FXIIa from either human or rabbit FXIIa, respectively. tested and no less than 3200-fold selectivity was observed for binding kinetics determined directly from the inhibition rabbit enzyme. Using the same method, the KI value for rHA- assays. Furthermore, the mode of FXIIa inhibition for rHA-

WT-inf on FXIIa was determined to be 0.12 nM (Supplemental Mut-inf was characterized using standard enzymatic methods at ASPET Journals on September 28, 2021 Fig. 2). rHA-Mut-inf is thus comparable to rHA-WT-inf for and was shown to be peptide substrate competitive (Fig. 1C). on-target (FXIIa-inhibitory) activity and is much more selec- These findings are in accordance with the earlier character- tive than rHA-WT-inf against other serine proteases. ization of the rHA-WT-inf mode of inhibition (Xu et al., 2014) Mode of Action of rHA-Mut-inf. Figure 1A shows the and strongly support the idea that the inhibitory effects of inhibitory effects of rHA-Mut-inf on the human FXIIa- these molecules are mediated through their direct interac- mediated cleavage of a fluorescent substrate. As has been tions with the active site of FXIIa. previously (Xu et al., 2014) described for rHA-WT-inf, this Effects of rHA-Mut-inf in aPTT, PT, TGA, and TEG. inhibition is slow to reach equilibrium and occurs at concen- Functional activity of rHA-Mut-inf was examined in vitro in trations of inhibitor that are less than the concentration of human, cynomolgus macaque, and rabbit plasma in aPTT and enzyme used for this assay, necessitating taking these factors PT assays, as well as in ellagic acid–triggered TGA. rHA-Mut- into account in the analysis of the data to determine the KI. inf prolonged aPTT in a dose-dependent manner in all three Also, since under the conditions used in this experiment the species (Fig. 2A), with the potency in rabbit far exceeding that time to reach equilibrium was slow relative to the data- in human and cynomolgus macaque. Concentrations needed recording timescale, the binding kinetics of the rHA-Mut-inf- for doubling aPTT are 19, 8.5, and 0.26 mM, respectively, in FXIIa interaction has been elucidated directly from this human, cynomolgus macaque, and rabbit plasma. In the PT inhibition study. This slowly evolving inhibition is driven by assay (Fig. 2B), no change in PT was observed at concentra- the long half-life of dissociation of the rHA-Mut-inf-FXIIa tions ,5 mM and a very modest (∼10%) PT prolongation was 25 21 complex (half-life 5 4.1 hours, koff 5 4.8 Â 10 s ), although observed in human and cynomolgus macaque plasma at 5 21 21 the association rate (kon 5 6.6 Â 10 M × s ) is within 30 mM. rHA-Mut-inf reduced ellagic acid–triggered thrombin normal ranges for this type of interaction. generation in TGA in a dose-dependent manner (Fig. 2C). IC50 Previous studies (Xu et al., 2014; Kolyadko et al., 2015) of peak thrombin is 5.5, 0.69, and 0.05 mM, respectively, in demonstrated that the inhibition of rHA-WT-inf was not human, cynomolgus macaque, and rabbit plasma. directed against the zymogen FXII, but solely against FXIIa. Effects of rHA-Mut-inf on blood coagulation and fibrinolysis To confirm the activated enzyme specificity and binding were also assessed in the whole-blood TEG assay (Fig. 3), in a kinetics for rHA-Mut-inf we used SPR to kinetically monitor direct comparison with rHA-WT-inf, as described in Materials the interactions of FXIIa and FXII zymogen with immobilized and Methods. A threshold concentration (1.5 nM) of tPA was rHA-Mut-inf (Fig. 1B). Results indicated that rHA-Mut-inf is present in the assay system to generate a fibrinolysis phase directed against FXIIa, since no significant binding was without affecting the clotting phase appreciably. Representa- observed for zymogen FXII. In addition, kinetic data from tive TEG traces of the human blood samples treated with SPR (equilibrium dissociation constant [KD] 5 250 pM, kon 5 0 (vehicle), 1, 2, and 4 mM rHA-Mut-inf or rHA-WT-inf are 5 21 21 25 21 1.0 Â 10 M × s , koff 5 3.0 Â 10 s ) agree well with the shown in Fig. 3A. Both agents exerted a dose-dependent 470 Barbieri et al. Downloaded from jpet.aspetjournals.org at ASPET Journals on September 28, 2021

