throbber
CMLS, Cell. Mol. Life Sci. 59 (2002) 189–192
`1420-682X/02/020189-04 $ 1.50 + 0.20/0
`© Birkhäuser Verlag, Basel, 2002
`
`CMLS Cellular and Molecular Life Sciences
`
`Visions & Reflections
`
`Factor Xa – a promising target for drug development
`
`B. Kaiser
`
`Friedrich Schiller University Jena, Centre for Vascular Biology and Medicine Erfurt, Nordhäuser Str. 78,
`99089 Erfurt (Germany), Fax + 49 361 2113502, e-mail: kaiser@zmkh.ef.uni-jena.de
`
`Received 12 October 2001; received after revision 7 December 2001; accepted 4 January 2002
`
`Background
`
`Based on their pivotal role in the blood coagulation cas-
`cade, the serine proteases thrombin and factor Xa (FXa)
`have become a focus of interest for the development of
`new anticoagulant/antithrombotic drugs. Direct inhibi-
`tion of these key enzymes may offer new ways to affect
`the blood clotting process as well as other actions of co-
`agulation enzymes which may be important for the patho-
`genesis of various cardiovascular disorders.
`The trypsin-like serine protease FXa is an essential com-
`ponent of the prothrombinase complex which is assem-
`bled on phospholipid surfaces involving the calcium-de-
`pendent association of FXa and FVa. FX, which is a two-
`chain glycoprotein synthesised in the liver and secreted
`into the blood as a zymogen, is converted to the active
`protease by a complex of either tissue factor/FVIIa or
`FIXa/FVIIIa/phospholipid/calcium. The catalytic activity
`of free FXa is extremely low, but upon formation of the
`prothrombinase complex it is strongly enhanced so that
`prothrombin can be rapidly cleaved and sufficient
`amounts of thrombin can be generated at sites of vascular
`injury and thrombus growth [1]. Under physiological
`conditions, the activity of clotting enzymes is controlled
`by endogenous inhibitors such as antithrombin III (AT
`III) which effectively inactivates thrombin and FXa as
`well as other serine proteases by forming very stable
`complexes that block the active site of the enzymes. The
`reaction between AT III and its target enzymes is very
`strongly accelerated by interaction with glycosaminogly-
`cans, especially the binding of a sequence-specific he-
`parin pentasaccharide, which causes conformational
`changes in AT III [2, 3].
`Besides its important role in the plasmatic coagulation
`system with the catalytic conversion of prothrombin to
`
`thrombin, FXa induces cellular responses implicated in
`cardiovascular and inflammatory diseases. FXa has been
`shown to be a strong mitogen and it exerts mitogenic ac-
`tions via high-affinity binding sites on vascular smooth
`muscle cells. Although effector cell protease receptor-1
`(EPR-1) was identified and cloned as a cellular receptor
`for FXa [4, 5], whether EPR-1 is really involved in the
`cellular effects of FXa or whether alternative cellular re-
`ceptors for FXa exist such as members of the protease-ac-
`tivated receptor family remains a controversial issue
`[6–9]. The inhibition of FXa-mediated mitogenic effects
`by specific inhibitors found in experimental studies sug-
`gests that this class of drugs might be effective to reduce
`neointimal hyperplasia in vivo either by preventing the
`mitogenic effects of FXa and/or by inhibiting the genera-
`tion of thrombin which is also known to be a potent mi-
`togen [10, 11].
`Commonly used anticoagulant/antithrombotic drugs such
`as heparin have limited efficacy, e.g. because of the re-
`quirement for endogenous cofactors, their inactivation by
`platelet factor 4 or the induction of various side effects
`such as heparin-induced thrombocytopaenia. Oral antico-
`agulants such as warfarin have a slow onset of action,
`may cause a paradoxical increase in coagulation activity
`due to the early inhibition of protein C and S, and require
`individual treatment with continuous laboratory monitor-
`ing as well as dose adjustment. Alternative antithrom-
`botics would be highly desirable. An important aspect for
`the development of small-molecule inhibitors of FXa as
`well as thrombin is their ability to inactivate the clotting
`enzymes not only in plasma but also when they are bound
`to fibrin within a clot, an effect which is not seen with AT
`III and AT III-dependent inhibitors. Because FXa and ac-
`tive prothrombinase complexes are main determinants for
`the procoagulant activity of intravascular thrombi, effec-
`
`MYLAN - EXHIBIT 1048
`
`

