Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
HSV DRUGS
Document Type and Number:
WIPO Patent Application WO/2006/133198
Kind Code:
A2
Abstract:
Pharmaceutical compositions comprising glucocorticoid receptor II antagonists, their enantiomers and isomers, antisense, inhibitory RNA, and peptide mimetic compounds targeting EIF3/mov34 and/or GR II useful to treat Herpes Simplex Virus (Types 1 and 2) infections are disclosed. Methods of treating individuals infected with Herpes Simplex Virus (Types 1 and 2) are disclosed.

Inventors:
KIM JONG JOSEPH (US)
Application Number:
PCT/US2006/021931
Publication Date:
December 14, 2006
Filing Date:
June 06, 2006
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
VGX PHARMACEUTICALS INC (US)
KIM JONG JOSEPH (US)
International Classes:
A61K31/57
Domestic Patent References:
WO2004112724A1
Attorney, Agent or Firm:
DELUCA, Mark (1900 Market Street Philadelphia, PA, US)
Download PDF:
Claims:

CLAIMS

1. A pharmaceutical composition comprising: a pharmaceutically acceptable carrier or diluent; and a compound having a structure selected from glucocorticoid receptor II antagonists or their enantiomers and isomers at dosage levels effective in treating those infected with Herpes Simplex Virus (Types 1 and 2).

2. A pharmaceutical composition comprising: a pharmaceutically acceptable carrier or diluent; and a compound having a structure selected from the group consisting of mifepristone, Formula D1-D21, or their enantiomers and isomers, and pharmaceutically acceptable salts thereof at dosage levels effective in treating those infected with Herpes Simplex Virus (Types 1 and 2).

3. A pharmaceutical composition comprising: a pharmaceutically acceptable carrier or diluent; and antisense, inhibitory RNA, and peptide mimetic compounds targeting EIF3/mov34 and/or GR II.

4. The pharmaceutical composition of claims 1-2 comprising mifepristone or its enantiomers and isomers at dosage levels effective in treating Herpes Simplex Virus (Types 1 and 2).

5. The pharmaceutical composition of claims 1-2 comprising Compound Dl or its enantiomers and isomers at dosage levels effective in treating Herpes Simplex Virus (Types 1 and 2).

6. The pharmaceutical composition of claims 1-2 comprising Compound D2 or its enantiomers and isomers at dosage levels effective in treating Herpes Simplex Virus (Types 1 and 2).

7. The pharmaceutical composition of claims 1-2 comprising Compound D3 or its enantiomers and isomers at dosage levels effective in treating Herpes Simplex Virus (Types 1 and 2).

8. The pharmaceutical composition of claims 1-2 comprising Compound D4 or its enantiomers and isomers at dosage levels effective in treating Herpes Simplex Virus (Types 1 and 2).

9. The pharmaceutical composition of claims 1-2 comprising Composition D5 or its enantiomers and isomers at dosage levels effective in treating Herpes Simplex Virus (Types 1 and 2).

10. The pharmaceutical composition of claims 1-2 comprising Composition D6 or its enantiomers and isomers at dosage levels effective in treating Herpes Simplex Virus (Types 1 and 2).

11. The pharmaceutical composition of claims 1-2 comprising Composition D7 or its enantiomers and isomers at dosage levels effective in treating Herpes Simplex Virus (Types 1 and 2).

12. The pharmaceutical composition of claims 1-2 comprising Composition D8 or its enantiomers and isomers at dosage levels effective in treating Herpes Simplex Virus (Types 1 and 2). The pharmaceutical composition of claims 1-2 comprising Composition D9.

13. The pharmaceutical composition of claims 1-2 comprising Composition D9 or its enantiomers and isomers at dosage levels effective in treating Herpes Simplex Virus (Types 1 and 2).

14. The pharmaceutical composition of claims 1-2 comprising Composition DlO or its enantiomers and isomers at dosage levels effective in treating Herpes Simplex Virus (Types 1 and 2).

15. The pharmaceutical composition of claims 1-2 comprising Composition DI l or its enantiomers and isomers at dosage levels effective in treating Herpes Simplex Virus (Types 1 and 2).

16. The pharmaceutical composition of claims 1-2 comprising Composition D12 or its enantiomers and isomers at dosage levels effective in treating Herpes Simplex Virus (Types 1 and 2).

17. The pharmaceutical composition of claims 1-2 comprising Composition D13 or its enantiomers and isomers at dosage levels effective in treating Herpes Simplex Virus (Types 1 and 2).

18. The pharmaceutical composition of claims 1-2 comprising Composition D 14 or its enantiomers and isomers at dosage levels effective in treating Herpes Simplex Virus (Types 1 and 2).

19. The pharmaceutical composition of claims 1-2 comprising Composition Dl 5 or its enantiomers and isomers at dosage levels effective in treating Herpes Simplex Virus (Types 1 and 2).

20. The pharmaceutical composition of claims 1-2 comprising Composition D16 or its enantiomers and isomers at dosage levels effective in treating Herpes Simplex Virus (Types 1 and 2).

21. The pharmaceutical composition of claims 1-2 comprising Composition Dl 7 or its enantiomers and isomers at dosage levels effective in treating Herpes Simplex Virus (Types 1 and 2).

22. The pharmaceutical composition of claims 1-2 comprising Composition Dl 8 or its enantiomers and isomers at dosage levels effective in treating Herpes Simplex Virus (Types 1 and 2).

23. The pharmaceutical composition of claims 1-2 comprising Composition D19 Monodemethylated metabolite of mifepristone or its enantiomers and isomers at dosage levels effective in treating Herpes Simplex Virus (Types 1 and 2).

24. The pharmaceutical composition of claims 1-2 comprising Composition D20 Didemethylated metabolite of mifepristone or its enantiomers and isomers at dosage levels effective in treating Herpes Simplex Virus (Types 1 and 2).

25. The pharmaceutical composition of claims 1-2 comprising Composition D21 Hydroxylated metabolite of mifepristone or its enantiomers and isomers at dosage levels effective in treating Herpes Simplex Virus (Types 1 and 2).

26. The pharmaceutical composition of claims 4-25 further comprising a compound having a structure selected from the group consisting: acyclovir, ganciclovir, foscarnet, lamivudine, ribavirin, interferon alpha-2a, interferon alpha-2b, peginterferon alfa-2a, and peginterferon alfa-2b.

27. The pharmaceutical composition of claims 1-25 comprising: a pharmaceutically acceptable carrier or diluent; and, a compound having a structure selected from the group consisting of mifepristone, Formulas Dl -Dl 8, and pharmaceutically acceptable salts thereof and further comprising a compound having a structure selected from the group consisting: acyclovir, valcyclovir, famciclovir, ganciclovir, foscarnet, lamivudine, ribavirin, interferon alpha-2a, interferon alpha-2b, peginterferon alfa-2a, and peginterferon alfa-2b.

28. A method of treating an individual who is infected with HSV comprising the step of administering to said individual a therapeutically effective amount of a composition according to claims 1-25.

29. A method of preventing HSV infection in an individual identified as being a high risk individual, the method comprising the step of administering to said individual a prophylactically effective amount of a composition according to claims 1-25.

30. A method of using mifepristone and pharmaceutically acceptable salts thereof and formulated as a transdermal patch for treating individuals infected with Herpes Simplex Virus (Types 1 and 2).

31. A method of using mifepristone and pharmaceutically acceptable salts thereof and formulated as a subdermal delivery system for treating individuals infected with Herpes Simplex Virus (Types 1 and 2).

32. A method of using mifepristone and pharmaceutically acceptable salts thereof and formulated as a controlled/sustained release formulation for treating individuals infected with Herpes Simplex Virus (Types 1 and 2).

33. A method of using a combination of compounds having a structure selected from the group consisting of mifepristone and/or Formulas D1-D21, and pharmaceutically acceptable salts thereof and formulated as a transdermal patch for treating individuals infected with Herpes Simplex Virus (Types 1 and 2).

