throbber
Proc. Nati. Acad. Sci. USA
`Vol. 91, pp. 4082-4085, April 1994
`Medical Sciences
`
`Thalidomide is an inhibitor of angiogenesis
`(fibroblast growth factor/rabbit cornea)
`
`ROBERT J. D'AMATO*, MICHAEL S. LOUGHNAN, EVELYN FLYNN, AND JUDAH FOLKMAN
`Department of Surgery, Children's Hospital, Harvard Medical School, Boston, MA 02115
`Communicated by John A. Glomset, January 3, 1994
`
`Thalidomide is a potent teratogen causing
`ABSTRACT
`dysmelia (stunted limb growth) in humans. We have demon-
`strated that orally administered thalidomide is an inhibitor of
`aniogenesis induced by basic fibroblast growth factor in a
`rabbit cornea micropocket assay. Experiments including the
`analysis of thalidomide analogs revealed that the antanio-
`genic activity correlated with the teratogenicity but not with the
`sedative or the mild immunosuppressive properties of tha-
`lidomide. Electron microscopic examination of the corneal
`neovascularization ofthafidomide-treated rabbits revealed spe-
`cific ultrastructural changes similar to those seen in the de-
`formed limb bud vasculature of thalidomide-treated embryos.
`These experiments shed light on the mechanism of tha-
`lidomide's teratogenicity and hold promise for the potential use
`of thalidomide as an orally adminsered drug for the treatment
`of many diverse dease dependent on angiogenesis.
`
`Thalidomide is a potent teratogen. It was developed by
`Chemie Grunenthal in the 1950s as a sedative that appeared
`so nontoxic in rodent models that a LD50 could not be
`established. In 1961, McBride (1) and Lenz (2) described the
`association between limb defects in babies and maternal
`thalidomide usage. Although humans are exquisitely sensi-
`tive to the teratogenic effects of thalidomide, experiments in
`rodents failed to reveal similar effects (3, 4). Teratogenic
`effects could be experimentally reproduced by the adminis-
`tration of thalidomide to pregnant rabbits at an oral dose of
`100-300 mg per kg per day (5, 6). Over the past 30 years, the
`mechanism of thalidomide's teratogenicity has been exten-
`sively studied but has remained unsolved (7).
`We now postulate that the limb defects seen with thali-
`domide were secondary to an inhibition of blood vessel
`growth in the developing fetal limb bud. The limb bud is
`unique in requiring a complex interaction of both angiogen-
`esis and vasculogenesis during development (8). Vasculo-
`genesis is the formation of a capillary bed from endothelial
`cells that have differentiated from mesenchymal precursors.
`Angiogenesis is the formation of new blood vessels from
`sprouts ofpreexisting vessels. Therefore, the limb bud would
`be a particularly vulnerable target to a teratogen that inhib-
`ited endothelial cell function. We chose to examine the effect
`of thalidomide on growing vasculature in the chicken chori-
`oallantoic membrane and in the rabbit cornea.
`
`MATERIALS AND METHODS
`Chicken chorioallantoic membrane (CAM) assays were per-
`formed as described (9, 10) and the effects on the developing
`vasculature were recorded at 48 h after implantation of the
`0.5% carboxymethylcellulose pellet containing various
`drugs. Corneal neovascularization was induced by an im-
`planted pellet of poly(hydroxyethyl methacrylate) (Hydron;
`Interferon Sciences, New Brunswick, NJ) containing 650 ng
`
`The publication costs of this article were defrayed in part by page charge
`payment. This article must therefore be hereby marked "advertisement"
`in accordance with 18 U.S.C. §1734 solely to indicate this fact.
`
`ofthe potent angiogenic protein basic fibroblast growth factor
`(bFGF) bound to sucralfate (sucrose aluminum sulfate; Bukh
`Meditec, Copenhagen) (11). The addition of sucralfate to the
`pellet protects the bFGF from degradation (12) and provides
`for its slow release, thus producing consistent aggressive
`angiogenesis that is more pronounced than that induced by
`bFGF/Hydron alone. Release of bFGF from pellets contain-
`ing sucralfate/Hydron could be detected in vitro for up to 4
`days after the pellets were formed compared tojust 1 day for
`pellets with Hydron alone (11). Pellets were made by mixing
`110 pI of saline containing 12 Mg of recombinant bFGF
`(Takeda, Osaka) with 40 mg of sucralfate; this suspension
`was added to 80 pI of 12% (wt/vol) Hydron in ethanol.