`

`190
`
`B. Kaiser
`
`Factor Xa as drug target
`
`tive inactivation of the clot-bound enzyme is of particular
`importance [12, 13].
`The important role of FXa in the coagulation network at
`the stage of the common pathway of both the tissue fac-
`tor-activated extrinsic and the surface-activated intrinsic
`system as well as the amplification of the procoagulant
`action of FXa by prothrombinase complex formation
`leads to the expectation that inhibitors with a high affin-
`ity and selectivity towards the enzyme will be potentially
`valuable therapeutic agents for various cardiovascular in-
`dications. Compared to thrombin, FXa acts at an earlier
`level in the coagulation system and is not such a multi-
`functional protein, i.e. it exerts its action mainly on the
`substrate prothrombin. Inactivation of FXa by specific in-
`hibitors does not influence preformed thrombin but ef-
`fectively prevents the generation of thrombin whereas in
`the presence of thrombin inhibitor, considerable throm-
`bin formation was demonstrated. The increased thrombin
`activity observed after cessation of therapy with thrombin
`inhibitors might be responsible for the continuation of the
`thrombotic process and contribute to early rethrombosis.
`The apparently incomplete and only temporary suppres-
`sion of thrombin generation by these drugs is not ex-
`pected with FXa inhibitors because of their mechanism
`and site of action in the coagulation cascade [14, 15]. In
`addition, with the different mechanism of action, FXa in-
`hibitors might have a better efficacy/safety profile than
`specific thrombin inhibitors for which clinical trials
`showed a relatively narrow safety/efficacy margin which
`can result in bleeding complications at drug overdosage
`[16].
`
`Present state of development of FXa inhibitors
`
`Due to its critical role in the clotting cascade, FXa pre-
`sents one of the most popular coagulation enzymes for
`the design of new oral directly acting anticoagulants/an-
`tithrombotics. Based on the specific mechanism of action
`of FXa, an effective inhibitor must have an extremely
`high affinity for the enzyme. Numerous inhibitors of FXa
`have been described in the literature and investigated in
`mainly preclinical studies. Besides naturally occurring
`FXa inhibitors such as antistasin and tick anticoagulant
`peptide, a variety of peptide, peptidomimetic and non-
`peptide small-molecule FXa inhibitors with a high affin-
`ity and selectivity for the enzyme have been synthesised
`by several pharmaceutical companies and characterised
`biochemically and pharmacologically [for reviews see
`refs 17–21]. Structure-activity relationship studies have
`led to selective inhibitors of FXa that are active at sub-
`nanomolar concentrations [22, 23]. However, many of the
`small-molecule FXa inhibitors are highly basic moieties
`leading to poor pharmacokinetic properties and, espe-
`cially, limited oral bioavailability. More recent develop-
`
`ments have focused on compounds with less basic groups
`which may represent potent orally available FXa in-
`hibitors for prophylactic and/or therapeutic use in clinical
`states.
`Effective indirect inhibition of FXa can be achieved by
`heparin pentasaccharide which represents the minimum
`saccharide sequence in the heparin molecule required for
`an antithrombotic activity as well as by synthetic, struc-
`turally modified analogues [24–26]. The first and at pre-
`sent only available representative of this class of FXa
`inhibitors is the synthetic heparin pentasaccharide
`Org31540/SR90107A which provides potent anti-FXa
`activity through selective inhibition of the clotting en-
`zyme by high-affinity binding to AT III [25].
`
`Pharmacological profile
`
`In vitro experiments and the first clinical studies have
`shown that FXa inhibitors exert strong anticoagulant, an-
`tithrombotic and even antiproliferative actions [10, 11,
`14, 15, 27]. Due to the central position of FXa in the co-
`agulation cascade, the inactivation of this serine protease
`offers a more global control of clotting. FXa inhibitors
`strongly inhibit the generation of thrombin and, thus, also
`thrombin-mediated positive feedback reactions such as
`the activation of factors V and VIII, as well as throm-
`bin-mediated platelet reactions. Small-molecule FXa
`inhibitors can inactivate both free and especially clot-
`bound FXa. Because of the presence of FXa and active
`prothrombinase complexes in intravascular and mural
`thrombi [12, 13], the inactivation of FXa and the result-
`ing inhibition of thrombin formation may be an effective
`way to control clot-associated procoagulant activity. Re-
`sults from experimental studies indicate a role for FXa in
`the complex pathogenesis of restenosis and atherosclero-
`sis. Both thrombin and FXa most likely contribute to vas-
`cular smooth muscle cell proliferation in vivo, although
`the precise role of the serine proteases and especially the
`significance of their mitogenic activities for restenosis
`have still to be clarified.
`
`Clinical studies
`
`At present, there are no direct FXa inhibitors marketed as
`drugs, but several compounds are in early phase clinical tri-
`als. Published clinical data are available for DX-9065a, a
`small-molecule, direct FXa inhibitor, and for the synthetic
`pentasaccharide Org31540/SR90107A which provides po-
`tent antithrombotic activity through selective inhibition of
`FXa by high-affinity binding to AT III. Both drugs offer
`safe and predictable pharmacokinetic and pharmacody-
`namic profiles after subcutaneous (s.c.) and intravenous
`(i.v.) administration in healthy volunteers [28].
`
`