34. A method of using a combination of compounds having a structure selected from the group consisting of mifepristone and/or Formulas D1-D21, and pharmaceutically acceptable salts thereof and formulated as a subdermal delivery system for treating individuals infected with Herpes Simplex Virus (Types 1 and 2).

5. A method of using a combination of compounds having a structure selected from the group consisting of mifepristone and/or Formulas D1-D21, and pharmaceutically acceptable salts thereof and formulated as a controlled/sustained release formulation for treating individuals infected with Herpes Simplex Virus (Types 1 and 2).

Description:

HSV Drugs

CROSS-REFERENCE TO RELATED APPLICATION

The present application claims priority to U.S. Provisional Application No. 60/687,839, filed June 6, 2005, which is hereby incorporate by reference in its entirety.

SUMMARY OF THE INVENTION

The present invention further relates to pharmaceutical composition comprising: a pharmaceutically acceptable carrier or diluent; and, a compound that inhibits HSV replication, the compound having a structure selected from the group consisting of mifepristone, Formula Dl, Formula D2, Formula D3, Formula D4, Formula D5, and pharmaceutically acceptable salts thereof.

The present invention further relates to methods of treating an individual who has been infected with HSV. The method comprise the step of administering to the individual an amount of a pharmaceutical composition comprising a pharmaceutically acceptable carrier or diluent, and, a compound that inhibits HSV replication having a structure selected from the group consisting of mifepristone, Formula Dl, Formula D2, Formula D3, Formula D4, Formula D5, and pharmaceutically acceptable salts thereof effective to inhibit HSV replication in the individuals.

The present invention further relates to methods of preventing HSV infection in an individual at an elevated risk of becoming HSV infected. The method comprise the step of administering to the individual a prophylactically effective amount of a pharmaceutical composition that comprises a pharmaceutically acceptable carrier or diluent, and, a compound that inhibits HSV replication having a structure selected from the group consisting of mifepristone, Formula Dl, Formula D2, Formula D3, Formula D4, Formula D5, and pharmaceutically acceptable salts thereof effective to inhibit HSV replication.

DETAILED DESCRIPTION OF PREFERRED EMBODIMENTS

The present invention provides pharmaceutical compositions comprising a compound having a structure selected from the group consisting of mifepristone, Formulad Dl -D 18 and pharmaceutically acceptable salts thereof. The present invention provides methods of treating individuals infected with HSV by administering to them a therapeutically effective amount of such compositions. The present invention further provides methods of preventing HSV infection in individuals exposed to HSV 5 by administering to them a prophylactically effective amount of such compositions.

The present invention is useful to therapeutically treat an individual identified as infected with HSV in order to eliminate, reduce or stabilize viral titer. The present invention is useful to prophylactically treat a high risk individual from becoming infected with HSV.

The compounds of the invention may act as steroid hormone receptor antagonists that interactively blocks Rip-l/mov34, alone or in association with one or more steroid receptors, or other components, or one or more steroid receptors alone, preventing or inhibiting formation and translocation of the Rip-1 and/or steroid receptor or other EIF component complex.

As used herein, the term "high risk individual" is meant to refer to an individual who is suspected of having been exposed to the HSV virus. Such individuals include health care or other individuals who may have accidently exchanged blood with an HSV-infected individual, such as through an accidental needle stick, injuries that occur during emergency medical care, rescue or arrest and unprotected sexual contact. High risk individuals can be treated prophylactically before any detection of HSV infection can be made.

As used herein, the term "therapeutically effective amount" is meant to refer to an amount of a compound which produces a medicinal effect observed as reduction or reverse in viral titer when a therapeutically effective amount of a compound is administered to an individual who is infected with HSV. Therapeutically effective amounts are typically determined by the effect they have compared to the effect observed when a composition which includes no active ingredient is administered to a similarly situated individual.

As used herein, the term "prophylactically effective amount" is meant to refer to an amount of a compound which produces a medicinal effect observed as the prevention of HSV infection in an individual when a prophylactically effective amount of a compound is administered to a high risk individual. Prophylactically effective amounts are typically determined by the effect they have compared to the effect observed when a composition which includes no active ingredient is administered to a similarly situated individual.

The invention provides novel pharmaceutical compositions comprising antiviral compounds that are inhibitors of HSV replication. The antiviral compounds included in the pharmaceutical compositions of the present invention have a formula selected from the group consisting of mifepristone and Formulas Dl -D 18 as set forth below, or a pharmaceutically acceptable salt thereof at doses effective in treating HSV. The invention provides novel pharmaceutical compositions comprising antiviral compositions that inhibit HSV replication. In some preferred embodiments, the HSV replication inhibitor in the pharmaceutical compositions of the present invention has a formula of mifepristone as set forth in the section below entitled Formulae. In some preferred embodiments, the HSV replication inhibitor in the pharmaceutical compositions of the present invention has a formula of Formula 1 as set forth in the section below entitled Formulae. In some preferred embodiments, the HSV replication inhibitor in the pharmaceutical compositions of the present invention has a formula of Formula 2 as set forth in the section below entitled Formulae. In some preferred embodiments, the HSV replication inhibitor in the pharmaceutical compositions of the present invention has a formula of Formula 3 as set forth in the section below entitled Formulae. In some preferred embodiments, the HSV replication inhibitor in the pharmaceutical compositions of the present invention has a formula of Formula 4 as set forth in the section below entitled Formulae. In some preferred embodiments, the HSV replication inhibitor in the pharmaceutical compositions of the present invention has a formula of Formula 5 as set forth in the section below entitled Formulae. In some preferred embodiments, the HSV replication inhibitor in the pharmaceutical compositions of the present invention has a formula of Formula 6 as set forth in the section below entitled Formulae. In some

preferred embodiments, the HSV replication inhibitor in the pharmaceutical compositions of the present invention has a formula of Formula 7 as set forth in the section below entitled Formulae. In some preferred embodiments, the HSV replication inhibitor in the pharmaceutical compositions of the present invention has a formula of Formula 8 as set forth in the section below entitled Formulae. In some preferred embodiments, the HSV replication inhibitor in the pharmaceutical compositions of the present invention has a formula of Formula 10 as set forth in the section below entitled Formulae. In some preferred embodiments, the HSV replication inhibitor in the pharmaceutical compositions of the present invention has a formula of Formula 5 as set forth in the section below entitled Formulae. In some preferred embodiments, the HSV replication inhibitor in the pharmaceutical compositions of the present invention has a formula of Formula 11 as set forth in the section below entitled Formulae. In some preferred embodiments, the HSV replication inhibitor in the pharmaceutical compositions of the present invention has a formula of Formula 12 as set forth in the section below entitled Formulae. In some preferred embodiments, the HSV replication inhibitor in the pharmaceutical compositions of the present invention has a formula of Formula 13 as set forth in the section below entitled Formulae. In some preferred embodiments, the HSV replication inhibitor in the pharmaceutical compositions of the present invention has a formula of Formula 14 as set forth in the section below entitled Formulae. In some preferred embodiments, the HSV replication inhibitor in the pharmaceutical compositions of the present invention has a formula of Formula 15 as set forth in the section below entitled Formulae. In some preferred embodiments, the HSV replication inhibitor in the pharmaceutical compositions of the present invention has a formula of Formula 16 as set forth in the section below entitled Formulae. In some preferred embodiments, the HSV replication inhibitor in the pharmaceutical compositions of the present invention has a formula of Formula 17 as set forth in the section below entitled Formulae. In some preferred embodiments, the HSV replication inhibitor in the pharmaceutical compositions of the present invention has a formula of Formula 18 as set forth in the section below entitled Formulae.

In some embodiments the method of the invention additionally includes the use of the HSV replication inhibitor compositions of the invention in combination with other methodologies to treat HSV infection. In some embodiments, the HSV

replication inhibitor is administered in conjunction with other antiviral agents such as acyclovir, ganciclovir, foscarnet, lamivudine, ribavirin, interferon alpha-2a, interferon alpha-2b, peginterferon alfa-2a, and peginterferon alfa-2b.