Fig. 2. rHA-Mut-inf markedly prolonged aPTT and reduced intrinsically triggered thrombin generation in plasma with substantially more pro- nounced effects in rabbit than in higher species. rHA-Mut-inf at different Fig. 1. rHA-Mut-inf inhibits FXIIa specifically and reversibly with concentrations were spiked into human, cynomolgus macaque, and rabbit picomolar potency. (A) Time course of inhibition of FXIIa of plasma (black, red, green, respectively) for aPTT (A), PT (B), and ellagic fluorescent substrate by rHA-Mut-inf. Nanomolar concentrations of rHA- acid–triggered TGA (C) analyses. aPTT, PT, and TGA were conducted Mut-inf are indicated. Solid lines represent the nonlinear least squares using standard methods as described in Materials and Methods. Individ- analysis of the data, as described in Materials and Methods. (B) Direct ual plots shown are representative of replicate experiments, n = 2 (A), n = measurement of the interaction between rHA-Mut-inf and FXIIa (filled 2 (B), and n = 2 (C). circles) or between rHA-Mut-inf and FXII zymogen (open circles) via SPR. (C) Assessment of the mechanism of inhibition of FXIIa by rHA-Mut-inf. Solid lines represent the global analysis of the data using a model for compartment of its target. Since coagulation occurs within competitive inhibition. Inset is the double reciprocal plot of the same data, clearly indicating competitive inhibition. Concentrations in nanomolar the plasma, and since rHA-Mut-inf is a large molecule that is concentrations of rHA-Mut-inf are indicated. Individual plots shown are intractable for deriving free fraction in plasma via the representative of replicate experiments, n = 4 (A), n = 2 (B), and n = 3 (C). conventional equilibrium dialysis method, we assessed the impacts of human, cynomolgus macaque, or rabbit heat- inactivated plasma on the observed inhibition of the respective prolongation on R (Fig. 3B, left panel), in a similar manner, FXIIa protein by rHA-Mut-inf (Fig. 4; Table 2) to indirectly with rHA-Mut-inf being slightly more potent than rHA-WT-inf determine the free fraction in plasma. To minimize the impact at the highest concentration tested (4 mM). In contrast, of slow binding by rHA-Mut-inf on the determination of KI,each although rHA-WT-inf produced marked and dose-dependent species of FXIIa was preincubated in rHA-Mut-inf for 2 hours, inhibition of tPA-induced fibrinolysis (Ly60), rHA-Mut-inf did enabling FXIIa  rHA-Mut-inf interactions to reach equilib- not modulate fibrinolysis (Fig. 3B, right panel). rium prior to reaction initiation with substrate (Supplemental Effects of rHA-Mut-inf in Plasma Shift Studies. An Fig. 3). For the plasma of each species, a shift toward lower essential component of the calculation of target engagement potency was observed, with the largest shift occurring for of a therapeutic molecule is its unbound fraction in the human plasma, followed by cynomolgus macaque, and the FXIIa Inhibition by rHA-Mut-inf 471

Fig. 3. Unlike rHA-WT-inf, rHA-Mut-inf does not modulate fibrinolysis in the fibrinolysis of TEG. Both compounds were spiked at varying concentrations into freshly collected citrated human whole blood and

subjected to ellagic acid–triggered TEG in the presence of tPA, as Downloaded from described in Materials and Methods. n = 6 individual donors. (A) TEG traces post-treatment with rHA-Mut-inf (left) and rHA-WT-inf (right) from one representative donor. (B) Summary result of the effects of the two compounds on R (left graph), and on fibrinolysis (%Ly60, right graph). **P , 0.01, and ***P , 0.001 versus vehicle (0 mM).