`

`CMLS, Cell. Mol. Life Sci. Vol. 59, 2002
`
`Visions & Reflections
`
`191
`
`The number of clinical studies published on the direct
`FXa inhibitor DX-9065a is rather limited. Studies on the
`pharmacokinetic and pharmacodynamic profile of DX-
`9065a after i.v. and s.c. administration in healthy volun-
`teers or in patients with stable coronary artery disease
`showed a strong correlation between doses and plasma
`concentrations of the drug [29–31]. Given intravenously
`to healthy male volunteers, DX-9065a reduced platelet
`thrombus formation ex vivo using a perfusion chamber
`model [32].
`For the synthetic pentasaccharide Org31540/SR90107A
`in phase II and III clinical trials, a significant efficacy and
`safety was demonstrated in orthopaedic patients under-
`going total hip replacement [33–35], elective major knee
`surgery [36] or hip fracture surgery [37]. Furthermore,
`there are promising preliminary results with this drug in
`cardiology trials in patients undergoing percutaneous
`transluminal coronary angioplasty or with unstable
`angina or acute myocardial infarction (AMI) [38]. In pa-
`tients with AMI treated with aspirin and alteplase, cother-
`apy with Org31540/SR90107A was as safe and effective
`as unfractionated heparin in restoring coronary artery pa-
`tency [39].
`
`Concluding remarks
`
`The FXa inhibitors which are presently under develop-
`ment are structurally diverse compounds with different
`biochemical and pharmacological characteristics. De-
`spite the anticoagulant and antithrombotic effectiveness
`of direct and indirect FXa inhibitors demonstrated in
`comprehensive experimental studies, a general assess-
`ment of the therapeutic potential of this new class of
`drugs has to consider various additional aspects. Pharma-
`cokinetic characteristics such as oral bioavailability, bio-
`logical half-life, metabolic transformations or excretory
`routes are important factors for the clinical use of a given
`drug. Interactions with other drugs or endogenous factors
`as well as additional mechanisms of action have to be
`taken into consideration. A particular FXa inhibitor
`might be useful for a specific clinical indication and one
`drug might not be the optimum treatment for all throm-
`botic situations. FXa inhibitors inhibit the generation of
`thrombin very effectively but are expected to be ineffec-
`tive in clinical conditions where thrombin has already
`been formed. The complex pathogenesis of thromboem-
`bolic processes which involves both plasmatic and cellu-
`lar reactions requires a careful analysis of the usefulness
`of a combination of FXa inhibitors with other drugs
`showing different mechanisms and sites of action in order
`to enhance the therapeutic effect. Simultaneous direct in-
`hibition of thrombin and FXa by synthetic proteinase in-
`hibitors might represent a novel approach to develop an-
`tithrombotics with improved pharmacological properties
`
`[40]. Although the efficacy/safety profile of FXa in-
`hibitors may be better than that for heparin or antithrom-
`bin agents, these agents may also cause undesired side ef-
`fects such as bleeding complications. Furthermore, com-
`bination of FXa inhibitors with other drugs such as