The pharmaceutical compositions comprising HSV replication inhibitor compositions of the present invention may be administered by any means that enables the active agent to reach the agent's site of action in the body of the individual. Pharmaceutical compositions of the present invention may be administered by conventional routes of pharmaceutical administration. Pharmaceutical compositions may be administered parenterally, i.e. intravenous, subcutaneous, intramuscular, subdermally, transdermally. In some embodiments, the pharmaceutical compositions are administered orally. Pharmaceutical compositions are administered to the individual for a length of time effective to eliminate, reduce or stabilize viral titer. When used prophylactically, Pharmaceutical compositions are administered to the individual for a length of time during which monitoring for evidence of infection continues.

Pharmaceutical compositions of the present invention may be administered either as individual therapeutic agents or in combination with other therapeutic agents. They can be administered alone, but are generally administered with a pharmaceutical carrier selected on the basis of the chosen route of administration and standard pharmaceutical practice.

Dosage varies depending upon known factors such as the pharmacodynamic characteristics of the particular agent, and its mode and route of administration; age, health, and weight of the recipient; nature and extent of symptoms, kind of concurrent treatment, frequency of treatment, and the effect desired. Usually a daily dosage of active ingredient can be about 0.001 to 1 grams per kilogram of body weight, in some embodiments about 0.1 to 100 milligrams per kilogram of body weight. Ordinarily dosages are in the range of 0.5 to 50 milligrams per kilogram of body weight, and preferably 1 to 10 milligrams per kilogram per day. In some embodiments, the pharmaceutical compositions are given in divided doses 1 to 6 times a day or in sustained release form is effective to obtain desired results.

Dosage forms (composition) suitable for internal administration generally contain from about 1 milligram to about 500 milligrams of active ingredient per unit. In these pharmaceutical compositions the active ingredient will ordinarily be present in an amount of about 0.5-95 by weight based on the total weight of the composition. Generally, multiple administrations are performed.

Pharmaceutical compositions may be formulated by one having ordinary skill in the art with compositions selected depending upon the chosen mode of administration. Suitable pharmaceutical carriers are described in Remington's Pharmaceutical Sciences, A. Osol, a standard reference text in this field, which is incorporated herein by reference.

For parenteral administration, the compound can be formulated as a solution, suspension, emulsion or lyophilized powder in association with a pharmaceutically acceptable parenteral vehicle. Examples of such vehicles are water, saline, Ringer's solution, dextrose solution, and 5% human serum albumin. Liposomes and nonaqueous vehicles such as fixed oils may also be used. The vehicle or lyophilized powder may contain additives that maintain isotonicity (e.g., sodium chloride, mannitol) and chemical stability (e.g., buffers and preservatives). The formulation is sterilized by commonly used techniques. In some embodiments, a parenteral composition suitable for administration by injection is prepared by dissolving 1.5% by weight of active ingredient in 0.9% sodium chloride solution.

According to some embodiments of the present invention, the composition is administered to tissue of an individual by topically or by lavage. The compounds may be formulated as a cream, ointment, salve, douche, suppository or solution for topical administration or irrigation. The compounds may be formulated as a transdermal patch or subdermal implants. Formulations for such routes administration of pharmaceutical compositions are well known. Generally, additives for isotonicity can include sodium chloride, dextrose, mannitol, sorbitol and lactose.

In some cases, isotonic solutions such as phosphate buffered saline are used. Stabilizers include gelatin and albumin. In some embodiments, a vasoconstriction agent is added to the formulation. The pharmaceutical preparations according to the present invention are preferably provided sterile and pyrogen free. The pharmaceutical

preparations according to the present invention which are to be used as injectables are provided sterile, pyrogen free and particulate free.

A pharmaceutically acceptable formulation will provide the active ingredient(s) in proper physical form together with such excipients, diluents, stabilizers, preservatives and other ingredients as are appropriate to the nature and composition of the dosage form and the properties of the drug ingredient(s) in the formulation environment and drug delivery system.

In some embodiments, the invention relates to methods of treating patients suffering from HSV infection. In some embodiments, the invention relates to methods of preventing HSV infection in high risk individuals.

According to some embodiments of the invention, the patient is treated with other antiviral therapy in conjunction the administration of pharmaceutical compositions according to the invention. The use of multiple therapeutic approaches provides the patient with a broader based intervention.

According to some aspects of the present invention, in combination with administration of the composition that comprises the HSV replication inhibitor, the individual is also administered another agent. In some embodiments, in combination with administration of the composition, the individual additionally receives compositions that comprises acyclovir, valcyclovir, famciclovir, ganciclovir, foscarnet, lamivudine, ribavirin, interferon alpha-2a, interferon alpha-2b, peginterferon alfa-2a, and peginterferon alfa-2b.

Other antivirals may also be used delivered according to standard protocols using standard agents, dosages and regimens. In some embodiments, the pharmaceutical compositions contain one or more of the compounds selected from the group consisting of mifepristone, Formulas Dl -D 18, and pharmaceutically acceptable salts thereof. In some embodiments, the pharmaceutical compositions contain one or more of the compounds selected from the group consisting of mifepristone, Formulas Dl- D 18, and pharmaceutically acceptable salts thereof and at least one additional antiviral selected from the group consisting of: acyclovir, valcyclovir, famciclovir, ganciclovir, foscarnet, lamivudine, ribavirin, interferon alpha-2a, interferon alpha-2b, peginterferon alfa-2a, and peginterferon alfa-2b, together with a pharmaceutically acceptable carrier.

The pharmaceutical compositions according to the present invention may be administered as a single doses or in multiple doses. The pharmaceutical compositions of the present invention may be administered either as individual therapeutic agents or in combination with other therapeutic agents. The treatments of the present invention may be combined with conventional therapies, which may be administered sequentially or simultaneously.

In addition to treating HSV-infected individual, the present invention relates to methods of preventing HSV infection in high risk individuals who, for example, are suspected of having been exposed to the virus.

Additionally, the present invention is particularly useful to prevent recurrence of infection in patients who have been previously diagnosed as HSV positive but show no indication of infection.

Those having ordinary skill in the art can readily identify high risk individuals. Healthcare workers come into contact with infected blood and suffer needle sticks from syringes used on HSV infected individuals. Surgeons cut themselves during surgery. Lab workers, dentists and dental technicians come into contact with infected blood as do emergency medical and rescue workers and law enforcement officers. Individuals involved in athletics and sexually active individuals can also become exposed to the virus. Once any person comes into contact with infected blood, that individual is at an elevated risk of infection.

The present invention is not limited to any particular theory or mechanism of action and while it is currently believed that the compounds identified herein operate through blocking the steroid hormone receptor complex that comprises Rip-l/mov34, such explanation of the mechanism of action is not intended to limit the invention. The present invention is further illustrated by the following examples, which are not intended to be limiting in any way.

The unbound Mifepristone is metabolized by two-step demethylation or by hydroxylation, and the initial metabolic steps are catalysed by the cytochrome P450 (CYP) enzyme CYP3A4 (Reilly et al, 1999). Three metabolites of Mifepristone have been identified (Sarkar, 2002). This compound undergoes demethylation to produce

mono-demethylated and di-demethylated derivatives as well as hydroxylation of the propynyl group to yield hydroxylated metabolite. Studies have shown that the metabolism of Mifepristone to mono-demethylated and hydroxylated metabolites was rapid but removal of the second methyl group leading to the formation of di- demethylated derivative occurred much more slowly and to much lesser extent than removal of the first. Serum levels of the monodemethylated metabolite always exceeded those of Mifepristone (Sarkar, 2002). The concentrations of the didemethylated and hydroxylated metabolites equalled or exceeded those of Mifepristone when the ingested dose was 400 mg or more. Monodemethylation and hydroxylation were rapid high- capacity reactions, whereas didemethylation was a lower-capacity reaction (Sarkar, 2002).