smallest shift occurring for rabbit plasma. These shifts were jpet.aspetjournals.org used to estimate the amount of rHA-Mut-inf not bound to plasma and thus available for binding to the target FXIIa (Table 2). From these values, the percentage of FXIIa bound to rHA-Mut-inf at steady state was calculated using the concentration of rHA-Mut-inf that prolonged aPTT by 2-fold. Target engagement of 99.2% is needed for doubling aPTT in rabbit plasma, although target engagement of 99.9% is needed at ASPET Journals on September 28, 2021 for doubling aPTT in cynomolgus macaque and human plasma. Effects of rHA-Mut-inf in Rabbit MES Studies. Dose- dependent inhibition of rHA-Mut-inf in both thrombus forma- tion in the carotid artery and MES incidents in the MCA was observed (Fig. 5). The integrated blood flow (AUC, illustrated as the percentage of blood flow relative to baseline) was increased from 55.0 6 5.9% in the vehicle group (n 5 8) to 92.3 6 1.1% for the 1 mg/kg rHA-Mut-inf i.v. group (n 5 6, P , 0.01), with an ED50 of 0.174 mg/kg rHA-Mut-inf (Fig. 5, A and 6 Fig. 4. rHA-Mut-inf interacts with plasma causing an apparent decrease B). Clot weight was reduced from 5.86 0.39 mg (vehicle) to in the inhibition of FXIIa activity. The inhibition of human (A), 0.16 6 0.10 mg (1 mg/kg rHA-Mut-inf, P , 0.001), with an cynomolgus macaque (B), and rabbit (C) FXIIa activity by rHA-Mut-inf ED50 of 0.162 mg/kg rHA-Mut-inf (Fig. 5C). in the absence (black) or presence (red) of 30% heat-inactivated, species- Figure 5D illustrates a representative MES recording in the specific plasma. Individual plots shown are representative of replicated experiments, n = 4 (A), n = 2 (B), and n = 4 (C). MCA in rabbits treated with 0.1 mg/kg rHA-Mut-inf. The mean MES frequency was reduced from 4.35 6 1.13 (vehicle) to 0.0 6 0.0 (1 mg/kg rHA-Mut-inf, P , 0.01), with an ED of 50 The fold aPTT increases at 0.3 and 1 mg/kg (∼1.3-fold and 0.057 mg/kg rHA-Mut-inf (Fig. 5E). The incidence of MES ∼2.5-fold, respectively) with their specific drug exposures are detected in animals was also decreased from 100% (vehicle) to highly consistent with the in vitro spike-in study (Fig. 2A). A 0% (1 mg/kg rHA-Mut-inf) (Fig. 5F). very modest numerical increase in PT post-treatment was Pharmacokinetic and Pharmacodynamic Analysis of observed in all treatment groups, including vehicle, with the rHA-Mut-inf in the Rabbit MES Model. Figure 6A illus- 0.3 and 1 mg/kg groups reaching statistical significance at trates the dose-related increase in plasma rHA-Mut-inf 80 minutes (Fig. 6C). exposure, with 78.3 6 3.6, 192.2 6 10.4, and 478.5 6 47.0 nM observed at 20 minutes, and 76.3 6 2.9, 182.1 6 14.0, and 6 397.8 37.9 nM observed at 80 minutes, for 0.1, 0.3, and 1 mg/kg Discussion i.v. bolus dose, respectively. Thus, drug exposures during the course of arterial thrombosis and MES monitoring were FXII is an emerging promising target for a number of relatively stable. diseases. rHA-WT-inf has been used extensively for interro- Ex vivo aPTT confirmed the pharmacodynamic response for gating the role of FXII in thrombosis, hemostasis, and rHA-Mut-inf (Fig. 6B). Significant increase in aPTT was additional pathologic settings. However, studies from us (Xu observed for 0.3 and 1 mg/kg rHA-Mut-inf dosing groups. et al., 2014) and others (Kolyadko et al., 2015) demonstrated 472 Barbieri et al.

TABLE 2 Enzyme occupancy for FXIIa in three species for a putative target of aPTT doubling

a b c d Species KI KI-app 30% plasma % Free in 100% Plasma rHA-Mut-inf at 2Â aPTT % XIIaBound at 2Â aPTT

nM nM Human 0.08 4.19 0.55% 19,000 99.93% Cynomolgus macaque 0.20 2.05 3.21% 8500 99.93% Rabbit 0.22 0.82 9.97% 260 99.15%

a KI-app 30% plasma indicates the value of KI observed in the presence of 30% heat-inactivated plasma. bEstimated using the equation described in the Materials and Methods. cMut-inf at 2Â aPTT is the concentration of rHA-Mut-inf associated with a 2-fold increase in aPTT time. d % XIIaBound at 2Â aPTT is the percentage of XIIa in complex with rHA-Mut-inf at concentration yielding 2Â aPTT. that rHA-WT-inf carries significant off-target activity on FXa carry an improved selectivity profile. Our in vitro enzyme and plasmin that may skew its effect on thrombosis and assays (Table 1) demonstrated that the HA fusion with hemostasis in vivo at high concentrations. We were thus Inf4mut15, dubbed as rHA-Mut-inf in this report, has an interested in exploring alternative FXIIa inhibitors that may on-target effect on FXIIa that is comparable with that on Downloaded from jpet.aspetjournals.org at ASPET Journals on September 28, 2021

Fig. 5. Dose-dependent effect of rHA-Mut-inf on FeCl3-induced arterial thrombosis and cerebral MES. rHA-Mut-inf dose-dependently inhibited 30% FeCl3-induced carotid arterial thrombosis (vehicle, n = 8; 0.1 mg/kg i.v., n = 8; 0.3 mg/kg i.v., n = 9; and 1.0 mg/kg i.v., n = 6) as illustrated using carotid blood flow within 60 minutes upon FeCl3 injury (A), AUC of the integrated blood flow (B), and reduction in clot weight (C). MES was monitored simultaneously in the ipsilateral MCA to the FeCl3 injury. (D) Representative image for MES detection by TCD in a vehicle-treated animal is illustrated with a snapshot of TCD recording in a period of 4 seconds. The MES was indicated in an arrow determined by Sonora software and confirmed manually. Mean frequency (E) and incidence (F) of MES in MCA was also dose-dependently reduced. *P , 0.05, **P , 0.01, and ***P , 0.001 versus vehicle. FXIIa Inhibition by rHA-Mut-inf 473

its activity, resulting in a lesser prolongation of R compared with rHA-Mut-inf at 4 mM. Although the current tPA-induced fibrinolysis TEG may not mimic all components in vivo by the way in which FXIIa modulates fibrinolysis, the system has been previously benchmarked with a number of thrombin inhibitors yielding results that are consistent with expectations (Xu et al., 2013), and the contrast we observed between rHA-Mut- and WT-inf in fibrinolysis is consistent with their in vitro activity in plasmin in a purified system. Overall, these in vitro results demonstrate that rHA-Mut-inf indeed is a much more selective molecule than rHA-WT-inf, which is consistent with findings from another group using different methods (Kolyadko et al., 2015). As the potency of rHA-Mut-inf exceeds that of 3F7 (a monoclonal antibody inhibitor of FXIIa, KD5 6.2 nM) (Larsson et al., 2014) and FXII618 (a peptide inhibitor of FXIIa, KI 5 22 nM) (Baeriswyl et al., 2015), rHA-Mut-inf is an attractive tool molecule for future research on the role of FXII in vivo in various pathologic Downloaded from settings. With regard to the in vitro aPTT studies, rHA-Mut-inf was much less potent in human and nonhuman primate plasma compared with rabbit plasma (Fig. 2A). This is consistent with what was observed with 3F7, and with the hypothesis that FXII-independent activation of FXI may play a more prom- jpet.aspetjournals.org inent role in primates than in lower species (Gailani et al., 2015). For the in vitro PT studies, appreciable increases in PT were not observed except at very high concentrations (20- 30 mM), and these PT increases could possibly be due to the residual off-target activity of rHA-Mut-inf in FXa, FIIa, and/or FVIIa (Table 1). It is therefore important to monitor the actual

exposures of rHA-Mut-inf to appropriately interpret the at ASPET Journals on September 28, 2021 results of in vivo studies. With regard to the mode of inhibition, our previous analysis (Xu et al., 2014) as well as X-ray crystallography (Campos et al., 2004a, 2012) and docking studies (Kolyadko et al., 2015) with the active proteases suggest that the Infestin family of peptides acts as competitive, reversible inhibitors of the active site of the coagulation factors and additional serine proteases. Our kinetics studies with rHA-Mut-inf using the small tripeptide substrate (Fig. 1) confirmed this notion. Our SPR- based binding studies (Fig. 1B) further demonstrated that Fig. 6. Pharmacokinetic and pharmacodynamics responses for rHA-Mut-inf rHA-Mut-inf specifically binds FXIIa but not FXII zymogen. It treatment on FeCl3-induced arterial thrombosis and cerebral MES. Plasma is therefore unlikely that rHA-Mut-inf blocks the activation samples were prepared in animals of each experimental group in Fig. 5 prior of FXII zymogen to form FXIIa. to or 20 minutes after (time of FeCl3 injury) and 80 minutes after rHA-Mut-inf As rHA-Mut-inf is a highly selective inhibitor of FXIIa and i.v. bolus dosing. n = 8, 8, 9, and 6, for the vehicle group, and 0.1, 0.3, and 1.0 mg/kg treatment group, respectively. Plasma drug exposures are its mode of inhibition is akin to typical small-molecule illustrated in (A). Ex vivo aPTT and PT are depicted in (B) and (C), inhibitors of serine proteases, we were interested in using it respectively. Statistical analyses for aPTT and PT were conducted for each in assessing the level of target engagement or enzyme dosing group in comparison with its own baseline. **P , 0.01, ***P , 0.001 versus respective baselines. occupancy needed for FXIIa for the active site inhibitor mechanism. From a drug discovery standpoint, this is a very relevant question, as higher levels of requisite on-target WT-inf, with higher selectivity against FXa and plasmin, enzyme occupancy will dictate a more stringent selectivity which is consistent with the findings of another report of profile and often a higher hurdle for the absorption, distribu- in vitro characterization of the Infestin-4 mutants without HA tion, metabolism, and excretion properties of the molecule, fusion (Kolyadko et al., 2015). In the whole-blood TEG assay, especially for an intended oral agent. Using a novel plasma at 1 and 2 mM, rHA-Mut-inf and WT-inf prolonged clotting shift assay (Fig. 4), we first determined the percentage of time (R) to the same extent, whereas at 4 mM rHA-Mut-inf was plasma protein binding (likely mediated by HA) for rHA-Mut- more potent; rHA-Mut-inf did not modulate tPA-induced inf across species. We then selected a putative target of aPTT fibrinolysis, whereas rHA-WT-inf dose-dependently sup- doubling in calculating enzyme occupancy. Although the pressed fibrinolysis, likely due to its off-target activity on levels of aPTT prolongation needed for efficacy plasmin. It is possible that at the highest dose (4 mM) tested, via targeting FXII in humans are unknown, previous findings the strong inhibition of plasmin by rHA-WT-inf has dampened with a FXII monoclonal antibody (15H8, targeting heavy chain 474 Barbieri et al. of FXII) in baboon (Matafonov et al., 2014) suggest that 2Â in developing an oral small-molecule drug-targeting active aPTT prolongation is associated with appreciable antithrom- site of FXIIa. Alternative strategies and alternative modes of botic efficacy in nonhuman primate and is a reasonable aPTT inhibition in targeting FXII thus should be considered. target, recognizing that aPTT target for translatable efficacy for different modes of inhibition could be different. The Acknowledgments calculated enzyme occupancies for FXIIa active site inhibitors The authors thank Weixun Wang and Xu Wang for analysis of for doubling aPTT in human, cynomolgus macaque, and rabbit plasma rHA-Mut-inf levels. plasma were 99.9%, 99.9%, and 99.2%, respectively (Table 2). This is in contrast with the previous findings that 75% of the Authorship Contributions antisense oligonucleotide–mediated loss of FXII zymogen Participated in research design: Barbieri, Wang, Seiffert, Gutstein, (Revenko et al., 2011) or 82% of the small interfering and Chen Conducted experiments: Barbieri, Wang, Wu, Zhou, Ogawa, RNA–mediated loss of FXII zymogen (Cai et al., 2015) was O’Neill, Chu, and Castriota sufficient in delivering robust antithrombotic efficacy in Contributed new reagents or analytic tools: Barbieri, Wang, and various models in rodents, with the caveat that there could Chen be a species difference in target-based pharmacology. The very Performed data analysis: Barbieri, Wang, Wu, Zhou, Ogawa, high level of calculated enzyme occupancy needed for FXIIa O’Neill, Chu, Castriota, and Chen active site inhibitors is perhaps unsurprising considering that Wrote or contributed to the writing of the manuscript: Barbieri, FXIIa is the most upstream factor in the intrinsic cascade and Wang, Seiffert, Gutstein, and Chen Downloaded from any “leakage” with a tiny amount of FXIIa may be sub- References sequently cascaded and amplified. One precedent of a very Ankrom W, Wood HB, Xu J, Geissler W, Bateman T, Chatterjee MS, Feng KI, high level of target engagement for an active site inhibitor of a Metzger JM, Strapps WR, Tadin-Strapps M, et al. (2016) Preclinical and trans- coagulation factor is , a FXa inhibitor, requiring .99% lational evaluation of coagulation factor IXa as a novel therapeutic target. Phar- macol Res Perspect 4:e00207. calculated enzyme occupancy to deliver clinically relevant Baeriswyl V, Calzavarini S, Chen S, Zorzi A, Bologna L, Angelillo-Scherrer A, efficacy (Ankrom et al., 2016). A third potential reason could and Heinis C (2015) A synthetic factor XIIa inhibitor blocks selectively intrinsic jpet.aspetjournals.org coagulation initiation. ACS Chem Biol 10:1861–1870. be that thrombin-mediated activation of FXI will bypass FXII, Björkqvist J, Nickel KF, Stavrou E, and Renné T (2014) In vivo activation and thus requiring more complete inhibition of FXIIa and immedi- functions of the protease factor XII. Thromb Haemost 112:868–875. Bonati LH, Jongen LM, Haller S, Flach HZ, Dobson J, Nederkoorn PJ, Macdonald S, ate shut-down of thrombin generation. Gaines PA, Waaijer A, Stierli P, et al.; ICSS-MRI study group (2010) New As FXII deficiency confers no bleeding risk, targeting FXII ischaemic brain lesions on MRI after stenting or endarterectomy for symptomatic carotid stenosis: a substudy of the International Carotid Stenting Study (ICSS). for the treatment of ischemic stroke could be a promising Lancet Neurol 9:353–362. strategy. MES has been considered as an independent pre- Cai TQ, Wu W, Shin MK, Xu Y, Jochnowitz N, Zhou Y, Hoos L, Bentley R, Strapps W,

Thankappan A, et al. (2015) Factor XII full and partial null in rat confers robust at ASPET Journals on September 28, 2021 dictor of stroke or transient ischemic attack (Gao et al., 2004; antithrombotic efficacy with no bleeding. Blood Coagul Fibrinolysis 26:893–902. Markus et al., 2005), and MES originating from the carotid Campos IT, Guimarães BG, Medrano FJ, Tanaka AS, and Barbosa JA (2004a) artery is considered to be one of the primary sources of stroke Crystallization, data collection and phasing of infestin 4, a factor XIIa inhibitor. Acta Crystallogr D Biol Crystallogr 60:2051–2053. in patients (Bonati et al., 2010; Yavin et al., 2011). Our Campos IT, Tanaka-Azevedo AM, and Tanaka AS (2004b) Identification and char- findings in the rabbit MES study thus provided preclinical acterization of a novel factor XIIa inhibitor in the hematophagous insect, Triatoma infestans (Hemiptera: Reduviidae). FEBS Lett 577:512–516. evidence in supporting the therapeutic potential of FXIIa Campos IT, Souza TA, Torquato RJ, De Marco R, Tanaka-Azevedo AM, Tanaka AS, inhibition in the treatment and prevention of ischemic stroke and Barbosa JA (2012) The Kazal-type inhibitors infestins 1 and 4 differ in spec- ificity but are similar in three-dimensional structure. Acta Crystallogr D Biol and additional thromboembolic disorders. Our findings also Crystallogr 68:695–702. strengthened the findings of previous reports (Kleinschnitz Dobrovolskaia MA and McNeil SE (2015) Safe anticoagulation when heart and are “on vacation”. Ann Transl Med 3 (Suppl 1):S11. et al., 2006; Krupka et al., 2016) on the beneficial effects of Gailani D, Bane CE, and Gruber A (2015) Factor XI and contact activation as targets targeting FXII in cerebral ischemia and reperfusion from for antithrombotic therapy. J Thromb Haemost 13:1383–1395. Gao S, Wong KS, Hansberg T, Lam WW, Droste DW, and Ringelstein EB (2004) different models and species. On the other hand, as more than Microembolic signal predicts recurrent cerebral ischemic events in acute stroke 2.0Â aPTT prolongation was needed to fully restore blood flow patients with middle cerebral artery stenosis. Stroke 35:2832–2836. Göbel K, Pankratz S, Asaridou CM, Herrmann AM, Bittner S, Merker M, Ruck T, in this rabbit MES model (Fig. 5A; Fig. 6B), more than 99.0% Glumm S, Langhauser F, Kraft P, et al. (2016) Blood coagulation factor XII drives target engagement will be needed, based on our estimate from adaptive immunity during neuroinflammation via CD87-mediated modulation of dendritic cells. Nat Commun 7:11626. in vitro analysis (Table 2). It is to be noted that although our Hagedorn I, Schmidbauer S, Pleines I, Kleinschnitz C, Kronthaler U, Stoll G, Dickneite scheme of dosing at 20 minutes prior to vascular injury may G, and Nieswandt B (2010) Factor XIIa inhibitor recombinant human albumin ensure adequate and consistent coverage of rHA-Mut-inf Infestin-4 abolishes occlusive arterial thrombus formation without affecting bleeding. Circulation 121:1510–1517. during the study, based on the findings of a prior pharmaco- Hansson KM, Nielsen S, Elg M, and Deinum J (2014) The effect of corn trypsin kinetic study with rHA-WT-inf (Hagedorn et al., 2010), the inhibitor and inhibiting antibodies for FXIa and FXIIa on coagulation of plasma and whole blood. J Thromb Haemost 12:1678–1686. implications of FXIIa inhibition for stroke prophylaxis may Hopp S, Albert-Weissenberger C, Mencl S, Bieber M, Schuhmann MK, Stetter C, need to be further addressed in studies with tool molecules Nieswandt B, Schmidt PM, Monoranu CM, Alafuzoff I, et al. (2016) Targeting coagulation factor XII as a novel therapeutic option in brain trauma. Ann Neurol that can allow more flexible dosing paradigms. Another 79:970–982. limitation of our study is that neurologic deficits and ischemic Kleinschnitz C, Stoll G, Bendszus M, Schuh K, Pauer HU, Burfeind P, Renné C, Gailani D, Nieswandt B, and Renné T (2006) Targeting coagulation factor XII brain injury could not be assessed under the current experi- provides protection from pathological thrombosis in cerebral ischemia without – mental setting (i.e., a nonrecovery procedure and with FeCl3 interfering with hemostasis. J Exp Med 203:513 518. Kolyadko VN, Lushchekina SV, Vuimo TA, Surov SS, Ovsepyan RA, Korneeva VA, injury of the carotid artery for 60 minutes to end of study). Vorobiev II, Orlova NA, Minakhin L, Kuznedelov K, et al. (2015) New Infestin-4 In summary, rHA-Mut-inf is a highly potent and selective Mutants with Increased Selectivity against Factor XIIa. PLoS One 10:e0144940. Konings J, Govers-Riemslag JW, Philippou H, Mutch NJ, Borissoff JI, Allan P, active site inhibitor of FXIIa. Rabbit MES studies with rHA- Mohan S, Tans G, Ten Cate H, and Ariëns RA (2011) Factor XIIa regulates the Mut-inf suggest that FXIIa inhibition may effectively reduce structure of the fibrin clot independently of thrombin generation through direct interaction with fibrin. Blood 118:3942–3951. both thrombus formation and thrombus embolization. How- Korneeva VA, Trubetskov MM, Korshunova AV, Lushchekina SV, Kolyadko VN, ever, significant challenges in chemical tractability may exist Sergienko OV, Lunin VG, Panteleev MA, and Ataullakhanov FI (2014) Interactions FXIIa Inhibition by rHA-Mut-inf 475

outside the proteinase-binding loop contribute significantly to the inhibition of coagulation factor XII inhibits thrombosis in mice without increased risk of activated coagulation factor XII by its canonical inhibitor from corn. J Biol Chem bleeding. Blood 118:5302–5311. 289:14109–14120. Ringelstein EB, Droste DW, Babikian VL, Evans DH, Grosset DG, Kaps M, Markus Krupka J, May F, Weimer T, Pragst I, Kleinschnitz C, Stoll G, Panousis C, Dickneite HS, Russell D, and Siebler M; International Consensus Group on Microembolus G, and Nolte MW (2016) The coagulation factor XIIa inhibitor rHA-Infestin-4 im- Detection (1998) Consensus on microembolus detection by TCD. Stroke 29: proves outcome after cerebral ischemia/reperfusion injury in rats. PLoS One 11: 725–729. e0146783. Schmaier AH (2016) The contact activation and kallikrein/kinin systems: patho- Kuijpers MJ, van der Meijden PE, Feijge MA, Mattheij NJ, May F, Govers-Riemslag physiologic and physiologic activities. J Thromb Haemost 14:28–39. J, Meijers JC, Heemskerk JW, Renné T, and Cosemans JM (2014) Factor XII von Brühl ML, Stark K, Steinhart A, Chandraratne S, Konrad I, Lorenz M, Khandoga regulates the pathological process of thrombus formation on ruptured plaques. A, Tirniceriu A, Coletti R, Köllnberger M, et al. (2012) Monocytes, neutrophils, and Arterioscler Thromb Vasc Biol 34:1674–1680. cooperate to initiate and propagate in mice in vivo. JExp Lämmle B, Wuillemin WA, Huber I, Krauskopf M, Zürcher C, Pflugshaupt R, Med 209:819–835. and Furlan M (1991) Thromboembolism and bleeding tendency in congenital factor Wong PC, Watson CA, and Crain EJ (2008) Arterial antithrombotic and bleeding XII deficiency–a study on 74 subjects from 14 Swiss families. Thromb Haemost 65: time effects of apixaban, a direct factor Xa inhibitor, in combination with anti- 117–121. therapy in rabbits. J Thromb Haemost 6:1736–1741. Larsson M, Rayzman V, Nolte MW, Nickel KF, Björkqvist J, Jämsä A, Hardy MP, Woodruff RS, Xu Y, Layzer J, Wu W, Ogletree ML, and Sullenger BA (2013) Inhib- Fries M, Schmidbauer S, Hedenqvist P, et al. (2014) A factor XIIa inhibitory an- iting the intrinsic pathway of coagulation with a factor XII-targeting RNA tibody provides thromboprotection in extracorporeal circulation without increasing aptamer. J Thromb Haemost 11:1364–1373. bleeding risk. Sci Transl Med 6:222ra17. XuY,CaiTQ,CastriotaG,ZhouY,HoosL,JochnowitzN,LoewrigkeitC,CookJA, Long AT, Kenne E, Jung R, Fuchs TA, and Renné T (2016) Contact system revisited: Wickham A, Metzger JM, et al. (2014) Factor XIIa inhibition by Infestin-4: an interface between , coagulation, and innate immunity. J Thromb in vitro mode of action and in vivo antithrombotic benefit. Thromb Haemost 111: Haemost 14:427–437. 694–704. Markus HS, Droste DW, Kaps M, Larrue V, Lees KR, Siebler M, and Ringelstein EB Xu Y, Wu W, Wang L, Chintala M, Plump AS, Ogletree ML, and Chen Z (2013) (2005) Dual antiplatelet therapy with and in symptomatic ca- Differential profiles of thrombin inhibitors (, , , and rotid stenosis evaluated using doppler embolic signal detection: the Clopidogrel ) in the thrombin generation assay and thromboelastography in vitro. and Aspirin for Reduction of Emboli in Symptomatic Carotid Stenosis (CARESS) Blood Coagul Fibrinolysis 24:332–338. Downloaded from trial. Circulation 111:2233–2240. Yau JW, Stafford AR, Liao P, Fredenburgh JC, Roberts R, Brash JL, and Weitz JI Martini S, Nair V, Keller BJ, Eichinger F, Hawkins JJ, Randolph A, Böger CA, (2012) Corn coating attenuates the prothrombotic properties of Gadegbeku CA, Fox CS, Cohen CD, et al.; European Renal cDNA Bank; C-PROBE catheters in vitro and in vivo. Acta Biomater 8:4092–4100. Cohort; CKDGen Consortium (2014) Integrative biology identifies shared tran- Yavin D, Roberts DJ, Tso M, Sutherland GR, Eliasziw M, and Wong JH (2011) scriptional networks in CKD. J Am Soc Nephrol 25:2559–2572. Carotid endarterectomy versus stenting: a meta-analysis of randomized trials. Can Matafonov A, Leung PY, Gailani AE, Grach SL, Puy C, Cheng Q, Sun MF, McCarty J Neurol Sci 38:230–235. OJ, Tucker EI, Kataoka H, et al. (2014) Factor XII inhibition reduces thrombus Zamolodchikov D, Renné T, and Strickland S (2016) The Alzheimer’s disease peptide formation in a primate thrombosis model. Blood 123:1739–1746. b-amyloid promotes thrombin generation through activation of coagulation factor Müller F, Mutch NJ, Schenk WA, Smith SA, Esterl L, Spronk HM, Schmidbauer S, XII. J Thromb Haemost 14:995–1007. jpet.aspetjournals.org Gahl WA, Morrissey JH, and Renné T (2009) Platelet polyphosphates are proin- Zhang R and Windsor WT (2013) In vitro kinetic profiling of hepatitis C virus NS3 flammatory and procoagulant mediators in vivo. Cell 139:1143–1156. protease inhibitors by progress curve analysis. Methods Mol Biol 1030:59–79. Nickel KF, Labberton L, Long AT, Langer F, Fuchs TA, Stavrou EX, Butler LM, Zhou X, Kurowski S, Wu W, Desai K, Chu L, Gutstein DE, Seiffert D, and Wang X and Renné T (2016) The polyphosphate/factor XII pathway in cancer-associated (2016a) A rabbit model of cerebral microembolic signals for translational research: thrombosis: novel perspectives for safe anticoagulation in patients with malig- preclinical validation for aspirin and clopidogrel. J Thromb Haemost 14: nancies. Thromb Res 141 (Suppl 2):S4–S7. 1855–1866. Renné T and Gailani D (2007) Role of factor XII in hemostasis and thrombosis: Zhou X, Wu W, Chu L, Gutstein DE, Seiffert D, and Wang X (2016b) Apixaban clinical implications. Expert Rev Cardiovasc Ther 5:733–741. inhibits cerebral microembolic signals derived from carotid arterial thrombosis in Renné T, Pozgajová M, Grüner S, Schuh K, Pauer HU, Burfeind P, Gailani D, rabbits. J Pharmacol Exp Ther 358:405–412.

and Nieswandt B (2005) Defective thrombus formation in mice lacking coagulation at ASPET Journals on September 28, 2021 factor XII. J Exp Med 202:271–281. Renné T, Schmaier AH, Nickel KF, Blombäck M, and Maas C (2012) In vivo roles of Address correspondence to: Zhu Chen, Cardiometabolic Diseases, MRL, factor XII. Blood 120:4296–4303. K15-C409, Galloping Hill Road, Kenilworth, NJ 07033. E-mail: zchen17@ Revenko AS, Gao D, Crosby JR, Bhattacharjee G, Zhao C, May C, Gailani D, Monia hotmail.com BP, and MacLeod AR (2011) Selective depletion of plasma or The Journal of Pharmacology And Experimental Therapeutics, JPET#238493

Factor XIIa as a novel target for thrombosis: target engagement requirement and efficacy in a rabbit model of microembolic signals

Christopher M. Barbieri, Xinkang Wang, Weizhen Wu, Xueping Zhou, Aimie M. Ogawa,

Kim O’Neill, Donald Chu, Gino Castriota, Dietmar A. Seiffert, David E. Gutstein, Zhu

Chen

In Vitro Pharmacology (C.M.B., A.M.O., K.O., D.C.), Cardiometabolic Diseases (X.W.,

W.W., X.Z., G.C., D.A.S., D.E.G., Z.C.), Merck &Co., Inc., Kenilworth, NJ USA

The Journal of Pharmacology And Experimental Therapeutics, JPET#238493

A

B

Supplemental Figure 1. rHA-Mut-inf inhibition of selected human and rabbit serine

proteases. Concentration dependent effects of rHA-Mut-inf on selected human (A) or rabbit (B) serine proteases. The enzymes tested are indicated in the legend. Solid lines represent the nonlinear least squares analysis of the data using a four parameter dose- response analysis.

The Journal of Pharmacology And Experimental Therapeutics, JPET#238493

[rHA-WT-inf] 6000 (nM) 0 20 4000 8 3.2 2000 1.28 Fluorescence Units)(Arbitrary 0.51 0.21 0 0 2000 4000 6000 8000 0.08 Time (s)

Supplemental Figure 2. rHA-WT-inf inhibits human FXIIa with pM potency Time

course of inhibition of FXIIa proteolysis of fluorescent substrate by rHA-WT-inf.

Concentrations in nM of rHA-WT-inf are indicated. Solid lines represent the nonlinear

least squares analysis of the data as described in Materials and Methods.

The Journal of Pharmacology And Experimental Therapeutics, JPET#238493

A

B

C

Supplemental Figure 3. rHA-Mut-inf inhibition of human, cynomolgus, and rabbit

FXIIa Inhibition of human (A), cynomolgus (B), or rabbit (C) FXIIa protease activity by rHA-Mut-inf following a 2 hr pre-incubation of the enzymes with the inhibitor. The Journal of Pharmacology And Experimental Therapeutics, JPET#238493

Concentrations of rHA-Mut-inf are indicated in the legend. Solid lines represent the nonlinear least squares analysis of the data as described in Materials and Methods.