`Aliquots (10 Al) of this mixture were then pipetted onto
`Teflon pegs and allowed to dry producing approximately 17
`pellets. A pellet was implanted into corneal micropockets of
`each eye of an anesthetized female New Zealand White
`rabbit, 2 mm from the limbus, followed by a single topical
`application of erythromycin ointment on the surface of the
`cornea. Histologic examination on consecutive days demon-
`strated progressive blood vessel growth into the cornea
`toward the pellet with only rare inflammatory cells seen. This
`angiogenic response was not altered by severe immune
`suppression with total body irradiation, and pellets with
`sucralfate alone did not induce angiogenesis (data not
`shown). Unlike other models of corneal angiogenesis that
`utilize inflammation to stimulate neovascularization, the new
`vessels are primarily induced by the bFGF. The animals were
`fed daily from 2 days after implantation by gastric lavage with
`either drug suspended in 0.5% carboxymethylcellulose or
`vehicle alone. Thalidomide was purchased from Andrulus
`Pharmaceutical (Beltsville, MD) and EM-12 and Supidimide
`were kindly provided by Grunenthal (Stolberg, F.R.G.).
`Immunosuppressed animals received total body radiation of
`6 Gy for 6 min immediately prior to implantation of the
`pellets. This dose of radiation resulted in a marked leukocy-
`topenia with >80%o reduction in the leukocyte count by day
`2 and >90o reduction by day 3, results that are consistent
`with previous reports (13, 14).
`The animals were examined with a slit lamp every other
`day in a masked manner by the same corneal specialist
`(M.S.L.). The area of corneal neovascularization was deter-
`mined by measuring with a reticule the vessel length (L) from
`the limbus and the number of clock hours (C) of limbus
`involved. A formula was used to determine the area of a
`circular band segment: C/12 x 3.1416 [r2 - (r - L)2], where
`r = 6 mm, the measured radius of the rabbit cornea. We have
`utilized various mathematical models to determine the
`amount of vascularized cornea and have found that this
`formula provides the most accurate approximation ofthe area
`of the band of neovascularization that grows toward the
`pellet. Only the uniform contiguous band of neovasculariza-
`
`Abbreviations: bFGF, basic fibroblast growth factor; CAM, chicken
`chorioallantoic membrane; PGA, phthaloylglutamic anhydride; PG
`acid, phthaloylglutamic acid; TNF-a, tumor necrosis factor a.
`*To whom reprint requests should be addressed.
`
`4082
`
`DR. REDDY’S LABS., INC. EX. 1030 PAGE 1
`
`

`

`Medical Sciences: D'Amato et al.
`
`Proc. Natl. Acad. Sci. USA 91 (1994)
`
`4083
`
`genic dose (200 mg/kg) of thalidomide resulted in an inhibi-
`tion of the area of vascularized cornea that ranged from 30 to
`51% in three experiments with a median inhibition of 36%
`(Figs. 2A and 3) (n = 30 eyes; P = 0.0001, two-way ANOVA
`with ranked data). The inhibition of angiogenesis by tha-
`lidomide was seen after only two doses (Fig. 2B). The rabbits
`did not demonstrate obvious sedation and there were no signs
`of toxicity or weight loss. The teratogenic analog EM-12,
`which shares the other properties of thalidomide, was also
`inhibitory, with a median inhibition of 42% (n = 10 eyes; P
`= 0.002, one-way ANOVA with ranked data). Supidimide, a
`nonteratogenic analog that retains the sedative properties of
`thalidomide, exhibited no activity (area 107% of control; n =
`10 eyes; not statistically different from control). Other ana-
`logs, PGA and PG acid, displayed weaker inhibitory effects
`
`M Supidimide
`o Thalidomide
`El EM12
`0 Thalidomide-irradiated
`
`==4"~
`
`Animal treatment groups
`
`2
`
`4
`8
`6
`Days post-implantation
`
`10
`
`12
`
`A
`
`o 100-
`
`80
`
`60-
`
`40
`
`20-
`
`O-
`
`N ,
`
`vQC.)CZ)
`-F
`a)
`
`0
`
`0
`
`B
`
`40 -
`
`N4
`Pi
`
`E c.