In each group of different dosage, positive correlations were found between the individual mean alpha 1-acid glycoprotein (AAG) concentrations and the peak concentration of Mifepristone measured at 1-2 h, versus the plateau concentration of Mifepristone measured at 6 h. The in-vitro studies showed that AAG was saturated by Mifepristone concentrations exceeding 2.5 microM. In serum at 40 nM and 2.5 microM Mifepristone concentrations, 2.7% and 2.4%, respectively, of Mifepristone was not protein bound. These results suggest that AAG regulates in part the serum concentrations of Mifepristone, and Mifepristone exceeding the specific serum transport capacity is effectively metabolized.

Like Mifepristone, these metabolites are immunologically and biologically active and retain anti-progestational and anti-glucocorticoid properties. The relative binding affinities of the metabolites to the human glucocorticoid receptor are 61, 48 and 45% for the monodemethylated, hydroxylated, and didemethylated metabolites, respectively; each was higher than that of dexamethasone or Cortisol (23%).

EXAMPLES

1) Drugs which disrupt mov34, a member of EIF3 complex can be used as a treatment for HSV infection.

2) EIF3/ICP27 complex is important for viral protein translation of herpes simplex virus (HSV) families and other viruses.

3) GRII antagonist drugs, including Mifepristone, can target and disrupt function/structure of mov34, a member of EIF3 complex.

4) Use of other drug compounds to block/inhibit EIF3/mov34 (antisense, antibodies, inhibitory RNA)

5) Delivery of drugs via patch, sustained release, and subdermal delivery for treating HSV infection.

Herpes Simplex Virus (HSV) ICP-27 is important for viral protein translation

HSV gene expression is characterized by a temporal pattern of expression of three gene classes: immediate early (IE), early (E), and late (L) genes. IE genes are transcribed in the absence of de novo viral protein synthesis, E genes are activated by IE gene products, and L genes are activated by viral DNA synthesis (reviewed in Roizman and Knipe, 2001). The IE-infected cell protein 27 (ICP27) is essential for viral replication and expression of certain early and nearly all late viral genes (Rice et al., 1989, Sacks et al., 1985 and Uprichard and Knipe, 1996). ICP27 is a multi-functional protein in that it increases late viral gene transcription (Jean et al., 2001), binds to RNA (Mears and Rice, 1996), associates with RNA pol II (Zhou and Knipe, 2002), and shuttles from the nucleus to the cytoplasm (Mears and Rice, 1998 and Soliman et al., 1997). ICP27 has been shown to associate with cellular transcriptional proteins (Taylor and Knipe, 2004 and Zhou and Knipe, 2002), as well as viral transcriptional proteins ICP4 (Panagiotidis et al., 1997) and ICP8 (Taylor and Knipe, 2004 and Zhou and Knipe, 2002), and function in post- transcriptional processes, such as pre-mRNA splicing and niRNA export, through its interactions with cellular splicing and export factors involved in these pathways (Koffa et al., 2001). ICP27 directly affects the expression and stability of specific viral and cellular transcripts in both transfected (Brown et al., 1995) and infected cells (Cheung et al., 2000, Ellison et al., 2000 and Pearson et al., 2004). Furthermore, ICP27 is thought to function, along with the virion host shut-off (vhs) protein, in shut-off of cellular protein synthesis (Sacks et al., 1985 and Song et al., 2001), and the involvement of ICP27 in inhibition of pre-mRNA splicing provides a mechanism for shut-off of cellular protein synthesis (Sandri-Goldin, 1998). Proteomic studies involving immunoprecipitation of ICP27 and mass spectrometric identification of co-precipitated proteins show an

association of ICP27 with the cellular translation initiation factors poly A binding protein (PABP), eukaryotic initiation factor 3 (eIF3), and eukaryotic initiation factor 4G (eIF4G) in infected cells (Fontaine-Rodriguez et al, 2004). Immunoprecipitation-western blot studies confirmed these associations. Finally, purified MBP-tagged ICP27 (MBP-27) can interact with eIF3 subunits p47 and pi 16 in vitro. These results show that ICP27 may play a role in stimulating translation of certain viral and host mRNAs and/or in inhibiting host mRNA translation.

EIF3 is important for viral protein translation

The interaction of eIF4G and PABP is thought to facilitate the interaction between the 5' cap and 3' polyadenylated end of the mRNA, which enhances translation both in vitro and in vivo, and facilitates recruitment of the 4OS ribosomal subunit to the 5' end of the mRNA molecule [(reviewed in Prevot et al., 2003) and (Sonenberg and Dever, 2003)]. eIF3 is a multi-subunit component of the 4OS ribosome, and interaction of eIF4G with eIF3 leads to recruitment of mRNA to the 43 S complex (reviewed (Gallie, 2002). Thus, the interaction of ICP27 with both eIF3 and PABP could lead to the recruitment of these translation initiation factors to viral mRNA and stimulation of translation of these mRNAs. Moreover, both PABP and eIF3 p47 subunit have been shown to localize to both the cytoplasm and the nucleus (Afonina et al., 1998 and Shi et al., 2003). Therefore, ICP27 could recruit these proteins to nascent viral transcripts, which may facilitate viral mRNA export out of the nucleus, and increase the efficiency of translational initiation on these mRNAs.

PABP, eIF3, and eIF4G are known targets for modification by viruses. These cellular translation factors are altered by specific viral proteins, and as a result, host cell protein synthesis is shut down (reviewed in Bushell and Sarnow, 2002 and Daughenbaugh et al., 2003). Translation initiation factor eIF4G acts as a scaffolding protein for the cap-binding complex (eIF4F), and interacts with multiple translation initiation proteins including PABP and eIF3 (reviewed in Kawaguchi and Bailey-Serres, 2002). Furthermore, each of these translation initiation factors have been shown to function in viral translation

regulatory mechanisms, which require specific binding to viral proteins (reviewed in GaIHe 5 2002).

GRII antagonist drugs can disrupt function/structure of mov34, a member of EIF3 complex

Using a yeast two-hybrid system, the cDNA of a Vpr-interacting cellular factor, termed human Vpr Interacting Protein (hVIP/mov34) was cloned (Mahalingam et al., 1998) hVIP/mov34 has complete homology with a reported member of the eIF3 complex (Asano et al., 1997). eIF3 is a large multimeric complex that regulates transcriptional events and is essential for Gl/S and G2/M phase progression through the cell cycle. hVIP is thought to be a GR-responsive protein. Experimental results strongly suggest that hVIP is associated with the activated glucocorticoid receptor complex.

Glucocorticoids regulate diverse functions and are important to maintain central nervous system, cardiovascular, metabolic, and immune homeostasis. They also exert antiinflammatory and immunosuppressive effects, which have made them invaluable therapeutic agents in numerous diseases (Chrousos, 1995). The actions of these hormones are mediated by their specific intracellular receptors, such as the GR. Several host co- activators of the GR have been described that directly interact with GR and components of the transcription initiation complex to enhance the glucocorticoid signal to the transcription machinery (Shibata et al., 1997).

The GR is the prototypic member of the translocating class of steroid receptors that are ubiquitously expressed in almost all human tissues and organs. Unliganded GR is found in the cytoplasm and moves rapidly into the nucleus in response to hormone stimulation (Htun et al., 1996, McNally et al., 2000). GR interacts in the cytoplasm with a complex array of chaperone proteins, including HSP90 and HSP70, and ligand-dependent displacement of these proteins is thought to be intimately involved in the translocation process (Bamberger et al., 1996, Beato et al., 1996). Both GR and hVIP are known Vpr ligands. Steroid hormone receptor antagonists such as mifepristone prevent the GR from

moving into the nucleus in response to appropriate stimulation. In addition, mifepristone blocks the Vpr-induced nuclear entry of hVIP. HVIP had been reported as a potential Vpr ligand and demonstrated its role in cell cycle regulation as antisense of this gene induced cell cycle arrest at the G2/M phase (Mahalingam et al., 1998).