`thrombin inhibitors or platelet function inhibitors might
`not only show synergistic therapeutic effects, but could
`also enhance side effects. Other important points to con-
`sider and problems to be solved with the use of FXa in-
`hibitors are whether and how to monitor the effect of a
`given FXa inhibitor in clinical practice, i.e. is there an
`easy and reproducible assay, and how can the effect of the
`drug be neutralised in case of overdose or occurrence of
`undesired side effects?
`In conclusion, FXa inhibitors represent promising drugs
`for the prophylaxis and/or therapy of various throm-
`boembolic disorders. However, additional experimental
`studies and comprehensive clinical trials are required to
`demonstrate the inhibitory profile of FXa inhibitors, their
`effectiveness and especially their superiority over other
`commonly used drug regimens for cardiovascular indica-
`tions.
`
`1 Mann K. G. and Kalafatis M. (1995) The coagulation explo-
`sion. Cerebrovasc. Dis. 5: 93–97
`2 Olson S. T., Bjork I., Sheffer R., Craig P. A., Shore J. D. and
`Cjoay J. (1992) Role of the antithrombin-binding pentasaccha-
`ride in heparin acceleration of antithrombin-proteinase reac-
`tions: resolution of the antithrombin conformational change
`contribution to heparin rate enhancement. J. Biol. Chem. 267:
`12528–12538
`3 Desai U. R., Petitou M., Björk I. and Olson S. T. (1998) Mech-
`anism of heparin activation of antithrombin: evidence for an in-
`duced-fit model of allosteric activation involving two interac-
`tion subsites. Biochemistry 37: 13033–13041
`4 Altieri D. C. and Edgington T. S. (1990) Identification of effec-
`tor cell protease receptor-1: a leukocyte-distributed receptor for
`the serine protease factor Xa. J. Immunol. 145: 246–253
`5 Altieri D. C. (1994) Molecular cloning of effector cell protease
`receptor-1, a novel cell surface receptor for the protease factor
`Xa. J. Biol. Chem. 269: 3139–3142
`6 Blanc-Brude O. P., Chambers R. C., Leoni P., Dik W. A. and
`Laurent G. J. (2001) Factor Xa is a fibroblast mitogen via bind-
`ing to effector-cell protease receptor-1 and autocrine release of
`PDGF. Am. J. Physiol. Cell. Physiol. 281: C681–C689
`7 Zaman G. J. R. and Conway E. M. (2000) The elusive factor Xa
`receptor: failure to detect transcripts that correspond to the pub-
`lished sequence of EPR-1. Blood 96: 145–148
`8 Riewald M., Kravchenko V. V., Petrovan R. J., O’Brien P. J.,
`Brass L. F., Ulevitch R. J. et al. (2001) Gene induction by co-
`agulation factor Xa is mediated by activation of protease-acti-
`vated receptor 1. Blood 97: 3109–3116
`9 Kawabata A., Kuroda R., Nakaya Y., Kawai K., Nishikawa H.
`and Kawao N. (2001) Factor Xa-evoked relaxation in rat aorta:
`involvement of PAR-1. Biochem. Biophys. Res. Commun. 282:
`432–435
`10 Herbert J. M., Bono F., Hérault J. P., Avril C., Dol F., Mares A.
`M. et al. (1998) Effector protease receptor 1 mediates the mito-
`genic activity of factor Xa for vascular smooth muscle cells in
`vitro and in vivo. J. Clin. Invest. 101: 993–1000
`11 Kaiser B., Paintz M., Scholz O., Kunitada S. and Fareed J.
`(2000) A synthetic inhibitor of factor Xa, DX-9065a, reduces
`
`