`
`° 30-
`
`N C
`
`o
`
`0a 20-
`co
`-a
`a)
`
`0
`
`0 .
`
`: 0'-
`0
`
`(A) Inhibition of bFGF-induced corneal neovasculariza-
`FIG. 2.
`tion by thalidomide and related analogs expressed as percent of
`median control on day 8. Pellets containing bFGF and sucralfate
`were implanted into micropockets of both corneas of rabbits (18).
`Vessel ingrowth into the clear cornea from the limbus was first noted
`on day 2 and treatments (200 mg/kg orally) were begun on this day.
`The area of corneal neovascularization was measured from day 4
`through day 12. Day 8 measurements were used for comparison
`between groups. No regression of vessels and near maximal neovas-
`cularization was seen at this time point. Statistical analysis was
`performed with ANOVA with ranked data to account for interex-
`perimental variation and to guard against a nonnormal distribution of
`data (i.e., outliers) by utilizing a nonparametric method. (b) Time
`course of inhibition of neovascularization with thalidomide. Mean
`areas of corneal neovascularization with standard error bars are
`presented from one experiment that is representative of the three
`experiments performed with thalidomide on nonirradiated animals.
`Data presented from the first time point after administration of the
`drug through the completion of the study are statistically different (n
`= 10 eyes; P < 0.005 for all time points, one-way ANOVA with
`ranked data).
`
`tion adjacent to the pellet was measured. The noncontiguous
`neovascularization, which can be seen superiorly, was not
`quantified due to its irregular shape. These vessels that often
`grow concurrently toward the pellet from the superior limbus
`arise from vessels ofthe superior rectus supplying the limbus,
`are directly induced by the bFGF/sucralfate pellet, and are
`histologically identical to the inferior limbal vessels. How-
`ever, it should be noted that this superior neovascularization
`was commonly seen in control animals and was never seen in
`thalidomide-treated animals. Thus, the total inhibition of
`neovascularization is conservatively underestimated.
`
`RESULTS
`Our initial investigations were performed on the CAM.
`Neither thalidomide nor EM-12, a related teratogenic analog
`(15), exhibited any inhibitory activity on blood vessel growth.
`This result was expected as it has been proposed that
`thalidomide must be metabolized by the liver to form an
`epoxide that may be the active teratogenic metabolite (16).
`Other thalidomide analogs that have been shown to be
`teratogenic in rodents (17), including phthaloylglutamic an-
`hydride (PGA) and phthaloylglutamic acid (PG acid), were
`also analyzed (Fig. 1). Interestingly, weak antiangiogenic
`activity of the developing vasculature was seen with both PG
`acid and PGA when 100 jig of either compound was placed
`on the CAM in a pellet of 0.5% carboxymethylcellulose.
`Despite frequent mild scarring, avascular zones were pro-
`duced in 15% of the CAMs with PGA compared to control
`0.5% carboxymethylcellulose pellets in which no avascular
`zones were seen (data not shown).
`Based on these initial findings, we decided to test tha-
`lidomide's effect on angiogenesis induced by bFGF in the
`rabbit corneal micropocket model. Treatment with a terato-
`
`Thalidomide
`
`0
`
`N
`
`a
`
`N
`
`01
`
`1 C
`
`11
`
`0 0
`
`c
`
`c
`
`0o
`
`N
`
`'o
`
`EM-12
`
`Phthaloyl Glutamic
`Anhydride (PGA)
`
`,c
`,>
`ll, 0 / b
`0
`
`Phthaloyl
`Glutamic Acid
`(PG Acid)
`
`0 C
`
`-OH
`
`e"
`
`OH
`
`01
`
`1 C c
`
`11
`0
`
`01
`
`1
`
`11
`0
`
`01
`
`1
`
`SJCN
`
`02
`
`N
`
`Supidimide
`
`FIG. 1.
`
`Structure of thalidomide and related analogs.
`
`DR. REDDY’S LABS., INC. EX. 1030 PAGE 2
`
`

`

`4084
`
`Medical Sciences: D'Amato et al.
`
`Proc. Natl. Acad. Sci. USA 91 (1994)
`
`than thalidomide (data not shown). The density of vessel
`ingrowth in thalidomide-treated animals was also markedly
`reduced. Due to the lack of an objective grading scale, these
`results are not presented.
`Thalidomide has immunosuppressive properties that might
`have indirectly affected angiogenesis. Recently, thalidomide
`has been used for its immunosuppressive properties in the
`treatment of leproma reactions (19) and chronic graft versus
`host disease (18, 20-23). However, in humans its immuno-
`suppressive properties are weak and delayed with little effect
`in acute graft versus host disease (24). Because the effect of
`thalidomide on the immune system is similar but weaker than
`that of cyclosporin A (25), we tested cyclosporin A at the
`highest tolerated dose of 25 mg/kg. No statistically signifi-
`cant effect was observed compared to control. To investigate
`
`.I]
`
`further the immune interactions, we pretreated the rabbits
`with the maximally tolerated immunosuppressive dose of
`total body irradiation. Immunosuppressed animals re-
`sponded equally well to thalidomide, with a median inhibition
`of neovascularization of 52% (n = 12; P = 0.0001, one-way
`ANOVA with ranked data) as compared to irradiated place-
`bo-treated controls (Fig. 2A).
`Electron microscopic examination of corneas from tha-
`lidomide-treated and control animals revealed ultrastructural
`differences. Vessels in the thalidomide-treated group dem-
`
`ti½*
`
`Ai
`
`A4-
`
`A
`
`B
`
`*L
`
`'o
`'O"
`I.- 1 '4
`
`.:!
`
`f..... ;7 .1
`
`:fsi,
`-..;z ebi,r,4
`
`"",::,
`
`..
`
`Representative corneas at 8 days after implantation of
`FIG. 3.
`bFGF pellets from control (A) and thalidomide-treated (B) rabbits.
`There is prominent corneal neovascularization (arrows) in the con-
`trol with associated corneal clouding, which was demonstrated
`histologically to be stromal edema without inflammation. The tha-
`lidomide-treated animal has markedly less neovascularization with
`minimal corneal edema.
`
`Electron micrographs of corneal neovascularization ob-
`FIG. 4.
`served in a thalidomide-treated rabbit 10 days after implntation of
`a pellet containing bFGF. (A) High-magnification (x40,000) view of
`typical fenestrations (arrow) in an endothelial cell from corneal
`neovascularization in thalidomide-treated rabbit. (B) High-
`magnification (x60,000) view of an area of cell thinning (asterisk)
`adjacent to a cell junction in thalidomide-treated corneal neovascu-
`larization. These changes were not seen in control day 10 corneal
`neovascularization. (Bars = 0.1 amn.)
`
`DR. REDDY’S LABS., INC. EX. 1030 PAGE 3
`
`

`

`Medical Sciences: D'Amato et al.
`
`Proc. Natl. Acad. Sci. USA 91 (1994)
`
`4085
`
`onstrated fenestrations not seen in control animals (Fig. 4A).
`Fenestrations have been previously reported to be specific to
`regressing corneal blood vessels after removal of the angio-
`genic stimulus (26). However, in that model, endothelial cell
`regression was associated with platelet plugging and cellular
`hypoxic changes such as swollen mitochondria, which were
`not seen in the thalidomide-treated animals. Interestingly,
`histologic changes previously described in the vasculature of
`the limb buds from chicken embryos treated with thalidomide
`(27) were also seen in the corneal neovascularization of our
`thalidomide-treated rabbits including vesicular projections
`into the lumen and extreme thinning of cell processes (Fig.
`4B). In general, the corneal neovascularization from thali-
`domide-treated rabbits appeared more immature than that
`observed in control animals with poorly formed cell junc-
`tions, incomplete basement membrane, and fewer associated
`pericytes. These findings support the hypothesis that tha-
`lidomide has a direct effect on the growing vasculature.
`
`DISCUSSION
`Orally administered thalidomide is an inhibitor of angiogen-
`esis induced by bFGF in the rabbit cornea micropocket
`assay. The mechanism by which thalidomide inhibits angio-
`genesis is unknown. Thalidomide has shown no effect on
`bFGF-induced proliferation of endothelial cells in culture
`(data not shown). Current studies are focused on the identi-
`fication of the most active thalidomide metabolite. The
`formation of an active metabolite by the liver in vivo provides
`an explanation of the observation that the effect of tha-
`lidomide on growing vessels is seen only when given sys-
`temically.
`Thalidomide has been shown to suppress tumor necrosis
`factor a (TNF-a) production from macrophages (28). How-
`ever, macrophages were rarely seen in histologic examina-
`tions of our model of corneal neovascularization. Further-
`more, studies examining the role of TNF-a in corneal angio-
`genesis have failed to detect TNF-a production in a model of
`inflammatory corneal angiogenesis in which macrophages
`were prominent (29). TNF-a is only weakly angiogenic in
`vivo. It acts by inducing secondary inflammation in contrast
`to bFGF, which stimulates angiogenesis without inflamma-
`tion (30). Thus, the ability of thalidomide to inhibit angio-
`genesis induced by pharmacologic doses of bFGF supports
`the hypothesis that thalidomide directly inhibits an essential
`component of angiogenesis and does not operate through
`effects on TNF-a production.
`In conclusion, thalidomide is a potent angiogenesis inhib-
`itor in vivo. In this model of corneal angiogenesis, we have
`tested many putative angiogenesis inhibitors (including an-
`timitotic agents, cis-retinoic acid, tamoxifen, and others).
`Thalidomide was the only agent capable of inhibiting angio-
`genesis after oral administration. Evaluation of thalidomide
`analogs demonstrated a correlation between teratogenicity
`and antiangiogenic potential. The weak and delayed immu-
`nosuppressive action of thalidomide when used clinically, its
`inhibition of angiogenesis in radiation-immunosuppressed
`animals, and the lack of effect of the functionally related
`immunosuppressive agent cyclosporin A argue for a direct
`effect of thalidomide on angiogenesis. Further support for
`this hypothesis is derived from the ultrastructural changes
`seen in thalidomide-treated animals. There are clear impli-
`cations for the use of this drug in the treatment of pathologic
`angiogenesis that occurs in diabetic retinopathy, macular
`
`degeneration, and solid tumors. Because antiangiogenic ther-
`apy is likely to be long-term, there is a need for an orally
`efficacious inhibitor.
`
`Special thanks to Klio Chatzistefanou, Geri Jackson, Evelyn
`Gonzalez, Pat D'Amore, Helene Sage, Michael O'Reilly, Michael
`Kaplan, Tony Adamis, Elizabeth N. Allred, Ramzi Cotran, and
`Dianna Ausprunk for their assistance and advice. We also thank E.
`Frankus and K. Zwingenberger of Grunenthal GMBH for providing
`technical information, EM-12, and supidimide. R.J.D. is a Howard
`Hughes Medical Institute physician research fellow. M.S.L. is partly
`supported by the Ruth Rae Davidson corneal fellowship endowment.
`Animal studies were reviewed and approved by the animal care and
`use committee of Children's Hospital and are in accordance with the
`guidelines of the Department of Health and Human Services.
`
`1.
`2.
`3.
`
`4.
`5.
`6.
`
`7.
`8.
`
`9.
`
`McBride, W. G. (1961) Lancet u1, 1358.
`Lenz, W. (1962) Lancet 1, 45.
`Cahen, R., Sautai, M., Montagne, J. & Pessonier, J. (1964)
`Med. Exp. 10, 201-224.
`Christie, G. A. (1962) Lancet ii, 249.
`Helm, F. (1966) Arzneim. Forsch. 16, 1232-1237.
`Stertz, H., Nothduft, H., Lexa, P. & Ockenfels, H. (1987)
`Arch. Toxicol. 60, 376-381.
`Stepens, T. D. (1988) Teratology 38, 229-239.
`Seifert, R., Zhao, B., Christ, B. (1992) Anat. Embryol. 186,
`601-610.
`Auerbach, R., Kubai, L., Knighton, D. & Folkman, J. (1974)
`Dev. Biol. 41, 391-394.
`Crum, R., Szabo, S. & Folkman, J. (1985) Science 230, 1375-
`1378.
`Gonzalez, E., Adamis, T. & Folkman, J. (1994) Invest. Oph-
`thalmol. Vis. Sci. 33 Suppl., 424.
`Folkman, J., Szabo, S., Stovroff, M., McNeil, P., Li, W. &
`Shing, Y. (1991) Ann. Surg. 214, 414-427.
`13. Bogman, M., Cornelissen, I., Berden, J., De Jong, J. & Koene,
`R. (1984) J. Immunol. Methods 70, 31-38.
`Bigelow, C., Adler, L., Appelbaum, F. (1987) Transplantation
`44, 351-354.
`Helm, F., Frankus, E., Friderichs, E., Graudums, I. & Flohe,
`L. (1981) Arzneim. Forsch. Drug Res. 31, 941-949.
`Blake, D. A. & Balasubramanian, V. (1981) Proc. Natl. Acad.
`Sci. USA 78, 2545-2548.
`Kocher-Becker, U., Kocher, W. & Ockenfels, H. (1984) Z.
`Naturforsch. 36, 904-909.
`Vogelsang, G. B., Taylor, S., Gordon, G. & Hess, A. D. (1986)
`Transp. Proc. 18, 904-906.
`Sheskin, J. (1965) Clin. Pharm. Ther. 6, 303-306.
`Field, E. O., Gibbs, J. E., Tucker, D. F. & Hellman, K. (1966)
`Nature (London) 211, 1308-1313.
`Lim, S. H., McWannell, A., Vora, A. J. & Boughton, B. J.
`(1988) Lancet 1, 117.
`McCarthy, D. M., Kanfer, E. J. & Barrett, A. J. (1987)
`Biomed. Pharmacother. 43, 693-697.
`Saurat, J. H., Camenzind, M., Helg, C. & Chapuis, B. (1988)
`Lancet 1, 359.
`McCarthy, D. M., Kanfer, E. J. & Barrett, A. J. (1989)
`Biomed. Pharmacother. 43, 693-697.
`Keenan, R., Eiras, G., Burckart, G., Stuart, R., Hardesty, R.,
`Vogelsang, G., Griffith, B. & Zeevi, A. (1991) Transplantation
`52, 908-910.
`Ausprunk, D., Falterman, K. & Folkman, J. (1978) Lab. Invest.
`38, 284-294.
`Jurand, A. (1966) J. Embryol. Exp. Morphol. 16, 289-300.
`Sampaio, E., Sarno, E., Galilly, R., Cohn, Z. & Kaplan, G.
`(1991) J. Exp. Med. 173, 699-703.
`Sunderkotter, C., Roth, J. & Sorg, C. (1990)Am. J. Pathol. 137,
`511-515.
`Frater-Schroder, M., Risau, W., Hallman, R., Gautschi, P. &
`Bohlen, P. (1987) Proc. Natl. Acad. Sci USA 84, 5277-5281.
`
`10.
`
`11.
`
`12.
`
`14:
`
`15.
`
`16.
`
`17.
`
`18.
`
`19.
`20.
`
`21.
`
`22.
`
`23.
`
`24.
`
`25.
`
`26.
`
`27.
`28.
`
`29.
`
`30.
`
`DR. REDDY’S LABS., INC. EX. 1030 PAGE 4
`
`

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