Glucocorticoids have been demonstrated to mimic the effects of Vpr in HIV infection; Glucocorticoid antagonist mifepristone has been shown to revert these effects of Vpr (Ayyavoo et al., 1997, Ayyavoo et al., 2002, Kino et al., 1999, Sherman et al., 2000). Moreover, mifepristone has been shown to block the nuclear translocation of hVIP induced by Vpr in cells by inhibiting GR as a complex with hVIP/mov34. Published results demonstrate that mifepristone inhibits the translocation of hVIP induced by the expression of Vpr and strongly suggested that mifepristone and other GR antagonists can directly affect hVIP/mov34. Taking this further, these results show that mifepristone and other GR antagonist compounds could bind to hVIP/mov34/EIF3 and block its interaction with ICP-27 protein of HSV. In addition, these results implicate the use of other drug compounds to block/inhibit EIF3/mov34 (antisense, antibodies, inhibitory RNA) as a potential treatment for viral pathogens like Herpes Simplex Virus (HSV).

Transdermal Drug Delivery

The skin is the largest and most accessible organ of the human body. The permeability of the skin and its ability to deliver drugs to the blood circulation makes it an ideal drug delivery route. Transdermal drug delivery is an increasingly important method of drug administration. Transdermal drug delivery devices typically involve a carrier (such as a liquid, gel, or solid matrix, or a pressure sensitive adhesive) into which the drug to be delivered is incorporated. The drug-containing carrier is then placed on the skin and the drug, along with any adjuvants and excipients, is delivered to the skin.

Typically the portions of the carrier that are not in contact with the skin are covered by a backing. The backing serves to protect the carrier (and the components contained in the carrier, including the drug) from the environment and prevents loss of the ingredients of the drug delivery device to the environment. Backing materials that have found use in

transdermal drug delivery devices include metal foils, metalized plastic films, and single layered and multilayered polymeric films.

Transdermal drug delivery utilizes the skin for the delivery of the drug molecules from the surface of the skin, through its layers, to the circulatory system. The transdermal drug delivery technology comprises of a controlling system that regulates the rate of drug delivery to the skin, and another that uses the skin to control the absorption rate.

Transdermal drug delivery occurs in two ways: passive and active transdermal delivery. Passive systems allow the drug to diffuse through the skin into the bloodstream using a simple concentration gradient as a driving force. Active delivery system requires a physical force to facilitate the movement of drug molecules across the skin.

The first transdermal patch was introduced in 1981. Subsequently, the applications of transdermal drug delivery have been expanded to include more products in multiple therapeutic areas. Numerous kinds of medications have been administered through the use of a patch, notably scopolamine for preventing motion sickness, nicotine derivatives intended to discourage an addicted smoker from continuing the smoking habit and estrogen hormones.

Prior art teaches us methods to load and deliver drugs via transdermal routes. U.S. Patent No. 5,223,261 describes a loading and using a transdermal delivery system for delivering estradiol. U.S. Patent No. 5,380,760 describes a transdermal delivery system for delivering prostaglandin. U.S. Patent No. 5,702,720 describes a transdermal delivery system for delivering flurbiprofen. U.S. Patent No. 6,132,760 describes a transdermal delivery system for delivering testosterone.

The amount of drug that constitutes a therapeutically effective amount varies according to the condition being treated, any drugs being coadministered with the drug, desired duration of treatment, the surface area and location of the skin over which the device is to be placed, and the selection of adjuvant and other components of the transdermal delivery

device. Accordingly, it is not practical to enumerate particular preferred amounts but such can be readily determined by those skilled in the art with due consideration of these and other appropriate factors. Generally, however, the drug is present in the adhesive layer in an amount of about 2 to about 9 percent, preferably about 2.5 to about 6.5 percent, by weight based on the total weight of the adhesive layer. A device of the invention preferably contains a therapeutically effective amount of the drug dissolved in the adhesive layer.

The adhesive layer of the device of the invention also comprises one or more polymers, typically one or more copolymers. The polymer(s) utilized in the practice of the invention should be substantially chemically inert to the drug, and is preferably a pressure sensitive skin adhesive. Examples of suitable types of adhesives include acrylates, natural and synthetic rubbers, polysiloxanes, polyurethanes, and other pressure sensitive skin adhesives known in the art, either alone or in combination. Preferably the adhesive is an acrylate copolymer.

Delivery of Mifepristone/GR II Antagonists via Transdermal Patch

The present invention provides transdermal drug delivery devices containing mifepristone, Compositions D1-D5 or other GRII antagonists (Drugs). The drug is present in the adhesive layer in a therapeutically effective amount, i.e., an amount effective to allow the device to deliver sufficient amount of the drug to achieve a desired therapeutic result in the treatment of a condition.

A delivery of mifepristone via a transdermal patch would reduce the number of drugs a patient must take orally and improve compliance. The transdermal drug delivery would be most appropriate in cases where low systemic and steady state drug concentration is desirable. This delivery method could enhance patient compliance and could reduce the effects of potential drug toxicities.

There are several advantages of delivering anti-viral drugs via transdermal delivery systems. Transdermal drug delivery is not subjected to first-pass effect and does not

cause frequent drag concentration alterations as compared to the drugs delivered through the oral route. This reduces the required dose in comparison to the oral drug delivery. Medications delivered via skin patches avoid liver metabolism and hence allow for lower doses of medication. It also avoids potential toxicity of the drag to the liver. The transdermal drag delivery also offers the flexibility of terminating the drag administration by simply removing the patch from the skin. This delivery system releases a controlled amount of a drag over a long period of time. Transdermal patch systems exhibit slow controlled drag release and absorption and the plasma drag concentration does not vary significantly over time. This delivery method would enhance patient compliance and thereby a reduction of drag resistant viruses as well as reduce the effects of potential drag toxicities.

Subdermal Drug Delivery (Implantable Devices)

A principal advantage of employing sustained-release compositions is that many therapeutic agents would otherwise be rapidly metabolized or cleared from the patient's system necessitating frequent administration of the drag to maintain a therapeutically effective concentration.

Accordingly, a variety of sustained release devices have been designed for oral, rectal and subcutaneous administration. "Matrix" type devices typically consist of an active compound dispersed in a matrix of carrier material which may be either porous or non- porous, solid or semi-solid, and permeable or impermeable to the active compound. These devices are rather easily prepared; however, they are not suitable for administering some pharmacologically active compounds. In addition, the rate of release of the active compound decreases with time. "Reservoir" type devices consist of a central reservoir of active compound surrounded by a rate controlling membrane (rcm). The rcm is generally a porous or a non-porous material which is non-biodegradable. In the case of the transdermal devices of this type, to maintain an effective concentration of active compound, the rate controlling membrane must have a large surface area. Thus, a common disadvantage of these devices is that their large size makes administration quite inconvenient. Other sustained release devices are hybrid-type devices which contain a

matrix core surrounded by a rcm. Yet other devices are mechanical in nature, and include active compound-filled electrical or osmotic pumps.

The subdermally implantable devices of the present invention can be prepared in a variety of sizes and shapes to accommodate such factors as the specific implantation site and the desired release rate of the drug. In a preferred embodiment wherein the drug is a contraceptive agent, the device is substantially cylindrical in shape having a preferred overall length of from about 4.2 cm to about 4.6 cm, and a preferred overall diameter of from about 2.3 mm to about 2.7 mm. In such a case, the central core is rod-shaped, and has a preferred length of from about 3.8 cm to about 4.2 cm, and a preferred diameter of from about 2.0 mm to about 2.2 mm. These dimensions can be modified depending upon such factors as the implantation site and method of implantation, the subject, the condition to be treated, the drug, and the desired release rate of the drug, etc. For example, the length of the implantable device can be varied to deliver different amounts of the drug.

Prior art teaches us methods to load and deliver drugs via subdermal routes. The subdermally implantable devices according to the present invention can be easily fabricated in accordance with standard techniques. Once the drug is mixed with the matrix material to achieve a substantially uniform dispersion, the desired shape of the resultant dispersion is achieved by molding, casting extrusion, or other appropriate process. When the matrix material contains polymers such as silicone elastomers, an additional curing step may be necessary. The intermediate layer is then applied to the thus-shaped matrix, e.g., by swelling, coating or laminating according to known techniques, a polymeric tube in water and then placing it over the matrix and allowing the polymer to dry in place, or by mechanical lapping. The outer layer can likewise be applied in a variety of ways such as by mechanical stretching, swelling or dipping. See, for example, U.S. Pat. Nos. 3,832,252, 3,854,480 and 4,957,119. U.S. Patent No. 5,756,115 describes a loading and using a subdermal delivery system for delivering contraceptives. The dimensions of the implant are also determined on the basis of the

implantation method. The devices of the present invention can be implanted into a subject in accordance with standard procedures.

The present invention provides subdermal drug delivery devices containing mifepristone, Compositions D1-D5 or other GRII antagonists (Drugs). The drug is present in the implantable devices in a therapeutically effective amount, i.e., an amount effective to allow the device to deliver sufficient amount of the drug to achieve a desired therapeutic result in the treatment of a condition.

Sustained and Controlled Release Drug Delivery

To improve the effectiveness of drug therapy and to reduce possible systematic side effects, many attempts have been made to deliver drugs in a controlled profile to human patients. The advantages of controlled release dosage forms are well known in both the pharmaceutical and medical sciences. The therapeutic benefits of controlled-release dosage forms include the pharmacokinetic ability to maintain a preplanned blood level of an administered drug over a comparatively longer period of time. The therapeutic benefits include also a simultaneous increase in patient compliance and a reduction in the number of doses of drug administered to a patient.

The prior art made available controlled release dosage that sought to provide a drug release rate profile that matched the blood physiological and chrono-pharmacological requirements needed for therapy. For example, an osmotic dosage form for delivering various drugs to a patient environment of use is presented in U.S. Pat. No. 3,845,770 issued to patentees Theeuwes and Higuchi, and in U.S. Pat. No. 3,916,899 issued to the same patentees. The dosage forms disclosed in these patents are manufactured comprising a wall that surrounds a compartment comprising a drug with an exit in the wall for delivering the drug to a patient. In U.S. Pat. Nos. 4,008,719; 4,014,334; 4,058,122; 4,116,241; and 4,160,452 patentees Theeuwes and Ayer made available dosage forms comprising an inside and an outside wall made of poly(cellulose acylate) for delivering a dosage of drug to a patient in need thereof.

Additional semipermeable polymers comprise acetaldehyde dimethylcellulose acetate; cellulose acetate ethylcarbamate; cellulose acetate methylcarbamate; cellulose diacetate propylcarbamate; cellulose acetate diethylaminoacetate; ethyl acrylate methyl methacrylate, semipermeable polyamide; semipermeable polyurethane; semipermeable sulfonated polystyrene; semipermeable crosslinked selective polymer formed by the coprecipation of a polyanion and polycation, as disclosed in U.S. Pat. Nos. 3,173,876; 3,276,586; 3,541,005; 3,541,006 and 3,546,876; semipermeable polymers as disclosed by Loeb and Sourirajan in U.S. Pat. No. 3,133,132; semipermeable, lightly crosslinked polystyrenes; semipermeable crosslinked poly (sodium styrene sulfonate); semipermeable crosslinked poly (vinylbenzyltrimethyl ammonium chloride); and semipermeable polymers possessing a fluid permeability in the range of 2.5.times.lO.sup.-8 to 5.times.lO.sup.-2 (cm.sup.2 /hr.multidot.atm), expressed per atmosphere of hydrostatic or osmotic pressure difference across the semipermeable exterior wall 12. The polymers are known to the polymer art in U.S. Pat. Nos. 3,845,770; 3,916,899 and 4,160,020; and in Handbook of Common Polymers, by Scott, J. R. and Roff, W. J. 1971, CRC Press, Cleveland, Ohio. Wall 12, in a present manufacture can be coated from a substantially single solvent system, such as acetone if coated from a solution, or water if coated as a dispersion.

The present invention provides delivery of mifepristone, Compositions D1-D5 or other GRII antagonists (Drugs) via a sustained release or controlled release delivery techniques.

References

Ackermann et al., 1984 M. Ackermann, D.K. Braun, L. Pereira and B. Roizman, Characterization of herpes simplex virus 1 alpha proteins 0, 4, and 27 with monoclonal antibodies, J. Virol. 52 (1984), pp. 108-118.

Afonina et al., 1998 E. Afonina, R. Stauber and G.N. Pavlakis, The human poly(A)- binding protein 1 shuttles between the nucleus and the cytoplasm, J. Biol. Chem. 273 (1998), pp. 13015-13021

AIi N, Siddiqui A. 1995. Interaction of polypyrimidine tract-binding protein with the 59 noncoding region of the hepatitis C virus RNA genome and its functional requirement in internal initiation of translation. J Virol 69:6367-6375.

AIi N, Siddiqui A. 1997. The La antigen binds 59 noncoding region of the hepatitis C virus RNA in the context of the initiator AUG codon and stimulates internal ribosome entry site-mediated translation. Proc Natl Acad Sci USA 94:2249-2254.

Asano, K., Vornlocher, H. P., Richter-Cook, N. J., Merricj, W. C, Hinnebusch, A. G., and Hershey, W. B. (1997) J. Biol. Chem. 272, 27042-27052.

Asano et al., 1997 K. Asano, H.P. Vornlocher, NJ. Richter-Cook, W.C. Merrick, A.G. Hinnebusch and J. W. Hershey, Structure of cDNAs encoding human eukaryotic initiation factor 3 subunits. Possible roles in RNA binding and macromolecular assembly, J. Biol. Chem. 272 (1997), pp. 27042-27052.

Ayyavoo, V., Mahboubi, A., Mahalingam, S., Ramalingam, R., Kudchodkar, S., Williams, W. V., Green, D. R., and Weiner, D. B. (1997) Nat. Med. 3, 1117-1123.

Ayyavoo, V., Muthumani, K., Kudchodkar, S. B., Zhang, D., Ramanathan, M. P., Dayes, N. S., Kim, J. J., Sin, J. L, Montaner, L. J., and Weiner, D. B. (2002) Int. Immunol. 14, 13-22.

Bamberger, C. M., Schulte, H. M., and Chrousos, G. P. (1996) Endocr. Rev. 17, 245-261. Beato, M., and Sanchez-Pacheco, A. (1996) Endocr. Rev. 17, 587-609.

Blackwell, JL and Brinton, MA. 1997. Translation elongation factor- 1 alpha interacts with the 3' stem-loop region of West Nile Virus genomic RNA. J Virol 71:6433-6444.

Brown et al., 1995 CR. Brown, M.S. Nakamura, J.D. Mosca, G.S. Hayward, S.E. Straus and L.P. Perera, Herpes simplex virus trans-regulatory protein ICP27 stabilizes and binds to 3' ends of labile niRNA, J. Virol. 69 (1995), pp. 7187-7195.

Brown ES, Zhang H, Ping LH, Lemon SM. 1992. Secondary structure of the 59 nontranslated regions of hepatitis C virus and pestivirus genomic RNAs. Nucleic Acids Res 20:5041-5045.

Buratti E, Tisminetzky S 5 Zotti M, Baralle FE. 1998. Functional analysis of the interaction between HSV 59 UTR and putative subunits of eukaryotic initiation factor eIF3. Nucleic Acids Res 26:3179-3187.

Bushell and Sarnow, 2002 M. Bushell and P. Sarnow, Hijacking the translation apparatus by RNA viruses, J. Cell Biol. 158 (2002), pp. 395-399.

Chen et al., 2002 LH. Chen, K.S. Sciabica and R.M. Sandri-Goldin, ICP27 interacts with the RNA export factor Aly/REF to direct herpes simplex virus type 1 intronless mRNAs to the TAP export pathway, J. Virol. 76 (2002), pp. 12877-12889. Cheung et al., 2000 P. Cheung, K.S. Ellison, R. Verity and J.R. Smiley, Herpes simplex virus ICP27 induces cytoplasmic accumulation of unspliced polyadenylated alpha-globin pre-mRNA in infected HeLa cells, J. Virol. 74 (2000), pp. 2913-2919.

Chrousos, G. P. (1995) N. Engl. J. Med. 332, 1351-1362.

Croxatto, HB, Salvatierra AM, Croxatto, HD, Fuentealba B. Effects of continuous treatment with low dose mifepristone throughout one menstrual cycle. Human Reprod. 1992. 7:945-50.

Daughenbaugh et al., 2003 K.F. Daughenbaugh, CS. Fraser, J. W. Hershey and M.E. Hardy, The genome-linked protein VPg of the Norwalk virus binds eIF3, suggesting its role in translation initiation complex recruitment, EMBO J. 22 (2003), pp. 2852-2859.

Daughenbaugh, KF. et al. 2003. EMBO J. The genome-linked protein VPg of the Norwalk virus binds eIF3, suggesting its role in translation initiation complex recruitment. 22:2852-2859.

Fletcher, SP et al. 2002. Pestivirus internal ribosome entry site (IRES) structure and function: Elements in the 5' untranslated region important for IRES function. J Virol 76: 5024-5033.

Ellison et al., 2000 K.S. Ellison, S.A. Rice, R. Verity and J.R. Smiley, Processing of alpha-globin and ICPO mRNA in cells infected with herpes simplex virus type 1 ICP27 mutants, J. Virol. 74 (2000), pp. 7307-7319.

Foldesi, L, G. Falkay and L. Kovacs, Determination of RU486 (mifepristone) in blood by radioreceptor assay: a pharmacokinetic study. Contraception 56 (1996), pp. 27-32.

Fontaine-Rodriguez et al, 2004 TJ. Taylor, M. Olesky and D.M. Knipe, Proteomics of herpes simplex virus infected cell protein 27: association with translation initiation factors, J. Virol. 330 (2004), pp. 487-492. Abstract-

Fukushi S, Okada M, Kageyama T, Hoshino FB, Katayama K. 1999. Specific interaction of a 25-kilodalton cellular protein, a 4OS ribosomal subunit protein, with the internal ribosome entry site of hepatitis C virus genome. Virus Genes 19:153-161.

Gallie, 2002 D.R. Gallie, Protein-protein interactions required during translation, Plant MoI. Biol. 50 (2002), pp. 949-970.

Hardwicke and Sandri-Goldin, 1994 M. A. Hardwicke and R.M. Sandri-Goldin, The herpes simplex virus regulatory protein ICP27 contributes to the decrease in cellular mRNA levels during infection, J. Virol. 68 (1994), pp. 4797-4810.

Heikinheimo, O., Kekkone R. Dose-response relationships of RU486. Ann Med. 1993. 25: 71-6.

Heikinheimo, O., Kontula K, H. Croxatto Spitz I, Luukkainen T, Lahteenmaki P Pharmacokinetics of the antiprogestin RU486 in women during multiple doses administration. J. Steroid Biochem 32 (1989), pp. 22-25.

Htun, H., Barsony, J., Renyi, L, Gould, D. L., and Hager, G. L. (1996) Proc. Natl. Acad. Sci. U. S. A. 93, 4845-4850.

Honda M, Beard MR, Ping LH, Lemon SM. 1999. A phylogenetically conserved stem- loop structure at the 59 border of the internal ribosome entry site of hepatitis C virus is required for cap-independent viral translation. J Virol 73 : 1165-1174.

Honda M, Brown EA, Lemon SM. 1996a. Stability of a stem-loop involving the initiator AUG controls the efficiency of internal initiation of translation on hepatitis C virus RNA. RNA 2:955-968.

Honda M, Ping LH, Rijnbrand RC, Amphlett E, Clarke B, Rowlands D, Lemon SM. 1996b. Structural requirements for initiation of translation by internal ribosome entry within genome-length hepatitis C virus RNA. Virology 222:31-42.

Jang et al.1988. A segment of the 5' nontranslated region of encephalomyocarditis virus RNA directs internal entry of ribosomes during in vitro translation, J. Virol. 62: 2636- 2643.

Jean et al, 2001 S. Jean, K.M. Le Van, B. Song, M. Levine and D.M. Knipe, Herpes simplex virus 1 ICP27 is required for transcription of two viral late (gamma2) genes in infected cells, Virology 283 (2001), pp. 273-284.

Kawaguchi and Bailey-Serres, 2002 R. Kawaguchi and J. Bailey-Serres, Regulation of translational initiation in plants, Curr. Opin. Plant Biol. 5 (2002), pp. 460-465.

Kekkonen, R., O. Heikinheimo, E. Mandelin and P. Lahteenmaki, Pharmacokinetics of mifepristone after low oral doses. Contraception 54 (1996), pp. 229-234.

Kino, T., Gragerov, A., Kopp, J. B., Stauber, R. H., Pavlakis, G. N., and Chrousos, G. P. (1999) J. Exp. Med. 189, 51-62.

Kieft JS, Zhou K, Jubin R, Murray MG, Lau JY, Doudna JA. 1999. The hepatitis C virus internal ribosome entry site adopts an ion-dependent tertiary fold. J MoI Biol 292:513-529.

Koffa et al., 2001 M.D. Koffa, J.B. Clements, E. Izaurralde, S. Wadd, S.A. Wilson, LW. Mattaj and S. Kuersten, Herpes simplex virus ICP27 protein provides viral mRNAs with access to the cellular mRNA export pathway, EMBO J. 20 (2001), pp. 5769-5778.

Le SY, Sonenberg N, Maizel JV Jr. 1995. Unusual folded regions and ribosome landing pad within hepatitis C virus and pestivirus RNAs. Gene 154:137-143.

Mahalingam, S., Ayyavoo, V., Patel, M., Kieber-Emmons, T., Kao, G. D., Muschel, R. J., and Weiner, D. B. (1998) Proc. Natl. Acad. Sci. U. S. A. 98, 3419-3424.

Malancha, TA, and Sudhanshu, V., 2000. Mov34 protein from mouse brain interacts with the 3' noncoding region of Japanese encephalitis virus. J Virol 74: 5108-5115.

McLauchlan et al., 1992 J. McLauchlan, A. Phelan, C. Loney, R.M. Sandri-Goldin and J.B. Clements, Herpes simplex virus IE63 acts at the posttranscriptional level to stimulate viral mRNA 3' processing, J. Virol. 66 (1992), pp. 6939-6945.

McNaIIy, J. G., Muller, W. G., Walker, D., Wolford, R., and Hager, G. L. (2000) Science 287, 1262-1265.

Mears and Rice, 1996 W.E. Mears and S.A. Rice, The RGG box motif of the herpes simplex virus ICP27 protein mediates an RNA-binding activity and determines in vivo methylation, J. Virol. 70 (1996), pp. 7445-7453.

Mears and Rice, 1998 W.E. Mears and S.A. Rice, The herpes simplex virus immediate- early protein ICP27 shuttles between nucleus and cytoplasm, Virology 242 (1998), pp. 128-137.

Methot et al., 1997 N. Methot, E. Rom, H. Olsen and N. Sonenberg, The human homologue of the yeast Prtl protein is an integral part of the eukaryotic initiation factor 3 complex and interacts with pi 70, J. Biol. Chem. 272 (1997), pp. 1110-1116.

Kieft JS, Zhou K, Jubin R, Doudna JA. 2001. Mechanism of ribosome recruitment by hepatitis C virus IRES. RNA. 7:194-206.

O'Connor JB, Brian DA 2000. Virology. 269:172-82.

Panagiotidis et al., 1997 CA. Panagiotidis, E.K. Lium and SJ. Silverstein, Physical and functional interactions between herpes simplex virus immediate-early proteins ICP4 and ICP27, J. Virol. 71 (1997), pp. 1547-1557.

Pearson et al., 2004 A. Pearson, D.M. Knipe and D.M. Coen, ICP27 selectively regulates the cytoplasmic localization of a subset of viral transcripts in herpes simplex virus type 1- infected cells, J. Virol. 78 (2004), pp. 23-32.

Perkins et al., 2003 K.D. Perkins, J. Gregonis, S. Borge and S.A. Rice, Transactivation of a viral target gene by herpes simplex virus ICP27 is posttranscriptional and does not require the endogenous promoter or polyadenylation site, J. Virol. 77 (2003), pp. 9872- 9884.