`

`192
`
`B. Kaiser
`
`Factor Xa as drug target
`
`proliferation of vascular smooth muscle cells in vivo in rats.
`Thromb. Res. 98: 175–185
`12 Prager N. A., Abendschein D. R., McKenzie C. R. and Eisen-
`berg P. R. (1995) Role of thrombin compared with factor Xa in
`the procoagulant activity of whole blood clots. Circulation 92:
`962–967
`13 Ghigliotti G., Waissbluth A. R., Speidel C., Abendschein D. R.
`and Eisenberg P. R. (1998) Prolonged activation of prothrom-
`bin on the vascular wall after arterial injury. Arterioscler.
`Thromb. Vasc. Biol. 18: 250–57
`14 Kaiser B. (1998) Thrombin and factor Xa inhibitors. Drugs Fu-
`ture 23: 423–436
`15 Kaiser B. and Hauptmann J. (1994) Factor Xa inhibitors as
`novel antithrombotic agents: facts and perspectives. Cardio-
`vasc. Drug Rev. 12: 225–236
`16 Kaiser B., Callas D., Walenga J. M. and Fareed J. (1998) Syn-
`thetic and recombinant antithrombin drugs. Exp. Opin. Invest.
`Drugs 7: 963–985
`17 Scarborough R. M. (1998) Coagulation factor Xa: the pro-
`thrombinase complex as an emerging therapeutic target for
`small molecule inhibitors. J. Enzyme Inhibit. 14: 15–25
`18 Ewing W. R., Pauls H. W. and Spada A. P. (1999) Progress in the
`design of inhibitors of coagulation factor Xa. Drugs Future 24:
`771–787
`19 Al-Obeidi F. and Ostrem J. A. (1999) Factor Xa inhibitors. Ex-
`pert. Opin. Ther. Patents 9: 931–953
`20 Zhu B. Y. and Scarborough R. M. (2000) Factor Xa inhibitors:
`recent advances in anticoagulant agents. Annu. Rep. Med.
`Chem. 35: 83–102
`21 Vacca J. P. (2000) New advances in the discovery of thrombin
`and factor Xa inhibitors. Curr. Opin. Chem. Biol. 4: 394–400
`22 Rai R., Sprengeler P. A., Elrod K. C. and Young W. B. (2001)
`Perspectives on factor Xa inhibition. Curr. Med. Chem. 8:
`101–119
`23 Betz A. (2001) Recent advances in factor Xa inhibitors. Expert
`Opin. Ther. Pat. 11: 1007–1017
`24 Choay J., Petitou M., Lormeau J. C., Sinay P., Casu B. and Gatti
`G. (1983) Structure-activity relationship in heparin: a synthetic
`pentasaccharide with high affinity for antithrombin III and elic-
`iting high anti-factor Xa activity. Biochem. Biophys. Res.
`Commun. 116: 492–499
`25 Van Boeckel C. A. A. and Petitou M. (1993) The unique an-
`tithrombin III binding domain of heparin: a lead to new syn-
`thetic antithrombotics. Angew. Chem. Int. Ed. Engl. 32:
`1671–1690
`26 Petitou M., Duchaussoy P., Jaurand G., Gourvenec F., Leder-
`man I., Strassel J. M. et al. (1997) Synthesis and pharmacolog-
`ical properties of a close analogue of an antithrombotic pen-
`tasaccharide (SR 90107A/ORG 31540). J. Med. Chem. 40:
`1600–1607
`27 Walenga J. M., Jeske W. P., Hoppensteadt D. and Kaiser B.
`(1999) Factor Xa inhibitors: today and beyond. Curr. Opin. Car-
`diovasc. Pulm. Renal Invest. Drugs 1: 13–27
`28 Porcari A. R., Chi L. and Leadley R. (2000) Recent advances in
`clinical trials of the direct and indirect selective factor Xa in-
`hibitors. Expert Opin. Invest. Drugs 9: 1595–1600
`29 Murayama N., Tanaka M., Kunitada S., Yamada H., Inoue T.,
`Terada Y. et al. (1999) Tolerability, pharmacokinetics, and phar-
`
`macodynamics of DX-9065a, a new synthetic potent anticoag-
`ulant and specific factor Xa inhibitor, in healthy male volun-
`teers. Clin. Pharmacol. Ther. 66: 258–264
`30 Depasse F., Caplain H., Kunitada S. and Samama M. M. (2001)
`Assessment of the pharmacodynamic profile of DX9065a, a di-
`rect factor Xa inhibitor, in 36 healthy male subjects: a first
`study using subcutaneous route (abstract). Thromb. Haemost.
`(Suppl.) P3098
`31 Dyke C. K., Becker R. C., Hasselblad V., Bovill E. G., Kunitada
`S., Robertson T. L. et al. (2001) Pharmacokinetic and pharma-
`codynamic responses of the novel factor Xa inhibitor DX-
`9065a: results from the Xa neutralization for atherosclerotic