Pestova TV, Shatsky IN, Fletcher SP, Jackson RJ, Hellen CUT. 1998. Aprokaryotic-like mode of cytoplasmic eukaryotic ribosome binding to the initiation codon during internal translation initiation of hepatitis C and classical swine fever virus RNAs. Genes & Dev 12:67-83.

Pelletier, J., Sonenberg, N. 1988. Internal initiation of translation of eukaryotic mRNA directed by a sequence derived from poliovirus RNA. Nature. 334: 320-325.

Pickering JM, Thomas HC, Karayiannis P. 1997. Predicted secondary structure of the hepatitis G virus and GB virus-A 59 untranslated regions consistent with an internal ribosome entry site. J Viral Hepat 4:175-184.

Prevot et al., 2003 D. Prevot, JX. Darlix and T. Ohlmann, Conducting the initiation of protein synthesis: the role of eIF4G, Biol. Cell 95 (2003), pp. 141-156.

Psaridi L, Georgopoulou U, Varaklioti A, Mavromara P. 1999. Mutationalanalysis of a conserved tetraloop in the 59 untranslated region of hepatitis C virus identifies a novel RNA element essential for the internal ribosome entry site function. FEBS Lett 453:49- 53.

Raman S, Bouma P, Williams GD, Brian DA. 2003. J Virol. 77:6720-30.

Rice and Knipe, 1990 S.A. Rice and D.M. Knipe, Genetic evidence for two distinct transactivation functions of the herpes simplex virus alpha protein ICP27, J. Virol. 64 (1990), pp. 1704-1715.

Rice et al., 1989 S.A. Rice, L.S. Su and D.M. Knipe, Herpes simplex virus alpha protein ICP27 possesses separable positive and negative regulatory activities, J. Virol. 63 (1989), pp. 3399-3407.

Rijnbrand R, Bredenbeek P, van der Straaten T, Whetter L, Inchauspe G, Lemon S, Spaan W. 1995. Almost the entire 59 non-translated region of hepatitis C virus is required for cap-independent trans-lation. FEBS Lett 365:115-119.

Rijnbrand RC, Lemon SM. 2000. Internal ribosome entry site-mediated translation in hepatitis C virus replication. Curr Top Microbiol Immunol 242:85-116.

Roizman and Knipe, 2001 B. Roizman and D.M. Knipe, Herpes simplex viruses and their replication In: D.M. Knipe and P.M. Howley, Editors, Fields Virology (4th ed.), Lippincott, Williams and Wilkins, Philadelphia, PA (2001), pp. 2399-2460.

Sacks et al., 1985 W.R. Sacks, CC. Greene, D.P. Aschman and P. A. Schaffer, Herpes simplex virus type 1 ICP27 is an essential regulatory protein, J. Virol. 55 (1985), pp. 796-805.

Sandri-Goldin, 1998 R.M. Sandri-Goldin, ICP27 mediates HSV RNA export by shuttling through a leucine-rich nuclear export signal and binding viral intronless RNAs through an RGG motif, Genes Dev. 12 (1998), pp. 868-879.

Sarkar, N. N.. Mifepristone: bioavailability, pharmacokinetics and use-effectiveness. 2002. Euro J Obst Gyneco and Repro Bio. 101 :113-120.

Sherman, M. P., de Noronha, C. M., Pearce, D., and Greene, W. C. (2000) J. Virol. 2000 74, 8159-8165.

Shi et al., 2003 J. Shi, Y. Feng, A.C. Goulet, R.R. Vaillancourt, N.A. Sachs, J.W. Hershey and M. A. Nelson, The p34cdc2-related cyclin-dependent kinase 11 interacts with the p47 subunit of eukaryotic initiation factor 3 during apoptosis, J. Biol. Chem. 278 (2003), pp. 5062-5071.

Shibata, H., Spencer, T. E., Onate, T. E., Genster, S. Y., Tsai, S. Y., Tsai, M. J., and O'Malley, B. W. (1997) Recent Prog. Horm. Res. 52, 141-164.

Sizova DV, Kolupaeva VG, Pestova TV, Shatsky IN, Hellen CUT. 1998. Specific interaction of eukaryotic translation initiation factor 3 with the 59 nontranslated regions of hepatitis C virus and classicals wine fever virus RNAs. J Virol 72:4775-4782.

Soliman et al., 1997 T.M. Soliman, R.M. Sandri-Goldin and SJ. Silverstein, Shuttling of the herpes simplex virus type 1 regulatory protein ICP27 between the nucleus and cytoplasm mediates the expression of late proteins, J. Virol. 71 (1997), pp. 9188-9197.

Sonenberg and Dever, 2003 N. Sonenberg and T.E. Dever, Eukaryotic translation initiation factors and regulators, Curr. Opin. Struct. Biol. 13 (2003), pp. 56-63.

Song et al., 2001 B. Song, K.C. Yeh, JJ. Liu and D.M. Knipe, Herpes simplex virus gene products required for viral infection of expression of Gl -phase functions, Virology 290 (2001), pp. 320-328.

Swahn, M.L., G. Wang, A.R. Aedo, S.Z. Cekan and M. Bygdeman, Plasma levels of anti progestin RU486 following oral administration to non-pregnant and early pregnant women. Contraception 34 (1986), pp. 469^481.

Tang S, Collier AJ, Elliott RM. 1999. Alterations to both the primary and predicted secondary structure of stem-loop IIIc of the hepatitis C virus Ib 59 untranslated region (59UTR) lead to mutants severely defective in translation which cannot be complemented in transby the wild-type 59UTR sequence. J Virol 73:2359-2364.

Taylor and Knipe, 2004 TJ Taylor and D. M. Knipe, Proteomics of herpes simplex virus replication compartments: association of cellular DNA replication, repair, recombination, and chromatin remodeling proteins with ICP8, J. Virol. 78 (2004), pp. 5856-5866.

Uprichard and Knipe, 1996 S.L. Uprichard and D.M. Knipe, Herpes simplex virus ICP27 mutant viruses exhibit reduced expression of specific DNA replication genes, J. Virol. 70 (1996), pp. 1969-1980.

Varaklioti A, Georgopoulou U, Kakkanas A, Psaridi L, Serwe M, Casel-mann WH, Mavromara P. 1998. Mutational analysis of two un-structured domains of the 59 untranslated region of HSV RNA. Biochem Biophys Res Commun 253:678-685.

Wang C, Le SY, AIi N, Siddiqui A. 1995. An RNA pseudoknot is an essential structural element of the internal ribosome entry site located within the hepatitis C virus 59 noncoding region. RNAl:526-537.

Wang C, Sarnow P, Siddiqui A. 1994. A conserved helical element is essential for internal initiation of translation of hepatitis C virus RNA. J Virol 68:7301-7307.

Yen JH, Chang SC, Hu CR, Chu SC, Lin SS, Hsieh YS, Chang MF. 1995. Cellular proteins specifically bind to the 59-noncoding region of hepatitis C Virus RNA. Virology 208:723-732. 206 J.S. Kieft et al.

Zhou and Knipe, 2002 C. Zhou and D.M. Knipe, Association of herpes simplex virus type 1 ICP8 and ICP27 proteins with cellular RNA polymerase II holoenzyme, J. Virol. 76 (2002), pp. 5893-5904.

COMPOUNDS Dl:

Dl: Pregna-4,6-diene-3,20-dione

Sigma Product Number: Rl 9,725-4. MDL Number: MFCDOOl 99858.

D2:

D2: 17-α-ethynyI-17-β-hydroxyestr-5 (10)-En-3-one

Sigma Product Number: Rl 8,844-1. MDL Number: MFCD00199015.

D3:

D4:

D5:

D5: Combination of Hydrocortisone Acetate and Zidovudine

Hydrocortisone Acetate Sigma Product Number: H4126 Zidovudine Sigma Product Number: 11546

D 19: Monodemethylated

D20: Didemethylated