`disease understanding (XaNADU) IB trial (abstract). Eur.
`Heart J. 22 (Suppl.): 664
`32 Shimbo D., Osende J., Chen J., Mukherjee J., Glenn J., Robbins
`J. et al. (2001) Antithrombotic effects of DX-9065a, a direct
`factor Xa inhibitor: a comparative study in humans vs low mo-
`lecular weight heparin (abstract). Thromb. Haemost. (Suppl.):
`P1392
`33 Turpie A. G. G. (2001) Pentasaccharide Org31540/SR90107A
`clinical trials update: lessons for practice. Am. Heart J. 142
`(Suppl. 2): S9–S15
`34 Lassen M. R. (2001) Efficacy of the first synthetic factor Xa in-
`hibitor, pentasaccharide Org31540/SR90107A, versus low
`molecular weight heparin (LMWH) in the prevention of venous
`thromboembolism (VTE) following elective hip replacement
`surgery: the Ephesus study (abstract). Thromb. Haemost.
`(Suppl.): OC45
`35 Turpie A. G. G. (2001) Efficacy of the first synthetic factor Xa
`inhibitor, pentasaccharide Org31540/SR90107A, versus low
`molecular weight heparin (LMWH) in the prevention of venous
`thromboembolism (VTE) following elective hip replacement
`surgery: the Pentathlon 2000 study (abstract). Thromb.
`Haemost. (Suppl.): OC48
`36 Bauer K. (2001) Efficacy of the first synthetic factor Xa in-
`hibitor, pentasaccharide Org31540/SR90107A, versus low
`molecular weight heparin (LMWH) in the prevention of venous
`thromboembolism (VTE) following elective major knee
`surgery: the Pentamaks study (abstract). Thromb. Haemost.
`(Suppl.): OC46
`37 Eriksson B. I. (2001) Efficacy of the first synthetic factor Xa
`inhibitor, pentasaccharide Org31540/SR90107A, versus low
`molecular weight heparin (LMWH) in the prevention of venous
`thromboembolism (VTE) following hip fracture surgery: the
`Penthifra study (abstract). Thromb. Haemost. (Suppl.): OC47
`38 Turpie A. G. G. (2001) The role in cardiology of the first syn-
`thetic factor Xa inhibitor: lessons from the results of the clini-
`cal programme
`in venous
`thromboembolism (abstract).
`Thromb. Haemost. (Suppl.): SY112
`39 Coussement P. K., Bassand J. P., Convens C., Vrolix M., Boland
`J., Grollier G. et al. (2001) A synthetic factor-Xa inhibitor
`(ORG31540/SR90107A) as an adjunct to fibrinolysis in acute
`myocardial infarction: the PENTALYSE study. Eur. Heart J. 22:
`1716–1724
`40 Nar H., Bauer M., Schmid A., Stassen J. M., Wienen W.,
`Priepke H. W. M. et al. (2001) Structural basis for inhibition
`promiscuity of dual specific thrombin and factor Xa blood co-
`agulation inhibitors. Structure 9: 29–37
`
`

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


Or .

Accessing this document will incur an additional charge of $.

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

Accept $ Charge
throbber

Still Working On It

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

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

throbber

A few More Minutes ... Still Working

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

Thank you for your continued patience.

This document could not be displayed.

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

Your account does not support viewing this document.

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

Your account does not support viewing this document.

Set your membership status to view this document.

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

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

Become a Member

One Moment Please

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

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

Your document is on its way!

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

Sealed Document

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

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


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket